Network


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

Hotspot


Dive into the research topics where Javier Marín-Prida is active.

Publication


Featured researches published by Javier Marín-Prida.


Brain Research Bulletin | 2011

C-Phycocyanin is neuroprotective against global cerebral ischemia/reperfusion injury in gerbils

Giselle Pentón-Rol; Javier Marín-Prida; Gilberto L. Pardo-Andreu; Gregorio Martínez-Sánchez; Emilio Felino Acosta-Medina; Alain Valdivia-Acosta; Nielsen Lagumersindez-Denis; Efraín Rodríguez-Jiménez; Alexey Llópiz-Arzuaga; Pedro Lopez-Saura; Gerardo Guillén-Nieto; Eduardo Pentón-Arias

Although the huge economic and social impact and the predicted incidence increase, neuroprotection for ischemic stroke remains as a therapeutically empty niche. In the present study, we investigated the rationale of the C-Phycocyanin (C-PC) treatment on global cerebral ischemia/reperfusion (I/R) injury in gerbils. We demonstrated that C-PC given either prophylactically or therapeutically was able to significantly reduce the infarct volume as assessed by triphenyltetrazolium chloride (TTC) staining and the neurological deficit score 24h post-stroke. In addition, C-PC exhibited a protective effect against hippocampus neuronal cell death, and significantly improved the functional outcome (locomotor behavior) and gerbil survival after 7 days of reperfusion. Malondialdehyde (MDA), peroxidation potential (PP) and ferric reducing ability of plasma (FRAP) were assayed in serum and brain homogenates to evaluate the redox status 24h post-stroke. The treatment with C-PC prevented the lipid peroxidation and the increase of FRAP in both tissue compartments. These results suggest that the protective effects of C-PC are most likely due to its antioxidant activity, although its anti-inflammatory and immuno-modulatory properties reported elsewhere could also contribute to neuroprotection. To our knowledge, this is the first report of the neuroprotective effect of C-PC in an experimental model of global cerebral I/R damage, and strongly indicates that C-PC may represent a potential preventive and acute disease modifying pharmacological agent for stroke therapy.


International Immunopharmacology | 2011

C-Phycocyanin ameliorates experimental autoimmune encephalomyelitis and induces regulatory T cells

Giselle Pentón-Rol; Gregorio Martínez-Sánchez; Majel Cervantes-Llanos; Nielsen Lagumersindez-Denis; Emilio Felino Acosta-Medina; Viviana Falcón-Cama; Ruby Alonso-Ramírez; Carmen Valenzuela-Silva; Efraín Rodríguez-Jiménez; Alexey Llópiz-Arzuaga; Javier Marín-Prida; Pedro Lopez-Saura; Gerardo Guillén-Nieto; Eduardo Pentón-Arias

For decades Experimental Autoimmune Encephalitis (EAE) has remained as an unsurpassed multiple sclerosis (MS) animal model. C-Phycocyanin (C-Pc) has been reported to exhibit pharmacological properties that may be expected to symptomatically improve EAE and MS. However, in this paper we reveal a basic underlying mechanism that may provide a new approach to the rationale of the overall beneficial effect of this natural antioxidant. We demonstrate that C-Pc is able to trigger mechanisms preventing or downgrading EAE expression and induces a regulatory T cell (Treg) response, in peripheral blood mononuclear cells (PBMC) from MS patients. These results agree with reports suggesting that Treg limit acute MS attacks and that C-Pc may act as a neuroprotector and thereby reverts the organic and functional damage in neurodegenerative disorders of the central nervous system (CNS). Moreover, evidence is provided on the antioxidant activity of C-Pc within the CNS, intended to improve the myelin and axonal damage of EAE induced Lewis rats. Our results indicate that specific Treg activation may represent a central and essential mechanism in supporting the therapeutic potential of C-Pc for MS and may lead to new and more effective therapies; this property would then complement and enhance other proven active principles such as interferons (IFN), giving rise to combined therapies.


Brain Research Bulletin | 2012

C-Phycocyanin protects SH-SY5Y cells from oxidative injury, rat retina from transient ischemia and rat brain mitochondria from Ca2+/phosphate-induced impairment

Javier Marín-Prida; Giselle Pentón-Rol; Fernando P. Rodrigues; Luciane C. Alberici; Karina Stringhetta; Andréia Machado Leopoldino; Zeki Naal; Ana Cristina M. Polizello; Alexey Llópiz-Arzuaga; Marcela Nunes Rosa; José Luiz Liberato; Wagner Ferreira dos Santos; Sérgio A. Uyemura; Eduardo Pentón-Arias; Carlos Curti; Gilberto L. Pardo-Andreu

