Network


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

Hotspot


Dive into the research topics where Tiziana Mennini is active.

Publication


Featured researches published by Tiziana Mennini.


Proceedings of the National Academy of Sciences of the United States of America | 2001

Erythropoietin prevents neuronal apoptosis after cerebral ischemia and metabolic stress

Anna Leena Sirén; Maddalena Fratelli; Michael Brines; Christoph G. Goemans; Simona Casagrande; Piotr Lewczuk; Sonja Keenan; Christoph H. Gleiter; Claudio Pasquali; Annalisa Capobianco; Tiziana Mennini; Rolf Heumann; Anthony Cerami; Hannelore Ehrenreich; Pietro Ghezzi

Erythropoietin (EPO) promotes neuronal survival after hypoxia and other metabolic insults by largely unknown mechanisms. Apoptosis and necrosis have been proposed as mechanisms of cellular demise, and either could be the target of actions of EPO. This study evaluates whether antiapoptotic mechanisms can account for the neuroprotective actions of EPO. Systemic administration of EPO (5,000 units/kg of body weight, i.p.) after middle-cerebral artery occlusion in rats dramatically reduces the volume of infarction 24 h later, in concert with an almost complete reduction in the number of terminal deoxynucleotidyltransferase-mediated dUTP nick-end labeling of neurons within the ischemic penumbra. In both pure and mixed neuronal cultures, EPO (0.1–10 units/ml) also inhibits apoptosis induced by serum deprivation or kainic acid exposure. Protection requires pretreatment, consistent with the induction of a gene expression program, and is sustained for 3 days without the continued presence of EPO. EPO (0.3 units/ml) also protects hippocampal neurons against hypoxia-induced neuronal death through activation of extracellular signal-regulated kinases and protein kinase Akt-1/protein kinase B. The action of EPO is not limited to directly promoting cell survival, as EPO is trophic but not mitogenic in cultured neuronal cells. These data suggest that inhibition of neuronal apoptosis underlies short latency protective effects of EPO after cerebral ischemia and other brain injuries. The neurotrophic actions suggest there may be longer-latency effects as well. Evaluation of EPO, a compound established as clinically safe, as neuroprotective therapy in acute brain injury is further supported.


Journal of Experimental Medicine | 2003

Erythropoietin selectively attenuates cytokine production and inflammation in cerebral ischemia by targeting neuronal apoptosis.

Pia Villa; Paolo Bigini; Tiziana Mennini; Davide Agnello; Teresa Laragione; Alfredo Cagnotto; Barbara Viviani; Marina Marinovich; Anthony Cerami; Thomas Coleman; Michael Brines; Pietro Ghezzi

Ischemic brain injury resulting from stroke arises from primary neuronal losses and by inflammatory responses. Previous studies suggest that erythropoietin (EPO) attenuates both processes. Although EPO is clearly antiapoptotic for neurons after experimental stroke, it is unknown whether EPO also directly modulates EPO receptor (EPO-R)–expressing glia, microglia, and other inflammatory cells. In these experiments, we show that recombinant human EPO (rhEPO; 5,000 U/kg body weight, i.p.) markedly reduces astrocyte activation and the recruitment of leukocytes and microglia into an infarction produced by middle cerebral artery occlusion in rats. In addition, ischemia-induced production of the proinflammatory cytokines tumor necrosis factor, interleukin 6, and monocyte chemoattractant protein 1 concentration is reduced by >50% after rhEPO administration. Similar results were also observed in mixed neuronal-glial cocultures exposed to the neuronal-selective toxin trimethyl tin. In contrast, rhEPO did not inhibit cytokine production by astrocyte cultures exposed to neuronal homogenates or modulate the response of human peripheral blood mononuclear cells, rat glial cells, or the brain to lipopolysaccharide. These findings suggest that rhEPO attenuates ischemia-induced inflammation by reducing neuronal death rather than by direct effects upon EPO-R–expressing inflammatory cells.


