Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
2.
Eur J Pharmacol ; 740: 144-50, 2014 Oct 05.
Article in English | MEDLINE | ID: mdl-25034807

ABSTRACT

We previously reported that melanocortins induce neuroprotection in experimental acute and chronic neurodegenerative conditions, including Alzheimer׳s disease (AD) of mild severity. Here we investigated whether melanocortins afford neuroprotection and counteract cognitive decline in AD with a medium level of severity by using 24 week-old (at the start of the study) APPSwe transgenic mice (Tg2576). Saline-treated (days 1-50) control Tg2576 mice showed an impairment in spatial learning and memory, associated (at day 50, end of the study) with hippocampus at low levels of the synaptic activity-dependent gene Zif268, relevant brain changes such as cerebral cortex/hippocampus increased level of ß-amyloid (Aß) deposit, and neuronal loss, in comparison with wild-type animals. Treatment of Tg2576 mice (once daily at days 1-50) with a nanomolar dose of the melanocortin analog [Nle4,D-Phe7]α-melanocyte-stimulating hormone (NDP-α-MSH) reduced cerebral cortex/hippocampus level of Aß deposit, decreased neuronal loss, increased hippocampus Zif268 expression and improved cognitive functions, relative to saline-treated Tg2576 mice. Pharmacological blockade of melanocortin MC4 receptors with the MC4 receptor antagonist HS024 prevented all favorable effects of NDP-α-MSH. Our data indicate that MC4 receptor-stimulating melanocortins are able to counteract cognitive decline in experimental AD of medium severity through induction of neuroprotection and improvement of synaptic transmission. After further studies, these agents could gain a role as disease modifying therapeutics for AD.


Subject(s)
Alzheimer Disease/drug therapy , Cognition Disorders/drug therapy , Neuroprotective Agents/therapeutic use , alpha-MSH/analogs & derivatives , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Animals , Cognition Disorders/metabolism , Disease Models, Animal , Early Growth Response Protein 1/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Male , Memory/drug effects , Mice, Transgenic , Neocortex/drug effects , Neocortex/metabolism , Peptides, Cyclic/pharmacology , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/metabolism , Spatial Learning/drug effects , alpha-MSH/therapeutic use
3.
Neurobiol Aging ; 35(3): 537-47, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24094579

ABSTRACT

Besides specific triggering causes, Alzheimer's disease (AD) involves pathophysiological pathways that are common to acute and chronic neurodegenerative disorders. Melanocortins induce neuroprotection in experimental acute neurodegenerative conditions, and low melanocortin levels have been found in occasional studies performed in AD-type dementia patients. Here we investigated the possible neuroprotective role of melanocortins in a chronic neurodegenerative disorder, AD, by using 12-week-old (at the start of the study) triple-transgenic (3xTg-AD) mice harboring human transgenes APPSwe, PS1M146V, and tauP301L. Treatment of 3xTg-AD mice, once daily until the end of the study (30 weeks of age), with the melanocortin analog [Nle(4),D-Phe(7)]-α-melanocyte-stimulating hormone (NDP-α-MSH) reduced cerebral cortex/hippocampus phosphorylation/level of all AD-related biomarkers investigated (mediators of amyloid/tau cascade, oxidative/nitrosative stress, inflammation, apoptosis), decreased neuronal loss, induced over-expression of the synaptic activity-dependent gene Zif268, and improved cognitive functions, relative to saline-treated 3xTg-AD mice. Pharmacological blockade of melanocortin MC4 receptors prevented all neuroprotective effects of NDP-α-MSH. Our study identifies, for the first time, a class of drugs, MC4 receptor-stimulating melanocortins, that are able to counteract the progression of experimental AD by targeting pathophysiological mechanisms up- and down-stream of ß-amyloid and tau. These data could have important clinical implications.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Melanocortins/pharmacology , Melanocortins/therapeutic use , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Amyloid beta-Peptides/metabolism , Animals , Apoptosis/genetics , Disease Models, Animal , Disease Progression , Humans , Male , Mice , Mice, Transgenic , Molecular Targeted Therapy , Oxidative Stress/genetics , Receptor, Melanocortin, Type 4/physiology , tau Proteins/metabolism
4.
Neurobiol Learn Mem ; 104: 82-91, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23726868

