Network


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

Hotspot


Dive into the research topics where Sandra Zárate is active.

Publication


Featured researches published by Sandra Zárate.


American Journal of Physiology-endocrinology and Metabolism | 2009

Estrogens exert a rapid apoptotic action in anterior pituitary cells

Sandra Zárate; Gabriela Jaita; Verónica Zaldivar; Daniela Radl; Guadalupe Eijo; Jimena Ferraris; Daniel Pisera; Adriana Seilicovich

It is now accepted that estrogens not only stimulate lactotrope proliferation but also sensitize anterior pituitary cells to proapoptotic stimuli. In addition to their classical mechanism of action through binding to intracellular estrogen receptors (ERs), there is increasing evidence that estrogens exert rapid actions mediated by cell membrane-localized ERs (mERs). In the present study, we examined the involvement of membrane-initiated steroid signaling in the proapoptotic action of estradiol in primary cultures of anterior pituitary cells from ovariectomized rats by using estren, a synthetic estrogen with no effect on classical transcription and a cell-impermeable 17beta-estradiol conjugate (E2-BSA). Both compounds induced cell death of anterior pituitary cells after 60 min of incubation as assessed by flow cytometry and the [3-(4,5-dimethylthiazol-2-yl)]-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium assay. Estren, E2, and E2-BSA induced apoptosis of lactotropes and somatotropes as evaluated by the deoxynucleotidyltransferase-mediated dUTP nick end-labeling assay and immunodetection of prolactin (PRL) and growth hormone (GH). The proapoptotic effect of E2-BSA was abrogated by ICI-182,780, an antagonist of ERs. The expression of membrane-associated ERalpha was observed in PRL- and GH-bearing cells. Our results indicate that estradiol is able to exert a rapid apoptotic action in anterior pituitary cells, especially lactotropes and somatotropes, by a mechanism triggered by mERs. This mechanism could be involved in anterior pituitary cell turnover.


Neuroendocrinology | 2009

Estradiol Increases the Bax/Bcl-2 Ratio and Induces Apoptosis in the Anterior Pituitary Gland

Verónica Zaldivar; María L. Magri; Sandra Zárate; Gabriela Jaita; Guadalupe Eijo; Daniela Radl; Jimena Ferraris; Daniel Pisera; Adriana Seilicovich

Background: Estrogens are recognized as acting as modulators of pituitary cell renewal, sensitizing cells to mitogenic and apoptotic signals, thus participating in anterior pituitary homeostasis during the estrous cycle. The balance of pro- and antiapoptotic proteins of the Bcl-2 family is known to regulate cell survival and apoptosis. Aims: In order to understand the mechanisms underlying apoptosis during the estrous cycle, we evaluated the expression of the proapoptotic protein Bax and the antiapoptotic proteins Bcl-2 and Bcl-xL in the anterior pituitary gland in cycling female rats as well as the influence of estradiol on the expression of these proteins in anterior pituitary cells of ovariectomized rats. Methods/Results: As determined by Western blot, the expression of Bax was higher in anterior pituitary glands from rats at proestrus than at diestrus I, Bcl-2 protein levels showed no difference and Bcl-xL expression was lower, thus increasing the Bax/Bcl-2 ratio at proestrus. Assessed by annexin V binding and flow cytometry, the percentage of apoptotic anterior pituitary cells was higher in rats at proestrus than at diestrus I. Chronic estrogen treatment in ovariectomized rats enhanced the Bax/Bcl-2 ratio and induced apoptosis. Moreover, incubation of cultured anterior pituitary cells from ovariectomized rats with 17β-estradiol for 24 h increased the Bax/Bcl-2 ratio, decreased Bcl-xL expression and induced apoptosis. Conclusion: Our results demonstrate that estradiol increases the ratio between proapoptotic and antiapoptotic proteins of the Bcl-2 family. This effect could participate in the sensitizing action of estrogens to proapoptotic stimuli and therefore be involved in the high apoptotic rate observed at proestrus in the anterior pituitary gland.


Neuroendocrinology | 2004

Tumor Necrosis Factor-Alpha-Induced Nitric Oxide Restrains the Apoptotic Response of Anterior Pituitary Cells

Marianela Candolfi; Gabriela Jaita; Verónica Zaldivar; Sandra Zárate; Daniel Pisera; Adriana Seilicovich

We previously reported that tumor necrosis factor-α (TNF-α) inhibits cell proliferation whereas it stimulates apoptosis of anterior pituitary cells in an estrogen-dependent manner. Also, we showed that nitric oxide (NO) mediates the inhibitory effect of TNF-α on prolactin release. Here, we studied the effect of TNF-α on nitric oxide synthase (NOS) activity and expression in anterior pituitary cells from cycling and ovariectomized (OVX) rats, and the role of NO in TNF-α induced apoptosis of anterior pituitary cells. NOS activity was higher in anterior pituitary cells from rats in proestrus than in diestrus and was stimulated by 17β-estradiol (10–9 M, E2). TNF-α (50 ng/ml) stimulated NOS activity in anterior pituitary cells from rats at both stages of the estrous cycle and in cells from OVX rats cultured either with or without E2. Inducible NOS (iNOS) gene expression was higher in anterior pituitary cells from rats in proestrus than in diestrus and its expression was enhanced by TNF-α. Acute administration of E2 to OVX rats increased endothelial NOS (eNOS) expression in the anterior pituitary gland. Also, E2 increased eNOS mRNA in dispersed anterior pituitary cells from OVX rats, and this effect was blocked by TNF-α. nNOS expression in the anterior pituitary gland was higer at proestrus than at diestrus but eNOS expression was similar at both stages. TNF-α decreased eNOS mRNA in anterior pituitary cells from rats at proestrus or diestrus. In anterior pituitary cells from OVX rats, TNF-α failed to induce apoptosis but was able to induce it when cells were incubated with NAME or NMMA, NOS inhibitors that did not affect cell viability per se. In the presence of E2, NAME induced apoptosis and enhanced the proapoptotic effect of TNF-α. In conclusion, our study shows that TNF-α upregulates iNOS gene expression whereas it downregulates estrogen-induced eNOS expression in anterior pituitary cells. Endogenous NO may restrain rather than mediate the proapoptotic effect of TNF-α in anterior pituitary cells.


Neuroendocrinology | 2008

Apoptosis of Lactotrophs Induced by D2 Receptor Activation Is Estrogen Dependent

Daniela Radl; Sandra Zárate; Gabriela Jaita; Jimena Ferraris; Verónica Zaldivar; Guadalupe Eijo; Adriana Seilicovich; Daniel Pisera

Background/Aims: Dopamine (DA) inhibits prolactin release and reduces lactotroph proliferation by activating D2 receptors. DA and its metabolite, 6-hydroxydopamine (6-OHDA), induce apoptosis in different cell types. DA receptors and DA transporter (DAT) were implicated in this action. Considering that estradiol sensitizes anterior pituitary cells to proapoptotic stimuli, we investigated the effect of estradiol on the apoptotic action of DA and 6-OHDA in anterior pituitary cells, and the involvement of the D2 receptor and DAT in the proapoptotic effect of DA. Methods: Viability of cultured anterior pituitary cells from ovariectomized rats was determined by MTS assay. Apoptosis was evaluated by Annexin-V/flow cytometry and TUNEL. Lactotrophs were identified by immunocytochemistry. Results: DA induced apoptosis of lactotrophs in an estrogen-dependent manner. In contrast, estradiol was not required to trigger the apoptotic action of 6-OHDA. Cabergoline, a D2 receptor agonist, induced lactotroph apoptosis, while sulpiride, a D2 receptor antagonist, blocked DA-induced cell death. The blockade of DAT by GBR12909 did not affect the apoptotic action of DA, but inhibited 6-OHDA-induced apoptosis. Conclusion: These data show that DA, through D2 receptor activation, induces apoptosis of estrogen-sensitized anterior pituitary cells, and suggest that DA contributes to the control of lactotroph number not only by inhibiting proliferation but also by inducing apoptosis.


Neuroendocrinology | 2010

Estrogen Receptors and Signaling Pathways in Lactotropes and Somatotropes

Sandra Zárate; Adriana Seilicovich

Estrogens are crucial determinants in the regulation of anterior pituitary function and maintenance of tissue homeostasis. Estrogen actions in this gland are exerted through both classical and non-classical mechanisms of action. This review summarizes the expression of classical α- and β-estrogen receptors and variant isoforms of estrogen receptors in anterior pituitary cell subpopulations. We also analyze estrogen receptor signaling pathways involved in estrogenic actions in the anterior pituitary gland, especially in lactotropes and somatotropes. Complex interactions between multiple signaling pathways are involved in estrogen regulation of hormone secretion, cell proliferation and cell death in this gland. Insight into these pituitary responses to estrogens would help to understand pituitary function and tumorigenesis.


Frontiers of Hormone Research | 2010

Role of Estrogens in Anterior Pituitary Gland Remodeling during the Estrous Cycle

Sandra Zárate; Verónica Zaldivar; Gabriela Jaita; L. Magri; Daniela Radl; Daniel Pisera; Adriana Seilicovich

In this review, we analyze the action of estrogens leading to the remodeling of the anterior pituitary gland, especially during the estrous cycle. Proliferation and death of anterior pituitary cells and especially lactotropes is regulated by estrogens, which act by sensitizing these cells to both mitotic and apoptotic stimuli such as TNF-alpha, FasL and dopamine. During the estrous cycle, the changing pattern of gonadal steroids is thought to modulate both cell proliferation and death in the anterior pituitary gland, estrogens being key players in cell turnover. The mechanisms involved in estrogen-modulated cell renewal in the anterior pituitary gland during the estrous cycle could include an increase in the expression of proapoptotic cytokines as well as the increase in the Bax/Bcl-2 ratio at proestrus, when estrogen levels are highest and a peak of apoptosis, in particular of lactotropes, is evident in this gland. Estrogens exert rapid antimitogenic and proapoptotic actions in the anterior pituitary through membrane-associated estrogen receptors, a mechanism that might also be involved in remodeling of this gland during the estrous cycle.


Neuroendocrinology | 2011

Estradiol Increases the Expression of TNF-α and TNF Receptor 1 in Lactotropes

Verónica Zaldivar; María L. Magri; Sandra Zárate; Gabriela Jaita; Guadalupe Eijo; Daniela Radl; Jimena Ferraris; Daniel Pisera; Adriana Seilicovich

Background: Estrogens are recognized modulators of pituitary cell renewal, sensitizing cells to mitogenic and apoptotic signals. Tumor necrosis factor-α (TNF-α) is a proinflammatory cytokine that plays an important role in tissue homeostasis modulating cell proliferation, differentiation and death. We previously demonstrated that TNF-α-induced apoptosis of anterior pituitary cells from female rats is estrogen-dependent and predominant in cells from rats at proestrus when estradiol levels are the highest. Aims: Considering that one of the mechanisms involved in the apoptotic action of estrogens can result from increased expression of cytokines and/or their receptors, the aim of the present study was to evaluate the effect of estrogens on the expression of TNF-α and its receptor, TNF receptor 1 (TNFR1), in anterior pituitary cells. Methods/Results: TNFR1 expression, determined by Western blot, was higher in anterior pituitary glands from rats at proestrus than at diestrus. Incubation of anterior pituitary cells from ovariectomized rats with 17β-estradiol enhanced TNFR1 protein expression. As determined by double immunocytochemistry, the expression of TNF-α and TNFR1 was detected in prolactin-, GH-, LH- and ACTH-bearing cells. 17β-estradiol increased the percentage of TNF-α and TNFR1-immunoreactive lactotropes but did not modify the number of GH-bearing cells expressing TNF-α or TNFR1. Conclusion: Our results demonstrate that estradiol increases the expression of TNF-α and TNFR1 in anterior pituitary cells, especially in lactotropes. The sensitizing action of estrogens to proapoptotic stimuli at proestrus in the anterior pituitary gland may involve changes in the expression of the TNF-α/TNFR1 system.


Frontiers in Aging Neuroscience | 2017

Role of Estrogen and Other Sex Hormones in Brain Aging. Neuroprotection and DNA Repair

Sandra Zárate; Tinna Stevnsner; Ricardo Gredilla

Aging is an inevitable biological process characterized by a progressive decline in physiological function and increased susceptibility to disease. The detrimental effects of aging are observed in all tissues, the brain being the most important one due to its main role in the homeostasis of the organism. As our knowledge about the underlying mechanisms of brain aging increases, potential approaches to preserve brain function rise significantly. Accumulating evidence suggests that loss of genomic maintenance may contribute to aging, especially in the central nervous system (CNS) owing to its low DNA repair capacity. Sex hormones, particularly estrogens, possess potent antioxidant properties and play important roles in maintaining normal reproductive and non-reproductive functions. They exert neuroprotective actions and their loss during aging and natural or surgical menopause is associated with mitochondrial dysfunction, neuroinflammation, synaptic decline, cognitive impairment and increased risk of age-related disorders. Moreover, loss of sex hormones has been suggested to promote an accelerated aging phenotype eventually leading to the development of brain hypometabolism, a feature often observed in menopausal women and prodromal Alzheimer’s disease (AD). Although data on the relation between sex hormones and DNA repair mechanisms in the brain is still limited, various investigations have linked sex hormone levels with different DNA repair enzymes. Here, we review estrogen anti-aging and neuroprotective mechanisms, which are currently an area of intense study, together with the effect they may have on the DNA repair capacity in the brain.


PLOS ONE | 2012

Estrogens induce expression of membrane-associated estrogen receptor α isoforms in lactotropes.

Sandra Zárate; Gabriela Jaita; Jimena Ferraris; Guadalupe Eijo; María L. Magri; Daniel Pisera; Adriana Seilicovich

Estrogens are key to anterior pituitary function, stimulating hormone release and controlling cell fate to achieve pituitary dynamic adaptation to changing physiological conditions. In addition to their classical mechanism of action through intracellular estrogen receptors (ERs), estrogens exert rapid actions via cell membrane-localized ERs (mERs). We previously showed that E2 exerts a rapid pro-apoptotic action in anterior pituitary cells, especially in lactotropes and somatotropes, through activation of mERs. In the present study, we examined the involvement of mERα in the rapid pro-apoptotic action of estradiol by TUNEL in primary cultures of anterior pituitary cells from ovariectomized rats using a cell-impermeable E2 conjugate (E2-BSA) and an ERα selective antagonist (MPP dihydrochloride). We studied mERα expression during the estrous cycle and its regulation by gonadal steroids in vivo by flow cytometry. We identified ERα variants in the plasma membrane of anterior pituitary cells during the estrous cycle and studied E2 regulation of these mERα variants in vitro by surface biotinylation and Western Blot. E2-BSA-induced apoptosis was abrogated by MPP in total anterior pituitary cells and lactotropes. In cycling rats, we detected a higher number of lactotropes and a lower number of somatotropes expressing mERα at proestrus than at diestrus. Acute E2 treatment increased the percentage of mERα-expressing lactotropes whereas it decreased the percentage of mERα-expressing somatotropes. We detected three mERα isoforms of 66, 39 and 22 kDa. Expression of mERα66 and mERα39 was higher at proestrus than at diestrus, and short-term E2 incubation increased expression of these two mERα variants. Our results indicate that the rapid apoptotic action exerted by E2 in lactotropes depends on mERα, probably full-length ERα and/or a 39 kDa ERα variant. Expression and activation of mERα variants in lactotropes could be one of the mechanisms through which E2 participates in anterior pituitary cell renewal during the estrous cycle.


Journal of Neuroendocrinology | 2011

Inhibition of nuclear factor-kappa B sensitises anterior pituitary cells to tumour necrosis factor-α- and lipopolysaccharide-induced apoptosis.

Guadalupe Eijo; Sandra Zárate; Gabriela Jaita; Jimena Ferraris; María L. Magri; Verónica Zaldivar; Daniela Radl; V. Boti; Daniel Pisera; Adriana Seilicovich

Nuclear factor‐kappa B (NF‐κB), an important pro‐inflammatory factor, is a crucial regulator of cell survival. Both lipopolysaccharide (LPS) and tumour necrosis factor (TNF)‐α activate NF‐κB signalling. Oestrogens were shown to suppress NF‐κB activation. Oestrogens exert a sensitising action to pro‐apoptotic stimuli such as LPS and TNF‐α in anterior pituitary cells. In the present study, we show by western blotting that 17β‐oestradiol (E2) decreases TNF‐α‐induced NF‐κB/p65 and p50 nuclear translocation in primary cultures of anterior pituitary cells from ovariectomised (OVX) rats. Also, the in vivo administration of E2 decreases LPS‐induced NF‐κB/p65 and p50 nuclear translocation. To investigate whether the inhibition of NF‐κB pathway sensitises anterior pituitary cells to pro‐apoptotic stimuli, we used an inhibitor of NF‐κB activity, BAY 11‐7082 (BAY). BAY, at a concentration that fails to induce apoptosis, has permissive action on TNF‐α‐induced apoptosis of lactotrophs and somatotrophs from OVX rats, as assessed by terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL). Pharmacological inhibition of NF‐κB signalling enhances E2‐sensitising effect to TNF‐α‐induced apoptosis in lactotrophs but not in somatotrophs. In vivo administration of BAY allowed LPS‐induced apoptosis in anterior pituitary cells from OVX rats (determined by fluorescence activated cell sorting). Furthermore, LPS‐induced expression of Bcl‐xL in pituitaries of OVX rats is decreased by E2 administration. Our results show that inhibition of the NF‐κB signalling pathway sensitises anterior pituitary cells to the pro‐apoptotic action of LPS and TNF‐α. Because E2 inhibits LPS‐ and TNF‐α‐activated NF‐κB nuclear translocation, the present study suggests that E2 sensitises anterior pituitary cells to TNF‐α‐ and LPS‐induced apoptosis by inhibiting NF‐κB activity.

Collaboration


Dive into the Sandra Zárate's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel Pisera

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Gabriela Jaita

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Guadalupe Eijo

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Jimena Ferraris

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar

Daniela Radl

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

María L. Magri

University of Buenos Aires

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge