1
|
Mansız-Kaplan B, Kotanoğlu MS, Gürsoy K, Vural S, Koca G, Nacır B, Yumuşak N, Kara H, Yüksel S, Korkmaz M. Evaluation of the effect of pitavastatin on motor deficit and functional recovery in sciatic nerve injury: A CatWalk study. Turk J Phys Med Rehabil 2023; 69:334-343. [PMID: 37674804 PMCID: PMC10478549 DOI: 10.5606/tftrd.2023.11002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 01/12/2023] [Indexed: 09/08/2023] Open
Abstract
Objectives This study aims to investigate the electrophysiological, scintigraphic, and histopathological effects of pitavastatin and its impact on functional status in rats with sciatic nerve injury. Materials and methods A total of 30 Wistar albino rats were divided into three equal groups including 10 rats in each group: sham group (no injury), control group (nerve injury induced), and pitavastatin group (nerve injury induced and 2 mg/kg of pitavastatin administered orally once a day for 21 days). Before and at the end of intervention, quantitative gait analysis with the CatWalk system and sciatic nerve conduction studies were performed. After the intervention, the gastrocnemius muscle was scintigraphically evaluated, and the sciatic nerve was histopathologically examined. Results There was no significant difference in the sciatic nerve conduction before the intervention and Day 21 among the groups (p>0.05). According to the quantitative gait analysis, there were significant differences in the control group in terms of the individual, static, dynamic, and coordination parameters (p<0.05). The histopathological examination revealed a significant difference in the total myelinated axon count and mean axon diameter among the groups (p<0.001). Conclusion Pitavastatin is effective in nerve regeneration and motor function recovery in rats with sciatic nerve injury.
Collapse
Affiliation(s)
- Başak Mansız-Kaplan
- Department of Ergotherapy, Ankara Medipol University, School of Health Sciences, Ankara, Türkiye
| | - Mustafa Sırrı Kotanoğlu
- Department of Anesthesiology and Reanimation, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| | - Koray Gürsoy
- Department of Plastic Reconstructive and Aesthetic Surgery, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| | - Seçil Vural
- Department of Physical Medicine and Rehabilitation, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| | - Gökhan Koca
- Department of Nuclear Medicine, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| | - Barış Nacır
- Department of Physical Medicine and Rehabilitation, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| | - Nihat Yumuşak
- Department of Pathology, Harran University Faculty of Veterinary Medicine, Urfa, Türkiye
| | - Halil Kara
- Department of Pharmacology, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Türkiye
| | - Selcen Yüksel
- Department of Biostatistics, Yıldırım Beyazıt University Faculty of Medicine, Ankara, Türkiye
| | - Meliha Korkmaz
- Department of Nuclear Medicine, University of Health Sciences, Ankara Training and Research Hospital, Ankara, Türkiye
| |
Collapse
|
2
|
Duan H, Jing Y, Li Y, Lian Y, Li J, Li Z. Rehabilitation treatment of multiple sclerosis. Front Immunol 2023; 14:1168821. [PMID: 37090712 PMCID: PMC10117641 DOI: 10.3389/fimmu.2023.1168821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 03/27/2023] [Indexed: 04/09/2023] Open
Abstract
Multiple sclerosis is a slowly progressive disease, immunosuppressants and other drugs can delay the progression and progression of the disease, but the most patients will be left with varying degrees of neurological deficit symptoms, such as muscle weakness, muscle spasm, ataxia, sensory impairment, dysphagia, cognitive dysfunction, psychological disorders, etc. From the early stage of the disease to the stage of disease progression, professional rehabilitation treatment can reduce the functional dysfunction of multiple sclerosis patients, improve neurological function, and reduce family and social burdens. With the development of various new rehabilitation technologies such as transcranial magnetic stimulation, virtual reality technology, robot-assisted gait, telerehabilitation and transcranial direct current stimulation, the advantages of rehabilitation therapy in multiple sclerosis treatment have been further established, and more treatment means have also been provided for patients.
Collapse
|
3
|
Ainatzoglou A, Stamoula E, Dardalas I, Siafis S, Papazisis G. The Effects of PDE Inhibitors on Multiple Sclerosis: a Review of in vitro and in vivo Models. Curr Pharm Des 2021; 27:2387-2397. [PMID: 33655851 DOI: 10.2174/1381612827666210303142356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 02/11/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is a chronic inflammatory and immune-mediated disease, whose current therapeutic means are mostly effective in the relapsing-remitting form of MS, where inflammation is still prominent, but fall short of preventing long term impairment. However, apart from inflammationmediated demyelination, autoimmune mechanisms play a major role in MS pathophysiology, constituting a promising pharmacological target. Phosphodiesterase (PDE) inhibitors have been approved for clinical use in psoriasis and have undergone trials suggesting their neuroprotective effects, rendering them eligible as an option for accessory MS therapy. OBJECTIVE In this review, we discuss the potential role of PDE inhibitors as a complementary MS therapy. METHODS We conducted a literature search through which we screened and comparatively assessed papers on the effects of PDE inhibitor use, both in vitro and in animal models of MS, taking into account a number of inclusion and exclusion criteria. RESULTS In vitro studies indicated that PDE inhibitors promote remyelination and axonal sustenance, while curbing inflammatory cell infiltration, hindering oligodendrocyte and neuronal loss and suppressing cytokine production. In vivo studies underlined that these agents alleviate symptoms and reduce disease scores in MS animal models. CONCLUSION PDE inhibitors proved to be effective in addressing various aspects of MS pathogenesis both in vitro and in vivo models. Given the latest clinical trials proving that the PDE4 inhibitor Ibudilast exerts neuroprotective effects in patients with progressive MS, research on this field should be intensified and selective PDE4 inhibitors with enhanced safety features should be seriously considered as prospective complementary MS therapy.
Collapse
Affiliation(s)
- Alexandra Ainatzoglou
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Eleni Stamoula
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioannis Dardalas
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Spyridon Siafis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgios Papazisis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
4
|
Di Benedetto G, Iannucci LF, Surdo NC, Zanin S, Conca F, Grisan F, Gerbino A, Lefkimmiatis K. Compartmentalized Signaling in Aging and Neurodegeneration. Cells 2021; 10:464. [PMID: 33671541 PMCID: PMC7926881 DOI: 10.3390/cells10020464] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022] Open
Abstract
The cyclic AMP (cAMP) signalling cascade is necessary for cell homeostasis and plays important roles in many processes. This is particularly relevant during ageing and age-related diseases, where drastic changes, generally decreases, in cAMP levels have been associated with the progressive decline in overall cell function and, eventually, the loss of cellular integrity. The functional relevance of reduced cAMP is clearly supported by the finding that increases in cAMP levels can reverse some of the effects of ageing. Nevertheless, despite these observations, the molecular mechanisms underlying the dysregulation of cAMP signalling in ageing are not well understood. Compartmentalization is widely accepted as the modality through which cAMP achieves its functional specificity; therefore, it is important to understand whether and how this mechanism is affected during ageing and to define which is its contribution to this process. Several animal models demonstrate the importance of specific cAMP signalling components in ageing, however, how age-related changes in each of these elements affect the compartmentalization of the cAMP pathway is largely unknown. In this review, we explore the connection of single components of the cAMP signalling cascade to ageing and age-related diseases whilst elaborating the literature in the context of cAMP signalling compartmentalization.
Collapse
Affiliation(s)
- Giulietta Di Benedetto
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Liliana F. Iannucci
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Nicoletta C. Surdo
- Neuroscience Institute, National Research Council of Italy (CNR), 35121 Padova, Italy;
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
| | - Sofia Zanin
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Filippo Conca
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Francesca Grisan
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Biology, University of Padova, 35122 Padova, Italy
| | - Andrea Gerbino
- Department of Biosciences, Biotechnology and Biopharmaceutics, University of Bari, 70121 Bari, Italy;
| | - Konstantinos Lefkimmiatis
- Veneto Institute of Molecular Medicine, Foundation for Advanced Biomedical Research, 35129 Padova, Italy; (L.F.I.); (S.Z.); (F.C.); (F.G.)
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| |
Collapse
|
5
|
Duarte LF, Altamirano-Lagos MJ, Tabares-Guevara JH, Opazo MC, Díaz M, Navarrete R, Muza C, Vallejos OP, Riedel CA, Bueno SM, Kalergis AM, González PA. Asymptomatic Herpes Simplex Virus Type 1 Infection Causes an Earlier Onset and More Severe Experimental Autoimmune Encephalomyelitis. Front Immunol 2021; 12:635257. [PMID: 33679788 PMCID: PMC7928309 DOI: 10.3389/fimmu.2021.635257] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple sclerosis (MS) is an increasingly prevalent progressive autoimmune and debilitating chronic disease that involves the detrimental recognition of central nervous system (CNS) antigens by the immune system. Although significant progress has been made in the last decades on the biology of MS and the identification of novel therapies to treat its symptoms, the etiology of this disease remains unknown. However, recent studies have suggested that viral infections may contribute to disease onset. Interestingly, a potential association between herpes simplex virus type 1 (HSV-1) infection and MS has been reported, yet a direct relationship among both has not been conclusively demonstrated. Experimental autoimmune encephalomyelitis (EAE) recapitulates several aspects of MS in humans and is widely used to study this disease. Here, we evaluated the effect of asymptomatic brain infection by HSV-1 on the onset and severity of EAE in C57BL/6 mice. We also evaluated the effect of infection with an HSV-1-mutant that is attenuated in neurovirulence and does not cause encephalitis. Importantly, we observed more severe EAE in mice previously infected either, with the wild-type (WT) or the mutant HSV-1, as compared to uninfected control mice. Also, earlier EAE onset was seen after WT virus inoculation. These findings support the notion that a previous exposure to HSV-1 can accelerate and enhance EAE, which suggests a potential contribution of asymptomatic HSV-1 to the onset and severity of MS.
Collapse
MESH Headings
- Animals
- Antibodies, Viral/blood
- Asymptomatic Diseases
- Blood-Brain Barrier/metabolism
- Blood-Brain Barrier/virology
- Capillary Permeability
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/virology
- Female
- Herpes Simplex/genetics
- Herpes Simplex/immunology
- Herpes Simplex/metabolism
- Herpes Simplex/virology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/pathogenicity
- Inflammation Mediators/metabolism
- Mice, Inbred C57BL
- Mutation
- Severity of Illness Index
- Time Factors
- Virulence
- Mice
Collapse
Affiliation(s)
- Luisa F. Duarte
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María J. Altamirano-Lagos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge H. Tabares-Guevara
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Ma. Cecilia Opazo
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Máximo Díaz
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Romina Navarrete
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina Muza
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Omar P. Vallejos
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M. Bueno
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Escuela de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
6
|
Hromádka R, Kejík Z, Jakubek M, Kaplánek R, Šandriková V, Urban M, Martásek P, Král V. Pigments from Filamentous Ascomycetes for Combination Therapy. Curr Med Chem 2018; 26:3812-3834. [PMID: 29600749 DOI: 10.2174/0929867325666180330091933] [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: 01/04/2018] [Revised: 03/21/2018] [Accepted: 03/21/2018] [Indexed: 11/22/2022]
Abstract
Filamentous ascomycetes (Neurospora and Monascus) have been studied for a long time because of their production of secondary metabolites such as microbial pigments. The ascomycetes represent an interesting group of compounds with high potential for medicinal applications. Many recent studies have shown their efficacy in the treatment of serious pathological states such as oncological diseases, neurodegenerative diseases and hyperlipidaemia. Nevertheless, the clinical usability of ascomycetes is still limited. However, this problem can be solved by the use of these compounds with combinations of other therapeutic agents. This strategy can suppress their side effects and improve their therapeutic efficacy. Moreover, their co-application can significantly enhance conventional therapies that are used. This review summarizes and discusses the general principles of this approach, introduced and supported by numerous examples. In addition, the prediction of the future potential application of this methodology is included.
Collapse
Affiliation(s)
- Róbert Hromádka
- C2P s.r.o. Jungmannova 101 503 51 Chlumec nad Cidlinou, Czech Republic
| | - Zdeněk Kejík
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague 2, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic.,Department of Analytical Chemistry, Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic.,Department of Analytical Chemistry, Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| | - Viera Šandriková
- C2P s.r.o. Jungmannova 101 503 51 Chlumec nad Cidlinou, Czech Republic
| | - Marian Urban
- Food Research Institute Prague, Radiova 1285/7, 1285/7, Prague 10, Czech Republic
| | - Pavel Martásek
- Department of Pediatrics and Adolescent Medicine, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague 2, Czech Republic
| | - Vladimír Král
- BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, 252 50 Vestec, Czech Republic.,Department of Analytical Chemistry, Faculty of Chemical Engineering, University of Chemistry and Technology, Prague, Technická 5, 166 28 Prague 6, Czech Republic
| |
Collapse
|
7
|
Combined Treatment with Methylprednisolone and Human Bone Marrow-Derived Mesenchymal Stem Cells Ameliorate Experimental Autoimmune Encephalomyelitis. Tissue Eng Regen Med 2018; 15:183-194. [PMID: 30603546 DOI: 10.1007/s13770-017-0101-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 10/16/2017] [Accepted: 11/19/2017] [Indexed: 10/18/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Although advances have been made in the treatment of MS, such as the use of IFN-β, glucocorticoids and stem cells, the therapeutic effects of these treatments are not sufficient. In the present study, we evaluated whether the combination of methylprednisolone (MP) and human bone marrow-derived mesenchymal stem cells (BM-MSCs) could enhance the therapeutic effectiveness in experimental autoimmune encephalomyelitis (EAE), a model for MS. EAE was induced by immunizing C57BL/6 mice with myelin oligodendrocyte glycoprotein 35-55 (MOG 35-55). The immunized mice received an intraperitoneal injection of MP (20 mg/kg), an intravenous injection of BM-MSCs (1 × 106 cells) or both on day 14 after immunization. Combination treatment significantly ameliorated the clinical symptoms, along with attenuating inflammatory infiltration and demyelination, compared to either treatment alone. Secretion of pro-inflammatory cytokines (IFN-γ, TNF-α, IL-17) was significantly reduced, and anti-inflammatory cytokines (IL-4, IL-10) was significantly increased by the combination treatment as compared to either treatment alone. Flow cytometry analysis of MOG-reactivated T cells in spleen showed that combination treatment reduced the number of CD4+CD45+ and CD8+ T cells, and increased the number of CD4+CD25+Foxp3+ regulatory T cells. Furthermore, combination treatment enhanced apoptosis in MOG-reactivated CD4+ T cells, a key cellular subset in MS pathogenesis. Combination treatment with MP and BM-MSCs provides a novel treatment protocol for enhancing therapeutic effects in MS.
Collapse
|
8
|
Saeedi Saravi SS, Saeedi Saravi SS, Arefidoust A, Dehpour AR. The beneficial effects of HMG-CoA reductase inhibitors in the processes of neurodegeneration. Metab Brain Dis 2017; 32:949-965. [PMID: 28578514 DOI: 10.1007/s11011-017-0021-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Statins, cholesterol lowering drugs, have been demonstrated to exert beneficial effects in other conditions such as primary and progressing neurodegenerative diseases beyond their original role. Observation that statins ameliorate the neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS) and cerebral ischemic stroke, the neuroprotective effects of these drugs are thought to be linked to their anti-inflammatory, anti-oxidative, and anti-excitotoxic properties. Despite the voluminous literature on the clinical advantages of 3-hydroxy-3-methylglutaryl Co-enzyme A reductase (HMGCR) inhibitors (statins) in cardiovascular system, the neuroprotective effects and the underlying mechanisms are little understood. Hence, the present review tries to provide a critical overview on the statin-induced neuroprotection, which are presumed to be associated with the ability to reduce cholesterol, Amyloid-β and apolipoprotein E (ApoE) levels, decrease reactive oxygen and nitrogen species (ROS and RNS) formation, inhibit excitotoxicity, modulate matrix metalloproteinases (MMPs), stimulate endothelial nitric oxide synthase (eNOS), and increase cerebral blood perfusion. This review is also aimed to illustrate that statins protect neurons against the neuro-inflammatory processes through balancing pro-inflammatory/anti-inflammatory cytokines. Ultimately, the beneficial role of statins in ameliorating the development of PD, AD, MS and cerebral ischemic stroke has been separately reviewed.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sobhan Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Arefidoust
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Nunley KA, Orchard TJ, Ryan CM, Miller R, Costacou T, Rosano C. Statin use and cognitive function in middle-aged adults with type 1 diabetes. World J Diabetes 2017; 8:286-296. [PMID: 28694929 PMCID: PMC5483427 DOI: 10.4239/wjd.v8.i6.286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 02/17/2017] [Accepted: 05/05/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To test associations between statin use and cognitive impairment in adults with childhood-onset type 1 diabetes (T1D).
METHODS In 2010-13, n = 108 middle-aged participants from ongoing observational Pittsburgh Epidemiology of Diabetes Complications Study underwent neurocognitive assessment (mean age and T1D duration of 49 and 41 years, respectively). All were diagnosed with childhood-onset (i.e., prior to age 18) T1D between 1950 and 1980 and were seen within one year of diagnosis at Children’s Hospital of Pittsburgh. Self-reported statin use (yes/no and if yes, name of statin) was collected biennially from parent study baseline (1986-1988) to time of neurocognitive testing. Logistic regression models tested associations between statin use groups and cognitive impairment (defined as having two or more cognitive test scores 1.5SD or worse than published norms) while linear regression models tested associations between statin use groups and cognitive domain z-scores (domains: Verbal IQ, memory, executive function, psychomotor speed, and visuo-construction). All models controlled for education and age. To address confounding by indication, models were repeated using a propensity score for statin use.
RESULTS Of the 108 participants, 51 reported never using statins. Median duration of statin use among the 57 ever users was 6 years. These 57 ever statin users were split to create two groups (≤ or > median years of statin use): 1-6 years (n = 25), and 7-12 years (n = 32). Compared with never users, using statins 1-6 years tripled the odds of cognitive impairment (OR = 3.16; 95%CI: 0.93-10.72; P = 0.06) and using statins 7-12 years almost quintupled the odds of cognitive impairment (OR = 4.84; 95%CI: 1.63-14.44; P = 0.005). Compared with never users, using statins 1-6 or 7-12 years was related to worse performance in the memory domain (β = -0.52; P = 0.003, and -0.39; P = 0.014, respectively). Adjusting for coronary artery disease, low density lipoprotein cholesterol, and Apo E4 status did not substantially alter results, and none of these covariates were significantly related to cognitive outcomes (all P > 0.05). Propensity score analyses support that associations between poor cognitive outcomes and statin use were not due merely to confounding by indication.
CONCLUSION Statin use was associated with cognitive impairment, particularly affecting memory, in these middle-aged adults with childhood-onset T1D, whom at this age, should not yet manifest age-related memory deficits.
Collapse
|
10
|
Togha M, Jahanshahi M, Alizadeh L, Jahromi SR, Vakilzadeh G, Alipour B, Gorji A, Ghaemi A. Rapamycin Augments Immunomodulatory Properties of Bone Marrow-Derived Mesenchymal Stem Cells in Experimental Autoimmune Encephalomyelitis. Mol Neurobiol 2017; 54:2445-2457. [PMID: 26971291 DOI: 10.1007/s12035-016-9840-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Accepted: 03/04/2016] [Indexed: 12/29/2022]
Abstract
The immunomodulatory and anti-inflammatory properties of bone marrow-derived mesenchymal stem cells (BM-MSCs) have been considered as an appropriate candidate for treatment of autoimmune diseases. Previous studies have revealed that treatment with BM-MSCs may modulate immune responses and alleviate the symptoms in experimental autoimmune encephalomyelitis (EAE) mice, an animal model of multiple sclerosis. Therefore, the present study was designed to examine immunomodulatory effects of BM-MSCs in the treatment of myelin oligodendrocyte glycoprotein (MOG) 35-55-induced EAE in C57BL/6 mice. MSCs were obtained from the bone marrow of C57BL mice, cultured with DMEM/F12, and characterized with flow cytometry for the presence of cell surface markers for BM-MSCs. Following three passages, BM-MSCs were injected intraperitoneally into EAE mice alone or in combination with rapamycin. Immunological and histopathological effects of BM-MSCs and addition of rapamycin to BM-MSCs were evaluated. The results demonstrated that adding rapamycin to BM-MSCs transplantation in EAE mice significantly reduced inflammation infiltration and demyelination, enhanced the immunomodulatory functions, and inhibited progress of neurological impairments compared to BM-MSC transplantation and control groups. The immunological effects of rapamycin and BM-MSC treatments were associated with the inhibition of the Ag-specific lymphocyte proliferation, CD8+ cytolytic activity, and the Th1-type cytokine (gamma-interferon (IFN-γ)) and the increase of Th-2 cytokine (interleukin-4 (IL-4) and IL-10) production. Addition of rapamycin to BM-MSCs was able to ameliorate neurological deficits and provide neuroprotective effects in EAE. This suggests the potential of rapamycin and BM-MSC combined therapy to play neuroprotective roles in the treatment of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Mansoureh Togha
- Iranian Center of Neurological Research, Neuroscience Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Jahanshahi
- Neuroscience Research Center, Department of Anatomy, Faculty of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | | | - Soodeh Razeghi Jahromi
- Shefa Neuroscience Research Center, Tehran, Iran
- Multiple Sclerosis Research Center-Neuroscience Institute, Sina Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Gelareh Vakilzadeh
- School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahram Alipour
- Iranian Blood Transfusion Organization Research Center, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Tehran, Iran
- Epilepsy Research Center, Klinik und Poliklinik für Neurochirurgie, Department of Neurology, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | - Amir Ghaemi
- Infectious Diseases Research Center, Department of Microbiology, Golestan University of Medical Sciences, P.O. Box 49175-1141, Gorgan, Iran.
- Department of Virology, Institute Pasteur of Iran, Tehran, Iran.
| |
Collapse
|
11
|
Tsai YF, Chu TC, Chang WY, Wu YC, Chang FR, Yang SC, Wu TY, Hsu YM, Chen CY, Chang SH, Hwang TL. 6-Hydroxy-5,7-dimethoxy-flavone suppresses the neutrophil respiratory burst via selective PDE4 inhibition to ameliorate acute lung injury. Free Radic Biol Med 2017; 106:379-392. [PMID: 28263828 DOI: 10.1016/j.freeradbiomed.2017.03.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/26/2017] [Accepted: 03/01/2017] [Indexed: 01/11/2023]
Abstract
Over-activated neutrophils produce enormous oxidative stress and play a key role in the development of acute and chronic inflammatory diseases. 6-Hydroxy-5,7-dimethoxy-flavone (UFM24), a flavone isolated from the Annonaceae Uvaria flexuosa, showed inhibitory effects on human neutrophil activation and salutary effects on lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. UFM24 potently inhibited superoxide anion (O2•-) generation, reactive oxidants, and CD11b expression, but not elastase release, in N-formyl-l-methionyl-l-leucyl-l-phenylalanine (fMLF)-activated human neutrophils. However, UFM24 failed to scavenge O2•- and inhibit the activity of subcellular NADPH oxidase. fMLF-induced phosphorylation of protein kinase B (Akt) was inhibited by UFM24. Noticeably, UFM24 increased cyclic adenosine monophosphate (cAMP) concentration and protein kinase (PK) A activity in activated human neutrophils. PKA inhibitors significantly reversed the inhibitory effects of UFM24, suggesting that the effects of UFM24 were through cAMP/PKA-dependent inhibition of Akt activation. Additionally, activity of cAMP-related phosphodiesterase (PDE)4, but not PDE3 or PDE7, was significantly reduced by UFM24. Furthermore, UFM24 attenuated neutrophil infiltration, myeloperoxidase activity, and pulmonary edema in LPS-induced ALI in mice. In conclusion, our data demonstrated that UFM24 inhibits oxidative burst in human neutrophils through inhibition of PDE4 activity. UFM24 also exhibited significant protection against endotoxin-induced ALI in mice. UFM24 has potential as an anti-inflammatory agent for treating neutrophilic lung damage.
Collapse
Affiliation(s)
- Yung-Fong Tsai
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Tzu-Chi Chu
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Yi Chang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan
| | - Yang-Chang Wu
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, Taiwan; Chinese Medicine Research and Development Center and Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Fang-Rong Chang
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, 807 Taiwan
| | - Shun-Chin Yang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anesthesiology, Taipei Veterans General Hospital and National Yang-Ming University, Taipei 112, Taiwan
| | - Tung-Ying Wu
- Chinese Medicine Research and Development Center and Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan
| | - Yu-Ming Hsu
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chun-Yu Chen
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Shih-Hsin Chang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan
| | - Tsong-Long Hwang
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan 333, Taiwan; Department of Anesthesiology, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan; Research Center for Food and Cosmetic Safety, Research Center for Chinese Herbal Medicine, and Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| |
Collapse
|
12
|
Biscola NP, Cartarozzi LP, Ulian-Benitez S, Barbizan R, Castro MV, Spejo AB, Ferreira RS, Barraviera B, Oliveira ALR. Multiple uses of fibrin sealant for nervous system treatment following injury and disease. J Venom Anim Toxins Incl Trop Dis 2017; 23:13. [PMID: 28293254 PMCID: PMC5348778 DOI: 10.1186/s40409-017-0103-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 02/23/2017] [Indexed: 12/14/2022] Open
Abstract
Lesions to the nervous system often produce hemorrhage and tissue loss that are difficult, if not impossible, to repair. Therefore, scar formation, inflammation and cavitation take place, expanding the lesion epicenter. This significantly worsens the patient conditions and impairment, increasing neuronal loss and glial reaction, which in turn further decreases the chances of a positive outcome. The possibility of using hemostatic substances that also function as a scaffold, such as the fibrin sealant, reduces surgical time and improve postoperative recovery. To date, several studies have demonstrated that human blood derived fibrin sealant produces positive effects in different interventions, becoming an efficient alternative to suturing. To provide an alternative to homologous fibrin sealants, the Center for the Study of Venoms and Venomous Animals (CEVAP, Brazil) has proposed a new bioproduct composed of certified animal components, including a thrombin-like enzyme obtained from snake venom and bubaline fibrinogen. Thus, the present review brings up to date literature assessment on the use of fibrin sealant for nervous system repair and positions the new heterologous bioproduct from CEVAP as an alternative to the commercial counterparts. In this way, clinical and pre-clinical data are discussed in different topics, ranging from central nervous system to peripheral nervous system applications, specifying positive results as well as future enhancements that are necessary for improving the use of fibrin sealant therapy.
Collapse
Affiliation(s)
- Natalia Perussi Biscola
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil.,Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil.,Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Luciana Politti Cartarozzi
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Suzana Ulian-Benitez
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil.,Neuro Development Lab, School of Biosciences, University of Birmingham, Birmingham, England UK
| | - Roberta Barbizan
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil.,The School of Medicine at Mucuri (FAMMUC), Federal University of Jequitinhonha and Mucuri Valleys (UFVJM), 39803-371 Teófilo Otoni, MG Brazil
| | - Mateus Vidigal Castro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Aline Barroso Spejo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| | - Rui Seabra Ferreira
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil.,Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
| | - Benedito Barraviera
- Graduate Program in Tropical Diseases, Botucatu Medical School, Univ Estadual Paulista (UNESP), Botucatu, SP Brazil.,Center for the Study of Venoms and Venomous Animals (CEVAP), Univ Estadual Paulista (UNESP), Botucatu, SP Brazil
| | - Alexandre Leite Rodrigues Oliveira
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas (UNICAMP), Laboratory of Nerve Regeneration, CEP 13083-970 Campinas, SP Brazil
| |
Collapse
|
13
|
Titus DJ, Oliva AA, Wilson NM, Atkins CM. Phosphodiesterase inhibitors as therapeutics for traumatic brain injury. Curr Pharm Des 2015; 21:332-42. [PMID: 25159077 DOI: 10.2174/1381612820666140826113731] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 08/25/2014] [Indexed: 11/22/2022]
Abstract
Developing therapeutics for traumatic brain injury remains a challenge for all stages of recovery. The pathological features of traumatic brain injury are diverse, and it remains an obstacle to be able to target the wide range of pathologies that vary between traumatic brain injured patients and that evolve during recovery. One promising therapeutic avenue is to target the second messengers cAMP and cGMP with phosphodiesterase inhibitors due to their broad effects within the nervous system. Phosphodiesterase inhibitors have the capability to target different injury mechanisms throughout the time course of recovery after brain injury. Inflammation and neuronal death are early targets of phosphodiesterase inhibitors, and synaptic dysfunction and circuitry remodeling are late potential targets of phosphodiesterase inhibitors. This review will discuss how signaling through cyclic nucleotides contributes to the pathology of traumatic brain injury in the acute and chronic stages of recovery. We will review our current knowledge of the successes and challenges of using phosphodiesterase inhibitors for the treatment of traumatic brain injury and conclude with important considerations in developing phosphodiesterase inhibitors as therapeutics for brain trauma.
Collapse
Affiliation(s)
| | | | | | - Coleen M Atkins
- Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida, 33136, USA.
| |
Collapse
|
14
|
Mestre L, Redondo M, Carrillo-Salinas FJ, Morales-García JA, Alonso-Gil S, Pérez-Castillo A, Gil C, Martínez A, Guaza C. PDE7 inhibitor TC3.6 ameliorates symptomatology in a model of primary progressive multiple sclerosis. Br J Pharmacol 2015; 172:4277-90. [PMID: 25994655 DOI: 10.1111/bph.13192] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Revised: 04/06/2015] [Accepted: 05/13/2015] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND AND PURPOSE cAMP plays an important role in the transduction of signalling pathways involved in neuroprotection and immune regulation. Control of the levels of this nucleotide by inhibition of cAMP-specific PDEs such as PDE7 may affect the pathological processes of neuroinflammatory diseases like multiple sclerosis (MS). In the present study, we evaluated the therapeutic potential of the selective PDE7 inhibitor, TC3.6, in a model of primary progressive multiple sclerosis (PPMS), a rare and severe variant of MS. EXPERIMENTAL APPROACH Theiler's murine encephalomyelitis virus-induced demyelinated disease (TMEV-IDD) is one of the models used to validate the therapeutic efficacy of new drugs in MS. As recent studies have analysed the effect of PDE7 inhibitors in the EAE model of MS, here the TMEV-IDD model was used to test their efficacy in a progressive variant of MS. Mice were subjected to two protocols of TC3.6 administration: on the pre-symptomatic phase and once the disease was established. KEY RESULTS Treatment with TC3.6 ameliorated the disease course and improved motor deficits of infected mice. This was associated with down-regulation of microglial activation and reduced cellular infiltrates. Decreased expression of pro-inflammatory mediators such as COX-2 and the cytokines, IL-1β, TNF-α, IFN-γ and IL-6 in the spinal cord of TMEV-infected mice was also observed after TC3.6 administration. CONCLUSION These findings support the importance of PDE7 inhibitors, and specifically TC3.6, as a novel class of agents with therapeutic potential for PPMS. Preclinical studies are needed to determine whether their effects translate into durable clinical benefits.
Collapse
Affiliation(s)
- L Mestre
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal-CSIC, Madrid, Spain
| | - M Redondo
- Departamento de Química Médica I, Instituto de Química Médica-CSIC, Madrid, Spain
| | - F J Carrillo-Salinas
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal-CSIC, Madrid, Spain
| | - J A Morales-García
- Departamento de Modelos Experimentales de Enfermedades Humanas, Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - S Alonso-Gil
- Departamento de Modelos Experimentales de Enfermedades Humanas, Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - A Pérez-Castillo
- Departamento de Modelos Experimentales de Enfermedades Humanas, Instituto de Investigaciones Biomédicas, CSIC-UAM, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - C Gil
- Departamento Biología Físico-Química, Centro de Investigaciones Biológicas-CSIC, Madrid, Spain
| | - A Martínez
- Departamento Biología Físico-Química, Centro de Investigaciones Biológicas-CSIC, Madrid, Spain
| | - C Guaza
- Departamento de Neurobiología Funcional y de Sistemas, Instituto Cajal-CSIC, Madrid, Spain
| |
Collapse
|
15
|
Poppinga WJ, Muñoz-Llancao P, González-Billault C, Schmidt M. A-kinase anchoring proteins: cAMP compartmentalization in neurodegenerative and obstructive pulmonary diseases. Br J Pharmacol 2014; 171:5603-23. [PMID: 25132049 PMCID: PMC4290705 DOI: 10.1111/bph.12882] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Revised: 07/14/2014] [Accepted: 08/10/2014] [Indexed: 12/25/2022] Open
Abstract
The universal second messenger cAMP is generated upon stimulation of Gs protein-coupled receptors, such as the β2 -adreneoceptor, and leads to the activation of PKA, the major cAMP effector protein. PKA oscillates between an on and off state and thereby regulates a plethora of distinct biological responses. The broad activation pattern of PKA and its contribution to several distinct cellular functions lead to the introduction of the concept of compartmentalization of cAMP. A-kinase anchoring proteins (AKAPs) are of central importance due to their unique ability to directly and/or indirectly interact with proteins that either determine the cellular content of cAMP, such as β2 -adrenoceptors, ACs and PDEs, or are regulated by cAMP such as the exchange protein directly activated by cAMP. We report on lessons learned from neurons indicating that maintenance of cAMP compartmentalization by AKAP5 is linked to neurotransmission, learning and memory. Disturbance of cAMP compartments seem to be linked to neurodegenerative disease including Alzheimer's disease. We translate this knowledge to compartmentalized cAMP signalling in the lung. Next to AKAP5, we focus here on AKAP12 and Ezrin (AKAP78). These topics will be highlighted in the context of the development of novel pharmacological interventions to tackle AKAP-dependent compartmentalization.
Collapse
Affiliation(s)
- W J Poppinga
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| | - P Muñoz-Llancao
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
- Laboratory of Cell and Neuronal Dynamics (Cenedyn), Department of Biology, Faculty of Sciences, Universidad de ChileSantiago, Chile
- Department of Neuroscience, Section Medical Physiology, University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| | - C González-Billault
- Laboratory of Cell and Neuronal Dynamics (Cenedyn), Department of Biology, Faculty of Sciences, Universidad de ChileSantiago, Chile
| | - M Schmidt
- Department of Molecular Pharmacology, University of GroningenGroningen, The Netherlands
- Groningen Research Institute for Asthma and COPD (GRIAC), University Medical Center Groningen, University of GroningenGroningen, The Netherlands
| |
Collapse
|
16
|
Braun D, Madrigal JLM, Feinstein DL. Noradrenergic regulation of glial activation: molecular mechanisms and therapeutic implications. Curr Neuropharmacol 2014; 12:342-52. [PMID: 25342942 PMCID: PMC4207074 DOI: 10.2174/1570159x12666140828220938] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 07/12/2014] [Accepted: 07/16/2014] [Indexed: 01/07/2023] Open
Abstract
It has been known for many years that the endogenous neurotransmitter noradrenaline (NA) exerts anti-inflammatory and neuroprotective effects both in vitro and in vivo. In many cases the site of action of NA are beta-adrenergic receptors (βARs), causing an increase in intracellular levels of cAMP which initiates a broad cascade of events including suppression of inflammatory transcription factor activities, alterations in nuclear localization of proteins, and induction of patterns of gene expression mediated through activity of the CREB transcription factor. These changes lead not only to reduced inflammatory events, but also contribute to neuroprotective actions of NA by increasing expression of neurotrophic substances including BDNF, GDNF, and NGF. These properties have prompted studies to determine if treatments with drugs to raise CNS NA levels could provide benefit in various neurological conditions and diseases having an inflammatory component. Moreover, increasing evidence shows that disruptions in endogenous NA levels occurs in several diseases and conditions including Alzheimer's disease (AD), Parkinson's disease (PD), Down's syndrome, posttraumatic stress disorder (PTSD), and multiple sclerosis (MS), suggesting that damage to NA producing neurons is a common factor that contributes to the initiation or progression of neuropathology. Methods to increase NA levels, or to reduce damage to noradrenergic neurons, therefore represent potential preventative as well as therapeutic approaches to disease.
Collapse
Affiliation(s)
- David Braun
- Department of Anesthesiology, University of Illinois at Chicago, Chicago IL, USA, 60612
| | - Jose L M Madrigal
- Departamento de Farmacología, Universidad Complutense de Madrid, Spain
| | - Douglas L Feinstein
- Department of Anesthesiology, University of Illinois at Chicago, Chicago IL, USA, 60612 ; Jesse Brown VA Medical Center, Chicago IL, USA, 60612
| |
Collapse
|
17
|
Ulivieri C, Baldari CT. Statins: From cholesterol-lowering drugs to novel immunomodulators for the treatment of Th17-mediated autoimmune diseases. Pharmacol Res 2014; 88:41-52. [DOI: 10.1016/j.phrs.2014.03.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 03/04/2014] [Accepted: 03/05/2014] [Indexed: 12/13/2022]
|
18
|
Interferon β-secreting mesenchymal stem cells combined with minocycline attenuate experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 274:20-7. [DOI: 10.1016/j.jneuroim.2014.06.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 05/21/2014] [Accepted: 06/02/2014] [Indexed: 12/30/2022]
|
19
|
González-García C, Bravo B, Ballester A, Gómez-Pérez R, Eguiluz C, Redondo M, Martínez A, Gil C, Ballester S. Comparative assessment of PDE 4 and 7 inhibitors as therapeutic agents in experimental autoimmune encephalomyelitis. Br J Pharmacol 2014; 170:602-13. [PMID: 23869659 DOI: 10.1111/bph.12308] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 07/01/2013] [Accepted: 07/08/2013] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE PDE4 inhibition suppresses experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). However, side effects hinder PDE4 inhibitors clinical use. PDE7 inhibition might constitute an alternative therapeutic strategy, but few data about the anti-inflammatory potential of PDE7 inhibitors are currently available. We have used the EAE model to perform a comparative evaluation of PDE4 and PDE7 inhibition as strategies for MS treatment. EXPERIMENTAL APPROACH Two PDE7 inhibitors, the sulfonamide derivative BRL50481 and the recently described quinazoline compound TC3.6, were assayed to modulate EAE in SJL mice, in comparison with the well-known PDE4 inhibitor Rolipram. We evaluated clinical signs, presence of inflammatory infiltrates in CNS and anti-inflammatory markers. We also analysed the effect of these inhibitors on the inflammatory profile of spleen cells in vitro. KEY RESULTS TC3.6 prevented EAE with efficacy similar to Rolipram, while BRL50481 had no effect on the disease. Differences between both PDE7 inhibitors are discussed. Data from Rolipram and TC3.6 showed that PDE4 and PDE7 inhibition work through both common and distinct pathways. Rolipram administration caused an increase in IL-10 and IL-27 expression which was not found after TC3.6 treatment. On the other hand, both inhibitors reduced IL-17 levels, prevented infiltration in CNS and increased the expression of the T regulator cell marker Foxp3. CONCLUSIONS AND IMPLICATIONS These results provide new information about the effects of Rolipram on EAE, underline PDE7 inhibition as a new therapeutic target for inflammatory diseases and show the value of TC3.6 to prevent EAE, with possible consequences for new therapeutic tools in MS.
Collapse
Affiliation(s)
- C González-García
- Unidad de Regulación Génica, UFIEC, Instituto de Salud Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Maternal pravastatin prevents altered fetal brain development in a preeclamptic CD-1 mouse model. PLoS One 2014; 9:e100873. [PMID: 24963809 PMCID: PMC4071009 DOI: 10.1371/journal.pone.0100873] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 05/30/2014] [Indexed: 12/14/2022] Open
Abstract
Objective Using an animal model, we have previously shown that preeclampsia results in long-term adverse neuromotor outcomes in the offspring, and this phenotype was prevented by antenatal treatment with pravastatin. This study aims to localize the altered neuromotor programming in this animal model and to evaluate the role of pravastatin in its prevention. Materials and Methods For the preeclampsia model, pregnant CD-1 mice were randomly allocated to injection of adenovirus carrying sFlt-1 or its control virus carrying mFc into the tail vein. Thereafter they received pravastatin (sFlt-1-pra “experimental group”) or water (sFlt-1 “positive control”) until weaning. The mFc group (“negative control”) received water. Offspring at 6 months of age were sacrificed, and whole brains underwent magnetic resonance imaging (MRI). MRIs were performed using an 11.7 Tesla vertical bore MRI scanner. T2 weighted images were acquired to evaluate the volumes of 28 regions of interest, including areas involved in adaptation and motor, spatial and sensory function. Cytochemistry and cell quantification was performed using neuron-specific Nissl stain. One-way ANOVA with multiple comparison testing was used for statistical analysis. Results Compared with control offspring, male sFlt-1 offspring have decreased volumes in the fimbria, periaquaductal gray, stria medullaris, and ventricles and increased volumes in the lateral globus pallidus and neocortex; however, female sFlt-1 offspring showed increased volumes in the ventricles, stria medullaris, and fasciculus retroflexus and decreased volumes in the inferior colliculus, thalamus, and lateral globus pallidus. Neuronal quantification via Nissl staining exhibited decreased cell counts in sFlt-1 offspring neocortex, more pronounced in males. Prenatal pravastatin treatment prevented these changes. Conclusion Preeclampsia alters brain development in sex-specific patterns, and prenatal pravastatin therapy prevents altered neuroanatomic programming in this animal model.
Collapse
|
21
|
Kranz K, Warnecke A, Lenarz T, Durisin M, Scheper V. Phosphodiesterase type 4 inhibitor rolipram improves survival of spiral ganglion neurons in vitro. PLoS One 2014; 9:e92157. [PMID: 24642701 PMCID: PMC3958480 DOI: 10.1371/journal.pone.0092157] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Accepted: 02/17/2014] [Indexed: 12/11/2022] Open
Abstract
Sensorineural deafness is caused by damage of hair cells followed by degeneration of the spiral ganglion neurons and can be moderated by cochlear implants. However, the benefit of the cochlear implant depends on the excitability of the spiral ganglion neurons. Therefore, current research focuses on the identification of agents that will preserve their degeneration. In this project we investigated the neuroprotective effect of Rolipram as a promising agent to improve the viability of the auditory neurons. It is a pharmaceutical agent that acts by selective inhibition of the phosphodiesterase 4 leading to an increase in cyclic AMP. Different studies reported a neuroprotective effect of Rolipram. However, its significance for the survival of SGN has not been reported so far. Thus, we isolated spiral ganglion cells of neonatal rats for cultivation with different Rolipram concentrations and determined the neuronal survival rate. Furthermore, we examined immunocytologically distinct proteins that might be involved in the neuroprotective signalling pathway of Rolipram and determined endogenous BDNF by ELISA. When applied at a concentration of 0.1 nM, Rolipram improved the survival of SGN in vitro. According to previous studies, our immunocytological data showed that Rolipram application induces the phosphorylation and thereby activation of the transcription factor CREB. This activation can be mediated by the cAMP-PKA-signalling pathway as well as via ERK as a part of the MAP-kinase pathway. However, only in cultures pre-treated with BDNF, an endogenous increase of BDNF was detected. We conclude that Rolipram has the potential to improve the vitality of neonatal auditory nerve cells in vitro. Further investigations are necessary to prove the effect of Rolipram in vivo in the adult organism after lesion of the hair cells and insertion of cochlear implants.
Collapse
Affiliation(s)
- Katharina Kranz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Martin Durisin
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| | - Verena Scheper
- Department of Otolaryngology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
22
|
Carver AR, Tamayo E, Perez‐Polo JR, Saade GR, Hankins GD, Costantine MM. The effect of maternal pravastatin therapy on adverse sensorimotor outcomes of the offspring in a murine model of preeclampsia. Int J Dev Neurosci 2013; 33:33-40. [DOI: 10.1016/j.ijdevneu.2013.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/10/2013] [Accepted: 11/05/2013] [Indexed: 12/13/2022] Open
Affiliation(s)
- Alissa R. Carver
- University of Texas Medical Branch301 University DriveGalvestonTX77555USA
| | - Esther Tamayo
- University of Texas Medical Branch301 University DriveGalvestonTX77555USA
| | | | - George R. Saade
- University of Texas Medical Branch301 University DriveGalvestonTX77555USA
| | - Gary D.V. Hankins
- University of Texas Medical Branch301 University DriveGalvestonTX77555USA
| | | |
Collapse
|
23
|
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
|
24
|
Medina-Rodríguez EM, Arenzana FJ, Pastor J, Redondo M, Palomo V, García de Sola R, Gil C, Martínez A, Bribián A, de Castro F. Inhibition of endogenous phosphodiesterase 7 promotes oligodendrocyte precursor differentiation and survival. Cell Mol Life Sci 2013; 70:3449-62. [PMID: 23661015 PMCID: PMC11113628 DOI: 10.1007/s00018-013-1340-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 04/05/2013] [Accepted: 04/08/2013] [Indexed: 12/21/2022]
Abstract
During the development of the central nervous system (CNS), oligodendrocyte precursors (OPCs) are generated in specific sites within the neural tube and then migrate to colonize the entire CNS, where they differentiate into myelin-forming oligodendrocytes. Demyelinating diseases such as multiple sclerosis (MS) are characterized by the death of these cells. The CNS reacts to demyelination and by promoting spontaneous remyelination, an effect mediated by endogenous OPCs, cells that represent approximately 5-7 % of the cells in the adult brain. Numerous factors influence oligodendrogliogenesis and oligodendrocyte differentiation, including morphogens, growth factors, chemotropic molecules, extracellular matrix proteins, and intracellular cAMP levels. Here, we show that during development and in early adulthood, OPCs in the murine cerebral cortex contain phosphodiesterase-7 (PDE7) that metabolizes cAMP. We investigated the effects of different PDE7 inhibitors (the well-known BRL-50481 and two new ones, TC3.6 and VP1.15) on OPC proliferation, survival, and differentiation. While none of the PDE7 inhibitors analyzed altered OPC proliferation, TC3.6 and VP1.15 enhanced OPC survival and differentiation, processes in which ERK intracellular signaling played a key role. PDE7 expression was also observed in OPCs isolated from adult human brains and the differentiation of these OPCs into more mature oligodendroglial phenotypes was accelerated by treatment with both new PDE7 inhibitors. These findings reveal new roles for PDE7 in regulating OPC survival and differentiation during brain development and in adulthood, and they may further our understanding of myelination and facilitate the development of therapeutic remyelination strategies for the treatment of MS.
Collapse
Affiliation(s)
- E. M. Medina-Rodríguez
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca “La Peraleda”, s/n, 45071 Toledo, Spain
| | - F. J. Arenzana
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca “La Peraleda”, s/n, 45071 Toledo, Spain
| | - J. Pastor
- Neurofisiología Clínica, Hospital Universitario La Princesa, Madrid, Spain
| | - M. Redondo
- Instituto de Química Médica, CSIC, Juan de la Cierva, Madrid, Spain
| | - V. Palomo
- Instituto de Química Médica, CSIC, Juan de la Cierva, Madrid, Spain
| | | | - C. Gil
- Instituto de Química Médica, CSIC, Juan de la Cierva, Madrid, Spain
| | - A. Martínez
- Instituto de Química Médica, CSIC, Juan de la Cierva, Madrid, Spain
| | - A. Bribián
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca “La Peraleda”, s/n, 45071 Toledo, Spain
- Institute of Bioengineering of Catalonia, Parc Cientific de Barcelona & Cell Biology Department, Universidad de Barcelona, Barcelona, Spain
| | - F. de Castro
- Grupo de Neurobiología del Desarrollo-GNDe, Hospital Nacional de Parapléjicos, Finca “La Peraleda”, s/n, 45071 Toledo, Spain
| |
Collapse
|
25
|
Cloutier FC, Rouleau DM, Hébert-Davies J, Beaumont PH, Beaumont E. Atorvastatin is beneficial for muscle reinnervation after complete sciatic nerve section in rats. J Plast Surg Hand Surg 2013; 47:446-50. [PMID: 23848426 DOI: 10.3109/2000656x.2013.778205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nerve regeneration and functional recovery are often incomplete after peripheral neurotmetic lesion. Atorvastatin has been shown to be neuroprotective after transient ischaemia or traumatic injury. The aim of this study was to establish if systemic administration of Atorvastatin could improve functional muscle reinnervation after complete sciatic nerve section. Sixteen female Sprague-Dawley rats were used in this study. After a complete right sciatic nerve section, end-to-end microsuture repair was performed and fibrin glue was added. Three groups were studied: (1) sutures (S) + fibrin glue (F) only + saline administration for 14 days; (2) S+F+Atorvastatin administration for 14 days; and (3) uninjured nerve. Five months later, the sciatic nerve and the gastrocnemius muscle were isolated to perform in vivo electrophysiological measurements. Better kinematics was observed in atorvastatin-treated rats 5 months after its administration. Indeed, a larger excursion of the hip-ankle-toe angle during walking was observed. This effect was associated with the preservation of electromyographic activity (2.91 mV vs 0.77 mV) and maximal muscle force (85.1 g vs 28.6 g) on stimulation of the proximal nerve section. Five months after a neurotmetic lesion, the recovery is incomplete when using suture and fibrin glue only. Furthermore, the systemic administration of Atorvastatin for 14 days after lesion was beneficial in improving locomotion capability associated with the re-establishment of muscle strength and EMG activity.
Collapse
Affiliation(s)
- Frédéric-Charles Cloutier
- Centre de Recherche, Hôpital du Sacré-Coeur de Montréal and Département de Chirurgie, Université de Montréal , Montréal, Québec , Canada
| | | | | | | | | |
Collapse
|
26
|
Hou Y, Ryu CH, Park KY, Kim SM, Jeong CH, Jeun SS. Effective combination of human bone marrow mesenchymal stem cells and minocycline in experimental autoimmune encephalomyelitis mice. Stem Cell Res Ther 2013; 4:77. [PMID: 23826999 PMCID: PMC3854709 DOI: 10.1186/scrt228] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Accepted: 07/01/2013] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common inflammatory demyelinating disorder of the central nervous system (CNS). Minocycline ameliorates the clinical severity of MS and exhibits antiinflammatory, neuroprotective activities, and good tolerance for long-term use, whereas it is toxic to the CNS. Recently, the immunomodulation and neuroprotection capabilities of human bone marrow mesenchymal stem cells (hBM-MSCs) were shown in experimental autoimmune encephalomyelitis (EAE). In this study, we evaluated whether the combination of hBM-MSCs and a low-dose minocycline could produce beneficial effects in EAE mice. METHODS The sensitivity of hBM-MSCs to minocycline was determined by an established cell-viability assay. Minocycline-treated hBM-MSCs were also characterized with flow cytometry by using MSC surface markers and analyzed for their multiple differentiation capacities. EAE was induced in C57BL/6 mice by using immunization with MOG35-55. Immunopathology assays were used to detect the inflammatory cells, demyelination, and neuroprotection. Interferon gamma (IFN-γ)/tumor necrosis factor alpha (TNF-α) and interleukin-4 (IL-4)/interleukin-10 (IL-10), the hallmark cytokines that direct Th1 and Th2 development, were detected with enzyme-linked immunosorbent assay (ELISA). terminal dUTP nick-end labeling (TUNEL) staining was performed to elucidate the cell apoptosis in the spinal cords of EAE mice. RESULTS Minocycline did not affect the viability, surface phenotypes, or differentiation capacity of hBM-MSCs, while minocycline affected the viability of astrocytes at a high dose. In vivo efficacy experiments showed that combined treatment, compared to the use of minocycline or hBM-MSCs alone, resulted in a significant reduction in clinical scores, along with attenuation of inflammation, demyelination, and neurodegeneration. Moreover, the combined treatment with hBM-MSCs and minocycline enhanced the immunomodulatory effects, which suppressed proinflammatory cytokines (IFN-γ, TNF-α) and conversely increased anti-inflammatory cytokines (IL-4, IL-10). In addition, TUNEL staining also demonstrated a significant decrease of the number of apoptotic cells in the combined treatment compared with either treatment alone. CONCLUSIONS The combination of hBM-MSCs and minocycline provides a novel experimental protocol to enhance the therapeutic effects in MS.
Collapse
|
27
|
Paintlia AS, Paintlia MK, Mohan S, Singh AK, Singh I. AMP-activated protein kinase signaling protects oligodendrocytes that restore central nervous system functions in an experimental autoimmune encephalomyelitis model. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 183:526-41. [PMID: 23759513 DOI: 10.1016/j.ajpath.2013.04.030] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Revised: 04/22/2013] [Accepted: 04/29/2013] [Indexed: 12/12/2022]
Abstract
AMP-activated protein kinase (AMPK) signaling is reported to protect neurons under pathologic conditions; however, its effect on oligodendrocytes (OLs) remains to be elucidated. We investigated whether AMPK signaling protects OLs to restore central nervous system (CNS) functions in an experimental autoimmune encephalomyelitis (EAE), a murine model of multiple sclerosis. Increased inflammation and demyelination in the CNS and peripheral immune responses were consistent with the observed clinical impairments in EAE animals, which were attenuated by treatment with metformin compared with vehicle. In addition, expressions of neurotrophic factors and of signatory genes of OL lineages were increased in the CNS of metformin-treated EAE animals. Likewise, metformin attenuated inflammatory response and enhanced expressions of neurotrophic factors, thereby protecting OLs via AMPK activation in mixed glial cultures stimulated with lipopolysaccharide/interferon γ in vitro, as evidenced by analysis of the expression of signatory genes of O1(+)/MBP(+) OLs and their cellular populations. Metformin also attenuated oxidative stress and malondialdehyde-containing protein levels, with corresponding induction of antioxidative defenses in OLs exposed to cytokines via AMPK activation. These effects of metformin were evident in the CNS of EAE animals. These data provide evidence that AMPK signaling is crucial to protect OLs and, thus, CNS functions in EAE animals. We conclude that AMPK activators, including metformin, have the potential to limit neurologic deficits in multiple sclerosis and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
28
|
Striatal TH-immunopositive fibers recover after an intrastriatal injection of 6-hydroxydopamine in golden hamsters treated with prednisolone: roles of tumor necrosis factor-α and inducible nitric oxide synthase in neurodegeneration. Neurosci Res 2013; 76:83-92. [PMID: 23471013 DOI: 10.1016/j.neures.2013.02.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 02/20/2013] [Indexed: 11/23/2022]
Abstract
Neuroinflammation has been implicated in the pathology of neurodegenerative processes such as Parkinson's disease (PD). Using the golden hamster (GH) 6-hydroxydopamine (6-OHDA) model, we investigated whether the attenuation of neuroinflammation influences the onset and progression of dopamine cell degeneration. 6-OHDA-injected GH received a treatment of minocycline (MINO), prednisolone (Pred) or a combination of minocycline and prednisolone (MINO+Pred). Immunohistochemistry for tyrosine hydroxylase (TH), Iba-1 and glial fibrillary acidic protein (GFAP) was used to evaluate lesions in the nigrostriatal axis and the amount of activated microglia and astroglia, respectively. RT-PCR was used to measure mRNA levels of cytokines and trophic neuroprotective factors. The three anti-inflammatory treatments dramatically reduced activated microglia in the nigrostriatal axis. In addition, TH-immunostaining showed that the positive areas in the ipsilateral striatum of either MINO or Pred groups were higher than that of control. However, only in Pred group this recovery was significant. mRNA measurements demonstrated lower levels of TNF-α, iNOS, BDNF and GDNF in Pred group when compared with controls. The results suggest that TH-immunopositive fibers have the ability to recover after 6-OHDA-induced toxicity of dopaminergic neurons, and this recovery may be due to a decrease in the microglial production of TNF-α and iNOS.
Collapse
|
29
|
Shunmugavel A, Martin MM, Khan M, Copay AG, Subach BR, Schuler TC, Singh I. Simvastatin ameliorates cauda equina compression injury in a rat model of lumbar spinal stenosis. J Neuroimmune Pharmacol 2013; 8:274-86. [PMID: 23188522 PMCID: PMC3587651 DOI: 10.1007/s11481-012-9419-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Accepted: 11/05/2012] [Indexed: 12/29/2022]
Abstract
Lumbar spinal stenosis (LSS) is the leading cause of morbidity and mortality worldwide. LSS pathology is associated with secondary injury caused by inflammation, oxidative damage and cell death. Apart from laminectomy, pharmacological therapy targeting secondary injury is limited. Statins are FDA-approved cholesterol-lowering drug. They also show pleiotropic anti-inflammatory, antioxidant and neuroprotective effects. To investigate the therapeutic efficacy of simvastatin in restoring normal locomotor function after cauda equina compression (CEC) in a rat model of LSS, CEC injury was induced in rats by implanting silicone gels into the epidural spaces of L4 and L6. Experimental group was treated with simvastatin (5 mg/kg body weight), while the injured (vehicle) and sham operated (sham) groups received vehicle solution. Locomotor function in terms of latency on rotarod was measured for 49 days and the threshold of pain was determined for 14 days. Rats were sacrificed on day 3 and 14 and the spinal cord and cauda equina fibers were extracted and studied by histology, immunofluorescence, electron microscopy (EM) and TUNEL assay. Simvastatin aided locomotor functional recovery and enhanced the threshold of pain after the CEC. Cellular Infiltration and demyelination decreased in the spinal cord from the simvastatin group. EM revealed enhanced myelination of cauda equina in the simvastatin group. TUNEL assay showed significantly decreased number of apoptotic neurons in spinal cord from the simvastatin group compared to the vehicle group. Simvastatin hastens the locomotor functional recovery and reduces pain after CEC. These outcomes are mediated through the neuroprotective and anti-inflammatory properties of simvastatin. The data indicate that simvastatin may be a promising drug candidate for LSS treatment in humans.
Collapse
Affiliation(s)
- Anandakumar Shunmugavel
- Department of Pediatrics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | | | | | | | | | | | | |
Collapse
|
30
|
Paintlia AS, Mohan S, Singh I. Combinatorial Effect of Metformin and Lovastatin Impedes T-cell Autoimmunity and Neurodegeneration in Experimental Autoimmune Encephalomyelitis. ACTA ACUST UNITED AC 2013; 4. [PMID: 24324917 DOI: 10.4172/2155-9899.1000149] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Multiple Sclerosis (MS) is an incurable central nervous system (CNS) demyelinating disease affecting several million people worldwide. Due to the multifactorial and complex pathology of MS, FDA approved drugs often show limited efficacy inpatients. We earlier documented that both lovastatin (cholesterol lowering drug) and metformin (anti-diabetic drug) attenuate experimental autoimmune encephalomyelitis (EAE), a widely used model of MS via different mechanisms of action. Since combination therapy of two or more agents has advantage over monotherapy, we here assessed the therapeutic efficacy of metformin and lovastatin combination in EAE. We found that suboptimal doses of these drugs in combination had additive effect to attenuate established EAE in treated animals than their individual treatments. Histological, immunohistochemistry and western blotting analyses revealed that the observed demyelination and axonal loss as evident from reduced levels of myelin and neurofilament proteins in the spinal cords of EAE animals were attenuated by treatment with these drugs in combination. Accordingly, the observed infiltration of myelin reactive T cells (CD4 and CD8) and macrophages (CD68) as well as the increased expression of their signatory cytokines in the spinal cords of EAE animals were attenuated by this regimen as revealed by enzyme-linked immune-sorbent assay and real-time PCR analyses. In the periphery, this regimen biased the class of elicited anti-myelin basic protein immunoglobulins from IgG2a to IgG1 and IgG2b, suggesting a Th1 to Th2 shift which was further supported by the increased expression of their signatory cytokines in EAE animals. Taken together, these data imply that metformin and lovastatin combination attenuates T-cell autoimmunity and neurodegeneration in treated EAE animals thereby suggesting that the oral administration of these FDA approved drugs in combination has potential to limit MS pathogenesis.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Darby Children's Research Institute, Department of Pediatrics, Medical University of South Carolina Charleston, South Carolina 29425, USA
| | | | | |
Collapse
|
31
|
Zhong D, Liu Z, Xie S, Zhang W, Zhang Y, Xue W. Study on poly(D,L-lactic) microspheres embedded in calcium alginate hydrogel beads as dual drug delivery systems. J Appl Polym Sci 2012. [DOI: 10.1002/app.38797] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
32
|
Yanuck D, Mihos CG, Santana O. Mechanisms and Clinical Evidence of the Pleiotropic Effects of the Hydroxy-Methyl-Glutaryl-CoA Reductase Inhibitors in Central Nervous System Disorders: A Comprehensive Review. Int J Neurosci 2012; 122:619-29. [DOI: 10.3109/00207454.2012.704455] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
|
33
|
Torres KJ, Göttle P, Kremer D, Rivera JF, Aguirre-Cruz L, Corona T, Hartung HP, Küry P. Vinpocetine inhibits oligodendroglial precursor cell differentiation. Cell Physiol Biochem 2012; 30:711-22. [PMID: 22854710 DOI: 10.1159/000341451] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/04/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In multiple sclerosis during periods of remission a limited degree of myelin repair can be observed mediated by oligodendroglial precursor cells. Phosphodiesterase inhibitors act as anti-inflammatory agents and might hold promise for future multiple sclerosis treatment. AIMS To investigate whether phosphodiesterase inhibitors could also influence myelin repair. METHODS We stimulated primary oligodendroglial precursor cells with cilostazol, rolipram and vinpocetine and assessed their effects on repair related cellular processes. RESULTS We found that vinpocetine exerted a strong negative effect on myelin expression while cilostazol and rolipram did not show such effects. In addition, vinpocetine decreased morphological complexities suggesting an overall negative impact on oligodendroglial cell maturation. We provide evidence that this is not mediated via a blockade of phosphodiesterase-1 but rather by inhibition of IĸB kinase. CONCLUSION These findings suggest that vinpocetine via IĸB inhibition exerts a strong negative impact on oligodendroglial cell maturation and may therefore provide the rationale to restrict its application during periods of remission in multiple sclerosis patients. This is of particular interest since vinpocetine is widely used as a health supplement thought to act as a cognitive and memory enhancer for healthy people and patients with neurological or muscle diseases.
Collapse
Affiliation(s)
- Klintsy Julieta Torres
- Heinrich-Heine-University, Medical Faculty, Department of Neurology, Düsseldorf, Germany
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Rolipram promotes remyelination possibly via MEK-ERK signal pathway in cuprizone-induced demyelination mouse. Exp Neurol 2012; 237:304-11. [PMID: 22836144 DOI: 10.1016/j.expneurol.2012.07.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 07/16/2012] [Accepted: 07/18/2012] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Rolipram, a 3'-5'-cyclic adenosine monophosphate (cAMP)-dependent phosphodiesterase 4 (PDE4) inhibitor, has long been studied for its immune modulating effects in the treatment of experimental autoimmune encephalomyelitis (EAE). In the current study, we investigated the effects of rolipram on remyelination after cuprizone- or lysolecithin-induced demyelination and the signal transduction pathways potentially modulating this response. MATERIALS AND METHODS Cuprizone-induced demyelination in mice and lysolecithin (LPC)-induced demyelination in rat cerebellum slice culture were treated with rolipram. Demyelination was evaluated by Luxol fast blue (LFB) or myelin basic protein (MBP) staining and western blot. Oligodendroglial cells were cultured with different concentrations of rolipram, and 2', 3'-cyclic nucleotide phosphodiesterase (CNPase) activity, MBP expression, and extracellular signal-regulated kinase (ERK) phosphorylation were measured. RESULTS Rolipram antagonized lysolecithin (LPC)-induced demyelination in rat cerebellar slice cultures and cuprizone-fed mice. In vitro, rolipram treatment promoted oligodendrocyte precursor cell (OPC) maturation, an effect that was partially blocked by the inhibitors of the mitogen activated protein kinase kinase (MEK). CONCLUSION Rolipram promotes the maturation of OPCs, facilitates remyelination, and increases ERK phosphorylation. All of these actions are involved in an action against cuprizone-induced demyelination that may occur partly via the MEK-ERK pathway. Importantly, this may have therapeutic implications for MS.
Collapse
|
35
|
Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull 2012; 87:10-20. [PMID: 22024597 PMCID: PMC9827422 DOI: 10.1016/j.brainresbull.2011.10.004] [Citation(s) in RCA: 739] [Impact Index Per Article: 61.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/02/2011] [Accepted: 10/10/2011] [Indexed: 01/11/2023]
Abstract
Microglia are activated in response to a number of different pathological states within the CNS including injury, ischemia, and infection. Microglial activation results in their production of pro-inflammatory cytokines such as IL-1, IL-6, and TNF-α. While release of these factors is typically intended to prevent further damage to CNS tissue, they may also be toxic to neurons and other glial cells. Mounting evidence indicates that chronic microglial activation may also contribute to the development and progression of neurodegenerative disorders. Unfortunately, determining the role of pro-inflammatory cytokines in these disorders has been complicated by their dual roles in neuroprotection and neurodegeneration. The purpose of this review is to summarize current understanding of the involvement of cytokines in neurodegenerative disorders and their potential signaling mechanisms in this context. Taken together, recent findings suggest that microglial activation and pro-inflammatory cytokines merit interest as targets in the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Joshua A. Smith
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC 29425, USA
| | - Arabinda Das
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC 29425, USA
| | - Swapan K. Ray
- Department of Pathology, Microbiology, and Immunology, University of South Carolina School of Medicine, Columbia, SC 29209, USA
| | - Naren L. Banik
- Division of Neurology, Department of Neurosciences, Medical University of South Carolina, 96 Jonathan Lucas Street, Suite 309 CSB, P.O. Box 250606, Charleston, SC 29425, USA,Corresponding author. Tel.: +1 843 792 7594; fax: +1 843 792 5137. (N.L. Banik)
| |
Collapse
|
36
|
Van Der Putten C, Kuipers HF, Zuiderwijk-Sick EA, Van Straalen L, Kondova I, Van Den Elsen PJ, Bajramovic JJ. Statins amplify TLR-induced responses in microglia via inhibition of cholesterol biosynthesis. Glia 2011; 60:43-52. [PMID: 21964955 DOI: 10.1002/glia.21245] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2011] [Accepted: 08/29/2011] [Indexed: 02/06/2023]
Abstract
Statins inhibit the endogenous intracellular mevalonate pathway and exposure to statins affects innate and adaptive immune responses. Different statins are currently under evaluation as (co)therapy in neuro-inflammatory diseases like multiple sclerosis. However, there are important discrepancies in the reported effects of statins on innate immune responses in different cell types. Studies to characterize such responses in clinically relevant primary cells are currently lacking. In this study, we investigated the effect of statins on Toll-like receptor (TLR)-induced responses of microglia, the resident macrophages of the central nervous system (CNS). Exposure of primary microglia from adult rhesus monkeys to different statins strongly amplified pro-inflammatory cytokine protein and mRNA levels in response to myeloid differentiation primary response gene 88-dependent TLR activation in particular. Rather than affecting nuclear facor-κB activation levels, statin exposure affected stress-activated protein/Jun-amino-terminal and p38 kinase signaling pathways. Mechanistic studies using specific pathway inhibitors and rescue experiments show that statin-induced inhibition of cholesterol biosynthesis, rather than inhibition of isoprenylation, was mainly responsible for the amplified TLR responses. Additionally, microglia were more sensitive to statin-mediated effects than bone marrow-derived macrophages of the same donor. This correlated to lower intrinsic microglial expression levels of 3-hydroxy-3-methylglutaryl coenzyme A reductase, the enzyme targeted by statins. Amplification of TLR-induced responses in microglia by statin exposure might contribute to the generation of a more pro-inflammatory CNS microenvironment which can be of relevance for the pathogenesis of neuroinflammatory disorders.
Collapse
|
37
|
Saher G, Quintes S, Nave KA. Cholesterol: a novel regulatory role in myelin formation. Neuroscientist 2011; 17:79-93. [PMID: 21343408 DOI: 10.1177/1073858410373835] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Myelin consists of tightly compacted membranes that form an insulating sheath around axons. The function of myelin for rapid saltatory nerve conduction is dependent on its unique composition, highly enriched in glycosphingolipids and cholesterol. Cholesterol emerged as the only integral myelin component that is essential and rate limiting for the development of CNS and PNS myelin. Experiments with conditional mouse mutants that lack cholesterol biosynthesis in oligodendrocytes revealed that only minimal changes of the CNS myelin lipid composition are tolerated. In Schwann cells of the PNS, protein trafficking and myelin compaction depend on cholesterol. In this review, the authors summarize the role of cholesterol in myelin biogenesis and myelin disease.
Collapse
Affiliation(s)
- Gesine Saher
- Max Planck Institute of Experimental Medicine, Neurogenetics, Göttingen, Germany.
| | | | | |
Collapse
|
38
|
Butterfield DA, Barone E, Mancuso C. Cholesterol-independent neuroprotective and neurotoxic activities of statins: perspectives for statin use in Alzheimer disease and other age-related neurodegenerative disorders. Pharmacol Res 2011; 64:180-6. [PMID: 21536132 DOI: 10.1016/j.phrs.2011.04.007] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2011] [Accepted: 04/14/2011] [Indexed: 01/24/2023]
Abstract
Statins, long known to be beneficial in conditions where dyslipidemia occurs by lowering serum cholesterol levels, also have been proposed for use in neurodegenerative conditions, including Alzheimer disease. However, it is not clear that the purported effectiveness of statins in neurodegenerative disorders is directly related to cholesterol-lowering effects of these agents; rather, the pleiotropic functions of statins likely play critical roles. Moreover, it is becoming more apparent with additional studies that statins can have deleterious effects in preclinical studies and lack effectiveness in various recent clinical trials. This perspective paper outlines pros and cons of the use of statins in neurodegenerative disorders, with particular emphasis on Alzheimer disease.
Collapse
Affiliation(s)
- D Allan Butterfield
- Department of Chemistry, Center of Membrane Sciences, University of Kentucky, Lexington, KY 40506, USA; Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506, USA.
| | | | | |
Collapse
|
39
|
Awad AM, Sellner J, Hemmer B, Stüve O. Role of statins in the treatment of multiple sclerosis: an update. Neurodegener Dis Manag 2011. [DOI: 10.2217/nmt.11.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY This article discusses the role of statins in the treatment of multiple sclerosis. Statins are promising agents as adjunctive therapies in immune-mediated disorders such as multiple sclerosis owing to their immunomodulatory, anti-inflammtory and neuroprotective characteristics. On the other hand, there are some conflicting data that warrant further investigation before sound conclusions can be made. Clinical trials are planned and being conducted to help shed more light on the potential benefit and the optimal dose of statins in treating multiple sclerosis.
Collapse
Affiliation(s)
- Amer M Awad
- Baton Rouge Neurology Associates, Baton Rouge General Medical Center, Baton Rouge, LA, USA
| | - Johann Sellner
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Neurology, Christian-Doppler-Klinik, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX, USA
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| |
Collapse
|
40
|
Abstract
The fundamental role of inflammatory immune processes in the pathology of multiple sclerosis (MS) provides the rationale for immunomodulatory therapies that attempt to shift the immune system from pro-inflammatory to anti-inflammatory pathways and induce regulatory mechanisms. Growing understanding of immune cellular and molecular mechanisms together with modern biotechnology engendered promising immunomodulatory treatment strategies, with novel mechanisms of actions and different levels of specificity. These include inhibitory molecules, monoclonal antibodies, cell therapies and agents that are administered orally or by infrequent infusions. Several of these treatments have demonstrated impressive efficacy in Phase II and III clinical trials by reducing disease activity and accumulation of disability. However, with the advent of potent therapies, rare but severe adverse effects, such as CNS infections and malignancies, have occurred. This article describes current and upcoming immunomodulatory strategies for MS therapy. The potential of immunomodulatory treatments to counteract the inflammatory characteristics of MS and support neuroprotective processes is discussed.
Collapse
Affiliation(s)
- Rina Aharoni
- The Department of Immunology, The Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
41
|
Mix E, Meyer-Rienecker H, Hartung HP, Zettl UK. Animal models of multiple sclerosis--potentials and limitations. Prog Neurobiol 2010; 92:386-404. [PMID: 20558237 PMCID: PMC7117060 DOI: 10.1016/j.pneurobio.2010.06.005] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Revised: 06/01/2010] [Accepted: 06/07/2010] [Indexed: 12/17/2022]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is still the most widely accepted animal model of multiple sclerosis (MS). Different types of EAE have been developed in order to investigate pathogenetic, clinical and therapeutic aspects of the heterogenic human disease. Generally, investigations in EAE are more suitable for the analysis of immunogenetic elements (major histocompatibility complex restriction and candidate risk genes) and for the study of histopathological features (inflammation, demyelination and degeneration) of the disease than for screening of new treatments. Recent studies in new EAE models, especially in transgenic ones, have in connection with new analytical techniques such as microarray assays provided a deeper insight into the pathogenic cellular and molecular mechanisms of EAE and potentially of MS. For example, it was possible to better delineate the role of soluble pro-inflammatory (tumor necrosis factor-α, interferon-γ and interleukins 1, 12 and 23), anti-inflammatory (transforming growth factor-β and interleukins 4, 10, 27 and 35) and neurotrophic factors (ciliary neurotrophic factor and brain-derived neurotrophic factor). Also, the regulatory and effector functions of distinct immune cell subpopulations such as CD4+ Th1, Th2, Th3 and Th17 cells, CD4+FoxP3+ Treg cells, CD8+ Tc1 and Tc2, B cells and γδ+ T cells have been disclosed in more detail. The new insights may help to identify novel targets for the treatment of MS. However, translation of the experimental results into the clinical practice requires prudence and great caution.
Collapse
Key Words
- apc, antigen-presenting cell
- at-eae, adoptive transfer eae
- bbb, blood–brain barrier
- bdnf, brain-derived neurotrophic factor
- cd, cluster of differentiation
- cns, central nervous system
- cntf, ciliary neurotrophic factor
- eae, experimental autoimmune encephalomyelitis
- hla, human leukocyte antigen
- ig, immunoglobulin
- il, interleukin
- ifn, interferon
- ivig, intravenous immunoglobulin
- mab, monoclonal antibody
- mbp, myelin basic protein
- mhc, major histocompatibility complex
- mog, myelin oligodendrocyte glycoprotein
- mp, methylprednisolone
- mri, magnetic resonance imaging
- ms, multiple sclerosis
- nk, natural killer
- odc, oligodendrocyte
- qtl, quantitative trait locus
- plp, proteolipid protein
- tc, cytotoxic t cell
- tcr, t cell receptor
- tgf, transforming growth factor
- th cell, helper t cell
- tnf, tumor necrosis factor
- animal model
- autoimmunity
- experimental autoimmune encephalomyelitis
- immunogenetics
- immunomodulatory therapy
- multiple sclerosis
Collapse
MESH Headings
- Animals
- Animals, Genetically Modified
- Clinical Trials as Topic
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/physiopathology
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Gene Expression Profiling
- History, 19th Century
- History, 20th Century
- History, 21st Century
- Humans
- Microarray Analysis
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/physiopathology
- Multiple Sclerosis/therapy
Collapse
Affiliation(s)
- Eilhard Mix
- Department of Neurology, University of Rostock, Germany
| | | | - Hans-Peter Hartung
- Department of Neurology, Heinrich-Heine-University, Moorenstr. 5, 40225 Duesseldorf, Germany
| | - Uwe K. Zettl
- Department of Neurology, University of Rostock, Germany
| |
Collapse
|
42
|
Zhao Z, Zhao S, Xu N, Yu C, Guan S, Liu X, Huang L, Liao W, Jia W. Lovastatin improves neurological outcome after nucleus basalis magnocellularis lesion in rats. Neuroscience 2010; 167:954-63. [DOI: 10.1016/j.neuroscience.2010.02.054] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Revised: 02/13/2010] [Accepted: 02/19/2010] [Indexed: 12/15/2022]
|
43
|
Van der Walt A, Butzkueven H, Kolbe S, Marriott M, Alexandrou E, Gresle M, Egan G, Kilpatrick T. Neuroprotection in multiple sclerosis: a therapeutic challenge for the next decade. Pharmacol Ther 2010; 126:82-93. [PMID: 20122960 DOI: 10.1016/j.pharmthera.2010.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2010] [Accepted: 01/19/2010] [Indexed: 12/20/2022]
Abstract
Multiple sclerosis (MS) is the commonest cause of progressive neurological disability amongst young, Caucasian adults. MS is considered to be an auto-immune disease that results from an attack against myelin, the layer which surrounds axons. The pathophysiology of MS is complex, with both demyelination and axonal degeneration contributing to what is essentially an inflammatory neurodegenerative disease. Axonal loss is increasingly being accepted as the histopathological correlate of neurological disability. Currently, the underpinnings of neurodegeneration in MS, and how to promote neuroprotection are only partly understood. No established treatments that directly reduce nervous system damage or enhance its repair are currently available. Moreover, the ability of currently available immunomodulatory therapies used to treat MS, such as interferon-beta, to prevent long-term disability is uncertain. Results from short-term randomized-controlled trials suggest a partial benefit with regards to disability outcomes, but this is yet to be established in long-term studies. Novel neuroprotective agents have been identified in preclinical studies but their development is being hampered by the absence of appropriate clinical platforms to test them. In this article, we will discuss some of the principal therapeutic candidates that could provide neuroprotection in MS and emerging methodologies by which to test them.
Collapse
Affiliation(s)
- Anneke Van der Walt
- The Royal Melbourne Hospital, Grattan St. Parkville, Melbourne, Australia; Centre for Neuroscience, University of Melbourne, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
44
|
|
45
|
Staines DR, Brenu EW, Marshall-Gradisnik S. Postulated vasoactive neuropeptide immunopathology affecting the blood-brain/blood-spinal barrier in certain neuropsychiatric fatigue-related conditions: A role for phosphodiesterase inhibitors in treatment? Neuropsychiatr Dis Treat 2009; 5:81-9. [PMID: 19557103 PMCID: PMC2695238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Neuropsychiatric symptoms occur in a number of neurological fatigue-related conditions including multiple sclerosis (MS), Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and chronic fatigue syndrome (CFS). These conditions have been attributed variably to neuroinflammatory and neurodegenerative processes. While autoimmune pathology, at least in part, has long been suspected in these conditions proof has been elusive. Autoimmune pathomechanisms affecting the blood-brain barrier (BBB) or blood-spinal barrier (BSB) may predispose the BBB/BSB to 'leakiness' and be a precursor to additional autoimmune events resulting in neuroinflammatory or neurodegenerative processes. The aim of the paper is to postulate immunopathology of the cerebrospinal perivascular compartment involving certain vasoactive neuropeptides, specifically pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP), in the etiology of certain neuropsychiatric fatigue-related conditions such as MS, ALS, PD, and CFS. Vasoactive neuropeptides (VNs) such as PACAP and VIP have critical roles as neurotransmitters, vasodilators including perfusion and hypoxia regulators, and immune and nociception modulators. PACAP and VIP are widely distributed in the central nervous system (CNS) and have key roles in CNS blood vessels including maintaining functional integrity of the BBB and BSB. Autoimmunity affecting these VNs would likely have a detrimental effect on BBB and BSB functioning arguably predisposing to further pathological processes. Virchow-Robin spaces (VRS) are perivascular compartments surrounding small vessels within the CNS which contribute to the BBB and BSB integrity and contain PACAP and VIP receptors. Autoimmunity of these receptors would likely affect BBB and VRS function and therefore may contribute to the etiology of these conditions by affecting CNS and immunological homeostasis, including promoting neuropsychological symptomatology. PACAP and VIP, as potent activators of adenylate cyclase (AC), have a key role in cyclic adenosine monophosphate (cAMP) production affecting regulatory T cell (Treg) and other immune functions. Phosphodiesterase enzymes (PDEs) catalyze cAMP and PDE inhibitors (PDEIs) maintain cAMP levels and have proven and well known therapeutic benefit in animal models such as experimental allergic encephalomyelitis (EAE). Therefore PDEIs may have a role in therapy for certain neuropsychiatric fatigue-related conditions.
Collapse
Affiliation(s)
- Donald R Staines
- Queensland Health, Gold Coast Population Health Unit, Southport, Gold Coast, Queensland, Australia.
| | | | | |
Collapse
|
46
|
Statins: mechanisms of neuroprotection. Prog Neurobiol 2009; 88:64-75. [PMID: 19428962 DOI: 10.1016/j.pneurobio.2009.02.002] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Revised: 01/09/2009] [Accepted: 02/10/2009] [Indexed: 12/17/2022]
Abstract
Clinical trials report that the class of drugs known as statins may be neuroprotective in Alzheimer's and Parkinson's disease, and further trials are currently underway to test whether these drugs are also beneficial in multiple sclerosis and acute stroke treatment. Since statins are well tolerated and have relatively few side effects, they may be considered as viable drugs to ameliorate neurodegenerative diseases. However, the mechanism of their neuroprotective effects is only partly understood. In this article, we review the current data on the neuroprotective effects of statins and their underlying mechanisms. In the first section, we detail the mechanisms by which statins affect cellular signalling. The primary action of statins is to inhibit cellular cholesterol synthesis. However, the cholesterol synthesis pathway also has several by-products, the non-sterol isoprenoids that are also important in cellular functioning. Furthermore, reduced cholesterol levels may deplete the cholesterol-rich membrane domains known as lipid rafts, which in turn could affect cellular signalling. In the second section, we summarize how the effects on signalling translate into general neuroprotective effects through peripheral systems. Statins improve blood-flow, reduce coagulation, modulate the immune system and reduce oxidative damage. The final section deals with the effects of statins on the central nervous system, particularly during Alzheimer's and Parkinson's disease, stroke and multiple sclerosis.
Collapse
|
47
|
Abstract
Traumatic brain injury (TBI) remains one of the leading causes of mortality and morbidity worldwide in individuals under the age of 45 years, and, despite extensive efforts to develop neuroprotective therapies, there has been no successful outcome in any trial of neuroprotection to date. In addition to recognizing that many TBI clinical trials have not been optimally designed to detect potential efficacy, the failures can be attributed largely to the fact that most of the therapies investigated have been targeted toward an individual injury factor. The contemporary view of TBI is that of a very heterogenous type of injury, one that varies widely in etiology, clinical presentation, severity, and pathophysiology. The mechanisms involved in neuronal cell death after TBI involve an interaction of acute and delayed anatomic, molecular, biochemical, and physiological events that are both complex and multifaceted. Accordingly, neuropharmacotherapies need to be targeted at the multiple injury factors that contribute to the secondary injury cascade, and, in so doing, maximize the likelihood of a successful outcome. This review focuses on a number of such multifunctional compounds that have shown considerable success in experimental studies and that show maximum promise for success in clinical trials.
Collapse
Affiliation(s)
- Robert Vink
- School of Medical Sciences, University of Adelaide, Adelaide, South Australia 5005, Australia.
| | | |
Collapse
|