Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
2.
Diabetes ; 62(6): 1843-54, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23349486

ABSTRACT

Obesity is associated with increased adipose tissue macrophage (ATM) infiltration, and rodent studies suggest that inflammatory factors produced by ATMs contribute to insulin resistance and type 2 diabetes. However, a relationship between ATM content and insulin resistance has not been clearly established in humans. Since thiazolidinediones attenuate adipose tissue inflammation and improve insulin sensitivity, we examined the temporal relationship of the effects of pioglitazone on these two parameters. The effect of 10 and 21 days of pioglitazone treatment on insulin sensitivity in 26 diabetic subjects was assessed by hyperinsulinemic-euglycemic clamp studies. Because chemoattractant factors, cytokines, and immune cells have been implicated in regulating the recruitment of ATMs, we studied their temporal relationship to changes in ATM content. Improved hepatic and peripheral insulin sensitivity was seen after 21 days of pioglitazone. We found early reductions in macrophage chemoattractant factors after only 10 days of pioglitazone, followed by a 69% reduction in ATM content at 21 days and reduced ATM activation at both time points. Although markers for dendritic cells and neutrophils were reduced at both time points, there were no significant changes in regulatory T cells. These results are consistent with an association between adipose macrophage content and systemic insulin resistance in humans.


Subject(s)
Adipose Tissue/cytology , Hypoglycemic Agents/therapeutic use , Macrophages/drug effects , Thiazolidinediones/therapeutic use , Adipose Tissue/metabolism , Female , Flow Cytometry , Humans , Male , Middle Aged , Obesity/drug therapy , Obesity/metabolism , Pioglitazone
3.
Neurosci Lett ; 441(1): 44-9, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18597941

ABSTRACT

Dopaminergic neurons in the substantia nigra (SN) selectively die in Parkinson's disease (PD), but it is unclear how and why this occurs. Recent findings implicate prostaglandin E(2) (PGE(2)) and two of its four receptors, namely EP1 and EP2, as mediators of degenerative and protective events in situations of acute and chronic neuronal death. EP1 activation can exacerbate excitotoxic damage in stroke models and our recent study showed that EP1 activation may explain the selective sensitivity of dopaminergic neurons to oxidative stress. Conversely, EP2 activation may be neuroprotective, although toxic effects have also been demonstrated. Here we investigated if and how EP2 activation might alter the survival of dopaminergic neurons following selective low-level oxidative injury evoked by the neurotoxin 6-hydroxydopamine (6-OHDA) in primary neuronal cultures prepared from embryonic rat midbrain. We found that cultured dopaminergic neurons displayed EP2 receptors. Butaprost, a selective EP2 agonist, significantly reduced 6-OHDA neurotoxicity. EP2 receptors are coupled to stimulatory G-proteins (Gs), which activate adenylate cyclase, increasing cAMP synthesis, which then activates protein kinase A (PKA). Both dibutyryl cAMP and forskolin reduced dopaminergic cell loss after 6-OHDA exposure. Conversely, KT5720 and H-89, two structurally distinct high-affinity PKA inhibitors, abolished the protective effect of butaprost, implicating cAMP-dependent PKA activity in the neuroprotection by EP2 activation. Finally, we show that melanized dopaminergic neurons in the human SN express EP2. This pathway warrants consideration as a neuroprotective strategy for PD.


Subject(s)
Adrenergic Agents/toxicity , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Oxidopamine/toxicity , Receptors, Prostaglandin E/physiology , Alprostadil/analogs & derivatives , Alprostadil/pharmacology , Animals , Cells, Cultured , Colforsin/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Humans , Mesencephalon/cytology , Rats , Receptors, Prostaglandin E, EP2 Subtype , Substantia Nigra/pathology , Tyrosine 3-Monooxygenase/metabolism
4.
J Neurosci Res ; 85(14): 3109-17, 2007 Nov 01.
Article in English | MEDLINE | ID: mdl-17868147

ABSTRACT

Oxidative stress and increased cyclooxygenase-2 (COX-2) activity are both implicated in the loss of dopaminergic neurons from the substantia nigra (SN) in idiopathic Parkinson's disease (PD). Prostaglandin E(2) (PGE(2)) is one of the key products of COX-2 activity and PGE(2) production is increased in PD. However, little is known about its role in the selective death of dopaminergic neurons. Previously, we showed that oxidative stress evoked by low concentrations of 6-hydroxydopamine (6-OHDA) was selective for dopaminergic neurons in culture and fully dependent on COX-2 activity. We postulated that this loss was mediated by PGE(2) acting through its receptors, EP1, EP2, EP3, and EP4. Using double-label immunohistochemistry for specific EP receptors and tyrosine hydroxylase (TH), we identified EP1 and EP2 receptors on dopaminergic neurons in rat SN. EP2 receptors were also found in non-dopaminergic neurons of this nucleus, as were EP3 receptors, whereas the EP4 receptor was absent. PGE(2), 16-phenyl tetranor PGE(2) (a stable synthetic analogue), and 17-phenyl trinor PGE(2) (an EP1 receptor-selective agonist) were significantly toxic to dopaminergic cells at nanomolar concentrations; EP2- and EP3-selective agonists were not. We challenged dopaminergic neurons in embryonic rat mesencephalic primary neuronal cultures and tested whether these receptors mediate selective 6-OHDA toxicity. The nonselective EP1-3 receptor antagonist AH-6809 and two selective EP1 antagonists, SC-19220 and SC-51089, completely prevented the 40%-50% loss of dopaminergic neurons caused by exposure to 5 muM 6-OHDA. Together, these results strongly implicate PGE(2) activation of EP1 receptors as a mediator of selective toxicity in this model of dopaminergic cell loss.


Subject(s)
Dinoprostone/toxicity , Dopamine/metabolism , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/drug effects , Receptors, Prostaglandin E/physiology , Analysis of Variance , Animals , Cell Count/methods , Cells, Cultured , Dose-Response Relationship, Drug , Embryo, Mammalian , Oxidopamine/toxicity , Prostaglandin Antagonists/pharmacology , Prostaglandins, Synthetic/pharmacology , Rats , Receptors, Prostaglandin E/agonists , Receptors, Prostaglandin E/antagonists & inhibitors , Receptors, Prostaglandin E, EP1 Subtype , Substantia Nigra/cytology , Sympatholytics/toxicity , Tyrosine 3-Monooxygenase/metabolism
5.
J Neurosci Res ; 81(1): 121-31, 2005 Jul 01.
Article in English | MEDLINE | ID: mdl-15931668

ABSTRACT

Cyclooxygenase (COX), a key enzymatic mediator of inflammation, is present in microglia and surviving dopaminergic neurons in Parkinson's disease (PD), but its role and place in the chain of neurodegenerative events is unclear. Epidemiologic evidence showed that regular use of nonsteroidal antiinflammatory drugs (NSAIDs), specifically non-aspirin COX inhibitors like ibuprofen, lowers the risk for PD; however, the putative cause-and-effect relationship between COX activity in activated microglia and neuronal loss was challenged recently. We examined whether neuronal COX activity is involved directly in dopaminergic cell death after neurotoxic insult. Using low concentrations of 6-hydroxydopamine (6-OHDA) and 1-methyl-4-phenylpyridium ion (MPP+), neurotoxicants used to model selective dopaminergic cell loss in PD, and cultures of embryonic rat mesencephalic neurons essentially devoid of glia, we tested whether the nonselective COX inhibitor ibuprofen attenuated 6-OHDA and MPP+ neurotoxicity. At levels close to its IC50 for both COX isoforms, ibuprofen protected dopaminergic neurons against 6-OHDA but not MPP+ toxicity. Experiments with selective inhibitors of COX-1 (SC-560) and COX-2 (NS-398 and Cayman 10404), indicated that COX-2, but not COX-1, was involved in 6-OHDA toxicity. Accordingly, 6-OHDA, but not MPP+, increased prostaglandin (PG) levels twofold and this increase was blocked by ibuprofen. At concentrations well above its IC50 for COX, ibuprofen also prevented MPP+ toxicity, but had only limited efficacy against loss of structural complexity. Taken together, our data suggest that selective 6-OHDA toxicity to dopaminergic neurons is associated with neuronal COX-2, whereas MPP+ toxicity is COX independent. This difference may be important for understanding and manipulating mechanisms of dopaminergic cell death.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Cyclooxygenase Inhibitors/pharmacology , Ibuprofen/pharmacology , Neurons/drug effects , Oxidopamine/toxicity , Prostaglandin-Endoperoxide Synthases/drug effects , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Dopamine/metabolism , Isoenzymes , Nerve Degeneration/enzymology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Neurotoxins/toxicity , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/metabolism , Rats
6.
Brain Res Dev Brain Res ; 140(1): 1-13, 2003 Jan 10.
Article in English | MEDLINE | ID: mdl-12524172

ABSTRACT

It has been established that thyroid hormone and neurotrophic factors both orchestrate developmental events in the brain. However, it is not clear how these two influences are related. In this study, we investigated the effects of thyroid hormone on cerebellar development and the coincident expression of transforming growth factor-alpha (TGF-alpha), a ligand in the epidermal growth factor (EGF) family, and the epidermal growth factor receptor (EGFR). Profiles of thyroid hormone expression were measured in postnatal animals and were found to peak at postnatal day 15 (P15). These levels dropped below detectable levels when mice were made hypothyroid with propylthiouracil (PTU). TGF-alpha and EGFR expression, as determined by RNAse protection assay, was maximal at P6 in normal animals, but remained low in hypothyroid animals, suggesting that thyroid hormone was responsible for their induction. In situ hybridization and immunohistochemical analysis of EGFR expression revealed that this receptor was present on granule cells within the inner zone of the external granule cell layer (EGL), suggesting that EGFR-ligands were not inducing granule cell proliferation. The persistence of EGFR expression on migrating granule cells and subsequent down-regulation of expression in the internal granule cell layer (IGL) implicates a role for EGFR-ligands in differentiation and/or migration. In hypothyroid animals, we observed a delayed progression of granule cell migration, consistent with the persistence of EGFR labeling in the EGL, and in the 'pile-up' of labeled cells at the interface between the molecular layer and the Purkinje cell layer. Taken together, these results implicate thyroid hormone in the coordinated expression of TGF-alpha and EGFR, which are positioned to play a role in post-mitotic developmental events in the cerebellum.


Subject(s)
Cerebellum/growth & development , ErbB Receptors/genetics , Gene Expression Regulation/physiology , Hypothyroidism/physiopathology , Thyroid Gland/physiology , Thyroxine/blood , Aging , Animals , Cerebellum/cytology , Female , Glial Fibrillary Acidic Protein/analysis , Hypothyroidism/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mitosis , Propylthiouracil , Reference Values , Transcription, Genetic
7.
Parkinsonism Relat Disord ; 8(6): 407-11, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12217628

ABSTRACT

We optimized a mesencephalic cell culture system to employ low concentrations of 6-hydroxydopamine (6-OHDA) and 1-methyl-4 phenylpyridinium (MPP+), neurotoxins known to trigger oxidative stress in dopaminergic cells. Both 6-OHDA and MPP(+) at 5 micro M reproducibly reduced the survival of dopaminergic neurons by 50-70% (p<0.02) without affecting the survival of the non-dopaminergic neuronal population. We found that 1mM of the non-steroidal anti-inflammatory drug (NSAID), acetylsalicylic acid (ASA), significantly (p<0.05) increased the survival of dopaminergic neurons exposed to either neurotoxin. The mechanisms underlying neuroprotection by ASA may be of therapeutic import in Parkinson's disease.


Subject(s)
1-Methyl-4-phenylpyridinium/toxicity , Aspirin/pharmacology , Dopamine/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxidopamine/toxicity , Animals , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Dopamine/analysis , Dose-Response Relationship, Drug , Female , Neurons/chemistry , Neurons/pathology , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...