Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
Eur J Pharmacol ; 787: 127-33, 2016 Sep 15.
Article in English | MEDLINE | ID: mdl-27090928

ABSTRACT

Primary open-angle glaucoma (POAG) is a common form of glaucoma in which retinal ganglion cells (RGCs) die at varying intervals. Primary open-angle glaucoma is often associated with an increased intraocular pressure (IOP), which when reduced, can slow down the progression of the disease. However, it is essential to develop better modes of treatments for glaucoma patients. In this overview, we discuss the hypothesis that RGC mitochondria are affected during the initiation of POAG, by becoming gradually weakened, but at different rates because of their specific receptor profiles. With this in mind, we argue that neuroprotection in the context of glaucoma should focus on preserving RGC mitochondrial function and suggest a number of ways by which this can theoretically be achieved. Since POAG is a chronic disease, any neuroprotective treatment strategy must be tolerated over many years. Theoretically, topically applied substances should have the fewest side effects, but it is questionable whether sufficient compounds can reach RGC mitochondria to be effective. Therefore, other delivery procedures that might result in greater concentrations of neuroprotectants reaching RGC mitochondria are being developed. Red-light therapy represents another therapeutic alternative for enhancing RGC mitochondrial functions and has the advantage of being both non-toxic and non-invasive.


Subject(s)
Glaucoma/drug therapy , Glaucoma/etiology , Mitochondria/drug effects , Neuroprotective Agents/pharmacology , Animals , Glaucoma/metabolism , Glaucoma/pathology , Humans , Mitochondria/metabolism , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology
2.
Regen Med ; 7(3): 309-22, 2012 May.
Article in English | MEDLINE | ID: mdl-22594325

ABSTRACT

AIMS: A major limiting factor for cell therapy in Parkinson's disease is that the survival of grafted dopaminergic neurons is very poor, which may be improved by administration of GDNF, for which the carotid body is a good source. MATERIALS & METHODS: Rats with total unilateral dopaminergic denervation were grafted with a cell suspension of rat dopaminergic neuroblasts with or without cell aggregates from the rat carotid body. At 1, 2 and 3 months after grafting, the rats were tested in the cylinder and the rotometer and killed 4 months after grafting. RESULTS: We observed that the survival of dopaminergic neurons and graft-derived dopaminergic innervation were higher in rats that received mixed grafts. Both grafted groups showed complete recovery in the amphetamine-induced rotation test. However, rats with cografts performed significantly better in the cylinder test. CONCLUSION: Cografting of carotid body cells may constitute a useful strategy for cell therapy in Parkinson's disease.


Subject(s)
Carotid Body/cytology , Carotid Body/transplantation , Dopaminergic Neurons/physiology , Dopaminergic Neurons/transplantation , Nerve Fibers/metabolism , Amphetamine , Animals , Cell Aggregation , Cell Survival , Dopaminergic Neurons/cytology , Female , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Mesencephalon/cytology , Mesencephalon/transplantation , Motor Activity/physiology , Neostriatum/cytology , Neostriatum/transplantation , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Rotation , Substantia Nigra/cytology , Time Factors , Tyrosine 3-Monooxygenase/metabolism
3.
Neurobiol Dis ; 47(2): 268-79, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22542954

ABSTRACT

It has recently been shown that the dopaminergic cell loss induced by neurotoxins is enhanced by brain angiotensin II (AII) via type 1 receptors (AT1). However, the mechanisms involved in the dopaminergic degeneration and the brain inflammatory effects of AII have not been clarified. The RhoA-Rho-Kinase (ROCK) pathway may play a critical role in the inflammatory and oxidative effects of AII. In the substantia nigra of mice, administration of the dopaminergic neurotoxin MPTP induced an increase in the expression of RhoA and ROCK II mRNA levels and ROCK activity, which were inhibited by AT1 receptor deletion (i.e., in AT1a null mice treated with MPTP). Administration of the ROCK inhibitor Y-27632 or AT1 deletion induced a significant decrease in MPTP-induced microglial activation and dopaminergic cell death. In rat primary mesencephalic cultures treated with MPP(+), the increase in dopaminergic cell loss induced by AII administration was also inhibited by treatment with Y27632. Intense expression of ROCK II was observed in the microglial cells in the substantia nigra of mice treated with MPTP, and the major role of the microglial ROCK was confirmed by comparing mesencephalic cultures with and without microglia. Activation of the RhoA/ROCK pathway is involved in the MPTP-induced dopaminergic degeneration, and in the enhancing effect of AII/AT1 activation on the microglial response and dopaminergic degeneration. ROCK inhibitors and AT1 receptor antagonists may provide new neuroprotective strategies against the progression of Parkinson's disease.


Subject(s)
Angiotensins/physiology , Dopaminergic Neurons/metabolism , Microglia/metabolism , Receptor, Angiotensin, Type 1/physiology , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , Amides/pharmacology , Amides/therapeutic use , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dopaminergic Neurons/enzymology , Dopaminergic Neurons/pathology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/enzymology , Microglia/pathology , Parkinson Disease/physiopathology , Parkinson Disease/prevention & control , Pyridines/pharmacology , Pyridines/therapeutic use , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein
4.
J Neuroinflammation ; 9: 38, 2012 Feb 22.
Article in English | MEDLINE | ID: mdl-22356806

ABSTRACT

BACKGROUND: Several recent studies have shown that angiotensin type 1 receptor (AT1) antagonists such as candesartan inhibit the microglial inflammatory response and dopaminergic cell loss in animal models of Parkinson's disease. However, the mechanisms involved in the neuroprotective and anti-inflammatory effects of AT1 blockers in the brain have not been clarified. A number of studies have reported that AT1 blockers activate peroxisome proliferator-activated receptor gamma (PPAR γ). PPAR-γ activation inhibits inflammation, and may be responsible for neuroprotective effects, independently of AT1 blocking actions. METHODS: We have investigated whether oral treatment with telmisartan (the most potent PPAR-γ activator among AT1 blockers) provides neuroprotection against dopaminergic cell death and neuroinflammation, and the possible role of PPAR-γ activation in any such neuroprotection. We used a mouse model of parkinsonism induced by the dopaminergic neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and co-administration of the PPAR-γ antagonist GW9662 to study the role of PPAR-γ activation. In addition, we used AT1a-null mice lesioned with MPTP to study whether deletion of AT1 in the absence of any pharmacological effect of AT1 blockers provides neuroprotection, and investigated whether PPAR-γ activation may also be involved in any such effect of AT1 deletion by co-administration of the PPAR-γ antagonist GW9662. RESULTS: We observed that telmisartan protects mouse dopaminergic neurons and inhibits the microglial response induced by administration of MPTP. The protective effects of telmisartan on dopaminergic cell death and microglial activation were inhibited by co-administration of GW9662. Dopaminergic cell death and microglial activation were significantly lower in AT1a-null mice treated with MPTP than in mice not subjected to AT1a deletion. Interestingly, the protective effects of AT1 deletion were also inhibited by co-administration of GW9662. CONCLUSION: The results suggest that telmisartan provides effective neuroprotection against dopaminergic cell death and that the neuroprotective effect is mediated by PPAR-γ activation. However, the results in AT1-deficient mice show that blockage of AT1, unrelated to the pharmacological properties of AT1 blockers, also protects against dopaminergic cell death and neuroinflammation. Furthermore, the results show that PPAR-γ activation is involved in the anti-inflammatory and neuroprotective effects of AT1 deletion.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Encephalitis/prevention & control , PPAR gamma/metabolism , Receptor, Angiotensin, Type 1/deficiency , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Analysis of Variance , Anilides/pharmacology , Animals , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Encephalitis/etiology , Encephalitis/pathology , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Lectins , Leukocyte Common Antigens/metabolism , MPTP Poisoning/chemically induced , MPTP Poisoning/complications , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , PPAR gamma/antagonists & inhibitors , Telmisartan , Tyrosine 3-Monooxygenase/metabolism
5.
Age (Dordr) ; 34(4): 863-80, 2012 Aug.
Article in English | MEDLINE | ID: mdl-21713375

