Bio


Originally from Spain, Irene L. Llorente joined the Neurosurgery Department at Stanford University in 2022. Following her undergraduate degree in Molecular Biology at the University of Leon in Spain, Dr. Llorente completed a MS in Molecular Biology and Biomedicine and a Ph.D. in Neuroscience between the Universities of Leon (Spain) and Florence (Italy). She conducted a postdoctoral fellowship in the Neurology Department at the David Geffen School of Medicine at UCLA where she also started her independent career as a Research Assistant Professor. Her research interests are largely directed toward understanding the biology of white matter repair in central nervous system (CNS) disorders, with a special emphasis on human glial biology. She is particularly interested in leveraging the current technologies emerging in the stem cell field to develop more efficient and effective stem cell-based therapies for stroke and vascular dementia patients. These stem cell-based therapies will also apply to other CNS disorders including spinal cord injury, multiple sclerosis, and traumatic brain injury in the future.

Academic Appointments


Boards, Advisory Committees, Professional Organizations


  • Member, Society for Neuroscience (2009 - Present)
  • Member, International Society for Stem Cell Research (2013 - Present)
  • Member, American Society for Neural Therapy and Repair (2018 - Present)
  • Member, American Stroke Association (2013 - Present)
  • Member, European Network for CNS Transplantation and Restoration (NECTAR) (2022 - Present)

Professional Education


  • PhD, University of Leon, Spain and University of Florence, Italy., Neuroscience (2013)
  • Ms, University of Leon, Spain, Molecular Biology and Biomedicine (2009)
  • BS, University of Leon, Spain, Molecular Biology (2009)

2023-24 Courses


Stanford Advisees


All Publications


  • Patient-derived glial enriched progenitors repair functional deficits due to white matter stroke and vascular dementia in rodents SCIENCE TRANSLATIONAL MEDICINE Llorente, I. L., Xie, Y., Mazzitelli, J. A., Hatanaka, E. A., Cinkornpumin, J., Miller, D. R., Lin, Y., Lowry, W. E., Carmichael, S. 2021; 13 (590)

    Abstract

    Subcortical white matter stroke (WMS) accounts for up to 30% of all stroke events. WMS damages primarily astrocytes, axons, oligodendrocytes, and myelin. We hypothesized that a therapeutic intervention targeting astrocytes would be ideally suited for brain repair after WMS. We characterize the cellular properties and in vivo tissue repair activity of glial enriched progenitor (GEP) cells differentiated from human-induced pluripotent stem cells, termed hiPSC-derived GEPs (hiPSC-GEPs). hiPSC-GEPs are derived from hiPSC-neural progenitor cells via an experimental manipulation of hypoxia inducible factor activity by brief treatment with a prolyl hydroxylase inhibitor, deferoxamine. This treatment permanently biases these cells to further differentiate toward an astrocyte fate. hiPSC-GEPs transplanted into the brain in the subacute period after WMS in mice migrated widely, matured into astrocytes with a prorepair phenotype, induced endogenous oligodendrocyte precursor proliferation and remyelination, and promoted axonal sprouting. hiPSC-GEPs enhanced motor and cognitive recovery compared to other hiPSC-differentiated cell types. This approach establishes an hiPSC-derived product with easy scale-up capabilities that might be effective for treating WMS.

    View details for DOI 10.1126/scitranslmed.aaz6747

    View details for Web of Science ID 000642350100002

    View details for PubMedID 33883275

  • Customized Brain Cells for Stroke Patients Using Pluripotent Stem Cells STROKE Kokaia, Z., Llorente, I. L., Carmichael, S. 2018; 49 (5): 1091-1098

    View details for DOI 10.1161/STROKEAHA.117.018291

    View details for Web of Science ID 000430662100020

    View details for PubMedID 29669871

    View details for PubMedCentralID PMC5916498

  • Hydrogel-delivered brain-derived neurotrophic factor promotes tissue repair and recovery after stroke JOURNAL OF CEREBRAL BLOOD FLOW AND METABOLISM Cook, D. J., Nguyen, C., Chun, H. N., Llorente, I. L., Chiu, A. S., Machnicki, M., Zarembinski, T. I., Carmichael, S. T. 2017; 37 (3): 1030-1045

    Abstract

    Stroke is the leading cause of adult disability. Systemic delivery of candidate neural repair therapies is limited by the blood-brain barrier and off-target effects. We tested a bioengineering approach for local depot release of BDNF from the infarct cavity for neural repair in chronic periods after stroke. The brain release levels of a hyaluronic acid hydrogel + BDNF were tested in several stroke models in mouse (strains C57Bl/6, DBA) and non-human primate ( Macaca fascicularis) and tracked with MRI. The behavioral recovery effects of hydrogel + BDNF and the effects on tissue repair outcomes were determined. Hydrogel-delivered BDNF diffuses from the stroke cavity into peri-infarct tissue over 3 weeks in two mouse stroke models, compared with 1 week for direct BDNF injection. Hydrogel delivery of BDNF promotes recovery of motor function. Mapping of motor system connections indicates that hydrogel-BDNF induces axonal sprouting within existing cortical and cortico-striatal systems. Pharmacogenetic studies show that hydrogel-BDNF induces the initial migration of immature neurons into the peri-infarct cortex and their long-term survival. In chronic stroke in the non-human primate, hydrogel-released BDNF can be detected up to 2 cm from the infarct, a distance relevant to human functional recovery in stroke. The hydrogel can be tracked by MRI in mouse and primate.

    View details for DOI 10.1177/0271678X16649964

    View details for Web of Science ID 000394660400024

    View details for PubMedID 27174996

  • Nogo receptor blockade overcomes remyelination failure after white matter stroke and stimulates functional recovery in aged mice PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES OF THE UNITED STATES OF AMERICA Sozmen, E. G., Rosenzweig, S., Llorente, I. L., DiTullio, D. J., Machnicki, M., Vinters, H. V., Havton, L. A., Giger, R. J., Hinman, J. D., Carmichael, S. T. 2016; 113 (52): E8453-E8462

    Abstract

    White matter stroke is a distinct stroke subtype, accounting for up to 25% of stroke and constituting the second leading cause of dementia. The biology of possible tissue repair after white matter stroke has not been determined. In a mouse stroke model, white matter ischemia causes focal damage and adjacent areas of axonal myelin disruption and gliosis. In these areas of only partial damage, local white matter progenitors respond to injury, as oligodendrocyte progenitors (OPCs) proliferate. However, OPCs fail to mature into oligodendrocytes (OLs) even in regions of demyelination with intact axons and instead divert into an astrocytic fate. Local axonal sprouting occurs, producing an increase in unmyelinated fibers in the corpus callosum. The OPC maturation block after white matter stroke is in part mediated via Nogo receptor 1 (NgR1) signaling. In both aged and young adult mice, stroke induces NgR1 ligands and down-regulates NgR1 inhibitors during the peak OPC maturation block. Nogo ligands are also induced adjacent to human white matter stroke in humans. A Nogo signaling blockade with an NgR1 antagonist administered after stroke reduces the OPC astrocytic transformation and improves poststroke oligodendrogenesis in mice. Notably, increased white matter repair in aged mice is translated into significant poststroke motor recovery, even when NgR1 blockade is provided during the chronic time points of injury. These data provide a perspective on the role of NgR1 ligand function in OPC fate in the context of a specific and common type of stroke and show that it is amenable to systemic intervention to promote recovery.

    View details for DOI 10.1073/pnas.1615322113

    View details for Web of Science ID 000391090800011

    View details for PubMedID 27956620

    View details for PubMedCentralID PMC5206535

  • Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain BIOMATERIALS Moshayedi, P., Nih, L. R., Llorente, I. L., Berg, A. R., Cinkornpumin, J., Lowry, W. E., Segura, T., Carmichael, S. T. 2016; 105: 145-155

    Abstract

    Stem cell therapies have shown promise in promoting recovery in stroke but have been limited by poor cell survival and differentiation. We have developed a hyaluronic acid (HA)-based self-polymerizing hydrogel that serves as a platform for adhesion of structural motifs and a depot release for growth factors to promote transplant stem cell survival and differentiation. We took an iterative approach in optimizing the complex combination of mechanical, biochemical and biological properties of an HA cell scaffold. First, we optimized stiffness for a minimal reaction of adjacent brain to the transplant. Next hydrogel crosslinkers sensitive to matrix metalloproteinases (MMP) were incorporated as they promoted vascularization. Finally, candidate adhesion motifs and growth factors were systemically changed in vitro using a design of experiment approach to optimize stem cell survival or proliferation. The optimized HA hydrogel, tested in vivo, promoted survival of encapsulated human neural progenitor cells (iPS-NPCs) after transplantation into the stroke core and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. This HA hydrogel can be tracked in vivo with MRI. A hydrogel can serve as a therapeutic adjunct in a stem cell therapy through selective control of stem cell survival and differentiation in vivo.

