Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Neuropharmacology ; 79: 249-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24316465

ABSTRACT

Sartans (Angiotensin II AT(1) Receptor Blockers, ARBs) are powerful neuroprotective agents in vivo and protect against IL-1ß neurotoxicity in vitro. The purpose of this research was to determine the extent of sartan neuroprotection against glutamate excitotoxicity, a common cause of neuronal injury and apoptosis. The results show that sartans are neuroprotective, significantly reducing glutamate-induced neuronal injury and apoptosis in cultured rat primary cerebellar granule cells (CGCs). Telmisartan was the most potent sartan studied, with an order of potency telmisartan > candesartan > losartan > valsartan. Mechanisms involved reduction of pro-apoptotic caspase-3 activation, protection of the survival PI3K/Akt/GSK-3ß pathway and prevention of glutamate-induced ERK1/2 activation. NMDA receptor stimulation was essential for glutamate-induced cell injury and apoptosis. Participation of AT(1A) receptor was supported by glutamate-induced upregulation of AT(1A) gene expression and AT(1) receptor binding. Conversely, AT(1B) or AT(2) receptors played no role. Glutamate-induced neuronal injury and the neuroprotective effect of telmisartan were decreased, but not abolished, in CGCs obtained from AT(1A) knock-out mice. This indicates that although AT(1) receptors are necessary for glutamate to exert its full neurotoxic potential, part of the neuroprotective effect of telmisartan is independent of AT(1) receptor blockade. PPARγ activation was also involved in the neuroprotective effects of telmisartan, as telmisartan enhanced PPARγ nuclear translocation and the PPARγ antagonist GW9662 partially reversed the neuroprotective effects of telmisartan. The present results substantiate the therapeutic use of sartans, in particular telmisartan, in neurodegenerative diseases and traumatic brain disorders where glutamate neurotoxicity plays a significant role.


Subject(s)
Benzimidazoles/pharmacology , Benzoates/pharmacology , Glutamic Acid/toxicity , Neurons/drug effects , Neuroprotective Agents/pharmacology , PPAR gamma/metabolism , Receptor, Angiotensin, Type 1/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cells, Cultured , Cerebellum/drug effects , Cerebellum/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/physiology , PPAR gamma/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1/genetics , Telmisartan
2.
Neuropsychopharmacology ; 37(13): 2817-29, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22892395

ABSTRACT

Traumatic brain injury (TBI) results in complex pathological reactions, the initial lesion worsened by secondary inflammation and edema. Angiotensin II (Ang II) is produced in the brain and Ang II receptor type 1 (AT1R) overstimulation produces vasoconstriction and inflammation. Ang II receptor blockers (ARBs) are neuroprotective in models of stroke but little is known of their effect when administered in TBI models. We therefore performed controlled cortical impact (CCI) injury on mice to investigate whether the ARB candesartan would mitigate any effects of TBI. We administered candesartan or vehicle to mice 5 h before CCI injury. Candesartan treatment reduced the lesion volume after CCI injury by approximately 50%, decreased the number of dying neurons, lessened the number of activated microglial cells, protected cerebral blood flow (CBF), and reduced the expression of the cytokine TGFß1 while increasing expression of TGFß3. Candesartan-treated mice also showed better motor skills on the rotarod 3 days after injury, and improved performance in the Morris water maze 4 weeks after injury. These results indicate that candesartan is neuroprotective, reducing neuronal injury, decreasing lesion volume and microglial activation, protecting CBF and improving functional behavior in a mouse model of TBI. Co-treatment with a peroxisome proliferator-activated receptor-gamma (PPARγ) antagonist significantly reduced some of the beneficial effects of candesartan after CCI, suggesting that PPARγ activation may contribute to part or to all of the neuroprotective effect of candesartan. Overall, our data suggest that ARBs with dual AT1R-blocking and PPARγ activation properties may have therapeutic value in treating TBI.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Benzimidazoles/administration & dosage , Brain Injuries/drug therapy , Memory/drug effects , Motor Skills/drug effects , PPAR gamma/agonists , Tetrazoles/administration & dosage , Animals , Biphenyl Compounds , Brain Injuries/pathology , Infusion Pumps, Implantable , Male , Maze Learning/drug effects , Maze Learning/physiology , Memory/physiology , Mice , Mice, Inbred C57BL , Motor Skills/physiology , PPAR gamma/physiology , Random Allocation
3.
Cell Mol Neurobiol ; 32(8): 1353-65, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22843099

ABSTRACT

Commercially available Angiotensin II AT1 receptor antibodies are widely employed for receptor localization and quantification, but they have not been adequately validated. In this study, six commercially available AT1 receptor antibodies were characterized by established criteria: sc-1173 and sc-579 from Santa Cruz Biotechnology, Inc., AAR-011 from Alomone Labs, Ltd., AB15552 from Millipore, and ab18801 and ab9391 from Abcam. The immunostaining patterns observed were different for every antibody tested, and were unrelated to the presence or absence of AT1 receptors. The antibodies detected a 43 kDa band in western blots, corresponding to the predicted size of the native AT1 receptor. However, identical bands were observed in wild-type mice and in AT1A knock-out mice not expressing the target protein. Moreover, immunoreactivity detected in rat hypothalamic 4B cells not expressing AT1 receptors or transfected with AT1A receptor construct was identical, as revealed by western blotting and immunocytochemistry in cultured 4B cells. Additional prominent immunoreactive bands above and below 43 kDa were observed by western blotting in extracts from tissues of AT1A knock-out and wild-type mice and in 4B cells with or without AT1 receptor expression. In all cases, the patterns of immunoreactivity were independent of the AT1 receptor expression and different for each antibody studied. We conclude that, in our experimental setup, none of the commercially available AT1 receptor antibodies tested met the criteria for specificity and that competitive radioligand binding remains the only reliable approach to study AT1 receptor physiology in the absence of full antibody characterization.


Subject(s)
Antibodies/metabolism , Receptor, Angiotensin, Type 1/immunology , Receptor, Angiotensin, Type 1/metabolism , Animals , Binding Sites, Antibody/immunology , Cells, Cultured , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Protein Binding/immunology , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/biosynthesis
4.
J Neuroinflammation ; 9: 102, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22642771

ABSTRACT

BACKGROUND: Blockade of angiotensin II type 1 (AT1) receptors ameliorates brain inflammation, and reduces excessive brain interleukin-1 beta (IL-1ß) production and release from cortical microglia. The aim of this study was to determine whether, in addition, AT1 receptor blockade directly attenuates IL-1ß-induced inflammatory responses in neuronal cultures. METHODS: SK-N-SH human neuroblasts and primary rat cortical neurons were pretreated with telmisartan followed by exposure to IL-1ß. Gene expression was determined by reverse transcriptase (RT)-PCR, protein expression and kinase activation by western blotting, NADPH oxidase activity by the lucigenin method, prostaglandin E2 (PGE2) release by enzyme immunoassay, reactive oxygen species (ROS) generation by the dichlorodihydrofluorescein diacetate fluorescent probe assay, and peroxisome proliferator-activated receptor gamma (PPARγ) involvement was assessed with the antagonists GW9662 and T0070907, the agonist pioglitazone and the expression of PPARγ target genes ABCG1 and CD36. RESULTS: We found that SK-N-SH neuroblasts expressed AT1 but not AT2 receptor mRNA. Telmisartan reduced IL-1ß-induced cyclooxygenase-2 (COX-2) expression and PGE2 release more potently than did candesartan and losartan. Telmisartan reduced the IL-1ß-induced increase in IL-1R1 receptor and NADPH oxidase-4 (NOX-4) mRNA expression, NADPH oxidase activity, and ROS generation, and reduced hydrogen peroxide-induced COX-2 gene expression. Telmisartan did not modify IL-1ß-induced ERK1/2 and p38 mitogen-activated protein kinase (MAPK) phosphorylation or nuclear factor-κB activation but significantly decreased IL-1ß-induced c-Jun N-terminal kinase (JNK) and c-Jun activation. The JNK inhibitor SP600125 decreased IL-1ß-induced PGE2 release with a potency similar to that of telmisartan. The PPARγ agonist pioglitazone reduced IL-1ß-induced inflammatory reaction, whereas telmisartan did not activate PPARγ, as shown by its failure to enhance the expression of the PPARγ target genes ABCG1 and CD36, and the inability of the PPARγ antagonists GW9662 and T0070907 to modify the effect of telmisartan on COX-2 induction. The effect of telmisartan on IL-1ß-stimulated COX-2 and IL-1R1 mRNA expression and ROS production was replicated in primary rat cortical neurons. CONCLUSIONS: Telmisartan directly ameliorates IL-1ß-induced neuronal inflammatory response by inhibition of oxidative stress and the JNK/c-Jun pathway. Our results support the hypothesis that AT1 receptor blockers are directly neuroprotective, and should be considered for the treatment of inflammatory conditions of the brain.


Subject(s)
Benzimidazoles/pharmacology , Benzoates/pharmacology , Inflammation Mediators/pharmacology , Interleukin-1beta/toxicity , MAP Kinase Signaling System/physiology , NADPH Oxidases/physiology , Neurons/metabolism , Neurons/pathology , Proto-Oncogene Proteins c-jun/physiology , Animals , Benzimidazoles/therapeutic use , Benzoates/therapeutic use , Cell Line , Cells, Cultured , Humans , Inflammation Mediators/immunology , Inflammation Mediators/therapeutic use , Interleukin-1beta/antagonists & inhibitors , MAP Kinase Signaling System/immunology , Neurons/enzymology , Rats , Rats, Sprague-Dawley , Telmisartan
5.
Behav Brain Res ; 232(1): 84-92, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22503782

ABSTRACT

Centrally acting Angiotensin II AT(1) receptor blockers (ARBs) protect from stress-induced disorders and decrease anxiety in a model of inflammatory stress, the systemic injection of bacterial endotoxin lipopolysaccharide (LPS). In order to better understand the anxiolytic effect of ARBs, we treated rats with LPS (50 µg/kg) with or without 3 days of pretreatment with the ARB candesartan (1mg/kg/day), and studied cortical benzodiazepine (BZ) and corticotrophin-releasing factor (CRF) receptors. We compared the cortical BZ and CRF receptors expression pattern induced by LPS with that produced in restraint stress. Inflammation stress produced a generalized increase in cortical BZ(1) receptors and reduced mRNA expression of the GABA(A) receptor γ(2) subunit in cingulate cortex; changes were prevented by candesartan pretreatment. Moreover, restraint stress produced similar increases in cortical BZ(1) receptor binding, and candesartan prevented these changes. Treatment with candesartan alone increased cortical BZ(1) binding, and decreased γ(2) subunit mRNA expression in the cingulate cortex. Conversely, we did not find changes in CRF(1) receptor expression in any of the cortical areas studied, either after inflammation or restraint stress. Cortical CRF(2) receptor binding was undetectable, but CRF(2) mRNA expression was decreased by inflammation stress, a change prevented by candesartan. We conclude that stress promotes rapid and widespread changes in cortical BZ(1) receptor expression; and that the stress-induced BZ(1) receptor expression is under the control of AT(1) receptor activity. The results suggest that the anti-anxiety effect of ARBs may be associated with their capacity to regulate stress-induced alterations in cortical BZ(1) receptors.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Cerebral Cortex/metabolism , Inflammation/metabolism , Receptors, GABA-A/biosynthesis , Stress, Psychological/metabolism , Tetrazoles/pharmacology , Up-Regulation/drug effects , Adrenocorticotropic Hormone/blood , Animals , Autoradiography , Biphenyl Compounds , Cerebral Cortex/drug effects , Corticosterone/blood , Inflammation/chemically induced , Inflammation/psychology , Lipopolysaccharides/pharmacology , Male , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Corticotropin-Releasing Hormone/drug effects , Receptors, GABA-A/drug effects , Restraint, Physical
6.
J Hypertens ; 30(1): 87-96, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22124178

ABSTRACT

OBJECTIVE: Angiotensin II type 1 receptor (AT1) blockers (ARBs) reduce the bacterial endotoxin lipopolysaccharide (LPS)-induced innate immune response in human circulating monocytes expressing few AT1. To clarify the mechanisms of anti-inflammatory effects of ARBs with different peroxisome proliferator-activated receptor-γ (PPARγ)-activating potencies, we focused our study on telmisartan, an ARB with the highest PPARγ-stimulating activity. METHODS: Human circulating monocytes and monocytic THP-1 (human acute monocytic leukemia cell line) cells were exposed to 50 ng/ml LPS with or without pre-incubation with telmisartan. AT1 mRNA and protein expressions were determined by real-time PCR and membrane receptor binding assay, respectively. The expression of pro-inflammatory factors was determined by real-time PCR, western blot analysis and ELISA. PPARγ activation was measured by electrophoretic mobility shift assay and its role was determined by pharmacological inhibition and PPARγ gene silencing. RESULTS: In human monocytes, telmisartan significantly attenuated the LPS-induced expression of pro-inflammatory factors, the release of pro-inflammatory cytokines and prostaglandin E2, nuclear factor-κB activation and reactive oxygen species formation. In THP-1 cells, telmisartan significantly reduced LPS-induced tumor necrosis factor-α, inhibitor of κB-α, monocyte chemotactic protein-1 (MCP-1) and lectin-like oxidized low-density lipoprotein receptor-1 gene expression and MCP-1-directed migration. Telmisartan also stimulated the expression of the PPARγ target genes cluster of differentiation 36 and ATP-binding cassette subfamily G member 1 in monocytes. The anti-inflammatory effects of telmisartan were prevented by both PPARγ antagonism and PPARγ gene silencing. Anti-inflammatory effects of ARBs correlated with their PPARγ agonist potency. CONCLUSION: Our observations demonstrate that in human monocytes, ARBs inhibit the LPS-induced pro-inflammatory response to a major extent through the PPARγ activation pathway and may be beneficial for the treatment of cardiovascular and metabolic disorders in which inflammation plays a major role.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Benzoates/pharmacology , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Monocytes/drug effects , PPAR gamma/agonists , Base Sequence , Blotting, Western , Cell Line, Tumor , DNA Probes , Electrophoretic Mobility Shift Assay , Humans , Monocytes/immunology , Real-Time Polymerase Chain Reaction , Telmisartan
7.
Psychoneuroendocrinology ; 36(1): 1-18, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21035950

ABSTRACT

Poor adaptation to stress, alterations in cerebrovascular function and excessive brain inflammation play critical roles in the pathophysiology of many psychiatric and neurological disorders such as major depression, schizophrenia, post traumatic stress disorder, Parkinson's and Alzheimer's diseases and traumatic brain injury. Treatment for these highly prevalent and devastating conditions is at present very limited and many times inefficient, and the search for novel therapeutic options is of major importance. Recently, attention has been focused on the role of a brain regulatory peptide, Angiotensin II, and in the translational value of the blockade of its physiological AT(1) receptors. In addition to its well-known cardiovascular effects, Angiotensin II, through AT(1) receptor stimulation, is a pleiotropic brain modulatory factor involved in the control of the reaction to stress, in the regulation of cerebrovascular flow and the response to inflammation. Excessive brain AT(1) receptor activity is associated with exaggerated sympathetic and hormonal response to stress, vulnerability to cerebrovascular ischemia and brain inflammation, processes leading to neuronal injury. In animal models, inhibition of brain AT(1) receptor activity with systemically administered Angiotensin II receptor blockers is neuroprotective; it reduces exaggerated stress responses and anxiety, prevents stress-induced gastric ulcerations, decreases vulnerability to ischemia and stroke, reverses chronic cerebrovascular inflammation, and reduces acute inflammatory responses produced by bacterial endotoxin. These effects protect neurons from injury and contribute to increase the lifespan. Angiotensin II receptor blockers are compounds with a good margin of safety widely used in the treatment of hypertension and their anti-inflammatory and vascular protective effects contribute to reduce renal and cardiovascular failure. Inhibition of brain AT(1) receptors in humans is also neuroprotective, reducing the incidence of stroke, improving cognition and decreasing the progression of Alzheimer's disease. Blockade of AT(1) receptors offers a novel and safe therapeutic approach for the treatment of illnesses of increasing prevalence and socioeconomic impact, such as mood disorders and neurodegenerative diseases of the brain.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Anxiety/drug therapy , Brain Ischemia/drug therapy , Encephalitis/drug therapy , Stress, Psychological/drug therapy , Animals , Anxiety/complications , Brain Ischemia/complications , Encephalitis/complications , Humans , Models, Biological , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/physiology , Stress, Psychological/complications
8.
Neuropsychopharmacology ; 36(4): 857-70, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21150913

ABSTRACT

Brain inflammation has a critical role in the pathophysiology of brain diseases of high prevalence and economic impact, such as major depression, schizophrenia, post-traumatic stress disorder, Parkinson's and Alzheimer's disease, and traumatic brain injury. Our results demonstrate that systemic administration of the centrally acting angiotensin II AT(1) receptor blocker (ARB) candesartan to normotensive rats decreases the acute brain inflammatory response to administration of the bacterial endotoxin lipopolysaccharide (LPS), a model of brain inflammation. The broad anti-inflammatory effects of candesartan were seen across the entire inflammatory cascade, including decreased production and release to the circulation of centrally acting proinflammatory cytokines, repression of nuclear transcription factors activation in the brain, reduction of gene expression of brain proinflammatory cytokines, cytokine and prostanoid receptors, adhesion molecules, proinflammatory inducible enzymes, and reduced microglia activation. These effects are widespread, occurring not only in well-known brain target areas for circulating proinflammatory factors and LPS, that is, hypothalamic paraventricular nucleus and the subfornical organ, but also in the prefrontal cortex, hippocampus, and amygdala. Candesartan reduced the associated anorexic effects, and ameliorated associated body weight loss and anxiety. Direct anti-inflammatory effects of candesartan were also documented in cultured rat microglia, cerebellar granule cells, and cerebral microvascular endothelial cells. ARBs are widely used in the treatment of hypertension and stroke, and their anti-inflammatory effects contribute to reduce renal and cardiac failure. Our results indicate that these compounds may offer a novel and safe therapeutic approach for the treatment of brain disorders.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Brain/metabolism , Brain/pathology , Receptor, Angiotensin, Type 1/metabolism , Animals , Animals, Newborn , Benzimidazoles/administration & dosage , Benzimidazoles/pharmacology , Biphenyl Compounds , Brain/drug effects , Female , Inflammation/metabolism , Inflammation/pathology , Inflammation/prevention & control , Lipopolysaccharides/antagonists & inhibitors , Lipopolysaccharides/toxicity , Male , Rats , Rats, Inbred SHR , Rats, Sprague-Dawley , Rats, Wistar , Tetrazoles/administration & dosage , Tetrazoles/pharmacology
9.
J Hypertens ; 27(12): 2365-76, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19730394

ABSTRACT

OBJECTIVE: Inhibition of angiotensin II receptor type 1 (AT1) reduces chronic inflammation associated with hypertension. We asked whether AT1 receptor inhibition would reduce the innate inflammatory response induced by bacterial lipopolysaccharide (LPS). METHODS: We used unstimulated human circulating monocytes obtained from healthy donors by counterflow centrifugal elutriation. Monocytes were studied in vitro after incubation with LPS (50 ng/ml) with and without 1 mumol/l candesartan, an AT1 receptor blocker. Angiotensin II receptor mRNA expression was determined by reverse transcriptase-PCR and receptor binding by autoradiography; inflammatory factor mRNA expression was studied by reverse transcriptase-PCR and cytokine release by ELISA. RESULTS: Human monocytes did not express detectable AT1 receptors, and angiotensin II did not induce inflammatory factor mRNA expression or cytokine release. However, candesartan substantially reduced the LPS-induced expression of the mRNAs for the LPS recognition protein cluster of differentiation 14, the proinflammatory cytokines tumor necrosis factor alpha, interleukin-1 beta and interleukin-6 and the lectin-like oxidized low-density lipoprotein receptor. In addition, candesartan reduced the activation of the nuclear factor kappa B pathway, the tumor necrosis factor alpha and interleukin-6 secretion, and the ROS formation induced by LPS, without affecting the secretion of interleukin-10. CONCLUSION: We hypothesize that the anti-inflammatory effects of candesartan in these cells are likely mediated by mechanisms unrelated to AT1 receptor blockade. Our results demonstrate that candesartan significantly reduces the innate immune response to LPS in human circulating monocytes. The anti-inflammatory effects of candesartan may be of importance not only in hypertension but also in other inflammatory disorders.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Monocytes/drug effects , Receptor, Angiotensin, Type 1/drug effects , Tetrazoles/pharmacology , Biphenyl Compounds , Cells, Cultured , Cytokines/genetics , Cytokines/metabolism , Drug Therapy, Combination , Gene Expression/drug effects , Humans , Immunity, Innate/physiology , Monocytes/immunology , RNA, Messenger/metabolism , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/genetics , Receptor, Angiotensin, Type 1/metabolism
10.
Brain Behav Immun ; 23(7): 945-57, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19427376

ABSTRACT

Systemic lipopolysaccharide (LPS) administration induces an innate immune response and stimulates the hypothalamic-pituitary-adrenal axis. We studied Angiotensin II AT(1) receptor participation in the LPS effects with focus on the pituitary gland. LPS (50 microg/kg, i.p.) enhanced, 3h after administration, gene expression of pituitary CD14 and that of Angiotensin II AT(1A) receptors in pituitary and hypothalamic paraventricular nucleus (PVN); stimulated ACTH and corticosterone release; decreased pituitary CRF(1) receptor mRNA and increased all plasma and pituitary pro-inflammatory factors studied. The AT(1) receptor blocker (ARB) candesartan (1mg/kg/day, s.c. daily for 3 days before LPS) blocked pituitary and PVN AT(1) receptors, inhibited LPS-induced ACTH but not corticosterone secretion and decreased LPS-induced release of TNF-alpha, IL-1beta and IL-6 to the circulation. The ARB reduced LPS-induced pituitary gene expression of IL-6, LIF, iNOS, COX-2 and IkappaB-alpha; and prevented LPS-induced increase of nNOS/eNOS activity. The ARB did not affect LPS-induced TNF-alpha and IL-1beta gene expression, IL-6 or IL-1beta protein content or LPS-induced decrease of CRF(1) receptors. When administered alone, the ARB increased basal plasma corticosterone levels and basal PGE(2) mRNA in pituitary. Our results demonstrate that the pituitary gland is a target for systemically administered LPS. AT(1) receptor activity is necessary for the complete pituitary response to LPS and is limited to specific pro-inflammatory pathways. There is a complementary and complex influence of the PVN and circulating cytokines on the initial pituitary response to LPS. Our findings support the proposal that ARBs may be considered for the treatment of inflammatory conditions.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Cytokines/blood , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Receptor, Angiotensin, Type 1/metabolism , Adrenocorticotropic Hormone/blood , Analysis of Variance , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Autoradiography , Benzimidazoles/pharmacology , Biphenyl Compounds , Corticosterone/blood , Corticosterone/metabolism , Cytokines/genetics , Dinoprostone/genetics , Enzyme-Linked Immunosorbent Assay , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , In Situ Hybridization , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Male , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/drug effects , Pituitary-Adrenal System/metabolism , RNA, Messenger/physiology , Radioimmunoassay , Rats , Rats, Wistar , Receptor, Angiotensin, Type 1/immunology , Reverse Transcriptase Polymerase Chain Reaction , Tetrazoles/pharmacology
11.
Cell Mol Neurobiol ; 29(6-7): 781-92, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19259805

ABSTRACT

In addition to regulating blood pressure, angiotensin II (Ang II) exerts powerful pro-inflammatory effects in hypertension through stimulation of its AT(1) receptors, most clearly demonstrated in peripheral arteries and in the cerebral vasculature. Administration of Ang II receptor blockers (ARBs) decreases hypertension-related vascular inflammation in peripheral organs. In rodent models of genetic hypertension, ARBs reverse the inflammation in the cerebral microcirculation. We hypothesized that ARBs could be effective in inflammatory conditions beyond hypertension. Our more recent studies, summarized here, indicate that this is indeed the case. We used the model of systemic administration of the bacterial endotoxin lipopolysaccharide (LPS). LPS produces a robust initial inflammatory reaction, the innate immune response, in peripheral organs and in the brain. Pretreatment with the ARB candesartan significantly diminishes the response to LPS, including reduction of pro-inflammatory cytokine release to the general circulation and decreased production and release of the pro-inflammatory adrenal hormone aldosterone. In addition, the ARB very significantly decreased the LPS-induced gene expression of pro-inflammatory cytokines and microglia activation in the brain. Our results demonstrate that AT(1) receptor activity is essential for the unrestricted development of full-scale innate immune response in the periphery and in the brain. ARBs, due to their immune response-limiting properties, may be considered as therapeutically useful in a number of inflammatory diseases of the peripheral organs and the brain.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Arteritis/drug therapy , Encephalitis/drug therapy , Animals , Chronic Disease , Hypertension/drug therapy , Microglia , Renin-Angiotensin System
12.
Am J Physiol Regul Integr Comp Physiol ; 296(5): R1376-84, 2009 May.
Article in English | MEDLINE | ID: mdl-19225144

ABSTRACT

ANG II AT(1) receptor blockade reduces inflammation in hypertension. To determine whether ANG II AT(1) receptor blockers (ARBs) influence the innate immune inflammatory response in normotensive rats, we studied rat plasma and spleen after a 3-day subcutaneous pretreatment with the ARB candesartan followed by a single dose of the bacterial endotoxin LPS (50 microg/kg ip). Peripheral administration of LPS to rodents produced a generalized inflammatory response with increased release of TNF-alpha, IL-1beta, and IL-6 into the circulation. Candesartan pretreatment reduced the LPS-induced release of TNF-alpha, IL-1beta, and IL-6 into the circulation. The red pulp of rat spleen expressed large numbers of AT(1) receptors and the LPS receptors Toll-like receptor 4 and CD14. Candesartan administration significantly blocked AT(1) receptors. The ARB reduced the LPS-induced upregulation of CD14 gene expression; expression of TNF-alpha and IL-6 mRNA and protein; expression of IL-1beta and IkappaB-alpha mRNA; COX-2 mRNA and protein expression and PGE(2) concentration; inducible nitric oxide synthase (iNOS) gene and protein expression and iNOS activity; and Nox2 gene expression and 8-isoprostane levels. In addition, candesartan reduced the CD14 protein expression in saline- and LPS-treated rats. Our results suggest that AT(1) receptors are essential for the development of the full innate immune response to bacterial endotoxin. The ARB decreased the general peripheral inflammatory reaction to LPS and partially decreased the inflammatory response in the spleen. An unrestricted innate immune response to the bacterial endotoxin may have deleterious effects for the organism and may lead to development of chronic inflammatory disease. We postulate that ARBs may have therapeutic effects on inflammatory conditions.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Immunity, Innate/drug effects , Lipopolysaccharides/pharmacology , Spleen/drug effects , Tetrazoles/pharmacology , Animals , Biphenyl Compounds , Blood Pressure/drug effects , Cyclooxygenase 2/metabolism , Cytokines/blood , Dinoprostone/blood , Disease Models, Animal , Immunity, Innate/physiology , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/adverse effects , Male , Membrane Glycoproteins/metabolism , NADPH Oxidase 2 , NADPH Oxidases/metabolism , NF-kappa B/metabolism , Nitric Oxide Synthase Type II/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Receptor, Angiotensin, Type 1/drug effects , Spleen/metabolism , Toll-Like Receptor 4/metabolism
13.
Neuropharmacology ; 55(5): 704-11, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18588904

ABSTRACT

It has been proposed that striatonigral GABAergic transmission in the substantia nigra reticulata (SNr) is enhanced during Parkinson's disease and subsequent L-DOPA treatment. To evaluate this proposal we determined the effects of activating dopamine D1 receptors on depolarization induced [(3)H]-GABA release and on [(3)H]-cAMP accumulation in slices of SNr of rats with unilateral 6-OHDA lesions with and without l-DOPA treatment. Denervation increased depolarization induced D1-stimulated [(3)H]-GABA release, while repeated L-DOPA treatment further enhanced this response. Both also enhanced the effects of forskolin on [(3)H]-cAMP production and [(3)H]-GABA release, while neither modified the stimulating effects of 8-Br-cAMP on the release. These results shown that, after 6-OHDA lesions and l-DOPA treatment, cAMP signaling is enhanced. Furthermore, the results suggest that activation of sites in the signaling cascade downstream of cAMP synthesis is not required to increase release.


Subject(s)
Cyclic AMP/metabolism , Functional Laterality/physiology , Levodopa/therapeutic use , Parkinsonian Disorders , Receptors, Dopamine D1/physiology , Substantia Nigra/metabolism , gamma-Aminobutyric Acid/metabolism , 1-Methyl-3-isobutylxanthine/pharmacology , Analysis of Variance , Animals , Antiparkinson Agents/therapeutic use , Behavior, Animal/drug effects , Disease Models, Animal , Dopamine Agents/pharmacology , Male , Oxidopamine , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/drug therapy , Parkinsonian Disorders/pathology , Phosphodiesterase Inhibitors/pharmacology , Rats , Rats, Wistar , Substantia Nigra/drug effects , Time Factors , Tritium/metabolism
14.
Endocrinology ; 149(10): 5177-88, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18556352

ABSTRACT

Peripheral administration of bacterial endotoxin [lipopolysaccharide (LPS)] to rodents produces an innate immune response and hypothalamic-pituitary-adrenal axis stimulation. Renin-angiotensin-aldosterone system inhibition by angiotensin II AT1 receptor blockade has antiinflammatory effects in the vasculature. We studied whether angiotensin II receptor blockers (ARBs) prevent the LPS response. We focused on the adrenal gland, one organ responsive to LPS and expressing a local renin-angiotensin-aldosterone system. LPS (50 microg/kg, ip) produced a generalized inflammatory response with increased release of TNF-alpha and IL-6 to the circulation, enhanced adrenal aldosterone synthesis and release, and enhanced adrenal cyclooxygenase-2, IL-6, and TNF-alpha gene expression. ACTH and corticosterone release were also increased by LPS. Pretreatment with the ARB candesartan (1 mg/kg.d, sc for 3 d before the LPS administration) decreased LPS-induced cytokine release to the circulation, adrenal aldosterone synthesis and release, and cyclooxygenase-2 and IL-6 gene expression. Candesartan did not prevent the LPS-induced ACTH and corticosterone release. Our results suggest that AT1 receptors are essential for the development of the full innate immune and stress responses to bacterial endotoxin. The ARB decreased the general peripheral inflammatory response to LPS, partially decreased the inflammatory response in the adrenal gland, prevented the release of the pro-inflammatory hormone aldosterone, and protected the antiinflammatory effects of glucocorticoid release. An unrestricted innate immune response to the bacterial endotoxin may have deleterious effects for the organism and may lead to development of chronic inflammatory disease. We postulate that the ARBs may have therapeutic effects on inflammatory conditions.


Subject(s)
Adrenal Glands/immunology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Benzimidazoles/pharmacology , Inflammation/drug therapy , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Adrenal Glands/drug effects , Adrenocorticotropic Hormone/blood , Aldosterone/blood , Animals , Biomarkers/blood , Biphenyl Compounds , Blood Pressure/drug effects , Corticosterone/blood , Inflammation/chemically induced , Inflammation/immunology , Interleukin-6/blood , Lipopolysaccharide Receptors/metabolism , Lipopolysaccharides/pharmacology , Male , Rats , Rats, Wistar , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/immunology , Tumor Necrosis Factor-alpha/blood
15.
Ann N Y Acad Sci ; 1148: 360-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19120129

ABSTRACT

Angiotensin II AT(1) receptor blockers (ARBs) are commonly used in the clinical treatment of hypertension. Subcutaneous or oral administration of the ARB candesartan inhibits brain as well as peripheral AT(1) receptors, indicating transport across the blood-brain barrier. Pretreatment with candesartan profoundly modifies the response to stress. The ARB prevents the peripheral and central sympathetic activation characteristic of isolation stress and abolishes the activation of the hypothalamic-pituitary-adrenal axis during isolation. In addition, candesartan prevents the isolation-induced decrease in cortical corticotropin-releasing factor 1 and benzodiazepine receptors induced by isolation. When administered before cold-restraint stress, candesartan totally prevents the production of gastric ulcerations. This preventive effect of candesartan is the consequence of profound anti-inflammatory effects, reduction of sympathetic stimulation, and preservation of blood flow to the gastric mucosa. The ARB does not reduce the hypothalamic-pituitary-adrenal axis stimulation during cold restraint. Preservation of the effects of endogenous glucocorticoids is essential for protection of the gastric mucosa during cold restraint. Administration of the ARB to nonstressed rats decreases anxiety in the elevated plus-maze. Our results demonstrate that Angiotensin II, through AT(1) receptor stimulation, is a major stress hormone, and that ARBs, in addition to their antihypertensive effects, may be considered for the treatment of stress-related disorders.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/therapeutic use , Antihypertensive Agents/therapeutic use , Benzimidazoles/therapeutic use , Receptor, Angiotensin, Type 1/metabolism , Stress, Psychological/drug therapy , Tetrazoles/therapeutic use , Adrenocorticotropic Hormone/metabolism , Angiotensin II/metabolism , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Angiotensin II Type 1 Receptor Blockers/metabolism , Angiotensin II Type 2 Receptor Blockers , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/metabolism , Benzimidazoles/administration & dosage , Benzimidazoles/metabolism , Biphenyl Compounds , Brain/anatomy & histology , Brain/metabolism , Rats , Receptor, Angiotensin, Type 2/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, GABA-A/metabolism , Social Isolation , Stomach Ulcer/metabolism , Stomach Ulcer/prevention & control , Tetrazoles/administration & dosage , Tetrazoles/metabolism
16.
Neurochem Res ; 31(4): 555-61, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16758365

ABSTRACT

In rat striatal slices labelled with [(3)H]-adenine and in the presence of 1 mM 3-isobutyl-1-methylxantine (IBMX), cyclic [(3)H]-AMP ([(3)H]-cAMP) accumulation induced by the dopamine D(1) receptor agonist SKF-81297 (1 microM; 177 +/- 13% of basal) was inhibited by the general muscarinic agonist carbachol (maximum inhibition 72 +/- 3%, IC(50) 0.30 +/- 0.06 microM). The muscarinic toxin 7 (MT-7), a selective antagonist at muscarinic M(1) receptors, reduced the effect of SKF-81297 by 40+/-7% (IC(50) 251+/- 57 pM) and enhanced the inhibitory action of a submaximal (1 microM) concentration of carbachol (69 +/- 4% vs. 40 +/- 7% inhibition, IC(50) 386 +/- 105 pM). The toxin MT-1, agonist at M(1) receptors, stimulated [(3)H]-cAMP accumulation in a modest but significant manner (137 +/- 11% of basal at 400 nM), an action additive to that of D(1) receptor activation and blocked by MT-7 (10 nM). The effects of MT-7 on D(1) receptor-induced [(3)H]-cAMP accumulation and the carbachol inhibition were mimicked by the PKC inhibitors Ro-318220 (200 nM) and Gö-6976 (200 nM). Taken together our results indicate that in addition to the inhibitory role of M(4) receptors, in rat striatum acetylcholine stimulates cAMP formation through the activation of M(1 )receptors and PKC stimulation.


Subject(s)
Corpus Striatum/metabolism , Cyclic AMP/metabolism , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M4/metabolism , Receptors, Dopamine D1/metabolism , Alkaloids/metabolism , Animals , Benzazepines/metabolism , Carbachol/metabolism , Cholinergic Agonists/metabolism , Corpus Striatum/cytology , Dopamine Agonists/metabolism , Furans/metabolism , Male , Muscarinic Antagonists/metabolism , N-Methylscopolamine/metabolism , Naphthalenes/metabolism , Parasympatholytics/metabolism , Piperidines/metabolism , Pirenzepine/metabolism , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Radioligand Assay , Rats , Rats, Wistar
17.
Neuropsychopharmacology ; 31(6): 1123-34, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16205776

ABSTRACT

Long-term pretreatment with an angiotensin II AT1 antagonist blocks angiotensin II effects in brain and peripheral organs and abolishes the sympathoadrenal and hypothalamic-pituitary-adrenal responses to isolation stress. We determined whether AT1 receptors were also important for the stress response of higher regulatory centers. We studied angiotensin II and corticotropin-releasing factor (CRF) receptors and benzodiazepine binding sites in brains of Wistar Hannover rats. Animals were pretreated for 13 days with vehicle or a central and peripheral AT1 antagonist (candesartan, 0.5 mg/kg/day) via osmotic minipumps followed by 24 h of isolation in metabolic cages, or kept grouped throughout the study (grouped controls). In another study, we determined the influence of a similar treatment with candesartan on performance in an elevated plus-maze. AT1 receptor blockade prevented the isolation-induced increase in brain AT1 receptors and decrease in AT2 binding in the locus coeruleus. AT1 receptor antagonism also prevented the increase in tyrosine hydroxylase mRNA in the locus coeruleus. Pretreatment with the AT1 receptor antagonist completely prevented the decrease in cortical CRF1 receptor and benzodiazepine binding produced by isolation stress. In addition, pretreatment with candesartan increased the time spent in and the number of entries to open arms of the elevated plus-maze, measure of decreased anxiety. Our results implicate a modulation of upstream neurotransmission processes regulating cortical CRF1 receptors and the GABA(A) complex as molecular mechanisms responsible for the anti-anxiety effect of centrally acting AT1 receptor antagonists. We propose that AT1 receptor antagonists can be considered as compounds with possible therapeutic anti-stress and anti-anxiety properties.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/administration & dosage , Benzimidazoles/administration & dosage , Benzodiazepines/pharmacology , Cerebral Cortex/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Social Isolation/psychology , Stress, Physiological/drug therapy , Tetrazoles/administration & dosage , Amphibian Proteins , Analysis of Variance , Angiotensin II/metabolism , Animals , Autoradiography/methods , Behavior, Animal , Biphenyl Compounds , Disease Models, Animal , Flunitrazepam/pharmacokinetics , GABA Modulators/pharmacokinetics , In Situ Hybridization/methods , Male , Maze Learning/drug effects , Peptide Hormones , Peptides/pharmacology , Protein Binding/drug effects , Pyrimidines/pharmacology , Pyrroles/pharmacology , RNA, Messenger/metabolism , Rats , Receptor, Angiotensin, Type 2/drug effects , Stress, Physiological/physiopathology , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism
18.
Neurosci Lett ; 364(3): 179-84, 2004 Jul 08.
Article in English | MEDLINE | ID: mdl-15196671

ABSTRACT

In striatal membranes bearing significant levels of histamine H3 receptors (72 +/- 14 fmol/mg protein), the H3 agonist immepip (1 microM) increased [35S]GTPgammaS binding to 119 +/- 2% of basal, an effect prevented by the H3 antagonist clobenpropit and by pre-treatment with pertussis toxin. In slices labelled with [3H]adenine and in the presence of 1 mM isobutylmethylxantine (IBMX), the selective dopamine D1-like (D1/D5) receptor agonist SKF-81297 stimulated cyclic [3H]AMP ([3H]cAMP) accumulation (maximal stimulation 205 +/- 24% of basal, EC50 113 +/- 12 nM), an effect fully blocked by the D1/D5 antagonist SCH-23390. The accumulation of [3H]cAMP induced by 1 microM SKF-81297 was inhibited in a concentration-dependent manner by the selective H3 receptor agonist immepip (maximal inhibition 60+/-5%, IC50 13 +/- 5 nM). The inhibitory action of 100 nM immepip was reversed in a concentration-dependent manner by the H3 antagonist thioperamide (EC50 13 +/- 3 nM, Ki 1.4 +/- 0.3 nM). Forskolin-induced [3H]cAMP accumulation (726 +/- 57% of basal) was also reduced by H3 receptor activation, although to a lesser extent (19.1 +/- 3.2% inhibition), an action not affected by the absence of either IBMX or Ca2+ ions in the incubation medium. Neither the density of [3H]SCH-23390 binding sites (D1 receptors) nor the inhibition by SKF-81297 were affected by 1 microM immepip, ruling out a direct interaction between D1 and H3 receptors. These results indicate that through H3 receptors coupled to Galphai/o proteins, histamine modulates cAMP formation in striatal neurones that possess D1 receptors, most probably GABAergic striato-nigral neurones.


Subject(s)
Corpus Striatum/metabolism , Cyclic AMP/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Histamine H3/metabolism , Animals , Male , Neurons/metabolism , Organ Culture Techniques , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...