Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neuroimmunol ; 321: 125-137, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29957383

ABSTRACT

Apoptosis is one form of cell death that is intimately related to health and pathological conditions. In most neuroinflammatory and/or neurodegenerative diseases, apoptosis is associated with disease development and pathology and inhibition of this process leads to considerable amelioration. It is becoming evident that apoptosis also participates in the pathogenesis of Multiple Sclerosis (MS) and its animal model, Experimental Autoimmune Encephalomyelitis (EAE). Drugs such as Sildenafil, a Phosphodiesterase type 5 Inhibitor (PDE5I), have proven to be neuroprotective in MS models. However, it is not known whether Sildenafil is able to modulate cell death, specifically apoptosis, in EAE mice. Therefore, the aim of this study was to determine the effects of Sildenafil on extrinsic and intrinsic apoptosis pathways in the spinal cord of C57BL/6 mice with EAE. TUNEL analysis showed that EAE mice had elevated number of TUNEL+ cells and that treatment with Sildenafil led to reduced number of dying cells, indicating that Sildenafil was able to inhibit cell death. We observed that both extrinsic and intrinsic pathways of apoptosis were governing the dynamics of EAE progression. We showed that in EAE mice there were increased levels of extrinsic (Caspase-8, -3, TNF-α, FADD) and intrinsic (Caspase-9, Bax and Cytochrome C) apoptosis markers. Bcl-2, an anti-apoptotic protein, was downregulated in EAE mice. We also demonstrated that EAE mice had increased levels of non-caspase mediators of cell survival/cell death (p-IκBα and p-MAPK-p38). Besides, EAE mice presented augmented demyelination. Nevertheless, this is the first research to demonstrate that Sildenafil, when administered concomitant to disease induction, modulated the expression of pro- and anti-apoptotic proteins of the extrinsic and intrinsic pathways, as well as diminished the expression of non-caspase mediators and promoted remyelination in the spinal cord, indicating neuroprotective effects. Thus, the present study demonstrated that Sildenafil inhibits apoptosis by two distinct, although interconnected, mechanisms: directly by modulating caspase expression (through extrinsic and intrinsic pathways) and indirectly by modulating the expression of molecules involved in cell death and/or cell survival.


Subject(s)
Apoptosis/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/immunology , Sildenafil Citrate/therapeutic use , Spinal Cord/drug effects , Spinal Cord/immunology , Animals , Apoptosis/physiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Phosphodiesterase 5 Inhibitors/pharmacology , Phosphodiesterase 5 Inhibitors/therapeutic use , Sildenafil Citrate/pharmacology , Spinal Cord/pathology
2.
Inflammopharmacology ; 26(2): 599-609, 2018 Apr.
Article in English | MEDLINE | ID: mdl-28409388

ABSTRACT

BACKGROUND AND AIM: While diethylcarbamazine citrate (DEC) displays important anti-inflammatory effects in experimental models of liver injury, the mechanisms of its action remain poorly understood. The aim of the present study was to investigate the fibrolytic potential of DEC. METHODS: Mice receive two injections of carbon tetrachloride (CCl4) per week for 8 weeks. DEC 50 mg/kg body weight was administered through drinking water during the last 12 days of liver injury. RESULTS: The expression of hepatic stellate cells (HSCs) activation markers, including smooth muscle α-actin (α-SMA), collagen I, transforming growth factor-ß 1 (TGF-ß1), matrix metalloproteinase-2 (MMP-2) and tissue inhibitor of metalloproteinase-1 (TIMP-1) was assessed. The influence of DEC on the intracellular MAPK pathways of the HSCs (JNK and p38 MAPK) was also estimated. DEC inhibited HSCs activation measured as the production of α-SMA and collagen I. In addition, it down regulated the production of TGF-ß1 and TIMP-1, and concomitantly increased MMP-2 activity. Furthermore, DEC significantly inhibited the activation of the JNK and p38 MAPK signaling pathways. CONCLUSIONS: In conclusion, DEC significantly attenuated the severity of CCl4-induced liver injury and the progression of liver fibrosis, exerting a potential fibrolytic effect in the CCl4-induced fibrosis model.