ABSTRACT

It has been previously reported that brain hydrogen sulfide (H2S) synthesis is severely decreased in Alzheimer's disease (AD) patients, and plasma H2S levels are negatively correlated with the severity of AD. Here we extensively investigated whether treatment with a H2S donor and spa-waters rich in H2S induces neuroprotection and slows down progression of AD. Studies with sodium hydrosulfide (a H2S donor) and Tabiano's spa-water were carried out in three experimental models of AD. Short-term and long-term treatments with sodium hydrosulfide and/or Tabiano's spa-water significantly protected against impairment in learning and memory in rat models of AD induced by brain injection of ß-amyloid1-40 (Aß) or streptozotocin, and in an AD mouse model harboring human transgenes APPSwe, PS1M146V and tauP301L (3xTg-AD mice). The improvement in behavioral performance was associated with hippocampus was size of Aß plaques and preservation of the morphological picture, as found in AD rats. Further, lowered concentration/phosphorylation levels of proteins thought to be the central events in AD pathophysiology, namely amyloid precursor protein, presenilin-1, Aß1-42 and tau phosphorylated at Thr181, Ser396 and Ser202, were detected in 3xTg-AD mice treated with spa-water. The excitotoxicity-triggered oxidative and nitrosative stress was counteracted in 3xTg-AD mice, as indicated by the decreased levels of malondialdehyde and nitrites in the cerebral cortex. Hippocampus reduced activity of c-jun N-terminal kinases, extracellular signal-regulated kinases and p38, which have an established role not only in phosphorylation of tau protein but also in inflammation and apoptosis, was also found. Consistently, decrease in tumor necrosis factor-α level, up-regulation of Bcl-2, and down-regulation of BAX and the downstream executioner caspase-3, also occurred in the hippocampus of 3xTg-AD mice after treatment with Tabiano's spa-water, thus suggesting that it is also able to modulate inflammation and apoptosis. Our findings indicate that appropriate treatments with H2S donors and Tabiano's spa-waters, and may be other spa-waters rich in H2S content, might represent an innovative approach to slow down AD progression in humans by targeting multiple pathophysiological mechanisms.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Disease Progression , Hydrogen Sulfide/therapeutic use , Alzheimer Disease/etiology , Amyloid beta-Peptides/drug effects , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/pharmacology , Animals , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/pathology , Male , Memory/drug effects , Mice , Mice, Transgenic , Neuroprotective Agents/therapeutic use , Rats , Rats, Wistar
5.
Pharmacol Res ; 72: 1-8, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23535516

ABSTRACT

The janus kinases (JAK), extracellular signal-regulated kinases (ERK) and signal transducers and activators of transcription (STAT) pathways have been shown to play a cardioprotective role. We previously gave evidence that melanocortins afford cardioprotection in conditions of myocardial ischemia/reperfusion. Here we aimed to investigate the influence of melanocortins on the JAK/ERK/STAT signaling in cardiac and systemic responses to prolonged myocardial ischemia/reperfusion. Ischemia was produced in rats by ligature of the left anterior descending coronary artery for 30 min. At the end of the 2-h reperfusion, western blot analysis of the cardioprotective transcription factors pJAK2, pERK1/2, pTyr-STAT3 and pSer-STAT3, the inflammatory mediator tumor necrosis factor-α (TNF-α), the pro-apoptotic factors BAX and c-jun N-terminal kinases (pJNK), the anti-apoptotic protein Bcl-XL, as well as of the cardioprotective enzyme heme oxygenase-1 (HO-1), was performed in the left ventricle and spleen. Intravenous treatment, during coronary artery occlusion, with the melanocortin analogs [Nle(4), D-Phe(7)]α-melanocyte-stimulating hormone (NDP-α-MSH) and adrenocorticotropic hormone 1-24 [ACTH-(1-24)], induced a left ventricle up-regulation of pJAK2, pERK1/2 and pTyr-STAT3 (JAK-dependent), and a reduction in pJNK and TNF-α levels; these effects of NDP-α-MSH and ACTH-(1-24) were associated with over-expression of the pro-survival proteins HO-1 and Bcl-XL, and marked decrease of the myocardial infarct size. Melanocortin treatment did not affect left ventricle pSer-STAT3 (ERK1/2-dependent) and BAX levels. In the spleen, NDP-α-MSH and ACTH-(1-24) induced similar effects on the expression of the above transcription factors/proteins, except for pERK1/2 (down-regulated) and HO-1 (unaffected). Blockade of JAK and ERK pathways with AG490 and U0126, respectively, abrogated the myocardial infarct size reduction by NDP-α-MSH. These results indicate that melanocortins inhibit local and systemic inflammatory and apoptotic cascades triggered by prolonged myocardial ischemia/reperfusion, with consequent reduction in myocardium infarct size, seemingly via activation of the JAK/STAT signaling and with modulation of an ERK (STAT unrelated) signaling pathway.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Janus Kinases/metabolism , Melanocortins/therapeutic use , Myocardial Ischemia/drug therapy , Myocardial Reperfusion , STAT Transcription Factors/metabolism , Animals , Apoptosis/drug effects , Heart/drug effects , Heart/physiopathology , Male , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Myocardial Reperfusion/methods , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Wistar , Signal Transduction/drug effects
6.
Eur J Pharmacol ; 707(1-3): 78-86, 2013 May 05.
Article in English | MEDLINE | ID: mdl-23535605

ABSTRACT

In experimental cerebral ischemia, melanocortin MC4 receptor agonists induce neuroprotection and neurogenesis with subsequent long-lasting functional recovery. Here we investigated the molecular mechanisms underlying melanocortin-induced neurogenesis. Gerbils were subjected to transient global cerebral ischemia, then they were treated every 12 h, and until sacrifice, with 5-bromo-2'-deoxyuridine (BrdU; to label proliferating cells), and the melanocortin analog [Nle(4),d-Phe(7)]α-melanocyte-stimulating hormone (NDP-α-MSH) or saline. NDP-α-MSH increased hippocampus dentate gyrus (DG) expression of Wnt-3A, ß-catenin, Sonic hedgehog (Shh), Zif268, interleukin-10 (IL-10) and doublecortin (DCX), as detected at days 3, 6 and 10 after the ischemic insult. Further, an elevated number of BrdU immunoreactive cells was found at days 3 and 10, and an improved histological picture with reduced neuronal loss at day 10, associated with learning and memory recovery. Pharmacological blockade of the Wnt-3A/ß-catenin and Shh pathways, as well as of melanocortin MC4 receptors, prevented all effects of NDP-α-MSH. These data indicate that, in experimental brain ischemia, treatment with melanocortins acting at MC4 receptors induces neural stem/progenitor cell proliferation in the DG by promptly and effectively triggering the canonical Wnt-3A/ß-catenin and Shh signaling pathways. Activation of these pathways is associated with up-regulation of the repair factor Zif268 and the neurogenesis facilitating factor IL-10, and it seems to address mainly toward a neuronal fate, as indicated by the increase in DCX positive cells.


Subject(s)
Brain Ischemia/drug therapy , Receptor, Melanocortin, Type 4/agonists , Wnt Signaling Pathway/drug effects , alpha-MSH/analogs & derivatives , Animals , Brain Ischemia/pathology , Cell Proliferation/drug effects , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Disease Models, Animal , Doublecortin Domain Proteins , Gerbillinae , Hedgehog Proteins/metabolism , Male , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurogenesis/drug effects , Neurons/drug effects , Neurons/metabolism , Neuropeptides/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation/drug effects , Wnt3A Protein/metabolism , alpha-MSH/pharmacology
7.
Front Neuroendocrinol ; 33(2): 179-93, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22531139

ABSTRACT

Melanocortin peptides with the adrenocorticotropin/melanocyte-stimulating hormone (ACTH/MSH) sequences and synthetic analogs have protective and life-saving effects in experimental conditions of circulatory shock, myocardial ischemia, ischemic stroke, traumatic brain injury, respiratory arrest, renal ischemia, intestinal ischemia and testicular ischemia, as well as in experimental heart transplantation. Moreover, melanocortins improve functional recovery and stimulate neurogenesis in experimental models of cerebral ischemia. These beneficial effects of ACTH/MSH-like peptides are mostly mediated by brain melanocortin MC(3)/MC(4) receptors, whose activation triggers protective pathways that counteract the main ischemia/reperfusion-related mechanisms of damage. Induction of signaling pathways and other molecular regulators of neural stem/progenitor cell proliferation, differentiation and integration seems to be the key mechanism of neurogenesis stimulation. Synthesis of stable and highly selective agonists at MC(3) and MC(4) receptors could provide the potential for development of a new class of drugs for a novel approach to management of severe ischemic diseases.


Subject(s)
Hypoxia/drug therapy , Melanocortins/therapeutic use , Animals , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Humans , Hypoxia/pathology , Melanocortins/metabolism , Melanocortins/pharmacology , Models, Biological , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism , Receptors, Melanocortin/physiology , Severity of Illness Index , Shock/drug therapy , Shock/prevention & control
8.
Anesthesiology ; 116(3): 692-700, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22266570

ABSTRACT

BACKGROUND: Melanocortin peptides improve hemodynamic parameters and prevent death during severe hemorrhagic shock. In the present research we determined influences of a synthetic melanocortin 1/4 receptor agonist on the molecular changes that occur in rat liver during hemorrhage. METHODS: Controlled-volume hemorrhage was performed in adult rats under general anesthesia by a stepwise blood withdrawal until mean arterial pressure fell to 40 mmHg. Then rats received either saline or the synthetic melanocortin 1/4 receptor agonist Butir-His-D-Phe-Arg-Trp-Sar-NH2 (Ro27-3225; n = 6-8 per group). Hemogasanalysis was performed throughout a 60-min period. Gene expression in liver samples was determined at 1 or 3 h using quantitative real-time polymerase chain reaction. RESULTS: At 1 h, in saline-treated shocked rats, there were significant increases in activating transcription factor 3 (Atf3), early growth response 1 (Egr1), heme oxygenase (decycling) 1 (Hmox1), FBJ murine osteosarcoma viral oncogene homolog (Fos), and jun oncogene (Jun). These changes were prevented by Ro27-3225 (mean ± SEM: Atf3 152.83 ± 58.62 vs. 579.00 ± 124.13, P = 0.002; Egr1 13.21 ± 1.28 vs. 26.63 ± 1.02, P = 0.001; Hmox1 3.28 ± 0.31 vs. 166.54 ± 35.03, P = 0.002; Fos 4.36 ± 1.03 vs. 14.90 ± 3.44, P < 0.001; Jun 6.62 ± 1.93 vs. 15.07 ± 2.09, P = 0.005; respectively). Increases in alpha-2-macroglobulin (A2m), heat shock 70kD protein 1A (Hspa1a), erythropoietin (Epo), and interleukin-6 (Il6) occurred at 3 h in shocked rats and were prevented by Ro27-3225 treatment (A2m 6.90 ± 0.82 vs. 36.73 ± 4.00, P < 0.001; Hspa1a 10.34 ± 3.28 vs. 25.72 ± 3.64, P = 0.001; Epo 0.49 ± 0.13 vs. 2.37 ± 0.73, P = 0.002; Il6 1.05 ± 0.15 vs. 1.88 ± 0.23, P < 0.001; respectively). Further, at 3 h in shocked rats treated with Ro27-3225 there were significant increases in tight junction protein 1 (Tjp1; 27.30 ± 2.43 vs. 5.03 ± 1.68, P < 0.001) and nuclear receptor subfamily 4, group A, member 1 (Nr4a1; 91.03 ± 16.20 vs. 30.43 ± 11.0, P = 0.01) relative to sham animals. Treatment with Ro27-3225 rapidly restored blood pressure, hemogasanalysis parameters, and lactate blood levels. CONCLUSIONS: Melanocortin treatment significantly prevents most of the systemic and hepatic detrimental changes induced by hemorrhage.


Subject(s)
Melanocortins/therapeutic use , Peptides/therapeutic use , Shock, Hemorrhagic/drug therapy , Shock, Hemorrhagic/metabolism , Animals , Melanocortins/metabolism , Peptides/metabolism , Rats , Rats, Wistar , Receptor, Melanocortin, Type 1/agonists , Receptor, Melanocortin, Type 1/physiology , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/physiology , Shock, Hemorrhagic/genetics , Treatment Outcome
9.
Acta Neuropathol ; 122(4): 443-53, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21927944

