Network


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

Hotspot


Dive into the research topics where M. Ruth Pazos is active.

Publication


Featured researches published by M. Ruth Pazos.


Brain | 2009

Microglial CB2 cannabinoid receptors are neuroprotective in Huntington's disease excitotoxicity

Javier Palazuelos; Tania Aguado; M. Ruth Pazos; Boris Julien; Carolina Carrasco; Eva Resel; Onintza Sagredo; Cristina Benito; Julián Romero; Iñigo Azcoitia; Javier Fernández-Ruiz; Manuel Guzmán; Ismael Galve-Roperh

Cannabinoid-derived drugs are promising agents for the development of novel neuroprotective strategies. Activation of neuronal CB(1) cannabinoid receptors attenuates excitotoxic glutamatergic neurotransmission, triggers prosurvival signalling pathways and palliates motor symptoms in animal models of neurodegenerative disorders. However, in Huntingtons disease there is a very early downregulation of CB(1) receptors in striatal neurons that, together with the undesirable psychoactive effects triggered by CB(1) receptor activation, foster the search for alternative pharmacological treatments. Here, we show that CB(2) cannabinoid receptor expression increases in striatal microglia of Huntingtons disease transgenic mouse models and patients. Genetic ablation of CB(2) receptors in R6/2 mice, that express human mutant huntingtin exon 1, enhanced microglial activation, aggravated disease symptomatology and reduced mice lifespan. Likewise, induction of striatal excitotoxicity in CB(2) receptor-deficient mice by quinolinic acid administration exacerbated brain oedema, microglial activation, proinflammatory-mediator state and medium-sized spiny neuron degeneration. Moreover, administration of CB(2) receptor-selective agonists to wild-type mice subjected to excitotoxicity reduced neuroinflammation, brain oedema, striatal neuronal loss and motor symptoms. Studies on ganciclovir-induced depletion of astroglial proliferation in transgenic mice expressing thymidine kinase under the control of the glial fibrillary acidic protein promoter excluded the participation of proliferating astroglia in CB(2) receptor-mediated actions. These findings support a pivotal role for CB(2) receptors in attenuating microglial activation and preventing neurodegeneration that may pave the way to new therapeutic strategies for neuroprotection in Huntingtons disease as well as in other neurodegenerative disorders with a significant excitotoxic component.


Brain | 2011

Loss of striatal type 1 cannabinoid receptors is a key pathogenic factor in Huntington’s disease

Cristina Blázquez; Anna Chiarlone; Onintza Sagredo; Tania Aguado; M. Ruth Pazos; Eva Resel; Javier Palazuelos; Boris Julien; María Salazar; Christine Börner; Cristina Benito; Carolina Carrasco; María Diez-Zaera; Paola Paoletti; Miguel Díaz-Hernández; Carolina Ruiz; Michael Sendtner; José J. Lucas; Justo García de Yébenes; Giovanni Marsicano; Krisztina Monory; Beat Lutz; Julián Romero; Jordi Alberch; Silvia Ginés; Jürgen Kraus; Javier Fernández-Ruiz; Ismael Galve-Roperh; Manuel Guzmán

Endocannabinoids act as neuromodulatory and neuroprotective cues by engaging type 1 cannabinoid receptors. These receptors are highly abundant in the basal ganglia and play a pivotal role in the control of motor behaviour. An early downregulation of type 1 cannabinoid receptors has been documented in the basal ganglia of patients with Huntingtons disease and animal models. However, the pathophysiological impact of this loss of receptors in Huntingtons disease is as yet unknown. Here, we generated a double-mutant mouse model that expresses human mutant huntingtin exon 1 in a type 1 cannabinoid receptor-null background, and found that receptor deletion aggravates the symptoms, neuropathology and molecular pathology of the disease. Moreover, pharmacological administration of the cannabinoid Δ(9)-tetrahydrocannabinol to mice expressing human mutant huntingtin exon 1 exerted a therapeutic effect and ameliorated those parameters. Experiments conducted in striatal cells show that the mutant huntingtin-dependent downregulation of the receptors involves the control of the type 1 cannabinoid receptor gene promoter by repressor element 1 silencing transcription factor and sensitizes cells to excitotoxic damage. We also provide in vitro and in vivo evidence that supports type 1 cannabinoid receptor control of striatal brain-derived neurotrophic factor expression and the decrease in brain-derived neurotrophic factor levels concomitant with type 1 cannabinoid receptor loss, which may contribute significantly to striatal damage in Huntingtons disease. Altogether, these results support the notion that downregulation of type 1 cannabinoid receptors is a key pathogenic event in Huntingtons disease, and suggest that activation of these receptors in patients with Huntingtons disease may attenuate disease progression.


British Journal of Clinical Pharmacology | 2013

Cannabidiol for neurodegenerative disorders: important new clinical applications for this phytocannabinoid?

Javier Fernández-Ruiz; Onintza Sagredo; M. Ruth Pazos; Concepción García; Roger G. Pertwee; Raphael Mechoulam; José Martínez-Orgado

Cannabidiol (CBD) is a phytocannabinoid with therapeutic properties for numerous disorders exerted through molecular mechanisms that are yet to be completely identified. CBD acts in some experimental models as an anti‐inflammatory, anticonvulsant, anti‐oxidant, anti‐emetic, anxiolytic and antipsychotic agent, and is therefore a potential medicine for the treatment of neuroinflammation, epilepsy, oxidative injury, vomiting and nausea, anxiety and schizophrenia, respectively. The neuroprotective potential of CBD, based on the combination of its anti‐inflammatory and anti‐oxidant properties, is of particular interest and is presently under intense preclinical research in numerous neurodegenerative disorders. In fact, CBD combined with Δ9‐tetrahydrocannabinol is already under clinical evaluation in patients with Huntingtons disease to determine its potential as a disease‐modifying therapy. The neuroprotective properties of CBD do not appear to be exerted by the activation of key targets within the endocannabinoid system for plant‐derived cannabinoids like Δ9‐tetrahydrocannabinol, i.e. CB1 and CB2 receptors, as CBD has negligible activity at these cannabinoid receptors, although certain activity at the CB2 receptor has been documented in specific pathological conditions (i.e. damage of immature brain). Within the endocannabinoid system, CBD has been shown to have an inhibitory effect on the inactivation of endocannabinoids (i.e. inhibition of FAAH enzyme), thereby enhancing the action of these endogenous molecules on cannabinoid receptors, which is also noted in certain pathological conditions. CBD acts not only through the endocannabinoid system, but also causes direct or indirect activation of metabotropic receptors for serotonin or adenosine, and can target nuclear receptors of the PPAR family and also ion channels.


Neuropharmacology | 2013

Mechanisms of cannabidiol neuroprotection in hypoxic–ischemic newborn pigs: Role of 5HT1A and CB2 receptors

M. Ruth Pazos; Nagat Mohammed; Hector Lafuente; Martín Santos; Eva Martínez-Pinilla; Estefanía Moreno; Elsa M. Valdizán; Julián Romero; Angel Pazos; Rafael Franco; Cecilia J. Hillard; Francisco J. Alvarez; José Martínez-Orgado

The mechanisms underlying the neuroprotective effects of cannabidiol (CBD) were studied in vivo using a hypoxic-ischemic (HI) brain injury model in newborn pigs. One- to two-day-old piglets were exposed to HI for 30 min by interrupting carotid blood flow and reducing the fraction of inspired oxygen to 10%. Thirty minutes after HI, the piglets were treated with vehicle (HV) or 1 mg/kg CBD, alone (HC) or in combination with 1 mg/kg of a CB₂ receptor antagonist (AM630) or a serotonin 5HT(1A) receptor antagonist (WAY100635). HI decreased the number of viable neurons and affected the amplitude-integrated EEG background activity as well as different prognostic proton-magnetic-resonance-spectroscopy (H(±)-MRS)-detectable biomarkers (lactate/N-acetylaspartate and N-acetylaspartate/choline ratios). HI brain damage was also associated with increases in excitotoxicity (increased glutamate/N-acetylaspartate ratio), oxidative stress (decreased glutathione/creatine ratio and increased protein carbonylation) and inflammation (increased brain IL-1 levels). CBD administration after HI prevented all these alterations, although this CBD-mediated neuroprotection was reversed by co-administration of either WAY100635 or AM630, suggesting the involvement of CB₂ and 5HT(1A) receptors. The involvement of CB₂ receptors was not dependent on a CBD-mediated increase in endocannabinoids. Finally, bioluminescence resonance energy transfer studies indicated that CB₂ and 5HT(1A) receptors may form heteromers in living HEK-293T cells. In conclusion, our findings demonstrate that CBD exerts robust neuroprotective effects in vivo in HI piglets, modulating excitotoxicity, oxidative stress and inflammation, and that both CB₂ and 5HT(1A) receptors are implicated in these effects.


Journal of Neuroscience Research | 2011

Neuroprotective Effects of Phytocannabinoid-Based Medicines in Experimental Models of Huntington's Disease

Onintza Sagredo; M. Ruth Pazos; Valentina Satta; José A. Ramos; Roger G. Pertwee; Javier Fernández-Ruiz

We studied whether combinations of botanical extracts enriched in either Δ9‐tetrahydrocannabinol (Δ9‐THC) or cannabidiol (CBD), which are the main constituents of the cannabis‐based medicine Sativex, provide neuroprotection in rat models of Huntingtons disease (HD). We used rats intoxicated with 3‐nitropropionate (3NP) that were given combinations of Δ9‐THC‐ and CBD‐enriched botanical extracts. The issue was also studied in malonate‐lesioned rats. The administration of Δ9‐THC‐ and CBD‐enriched botanical extracts combined in a ratio of 1:1 as in Sativex attenuated 3NP‐induced GABA deficiency, loss of Nissl‐stained neurons, down‐regulation of CB1 receptor and IGF‐1 expression, and up‐regulation of calpain expression, whereas it completely reversed the reduction in superoxide dismutase‐1 expression. Similar responses were generally found with other combinations of Δ9‐THC‐ and CBD‐enriched botanical extracts, suggesting that these effects are probably related to the antioxidant and CB1 and CB2 receptor‐independent properties of both phytocannabinoids. In fact, selective antagonists for both receptor types, i.e., SR141716 and AM630, respectively, were unable to prevent the positive effects on calpain expression caused in 3NP‐intoxicated rats by the 1:1 combination of Δ9‐THC and CBD. Finally, this combination also reversed the up‐regulation of proinflammatory markers such as inducible nitric oxide synthase observed in malonate‐lesioned rats. In conclusion, this study provides preclinical evidence in support of a beneficial effect of the cannabis‐based medicine Sativex as a neuroprotective agent capable of delaying disease progression in HD, a disorder that is currently poorly managed in the clinic, prompting an urgent need for clinical trials with agents showing positive results in preclinical studies.


Pediatric Research | 2007

The Cannabinoid Agonist Win55212 Reduces Brain Damage in an In Vivo Model of Hypoxic-Ischemic Encephalopathy in Newborn Rats

David Fernández-López; M. Ruth Pazos; Rosa M. Tolón; M Angeles Moro; Julián Romero; Ignacio Lizasoain; José Martínez-Orgado

Neonatal hypoxic-ischemic encephalopathy (NHIE) is a devastating condition for which effective therapeutic treatments are still unavailable. Cannabinoids emerge as neuroprotective substances in adult animal studies; therefore, we aimed herein to test whether cannabinoids might reduce brain damage induced by hypoxia-ischemia (HI) in newborn rats. Thus, 7-d-old Wistar rats (P7) were exposed to 8% O2 for 120 min after left carotid artery ligature, then received s.c. vehicle (VEH) (HI+VEH), the cannabinoid agonist WIN55212 (WIN) (0.1 mg/kg), or WIN with the CB1 or CB2 receptor antagonist SR141617 (SR1) (3 mg/kg) or SR141588 (SR2) (2 mg/kg). Brain damage was assessed by magnetic resonance imaging (MRI) at 1, 3, and 7 d after the insult. At the end of the experiment, MRI findings were corroborated by histology (Nissl staining). HI+VEH showed an area of cytotoxic and vasogenic edema at 24 h after the insult, then evolving to necrosis. HI+WIN showed a similar damaged area at 24 h after the insult, but the final necrotic area was reduced by 66%. Coadministration of either SR1 or SR2 reversed the effects of WIN. In conclusion, likely by activating CB1 and CB2 receptors, WIN afforded robust neuroprotection in newborn rats after HI.


Pediatric Research | 2011

Cannabidiol reduces brain damage and improves functional recovery after acute hypoxia-ischemia in newborn pigs.

Hector Lafuente; Francisco J. Alvarez; M. Ruth Pazos; Antonia Alvarez; M Carmen Rey-Santano; Victoria Mielgo; Xabier Murgia-Esteve; Enrique Hilario; José Martínez-Orgado

Newborn piglets exposed to acute hypoxia-ischemia (HI) received i.v. cannabidiol (HI + CBD) or vehicle (HI + VEH). In HI + VEH, 72 h post-HI brain activity as assessed by amplitude-integrated EEG (aEEG) had only recovered to 42 ± 9% of baseline, near-infrared spectroscopy (NIRS) parameters remained lower than normal, and neurobehavioral performance was abnormal (27.8 ± 2.3 points, normal 36). In the brain, there were fewer normal and more pyknotic neurons, while astrocytes were less numerous and swollen. Cerebrospinal fluid concentration of neuronal-specific enolase (NSE) and S100β protein and brain tissue percentage of TNFα(+) cells were all higher. In contrast, in HI + CBD, aEEG had recovered to 86 ± 5%, NIRS parameters increased, and the neurobehavioral score normalized (34.3 ± 1.4 points). HI induced histological changes, and NSE and S100β concentration and TNFα(+) cell increases were suppressed by CBD. In conclusion, post-HI administration of CBD protects neurons and astrocytes, leading to histological, functional, biochemical, and neurobehavioral improvements.


Recent Patents on Cns Drug Discovery | 2012

Cannabinoids: Novel Medicines for the Treatment of Huntingtons Disease

Onintza Sagredo; M. Ruth Pazos; Sara Valdeolivas; Javier Fernández-Ruiz

Cannabinoid pharmacology has experienced a notable increase in the last 3 decades which is allowing the development of novel cannabinoid-based medicines for the treatment of different human pathologies, for example, Cesamet® (nabilone) or Marinol® (synthetic Δ9-tetrahydrocannabinol for oral administration) that were approved in 80s for the treatment of nausea and vomiting associated with chemotherapy treatment in cancer patients and in 90s for anorexiacachexia associated with AIDS therapy. Recently, the british company GW Pharmaceuticals plc has developed an oromucosal spray called Sativex®, which is constituted by an equimolecular combination of Δ9-tetrahydrocannabinol- and cannabidiol- enriched botanical extracts. Sativex® has been approved for the treatment of specific symptoms (i.e. spasticity and pain) of multiple sclerosis patients in various countries (i.e. Canada, UK, Spain, New Zealand). However, this cannabis- based medicine has been also proposed to be useful in other neurological disorders given the analgesic, antitumoral, anti-inflammatory, and neuroprotective properties of their components demonstrated in preclinical models. Numerous clinical trials are presently being conducted to confirm this potential in patients. We are particularly interested in the case of Huntingtons disease (HD), an autosomal-dominant inherited disorder caused by an excess of CAG repeats in the genomic allele resulting in a polyQ expansion in the encoded protein called huntingtin, and that affects primarily striatal and cortical neurons thus producing motor abnormalities (i.e. chorea) and dementia. Cannabinoids have been studied for alleviation of hyperkinetic symptoms, given their inhibitory effects on movement, and, in particular, as disease-modifying agents due to their anti-inflammatory, neuroprotective and neuroregenerative properties. This potential has been corroborated in different experimental models of HD and using different types of cannabinoid agonists, including the phytocannabinoids present in Sativex®, and we are close to initiate a clinical trial with this cannabis-based medicine to evaluate its capability as a disease-modifying agent in a population of HD patients. The present review will address all preclinical evidence supporting the potential of Sativex® for the treatment of disease progression in HD patients. The article presents some promising patents on the cannabinoids.


Journal of Biological Chemistry | 2014

Targeting CB2-GPR55 Receptor Heteromers Modulates Cancer Cell Signaling

Estefanía Moreno; Clara Andradas; Mireia Medrano; María M. Caffarel; Eduardo Pérez-Gómez; Sandra Blasco-Benito; María Gómez-Cañas; M. Ruth Pazos; Andrew J. Irving; Carme Lluis; Enric I. Canela; Javier Fernández-Ruiz; Manuel Guzmán; Peter J. McCormick; Cristina Sánchez

Background: Cannabinoid receptor CB2 (CB2R) and GPR55 are overexpressed in cancer cells and control cell fate. Results: In cancer cells, CB2R and GPR55 form heteromers that impact the signaling of each protomer. Conclusion: CB2R-GPR55 heteromers drive biphasic signaling responses as opposed to the individual receptors via cross-antagonism. Significance: These heteromers may explain some of the biphasic effects of cannabinoids and, therefore, constitute potential new targets in oncology. The G protein-coupled receptors CB2 (CB2R) and GPR55 are overexpressed in cancer cells and human tumors. Because a modulation of GPR55 activity by cannabinoids has been suggested, we analyzed whether this receptor participates in cannabinoid effects on cancer cells. Here we show that CB2R and GPR55 form heteromers in cancer cells, that these structures possess unique signaling properties, and that modulation of these heteromers can modify the antitumoral activity of cannabinoids in vivo. These findings unveil the existence of previously unknown signaling platforms that help explain the complex behavior of cannabinoids and may constitute new targets for therapeutic intervention in oncology.


European Journal of Medicinal Chemistry | 2016

Tricyclic pyrazoles. Part 8. Synthesis, biological evaluation and modelling of tricyclic pyrazole carboxamides as potential CB2 receptor ligands with antagonist/inverse agonist properties

Valeria Deiana; María Gómez-Cañas; M. Ruth Pazos; Javier Fernández-Ruiz; Battistina Asproni; Elena Cichero; Paola Fossa; Eduardo Muñoz; Francesco Deligia; Gabriele Murineddu; Moisés García-Arencibia; Gérard Aimé Pinna

Previous studies have investigated the relevance and structure-activity relationships (SARs) of pyrazole derivatives in relation with cannabinoid receptors, and the series of tricyclic 1,4-dihydroindeno[1,2-c]pyrazoles emerged as potent CB2 receptor ligands. In the present study, novel 1,4-dihydroindeno[1,2-c]pyrazole and 1H-benzo[g]indazole carboxamides containing a cyclopropyl or a cyclohexyl substituent were designed and synthesized to evaluate the influence of these structural modifications towards CB1 and CB2 receptor affinities. Among these derivatives, compound 15 (6-cyclopropyl-1-(2,4-dichlorophenyl)-N-(adamantan-1-yl)-1,4-dihydroindeno[1,2-c]pyrazole-3-carboxamide) showed the highest CB2 receptor affinity (Ki = 4 nM) and remarkable selectivity (KiCB1/KiCB2 = 2232), whereas a similar affinity, within the nM range, was seen for the fenchyl derivative (compound 10: Ki = 6 nM), for the bornyl analogue (compound 14: Ki = 38 nM) and, to a lesser extent, for the aminopiperidine derivative (compound 6: Ki = 69 nM). Compounds 10 and 14 were also highly selective for the CB2 receptor (KiCB1/KiCB2 > 1000), whereas compound 6 was relatively selective (KiCB1/KiCB2 = 27). The four compounds were also subjected to GTPγS binding analysis showing antagonist/inverse agonist properties (IC50 for compound 14 = 27 nM, for 15 = 51 nM, for 10 = 80 nM and for 6 = 294 nM), and this activity was confirmed for the three more active compounds in a CB2 receptor-specific in vitro bioassay consisting in the quantification of prostaglandin E2 release by LPS-stimulated BV2 cells, in the presence and absence of WIN55,212-2 and/or the investigated compounds. Modelling studies were also conducted with the four compounds, which conformed with the structural requirements stated for the binding of antagonist compounds to the human CB2 receptor.

Collaboration


Dive into the M. Ruth Pazos's collaboration.

Top Co-Authors

Avatar

Javier Fernández-Ruiz

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

José Martínez-Orgado

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Julián Romero

Universidad Francisco de Vitoria

View shared research outputs
Top Co-Authors

Avatar

María Gómez-Cañas

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Onintza Sagredo

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Manuel Guzmán

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Cristina Benito

Medical College of Wisconsin

View shared research outputs
Top Co-Authors

Avatar

Boris Julien

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Carolina Carrasco

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Concepción García

Complutense University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge