Miembros del Grupo
Coordinador
Investigadores
Luisa María del Mar
Castle Ramírez
Alfonso
Vazquez Miguez
Colaboradores
Diego
Sucunza Guibert
Líneas de Investigación
- Enfermedad de Gaucher y enfermedad de Parkinson: déficit de glucocerebrosidasa
- Heterómeros de receptores GPCRs como nuevas dianas farmacológicas para enfermedad de Parkinson
- Modelos de enfermedad de Parkinson en primates no humanos
- Neuroanatomía de ganglios basales en primates no humanos
- Terapia génica con vectores virales para el tratamiento de la enfermedad de Parkinson
Palabras Clave
- Adeno-associated viral vectors
- Alpha-synuclein
- Basal ganglia
- Cannabinoid receptors
- Dopamine
- Dyskinesia
- Gene Therapy
- Glucocerebrosidase
- GPCR heteromers
- MPTP
- Non-human primates
- Parkinson's disease
Publicaciones Científicas desde 2018
-
Autores: Pedrosa, M. A.; Labandeira, C. M.; Valenzuela, R.; et al.Título: AT1 receptor autoantibodies mediate effects of metabolic syndrome on dopaminergic vulnerabilityRevista: BRAIN BEHAVIOR AND IMMUNITYISSN: 0889-1591 Vol.108 2023 págs. 255 - 268ResumenThe metabolic syndrome has been associated to chronic peripheral inflammation and related with neuroinflammation and neurodegeneration, including Parkinson¿s disease. However, the responsible mechanisms are unclear. Previous studies have involved the brain renin-angiotensin system in progression of Parkinson¿s disease and the angiotensin receptor type 1 (AT1) has been recently revealed as a major marker of dopaminergic vulnerability in humans. Dysregulation of tissue renin-angiotensin system is a key common mechanism for all major components of metabolic syndrome. Circulating AT1 agonistic autoantibodies have been observed in several inflammation-related peripheral processes, and activation of AT1 receptors of endothelial cells, dopaminergic neurons and glial cells have been observed to disrupt endothelial blood -brain barrier and induce neurodegeneration, respectively. Using a rat model, we observed that metabolic syndrome induces overactivity of nigral pro-inflammatory renin-angiotensin system axis, leading to increase in oxidative stress and neuroinflammation and enhancing dopaminergic neurodegeneration, which was inhibited by treatment with AT1 receptor blockers (ARBs). In rats, metabolic syndrome induced the increase in circulating levels of LIGHT and other major pro-inflammatory cytokines, and 27-hydroxycholesterol. Furthermore, the rats showed a significant increase in serum levels of proinflammatory AT1 and angiotensin converting enzyme 2 (ACE2) autoantibodies, which correlated with levels of several metabolic syndrome parameters. We also found AT1 and ACE2 autoantibodies in the CSF of these rats. Effects of circulating autoantibodies were confirmed by chronic infusion of AT1 autoantibodies, which induced blood¿brain barrier disruption, an increase in the pro-inflammatory renin-angiotensin system activity in the substantia nigra and a significant enhancement in dopaminergic neuron death in two different rat models of Parkinson¿s disease. Observations in the rat models, were analyzed in a cohort of parkinsonian and non-parkinsonian patients with or without metabolic syndrome. Non-parkinsonian patients with metabolic syndrome showed significantly higher levels of AT1 autoantibodies than non-parkinsonian patients without metabolic syndrome. However, there was no significant difference between parkinsonian patients with metabolic syndrome or without metabolic syndrome, which showed higher levels of AT1 autoantibodies than non-parkinsonian controls. This is consistent with our recent studies, showing significant increase of AT1 and ACE2 autoantibodies in parkinsonian patients, which was related to dopaminergic degeneration and neuroinflammation. Altogether may lead to a vicious circle enhancing the progression of the disease that may be inhibited by strategies against production of these autoantibodies or AT1 receptor blockers (ARBs).
-
Autores: Fajardo Serrano, Ana (Autor de correspondencia); Rico Martin, Alberto Jose; Roda Recalde, Elvira; et al.Revista: BIOMEDICINESISSN: 2227-9059 Vol.10 N° 4 2022 págs. 746ResumenIt is without doubt that the gene therapy field is currently in the spotlight for the development of new therapeutics targeting unmet medical needs. Thus, considering the gene therapy scenario, neurological diseases in general and neurodegenerative disorders in particular are emerging as the most appealing choices for new therapeutic arrivals intended to slow down, stop, or even revert the natural progressive course that characterizes most of these devastating neurodegenerative processes. Since an extensive coverage of all available literature is not feasible in practical terms, here emphasis was made in providing some advice to beginners in the field with a narrow focus on elucidating the best delivery route available for fulfilling any given AAV-based therapeutic approach. Furthermore, it is worth nothing that the number of ongoing clinical trials is increasing at a breathtaking speed. Accordingly, a landscape view of preclinical and clinical initiatives is also provided here in an attempt to best illustrate what is ongoing in this quickly expanding field.
-
Autores: Lanciego Pérez, José Luis; Roda Recalde, Elvira; Jolinon, N.; et al.Título: Biodistribution and comparative performance of AAV-TT and AAV9 in the nonhuman primate brainRevista: MOLECULAR THERAPYISSN: 1525-0016 Vol.30 N° 4 2022 págs. 129 - 130
-
Autores: Fajardo Serrano, Ana (Autor de correspondencia); Rico Martin, Alberto Jose; Roda Recalde, Elvira; et al.Revista: INTERNATIONAL JOURNAL OF MOLECULAR SCIENCESISSN: 1422-0067 Vol.22 N° 12 2021 págs. 6389ResumenIt is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson's disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson's disease and related synucleinopathies.
-
Autores: Galán-Ganga, M.; Rodríguez-Cueto, C.; Merchán-Rubira, J.; et al.Revista: ACTA NEUROPATHOLOGICA COMMUNICATIONSISSN: 2051-5960 Vol.9 N° 1 2021 págs. 90ResumenTauopathies are a group of neurodegenerative diseases characterized by the alteration/aggregation of TAU protein, for which there is still no effective treatment. Therefore, new pharmacological targets are being sought, such as elements of the endocannabinoid system (ECS). We analysed the occurrence of changes in the ECS in tauopathies and their implication in the pathogenesis. By integrating gene expression analysis, immunofluorescence, genetic and adeno-associated virus expressing TAU mouse models, we found a TAU-dependent increase in CB2 receptor expression in hippocampal neurons, that occurs as an early event in the pathology and was maintained until late stages. These changes were accompanied by alterations in the endocannabinoid metabolism. Remarkably, CB2 ablation in mice protects from neurodegeneration induced by hTAU(P301L) overexpression, corroborated at the level of cognitive behaviour, synaptic plasticity, and aggregates of insoluble TAU. At the level of neuroinflammation, the absence of CB2 did not produce significant changes in concordance with a possible neuronal location rather than its classic glial expression in these models. These findings were corroborated in post-mortem samples of patients with Alzheimer's disease, the most common tauopathy. Our results show that neurons with accumulated TAU induce the expression of the CB2 receptor, which enhances neurodegeneration. These results are important for our understanding of disease mechanisms, providing a novel therapeutic strategy to be investigated in tauopathies.
-
Autores: Sucunza, D.; Rico Martin, Alberto Jose (Autor de correspondencia); Roda Recalde, Elvira; et al.Revista: INTERNATIONAL JOURNAL OF MOLECULAR SCIENCESISSN: 1422-0067 Vol.22 N° 9 2021 págs. 4825ResumenMutations in the GBA1 gene coding for glucocerebrosidase (GCase) are the main genetic risk factor for Parkinson's disease (PD). Indeed, identifying reduced GCase activity as a common feature underlying the typical neuropathological signatures of PD-even when considering idiopathic forms of PD-has recently paved the way for designing novel strategies focused on enhancing GCase activity to reduce alpha-synuclein burden and preventing dopaminergic cell death. Here we have performed bilateral injections of a viral vector coding for the mutated form of alpha-synuclein (rAAV9-SynA53T) for disease modeling purposes, both in mice as well as in nonhuman primates (NHPs), further inducing a progressive neuronal death in the substantia nigra pars compacta (SNpc). Next, another vector coding for the GBA1 gene (rAAV9-GBA1) was unilaterally delivered in the SNpc of mice and NHPs one month after the initial insult, together with the contralateral delivery of an empty/null rAAV9 for control purposes. Obtained results showed that GCase enhancement reduced alpha-synuclein burden, leading to improved survival of dopaminergic neurons. Data reported here support using GCase gene therapy as a disease-modifying treatment for PD and related synucleinopathies, including idiopathic forms of these disorders.
-
Autores: Blandini, F.; Cilia, R.; Cerri, S. ; et al.Revista: MOVEMENT DISORDERSISSN: 0885-3185 Vol.34 N° 1 2019 págs. 9 - 21ResumenGlucocerebrosidase is a lysosomal enzyme. The characterization of a direct link between mutations in the gene coding for glucocerebrosidase (GBA1) with the development of Parkinson's disease and dementia with Lewy bodies has heightened interest in this enzyme. Although the mechanisms through which glucocerebrosidase regulates the homeostasis of -synuclein remains poorly understood, the identification of reduced glucocerebrosidase activity in the brains of patients with PD and dementia with Lewy bodies has paved the way for the development of novel therapeutic strategies directed at enhancing glucocerebrosidase activity and reducing -synuclein burden, thereby slowing down or even preventing neuronal death. Here we reviewed the current literature relating to the mechanisms underlying the cross talk between glucocerebrosidase and -synuclein, the GBA1 mutation-associated clinical phenotypes, and ongoing therapeutic approaches targeting glucocerebrosidase. (c) 2018 International Parkinson and Movement Disorder Society
-
Autores: Erro, M. E. (Autor de correspondencia); Zelayaz, M. V.; Mendioroz, M.; et al.Título: Globular glial tauopathy caused by MAPT P301T mutation: clinical and neuropathological findingsRevista: JOURNAL OF NEUROLOGYISSN: 0340-5354 Vol.266 N° 10 2019 págs. 2396 - 2405ResumenObjective To describe the clinical, biochemical, and neuropathological findings of an autosomal dominant globular glial tauopathy caused by the P301T mutation at the MAPT gene. Methods Five patients from two unrelated pedigrees underwent clinical evaluation. Genetic analysis, brain pathological examination, and biochemical analysis of tau were performed. Results The patients studied were 3 men and 2 women with a mean age at onset of 52.2 years and mean disease duration of 5.2 years. Three patients presented a corticobasal syndrome, one patient an asymmetric pyramidal syndrome compatible with primary lateral sclerosis, and one patient a frontotemporal dementia. In both pedigrees (4 patients) Sanger sequencing showed the p.P301T mutation in exon 10 of the MAPT gene. Neuropathological findings consisted of atrophy of frontal and temporal lobes with marked spongiosis and astrogliosis, and abundant phosphorylated tau protein deposits in the frontal and temporal cortex, limbic area, basal ganglia, and brain stem. The most striking finding was the presence of oligodendroglial 4R phospho-tau globular positive inclusions in the white matter and cortex. Globose-type neurofibrillary neuronal tangles, and in particular astrocytic globular inclusions and coarse tufts, were present in the grey matter. Biochemical analysis of sarkosyl-insoluble fractions revealed two tau bands of 64 and 68 kDa and case-dependent bands of lower molecular weight. Conclusion This is the first pathological and biochemical study of the MAPT p.P301T mutation showing variable clinical manifestation and neuropathological phenotype of globular glial tauopathy not only among different families but also within families.
-
Autores: Pignataro, D.; Sucunza, D.; Rico Martin, Alberto Jose; et al.Revista: JOURNAL OF NEURAL TRANSMISSIONISSN: 0300-9564 Vol.125 N° 3 2018 págs. 575 - 589ResumenThe field of gene therapy has recently witnessed a number of major conceptual changes. Besides the traditional thinking that comprises the use of viral vectors for the delivery of a given therapeutic gene, a number of original approaches have been recently envisaged, focused on using vectors carrying genes to further modify basal ganglia circuits of interest. It is expected that these approaches will ultimately induce a therapeutic potential being sustained by gene-induced changes in brain circuits. Among others, at present, it is technically feasible to use viral vectors to (1) achieve a controlled release of neurotrophic factors, (2) conduct either a transient or permanent silencing of any given basal ganglia circuit of interest, (3) perform an in vivo cellular reprogramming by promoting the conversion of resident cells into dopaminergic-like neurons, and (4) improving levodopa efficacy over time by targeting aromatic l-amino acid decarboxylase. Furthermore, extensive research efforts based on viral vectors are currently ongoing in an attempt to better replicate the dopaminergic neurodegeneration phenomena inherent to the progressive intraneuronal aggregation of alpha-synuclein. Finally, a number of incoming strategies will soon emerge over the horizon, these being sustained by the underlying goal of promoting alpha-synuclein clearance, such as, for instance, gene therapy initiatives based on increasing the activity of glucocerebrosidase. To provide adequate proof-of-concept on safety and efficacy and to push forward true translational initiatives based on these different types of gene therapies before entering into clinical trials, the use of non-human primate models undoubtedly plays an instrumental role.
-
Autores: Reyes-Resina, I.; Aguinaga, D.; Labandeira-García, J. L.; et al.Revista: HISTOLOGY AND HISTOPATHOLOGYISSN: 0213-3911 Vol.33 N° 9 2018 págs. 909 - 917ResumenImmunochemical detection of G-protein-coupled receptors (GPCRs) in cells and tissues was a technical challenge for years. After the discovery of formation of GPCR dimers/trimers/tetramers in transfected cells, a most recent challenge has been to confirm receptor-receptor interactions in natural sources. The occurrence of dimers or higher order oligomers is important from a therapeutic point of view, mainly because their physiology/pharmacology is different from those of individual receptors. On the one hand, pathophysiological factors need to count more on GPCR dimers than on individual receptors. On the other hand, the expression of dimers, trimers, etc. may change in pathological conditions and/or along the course of a disease. This review will focus on G-protein-coupled receptor dimers, on how to detect them by novel histological techniques and on how the detection may be used in diagnosis and therapy of ailments of the central nervous system, for instance in neurodegenerative diseases and gliomas.
-
Autores: González-Dopeso Reyes, Iria María; Sucunza, D.; Rico Martin, Alberto Jose; et al.Revista: BRAIN STRUCTURE AND FUNCTIONISSN: 1863-2653 Vol.223 N° 1 2018 págs. 343 - 355ResumenGlucocerebrosidase (GCase) is a lysosomal enzyme encoded by the GBA1 gene. Mutations in GBA1 gene lead to Gaucher¿s disease, the most prevalent lysosomal storage disorder. GBA1 mutations reduce GCase activity, therefore promoting the aggregation of alpha-synuclein, a common neuropathological finding underlying Parkinson¿s disease (PD) and dementia with Lewy bodies. However, it is also worth noting that a direct link between GBA1 mutations and alpha-synuclein aggregation indicating cause and effect is still lacking, with limited experimental evidence to date. Bearing in mind that a number of strategies increasing GCase expression for the treatment of PD are currently under development, here we sought to analyze the baseline expression of GCase in the brain of Macaca fascicularis, which has often been considered as the gold-standard animal model of PD. Although as with other lysosomal enzymes, GCase is expected to be ubiquitously expressed, here a number of regional variations have been consistently found, together with several specific neurochemical phenotypes expressing very high levels of GCase. In this regard, the most enriched expression of GCase was constantly found in cholinergic neurons from the nucleus basalis of Meynert, dopaminergic cells in the substantia nigra pars compacta, serotoninergic neurons from the raphe nuclei, as well as in noradrenergic neurons located in the locus ceruleus. Moreover, it is also worth noting that moderate levels of expression were also found in a number of areas within the paleocortex and archicortex, such as the entorhinal cortex and the hippocampal formation, respectively.
-
Autores: García-Gutiérrez, M. S.; Navarrete, F.; Navarro Díez, Gemma; et al.Revista: NEUROTHERAPEUTICSISSN: 1933-7213 Vol.15 N° 3 2018 págs. 796 - 806ResumenRecent studies point to the cannabinoid CB2 receptors (CB2r) and the non-cannabinoid receptor GPR55 as potential key targets involved in the response to stress, anxiety, and depression. Considering the close relationship between neuropsychiatric disorders and suicide, the purpose of this study was to evaluate the potential alterations of CB2r and GPR55 in suicide victims. We analyzed gene and protein expression of both receptors by real-time PCR and western blot, respectively, in the dorsolateral prefrontal cortex (DLPFC) of 18 suicide victims with no clinical psychiatric history or treatment with anxiolytics or antidepressants, and 15 corresponding controls. We used in situ proximity ligation assay to evaluate whether the receptors formed heteromeric complexes and to determine the expression level of these heteromers, also assessing the co-expression of heteromers in neurons, astroglia, or microglia cells. CB2r and GPR55 gene expressions were significantly lower (by 33 and 41%, respectively) in the DLPFC of suicide cases. CB2r protein expression was higher, as were CB2-GPR55 heteroreceptor complexes. The results also revealed the presence of CB2-GPR55 receptor heteromers in both neurons and astrocytes, whereas microglial cells showed no expression. We did not observe any significant alterations of GPR55 protein expression. Additional studies will be necessary to evaluate if these alterations are reproducible in suicide victims diagnosed with different psychiatric disorders. Taken together, the results suggest that CB2r and GPR55 may play a relevant role in the neurobiology of suicide.
-
Autores: Navarrete, F. ; Garcia-Gutierrez, M. S.; Aracil-Fernandez, A.; et al.Revista: NEUROTHERAPEUTICSISSN: 1933-7213 Vol.15 N° 2 2018 págs. 459 - 469ResumenPrevious studies suggest that the endocannabinoid system plays an important role in the neuropathological basis of Parkinson's disease (PD). This study was designed to detect potential alterations in the cannabinoid receptors CB1 (CB(1)r) and CB2 (A isoform, CB(2A)r), and in monoacylglycerol lipase (MAGL) gene expression in the substantia nigra (SN) and putamen (PUT) of patients with PD. Immunohistochemical studies were performed to identify precise CB(2)r cellular localization in the SN of control and PD patients. To ensure the validity and reliability of gene expression data, the RNA integrity number (RIN) was calculated. CB(1)r, CB(2A)r, and MAGL gene expressions were evaluated by real-time polymerase chain reaction (real-time PCR) using Taqman assays. Immunohistochemical experiments with in situ proximity ligation assay (PLA) were used to detect the precise cellular localization of CB(2)r in neurons, astrocytes, and/or microglia. All RIN values from control and PD postmortem brain samples were > 6. CB(1)r gene expression was unchanged in the SN but significantly higher in the PUT of patients with PD. CB(2A)r gene expression was significantly increased (4-fold) in the SN but decreased in the PUT, whereas MAGL gene expression was decreased in the SN and increased in the PUT. Immunohistochemical analyses revealed that CB(2)r co-localize with astrocytes but not with neurons or microglial cells in the SN. The results of the present study suggest that CB(1)r, CB(2)r, and MAGL are closely related to the neuropathological processes of PD. Therefore, the pharmacological modulation of these targets could represent a new potential therapeutic tool for the management of PD.
-
Autores: Navarro Díez, Gemma; Borroto-Escuela, D.; Angelats, E.; et al.Revista: BRAIN BEHAVIOR AND IMMUNITYISSN: 0889-1591 Vol.67 2018 págs. 139 - 151ResumenEndocannabinoids are important regulators of neurotransmission and, acting on activated microglia, they are postulated as neuroprotective agents. Endocannabinoid action is mediated by CB1 and CB2 receptors, which may form heteromeric complexes (CB1-CB2Hets) with unknown function in microglia. We aimed at establishing the expression and signaling properties of cannabinoid receptors in resting and LPS/IFN-¿-activated microglia. In activated microglia mRNA transcripts increased (2 fold for CB1 and circa 20 fold for CB2), whereas receptor levels were similar for CB1 and markedly upregulated for CB2; CB1-CB2Hets were also upregulated. Unlike in resting cells, CB2 receptors became robustly coupled to Gi in activated cells, in which CB1-CB2Hets mediated a potentiation effect. Hence, resting cells were refractory while activated cells were highly responsive to cannabinoids. Interestingly, similar results were obtained in cultures treated with ß-amyloid (Aß1-42). Microglial activation markers were detected in the striatum of a Parkinson¿s disease (PD) model and, remarkably, in primary microglia cultures from the hippocampus of mutant ß-amyloid precursor protein (APPSw,Ind) mice, a transgenic Alzheimer¿s disease (AD) model. Also of note was the similar cannabinoid receptor signaling found in primary cultures of microglia from APPSw,Ind and in cells from control animals activated using LPS plus IFN-¿. Expression of CB1-CB2Hets was increased in the striatum from rats rendered dyskinetic by chronic levodopa treatment. In summary, our results showed sensitivity of activated microglial cells to cannabinoids, increased CB1-CB2Het expression in activated microglia and in microglia from the hippocampus of an AD model, and a correlation between levodopa-induced dyskinesia and striatal microglial activation in a PD model. Cannabinoid receptors and the CB1-CB2 heteroreceptor complex in activated microglia have potential as targets in the treatment of neurodegenerative diseases.
-
Autores: Costa-Besada, M. A. ; Valenzuela, R.; Garrido-Gil, P. ; et al.Revista: MOLECULAR NEUROBIOLOGYISSN: 0893-7648 Vol.55 N° 7 2018 págs. 5847 - 5867ResumenIn addition to the classical hormonal (tissue-to-tissue) renin-angiotensin system (RAS), there are a paracrine (cell-to-cell) and an intracrine (intracellular/nuclear) RAS. A local paracrine brain RAS has been associated with several brain disorders, including Parkinson's disease (PD). Classically, angiotensin II (Ang II) is the main RAS effector peptide and acts through two major receptors: Ang II type 1 and 2 (AT1 and AT2) receptors. It has been shown that enhanced activation of the Ang II/AT1 axis exacerbates dopaminergic cell death. Several new components of the RAS have more recently been discovered. However, the role of new Ang 1-7/Mas receptor RAS component was not investigated in the brain and particularly in the dopaminergic system. In the present study, we observed Mas receptor labeling in dopaminergic neurons and glial cells in rat mesencephalic primary cultures; substantia nigra of rats, monkeys, and humans; and human induced pluripotent stem (iPS) cells derived from healthy controls and sporadic PD patients. The present data support a neuroprotective role of the Ang 1-7/Mas receptor axis in the dopaminergic system. We observed that this axis is downregulated with aging, which may contribute to the aging-related vulnerability to neurodegeneration. We have also identified an intracellular Ang 1-7/Mas axis that modulates mitochondrial and nuclear levels of superoxide. The present data suggest that nuclear RAS receptors regulate the adequate balance between the detrimental and the protective arms of the cell RAS. The results further support that the brain RAS should be taken into account for the design of new therapeutic strategies for PD.
-
Autores: Rodriguez-Perez, A. I.; Sucunza, D. ; Pedrosa, M. A.; et al.Revista: NEUROTHERAPEUTICSISSN: 1933-7213 Vol.15 N° 4 2018 págs. 1063 - 1081ResumenThe loss of dopaminergic neurons and alpha-synuclein accumulation are major hallmarks of Parkinson's disease (PD), and it has been suggested that a major mechanism of alpha-synuclein toxicity is microglial activation. The lack of animal models that properly reproduce PD, and particularly the underlying synucleinopathy, has hampered the clarification of PD mechanisms and the development of effective therapies. Here, we used neurospecific adeno-associated viral vectors serotype 9 coding for either the wild-type or mutated forms of human alpha-synuclein (WT and SynA53T, respectively) under the control of a synapsin promoter to further induce a marked dopaminergic neuron loss together with an important microglial neuroinflammatory response. Overexpression of neuronal alpha-synuclein led to increased expression of angiotensin type 1 receptors and NADPH oxidase activity, together with a marked increase in the number of OX-6-positive microglial cells and expression of markers of phagocytic activity (CD68) and classical pro-inflammatory/M1 microglial phenotype markers such as inducible nitric oxide synthase, tumor necrosis factor alpha, interleukin-1 beta, and IL-6. Moreover, a significant decrease in the expression of markers of immunoregulatory/M2 microglial phenotype such as the enzyme arginase-1 was constantly observed. Interestingly, alpha-synuclein-induced changes in microglial phenotype markers and dopaminergic neuron death were inhibited by simultaneous treatment with the angiotensin type 1 blockers candesartan or telmisartan. Our results suggest the repurposing of candesartan and telmisartan as a neuroprotective strategy for PD.
-
Autores: Castro-Sánchez, S.; García-Yagüe, A. J. ; López-Royo, T.; et al.Revista: GLIAISSN: 0894-1491 Vol.66 N° 8 2018 págs. 1752 - 1762ResumenParkinson's disease (PD) is the second most common neurodegenerative disorder characterized by the degeneration of dopaminergic neurons of the substantia nigra and the accumulation of protein aggregates, called Lewy bodies, where the most abundant is alpha-synuclein (alpha-SYN). Mutations of the gene that codes for alpha-SYN (SNCA), such as the A53T mutation, and duplications of the gene generate cases of PD with autosomal dominant inheritance. As a result of the association of inflammation with the neurodegeneration of PD, we analyzed whether overexpression of wild-type alpha-SYN (alpha-SYNWT) or mutated alpha-SYN (alpha-SYNA53T) are involved in the neuronal dopaminergic loss and inflammation process, along with the role of the chemokine fractalkine (CX3CL1) and its receptor (CX3CR1). We generated in vivo murine models overexpressing human alpha-SYNWT or alpha-SYNA53T in wild type (Cx3cr1(+/+)) or deficient (Cx3cr1(-/-)) mice for CX3CR1 using unilateral intracerebral injection of adeno-associated viral vectors. No changes in CX3CL1 levels were observed by immunofluorescence or analysis by qRT-PCR in this model. Interestingly, the expression alpha-SYNWT induced dopaminergic neuronal death to a similar degree in both genotypes. However, the expression of alpha-SYNA53T produced an exacerbated neurodegeneration, enhanced in the Cx3cr1(-/-) mice. This neurodegeneration was accompanied by an increase in neuroinflammation and microgliosis as well as the production of pro-inflammatory markers, which were exacerbated in Cx3cr1(-/-) mice overexpressing alpha-SYNA53T. Furthermore, we observed that in primary microglia CX3CR1 was a critical factor in the modulation of microglial dynamics in response to alpha-SYNWT or alpha-SYNA53T. Altogether, our study reveals that CX3CR1 plays an essential role in neuroinflammation induced by alpha-SYNA53T.
-
Autores: Sierra San Nicolás, S.; Lanciego Pérez, José LuisLibro: Velázquez. Farmacología Básica y ClínicaISSN: 9786078546077 2018 págs. 259 - 270
-
Autores: Lanciego Pérez, José Luis2018
Proyectos desde 2018
-
Título: Terapia génica con glucocerebrosidasa para el tratamiento de la enfermedad de Parkinson.Código de expediente: 0011-1383-2019-000006Investigador principal: JOSE LUIS LANCIEGO PEREZ, JOSE LUIS LANCIEGO PEREZ.Financiador: GOBIERNO DE NAVARRAConvocatoria: 2019 - GN INDUSTRIA PROYECTOS CENTROS TECNOLOGICOS Y ORGANISMOS DE INVESTIGACIONFecha de inicio: 01-12-2018Fecha fin: 30-11-2019Importe concedido: 132.648,00€Otros fondos: -
-
Título: TERAPIA GENICA CON GLUCOCEREBROSIDASA PARA EL TRATAMIENTO DE SINUCLEINOPATIA PARKINSONIANA EN PRIMATES NO HUMANOSCódigo de expediente: BFU2017-82407-RInvestigador principal: JOSE LUIS LANCIEGO PEREZ.Financiador: MINISTERIO DE CIENCIA E INNOVACIÓNConvocatoria: 2017 MINECO RETOSFecha de inicio: 01-01-2018Fecha fin: 31-12-2020Importe concedido: 108.900,00€Otros fondos: Fondos FEDER
-
Título: Parkinsonismos asociados a enfermedad de Gaucher: glucocerebrosidasa para el aclaramiento de alfa?sinucleína.Código de expediente: 46/2017Investigador principal: JOSE LUIS LANCIEGO PEREZ.Financiador: GOBIERNO DE NAVARRA. DEPARTAMENTO DE SALUDConvocatoria: 2017 PROYECTOS DE I+D EN SALUDFecha de inicio: 15-12-2017Fecha fin: 14-12-2020Importe concedido: 74.106,01€Otros fondos: Fondos FEDER
-
Título: Comorbidity mechanisms of anxiety and Parkinson'sCódigo de expediente: 848002Investigador principal: JOSE LUIS LANCIEGO PEREZFinanciador: COMISIÓN EUROPEAConvocatoria: SC1-BHC-02-2019Fecha de inicio: 01-01-2020Fecha fin: 31-12-2023Importe concedido: 130.625,00€Otros fondos: -
-
Título: REPROPARK: New experimental therapeutic approaches for Parkinson¿s disease by direct DA neuronal reprogrammingCódigo de expediente: 340527Investigador principal: JOSE LUIS LANCIEGO PEREZ JOSE LUIS LANCIEGO PEREZFinanciador: COMISIÓN EUROPEAConvocatoria: FP7-IDEASFecha de inicio: 01-06-2014Fecha fin: 31-12-2019Importe concedido: 420.500,00€Otros fondos: -
-
Título: Cannabinoid receptor heteromers com a dianes terapeutiques en la malaltia de ParkinsonInvestigador principal: JOSE LUIS LANCIEGO PEREZ, JOSE LUIS LANCIEGO PEREZFinanciador: FUNDACIO "LA MARATO DE TV3"Convocatoria: 2014 PROYECTOS DE I+D FUNDACIÓN LA MARATO TV3Fecha de inicio: 04-03-2015Fecha fin: 30-11-2018Importe concedido: 149.056,25€