Brain Research | 2002

Erythropoietin exerts an anti-inflammatory effect on the CNS in a model of experimental autoimmune encephalomyelitis

Davide Agnello; Paolo Bigini; Pia Villa; Tiziana Mennini; Anthony Cerami; Michael Brines; Pietro Ghezzi

In recent work we reported that systemically administered erythropoietin (EPO) crosses the blood-brain barrier and has protective effects in animal models of cerebral ischemia, brain trauma and in a rat model of experimental autoimmune encephalomyelitis (EAE). Here we characterize the effect of systemic EPO on the inflammatory component of actively induced, acute EAE in Lewis rats. Administration of EPO at doses of 500-5000 U/kg bw i.p., daily from day 3 after immunization with myelin basic protein (MBP), delayed the onset of EAE and decreased its clinical score at peak time (days 12-13). Immunohistochemical analysis of the spinal cord using anti-glial fibrillary acidic protein (GFAP) and anti-CD11b antibodies showed that EPO markedly diminished inflammation and glial activation/proliferation. EAE induced significant levels of TNF and IL-6 in the spinal cord, where IL-6 was maximum at the onset of the disease (day 10) and TNF at its peak (day 12). EPO delayed the increase of TNF levels, without altering their peak levels, and markedly reduced those of IL-6 suggesting that the decreased inflammation and clinical score may be in part upon attenuation of IL-6. On the other hand, EPO was without effect in a model of adjuvant-induced arthritis in Lewis rats, suggesting a specificity towards autoimmune demyelinating diseases. These data suggest that EPO might act as a protective cytokine in inflammatory pathologies of the CNS.


Naunyn-schmiedebergs Archives of Pharmacology | 1979

m-Chlorophenylpiperazine: A central serotonin agonist causing powerful anorexia in rats

Rosario Samanin; Tiziana Mennini; A. Ferraris; C. Bendotti; Franco Borsini; Silvio Garattini

SummaryMeta-chlorophenylpiperazine inhibited serotonin and noradrenaline uptake by synaptosomes to the same extent with IC50 of 1.3×10−6 M and 5.8×10−6 M respectively. Dopamine uptake was lesss affected by meta-chlorophenylpiperazine (IC50 of 2.2×10−5 M). Unlike d-amphetamine and d-fenfluramine, the drug did not significantly increase monoamine release in synaptosomal preparations. On the other hand, metachlorophenylpiperazine showed an IC50 of 620 nM in displacing 3H-5HT binding to brain membranes. Meta-chlorophenylpiperazine produced a dose-dependent reduction of food intake and this effect was prevented by a pretreatment with methergoline, a serotonin antagonist. The effect of metachlorophenylpiperazine was not modified by an intraventricular injection of 6-hydroxydopamine, electrolytic lesions of nucleus medianus raphe or ventral noradrenergic bundle, nor by a pretreatment with penfluridol, propranolol or phentolamine. The data suggest that the decrease of food intake induced by metachlorophenylpiperazine depends on its ability to act as a serotonin agonist in the brain. The specificity of the effects on serotonin suggests that this compound could prove an important tool for studies aimed at elucidating the functional role of serotonin in the central nervous system.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Nonerythropoietic, tissue-protective peptides derived from the tertiary structure of erythropoietin.

Michael Brines; Nimesh S. A. Patel; Pia Villa; Courtenay Brines; Tiziana Mennini; Massimiliano De Paola; Zübeyde Erbayraktar; Serhat Erbayraktar; Bruno Sepodes; Christoph Thiemermann; Pietro Ghezzi; Michael A. Yamin; Carla Hand; Qiao wen Xie; Thomas Coleman; Anthony Cerami

Erythropoietin (EPO), a member of the type 1 cytokine superfamily, plays a critical hormonal role regulating erythrocyte production as well as a paracrine/autocrine role in which locally produced EPO protects a wide variety of tissues from diverse injuries. Significantly, these functions are mediated by distinct receptors: hematopoiesis via the EPO receptor homodimer and tissue protection via a heterocomplex composed of the EPO receptor and CD131, the β common receptor. In the present work, we have delimited tissue-protective domains within EPO to short peptide sequences. We demonstrate that helix B (amino acid residues 58–82) of EPO, which faces the aqueous medium when EPO is bound to the receptor homodimer, is both neuroprotective in vitro and tissue protective in vivo in a variety of models, including ischemic stroke, diabetes-induced retinal edema, and peripheral nerve trauma. Remarkably, an 11-aa peptide composed of adjacent amino acids forming the aqueous face of helix B is also tissue protective, as confirmed by its therapeutic benefit in models of ischemic stroke and renal ischemia–reperfusion. Further, this peptide simulating the aqueous surface of helix B also exhibits EPOs trophic effects by accelerating wound healing and augmenting cognitive function in rodents. As anticipated, neither helix B nor the 11-aa peptide is erythropoietic in vitro or in vivo. Thus, the tissue-protective activities of EPO are mimicked by small, nonerythropoietic peptides that simulate a portion of EPOs three-dimensional structure.


British Journal of Pharmacology | 1997

Carrier-dependent and Ca2+-dependent 5-HT and dopamine release induced by (+)-amphetamine, 3,4-methylendioxy-methamphetamine, p-chloroamphetamine and (+)-fenfluramine

Daniela Crespi; Tiziana Mennini; Marco Gobbi

1 The mechanism underlying 5‐hydroxytryptamine (5‐HT) and/or dopamine release induced by (+)‐amphetamine ((+)‐Amph), 3,4‐methylendioxymethamphetamine (MDMA), p‐chloroamphetamine (pCA) and (+)‐fenfluramine ((+)‐Fen) was investigated in rat brain superfused synaptosomes preloaded with the 3H neurotransmitters. 2 Their rank order of potency for [3H]‐5‐HT‐releasing activity was the same as for inhibition of 5‐HT uptake (pCAMDMA(+)‐Fen>>(+)‐Amph). Similarly, their rank order as [3H]‐dopamine releasers and dopamine uptake inhibitors was the same ((+)‐Amph>>pCA=MDMA>>(+)‐Fen). We also confirmed that the release induced by these compounds was prevented by selective transporter inhibitors (indalpine or nomifensine). 3 [3H]‐5‐HT and/or [3H]‐dopamine release induced by all these compounds was partially (31–80%), but significantly Ca2+‐dependent. Lack of extracellular Ca2+ did not alter uptake mechanisms nor did it modify the carrier‐dependent dopamine‐induced [3H]‐dopamine release. (+)‐Amph‐induced [3H]‐dopamine release and pCA‐ and MDMA‐induced [3H]‐5‐HT release were significantly inhibited by ω‐agatoxin‐IVA, a specific blocker of P‐type voltage‐operated Ca2+‐channels, similar to the previous results on (+)‐Fen‐induced [3H]‐5‐HT release. 4 Methiothepin inhibited the Ca2+‐dependent component of (+)‐Amph‐induced [3H]‐dopamine release with high potency (70 nM), as previously found with (+)‐Fen‐induced [3H]‐5‐HT release. The inhibitory effect of methiothepin was not due to its effects as a transporter inhibitor or Ca2+‐channel blocker and is unlikely to be due to its antagonist properties on 5‐HT1/2, dopamine or any other extracellular receptor. 5 These results indicate that the release induced by these compounds is both ‘carrier‐mediated’ and Ca2+‐dependent (possibly exocytotic‐like), with the specific carrier allowing the amphetamines to enter the synaptosome. The Ca2+‐dependent release is mediated by Ca2+‐influx (mainly through P‐type Ca2+‐channels), possibly triggered by the drug interacting with an unknown intracellular target, affected by methiothepin, common to both 5‐HT and dopamine synaptosomes.


Journal of Neurochemistry | 2008

Transgenic SOD1 G93A mice develop reduced GLT-1 in spinal cord without alterations in cerebrospinal fluid glutamate levels.

Caterina Bendotti; Massimo Tortarolo; Sachin K. Suchak; Novella Calvaresi; Lucia Carvelli; Antonio Bastone; T. Massimo Rizzi; Marcus Rattray; Tiziana Mennini

Glutamate‐induced excitotoxicity is suggested to play a central role in the development of amyotrophic lateral sclerosis (ALS), although it is still unclear whether it represents a primary cause in the cascade leading to motor neurone death. We used western blotting, immunocytochemistry and in situ hybridization to examine the expression of GLT‐1 in transgenic mice carrying a mutated (G93A) human copper–zinc superoxide dismutase (TgSOD1 G93A), which closely mimic the features of ALS. We observed a progressive decrease in the immunoreactivity of the glial glutamate transporter (GLT‐1) in the ventral, but not in the dorsal, horn of lumbar spinal cord. This effect was specifically found in 14‐ and 18‐week‐old mice that had motor function impairment, motor neurone loss and reactive astrocytosis. No changes in GLT‐1 were observed at 8 weeks of age, before the appearance of clinical symptoms. Decreases in GLT‐1 were accompanied by increased glial fibrillary acidic protein (GFAP) levels and no change in the levels of GLAST, another glial glutamate transporter. The glutamate concentration in the cerebrospinal fluid (CSF) of TgSOD1 G93A mice was not modified at any of the time points examined, compared with age‐matched controls. These findings indicate that the loss of GLT‐1 protein in ALS mice selectively occurs in the areas affected by neurodegeneration and reactive astrocytosis and it is not associated with increases of glutamate levels in CSF. The lack of changes in GLT‐1 at the presymptomatic stage suggests that glial glutamate transporter reduction is not a primary event leading to motor neurone loss.


Annals of the New York Academy of Sciences | 2006

From Fenfluramine Racemate to d-Fenfluramine

Silvio Garattini; Tiziana Mennini; Rosario Samanin

: Experiments using the binding of various ligands for monoamines to rat brain membranes and synaptosomal preparations for studying monoamine uptake and release have shown that d-fenfluramine is more potent than the l isomer in inhibiting 5-HT uptake, whereas d-norfenfluramine preferentially releases 5-HT from a reserpine-insensitive compartment. Studies on brain monoamine metabolism in intact animals have shown that the d and l isomers of fenfluramine at relatively low doses have a specific action on brain 5-HT and catecholamines, respectively. Based on the different ability of metergoline and ritanserin to displace 5-HT2 binding to rat brain membranes and to antagonize d-fenfluramines anorexia, evidence has been provided that d-fenfluramine preferentially uses 5-HT1 sites in the rat brain to cause anorexia in this animal species. Finally, characteristics, regional distribution, and pharmacological characterization of a high-affinity [3H]d-fenfluramine binding to rat brain membranes have been described. This binding appears to be different from 5-HT uptake sites ([3H]imipramine binding) and 5-HT receptors and is not regionally related to the endogenous levels of 5-HT in the rat brain. It is, however, preferentially displaced by some agents using 5-HT to cause anorexia in rats, raising the possibility that it is somewhat related to 5-HT mechanisms involved in feeding control.


Journal of Neuroimmunology | 2006

Delayed administration of erythropoietin and its non-erythropoietic derivatives ameliorates chronic murine autoimmune encephalomyelitis

Costanza Savino; Rosetta Pedotti; Fulvio Baggi; Federica Ubiali; Barbara Gallo; Sara Nava; Paolo Bigini; Sara Barbera; Elena Fumagalli; Tiziana Mennini; Annamaria Vezzani; Massimo Rizzi; Thomas Coleman; Anthony Cerami; Michael Brines; Pietro Ghezzi; Roberto Bianchi

Erythropoietin (EPO) mediates a wide range of neuroprotective activities, including amelioration of disease and neuroinflammation in rat models of EAE. However, optimum dosing parameters are currently unknown. In the present study, we used a chronic EAE model induced in mice by immunization with the myelin oligodendrocyte glycoprotein peptide (MOG35-55) to compare the effect of EPO given with different treatment schedules. EPO was administered intraperitoneally at 0.5, 5.0 or 50 microg/kg three times weekly starting from day 3 after immunization (preventive schedule), at the onset of clinical disease (therapeutic schedule) or 15 days after the onset of symptoms (late therapeutic schedule). The results show that EPO is effective even when given after the appearance of clinical signs of EAE, but with a reduced efficacy compared to the preventative schedule. To determine whether this effect requires the homodimeric EPO receptor (EPOR2)-mediated hematopoietic effect of EPO, we studied the effect of carbamylated EPO (CEPO) that does not bind EPOR2. CEPO, ameliorated EAE without changing the hemoglobin concentration. Another non-erythropoietic derivative, asialoEPO was also effective. Both EPO and CEPO equivalently decreased the EAE-associated production of TNF-alpha, IL-1beta and IL-1Ra in the spinal cord, and IFN-gamma by peripheral lymphocytes, indicating that their action involves targeting neuroinflammation. The lowest dosage tested appeared fully effective. The possibility to dissociate the anti-neuroinflammatory action of EPO from its hematopoietic action, which may cause undesired side effects in non-anemic patients, present new avenues to the therapy of multiple sclerosis.


Psychopharmacology | 1985

Anorectic effect of fenfluramine isomers and metabolites: Relationship between brain levels and in vitro potencies on serotonergic mechanisms

Tiziana Mennini; Silvio Garattini; Silvio Caccia

A study of the possible molecular mechanisms of action by which the isomers and metabolites of fenfluramine increase serotonin transmission, leading to anorectic activity, is presented. The actual brain levels of fenfluramine and norfenfluramine isomers after administration of equi-anorectic doses to rats are compared with their potencies in affecting serotonergic mechanisms in vitro. Isomers and metabolites of fenfluramine can have the same pharmacological action by influencing serotonin uptake, release and binding in a quantitatively different manner.

Collaboration


Dive into the Tiziana Mennini's collaboration.

Top Co-Authors

Avatar

Marco Gobbi

University of Strasbourg

View shared research outputs
Top Co-Authors

Avatar

Alfredo Cagnotto

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Paolo Bigini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Silvio Garattini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvio Caccia

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Elena Fumagalli

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge