Network


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

Hotspot


Dive into the research topics where Silvia Grottelli is active.

Publication


Featured researches published by Silvia Grottelli.


Journal of Nutritional Biochemistry | 2012

Inhibition of NF-κB nuclear translocation via HO-1 activation underlies α-tocopheryl succinate toxicity

Ilaria Bellezza; Arianna Tucci; Francesco Galli; Silvia Grottelli; Anna Lisa Mierla; Francesca Pilolli; Alba Minelli

α-Tocopheryl succinate (α-TOS) inhibits oxidative phosphorylation at the level of mitochondrial complex I and II, thus promoting cancer cell death through mitochondrial reactive oxygen species (ROS) generation. Redox imbalance activates NF-E2 p45-related factor 2 (Nrf2), a transcription factor involved in cell protection and detoxification responses. Here we examined the involvement of heme oxygenase-1 (HO-1) in the regulation of nuclear factor κB (NF-κB) signaling by short exposure to α-TOS in prostate cancer cells. A short-term (4 h) exposure to α-TOS causes a significant reduction in cell viability (76%±9%) and a moderate rise in ROS production (113%±8%). α-TOS alters glutathione (GSH) homeostasis by inducing a biphasic effect, i.e., an early (1 h) decrease in intracellular GSH content (56%±20%) followed by a threefold rise at 4 h. α-TOS increases nuclear translocation and electrophile-responsive/antioxidant-responsive elements binding activity of Nrf2, resulting in up-regulation of downstream genes cystine-glutamic acid exchange transporter and HO-1, while decreasing NF-κB nuclear translocation. This effect is suppressed by the pharmacological inhibition of HO-1 and mimicked by the end-products of HO activity, i.e., bilirubin and carbon monoxide. Results suggest a little understood mechanism for α-TOS-induced inhibition of NF-κB nuclear translocation due to HO-1 up-regulation.


Amino Acids | 2008

Focus on cyclo(His-Pro): history and perspectives as antioxidant peptide

Alba Minelli; Ilaria Bellezza; Silvia Grottelli; Francesco Galli

Summary.Cyclo(His-Pro) is an endogenous cyclic dipeptide structurally related to tyreotropin-releasing hormone that was originally discovered in brain. In the central nervous system it has been described to exert multiple biological activities, which seem to be related to a presynaptic dopaminergic mechanism and include among the others a leptin-like function. It can be found in several body fluids and in the gastrointestinal tract where it has been suggested to act as a gut peptide with influence on the entero-insular axis. The oral administration of cyclo(His-Pro) and zinc was described to improve with a synergistic mechanism the glycaemic control in diabetes.The most intriguing function of this cyclic dipeptide is related with its neuroprotective role that was first reported in traumatic injuries of the spinal cord, and then confirmed in other models of experimental injuries of the nervous system. The mechanism that lies behind the neuroprotective activity of cyclo(His-Pro) remain poorly understood. Recent in vitro studies on rat pheochromocytoma PC12 cells have shown that it is a protective factor against stress stimuli and there is early pre-clinical evidence strongly suggesting that it enhances the expression of small heat shock proteins and antioxidant protection at the cellular level.Future research is underway to better characterize the possible use of this cyclic dipeptide in the therapy of neurodegenerative and metabolic disorders.


Journal of Neurochemistry | 2009

Cyclo(His-Pro) up-regulates heme oxygenase 1 via activation of Nrf2-ARE signalling.

Alba Minelli; Carmela Conte; Silvia Grottelli; Ilaria Bellezza; Carla Emiliani; Juan P. Bolaños

Paraquat (1,1′‐dimethyl‐4,4′‐bipyridinium), a widely used non‐selective herbicide, is a redox cycling agent with adverse effects on dopamine systems. Epidemiological data have shown that exposure to paraquat is one of the several risk factors for Parkinson’s disease. We have already shown that cyclo(His‐Pro), an endogenous cyclic dipeptide produced by the cleavage of the thyrotropin releasing hormone, has a cytoprotective effect through a mechanism involving Nrf2 activation that decreases production of reactive oxygen species and increases glutathione synthesis. Using primary neuronal cultures and PC12 cells as targets of paraquat neurotoxicity, we addressed whether and how cyclo(His‐Pro) causes cellular protective response against paraquat‐mediated cell death. We found that cyclo(His‐Pro) attenuated reactive oxygen species production, and prevented glutathione depletion by up‐regulating Nrf2 gene expression, triggering its nuclear accumulation and activating the expression of heme oxygenase1. These protective effects were abolished by RNA interference‐mediated Nrf2 knock down whereas were unaffected by RNA interference‐mediated Keap1 knock down. Inhibition of heme oxygenase activity decreased cyclo(His‐Pro)‐induced neuroprotection. These results suggest that cyclo(His‐Pro), acting as a selective activator of the brain modulable Nrf2 pathway, may be a promising candidate as neuroprotective agent that act through induction of phase II genes.


The International Journal of Biochemistry & Cell Biology | 2012

Cyclo(His-Pro) exerts anti-inflammatory effects by modulating NF-κB and Nrf2 signalling.

Alba Minelli; Silvia Grottelli; Annalisa Mierla; Francesco Pinnen; Ivana Cacciatore; Ilaria Bellezza

Cyclo(His-Pro) is an endogenous cyclic dipeptide that exerts oxidative damage protection by selectively activating the transcription factor Nrf2 signalling pathway. Given the existence of a tight interplay of the Nrf2/NF-κB systems and that the pro-inflammatory response is governed by transcription factor NF-κB, here we sought to investigate whether and how cyclo(His-Pro) interferes with the cross-talk between the antioxidant Nrf2/heme oxygenase-1 and the pro-inflammatory NF-κB pathways. By knocking down the Nrf2 gene, we confirmed that cyclo(His-Pro) inhibits NF-κB nuclear accumulation induced by paraquat in rat pheochromocytoma PC12 cells via the Nrf2/heme oxygenase-1 pathway. The protection required functional heme oxygenase-1 activity, since zinc protoporphyrin IX, a heme oxygenase-1 inhibitor, prevented NF-κB inhibition, and the presence of exogenous carbon monoxide and bilirubin afforded cytoprotection against paraquat-induced toxicity by preventing NF-κB activation. Cyclooxygenase-2 and matrix metalloproteinase 3, two gene products governed by NF-κB, were down-regulated by cyclo(His-Pro) and up-regulated in heme oxygenase-1 knock-down cells. We validated the general mechanism underlying the anti-inflammatory effects by treating PC12 and murine microglial BV2 cells with different pro-inflammatory agents. Finally, cyclo(His-Pro) reduced 12-otetradecanoylphorbol-13-acetate-induced oedema in mouse ear inflammation model. Results, by showing that cyclo(His-pro) suppresses the pro-inflammatory NF-κB signalling via the Nrf2-mediated heme oxygenase-1 activation, contribute to the understanding of essential cellular pathways and allow the proposal of cyclo(His-Pro) as an in vivo anti-inflammatory compound.


Journal of Cellular and Molecular Medicine | 2009

Cyclo(His‐Pro) promotes cytoprotection by activating Nrf2‐mediated up‐regulation of antioxidant defence

Alba Minelli; Carmela Conte; Silvia Grottelli; Maria Bellezza; Ivana Cacciatore; Juan P. Bolaños

Hystidyl‐proline [cyclo(His‐Pro)] is an endogenous cyclic dipeptide produced by the cleavage of thyrotropin releasing hormone. Previous studies have shown that cyclo(His‐Pro) protects against oxidative stress, although the underlying mechanism has remained elusive. Here, we addressed this issue and found that cyclo(His‐Pro) triggered nuclear accumulation of NF‐E2‐related factor‐2 (Nrf2), a transcription factor that up‐regulates antioxidant‐/electrophile‐responsive element (ARE‐EpRE)‐related genes, in PC12 cells. Cyclo(His‐Pro) attenuated reactive oxygen species production, and prevented glutathione depletion caused by glutamate, rotenone, paraquat and β‐amyloid treatment. Moreover, real‐time PCR analyses revealed that cyclo(His‐Pro) induced the expression of a number of ARE‐related genes and protected cells against hydrogen peroxide‐mediated apoptotic death. Furthermore, these effects were abolished by RNA interference‐mediated Nrf2 knockdown. Finally, pharmacological inhibition of p‐38 MAPK partially prevented both cyclo(His‐Pro)‐mediated Nrf2 activation and cellular protection. These results suggest that the signalling mechanism responsible for the cytoprotective actions of cyclo(His‐Pro) would involve p‐38 MAPK activation leading to Nrf2‐mediated up‐regulation of antioxidant cellular defence.


The International Journal of Biochemistry & Cell Biology | 2014

Neuroinflammation and endoplasmic reticulum stress are coregulated by cyclo(His-Pro) to prevent LPS neurotoxicity.

Ilaria Bellezza; Silvia Grottelli; Anna Lisa Mierla; Ivana Cacciatore; Erika Fornasari; Luca Roscini; Gianluigi Cardinali; Alba Minelli

Many neurological and neurodegenerative diseases are associated with oxidative stress and glial inflammation, all related to endoplasmic reticulum stress. Cyclo(His-Pro) is an endogenous cyclic dipeptide that exerts cytoprotection by interfering with the Nrf2-NF-κB systems, the former presiding the antioxidant and the latter the pro-inflammatory cellular response. Here we investigated whether the cyclic dipeptide inhibits glial inflammation thus reducing the detrimental effect of inflammatory neurotoxins on neurons. We found that systemic administration of cyclo(His-Pro) exerts in vivo anti-inflammatory effects in the central nervous system by down-regulating hepatic and cerebral TNFα expression thereby counteracting LPS-induced gliosis. Mechanistic studies indicated that the cyclic dipeptide-mediated effects are achieved through the activation of Nrf2-driven antioxidant response and the inhibition of the pro-inflammatory NF-κB pathway. Moreover, by up-regulating Bip, cyclo(His-Pro) increases the ER stress sensitivity and triggers the unfolded protein response to alleviate the ER stress. These results unveil a novel potential therapeutic use of cyclo(His-Pro) against neuroinflammatory-related diseases and we might now consider its potential anti-inflammatory role in other neuropathological conditions.


Peptides | 2006

Phosphoproteomic analysis of the effect of cyclo-[His-Pro] dipeptide on PC12 cells

Alba Minelli; Ilaria Bellezza; Silvia Grottelli; Francesco Pinnen; Luigi Brunetti; Michele Vacca

The effects of dipeptide cyclo-[His-Pro] (CHP), known to participate in the appetite behavior and food intake control, have been investigated using PC12 cells in culture as model system. We found that only in the presence of experimental conditions that cause cellular stress the cyclic dipeptide affect cellular proliferation and protects from apoptosis. It greatly enhances the phosphorylation of hsp27, alpha-B-crystallin, Cdc2, and p-38 MAPK, whereas it decreases the phosphorylation of MEK1, Cav 2, GSK3a, PKB/Akt, PKCdelta, PKCgamma, and Erk2. PKA and PKG are involved in ERK1/2 deactivation via a receptor that appears to be dually coupled to Gs and Gq protein subfamilies.


Gene | 2014

α-Tocopheryl succinate pre-treatment attenuates quinone toxicity in prostate cancer PC3 cells.

Ilaria Bellezza; Silvia Grottelli; Leonardo Gatticchi; Anna Lisa Mierla; Alba Minelli

UNLABELLED α-Tocopheryl succinate is one of the most effective analogues of vitamin E for inhibiting cell proliferation and inducing cell death in a variety of cancerous cell lines while sparing normal cells or tissues. αTocopheryl succinate inhibits oxidative phosphorylation at the level of mitochondrial complexes I and II, thus enhancing reactive oxygen species generation which, in turn, induces the expression of Nrf2-driven antioxidant/detoxifying genes. The cytoprotective role of Nrf2 downstream genes/proteins prompted us to investigate whether and how α-tocopheryl succinate increases resistance of PC3 prostate cancer cells to pro-oxidant damage. A 4h α-tocopheryl succinate pre-treatment increases glutathione intracellular content, indicating that the vitamin E derivative is capable of training the cells to react to an oxidative insult. We found that α-tocopheryl succinate pre-treatment does not enhance paraquat-/hydroquinone-induced cytotoxicity whereas it exhibits an additional/synergistic effect on H₂O₂₋/docetaxel-induced cytotoxicity. While glutathione and heme oxygenase-1 are not involved in α-tocopheryl succinate-induced adaptive response to paraquat, NAD(P)H quinone oxidoreductase seems to be responsible, at least in part, for the lack of the additional response. Silencing the gene and/or the inhibition of NAD(P)H quinone oxidoreductase activity counteracts the α-tocopheryl succinate-induced adaptive response. In conclusion, the adaptive response to α-tocopheryl succinate shows that the activation of Nrf2 can promote the survival of cancer cells in an unfavourable environment.


Molecular Neurobiology | 2018

Peroxynitrite Activates the NLRP3 Inflammasome Cascade in SOD1(G93A) Mouse Model of Amyotrophic Lateral Sclerosis

Ilaria Bellezza; Silvia Grottelli; Egidia Costanzi; Paolo Scarpelli; Eva Pigna; Giulio Morozzi; Letizia Mezzasoma; Matthew J. Peirce; Viviana Moresi; Sergio Adamo; Alba Minelli

Neuroinflammation, characterized by the appearance of reactive microglial and astroglial cells, is one of the several pathogenic mechanisms of amyotrophic lateral sclerosis (ALS), a fast-progressing and fatal neurodegenerative disease. Cerebrospinal fluid and spinal cord of ALS patients and SOD1 mutant mice show high concentrations of IL-1β. This interleukin, expressed as an inactive precursor, undergoes a proteolytic maturation by caspase1, whose activation, in turn, depends on inflammasomes. Whether and how inflammasome is activated in ALS models is still to be clarified. The mechanism of inflammasome activation was studied in murine microglial cells overexpressing hSOD1(G93A) and verified in the spinal cord of hSOD1(G93A) mice. Murine microglial hSOD1(G93A) cells express all the inflammasome components and LPS activates caspase1 leading to an increase in the secretion of IL-1β. By activating NF-κB, LPS increases ROS and NO levels that spontaneously react to form peroxynitrite, thus leading to protein nitration. Reduction in peroxynitrite levels results in a decrease in caspase1 activity. Protein nitration and caspase1 activity are concomitantly increased in the spinal cord of pre-symptomatic SOD1(G93A) mice. Oxidative/nitrosative stress induces peroxynitrite formation that may be a key trigger of caspase1/inflammasome activation. Peroxynitrite formation may play a critical role in inflammasome activation and might be exploited as potential therapeutic target for ALS.


Mitochondrion | 2010

Adenosine A1 receptors contribute to mitochondria vulnerability to pro-oxidant stressors

Alba Minelli; Silvia Grottelli; Lanfranco Corazzi; Ilaria Bellezza; Maria Grazia Rambotti; Carla Emiliani; Bertil B. Fredholm

A(1) adenosine receptors are highly expressed in the central nervous system. Mitochondrial function is a major player in adenosine receptors-mediated effects. Here, by using mice with genetic deletion of the A(1) receptor, we addressed the existence of a relationship between mitochondria functions and adenosine A(1) receptor. Mitochondrial functions and effects of MPP(+) in primary mixed cultures are influenced by the presence of the A(1) receptor, demonstrating, for the first time, the mitochondrial localization of the adenosine A(1) receptor and suggesting a role for this receptor as a mitochondrial vulnerability factor.

Collaboration


Dive into the Silvia Grottelli's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
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