Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Type of study
Language
Publication year range
1.
PLoS One ; 8(8): e71326, 2013.
Article in English | MEDLINE | ID: mdl-23936499

ABSTRACT

Neuropathic pain (NP) is a debilitating condition associated with traumatic, metabolic, autoimmune and neurological etiologies. Although the triggers for NP are diverse, there are common underlying pathways, including activation of immune cells in the spinal cord and up-regulation of the N-methyl-D-aspartate receptor (NMDAR). Ketamine, a well-known NDMAR antagonist, reduces neuropathic pain in a sustained manner. Recent study has shown that the novel 11-amino acid peptide erythropoietin derivative ARA290 produces a similar, long-lasting relief of NP. Here, we show that both drugs also have similar effects on the expression of mRNA of the NMDAR, as well as that of microglia, astrocytes and chemokine (C-C motif) ligand 2, all-important contributors to the development of NP. Although the effects of ketamine and ARA 290 on NP and its molecular mediators suggest a common mechanism of action, ARA 290 has no affinity for the NMDAR and acts specifically via the innate repair receptor (IRR) involved in tissue protection. We speculated therefore, that the IRR might be critically involved in the action of ketamine on neuropathic pain. To evaluate this, we studied the effects of ketamine and ARA 290 on acute pain, side effects, and allodynia following a spared nerve injury model in mice lacking the ß-common receptor (ßcR), a structural component of the IRR. Ketamine (50 mg/kg) and ARA 290 (30 µg/kg) produced divergent effects on acute pain: ketamine produced profound antinociception accompanied with psychomotor side effects, but ARA290 did not, in both normal and knock out mice. In contrast, while both drugs were antiallodynic in WT mice, they had no effect on NP in mice lacking the ßcR. Together, these results show that an intact IRR is required for the effective treatment of NP with either ketamine or ARA 290, but is not involved in ketamine's analgesic and side effects.


Subject(s)
Cytokine Receptor Common beta Subunit/deficiency , Cytokine Receptor Common beta Subunit/genetics , Gene Knockout Techniques , Ketamine/pharmacology , Neuralgia/drug therapy , Animals , Behavior, Animal/drug effects , Female , Ketamine/therapeutic use , Mice , Mice, Inbred C57BL , Neuralgia/genetics , Neuralgia/metabolism , Nociception/drug effects , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , RNA, Messenger/genetics , RNA, Messenger/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism
2.
PLoS One ; 7(5): e37171, 2012.
Article in English | MEDLINE | ID: mdl-22629364

ABSTRACT

Iron chelators for the treatment of malaria have proven therapeutic activity in vitro and in vivo in both humans and mice, but their clinical use is limited by the unsuitable absorption and pharmacokinetic properties of the few available iron chelators. FBS0701, (S)3"-(HO)-desazadesferrithiocin-polyether [DADFT-PE], is an oral iron chelator currently in Phase 2 human studies for the treatment of transfusional iron overload. The drug has very favorable absorption and pharmacokinetic properties allowing for once-daily use to deplete circulating free iron with human plasma concentrations in the high µM range. Here we show that FBS0701 has inhibition concentration 50% (IC(50)) of 6 µM for Plasmodium falciparum in contrast to the IC(50) for deferiprone and deferoxamine at 15 and 30 µM respectively. In combination, FBS0701 interfered with artemisinin parasite inhibition and was additive with chloroquine or quinine parasite inhibition. FBS0701 killed early stage P. falciparum gametocytes. In the P. berghei Thompson suppression test, a single dose of 100 mg/kg reduced day three parasitemia and prolonged survival, but did not cure mice. Treatment with a single oral dose of 100 mg/kg one day after infection with 10 million lethal P. yoelii 17XL cured all the mice. Pretreatment of mice with a single oral dose of FBS0701 seven days or one day before resulted in the cure of some mice. Plasma exposures and other pharmacokinetics parameters in mice of the 100 mg/kg dose are similar to a 3 mg/kg dose in humans. In conclusion, FBS0701 demonstrates a single oral dose cure of the lethal P. yoelii model. Significantly, this effect persists after the chelator has cleared from plasma. FBS0701 was demonstrated to remove labile iron from erythrocytes as well as enter erythrocytes to chelate iron. FBS0701 may find clinically utility as monotherapy, a malarial prophylactic or, more likely, in combination with other antimalarials.


Subject(s)
Antimalarials/therapeutic use , Ethyl Ethers/therapeutic use , Iron Chelating Agents/therapeutic use , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Thiazoles/therapeutic use , Animals , Antimalarials/pharmacology , Disease Models, Animal , Ethyl Ethers/pharmacology , Iron Chelating Agents/pharmacology , Mice , Parasitemia/drug therapy , Thiazoles/pharmacology , Treatment Outcome
3.
J Neurotrauma ; 29(6): 1156-66, 2012 Apr 10.
Article in English | MEDLINE | ID: mdl-21545288

ABSTRACT

Pyroglutamate helix B surface peptide (pHBSP) is an 11 amino acid peptide, designed to interact with a novel cell surface receptor, composed of the classical erythropoietin (EPO) receptor disulfide linked to the beta common receptor. pHBSP has the cytoprotective effects of EPO without stimulating erythropoiesis. Effects on early cerebral hemodynamics and neurological outcome at 2 weeks post-injury were compared in a rat model of mild cortical impact injury (3m/sec, 2.5 mm deformation) followed by 50 min of hemorrhagic hypotension (MAP 40 mm Hg for 50 min). Rats were randomly assigned to receive 5000 U/kg of EPO, 30 µg/kg of pHBSP, or an inactive substance every 12 h for 3 days, starting at the end of resuscitation from the hemorrhagic hypotension, which was 110 min post-injury. Both treatments reduced contusion volume at 2 weeks post-injury, from 20.8±2.8 mm(3) in the control groups to 7.7±2.0 mm(3) in the EPO-treated group and 5.9±1.5 mm(3) in the pHBSP-treated group (p=0.001). Both agents improved recovery of cerebral blood flow in the injured brain following resuscitation, and resulted in more rapid recovery of performance on beam balancing and beam walking tests. These studies suggest that pHBSP has neuroprotective effects similar to EPO in this model of combined brain injury and hypotension. pHBSP may be more useful in the clinical situation because there is less risk of thrombotic adverse effects.


Subject(s)
Brain Injuries/drug therapy , Cerebrovascular Circulation/drug effects , Erythropoietin/pharmacology , Hemodynamics/drug effects , Neuroprotective Agents/pharmacology , Shock, Hemorrhagic/drug therapy , Animals , Brain Injuries/complications , Disease Models, Animal , Oligopeptides/pharmacology , Rats , Rats, Long-Evans , Recovery of Function/drug effects , Shock, Hemorrhagic/etiology
5.
J Investig Med ; 59(7): 1073-82, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20683348

ABSTRACT

The essential biological role of erythropoietin (EPO) in maintaining erythrocyte mass has been well understood for many years. Although EPO is required for the maturation of red cells, it also has strong procoagulant effects on the vascular endothelium and platelets, which limit erythrocyte losses after hemorrhage. Like other members of the type 1 cytokine superfamily, EPO has multiple biological activities. For the past 10 years, multiple investigators have shown that EPO acts as a locally produced antagonist of proinflammatory cytokines that are generated by the innate immune response in response to infection, trauma, or metabolic stress. Specifically, EPO inhibits apoptosis of cells surrounding a locus of injury, reduces the influx of inflammatory cells, and recruits tissue-specific stem cells and endothelial progenitor cells. Available evidence suggests that these multiple, nonerythropoietic effects of EPO are mediated by a tissue protective receptor (TPR) that is distinct from the homodimeric receptor responsible for erythropoiesis. Notably, activation of the TPR requires a higher concentration of EPO than is needed for maximal erythropoiesis. Unfortunately, these higher concentrations of EPO also stimulate hematopoietic and procoagulant pathways, which can cause adverse effects and, therefore, potentially limit the clinical use of EPO for tissue protection. To circumvent these problems, the EPO molecule has been successfully modified in a variety of ways to interact only with the TPR. Early clinical experience has shown that these compounds appear to be safe, and proof of concept trials are ready to begin.


Subject(s)
Erythropoiesis/drug effects , Erythropoietin/metabolism , Animals , Apoptosis , Coagulants/metabolism , Erythrocytes/cytology , Hemorrhage/metabolism , Humans , Hypoxia/pathology , Inflammation , Ischemia/pathology , Malaria, Cerebral/pathology , Neoplasms/pathology , Signal Transduction , Stem Cells/cytology , Thrombosis
SELECTION OF CITATIONS
SEARCH DETAIL
...