Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Enrique José Cobos is active.

Publication


Featured researches published by Enrique José Cobos.


Current Neuropharmacology | 2008

Pharmacology and Therapeutic Potential of Sigma1 Receptor Ligands

Enrique José Cobos; José Manuel Entrena; Francisco Rafael Nieto; Cruz Miguel Cendán; E Del Pozo

Sigma (σ) receptors, initially described as a subtype of opioid receptors, are now considered unique receptors. Pharmacological studies have distinguished two types of σ receptors, termed σ1 and σ2. Of these two subtypes, the σ1 receptor has been cloned in humans and rodents, and its amino acid sequence shows no homology with other mammalian proteins. Several psychoactive drugs show high to moderate affinity for σ1 receptors, including the antipsychotic haloperidol, the antidepressant drugs fluvoxamine and sertraline, and the psychostimulants cocaine and methamphetamine; in addition, the anticonvulsant drug phenytoin allosterically modulates σ1 receptors. Certain neurosteroids are known to interact with σ1 receptors, and have been proposed to be their endogenous ligands. These receptors are located in the plasma membrane and in subcellular membranes, particularly in the endoplasmic reticulum, where they play a modulatory role in intracellular Ca2+ signaling. Sigma1 receptors also play a modulatory role in the activity of some ion channels and in several neurotransmitter systems, mainly in glutamatergic neurotransmission. In accordance with their widespread modulatory role, σ1 receptor ligands have been proposed to be useful in several therapeutic fields such as amnesic and cognitive deficits, depression and anxiety, schizophrenia, analgesia, and against some effects of drugs of abuse (such as cocaine and methamphetamine). In this review we provide an overview of the present knowledge of σ1 receptors, focussing on σ1 ligand neuropharmacology and the role of σ1 receptors in behavioral animal studies, which have contributed greatly to the potential therapeutic applications of σ1 ligands.


Pain | 2009

Sigma-1 receptors are essential for capsaicin-induced mechanical hypersensitivity: Studies with selective sigma-1 ligands and sigma-1 knockout mice

José Manuel Entrena; Enrique José Cobos; Francisco Rafael Nieto; Cruz Miguel Cendán; Georgia Gris; Esperanza Del Pozo; Daniel Zamanillo; José M. Baeyens

ABSTRACT We evaluated the role of σ1 receptors on capsaicin‐induced mechanical hypersensitivity and on nociceptive pain induced by punctate mechanical stimuli, using wild‐type and σ1 receptor knockout (σ1‐KO) mice and selective σ1 receptor‐acting drugs. Mutation in σ1‐KO mice was confirmed by PCR analysis of genomic DNA and, at the protein level, by [3H](+)‐pentazocine binding assays. Both wild‐type and σ1‐KO mice not treated with capsaicin showed similar responses to different intensities of mechanical stimuli (0.05–8 g force), ranging from innocuous to noxious, applied to the hind paw. This indicates that σ1 gene inactivation does not modify the perception of punctate mechanical stimuli. The intraplantar (i.pl.) administration of capsaicin induced dose‐dependent mechanical allodynia in wild‐type mice (markedly reducing both the threshold force necessary to induce paw withdrawal and the latency to paw withdrawal induced by a given force). In contrast, capsaicin was completely unable to induce mechanical hypersensitivity in σ1‐KO mice. The high‐affinity and selective σ1 antagonists BD‐1063, BD‐1047 and NE‐100, administered subcutaneously (s.c.), dose‐dependently inhibited mechanical allodynia induced by capsaicin (1 μg,i.pl.), yielding ED50 (mg/kg) values of 15.80 ± 0.93, 29.31 ± 1.65 and 40.74 ± 7.20, respectively. The effects of the σ1 antagonists were reversed by the σ1 agonist PRE‐084 (32 mg/kg, s.c.). None of the drugs tested modified the responses induced by a painful mechanical punctate stimulus (4 g force) in nonsensitized animals. These results suggest that σ1 receptors are essential for capsaicin‐induced mechanical hypersensitivity, but are not involved in mechanical nociceptive pain.


The Journal of Pain | 2012

Role of Sigma-1 Receptors in Paclitaxel-Induced Neuropathic Pain in Mice

Francisco Rafael Nieto; Cruz Miguel Cendán; Cristina Sánchez-Fernández; Enrique José Cobos; José Manuel Entrena; Miguel Á. Tejada; Daniel Zamanillo; José Miguel Vela; José M. Baeyens

