Network


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

Hotspot


Dive into the research topics where Rafael Alonso is active.

Publication


Featured researches published by Rafael Alonso.


The Journal of Comparative Neurology | 2007

Distribution patterns of estrogen receptor α and β in the human cortex and hippocampus during development and adulthood

Miriam González; Alfredo Cabrera-Socorro; Carlos Gustavo Pérez-García; James D. Fraser; Francisco J. López; Rafael Alonso; Gundela Meyer

The expression of estrogen receptors (ERs) in the developing and adult human brain has not been clearly established, although estrogens are crucial for neuronal differentiation, synapse formation, and cognitive functions. By using immunohistochemistry, we have studied the distribution of ERα and ERβ in human cerebral cortex and hippocampus from early prenatal stages to adult life. ERα was detected in the cortex at 9 gestational weeks (GW), with a high expression in proliferating zones and the cortical plate. The staining intensity decreased gradually during prenatal development but increased again from birth to adulthood. In contrast, ERβ was first detected at 15 GW in proliferating zones, and at 16/17 GW, numerous ERβ immunopositive cells were also observed in the cortical plate. ERβ expression persisted in the adult cortex, being widely distributed throughout cortical layers II–VI. In addition, from around 15 GW to adulthood, ERα and ERβ were expressed in human hippocampus mainly in pyramidal cells of Ammons horn and in the dentate gyrus. Western blotting and immunohistochemistry in the adult cerebral cortex and hippocampus revealed lower protein expression of ERα compared with ERβ. Double immunostaining showed that during fetal life both ERs are expressed in neurons as well as in radial glia, although only ERα is expressed in the Cajal‐Retzius neurons of the marginal zone. These observations demonstrate that the expression of ERα and ERβ displays different spatial‐temporal patterns during human cortical and hippocampal development and suggest that both ERs may play distinct roles in several processes related to prenatal brain development. J. Comp. Neurol. J. Comp. Neurol. 503:790–802, 2007.


Journal of Neurochemistry | 2004

Plasma membrane oestrogen receptor mediates neuroprotection against β‐amyloid toxicity through activation of Raf‐1/MEK/ERK cascade in septal‐derived cholinergic SN56 cells

Borja Guerra; Mario Díaz; Rafael Alonso; Raquel Marin

Rapid oestrogen neuroprotection against β‐amyloid peptide (Aβ)‐induced toxicity, a main feature of Alzheimers disease, may be partially initiated at the plasma membrane. However, the mechanism by which this oestrogen effect occurs is unknown. In a septal murine cell line (SN56), we observed that short exposures to either 17β‐oestradiol (E2) or membrane impermeant E2 bound to horseradish peroxidase (E‐HRP) induced a biphasic stimulation of extracellular‐signal regulated protein kinase (ERK1/2) phosphorylation, with peak inductions detected around 4–8 min in the early phase and a second maximum around 8 h after treatment. ERK1/2 phosphorylation was abolished by ERK1/2 kinase (MEK) inhibitors PD98059 and U0126. Interestingly, PD98059 was also shown to block rapid E2‐related prevention of death in cells exposed to Aβ fragment 1–40 (Aβ1−40) for 24 h. In contrast, no neuroprotective effects were obtained when MEK inhibitor was used to selectively abolish the late phosphorylation phase. Furthermore, both ERK1/2 activation and E2‐associated protection were blocked by an inhibitor of Raf‐1 kinase. Raf‐1 may be involved in these effects because oestrogen caused the rapid serine 338 (Ser338) phosphorylation of this protein. In addition, the oestrogen receptor (ER) antagonist ICI 182 780 was also observed to block ERK1/2 phosphorylation. We propose a novel mechanism in SN56 cells by which rapid effects of oestrogen leading to neuroprotection are signalled through Raf‐1/MEK/ERK1/2 pathway, possibly by activation of a membrane‐related ER.


Current Neurovascular Research | 2005

Estrogen Activates Classical and Alternative Mechanisms to Orchestrate Neuroprotection

Raquel Marin; Borja Guerra; Rafael Alonso; Cristina M. Ramírez; Mario Díaz

Evidence for a protective role of estradiol in neurodegenerative diseases has steadily increased over the past decade, though the mechanisms of action and the participation of true estrogen receptors (ERs) have proven a complex score. The protective effects of estrogens take place partly through pathways involving canonical ER activation, which is constitutively expressed in many brain regions and is able to initiate gene transcription after specifically binding to estradiol. In addition, non-genomic (or alternative) signalling pathways, involving extranuclear ERs, respond to physiological concentration of estrogens to elicit neuroprotection. Often, rapid activation of intracellular signallers such as mitogen-activated protein kinase (MAPK) or phosphatidylinositol 3-kinase (PI3K) underlie alternative estrogen-induced neuroprotection upon activation of specific binding sites at the plasma membrane. Although the molecular characteristics of these unconventional ERs are still largely unknown, the generally held view maintains that plasma membrane ER (mER) originates from, or is related to, classical nuclear ERs. The present article will review some of the most recent evidence revealing the relevance of alternative mechanisms in estrogen-dependent neuroprotection. Special emphasis will be paid to cellular models of amyloid-beta toxicity where classical and alternative pathways activated by estrogens seem to coexist to orchestrate neuroprotection.


Molecular and Cellular Neuroscience | 2009

VDAC and ERα interaction in caveolae from human cortex is altered in Alzheimer's disease

Cristina M. Ramírez; Miriam González; Mario Díaz; Rafael Alonso; Isidre Ferrer; Gabriel Santpere; B. Puig; Gundela Meyer; Raquel Marin

Voltage-dependent anion channel (VDAC) is a mitochondrial porin also found in the neuronal membrane (pl-VDAC), where its function may be related to redox homeostasis and apoptosis. Murine models have evidenced pl-VDAC into caveolae in a complex with estrogen receptor alpha (mERalpha), which participates in neuroprotection against amyloid beta (Abeta), and whose integration into this hydrophobic domain remains unclear. Here, we have demonstrated in caveolae of human cortex and hippocampus the presence of pl-VDAC and mERalpha, in a complex with scaffolding caveolin-1 which likely provides mERalpha stability at the plasma membrane. In Alzheimers disease (AD) brains, VDAC was accumulated in caveolae, and it was observed in dystrophic neurites of senile plaques, whereas ERalpha was expressed in astrocytes surrounding the plaques. Together with previous data in murine neurons demonstrating the participation of pl-VDAC in Abeta-induced neurotoxicity, these data suggest that the channel may be involved in membrane dysfunctioning observed in AD neuropathology.


Molecular Membrane Biology | 2007

Voltage-dependent anion channel (VDAC) participates in amyloid beta-induced toxicity and interacts with plasma membrane estrogen receptor α in septal and hippocampal neurons

Raquel Marin; Cristina M. Ramírez; Miriam González; Elena González-Muñoz; Antonio Zorzano; Marta Camps; Rafael Alonso; Mario Díaz

Voltage-dependent anion channel (VDAC) is a porin known by its role in metabolite transport across mitochondria and participation in apoptotic processes. Although traditionally accepted to be located within mitochondrial outer membrane, some data has also reported its presence at the plasma membrane level where it seems to participate in regulation of normal redox homeostasis and apoptosis. Here, exposure of septal SN56 and hippocampal HT22 cells to specific anti-VDAC antibodies prior to amyloid beta (Aβ) peptide was observed to prevent neurotoxicity. In these cell lines, we identified a VDAC form associated with the plasma membrane that seems to be particularly abundant in caveolae. The two membrane-related isoforms of estrogen receptor α (mERα) (80 and 67 kDa), known in SN56 cells to participate in estrogen-induced neuroprotection against Aβ injury, were also observed to be present in caveolae. Interestingly, we demonstrated for the first time that both VDAC and mERα interact at the plasma membrane of these neurons as well as in microsomal fractions of the corresponding murine septal and hippocampal tissues. These proteins were also shown to associate with caveolin-1, thereby corroborating their presence in caveolar microdomains. Taken together, these results suggest that VDAC-mERα association at the plasma membrane level may participate in the modulation of Aβ-induced cell death.


Neuroscience | 2003

Estradiol prevents amyloid-β peptide-induced cell death in a cholinergic cell line via modulation of a classical estrogen receptor

Raquel Marin; B Guerra; J.G Hernández-Jiménez; X.-L Kang; J.D Fraser; F.J López; Rafael Alonso

The pathology of Alzheimers disease includes amyloid-beta peptide aggregation that contributes to degeneration of cholinergic neurons. Even though the underlying molecular mechanisms remain unclear, recent in vitro evidence supports a protective role for estrogens against several neurotoxic agents. Here we report that, in a murine cholinergic cell line (SN56), the massive cell death induced by 1-40 fragment of amyloid-beta peptide was prevented by 17beta-estradiol through a mechanism that may involve estrogen receptor activation. The protective effect of estradiol was observed in a dose-dependent manner, and was completely blocked by the pure antiestrogen ICI 182,780. In contrast, the inactive isomer 17alpha-estradiol consistently showed weaker neuroprotection than the native hormone that was unaffected by ICI 182,780 treatment. In addition, equivalent concentrations of 17beta-estradiol enhanced luciferase activity in cells transfected with a luciferase reporter gene driven by tandem estrogen response elements. Estrogen-induced luciferase activity was blocked by ICI 182,780, indicating estrogen receptor-dependent transcriptional activity. We also observed by reverse transcription-polymerase chain reaction, Western blot and immunocytochemistry that increasing concentrations of 17beta-estradiol enhanced the expression of estrogen receptor alpha mRNA and protein during amyloid-beta-induced toxicity. Under these conditions, it was found by confocal microscopy that the localization of estrogen receptor alpha in the absence of hormone was mainly extranuclear. However, the receptor was consistently observed also at the nuclear region after estrogen exposure. Overall, these data suggest that estrogen may exert neuroprotective effects against amyloid-beta-induced toxicity by activation of estrogen receptor-mediated pathways. In addition, intracellular estrogen receptors are up-regulated by their cognate hormone even during exposure to neurotoxic agents.


The Journal of Steroid Biochemistry and Molecular Biology | 2009

Role of estrogen receptor α in membrane-initiated signaling in neural cells: Interaction with IGF-1 receptor

Raquel Marin; Mario Díaz; Rafael Alonso; Amaya Sanz; María Ángeles Arévalo; Luis Miguel Garcia-Segura

The mechanisms of action of estradiol in the nervous system involve nuclear-initiated steroid signaling and membrane-initiated steroid signaling. Estrogen receptors (ERs) are involved in both mechanisms. ERalpha interacts with the signaling of IGF-1 receptor in neural cells: ERalpha transcriptional activity is regulated by IGF-1 receptor signaling and estradiol regulates IGF-1 receptor signaling. The interaction between ERalpha and the IGF-1 receptor in the brain may occur at the plasma membrane of neurons and glial cells. Caveolin-1 may provide the scaffolding for the interaction of different membrane-associated molecules, including voltage-dependent anion channel, ERalpha and IGF-I receptor.


Journal of Neurochemistry | 2003

An oestrogen membrane receptor participates in estradiol actions for the prevention of amyloid-β peptide1-40-induced toxicity in septal-derived cholinergic SN56 cells

Raquel Marin; B Guerra; Araceli Morales; Mario Díaz; Rafael Alonso

Although oestrogen [17β‐estradiol (E2)]‐related neuroprotection has been demonstrated in different models, the involvement of non‐classical oestrogen receptors (ERs) remains unexplored. Using the SN56 cholinergic cell line, we present evidence indicating that an ER associated with the plasma membrane participates in oestrogen‐dependent inhibition of cell death induced by amyloid‐β peptide (Aβ) toxicity. Similarly to E2 alone, a 15‐min exposure to estradiol‐horseradish peroxidase (E‐HRP) significantly reduced Aβ‐induced cell death. This effect was decreased by the ER antagonist ICI 182,780 as well as by MC‐20 antibody directed to a region neighbouring the ligand‐binding domain of ERα. Using confocal microscopy on unpermeabilized SN56 cells exposed to MC‐20 antibody, we identified a protein at the plasma membrane level. Western blot analysis of purified SN56 cell membrane fractions using MC‐20 antibody revealed the presence of one band with the same electrophoretic mobility as intracellular ERα. Using conjugated forms of the steroid, E‐HRP and E2 conjugated to bovine serum albumin‐FITC, we demonstrated by confocal microscopy that SN56 cells contain surface binding sites for E2. Binding of both conjugates was blocked by pre‐incubation with E2 and decreased by either ICI 182,780 or MC‐20 antibody in a concentration‐dependent manner. Thus, a membrane‐related ER that shares some structural homologies with ERα may participate in oestrogen‐mediated neuroprotection.


European Journal of Neuroscience | 2003

Estradiol modulates acetylcholine-induced Ca2+ signals in LHRH-releasing GT1-7 cells through a membrane binding site.

Araceli Morales; Mario Díaz; Ana B. Ropero; Angel Nadal; Rafael Alonso

Estrogen regulation of the female reproductive axis involves the rapid inhibition (< 30 min) of luteinizing hormone‐releasing hormone (LHRH) secretion from hypothalamic neurons. This fast time‐course suggests interactions with potential plasma membrane binding sites that could result in short‐term effects on LHRH neurons. Because LHRH release is calcium dependent, we have studied the acute effects of 17β‐estradiol (E2) and estradiol‐peroxidase (E‐HRP) on the elevations of intracellular calcium ([Ca2+]i) induced by acetylcholine (ACh) in LHRH‐producing GT1‐7 cells. Exposure to ACh (1–100 µm) induced transient increases of [Ca2+]i, whereas pretreatment with E2 or E‐HRP (10 nm) for 2 min reduced this response by 50–60%. The effect was specific for E2 as neither 17α‐estradiol (1 µm) nor the synthetic antiestrogens ICI182 780 (1 µm) or tamoxifen (1 µm) elicited any change on the ACh‐induced Ca2+ signal. Both the latency of the effect and the response to the membrane impermeant conjugate suggested a membrane‐mediated mechanism. Such membrane binding sites for E2 in GT1‐7 cells were demonstrated by visualizing the binding of E‐HRP and estradiol‐BSA‐fluorescein isothiocyanate (E‐BSA‐FITC) conjugates. Competition studies showed that E‐HRP binding was blocked by preincubation with E2, but not with 17α‐E2, ICI182 780, tamoxifen or progesterone, indicating that the plasma membrane binding site is highly specific for E2 and exhibits a pharmacological profile different from classical estrogen receptors. We conclude that ACh‐induced increase in [Ca2+]i in GT1‐7 cells is modulated acutely by physiological E2 concentrations in a manner which is compatible with the existence of an estrogen‐specific membrane binding site.


Journal of Neurochemistry | 2011

Voltage-dependent anion channel as a resident protein of lipid rafts: post-transductional regulation by estrogens and involvement in neuronal preservation against Alzheimer's disease.

Jose Luis Herrera; Mario Díaz; Juan Ramon Hernandez-Fernaud; Eduardo Salido; Rafael Alonso; Cecilia Fernández; Araceli Morales; Raquel Marin

J. Neurochem. (2011) 116, 820–827.

Collaboration


Dive into the Rafael Alonso's collaboration.

Top Co-Authors

Avatar

Raquel Marin

University of La Laguna

View shared research outputs
Top Co-Authors

Avatar

Mario Díaz

University of La Laguna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pedro Abreu

University of La Laguna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

B Guerra

University of La Laguna

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

C. Santana

University of La Laguna

View shared research outputs
Researchain Logo
Decentralizing Knowledge