Subject(s)
Biomarkers/metabolism , Carbon Tetrachloride/pharmacology , Diethylcarbamazine/pharmacology , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/chemically induced , Liver Cirrhosis/drug therapy , Actins/metabolism , Animals , Collagen Type I/metabolism , Down-Regulation/drug effects , Hepatic Stellate Cells/metabolism , Liver/drug effects , Liver/metabolism , Liver Cirrhosis/metabolism , Male , Matrix Metalloproteinase 2/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Signal Transduction/drug effects , Tissue Inhibitor of Metalloproteinase-1/metabolism , Transforming Growth Factor beta1/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
3.
Exp Neurol ; 298(Pt A): 31-41, 2017 12.
Article in English | MEDLINE | ID: mdl-28844606

ABSTRACT

Adenosine monophosphate-activated protein kinase (AMPK) is an evolutionarily conserved sensor of cellular energy status and has been reported to be involved in chronic inflammatory disorders. AMPK is expressed in immune cells, such as dendritic cells, macrophages, lymphocytes and neutrophils, and is an important regulator of inflammatory responses through the regulation of complex signaling networks in part by inhibiting downstream cascade pathways, such as nuclear factor kB, which is a key regulator of innate immunity and inflammation, as well as acting as a negative regulator of toll-like receptors. Recent data suggest that AMPK dysregulation may participate in neurodegenerative diseases, such as multiple sclerosis, Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis and neuropathies. However, there are conflicting reports on the benefits or detrimental effects of AMPK in distinct pathological conditions. This paper offers a review of the recent literature on the pharmacological modulation of the AMPK system as a potential molecular target in the management of neurodegenerative diseases.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Inflammation Mediators/metabolism , Neurodegenerative Diseases/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/chemistry , Animals , Enzyme Activation/physiology , Humans , Inflammation/metabolism , Inflammation/pathology , Neurodegenerative Diseases/pathology , Protein Structure, Secondary
4.
Neurosci Lett ; 630: 59-65, 2016 Sep 06.
Article in English | MEDLINE | ID: mdl-27466020

ABSTRACT

Astrocytes are effectively involved in the pathophysiological processes in the central nervous system (CNS) and may contribute to or protect against development of inflammatory and degenerative diseases. Sildenafil is a potent and selective phosphodiesterase-5 (PDE-5) inhibitor, which induces cyclic GMP accumulation. However, the mechanisms of actions on glial cells are not clear. The aim of the present work is to evaluate the role of sildenafil in lipopolysaccharide (LPS)-stimulated astrocytes. The cytoskeleton integrity and Ca(2+) waves were assessed as indicators of inflammatory state. Two main groups were done: (A) one prevention and (B) one restoration. Each of these groups: A1: control; A2: LPS for 24h; A3: sildenafil 1 or 10µM for 4h and then sildenafil 1 or 10µM+LPS for 24h. B1: control; B2: LPS for 24h; B3: LPS for 24h and then LPS+sildenafil 1 or 10µM for 24h. Cytoskeleton integrity was analyzed through GFAP immunolabeling and actin labeling with an Alexa 488-conjugated phalloidin probe. Calcium responses were assessed through a Ca(2+)-sensitive fluorophore Fura-2/AM. The results show that both preventive and restorative treatments with sildenafil (in both concentrations) reduced the Ca(2+) responses in intensity and induced a more organized actin fiber pattern, compared to LPS treated cells. This work demonstrated for the first time that astrocytes are a key part of the sildenafil protective effects in the CNS.


Subject(s)
Astrocytes/drug effects , Calcium Signaling/drug effects , Cytoskeleton/drug effects , Encephalitis/prevention & control , Phosphodiesterase 5 Inhibitors/pharmacology , Sildenafil Citrate/pharmacology , Actin Cytoskeleton/drug effects , Actin Cytoskeleton/metabolism , Adenosine Triphosphate/pharmacology , Animals , Astrocytes/metabolism , Cells, Cultured , Encephalitis/chemically induced , Encephalitis/metabolism , Lipopolysaccharides , Rats
5.
Brain Res ; 1644: 149-60, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27174003

ABSTRACT

The aim of the present study was to analyze the action of metformin on short-term memory, glial cell activation and neuroinflammation caused by experimental diabetic encephalopathy in C57BL/6 mice. Diabetes was induced by the intraperitoneal injection of a dose of 90mg/kg of streptozotocin on two successive days. Mice with blood glucose levels ≥200dl/ml were considered diabetic and were given metformin hydrochloride at doses of 100mg/kg and 200mg/kg (by gavage, twice daily) for 21 days. On the final day of treatment, the mice underwent a T-maze test. On the 22nd day of treatment all the animals were anesthetized and euthanized. Diabetic animals treated with metformin had a higher spatial memory score. The hippocampus of the diabetic animals presented reactive gliosis, neuronal loss, NF-kB signaling activation, and high levels of IL-1 and VEGF. In addition, the T-maze test scores of these animals were low. Treatment with metformin reduced the expression of GFAP, Iba-1 (astrocyte and microglial markers) and the inflammation markers (p-IKB, IL-1 and VEGF), while enhancing p-AMPK and eNOS levels and increasing neuronal survival (Fox-1 and NeuN). Treatment with metformin also improved the spatial memory scores of diabetic animals. In conclusion, the present study showed that metformin can significantly reduce neuroinflammation and can decrease the loss of neurons in the hippocampus of diabetic animals, which can subsequently promote improvements in spatial memory.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/psychology , Encephalitis/metabolism , Hippocampus/drug effects , Hypoglycemic Agents/administration & dosage , Memory, Short-Term/drug effects , Metformin/administration & dosage , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Blood Glucose , Diabetes Mellitus, Experimental/complications , Encephalitis/etiology , Hippocampus/pathology , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/metabolism , NF-KappaB Inhibitor alpha/metabolism , Neurons/drug effects , Neurons/pathology , Nitric Oxide Synthase Type III/metabolism , Peptide Fragments/metabolism , Streptozocin
6.
Cells Tissues Organs ; 201(3): 193-202, 2016.
Article in English | MEDLINE | ID: mdl-26978649

ABSTRACT

Lipopolysaccharide (LPS) injections during pregnancy are well established as models for pregnancy complications, including fetal growth restriction (FGR), thrombophilia, preterm labor and abortion. Indeed, inflammation, as induced by LPS injection has been described as a pivotal factor in cases of miscarriage related to placental tissue damage. The phosphodiesterase-5 inhibitor sildenafil (Viagra®) is currently used to treat FGR cases in women, while low-molecular weight heparin (Fragmin®) is a standard treatment for recurrent miscarriage (RM). However, the pathways and cellular dynamics involved in RM are not completely understood. The aim of this study was to evaluate the protective effect of sildenafil and dalteparin in a mouse model of LPS-induced abortion. Histopathology, ultrastructural analysis and immunofluorescence for P-selectin were studied in two different placental cell types: trophoblast cells and labyrinth endothelial cells. Treatment with sildenafil either alone or in combination with heparin showed the best response against LPS-induced injury during pregnancy. In conclusion, our results support the use of these drugs as future therapeutic agents that may protect the placenta against inflammatory injury in RM events. Analyses of the ultrastructure and placental immunophysiology are important to understand the mechanism underlying RM. These findings may spark future studies and aid in the development of new therapies in cases of RM.


Subject(s)
Abortion, Habitual/drug therapy , Anticoagulants/therapeutic use , Dalteparin/therapeutic use , Phosphodiesterase 5 Inhibitors/therapeutic use , Placenta/drug effects , Placenta/pathology , Sildenafil Citrate/therapeutic use , Abortion, Habitual/immunology , Abortion, Habitual/pathology , Animals , Disease Models, Animal , Female , Lipopolysaccharides/immunology , Male , Mice , Placenta/cytology , Placenta/immunology , Pregnancy , Trophoblasts/cytology , Trophoblasts/drug effects , Trophoblasts/immunology , Trophoblasts/pathology
7.
Exp Neurol ; 275 Pt 1: 143-53, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26515692

ABSTRACT

While it has recently been shown that sildenafil (Viagra®) has a protective effect on myelination/remyelination, the mechanism of this protection is still unknown. In general, cytokines, chemokines and metalloproteinases have a pro-inflammatory action, but can also exert a role in modulating glial cell activation, contributing to the balance of cell response. Investigating these molecules can contribute to clarifying the mechanisms of sildenafil neuroprotection. In addition, it is not known whether sildenafil is able to restore an already installed neurodegenerative process or if the treatment period is critical for its action. The aim of the present study was to evaluate, in a cuprizone (CPZ)-induced demyelination model, the effects and mechanisms of time-dependent treatment with sildenafil (beginning 15 days after neurodegeneration and continuing for 15 days, or starting concomitantly with neurodegeneration and continuing for 30 days) on neuroinflammation and remyelination. Neuroinflammation and demyelination induced by CPZ in rodents has been widely used as a model of multiple sclerosis (MS). In the present study, five male C57BL/6 mice aged 7-10 weeks were used per group. For four weeks, the groups received either cuprizone (CPZ) 0.2% mixed in feed or CPZ combined with the administration of sildenafil (Viagra®, Pfizer, 25 mg/kg) orally in drinking water, starting concurrently with (sild-T0) or 15 days (sild-T15) after the start of CPZ treatment. Control animals received pure food and water. The cerebella were dissected and processed for immunohistochemistry, immunofluorescence (frozen), Western blotting, Luxol fast blue staining and transmission electron microscopy. Magnetic resonance was performed for live animals, after the same treatment, using CPZ 0.3%. CPZ induced an increase in the expression of IL-1ß and a decrease in MCP-1, CCR-2, MBP and GST-pi, as well as promoting damage in the structure and ultra-structure of the myelin sheath. Interestingly, the administering of sild-T0 promoted a further increase of MMP-9, MCP-1, and CCR-2, possibly contributing to changes in the microglia phenotype, which becomes more phagocytic, cleaning myelin debris. It was also observed that, after sild-T0 treatment, the expression of GST-pi and MBP increased and the myelin structure was improved. However, sild-T15 was not efficient in all aspects, probably due to the short treatment period and to starting after the installation of the degenerative process. Therefore, the present study shows that sildenafil modulates inflammation, with the involvement of MMP-9, MCP-1, and CCR-2, and also contributes to myelin repair. These protective effects were dependent on the therapeutic strategy used. This clarification can strengthen research proposals into the mechanism of action of sildenafil and contribute to the control of neurodegenerative diseases such as MS.


Subject(s)
Chemokine CCL2/metabolism , Demyelinating Diseases/metabolism , Matrix Metalloproteinase 9/metabolism , Myelin Sheath/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Receptors, CCR2/metabolism , Signal Transduction/drug effects , Sildenafil Citrate/pharmacology , Animals , Cuprizone , Demyelinating Diseases/chemically induced , Mice , Multiple Sclerosis/chemically induced , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology
8.
Brain Res ; 1627: 119-33, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26404052

ABSTRACT

Sildenafil (Viagra®) has recently been found to have a neuroprotective effect, which occurs through the inhibition of inflammation and demyelination in the cerebellum. However, the mechanism of action of sildenafil remains unknown. AMPK, the regulatory protein of the lipid and glucose metabolism, plays a protective role by activating the eNOS enzyme. The production of a nanomolar concentration of NO by eNOS has an anti-inflammatory effect through the cGMP signaling pathway and plays an important role in the regulation of the nuclear transcription factor (NFkB), preventing the expression of inflammatory genes. The present study investigated whether AMPK-eNOS-NO-cGMP-IКßα-NFkB is involved in the mechanism of action of sildenafil in a cuprizone-demyelination model. Neuroinflammation and demyelination induced by cuprizone in rodents have been widely used as a model of MS. In the present study, five male C57BL/6 mice (7-10 weeks old) were used. Over a four week period, the groups received: cuprizone (CPZ) 0.2% mixed in feed; CPZ in the diet, combined with the administration of sildenafil (Viagra®, Pfizer, 25mg/kg) orally in drinking water, starting concurrently (sild-T0) or 15 days (sild-T15) after the start of CPZ. Control animals received pure food and water. The cerebella of the mice were dissected and processed for immunohistochemistry, immunofluorescence (frozen), western blotting and dosage of cytokines (Elisa). CPZ induced an increase in the expression of GFAP, IL-1ß TNF-α, total NFkB and inactive AMPK, and prompt microglia activation. CPZ also induced a reduction of IKßα. The administration of sildenafil reduced the expression of the pro-inflammatory cytokines IL-1ß and TNF-α and increased the expression of the anti-inflammatory cytokine IL-10. In addition, the administration of sildenafil reduced expression of GFAP, NFkB, inactive AMPK and iNOS, and increased IKßα. Interestingly, sildenafil also reduced levels of NGF. In general, the sild-T0 group was more effective than sild-T15 in improving clinical status and promoting the control of neuroinflammation. The present study offers evidence that sildenafil has anti-inflammatory and neuroprotective effects, which are probably achieved through modulation of AMPK-IKßα-NFκB signaling. In addition, eNOS may play a role in the sildenafil neuroprotective mechanism, contributing to the activation of AMPK. However, other pathways such as MAPK-NFkB and the downstream proteins AMPK (AMPK-SIRT1-NFκB) should also be further investigated. An understanding of these mechanisms of action is critical for the clinical use of sildenafil to control neuroinflammation in neurodegenerative diseases such as MS.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/therapeutic use , Demyelinating Diseases/drug therapy , NF-kappa B/metabolism , Nitric Oxide Synthase Type III/metabolism , Sildenafil Citrate/therapeutic use , Animals , Chelating Agents/toxicity , Cuprizone/toxicity , Cytokines/metabolism , Demyelinating Diseases/chemically induced , Disease Models, Animal , Encephalitis/chemically induced , Encephalitis/drug therapy , Enzyme Inhibitors/pharmacology , Glial Fibrillary Acidic Protein/metabolism , Male , Mice , Mice, Inbred C57BL
9.
Mediators Inflamm ; 2015: 940207, 2015.
Article in English | MEDLINE | ID: mdl-26770022

ABSTRACT

Phosphodiesterase type 5 inhibitors (PDE5-Is) have recently emerged as a potential therapeutic strategy for neuroinflammatory, neurodegenerative, and memory loss diseases. Mechanistically, PDE5-Is produce an anti-inflammatory and neuroprotection effect by increasing expression of nitric oxide synthases and accumulation of cGMP and activating protein kinase G (PKG), the signaling pathway of which is thought to play an important role in the development of several neurodiseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). The aim of this paper was to review present knowledge of the signaling pathways that underlie the use of PDE5-Is in neuroinflammation, neurogenesis, learning, and memory.


Subject(s)
Cognition/drug effects , Nerve Degeneration/drug therapy , Neuroimmunomodulation/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Animals , Blood-Brain Barrier/drug effects , Cognition/physiology , Humans , Mice , Nerve Degeneration/physiopathology , Neurogenesis/drug effects , Neurogenesis/physiology , Neuroglia/drug effects , Neuroimmunomodulation/physiology , Neuroprotective Agents/pharmacology , Nucleotides, Cyclic/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
10.
Mediators Inflamm ; 2014: 696383, 2014.
Article in English | MEDLINE | ID: mdl-25374445

ABSTRACT

This study investigated the anti-inflammatory effects of DEC on the CCl4-induced hepatotoxicity in C57BL/6 mice. Chronic inflammation was induced by i.p. administration of CCl4 0.5 µL/g of body weight through two injections a week for 6 weeks. DEC (50 mg/kg) was administered by gavage for 12 days before finishing the CCl4 induction. Histological analyses of the DEC-treated group exhibited reduced inflammatory process and prevented liver necrosis and fibrosis. Immunohistochemical and immunofluorescence analyses of the DEC-treated group showed reduced COX-2, IL1ß, MDA, TGF-ß, and αSMA immunopositivity, besides exhibiting decreased IL1ß, COX-2, NFκB, IFNγ, and TGFß expressions in the western blot analysis. The DEC group enhanced significantly the IL-10 expression. The reduction of hepatic injury in the DEC-treated group was confirmed by the COX-2 and iNOS mRNA expression levels. Based on the results of the present study, DEC can be used as a potential anti-inflammatory drug for chronic hepatic inflammation.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Chemical and Drug Induced Liver Injury/drug therapy , Diethylcarbamazine/pharmacology , Animals , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Collagen/metabolism , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Disease Models, Animal , Gene Expression/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
11.
Toxicol Appl Pharmacol ; 280(1): 159-68, 2014 Oct 01.
Article in English | MEDLINE | ID: mdl-24918699

ABSTRACT

The aim of the present study was to analyze the effect of a combination of metformin hydrochloride and melatonin on oxidative stress together with a biochemical and histopathological analysis of the livers of Wistar rats induced with PCOS. The results indicated that a combination of the drugs was more effective in the reduction of plasmatic levels of liver enzyme alanine aminotransferase, nitric oxide and total glutathione, and decreased the inflammatory response and histopathological damage, producing results that were significantly similar to animals from the control group. A mixture of the drugs produced more effective results against liver toxicity caused by PCOS, encouraging the normalization of biochemical parameters. During pregnancy, there was reduced oxidative stress compared to monotherapeutic use of these drugs. Interestingly, the combination of the drugs caused a physiological reaction similar to responses identified in healthy rats without induction of the PCOS control group. However, the clinical and physiological effectiveness of the combination should be further explored, especially with respect to the possible side effects on offspring.


Subject(s)
Antioxidants/administration & dosage , Liver/drug effects , Melatonin/administration & dosage , Metformin/administration & dosage , Oxidative Stress/drug effects , Polycystic Ovary Syndrome/drug therapy , Animals , Drug Therapy, Combination , Female , Liver/metabolism , Liver/pathology , Oxidative Stress/physiology , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/pathology , Pregnancy , Rats , Rats, Wistar , Treatment Outcome
12.
Brain Res Bull ; 104: 60-73, 2014 May.
Article in English | MEDLINE | ID: mdl-24727400

ABSTRACT

Nitric oxide (NO) is the main activator of the soluble guanylate cyclase (sGC)-guanosine 3'5' cyclic monophosphate (cGMP) pathway. The level of cGMP is regulated by phosphodiesterases (PDEs), which break down cGMP. It has been reported that levels of NO in the central nervous system (CNS) can greatly increase during demyelination and/or neuroinflammation. Controversially, in demyelination models, mice without iNOS may develop more severe cases of disease. Furthermore, cGMP accumulation caused by PDE inhibitors has an anti-inflammatory/neuroprotective effect in MS-models. The role of the NO-cGMP pathway in the nervous tissue is, therefore, complex and not fully understood. The aim of the present study was to contribute to existing knowledge of the role of this pathway in the CNS. Wild type (WT - C57BL/6) and iNOS(-/-) animals were treated with sildenafil (25mg/kg) for 8 weeks. Control animals were not treated. VCAM and ICAM (adhesion proteins), GFAP and Iba-1 (astrocyte and microglia markers, respectively), PKG (cGMP-dependent protein kinase), sGC, eNOS (constitutive endothelial NO sinthase) and GSTpi (a marker of mature oligodendrocytes) were evaluated in the cerebellum using immunohistochemistry or western blotting. Myelin was assessed by luxol fast blue staining and electron transmission microscopy. Treatment with sildenafil reduced ICAM and VCAM levels (anti-inflammatory effect) and increased GFAP and Iba-1 expression (clearance phenotype) in WT animals. The expression of VCAM, ICAM, GFAP, PKG and sGC was lower in iNOS(-/-) mice than in WT control animals. The treatment of iNOS(-/-) animals with sildenafil resulted in an increase of all proteins (pro-inflammatory effect). There was overexpression of eNOS in untreated iNOS(-/-) mice. The myelin structure of iNOS(-/-) animals was damaged in comparison with WT control. Sildenafil increased GSTpi and resulted in an improved myelin structure in iNOS(-/-) mice. In conclusion, NO-cGMP signaling plays a role in the regulation of inflammation and myelination processes. The accumulation of cGMP produced opposite effects in WT and iNOS(-/-) mice. This can be explained by the overexpression of eNOS in iNOS(-/-) mice, unbalancing cGMP signaling, or cGMP has a dual role in inflammation. Drugs that modulate the NO-sGC-cGMP pathway may be clinically beneficial in the treatment of neuroinflammatory/demyelinating disorders, but further studies of the regulation of this pathway are required.


Subject(s)
Cerebellum/metabolism , Myelin Sheath/ultrastructure , Neuroglia/metabolism , Signal Transduction/physiology , Animals , Cerebellum/ultrastructure , Cyclic GMP/metabolism , Inflammation/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Myelin Sheath/physiology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics
13.
Mediators Inflamm ; 2013: 321460, 2013.
Article in English | MEDLINE | ID: mdl-23970812

ABSTRACT

We recently demonstrated that sildenafil reduces the expression of cytokines, COX-2, and GFAP in a demyelinating model induced in wild-type (WT) mice. Herein, the understandings of the neuroprotective effect of sildenafil and the mediation of iNOS/NO system on inflammatory demyelination induced by cuprizone were investigated. The cerebella of iNOS(-/-) mice were examined after four weeks of treatment with cuprizone alone or combined with sildenafil. Cuprizone increased GFAP, Iba-1, TNF- α , COX-2, IL-1 ß , and IFN- γ expression, decreased expression of glutathione S-transferase pi (GSTpi), and damaged myelin in iNOS(-/-) mice. Sildenafil reduced Iba-1, IFN- γ , and IL-1 ß levels but had no effect on the expression of GFAP, TNF- α , and COX-2 compared to the cuprizone group. Sildenafil elevated GSTpi levels and improved the myelin structure/ultrastructure. iNOS(-/-) mice suffered from severe inflammation following treatment with cuprizone, while WT mice had milder inflammation, as found in the previous study. It is possible that inflammatory regulation through iNOS-feedback is absent in iNOS(-/-) mice, making them more susceptible to inflammation. Sildenafil has at least a partial anti-inflammatory effect through iNOS inhibition, as its effect on iNOS(-/-) mice was limited. Further studies are required to explain the underlying mechanism of the sildenafil effects.


Subject(s)
Demyelinating Diseases/metabolism , Inflammation/drug therapy , Myelin Sheath/chemistry , Neurons/drug effects , Nitric Oxide Synthase Type II/metabolism , Nitric Oxide/chemistry , Piperazines/pharmacology , Sulfones/pharmacology , Animals , Cuprizone/toxicity , Cyclic GMP/metabolism , Demyelinating Diseases/genetics , Glutathione S-Transferase pi/metabolism , Mice , Mice, Knockout , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Nitric Oxide Synthase Type II/genetics , Phosphodiesterase 5 Inhibitors/pharmacology , Purines/pharmacology , Sildenafil Citrate
14.
Cytokine ; 60(2): 540-51, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22749439

ABSTRACT

Sildenafil induces cGMP accumulation through phosphodiesterase-5 (PDE5) inhibition. cGMP-pathways protect oligodendrocytes and modulate astroglial and microglial reactions. Microglia and astrocytes play an important role in perpetuating multiple sclerosis (MS), a chronic inflammatory disease characterized by demyelination. Therefore, sildenafil can be a potential tool for MS treatment. The present study investigated the effects of sildenafil on the myelin structure and astrocyte/microglia-mediated neuroinflammation in an animal model of MS. Cuprizone-induced demyelination and neuroinflammation in rodents has been widely used as a model for MS. Herein, five male C57BL/6 mice (7-10 weeks old) were used per group. Over a 4-week period, the different groups received the following: (1) cuprizone (0.2%) mixed into the chow; (2) cuprizone in the chow and sildenafil (Viagra®; 3, 25 or 50mg/kg) in the drinking water; or (3) pure chow and water (control group). Cerebella were analyzed using transmission electron microscopy, western blotting, immunohistochemistry and luxol fast blue staining. Cuprizone induced tissue damage, with an increase in GFAP, Iba-1 and COX-2 and demyelination in comparison to the control group. However, cuprizone did not affect the expression of cytokines (TNF-α, IFN-γ, IL-1ß and IL-2). Sildenafil reduced GFAP (25 and 50mg/kg) and Iba-1 expression (25mg/kg) in comparison to the cuprizone group, indicating the modulation of astrocytes and microglia, respectively. Sildenafil preserved myelin and axons ultrastructure and strongly reduced IFN-γ, TNF-α, IL-1ß, IL-2 and COX-2 expression in comparison to the control and/or cuprizone groups. The results demonstrate a protective effect of sildenafil in the cerebellum. Thus, well-designed clinical trials may demonstrate that the oral administration of sildenafil can be suitable for individuals with MS and other neuroinflammatory/neurodegenerative diseases, providing additional benefits to current treatments.


Subject(s)
Cytokines/metabolism , Demyelinating Diseases/drug therapy , Demyelinating Diseases/prevention & control , Multiple Sclerosis/drug therapy , Piperazines/therapeutic use , Sulfones/therapeutic use , Animals , Cerebellum/drug effects , Cerebellum/enzymology , Cerebellum/pathology , Cerebellum/ultrastructure , Cuprizone , Cyclooxygenase 2/metabolism , Disease Models, Animal , Down-Regulation/drug effects , Fluorescent Antibody Technique , Glial Fibrillary Acidic Protein/metabolism , Immunoblotting , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Piperazines/pharmacology , Purines/pharmacology , Purines/therapeutic use , Sildenafil Citrate , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...