ABSTRACT

Melanocortins produce neuroprotection against ischemic stroke with subsequent long-lasting functional recovery, through melanocortin MC(4) receptor activation. Here we investigated whether the long-lasting beneficial effect of melanocortins in stroke conditions is associated with a stimulation of neurogenesis. Gerbils were subjected to transient global cerebral ischemia by occluding both common carotid arteries for 10 min; then, they were prepared for 5-bromo-2'-deoxyuridine (BrdU) labeling of proliferating cells. Delayed treatment (up to 9 h after the ischemic injury) for 11 days with the melanocortin analog [Nle(4),D-Phe(7)]α-melanocyte-stimulating hormone (NDP-α-MSH) improved learning and memory throughout the 50-day observation period. Immunohistochemical examination of the hippocampus on day 50 showed, in the dentate gyrus, an elevated number of BrdU immunoreactive cells colocalized with NeuN (used as indicator of mature neurons) and Zif268 (used as indicator of functionally integrated neurons). Retrospective analysis during the early stage of neural stem/progenitor cell development (days 3 and 4 after stroke) showed, in NDP-α-MSH-treated gerbils, a high degree of daily cell proliferation and revealed that NDP-α-MSH favorably affects Wnt-3A signaling pathways and doublecortin expression. Pharmacologic blockade of MC(4) receptors prevented all effects of NDP-α-MSH. These data indicate that treatment of cerebral ischemia with MC(4) receptor agonists induces, with a broad window of therapeutic opportunity, long-lasting functional recovery associated with a large number of mature and likely functional newborn neurons in brain injured areas. Our findings reveal previously undescribed effects of melanocortins which might have major clinical implications.


Subject(s)
Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Melanocortins/physiology , Nerve Degeneration/drug therapy , Nerve Degeneration/metabolism , Neurogenesis/physiology , Receptor, Melanocortin, Type 4/metabolism , Recovery of Function/physiology , Animals , Brain Ischemia/pathology , Disease Models, Animal , Gerbillinae , Male , Melanocortins/pharmacology , Nerve Degeneration/pathology , Receptor, Melanocortin, Type 4/physiology , Recovery of Function/drug effects , Time Factors
10.
Eur J Pharmacol ; 670(2-3): 479-86, 2011 Nov 30.
Article in English | MEDLINE | ID: mdl-21946115

ABSTRACT

Indirect evidence indicates that, in cerebral ischemia, melanocortins have neuroprotective effects likely mediated by MC4 receptors. To gain direct insight into the role of melanocortin MC4 receptors in ischemic stroke, we investigated the effects of a highly selective MC4 receptor agonist. Gerbils were subjected to transient global cerebral ischemia by occluding both common carotid arteries for 10 min. In saline-treated stroke animals, an impairment in learning and memory occurred that, at day 11 after stroke, was associated with hippocampus up-regulation of tumor necrosis factor-α (TNF-α), BAX, activated extracellular signal-regulated kinases (ERK1/2), c-jun N-terminal kinases (JNK1/2) and caspase-3, down-regulation of Bcl-2, and neuronal loss. Treatment for 11days with the selective melanocortin MC4 receptor agonist RO27-3225, as well as with the well known non-selective [Nle4,D-Phe7]α-melanocyte-stimulating hormone (NDP-α-MSH) as a reference non-selective melanocortin, counteracted the inflammatory and apoptotic responses, as indicated by the changes in TNF-α, BAX, ERK1/2, JNK1/2, caspase-3 and Bcl-2 protein expression. Furthermore, melanocortin treatment reduced neuronal loss and dose-dependently improved learning and memory. These positive effects were associated with overexpression of Zif268, an immediate early gene involved in injury repair, synaptic plasticity and memory formation. Pharmacological blockade of MC4 receptors with the selective MC4 receptor antagonist HS024 prevented all effects of RO27-3225 and NDP-α-MSH. These data give direct evidence that stimulation of MC4 receptors affords neuroprotection and promotes functional recovery from stroke, by counteracting prolonged and/or recurrent inflammatory and apoptotic responses, and likely by triggering brain repair pathways.


Subject(s)
Apoptosis/drug effects , Brain Ischemia/drug therapy , Brain Ischemia/pathology , Neurons/drug effects , Neurons/pathology , Peptides/pharmacology , Receptor, Melanocortin, Type 4/agonists , Amino Acid Sequence , Animals , Brain Ischemia/complications , Brain Ischemia/physiopathology , Cell Count , Gerbillinae , Inflammation/complications , Inflammation/drug therapy , Inflammation/pathology , Inflammation/physiopathology , Male , Memory/drug effects , Peptides/chemistry , Peptides/therapeutic use , Stroke/complications , Stroke/drug therapy , Stroke/pathology , Stroke/physiopathology
11.
Br J Pharmacol ; 162(4): 917-28, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21039420

ABSTRACT

BACKGROUND AND PURPOSE: Melanocortins reverse circulatory shock and improve survival by counteracting the systemic inflammatory response, and through the activation of the vagus nerve-mediated cholinergic anti-inflammatory pathway. To gain insight into the potential therapeutic value of melanocortins against multiple organ damage following systemic inflammatory response, here we investigated the effects of the melanocortin analogue [Nle4 D-Phe7]α-MSH (NDP-α-MSH) in a widely used murine model of multiple organ dysfunction syndrome (MODS). EXPERIMENTAL APPROACH: MODS was induced in mice by a single intraperitoneal injection of lipopolysaccharide followed, 6 days later (= day 0), by zymosan. After MODS or sham MODS induction, animals were randomized to receive intraperitoneally NDP-α-MSH (340 µg·kg⁻¹ day) or saline for up to 16 days. Additional groups of MODS mice were concomitantly treated with the melanocortin MC4 receptor antagonist HS024, or the nicotinic acetylcholine receptor antagonist chlorisondamine, and NDP-α-MSH. KEY RESULTS: At day 7, in the liver and lung NDP-α-MSH, significantly reduced mRNA expression of tumour necrosis factor-α (TNF-α), increased mRNA expression of interleukin-10 and improved the histological picture, as well as reduced TNF-α plasma levels; furthermore, NDP-α-MSH dose-dependently increased survival rate, as assessed throughout the 16 day observation period. HS024 and chlorisondamine prevented all the beneficial effects of NDP-α-MSH in MODS mice. CONCLUSIONS AND IMPLICATIONS: These data indicate that NDP-α-MSH protects against experimental MODS by counteracting the systemic inflammatory response, probably through brain MC4 receptor-triggered activation of the cholinergic anti-inflammatory pathway. These findings reveal previously undescribed effects of melanocortins and could have clinical relevance in the MODS setting.


Subject(s)
Inflammation Mediators/metabolism , Melanocortins/metabolism , Melanocyte-Stimulating Hormones/therapeutic use , Multiple Organ Failure/drug therapy , Multiple Organ Failure/metabolism , Pentetic Acid/analogs & derivatives , Protective Agents/therapeutic use , Animals , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Inflammation Mediators/blood , Interleukin-10/genetics , Interleukin-10/metabolism , Liver/drug effects , Liver/metabolism , Liver/pathology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Melanocyte-Stimulating Hormones/administration & dosage , Melanocyte-Stimulating Hormones/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Multiple Organ Failure/blood , Multiple Organ Failure/pathology , Nicotinic Antagonists/pharmacology , Pentetic Acid/administration & dosage , Pentetic Acid/antagonists & inhibitors , Pentetic Acid/therapeutic use , Protective Agents/administration & dosage , RNA, Messenger/metabolism , Random Allocation , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Survival Analysis , Tumor Necrosis Factor-alpha/blood , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
12.
Eur J Pharmacol ; 637(1-3): 124-30, 2010 Jul 10.
Article in English | MEDLINE | ID: mdl-20385118

ABSTRACT

Recently we reported that an efferent vagal fibre-mediated cholinergic protective pathway, activated by melanocortins acting at brain melanocortin MC(3) receptors, is operative in a condition of short-term myocardial ischemia/reperfusion associated with a high incidence of severe arrhythmias and death. Here we investigated melanocortin effects, and the role of the vagus nerve-mediated cholinergic protective pathway, in a rat model of prolonged myocardial ischemia/reperfusion associated with marked inflammatory and apoptotic reactions, and a large infarct size. Ischemia was produced in rats by ligature of the left anterior descending coronary artery for 30 min. At the end of the 2-h reperfusion, western blot analysis of the inflammatory and apoptotic markers tumor necrosis factor-alpha (TNF-alpha), c-jun N-terminal kinases (JNK) and caspase-3, as well as of the anti-apoptotic extracellular signal-regulated kinases (ERK 1/2), was performed in the left ventricle. In saline-treated ischemic rats there was an increase in TNF-alpha levels and in the activity of JNK and caspase-3 accompanied, despite an appreciable ERK 1/2 activation, by a large infarct size. Intravenous treatment, during coronary artery occlusion, with the melanocortin analog [Nle(4), D-Phe(7)]alpha-melanocyte-stimulating hormone (NDP-alpha-MSH) produced a reduction in TNF-alpha levels and in the activity of JNK and caspase-3, associated with marked activation of the pro-survival kinases ERK 1/2, and consequent attenuation of infarct size. Bilateral cervical vagotomy blunted the protective effects of NDP-alpha-MSH. These results indicate that melanocortins modulate the inflammatory and apoptotic cascades triggered by prolonged myocardial ischemia/reperfusion, and reduce infarct size, seemingly by activation of the vagus nerve-mediated cholinergic protective pathway.


Subject(s)
Apoptosis/drug effects , Inflammation/drug therapy , Inflammation/pathology , Melanocortins/pharmacology , Melanocortins/therapeutic use , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Vagus Nerve/drug effects , Animals , Blotting, Western , Caspase 3/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Myocardial Reperfusion Injury/enzymology , Myocardial Reperfusion Injury/metabolism , Rats , Rats, Wistar , Time Factors , Tumor Necrosis Factor-alpha/metabolism , Vagus Nerve/physiology
13.
Brain Behav Immun ; 23(6): 844-50, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19345727

ABSTRACT

Melanocortin peptides afford strong neuroprotection and improve functional recovery in experimental ischemic stroke; they also have established neurotrophic actions. The expression of the immediate early gene Zif268 is dependent on synaptic activity and is involved in injury repair and memory formation. Here, we investigated the role of Zif268 in learning and memory recovery after delayed treatment of ischemic stroke with the melanocortin analog [Nle(4), D-Phe(7)]alpha-MSH (NDP-alpha-MSH). A 10-min period of global cerebral ischemia was induced by occluding both common carotid arteries in gerbils. Treatment with a nanomolar dose of NDP-alpha-MSH (every 12h for 11 days) was performed starting 3h or 9h after stroke induction; where indicated, gerbils were pretreated with the melanocortin MC(4) receptor antagonist HS024. Animals were subjected to the Morris water-maze test (four sessions from 4 to 50 days after the ischemic episode). Fifty days after stroke, histological damage and Zif268 expression were investigated in the hippocampus. Treatment with NDP-alpha-MSH significantly reduced hippocampal damage, including neuronal death, and improved learning and memory recovery. This protective effect was long-lasting (50 days, at least) and associated with Zif268 overexpression, with both schedules of NDP-alpha-MSH treatment. Pharmacological blockade of MC(4) receptors prevented these effects. Our data indicate that MC(4) receptor-mediated actions of melanocortins could trigger repair mechanisms able to improve neuronal functionality and synaptic plasticity, and to promote long-lasting functional recovery from ischemic stroke with Zif268 gene involvement.


Subject(s)
Early Growth Response Protein 1/biosynthesis , Ischemic Attack, Transient/complications , Melanocortins/therapeutic use , Stroke/drug therapy , Stroke/etiology , Animals , Blotting, Western , Early Growth Response Protein 1/genetics , Gerbillinae , Learning/physiology , Male , Maze Learning/drug effects , Memory/physiology , Recovery of Function , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology
14.
J Cereb Blood Flow Metab ; 29(3): 512-23, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19018269

ABSTRACT

A vagus nerve-mediated, efferent cholinergic protective pathway activated by melanocortins is operative in circulatory shock and myocardial ischemia. Moreover, melanocortins have neuroprotective effects against brain damage after ischemic stroke. Here we investigated cerebral and systemic pathophysiologic reactions to focal cerebral ischemia in rats induced by intrastriatal microinjection of endothelin-1, and the possible protective role of the melanocortin-activated vagal cholinergic pathway. In the striatum and liver of saline-treated control rats, the activation of extracellular signal-regulated kinases, c-jun N-terminal kinases, and caspase-3, the increase in tumor necrosis factor-alpha (TNF-alpha) concentration and DNA fragmentation, as well as the increase in TNF-alpha plasma levels, occurred 10 and 20 h after the ischemic insult suggesting an activation of inflammatory and apoptotic responses. Treatment with [Nle(4), D-Phe(7)]alpha-melanocyte-stimulating hormone (NDP-alpha-MSH; 3 or 9 h after stroke) suppressed the inflammatory and apoptotic cascades at central and peripheral level. Bilateral vagotomy and pharmacologic blockade of peripheral nicotinic acetylcholine receptors blunted the protective effect of NDP-alpha-MSH. The present results show that focal brain ischemia in rats causes significant effects not only in the brain, but also in the liver. Moreover, our data support the hypothesis that a protective, melanocortin-activated, vagal cholinergic pathway is likely operative in conditions of ischemic stroke.


Subject(s)
Brain Ischemia/complications , Corpus Striatum/drug effects , Liver/drug effects , Stroke/prevention & control , Vagus Nerve/physiology , alpha-MSH/analogs & derivatives , Animals , Apoptosis/drug effects , Blotting, Western , Corpus Striatum/enzymology , Corpus Striatum/metabolism , Corpus Striatum/pathology , DNA Fragmentation/drug effects , Endothelin-1 , Liver/enzymology , Liver/metabolism , Liver/pathology , Male , Rats , Rats, Wistar , Receptors, Nicotinic/metabolism , Stroke/etiology , Stroke/metabolism , Stroke/pathology , Tumor Necrosis Factor-alpha/blood , Vagotomy , alpha-MSH/pharmacology , alpha-MSH/therapeutic use
15.
Eur J Pharmacol ; 570(1-3): 57-65, 2007 Sep 10.
Article in English | MEDLINE | ID: mdl-17588564

ABSTRACT

In gerbils subjected to transient global cerebral ischemia, melanocortin peptides produce long-lasting protection with a broad time window, and through the activation of central nervous system melanocortin MC(4) receptors. Here we aimed to investigate whether melanocortins are neuroprotective also in a rat model of focal cerebral ischemia induced by intrastriatal microinjection of endothelin-1. The vasoconstrictor agent endothelin-1 caused a significant impairment in spatial learning and memory, as well as in sensory-motor orientation and limb use, associated with severe striatal morphological damage including intense neuronal death and an almost complete myelin degradation. Treatment of ischemic rats with a nanomolar dose (340 microg/kg/day i.p. for 11 days, beginning 3 h or 9 h after endothelin-1 microinjection) of the melanocortin analog [Nle(4), D-Phe(7)]alpha-melanocyte-stimulating hormone (NDP-alpha-MSH) significantly reduced striatal damage, and improved subsequent functional recovery, with all scheduled NDP-alpha-MSH treatments. Pharmacological blockade of melanocortin MC(4) receptors prevented the protective effect of NDP-alpha-MSH. Our findings give evidence that melanocortins are neuroprotective, with a broad time window, also in a severe model of focal cerebral ischemia, and suggest that melanocortin MC(4) receptor agonists could produce neuroprotection in different experimental models of ischemic stroke.


Subject(s)
Brain Ischemia/drug therapy , Neuroprotective Agents/therapeutic use , alpha-MSH/analogs & derivatives , Animals , Brain/drug effects , Brain/pathology , Brain/physiopathology , Brain Ischemia/physiopathology , Male , Memory/drug effects , Psychomotor Performance/drug effects , Rats , Rats, Wistar , alpha-MSH/therapeutic use
16.
Eur J Pharmacol ; 538(1-3): 48-56, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16647700

ABSTRACT

Melanocortin peptides have been shown to produce neuroprotection in experimental ischemic stroke. The aim of the present investigation was to identify the therapeutic treatment window of melanocortins, and to determine whether these neuropeptides chronically protect against damage consequent to brain ischemia. A 10-min period of global cerebral ischemia in gerbils, induced by occluding both common carotid arteries, caused impairment in spatial learning and memory (Morris test: four sessions from 4 to 67 days after the ischemic episode), associated with neuronal death in the hippocampus. Treatment with a nanomolar dose (340 microg/kg i.p., every 12 h for 11 days) of the melanocortin analog [Nle(4), D-Phe(7)]alpha-melanocyte-stimulating hormone (NDP-alpha-MSH), starting 3-18 h after the ischemic episode, reduced hippocampal damage with improvement in subsequent functional recovery. The protective effect was long-lasting (67 days, at least) with all schedules of NDP-alpha-MSH treatment; however, in the latest treated (18 h) gerbils, some spatial memory deficits were detected. Pharmacological blockade of melanocortin MC(4) receptors prevented the protective effects of NDP-alpha-MSH. Our findings indicate that, in conditions of brain ischemia, melanocortins can provide strong and long-lasting protection with a broad therapeutic treatment window, and with involvement of melanocortin MC(4) receptors, 18 h being the approximately time-limit for stroke late treatment to be effective.


Subject(s)
Brain Ischemia/prevention & control , alpha-MSH/analogs & derivatives , Animals , Behavior, Animal/drug effects , Brain Ischemia/physiopathology , Drug Administration Schedule , Gerbillinae , Glial Fibrillary Acidic Protein/analysis , Hippocampus/chemistry , Hippocampus/drug effects , Hippocampus/pathology , Immunohistochemistry , Male , Maze Learning/drug effects , Memory/drug effects , Peptides, Cyclic/pharmacology , Proto-Oncogene Proteins c-bcl-2/analysis , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/physiology , Time Factors , alpha-MSH/administration & dosage , alpha-MSH/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...