Oxidative stress and mitochondrial impairment are essential in the ischemic stroke cascade and eventually lead to tissue injury. C-Phycocyanin (C-PC) has previously been shown to have strong antioxidant and neuroprotective actions. In the present study, we assessed the effects of C-PC on oxidative injury induced by tert-butylhydroperoxide (t-BOOH) in SH-SY5Y neuronal cells, on transient ischemia in rat retinas, and in the calcium/phosphate-induced impairment of isolated rat brain mitochondria (RBM). In SH-SY5Y cells, t-BOOH induced a significant reduction of cell viability as assessed by an MTT assay, and the reduction was effectively prevented by treatment with C-PC in the low micromolar concentration range. Transient ischemia in rat retinas was induced by increasing the intraocular pressure to 120mmHg for 45min, which was followed by 15min of reperfusion. This event resulted in a cell density reduction to lower than 50% in the inner nuclear layer (INL), which was significantly prevented by the intraocular pre-treatment with C-PC for 15min. In the RBM exposed to 3mM phosphate and/or 100μM Ca(2+), C-PC prevented in the low micromolar concentration range, the mitochondrial permeability transition as assessed by mitochondrial swelling, the membrane potential dissipation, the increase of reactive oxygen species levels and the release of the pro-apoptotic cytochrome c. In addition, C-PC displayed a strong inhibitory effect against an electrochemically-generated Fenton reaction. Therefore, C-PC is a potential neuroprotective agent against ischemic stroke, resulting in reduced neuronal oxidative injury and the protection of mitochondria from impairment.


Toxicology and Applied Pharmacology | 2013

Phycocyanobilin promotes PC12 cell survival and modulates immune and inflammatory genes and oxidative stress markers in acute cerebral hypoperfusion in rats

Javier Marín-Prida; Nancy Pavón-Fuentes; Alexey Llópiz-Arzuaga; Julio R. Fernández-Massó; Livan Delgado-Roche; Yssel Mendoza-Marí; Seydi Pedroso Santana; Alieski Cruz-Ramírez; Carmen Valenzuela-Silva; Marcelo Nazábal-Gálvez; Alberto Cintado-Benítez; Gilberto L. Pardo-Andreu; Nadia Polentarutti; Federica Riva; Eduardo Pentón-Arias; Giselle Pentón-Rol

Since the inflammatory response and oxidative stress are involved in the stroke cascade, we evaluated here the effects of Phycocyanobilin (PCB, the C-Phycocyanin linked tetrapyrrole) on PC12 cell survival, the gene expression and the oxidative status of hypoperfused rat brain. After the permanent bilateral common carotid arteries occlusion (BCCAo), the animals were treated with saline or PCB, taking samples 24h post-surgery. Global gene expression was analyzed with GeneChip Rat Gene ST 1.1 from Affymetrix; the expression of particular genes was assessed by the Fast SYBR Green RT-PCR Master Mix and Bioplex methods; and redox markers (MDA, PP, CAT, SOD) were evaluated spectrophotometrically. The PCB treatment prevented the H2O2 and glutamate induced PC12 cell injury assessed by the MTT assay, and modulated 190 genes (93 up- and 97 down-regulated) associated to several immunological and inflammatory processes in BCCAo rats. Furthermore, PCB positively modulated 19 genes mostly related to a detrimental pro-inflammatory environment and counteracted the oxidative imbalance in the treated BCCAo animals. Our results support the view of an effective influence of PCB on major inflammatory mediators in acute cerebral hypoperfusion. These results suggest that PCB has a potential to be a treatment for ischemic stroke for which further studies are needed.


Chemico-Biological Interactions | 2014

Clusianone, a naturally occurring nemorosone regioisomer, uncouples rat liver mitochondria and induces HepG2 cell death

Felippe H.Z. Reis; Gilberto L. Pardo-Andreu; Yanier Nuñez-Figueredo; Osmany Cuesta-Rubio; Javier Marín-Prida; Sérgio A. Uyemura; Carlos Curti; Luciane C. Alberici

Clusianone is a member of the polycyclic polyprenylated acylphloroglucinol family of natural products; its cytotoxic mechanism is unknown. Clusianone is a structural isomer of nemorosone, which is a mitochondrial uncoupler and a well-known cytotoxic anti-cancer agent; thus, we addressed clusianone action at the mitochondria and its potential cytotoxic effects on cancer cells. In the HepG2 hepatocarcinoma cell line, clusianone induced mitochondrial membrane potential dissipation, ATP depletion and phosphatidyl serine externalization; this later event is indicative of apoptosis induction. In isolated mitochondria from rat liver, clusianone promoted protonophoric mitochondrial uncoupling. This was evidenced by the dissipation of mitochondrial membrane potential, an increase in resting respiration, an inhibition of Ca(2+) influx, stimulation of Ca(2+) efflux in Ca(2+)-loaded mitochondria, a decrease in ATP and NAD(P)H levels, generation of ROS, and swelling of valinomycin-treated organelles in hyposmotic potassium acetate media. The cytotoxic and uncoupling actions of clusianone were appreciably less than those of nemorosone, likely due to the presence of an intra-molecular hydrogen bond with the juxtaposed carbonyl group at the C15 position. Therefore, clusianone is capable of pharmacologically increasing the leakage of protons from the mitochondria and with favorable cytotoxicity in relation to nemorosone.


Life Sciences | 2018

Beneficial effects of oral administration of C-Phycocyanin and Phycocyanobilin in rodent models of experimental autoimmune encephalomyelitis

Majel Cervantes-Llanos; Nielsen Lagumersindez-Denis; Javier Marín-Prida; Nancy Pavón-Fuentes; Viviana Falcón-Cama; Beatriz Piniella-Matamoros; Hanlet Camacho-Rodríguez; Julio R. Fernández-Massó; Carmen Valenzuela-Silva; Ivette Raíces-Cruz; Eduardo Pentón-Arias; Mauro M. Teixeira; Giselle Pentón-Rol

&NA; The only three oral treatments currently available for multiple sclerosis (MS) target the relapsing forms of the disease and concerns regarding efficacy, safety and tolerability limit their use. Identifying novel oral disease‐modifying therapies for MS, targeting both its inflammatory and neurodegenerative components is still a major goal. Aim: The scope of this study was to provide evidence that the oral administration of C‐Phycocyanin (C‐PC), the main biliprotein of the Spirulina platensis cyanobacteria and its tetrapyrrolic prosthetic group, Phycocyanobilin (PCB), exert ameliorating actions on rodent models of experimental autoimmune encephalomyelitis (EAE). Main methods: EAE was induced in Lewis rats using the spinal cord encephalitogen from Sprague Dawley rats and in C57BL6 mice with MOG35–55 peptide. Clinical signs, motor function, oxidative stress markers, cytokine levels by ELISA and transmission electron microscopy analysis were assessed. Key findings: Either prophylactic or early therapeutic administration of C‐PC to Lewis rats with EAE, significantly improved clinical signs and restored the motor function of the animals. Furthermore, C‐PC positively modulated oxidative stress markers measured in brain homogenate and serum and protected the integrity of cerebral myelin sheaths as shown by transmission electron microscopy analysis. In C57BL/6 mice with EAE, PCB orally improved clinical status of the animals and reduced the expression levels of brain IL‐6 and IFN‐&ggr; proinflammatory cytokines. Significance: These results, for the first time, support the fact that both C‐PC and PCB administered orally could potentially improve neuroinflammation, protect from demyelination and axonal loss, which may be translated into an improved quality of life for MS patients.


Toxicology in Vitro | 2017

The cytotoxic effects of VE-3N, a novel 1,4-dihydropyridine derivative, involve the mitochondrial bioenergetic disruption via uncoupling mechanisms

Javier Marín-Prida; Gilberto L. Pardo Andreu; Camila Pederiva Rossignoli; Michael González Durruthy; Estael Ochoa Rodríguez; Yamila Verdecia Reyes; Roberto Fernández Acosta; Sérgio A. Uyemura; Luciane C. Alberici

Several 1,4-dihydropyridine derivatives overcome the multidrug resistance in tumors, but their intrinsic cytotoxic mechanisms remain unclear. Here we addressed if mitochondria are involved in the cytotoxicity of the novel 1,4-dihydropyridine derivative VE-3N [ethyl 6-chloro-5-formyl-2-methyl-4-(3-nitrophenyl)-1,4-dihydropyridine-3-carboxylate] towards cancer cells by employing hepatic carcinoma (HepG2) cells and isolated rat liver mitochondria. In HepG2 cells, VE-3N induced mitochondrial membrane potential dissipation, ATP depletion, annexin V/propidium iodide double labeling, and Hoechst staining; events indicating apoptosis induction. In isolated rat liver mitochondria, VE-3N promoted mitochondrial uncoupling by exerting protonophoric actions and by increasing membrane fluidity. Mitochondrial uncoupling was evidenced by an increase in resting respiration, dissipation of mitochondrial membrane potential, inhibition of Ca2+ uptake, stimulation of Ca2+ release, decrease in ATP synthesis, and swelling of valinomycin-treated organelles in hyposmotic potassium acetate media. Furthermore, uncoupling concentrations of VE-3N in the presence of Ca2+ plus ruthenium red induced the mitochondrial permeability transition process. These results indicate that mitochondrial uncoupling is potentially involved in the VE-3N cytotoxic actions towards HepG2 cells. Considering that hepatocellular carcinoma is the most common form of liver cancer, our findings may open a new avenue for the development of VE-3N-based cancer therapies, and help to unravel the cytotoxic mechanisms of 1,4-dihydropyridines towards cancer cells.


Systems Research and Behavioral Science | 2018

C-Phycocyanin and Phycocyanobilin as Remyelination Therapies for Enhancing Recovery in Multiple Sclerosis and Ischemic Stroke: A Preclinical Perspective

Giselle Pentón-Rol; Javier Marín-Prida; Viviana Falcón-Cama

Myelin loss has a crucial impact on behavior disabilities associated to Multiple Sclerosis (MS) and Ischemic Stroke (IS). Although several MS therapies are approved, none of them promote remyelination in patients, limiting their ability for chronic recovery. With no available therapeutic options, enhanced demyelination in stroke survivors is correlated with a poorer behavioral recovery. Here, we show the experimental findings of our group and others supporting the remyelinating effects of C-Phycocyanin (C-PC), the main biliprotein of Spirulina platensis and its linked tetrapyrrole Phycocyanobilin (PCB), in models of these illnesses. C-PC promoted white matter regeneration in rats and mice affected by experimental autoimmune encephalomyelitis. Electron microscopy analysis in cerebral cortex from ischemic rats revealed a potent remyelinating action of PCB treatment after stroke. Among others biological processes, we discussed the role of regulatory T cell induction, the control of oxidative stress and pro-inflammatory mediators, gene expression modulation and COX-2 inhibition as potential mechanisms involved in the C-PC and PCB effects on the recruitment, differentiation and maturation of oligodendrocyte precursor cells in demyelinated lesions. The assembled evidence supports the implementation of clinical trials to demonstrate the recovery effects of C-PC and PCB in these diseases.


European Journal of Pharmacology | 2018

Anti-cancer activity of a new dihydropyridine derivative, VdiE-2N, in head and neck squamous cell carcinoma

Renata Nishida Goto; Lays Martin Sobral; Lucas Oliveira Sousa; Cristiana B. Garcia; Norberto Peporine Lopes; Javier Marín-Prida; Estael Ochoa-Rodríguez; Yamila Verdecia-Reyes; Gilberto L. Pardo-Andreu; Carlos Curti; Andréia Machado Leopoldino

ABSTRACT This study aims to examine the effects of a new 1,4‐dihydropyridine derivative, VdiE‐2N, on cell signaling pathways and mitochondrial events in head and neck squamous cell carcinoma (HNSCC) cells, and on a mice model of xenograft tumor growth/cell proliferation. Four HNSCC cell lines (HN13, HN12, HN6, and CAL27), HEK293 cells (human embryonic kidney 293 cells), and human oral healthy mucosa fibroblasts (OHMF) were used for in vitro assessment of cell viability (resazurin assay) and invasion capacity (modified Boyden chamber assay), and mitochondrial membrane potential (JC‐1 fluorescence assay), morphology (transmission electron microscopy), and number of mitochondria (MitoTracker® imaging). SET and pDRP1 proteins were analyzed by immunofluorescence, and proteins involved in cell death/survival pathways were analyzed by Western blotting. HN12 xenograft tumors were established in the flank of Balb/c nude mice, and their characteristics and sensitivity to VdiE‐2N were determined by immunohistochemistry and histology. VdiE‐2N decreased cell viability in HNSCC cells (IC50 = 9.56 and 22.45 &mgr;M for HN13 and HN12 cells, respectively) more strongly than it decreased cell viability in OHMF and HEK293 cells (IC50 = 32.90 and > 50 &mgr;M, respectively). In HN13 cells, VdiE‐2N dissipated mitochondrial membrane potential and altered the mitochondria size, shape, and number in a concentration‐dependent manner, as well as it induced apoptosis and reduced their invasion capacity. Treatment of mice bearing xenograft tumors with VdiE‐2N significantly diminished proliferation of cancer cells. Therefore, VdiE‐2N induces HNSCC cell death in vitro through mitochondria‐mediated apoptotic pathways and dampens tumor growth in vivo, thus supporting a potential anti‐cancer effect. Graphical abstract Figure. No Caption available.


European Journal of Pharmacology | 2014

JM-20, a novel benzodiazepine–dihydropyridine hybrid molecule, protects mitochondria and prevents ischemic insult-mediated neural cell death in vitro.

Yanier Nuñez-Figueredo; Jeney Ramírez-Sánchez; René Delgado-Hernández; Marlene Porto-Verdecia; Estael Ochoa-Rodríguez; Yamila Verdecia-Reyes; Javier Marín-Prida; Michael González-Durruthy; Sérgio A. Uyemura; Fernando P. Rodrigues; Carlos Curti; Diogo O. Souza; Gilberto L. Pardo-Andreu

Collaboration


Dive into the Javier Marín-Prida's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carmen Valenzuela-Silva

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Carlos Curti

University of São Paulo

View shared research outputs
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