UNLABELLED Sigma-1 (σ(1)) receptors play a role in different types of pain and in central sensitization mechanisms; however, it is unknown whether they are involved in chemotherapy-induced neuropathic pain. We compared the ability of paclitaxel to induce cold (acetone test) and mechanical (electronic Von Frey test) allodynia in wild-type (WT) and σ(1) receptor knockout (σ(1)-KO) mice. We also tested the effect on paclitaxel-induced painful neuropathy of BD-1063 (16-64 mg/kg, subcutaneously) and S1RA (32-128 mg/kg, subcutaneously), 2 selective σ(1) receptor antagonists that bind to the σ(1) receptor with high affinity and competitively. The responses to cold and mechanical stimuli were similar in WT and σ(1)-KO mice not treated with paclitaxel; however, treatment with paclitaxel (2 mg/kg, intraperitoneally, once per day during 5 consecutive days) produced cold and mechanical allodynia and an increase in spinal cord diphosphorylated extracellular signal-regulated kinase (pERK) in WT but not in σ(1)-KO mice. The administration of BD-1063 or S1RA 30 minutes before each paclitaxel dose prevented the development of cold and mechanical allodynia in WT mice. Moreover, the acute administration of both σ(1) receptor antagonists dose dependently reversed both types of paclitaxel-induced allodynia after they had fully developed. These results suggest that σ(1) receptors play a key role in paclitaxel-induced painful neuropathy. PERSPECTIVE Antagonists of the σ(1) receptor may have therapeutic value for the treatment and/or prevention of paclitaxel-induced neuropathic pain. This possibility is especially interesting in the context of chemotherapy-induced neuropathy, where the onset of nerve damage is predictable and preventive treatment could be administered.


Neuropharmacology | 2013

Potentiation of morphine-induced mechanical antinociception by σ1 receptor inhibition: Role of peripheral σ1 receptors

Cristina Sánchez-Fernández; Francisco Rafael Nieto; Rafael González-Cano; Antonia Artacho-Cordón; Lucía Romero; Ángeles Montilla-García; Daniel Zamanillo; José M. Baeyens; José Manuel Entrena; Enrique José Cobos

We studied the modulation of morphine-induced mechanical antinociception and side effects by σ₁ receptor inhibition. Both wild-type (WT) and σ₁ receptor knockout (σ₁-KO) mice showed similar responses to paw pressure (100-600 g). The systemic (subcutaneous) or local (intraplantar) administration of σ₁ antagonists (BD-1063, BD-1047, NE-100 and S1RA) was devoid of antinociceptive effects in WT mice. However, σ₁-KO mice exhibited an enhanced mechanical antinociception in response to systemic morphine (1-16 mg/kg). Similarly, systemic treatment of WT mice with σ₁ antagonists markedly potentiated morphine-induced antinociception, and its effects were reversed by the selective σ₁ agonist PRE-084. Although the local administration of morphine (50-200 μg) was devoid of antinociceptive effects in WT mice, it induced dose-dependent antinociception in σ₁-KO mice. This effect was limited to the injected paw. Enhancement of peripheral morphine antinociception was replicated in WT mice locally co-administered with σ₁ antagonists and the opioid. None of the σ₁ antagonists tested enhanced morphine-antinociception in σ₁-KO mice, confirming a σ₁-mediated action. Morphine-induced side-effects (hyperlocomotion and inhibition of gastrointestinal transit) were unaltered in σ₁-KO mice. These results cannot be explained by a direct interaction of σ₁ ligands with μ-opioid receptors or adaptive changes of μ-receptors in σ₁-KO mice, given that [(3)H]DAMGO binding in forebrain, spinal cord, and hind-paw skin membranes was unaltered in mutant mice, and none of the σ₁ drugs tested bound to μ-opioid receptors. These results show that σ₁ receptor inhibition potentiates morphine-induced mechanical analgesia but not its acute side effects, and that this enhanced analgesia can be induced at peripheral level.


Journal of Pharmacology and Experimental Therapeutics | 2013

Modulation of Peripheral μ-Opioid Analgesia by σ1 Receptors

Cristina Sánchez-Fernández; Ángeles Montilla-García; Rafael González-Cano; Francisco Rafael Nieto; Lucía Romero; Antonia Artacho-Cordón; Rosa Montes; Begoña Fernández-Pastor; Manuel Merlos; José M. Baeyens; José Manuel Entrena; Enrique José Cobos

We evaluated the effects of σ1-receptor inhibition on μ-opioid–induced mechanical antinociception and constipation. σ1-Knockout mice exhibited marked mechanical antinociception in response to several μ-opioid analgesics (fentanyl, oxycodone, morphine, buprenorphine, and tramadol) at systemic (subcutaneous) doses that were inactive in wild-type mice and even unmasked the antinociceptive effects of the peripheral μ-opioid agonist loperamide. Likewise, systemic (subcutaneous) or local (intraplantar) treatment of wild-type mice with the selective σ1 antagonists BD-1063 [1-[2-(3,4-dichlorophenyl)ethyl]-4-methylpiperazine dihydrochloride] or S1RA [4-[2-[[5-methyl-1-(2-naphthalenyl)1H-pyrazol-3-yl]oxy]ethyl] morpholine hydrochloride] potentiated μ-opioid antinociception; these effects were fully reversed by the σ1 agonist PRE-084 [2-(4-morpholinethyl)1-phenylcyclohexanecarboxylate) hydrochloride], showing the selectivity of the pharmacological approach. The μ-opioid antinociception potentiated by σ1 inhibition (by σ1-receptor knockout or σ1-pharmacological antagonism) was more sensitive to the peripherally restricted opioid antagonist naloxone methiodide than opioid antinociception under normal conditions, indicating a key role for peripheral opioid receptors in the enhanced antinociception. Direct interaction between the opioid drugs and σ1 receptor cannot account for our results, since the former lacked affinity for σ1 receptors (labeled with [3H](+)-pentazocine). A peripheral role for σ1 receptors was also supported by their higher density (Western blot results) in peripheral nervous tissue (dorsal root ganglia) than in several central areas involved in opioid antinociception (dorsal spinal cord, basolateral amygdala, periaqueductal gray, and rostroventral medulla). In contrast to its effects on nociception, σ1-receptor inhibition did not alter fentanyl- or loperamide-induced constipation, a peripherally mediated nonanalgesic opioid effect. Therefore, σ1-receptor inhibition may be used as a systemic or local adjuvant to enhance peripheral μ-opioid analgesia without affecting opioid-induced constipation.


Current Neuropharmacology | 2013

Bedside-to-Bench Behavioral Outcomes in Animal Models of Pain: Beyond the Evaluation of Reflexes.

Enrique José Cobos; Enrique Portillo-Salido

Despite the myriad promising new targets and candidate analgesics recently identified in preclinical pain studies, little translation to novel pain medications has been generated. The pain phenotype in humans involves complex behavioral alterations, including changes in daily living activities and psychological disturbances. These behavioral changes are not reflected by the outcome measures traditionally used in rodents for preclinical pain testing, which are based on reflexes evoked by sensory stimuli of different types (mechanical, thermal or chemical). These measures do not evaluate the impact of the pain experience on the global behavior or disability of the animals, and therefore only consider a limited aspect of the pain phenotype. The development of relevant new outcomes indicative of pain to increase the validity of animal models of pain has been increasingly pursued over the past few years. The aim has been to translate “bedside-to-bench” outcomes from the human pain phenotype to rodents, in order to complement traditional pain outcomes by providing a closer and more realistic measure of clinical pain in rodents. This review summarizes and discusses the most important nonstandard outcomes for pain assessment in preclinical studies. The advantages and drawbacks of these techniques are considered, and their potential impact on the validation of potential analgesics is evaluated.


Psychopharmacology | 2014

Sigma-1 receptor inhibition reverses acute inflammatory hyperalgesia in mice: role of peripheral sigma-1 receptors

Miguel Á. Tejada; Ángeles Montilla-García; Cristina Sánchez-Fernández; José Manuel Entrena; Gloria Perazzoli; JoséM. Baeyens; Enrique José Cobos

RationaleSigma-1 (σ1) receptor inhibition ameliorates neuropathic pain by inhibiting central sensitization. However, it is unknown whether σ1 receptor inhibition also decreases inflammatory hyperalgesia, or whether peripheral σ1 receptors are involved in this process.ObjectiveThe purpose of this study was to determine the role of σ1 receptors in carrageenan-induced inflammatory hyperalgesia, particularly at the inflammation site.ResultsThe subcutaneous (s.c.) administration of the selective σ1 antagonists BD-1063 and S1RA to wild-type mice dose-dependently and fully reversed inflammatory mechanical (paw pressure) and thermal (radiant heat) hyperalgesia. These antihyperalgesic effects were abolished by the s.c. administration of the σ1 agonist PRE-084 and also by the intraplantar (i.pl.) administration of this compound in the inflamed paw, suggesting that blockade of peripheral σ1 receptors in the inflamed site is involved in the antihyperalgesic effects induced by σ1 antagonists. In fact, the i.pl. administration of σ1 antagonists in the inflamed paw (but not in the contralateral paw) was sufficient to completely reverse inflammatory hyperalgesia. σ1 knockout (σ1-KO) mice did not develop mechanical hyperalgesia but developed thermal hypersensitivity; however, the s.c. administration of BD-1063 or S1RA had no effect on thermal hyperalgesia in σ1-KO mice, supporting on-target mechanisms for the effects of both drugs. The antiedematous effects of σ1 inhibition do not account for the decreased hyperalgesia, since carrageenan-induced edema was unaffected by σ1 knockout or systemic σ1 pharmacological antagonism.Conclusionsσ1 receptors play a major role in inflammatory hyperalgesia. Targeting σ1 receptors in the inflamed tissue may be useful for the treatment of inflammatory pain.


Inflammation Research | 2015

Sigma-1 receptor and inflammatory pain

Georgia Gris; Enrique José Cobos; Daniel Zamanillo; Enrique Portillo-Salido

IntroductionThe sigma-1 receptor (Sig-1R) is a unique ligand-regulated molecular chaperone that interacts with several protein targets such as G protein-coupled receptors and ion channels to modulate their activity. Sig-1R is located in areas of the central and peripheral nervous system that are key to pain control. Previous preclinical studies have suggested a potential therapeutic use of Sig-1R antagonists for the management of neuropathic pain.DiscussionRecent studies using pharmacological and genetic tools have explored the role of Sig-1R in inflammatory pain conditions. Mice lacking the Sig-1R have shown different patterns of phenotypic responses to inflammatory injury. Systemic or peripheral administration of several Sig-1R antagonists, including the selective Sig-1R antagonist S1RA, inhibited both mechanical and thermal hypersensitivity in several preclinical models of inflammatory pain. These recent studies are summarized in the present commentary.ConclusionCentral and peripheral pharmacological blockade of Sig-1R could be an effective option to treat inflammatory pain.


Archive | 2017

Sigma-1 Receptor Antagonists: A New Class of Neuromodulatory Analgesics

Cristina Sánchez-Fernández; José Manuel Entrena; José M. Baeyens; Enrique José Cobos

The sigma-1 receptor is a unique ligand-operated chaperone present in key areas for pain control, in both the peripheral and central nervous system. Sigma-1 receptors interact with a variety of protein targets to modify their function. These targets include several G-protein-coupled receptors such as the μ-opioid receptor, and ion channels such as the N-methyl-D-aspartate receptor (NMDAR). Sigma-1 antagonists modify the chaperoning activity of sigma-1 receptor by increasing opioid signaling and decreasing NMDAR responses, consequently enhancing opioid antinociception and decreasing the sensory hypersensitivity that characterizes pathological pain conditions. However, the participation in pain relief of other protein partners of sigma-1 receptors in addition to opioid receptors and NMDARs cannot be ruled out. The enhanced opioid antinociception by sigma-1 antagonism is not accompanied by an increase in opioid side effects , including tolerance, dependence or constipation, so the use of sigma-1 antagonists may increase the therapeutic index of opioids. Furthermore, sigma-1 antagonists (in the absence of opioids) have been shown to exert antinociceptive effects in preclinical models of neuropathic pain induced by nerve trauma or chemical injury (the antineoplastic paclitaxel), and more recently in inflammatory and ischemic pain. Although most studies attributed the analgesic properties of sigma-1 antagonists to their central actions, it is now known that peripheral sigma-1 receptors also participate in their effects. Overwhelming preclinical evidence of the role of sigma-1 receptors in pain has led to the development of the first selective sigma-1 antagonist with an intended indication for pain treatment, which is currently in Phase II clinical trials.


European Journal of Pain | 2009

245 ROLE OF SIGMA-1 RECEPTORS IN COLD ALLODYNIA INDUCED BY PACLITAXEL

Francisco Rafael Nieto; Cruz Miguel Cendán; José Manuel Entrena; Enrique José Cobos; Rafael González-Cano; D. Zamanillo; José M. Baeyens

This substance regulates various systems including excitatory glutamatergic nervous system activation of the N-methyl-Daspartate (NMDA) receptor related to nitric oxide (NO) production. Hyperalgesia caused by peripheral tissue damage is involved in glutamate-related neuronal plasticity of the spinal cord. However, its mechanism of action still is unknown. Therefore, we determined the analgesic effect of agmatine in relation to modification of glutamate-related neuronalplasticity using a rat inflammatory pain model. Methods: SD rats implanted with an intrathecal catheter were subjected to formalin-induced hyperalgesia. Pain behavior was assessed by counting spontaneous flinches per min after formalin injection into paws under halothane anesthesia. The rats were separated into six groups as follows, (1) Non-treated (saline, it. inj.); (2) Agmatine (30ug, it. inj.); (3) MK-801 (15ug, it. inj.); (4) AVS (OH radical scavenger, 100ug, it. inj); (5) Agmatine + MK-801; (6) Agmatine + AVS. Results: Rats showed biphasic pain behavior – the sum of the first (0–5 min) is 14/min and the sum of the second (20–60 min) is 147/min. All drugs diminished the sum of the second phase 32–68% compared with non-treated animals but did not affect the first phase. An additive effect of agmatine was obtained with MK801 but not with AVS. Conclusion: We suggest that the synergistic analgesia effect of agmatine may involve the inhibition of NMDA receptor(s) related to NO production, and that agmatine acts on multiple sites of glutamatergic neuronal activation in developing hyperalgesia.

Collaboration


Dive into the Enrique José Cobos's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge