Network


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

Hotspot


Dive into the research topics where Lucia Lisi is active.

Publication


Featured researches published by Lucia Lisi.


Journal of Neuroinflammation | 2011

The mTOR kinase inhibitor rapamycin decreases iNOS mRNA stability in astrocytes

Lucia Lisi; Pierluigi Navarra; Douglas L. Feinstein; Cinzia Dello Russo

BackgroundReactive astrocytes are capable of producing a variety of pro-inflammatory mediators and potentially neurotoxic compounds, including nitric oxide (NO). High amounts of NO are synthesized following up-regulation of inducible NO synthase (iNOS). The expression of iNOS is tightly regulated by complex molecular mechanisms, involving both transcriptional and post-transcriptional processes. The mammalian target of rapamycin (mTOR) kinase modulates the activity of some proteins directly involved in post-transcriptional processes of mRNA degradation. mTOR is a serine-threonine kinase that plays an evolutionarily conserved role in the regulation of cell growth, proliferation, survival, and metabolism. It is also a key regulator of intracellular processes in glial cells. However, with respect to iNOS expression, both stimulatory and inhibitory actions involving the mTOR pathway have been described. In this study the effects of mTOR inhibition on iNOS regulation were evaluated in astrocytes.MethodsPrimary cultures of rat cortical astrocytes were activated with different proinflammatory stimuli, namely a mixture of cytokines (TNFα, IFNγ, and IL-1β) or by LPS plus IFNγ. Rapamycin was used at nM concentrations to block mTOR activity and under these conditions we measured its effects on the iNOS promoter, mRNA and protein levels. Functional experiments to evaluate iNOS activity were also included.ResultsIn this experimental paradigm mTOR activation did not significantly affect astrocyte iNOS activity, but mTOR pathway was involved in the regulation of iNOS expression. Rapamycin did not display any significant effects under basal conditions, on either iNOS activity or its expression. However, the drug significantly increased iNOS mRNA levels after 4 h incubation in presence of pro-inflammatory stimuli. This stimulatory effect was transient, since no differences in either iNOS mRNA or protein levels were detected after 24 h. Interestingly, reduced levels of iNOS mRNA were detected after 48 hours, suggesting that rapamycin can modify iNOS mRNA stability. In this regard, we found that rapamycin significantly reduced the half-life of iNOS mRNA, from 4 h to 50 min when cells were co-incubated with cytokine mixture and 10 nM rapamycin. Similarly, rapamycin induced a significant up-regulation of tristetraprolin (TTP), a protein involved in the regulation of iNOS mRNA stability.ConclusionThe present findings show that mTOR controls the rate of iNOS mRNA degradation in astrocytes. Together with the marked anti-inflammatory effects that we previously observed in microglial cells, these data suggest possible beneficial effects of mTOR inhibitors in the treatment of inflammatory-based CNS pathologies.


Biochemical Pharmacology | 2009

Involvement of mTOR kinase in cytokine-dependent microglial activation and cell proliferation.

Cinzia Dello Russo; Lucia Lisi; Giuseppe Tringali; Pierluigi Navarra

Neuroinflammation plays a prominent role in the pathophysiology of several neurodegenerative disorders, including Multiple Sclerosis. Reactive microglial cells are always found in areas of active demyelination as well as in normal-appearing white matter. Microglia contribute to initiating and maintaining brain inflammation, and once activated release pro-inflammatory mediators potentially cytotoxic, like nitric oxide (NO). It is now evident that the mTOR signaling pathway regulates different functions in the innate immune system, contributing to macrophage activation. More recently, mTOR has been found to enhance the survival of EOC2 microglia during oxygen-glucose deprivation and increase NO synthase 2 (NOS2) expression during hypoxia in BV2 microglial cell line, thus suggesting an involvement in microglial pro-inflammatory activation. In the present study, we detected mTOR activation in response to two different stimuli, namely LPS and a mixture of cytokines, in primary cultures of rat cortical microglia. Moreover, mTOR inhibitors reduced NOS activity and NOS2 expression induced by cytokines, but not those induced by LPS. The mTOR inhibitor RAD001, in combination with cytokines, also reduced microglial proliferation and the intracellular levels of cyclooxygenase. Under basal conditions mTOR inhibition significantly reduced microglial viability. Interestingly, mTOR inhibitors did not display any relevant effect on astrocyte NOS2 activity or cell viability. In conclusion, mTOR selectively controls microglial activation in response to pro-inflammatory cytokines and appears to play a crucial role in microglial viability; thus these drugs may be a useful pharmacological tool to reduce neuroinflammation.


Asn Neuro | 2011

The anti-inflammatory effects of dimethyl fumarate in astrocytes involve glutathione and haem oxygenase-1

Shao Xia Lin; Lucia Lisi; Cinzia Dello Russo; Paul E. Polak; Anthony Sharp; Guy Weinberg; Sergey Kalinin; Douglas L. Feinstein

DMF (dimethyl fumarate) exerts anti-inflammatory and pro-metabolic effects in a variety of cell types, and a formulation (BG-12) is being evaluated for monotherapy in multiple sclerosis patients. DMF modifies glutathione (GSH) levels that can induce expression of the anti-inflammatory protein HO-1 (haem oxygenase-1). In primary astrocytes and C6 glioma cells, BG-12 dose-dependently suppressed nitrite production induced by either LI [LPS (lipopolysaccharide) at 1 μg/ml plus IFNγ (interferon γ) at 20 units/ml] or a mixture of pro-inflammatory cytokines, with greater efficacy in C6 cells. BG-12 reduced NOS2 (nitric oxide synthase 2) mRNA levels and activation of a NOS2 promoter, reduced nuclear levels of NF-κB (nuclear factor κB) p65 subunit and attenuated loss of IκBα (inhibitory κBα) in both cell types, although with greater effects in astrocytes. In astrocytes, LI decreased mRNA levels for GSHr (GSH reductase) and GCL (c-glutamylcysteine synthetase), and slightly suppressed GSHs (GSH synthetase) mRNAs. Co-treatment with BG-12 prevented those decreased and increased levels above control values. In contrast, LI reduced GSHp (GSH peroxidase) and GCL in C6 cells, and BG-12 had no effect on those levels. BG-12 increased nuclear levels of Nrf2 (nuclear factor-erythroid 2 p45 subunit-related factor 2), an inducer of GSH-related enzymes, in astrocytes but not C6 cells. In astrocytes, GSH was decreased by BG-12 at 2 h and increased at 24 h. Prior depletion of GSH using buthionine-sulfoximine increased the ability of BG-12 to reduce nitrites. In astrocytes, BG-12 increased HO-1 mRNA levels and effects on nitrite levels were blocked by an HO-1 inhibitor. These results demonstrate that BG-12 suppresses inflammatory activation in astrocytes and C6 glioma cells, but with distinct mechanisms, different dependence on GSH and different effects on transcription factor activation.


BioMed Research International | 2015

mTOR Kinase: A Possible Pharmacological Target in the Management of Chronic Pain

Lucia Lisi; Paola Aceto; Pierluigi Navarra; Cinzia Dello Russo

Chronic pain represents a major public health problem worldwide. Current pharmacological treatments for chronic pain syndromes, including neuropathic pain, are only partially effective, with significant pain relief achieved in 40–60% of patients. Recent studies suggest that the mammalian target of rapamycin (mTOR) kinase and downstream effectors may be implicated in the development of chronic inflammatory, neuropathic, and cancer pain. The expression and activity of mTOR have been detected in peripheral and central regions involved in pain transmission. mTOR immunoreactivity was found in primary sensory axons, in dorsal root ganglia (DRG), and in dorsal horn neurons. This kinase is a master regulator of protein synthesis, and it is critically involved in the regulation of several neuronal functions, including the synaptic plasticity that is a major mechanism leading to the development of chronic pain. Enhanced activation of this pathway is present in different experimental models of chronic pain. Consistently, pharmacological inhibition of the kinase activity turned out to have significant antinociceptive effects in several experimental models of inflammatory and neuropathic pain. We will review the main evidence from animal and human studies supporting the hypothesis that mTOR may be a novel pharmacological target for the management of chronic pain.


Glia | 2013

mTOR kinase, a key player in the regulation of glial functions: Relevance for the therapy of multiple sclerosis

Cinzia Dello Russo; Lucia Lisi; Douglas L. Feinstein; Pierluigi Navarra

The mammalian target of rapamycin (mTOR) is a serine/threonine protein kinase with a central role in the regulation of cell growth and proliferation, and several intracellular processes, such as mRNA transcription and translation, autophagy and cytoskeletal organization. The relevance of this pathway in the regulation of the immune system is well characterized. mTOR is essential for the proper activation and proliferation of effector T cells, restricts the development of regulatory T cells, and downregulates innate immune responses. Recently, a direct role of mTOR in the modulation of glial functions has also been recognized. Data from our group and others support the notion that mTOR is involved in microglial proinflammatory activation. The kinase regulates several intracellular processes in astrocytes, among which the rate of mRNA degradation of the inducible form of NO synthase. Therefore, the inhibition of mTOR kinase activity in glial cells results in anti‐inflammatory actions, suggesting possible beneficial effects of mTOR inhibitors (like rapamycin) in the treatment of inflammatory‐based pathologies of the central nervous system. In contrast, mTOR plays an important role in the regulation of oligodendrocyte development and myelination process as well as several neuronal functions, which may limit this therapeutic approach. Nevertheless, as reviewed here, there is robust evidence that rapamycin ameliorates the clinical course of both the relapsing‐remitting and the chronic experimental autoimmune encephalomyelitis (EAE), and significantly reduces the hyperalgesia observed before clinical development of EAE. These findings may have important clinical implications for the therapy of multiple sclerosis.


Journal of Neuroimmunology | 2012

Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis.

Lucia Lisi; Pierluigi Navarra; R. Cirocchi; Anthony Sharp; Egidio Stigliano; Douglas L. Feinstein; C. Dello Russo

Current treatments used in Multiple Sclerosis (MS) are partly effective in the early stages of the disease but display very limited benefits in patients affected by progressive MS. One possible explanation is that these therapies are unable to target the inflammatory component most active during the progressive phase of the disease, and compartmentalized behind the blood-brain barrier. Our findings show that Rapamycin ameliorates clinical and histological signs of chronic EAE when administered during ongoing disease. Moreover, Rapamycin significantly reduced the hyperalgesia observed before clinical development of EAE which, in turn, is completely abolished by the administration of the drug.


Pharmacogenomics | 2011

Novel sensitive, specific and rapid pharmacogenomic test for the prediction of abacavir hypersensitivity reaction: HLA-B*57:01 detection by real-time PCR.

Cinzia Dello Russo; Lucia Lisi; Alessia Lofaro; Simona Di Giambenedetto; Bruno Federico; Giordano Madeddu; Marianna Salerno; Maria Stella Mura; Antonella Pirazzoli; Andrea De Luca; Roberto Cauda; Pierluigi Navarra

AIM International HIV treatment guidelines recommend HLA-B*57:01 typing before abacavir administration, in order to reduce the incidence of abacavir hypersensitivity reactions, the major cause of early therapy discontinuation. A fast, sensitive and specific test for HLA-B*57:01 detection has been developed in the present study. MATERIALS & METHODS Two sets of sequence-specific primers were designed, and amplification rapidly detected by real-time PCR. RESULTS A total of 108 samples were analyzed in a single-blind fashion, and 41 samples were identified as positive. Complete agreement, with κ = 1 (standard error = 0.0962, p < 0.0001), was found, with a validated methodology used in the EPI109367 clinical trial funded by GlaxoSmithKline, and consisting of low-resolution sequence-specific oligonucleotide PCR, followed by high-resolution sequence-specific oligonucleotide PCR carried out on the HLA-B*57-positive samples. CONCLUSION We provided a detailed characterization of a novel HLA-B*57:01 screening test, which can be easily implemented by those laboratories already involved in the detection of viral load and virus genotyping. Original submitted 26 October 2010; Revision submitted 13 December 2010.


Asn Neuro | 2014

Proinflammatory-Activated Glioma Cells Induce a Switch in Microglial Polarization and Activation Status, From a Predominant M2b Phenotype to a Mixture of M1 and M2a/B Polarized Cells

Lucia Lisi; Egidio Stigliano; Libero Lauriola; Pierluigi Navarra; Cinzia Dello Russo

Malignant gliomas are primary brain tumors characterized by morphological and genetic complexities, as well as diffuse infiltration into normal brain parenchyma. Within gliomas, microglia/macrophages represent the largest tumor-infiltrating cell population, contributing by at least one-third to the total tumor mass. Bi-directional interactions between glioma cells and microglia may therefore play an important role on tumor growth and biology. In the present study, we have characterized the influence of glioma-soluble factors on microglial function, comparing the effects of media harvested under basal conditions with those of media obtained after inducing a pro-inflammatory activation state in glioma cells. We found that microglial cells undergo a different pattern of activation depending on the stimulus; in the presence of activated glioma-derived factors, i.e. a condition mimicking the late stage of pathology, microglia presents as a mixture of polarization phenotypes (M1 and M2a/b), with up-regulation of iNOS (inducible nitric oxide synthase), ARG (arginase) and IL (interleukine)-10. At variance, microglia exposed to basal glioma-derived factors, i.e. a condition resembling the early stage of pathology, shows a more specific pattern of activation, with increased M2b polarization status and up-regulation of IL-10 only. As far as viability and cell proliferation are concerned, both LI-CM [LPS (lipopolysaccharide)–IFNγ (interferon γ) conditioned media] and C-CM (control-conditioned media) induce similar effects on microglial morphology. Finally, in human glioma tissue obtained from surgical resection of patients with IV grade glioblastoma, we detected a significant amount of CD68 positive cells, which is a marker of macrophage/microglial phagocytic activity, suggesting that in vitro findings presented here might have a relevance in the human pathology as well.


Journal of Neurochemistry | 2012

Modulatory effects of the CCR5 antagonist maraviroc on microglial pro‐inflammatory activation elicited by gp120

Lucia Lisi; Antonella Tramutola; Andrea De Luca; Pierluigi Navarra; Cinzia Dello Russo

J. Neurochem. (2012) 120, 106–114.


Journal of Neuroimmunology | 2011

Trigeminal satellite cells express functional calcitonin gene-related peptide receptors, whose activation enhances interleukin-1β pro-inflammatory effects

Alice De Corato; Lucia Lisi; Alessandro Capuano; Giuseppe Tringali; Antonella Tramutola; Pierluigi Navarra; Cinzia Dello Russo

Calcitonin gene-related peptide (CGRP) is the main mediator of trigeminal pain signal. Functional CGRP receptors were detected in trigeminal satellite cells, a specialized type of glia found within the sensory ganglia. CGRP displayed modest pro-inflammatory effects per se on trigeminal satellite cells, while it significantly enhanced IL-1β actions, increasing the expression and activity of cycloxygenase 2 as well as the expression of the inducible form of nitric oxide synthase and IL-1β. CGRP effects were reverted by a specific CGRP receptor antagonist and mimicked by elevation of intracellular cAMP levels. CGRP exerted also minor proinflammatory effects on cortical astrocytes.

Collaboration


Dive into the Lucia Lisi's collaboration.

Top Co-Authors

Avatar

Pierluigi Navarra

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Cinzia Dello Russo

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Giuseppe Tringali

Sapienza University of Rome

View shared research outputs
Top Co-Authors

Avatar

Douglas L. Feinstein

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Giacomo Pozzoli

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Simona Di Giambenedetto

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar

Anthony Sharp

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Diego Currò

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Emilia Laudati

The Catholic University of America

View shared research outputs
Top Co-Authors

Avatar

Sergey Kalinin

University of Illinois at Chicago

View shared research outputs
Researchain Logo
Decentralizing Knowledge