    View details for DOI 10.1016/j.biomaterials.2016.07.028

    View details for Web of Science ID 000383299800014

    View details for PubMedID 27521617

    View details for PubMedCentralID PMC5003628

  • Temporal Patterning of Neurofilament Light as a Blood-Based Biomarker for Stroke: A Meta-Analysis Sanchez, J., Martirosian, R., Chong, D., Tiedt, S., Uphaus, T., Groschel, K., Hinman, J. D. WILEY. 2021: S57-S58
  • Reliable generation of glial enriched progenitors from human fibroblast-derived iPSCs STEM CELL RESEARCH Llorente, I. L., Hatanaka, E. A., Meadow, M. E., Xie, Y., Lowry, W. E., Carmichael, S. 2021; 55: 102458

    Abstract

    White matter stroke (WMS) occurs as small infarcts in deep penetrating blood vessels in the brain and affects the regions of the brain that carry connections, termed the subcortical white matter. WMS progresses over years and has devastating clinical consequences. Unlike large grey matter strokes, WMS disrupts the axonal architecture of the brain and depletes astrocytes, oligodendrocyte lineage cells, axons and myelinating cells, resulting in abnormalities of gait and executive function. An astrocytic cell-based therapy is positioned as a strong therapeutic candidate after WMS. In this study we report, the reliable generation of a novel stem cell-based therapeutic product, glial enriched progenitors (GEPs) derived from human induced pluripotent stem cells (hiPSCs). By transient treatment of hiPSC derived neural progenitors (hiPSC-NPCs) with the small molecule deferoxamine, a prolyl hydroxylase inhibitor, for three days hiPSC-NPCs become permanently biased towards an astrocytic fate, producing hiPSC-GEPs. In preparation for clinical application, we have developed qualification assays to ensure identity, safety, purity, and viability of the cells prior to manufacture. Using tailored q-RT-PCR-based assays, we have demonstrated the lack of pluripotency in our final therapeutic candidate cells (hiPSC-GEPs) and we have identified the unique genetic profile of hiPSC-GEPs that is clearly distinct from the parent lines, hiPSCs and iPSC-NPCs. After completion of the viability assay, we have stablished the therapeutic window of use for hiPSC-GEPs in future clinical applications (7 h). Lastly, we were able to reliably and consistently produce a safe therapeutic final product negative for contamination by any human or murine viral pathogens, selected bacteria, common laboratory mycoplasmas, growth of any aerobes, anaerobes, yeast, or fungi and 100 times less endotoxin levels than the maximum acceptable value. This study demonstrates the reliable and safe generation of patient derived hiPSC-GEPs that are clinically ready as a cell-based therapeutic approach for WMS.

    View details for DOI 10.1016/j.scr.2021.102458

    View details for Web of Science ID 000709214200003

    View details for PubMedID 34274773

    View details for PubMedCentralID PMC8444576

  • Using organotypic hippocampal slice cultures to gain insight into mechanisms responsible for the neuroprotective effects of meloxicam: a role for gamma aminobutyric and endoplasmic reticulum stress NEURAL REGENERATION RESEARCH Landucci, E., Llorente, I. L., Anuncibay-Soto, B., Pellegrini-Giampietro, D. E., Fernandez-Lopez, A. 2019; 14 (1): 65-66

    View details for DOI 10.4103/1673-5374.243704

    View details for Web of Science ID 000451333500012

    View details for PubMedID 30531073

    View details for PubMedCentralID PMC6263004

  • Bicuculline Reverts the Neuroprotective Effects of Meloxicam in an Oxygen and Glucose Deprivation (OGD) Model of Organotypic Hippocampal Slice Cultures NEUROSCIENCE Landucci, E., Llorente, I. L., Anuncibay-Soto, B., Pellegrini-Giampietro, D. E., Fernandez-Lopez, A. 2018; 386: 68-78