ABSTRACT

Recent studies have shown that renin-angiotensin system overactivation is involved in the aging process in several tissues as well as in longevity and aging-related degenerative diseases by increasing oxidative damage and inflammation. We have recently shown that angiotensin II enhances dopaminergic degeneration by increasing levels of reactive oxygen species and neuroinflammation, and that there is an aging-related increase in angiotensin II activity in the substantia nigra in rats, which may constitute a major factor in the increased risk of Parkinson's disease with aging. The mechanisms involved in the above mentioned effects and particularly a potential angiotensin-mitochondria interaction have not been clarified. The present study revealed that activation of mitochondrial ATP-sensitive potassium channels [mitoK(ATP)] may play a major role in the angiotensin II-induced effects on aging and neurodegeneration. Inhibition of mitoK(ATP) channels with 5-hydroxydecanoic acid inhibited the increase in dopaminergic cell death induced by angiotensin II, as well as the increase in superoxide/superoxide-derived reactive oxygen species levels and the angiotensin II-induced decrease in the mitochondrial inner membrane potential in cultured dopaminergic neurons. The present study provides data for considering brain renin-angiotensin system and mitoK(ATP) channels as potential targets for protective therapy in aging-associated diseases such as Parkinson's disease.


Subject(s)
Aging/physiology , Dopaminergic Neurons/pathology , Oxidative Stress/drug effects , Oxidopamine/pharmacology , Potassium Channels/metabolism , Reactive Oxygen Species/metabolism , Angiotensin II/pharmacology , Animals , Cells, Cultured/drug effects , Cells, Cultured/metabolism , Dopaminergic Neurons/metabolism , Fluorescence , Humans , Immunohistochemistry , Male , Parkinson Disease/etiology , Parkinson Disease/physiopathology , Rats , Reference Values , Renin-Angiotensin System/drug effects , Sensitivity and Specificity , Substantia Nigra/drug effects , Substantia Nigra/metabolism
6.
Mol Neurodegener ; 6(1): 58, 2011 Aug 16.
Article in English | MEDLINE | ID: mdl-21846363

ABSTRACT

BACKGROUND: There are sex differences in dopaminergic degeneration. Men are approximately two times as likely as premenopausal women of the same age to develop Parkinson's disease (PD). It has been shown that the local renin angiotensin system (RAS) plays a prominent role in sex differences in the development of chronic renal and cardiovascular diseases, and there is a local RAS in the substantia nigra and dopaminergic cell loss is enhanced by angiotensin via type 1 (AT1) receptors. RESULTS: In the present study, we observed that intrastriatal injection of 6-hydroxydopamine induced a marked loss of dopaminergic neurons in the substantia nigra of male rats, which was significantly higher than the loss induced in ovariectomized female rats given estrogen implants (i.e. rats with estrogen). However, the loss of dopaminergic neurons was significantly lower in male rats treated with the AT1 antagonist candesartan, and similar to that observed in female rats with estrogen. The involvement of the RAS in gender differences in dopaminergic degeneration was confirmed with AT1a-null mice lesioned with the dopaminergic neurotoxin MPTP. Significantly higher expression of AT1 receptors, angiotensin converting enzyme activity, and NADPH-oxidase complex activity, and much lower levels of AT2 receptors were observed in male rats than in female rats with estrogen. CONCLUSIONS: The results suggest that brain RAS plays a major role in the increased risk of developing PD in men, and that manipulation of brain RAS may be an efficient approach for neuroprotective treatment of PD in men, without the feminizing effects of estrogen.

7.
J Neuropathol Exp Neurol ; 69(11): 1130-42, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20940627

ABSTRACT

Angiotensin II acts via angiotensin type 1 receptors and is a major inducer of inflammation and oxidative stress. Local renin-angiotensin systems play a major role in the development of age-related disorders in several tissues. These processes are delayed, but not totally abolished, by blockade of angiotensin signaling. A specific receptor for renin and its precursor prorenin has recently been identified. We previously showed that neurotoxin-induced dopaminergic (DA) cell loss is decreased by inhibition of angiotensin receptors, but the location and functional effects of prorenin receptor (PRR) in the brain, including the DA system, are unknown. In the substantia nigra of Macaca fascicularis and in rat primary mesencephalic cultures, double immunofluorescence analysis revealed PRR immunoreactivity in neurons (including DA neurons) and microglia, but not in astrocytes. Administration of the PRR blocker, handle region peptide, led to a significant decrease in 6-hydroxydopamine-induced DA cell death in the cultures,whereas administration of renin with simultaneous blockade of angiotensin receptors led to an increase in 6-hydroxydopamine-induced cell death. These results suggest that active agent angiotensin II-independent PRR intracellular signaling may contribute to exacerbation of DA cell death in vivo. Therefore, potential neuroprotective strategies for DA neurons in Parkinson disease should address both angiotensin and PRR signaling.


Subject(s)
Dopamine/metabolism , Macaca fascicularis/anatomy & histology , Receptors, Cell Surface/metabolism , Substantia Nigra/metabolism , Adrenergic Agents/toxicity , Animals , Cell Death/physiology , Cells, Cultured , Drug Interactions/physiology , Embryo, Mammalian , Glial Fibrillary Acidic Protein/metabolism , Imidazoles/pharmacology , Male , Mesencephalon/cytology , Naphthyridines/pharmacology , Neurons/drug effects , Oxidopamine/toxicity , Pyridines/pharmacology , Rats , Receptors, Cell Surface/genetics , Renin/pharmacology , Tyrosine 3-Monooxygenase/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , Prorenin Receptor
8.
J Neurochem ; 109(2): 656-69, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19245663

ABSTRACT

The neurotoxin MPTP reproduces most of the biochemical and pathological hallmarks of Parkinson's disease. In addition to reactive oxygen species (ROS) generated as a consequence of mitochondrial complex I inhibition, microglial NADPH-derived ROS play major roles in the toxicity of MPTP. However, the exact mechanism regulating this microglial response remains to be clarified. The peptide angiotensin II (AII), via type 1 receptors (AT1), is one of the most important inflammation and oxidative stress inducers, and produces ROS by activation of the NADPH-oxidase complex. Brain possesses a local angiotensin system, which modulates striatal dopamine (DA) release. However, it is not known if AII plays a major role in microglia-derived oxidative stress and DA degeneration. The present study indicates that in primary mesencephalic cultures, DA degeneration induced by the neurotoxin MPTP/MPP(+) is amplified by AII and inhibited by AT1 receptor antagonists, and that protein kinase C, NADPH-complex activation and microglial activation are involved in this effect. In mice, AT1 receptor antagonists inhibited both DA degeneration and early microglial and NADPH activation. The brain angiotensin system may play a key role in the self-propelling mechanism of Parkinson's disease and constitutes an unexplored target for neuroprotection, as previously reported for vascular diseases.


Subject(s)
Angiotensins/physiology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Inflammation Mediators/physiology , MPTP Poisoning/metabolism , Parkinson Disease, Secondary/metabolism , Parkinson Disease, Secondary/pathology , Angiotensins/toxicity , Animals , Cells, Cultured , Disease Progression , Inflammation Mediators/toxicity , MPTP Poisoning/pathology , Male , Mice , Mice, Inbred C57BL , Rats
SELECTION OF CITATIONS
SEARCH DETAIL