    Abstract

    We previously demonstrated that the non-steroidal anti-inflammatory agent meloxicam has neuroprotective effects in an oxygen and glucose deprivation model (OGD) of rat organotypic hippocampal slice cultures. We wondered if GABAergic transmission changed the neuroprotective effects of meloxicam and if meloxicam was able to modulate endoplasmic reticulum stress (ER stress) in this model. Mortality was measured using propidium iodide. Western blot assays were performed to measure levels of cleaved and non-cleaved caspase-3 to quantify apoptosis, while levels of GRP78, GRP94 and phosphorylated eIF2α were used to detect unfolded protein response (UPR). Transcript levels of GRP78, GRP94 and GABAergic receptor α, β, and γ subunits were measured by real-time quantitative polymerase chain reaction (qPCR). In the present study, we show that the presence of meloxicam in a 30 min OGD assay, followed by 24 h of normoxic conditions, presented an antiapoptotic effect. The simultaneous presence of the GABAA receptor antagonist, bicuculline, in combination with meloxicam blocked the neuroprotective effect provided by the latter. However, in light of its effects on caspase 3 and PARP, bicuculline did not seem to promote the apoptotic pathway. Our results also showed that meloxicam modified the unfolded protein response (UPR), as well as the transcriptional response of different genes, including the GABAA receptor, alpha1, beta3 and gamma2 subunits. We concluded that meloxicam has a neuroprotective anti-apoptotic action, is able to enhance the UPR independently of the systemic anti-inflammatory response and its neuroprotective effect can be inhibited by blocking GABAA receptors.

    View details for DOI 10.1016/j.neuroscience.2018.06.024

    View details for Web of Science ID 000440325500006

    View details for PubMedID 29949743

  • Engineered HA hydrogel for stem cell transplantation in the brain: Biocompatibility data using a design of experiment approach DATA IN BRIEF Nih, L. R., Moshayedi, P., Llorente, I. L., Berg, A. R., Cinkornpumin, J., Lowry, W. E., Segura, T., Carmichael, S. 2017; 10: 202-209

    Abstract

    This article presents data related to the research article "Systematic optimization of an engineered hydrogel allows for selective control of human neural stem cell survival and differentiation after transplantation in the stroke brain" (P. Moshayedi, L.R. Nih, I.L. Llorente, A.R. Berg, J. Cinkornpumin, W.E. Lowry et al., 2016) [1] and focuses on the biocompatibility aspects of the hydrogel, including its stiffness and the inflammatory response of the transplanted organ. We have developed an injectable hyaluronic acid (HA)-based hydrogel for stem cell culture and transplantation, to promote brain tissue repair after stroke. This 3D biomaterial was engineered to bind bioactive signals such as adhesive motifs, as well as releasing growth factors while supporting cell growth and tissue infiltration. We used a Design of Experiment approach to create a complex matrix environment in vitro by keeping the hydrogel platform and cell type constant across conditions while systematically varying peptide motifs and growth factors. The optimized HA hydrogel promoted survival of encapsulated human induced pluripotent stem cell derived-neural progenitor cells (iPS-NPCs) after transplantation into the stroke cavity and differentially tuned transplanted cell fate through the promotion of glial, neuronal or immature/progenitor states. The highlights of this article include: (1) Data of cell and bioactive signals addition on the hydrogel mechanical properties and growth factor diffusion, (2) the use of a design of Experiment (DOE) approach (M.W. 2 Weible and T. Chan-Ling, 2007) [2] to select multi-factorial experimental conditions, and (3) Inflammatory response and cell survival after transplantation.

    View details for DOI 10.1016/j.dib.2016.11.069

    View details for Web of Science ID 000453159700032

    View details for PubMedID 27995155

    View details for PubMedCentralID PMC5154973

  • A Versatile Murine Model of Subcortical White Matter Stroke for the Study of Axonal Degeneration and White Matter Neurobiology JOVE-JOURNAL OF VISUALIZED EXPERIMENTS Nunez, S., Doroudchi, M., Gleichman, A. J., Ng, K. L., Llorente, I. L., Sozmen, E. G., Carmichael, S., Hinman, J. D. 2016

    Abstract

    Stroke affecting white matter accounts for up to 25% of clinical stroke presentations, occurs silently at rates that may be 5-10 fold greater, and contributes significantly to the development of vascular dementia. Few models of focal white matter stroke exist and this lack of appropriate models has hampered understanding of the neurobiologic mechanisms involved in injury response and repair after this type of stroke. The main limitation of other subcortical stroke models is that they do not focally restrict the infarct to the white matter or have primarily been validated in non-murine species. This limits the ability to apply the wide variety of murine research tools to study the neurobiology of white matter stroke. Here we present a methodology for the reliable production of a focal stroke in murine white matter using a local injection of an irreversible eNOS inhibitor. We also present several variations on the general protocol including two unique stereotactic variations, retrograde neuronal tracing, as well as fresh tissue labeling and dissection that greatly expand the potential applications of this technique. These variations allow for multiple approaches to analyze the neurobiologic effects of this common and understudied form of stroke.

    View details for DOI 10.3791/53404

    View details for Web of Science ID 000374636300011

    View details for PubMedID 27023377

    View details for PubMedCentralID PMC4829029

  • Hippocampus and cerebral cortex present a different autophagic response after oxygen and glucose deprivation in an ex vivo rat brain slice model NEUROPATHOLOGY AND APPLIED NEUROBIOLOGY Perez-Rodriguez, D., Anuncibay-Soto, B., Llorente, I. L., Perez-Garcia, C. C., Fernandez-Lopez, A. 2015; 41 (4): e68-e79

    Abstract

    To evaluate the neuroprotective role of autophagy in the cerebral cortex and hippocampus using an ex vivo animal model of stroke in brain slices.Brain slices were maintained for 30 min in oxygen and glucose deprivation (OGD) followed by 3 h in normoxic conditions to simulate the reperfusion that follows ischaemia in vivo (RL, reperfusion-like). Phagophore formation (Beclin 1 and LC3B) as well as autophagy flux (p62/SQSTM1, Atg5, Atg7 and polyubiquitin) markers were quantified by Western blot and/or qPCR. The release of lactate dehydrogenase (LDH) and glutamate in the medium was used as a measure of the mortality in the absence and in the presence of the autophagy inhibitor 3-methyladenine.Striking differences in the autophagy markers were observed between the hippocampus and cerebral cortex in normoxic conditions. OGD/RL induced increases both in the phagophore formation and in the autophagy flux in the first three hours in the cerebral cortex that were not observed in the hippocampus. The blocking of autophagy increased the OGD/RL-induced mortality, increased the glutamate release in both the cerebral cortex and hippocampus and abolished the OGD-induced decrease in the polyubiquitinated proteins in the cerebral cortex.We conclude that OGD induces a rapid autophagic response in the cerebral cortex that plays a neuroprotective role. Polyubiquitination levels and control of the glutamate release appear to be involved in the neuroprotective role of autophagy.

    View details for DOI 10.1111/nan.12152

    View details for Web of Science ID 000354465900001

    View details for PubMedID 24861158

  • GLUTAMATE RECEPTOR AND TRANSPORTER MODIFICATIONS IN RAT ORGANOTYPIC HIPPOCAMPAL SLICE CULTURES EXPOSED TO OXYGEN-GLUCOSE DEPRIVATION: THE CONTRIBUTION OF CYCLOOXYGENASE-2 NEUROSCIENCE Llorente, I. L., Landucci, E., Pellegrini-Giampietro, D. E., Fernandez-Lopez, A. 2015; 292: 118-128

    Abstract

    Meloxicam is a non-steroidal anti-inflammatory drug which has been reported to lessen the ischemic transcriptional effects in some of the glutamatergic system genes as well as to decrease the infarct volume in in vivo assays. In this study, we show how the presence of meloxicam decreases cell mortality in assays of oxygen-glucose deprivation (OGD) in rat organotypic hippocampal slices culture. Mortality was measured using propidium iodide. Transcript levels of some glutamatergic system genes, including vesicular and membrane glutamate transporters (VGLUT1, VGLUT2, GLAST-1A, GLT-1, and EAAC-1) and some glutamatergic receptor subunits (NMDA receptor, GluN1, GluN2A and GluN2B subunits and AMPA receptor, GluA1 and GluA2 subunits) were measured by real-time PCR (qPCR). The transcription of vesicular glutamate transporters and glutamatergic receptor subunits, but not membrane glutamate transporters, was modified by the presence of meloxicam. The study demonstrates the neuroprotective role of meloxicam in organotypic hippocampal slice cultures and shows how meloxicam is able to selectively increase or decrease the OGD-induced changes in the expression of the different glutamatergic system genes studied here. We suggest that the neuroprotective role of meloxicam could be due to a modification in the balance of the expression of some glutamatergic receptor subunits, leading to a different stoichiometry of receptors such as NMDA or AMPA. Thus, meloxicam would decrease the excitotoxicity induced by OGD.

    View details for DOI 10.1016/j.neuroscience.2015.02.040

    View details for Web of Science ID 000351664700011

    View details for PubMedID 25732138

  • Age-dependent modifications in vascular adhesion molecules and apoptosis after 48-h reperfusion in a rat global cerebral ischemia model AGE Anuncibay-Soto, B., Perez-Rodriguez, D., Llorente, I. L., Regueiro-Purrinos, M., Manuel Gonzalo-Orden, J., Fernandez-Lopez, A. 2014; 36 (5): 9703

    Abstract

    Stroke is one of the leading causes of death and permanent disability in the elderly. However, most of the experimental studies on stroke are based on young animals, and we hypothesised that age can substantially affect the stroke response. The two-vessel occlusion model of global ischemia by occluding the common carotid arteries for 15 min at 40 mmHg of blood pressure was carried out in 3- and 18-month-old male Sprague-Dawley rats. The adhesion molecules E- and P-selectin, cell adhesion molecules (CAMs), both intercellular (ICAM-1) and vascular (VCAM-1), as well as glial fibrillary acidic protein (GFAP), and cleaved caspase-3 were measured at 48 h after ischemia in the cerebral cortex and hippocampus using Western blot, qPCR and immunofluorescence techniques. Diametric expression of GFAP and a different morphological pattern of caspase-3 labelling, although no changes in the cell number, were observed in the neurons of young and old animals. Expression of E-selectin and CAMs was also modified in an age- and ischemia/reperfusion-dependent manner. The hippocampus and cerebral cortex had similar response patterns for most of the markers studied. Our data suggest that old and young animals present different time-courses of neuroinflammation and apoptosis after ischemic damage. On the other hand, these results suggest that neuroinflammation is dependent on age rather than on the different vulnerability described for the hippocampus and cerebral cortex. These differences should be taken into account in searching for therapeutic targets.

    View details for DOI 10.1007/s11357-014-9703-7

    View details for Web of Science ID 000343801300005

    View details for PubMedID 25182537

    View details for PubMedCentralID PMC4453934

  • Unfolded protein response to global ischemia following 48 h or reperfusion in the rat brain: the effect of age and meloxicam JOURNAL OF NEUROCHEMISTRY Llorente, I. L., Burgin, T. C., Perez-Rodriguez, D., Martinez-Villayandre, B., Perez-Garcia, C. C., Fernandez-Lopez, A. 2013; 127 (5): 701-710

    Abstract

    The unfolded protein response (UPR) in the hippocampal regions Cornu Ammonis 1 hippocampal region, Cornu Ammonis 3 hippocampal region, and dentate gyrus, as well as in the cerebral cortex of 3-month-old and 18-month-old rats were studied in a model of 15 min of global cerebral ischemia followed by 48 h of reperfusion. UPR was measured by quantifying the protein disulfide isomerase (PDI), C/EBP-homologous protein (CHOP), GRP78 and GRP94 transcripts using qPCR and the amounts of PDI and GRP78 by western blot. The study shows how the mRNA levels of these genes were similar in 3-month-old and 18-month-old sham-operated animals, but the ischemic insult elicited a noticeable increase in the expression of these genes in young animals that was scarcely appreciable in older animals. The striking increase in the mRNA levels of these genes in 3-month-old animals was abolished or even reverted by treatment with meloxicam, an anti-inflammatory agent. Western blot assays showed that the UPR was still detectable 48 h after ischemia in some of the studied areas, and provided evidence that the UPR is different between young and older animals. Western blot assays carried out in young animals also showed that meloxicam elicited different effects on the levels of PDI and GRP78 in the cerebral cortex and the hippocampus. We conclude that the UPR response to ischemic/reperfusion insult is age- and probably inflammation-dependent and could play an important role in ischemic vulnerability. The UPR appears to be strongly decreased in aged animals, suggesting a reduced ability for cell survival. In this study, we conclude that the unfolded protein response (UPR) to ischemic/reperfusion insult is age- and probably inflammation-dependent and could play an important role in ischemic vulnerability. The UPR strongly decreased in aged rats, suggesting a reduced ability for cell survival. The increase in the mRNA levels of UPR gene transcripts in 3-month-old animals was abolished or even reverted by treatment with meloxicam, an anti-inflammatory agent.

    View details for DOI 10.1111/jnc.12337

    View details for Web of Science ID 000326834600015

    View details for PubMedID 23763503

  • GABA(A) receptor chloride channels are involved in the neuroprotective role of GABA following oxygen and glucose deprivation in the rat cerebral cortex but not in the hippocampus BRAIN RESEARCH Llorente, I. L., Perez-Rodriguez, D., Martinez-Villayandre, B., Dos-Anjos, S., Darlison, M. G., Poole, A. V., Fernandez-Lopez, A. 2013; 1533: 141-151

    Abstract

    Assays on "ex vivo" sections of rat hippocampus and rat cerebral cortex, subjected to oxygen and glucose deprivation (OGD) and a three-hour reperfusion-like (RL) recovery, were performed in the presence of either GABA or the GABA(A) receptor binding site antagonist, bicuculline. Lactate dehydrogenase (LDH) and propidium iodide were used to quantify cell mortality. We also measured, using real-time quantitative polymerase chain reaction (qPCR), the early transcriptional response of a number of genes of the glutamatergic and GABAergic systems. Specifically, glial pre- and post-synaptic glutamatergic transporters (namely GLAST1a, EAAC-1, GLT-1 and VGLUT1), three GABAA receptor subunits (α1, β2 and γ2), and the GABAergic presynaptic marker, glutamic acid decarboxylase (GAD65), were studied. Mortality assays revealed that GABAA receptor chloride channels play an important role in the neuroprotective effect of GABA in the cerebral cortex, but have a much smaller effect in the hippocampus. We also found that GABA reverses the OGD-dependent decrease in GABA(A) receptor transcript levels, as well as mRNA levels of the membrane and vesicular glutamate transporter genes. Based on the markers used, we conclude that OGD results in differential responses in the GABAergic presynaptic and postsynaptic systems.

    View details for DOI 10.1016/j.brainres.2013.08.024

    View details for Web of Science ID 000325594900015

    View details for PubMedID 23969196

  • Age and meloxicam modify the response of the glutamate vesicular transporters (VGLUTs) after transient global cerebral ischemia in the rat brain BRAIN RESEARCH BULLETIN Llorente, I. L., Perez-Rodriguez, D., Burgin, T. C., Gonzalo-Orden, J. M., Martinez-Villayandre, B., Fernandez-Lopez, A. 2013; 94: 90-97

    Abstract

    This study analyzes how age and inflammation modify the response of the vesicular glutamate transporters (VGLUTs), VGLUT1-3 to global brain ischemia/reperfusion (I/R) in brain areas with different I/R vulnerabilities.Global ischemia was induced in 3- and 18-month-old male Sprague-Dawley rats and CA1 and CA3 hippocampal areas, dentate gyrus and cerebral cortex of sham-operated and I/R animals were removed 48 h after insult. Real-time PCR analysis revealed that I/R challenge resulted in a significant decrease of the VGLUT mRNA levels in young animals. Western blot assays showed a lessened age-dependent response to the ischemic damage in VGLUT1 and VGLUT3, while VGLUT2 presented an age and structure-dependent response to challenge. The use of the anti-inflammatory agent meloxicam following challenge showed that COX2 inhibition promotes the expression of VGLUTs in both sham and injured animals, which results in a lessened response to I/R injury.VGLUT1 and VGLUT3 presented an age-dependent response to ischemic damage, while this VGLUT response was age both and structure-dependent. In addition, COX-2 inhibition resulted in an increase of VGLUT1 and VGLUT2 protein amounts both in sham and injured animals together with a lessening of the transporters' response to ischemia.

    View details for DOI 10.1016/j.brainresbull.2013.02.006

    View details for Web of Science ID 000318754200012

    View details for PubMedID 23458738

  • Differential effect of transient global ischaemia on the levels of gamma-aminobutyric acid type A (GABAA) receptor subunit mRNAs in young and older rats NEUROPATHOLOGY AND APPLIED NEUROBIOLOGY Montori, S., Dos Anjos, S., Poole, A., Regueiro-Purrinos, M. M., Llorente, I. L., Darlison, M. G., Fernandez-Lopez, A., Martinez-Villayandre, B. 2012; 38 (7): 710-722

    Abstract

    This study has investigated how global brain ischaemia/reperfusion (I/R) modifies levels of mRNAs encoding γ-aminobutyric acid type A (GABA(A)) receptor α1, β2 and γ2 subunits and glutamic acid decarboxylase 65 (GAD65) in an age- and structure-dependent manner. Gene expression in response to treatment with the anti-inflammatory agent meloxicam was also investigated.Global ischaemia was induced in 3- and 18-month-old male Sprague-Dawley rats. CA1, CA3, and dentate gyrus (DG) hippocampal areas, cerebral cortex (CC) and caudate putamen (C-Pu) from sham-operated and I/R-injured animals were excised 48 h after the insult and prepared for quantitative polymerase chain reaction assays. Following I/R, meloxicam treatment was also carried out on young animals.Data revealed significant decreases in the levels of all GABA(A) receptor subunit transcripts in the hippocampus of both young and older injured animals compared with sham-operated ones. In contrast, there was either an increase or no change in GAD65 mRNA levels. GABA(A) receptor subunit transcript decreases were also observed in the CC and C-Pu in young injured animals but not in the CC of the older injured ones; interestingly, significant increases were observed in the C-Pu of older injured animals compared with controls. Meloxicam treatment following the insult resulted in a diminution of the previously described I/R response.The data indicate that I/R results in the modification of the levels of several gene transcripts involved in GABAergic signalling in both the pre- and postsynaptic components, of this neurotransmitter system, in an age- and structure-dependent manner.

    View details for DOI 10.1111/j.1365-2990.2012.01254.x

    View details for Web of Science ID 000311075000006

    View details for PubMedID 22289121

  • Age-dependent modifications in the mRNA levels of the rat excitatory amino acid transporters (EAATs) at 48 hour reperfusion following global ischemia BRAIN RESEARCH Montori, S., Martinez-Villayandre, B., Dos-Anjos, S., Llorente, I. L., Burgin, T. C., Fernandez-Lopez, A. 2010; 1358: 11-19

    Abstract

    This study reports the mRNA levels of some excitatory amino acid transporters (EAATs) in response to ischemia-reperfusion (I/R) in rat hippocampus and cerebral cortex. The study was performed in 3-month-old and 18-month-old animals to analyze the possible role of age in the I/R response of these transporters. The I/R resulted in a reduced transcription of both the neuronal EAAC1 (excitatory amino acid carrier-1) and the neuronal and glial GLT-1 (glial glutamate transporter 1), while the glial GLAST1a (l-glutamate/l-aspartate transporter 1a) transcription increased following I/R. The changes observed were more striking in 3-month-old animals than in 18-month-old animals. We hypothesize that increases in the GLAST1a mRNA levels following I/R insult can be explained by increases in glial cells, while the GLT-1 response to I/R mirrors neuronal changes. GLAST1a transcription increases in 3-month-old animals support the hypothesis that this transporter would be the main mechanism for extracellular glutamate clearance after I/R. Decreases in EAAC1 and GLT-1 mRNA levels would represent either neuronal changes due to the delayed neuronal death or a putative protective down-regulation of these transporters to decrease the amount of glutamate inside the neurons, which would decrease their glutamate release. This study also reports how the treatment with the anti-inflammatory agent meloxicam attenuates the transcriptional response to I/R in 3-month-old rats and decreases the survival of the I/R-injured animals.

    View details for DOI 10.1016/j.brainres.2010.08.020

    View details for Web of Science ID 000283814800002

    View details for PubMedID 20709031