Network


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

Hotspot


Dive into the research topics where Sara Morando is active.

Publication


Featured researches published by Sara Morando.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Mesenchymal stem cells impair in vivo T-cell priming by dendritic cells

Sabrina Chiesa; Silvia Morbelli; Sara Morando; Michela Massollo; Cecilia Marini; Arinna Bertoni; Francesco Frassoni; Soraya Tabera Bartolomé; Gianmario Sambuceti; Elisabetta Traggiai; Antonio Uccelli

Dendritic cells (DC) are highly specialized antigen-presenting cells characterized by the ability to prime T-cell responses. Mesenchymal stem cells (MSC) are adult stromal progenitor cells displaying immunomodulatory activities including inhibition of DC maturation in vitro. However, the specific impact of MSC on DC functions, upon in vivo administration, has never been elucidated. Here we show that murine MSC impair Toll-like receptor-4 induced activation of DC resulting in the inhibition of cytokines secretion, down-regulation of molecules involved in the migration to the lymph nodes, antigen presentation to CD4+ T cells, and cross-presentation to CD8+ T cells. These effects are associated with the inhibition of phosphorylation of intracellular mitogen-activated protein kinases. Intravenous administration of MSC decreased the number of CCR7 and CD49dβ1 expressing CFSE-labeled DC in the draining lymph nodes and hindered local antigen priming of DO11.10 ovalbumin-specific CD4+ T cells. Upon labeling of DC with technetium-99m hexamethylpropylene amine oxime to follow their in vivo biodistribution, we demonstrated that intravenous injection of MSC blocks, almost instantaneously, the migration of subcutaneously administered ovalbumin-pulsed DC to the draining lymph nodes. These findings indicate that MSC significantly affect DC ability to prime T cells in vivo because of their inability to home to the draining lymph nodes and further confirm MSC potentiality as therapy for immune-mediated diseases.


Journal of Neurochemistry | 2009

Neuroprotective mesenchymal stem cells are endowed with a potent antioxidant effect in vivo.

Cristina Lanza; Sara Morando; Adriana Voci; Laura Canesi; Maria Cristina Principato; Laura D. Serpero; Gianluigi Mancardi; Antonio Uccelli; Laura Vergani

Experimental autoimmune encephalomyelitis (EAE), an animal model for human multiple sclerosis, is characterized by demyelination, inflammation and neurodegeneration of CNS in which free radicals play a role. Recently, the efficacy of murine mesenchimal stem cells (MSCs) as treatment of EAE induced in mice by the encephalitogenic peptide MOG(35–55) was demonstrated. The present study analyzed some markers of oxidative stress, inflammation/degeneration and apoptosis such as metallothioneins (MTs), antioxidant enzymes (superoxide dismutase, catalase and glutathione‐S‐transferase), poly(ADP‐ribose) polymerase‐1 and p53 during EAE progression and following MSC treatment. Expression of the three brain MT isoforms increased significantly in EAE mice compared with healthy controls, but while expression of MT‐1 and MT‐3 increased along EAE course, MT‐2 was up‐regulated at the onset, but returned to levels similar to those of controls in chronic phase. The changes in the transcription and activity of the antioxidant enzymes and in expression of poly(ADP‐ribose) polymerase‐1 and p53 showed the same kinetics observed for MT‐1 and MT‐3 during EAE. Interestingly, i.v. administration of MSCs reduced the EAE‐induced increases in levels/activities of all these proteins. These results support an antioxidant and neuroprotective activity for MSCs that was also confirmed in vitro on neuroblastoma cells exposed to an oxidative insult.


PLOS ONE | 2009

Catastrophic NAD+ Depletion in Activated T Lymphocytes through Nampt Inhibition Reduces Demyelination and Disability in EAE

Santina Bruzzone; Floriana Fruscione; Sara Morando; Tiziana Ferrando; Alessandro Poggi; Anna Garuti; Agustina D'Urso; Martina Selmo; Federica Benvenuto; Michele Cea; Gabriele Zoppoli; Eva Moran; Debora Soncini; Alberto Ballestrero; Bernard Sordat; Franco Patrone; Raul Mostoslavsky; Antonio Uccelli; Alessio Nencioni

Nicotinamide phosphoribosyltransferase (Nampt) inhibitors such as FK866 are potent inhibitors of NAD+ synthesis that show promise for the treatment of different forms of cancer. Based on Nampt upregulation in activated T lymphocytes and on preliminary reports of lymphopenia in FK866 treated patients, we have investigated FK866 for its capacity to interfere with T lymphocyte function and survival. Intracellular pyridine nucleotides, ATP, mitochondrial function, viability, proliferation, activation markers and cytokine secretion were assessed in resting and in activated human T lymphocytes. In addition, we used experimental autoimmune encephalomyelitis (EAE) as a model of T-cell mediated autoimmune disease to assess FK866 efficacy in vivo. We show that activated, but not resting, T lymphocytes undergo massive NAD+ depletion upon FK866-mediated Nampt inhibition. As a consequence, impaired proliferation, reduced IFN-γ and TNF-α production, and finally autophagic cell demise result. We demonstrate that upregulation of the NAD+-degrading enzyme poly-(ADP-ribose)-polymerase (PARP) by activated T cells enhances their susceptibility to NAD+ depletion. In addition, we relate defective IFN-γ and TNF-α production in response to FK866 to impaired Sirt6 activity. Finally, we show that FK866 strikingly reduces the neurological damage and the clinical manifestations of EAE. In conclusion, Nampt inhibitors (and possibly Sirt6 inhibitors) could be used to modulate T cell-mediated immune responses and thereby be beneficial in immune-mediated disorders.


Molecular Medicine | 2012

Intravenous mesenchymal stem cells improve survival and motor function in experimental amyotrophic lateral sclerosis.

Antonio Uccelli; Marco Milanese; Maria Cristina Principato; Sara Morando; Tiziana Bonifacino; Laura Vergani; Debora Giunti; Adriana Voci; Enrico Carminati; Francesco Giribaldi; Claudia Caponnetto; Giambattista Bonanno

Despite some advances in the understanding of amyotrophic lateral sclerosis (ALS) pathogenesis, significant achievements in treating this disease are still lacking. Mesenchymal stromal (stem) cells (MSCs) have been shown to be effective in several models of neurological disease. To determine the effects of the intravenous injection of MSCs in an ALS mouse model during the symptomatic stage of disease, MSCs (1 × 106) were intravenously injected in mice expressing human superoxide dismutase 1 (SOD1) carrying the G93A mutation (SOD1/G93A) presenting with experimental ALS. Survival, motor abilities, histology, oxidative stress markers and [3H]d-aspartate release in the spinal cord were investigated. MSC injection in SOD1/G93A mice improved survival and motor functions compared with saline-injected controls. Injected MSCs scantly home to the central nervous system and poorly engraft. We observed a reduced accumulation of ubiquitin agglomerates and of activated astrocytes and microglia in the spinal cord of MSC-treated SOD1/G93A mice, with no changes in the number of choline acetyltransferase- and glutamate transporter type 1-positive cells. MSC administration turned around the upregulation of metallothionein mRNA expression and of the activity of the antioxidant enzyme glutathione S-transferase, both associated with disease progression. Last, we observed that MSCs reverted both spontaneous and stimulus-evoked neuronal release of (3H)d-aspartate, a marker of endogenous glutamate, which is upregulated in SOD1/G93A mice. These findings suggest that intravenous administration of MSCs significantly improves the clinical outcome and pathological scores of mutant SOD1/G93A mice, thus providing the rationale for their exploitation for the treatment of ALS.


Acta Neuropathologica | 2015

Fumarates modulate microglia activation through a novel HCAR2 signaling pathway and rescue synaptic dysregulation in inflamed CNS

Benedetta Parodi; Silvia Rossi; Sara Morando; Christian Cordano; Alberto Bragoni; Caterina Motta; Cesare Usai; Brian Wipke; Robert H. Scannevin; Giovanni Luigi Mancardi; Diego Centonze; Nicole Kerlero de Rosbo; Antonio Uccelli

Dimethyl fumarate (DMF), recently approved as an oral immunomodulatory treatment for relapsing-remitting multiple sclerosis (MS), metabolizes to monomethyl fumarate (MMF) which crosses the blood–brain barrier and has demonstrated neuroprotective effects in experimental studies. We postulated that MMF exerts neuroprotective effects through modulation of microglia activation, a critical component of the neuroinflammatory cascade that occurs in neurodegenerative diseases such as MS. To ascertain our hypothesis and define the mechanistic pathways involved in the modulating effect of fumarates, we used real-time PCR and biochemical assays to assess changes in the molecular and functional phenotype of microglia, quantitative Western blotting to monitor activation of postulated pathway components, and ex vivo whole-cell patch clamp recording of excitatory post-synaptic currents in corticostriatal slices from mice with experimental autoimmune encephalomyelitis (EAE), a model for MS, to study synaptic transmission. We show that exposure to MMF switches the molecular and functional phenotype of activated microglia from classically activated, pro-inflammatory type to alternatively activated, neuroprotective one, through activation of the hydroxycarboxylic acid receptor 2 (HCAR2). We validate a downstream pathway mediated through the AMPK–Sirt1 axis resulting in deacetylation, and thereby inhibition, of NF-κB and, consequently, of secretion of pro-inflammatory molecules. We demonstrate through ex vivo monitoring of spontaneous glutamate-mediated excitatory post-synaptic currents of single neurons in corticostriatal slices from EAE mice that the neuroprotective effect of DMF was exerted on neurons at pre-synaptic terminals by modulating glutamate release. By exposing control slices to untreated and MMF-treated activated microglia, we confirm the modulating effect of MMF on microglia function and, thereby, its indirect neuroprotective effect at post-synaptic level. These findings, whereby DMF-induced activation of a new HCAR2-dependent pathway on microglia leads to the modulation of neuroinflammation and restores synaptic alterations occurring in EAE, represent a possible novel mechanism of action for DMF in MS.


Journal of Neuropathology and Experimental Neurology | 2012

Blood-Brain Barrier Alterations in the Cerebral Cortex in Experimental Autoimmune Encephalomyelitis

Mariella Errede; Francesco Girolamo; Giovanni Ferrara; Maurizio Strippoli; Sara Morando; Valentina Boldrin; Marco Rizzi; Antonio Uccelli; Roberto Perris; Caterina Bendotti; Mario Salmona; Luisa Roncali; Daniela Virgintino

Abstract The pathophysiology of cerebral cortical lesions in multiple sclerosis (MS) is not understood. We investigated cerebral cortex microvessels during immune-mediated demyelination in the MS model chronic murine experimental autoimmune encephalomyelitis (EAE) by immunolocalization of the endothelial cell tight junction (TJ) integral proteins claudin-5 and occludin, a structural protein of caveolae, caveolin-1, and the blood-brain barrier–specific endothelial transporter, Glut 1. In EAE-affected mice, there were areas of extensivesubpial demyelination and well-demarcated lesions that extended to deeper cortical layers. Activation of microglia and absence of perivascular inflammatory infiltrates were common in these areas. Microvascular endothelial cells showed increased expression of caveolin-1 and a coincident loss of both claudin-5 and occludin normal junctional staining patterns. At a very early disease stage, claudin-5 molecules tended to cluster and form vacuoles that were also Glut 1 positive; the initially preserved occludin pattern became diffusely cytoplasmic at more advanced stages. Possible internalization of claudin-5 on TJ dismantling was suggested by its coexpression with the autophagosomal marker MAP1LC3A. Loss of TJ integrity was confirmed by fluorescein isothiocyanate–dextran experimentsthat showed leakage of the tracer into the perivascular neuropil. These observations indicate that, in the cerebral cortex of EAE-affected mice, there is a microvascular disease that differentially targets claudin-5 and occludin during ongoing demyelination despite only minimal inflammation.


Current Stem Cell Research & Therapy | 2011

Mesenchymal stem cells for multiple sclerosis: does neural differentiation really matter?

Antonio Uccelli; Sara Morando; Silvia Bonanno; Ivan Bonanni; Alessandro Leonardi; Gianluigi Mancardi

The lack of therapies fostering remyelination and regeneration of the neural network deranged by the autoimmune attack occurring in multiple sclerosis (MS), is raising great expectations about stem cells therapies for tissue repair. Mesenchymal stem cells (MSCs) have been proposed as a possible treatment for MS due to the reported capacity of transdifferentiation into neural cells and their ability at modulating immune responses. However, recent studies have demonstrated that many other functional properties are likely to play a role in the therapeutic plasticity of MSCs, including anti-apoptotic, trophic and anti-oxidant effects. These features are mostly based on the paracrine release of soluble molecules, often dictated by local environmental cues. Based on the modest evidence of long-term engraftment and the striking clinical effects that are observed immediately after MSCs administration in the experimental model of MS, we do not favor a major role for transdifferentiation as an important mechanism involved in the therapeutic effect of MSCs.


Journal of Neurochemistry | 2010

Alterations of glutamate release in the spinal cord of mice with experimental autoimmune encephalomyelitis

Antonella Marte; Anna Cavallero; Sara Morando; Antonio Uccelli; Maurizio Raiteri; Ernesto Fedele

J. Neurochem. (2010) 115, 343–352.


Acta Neuropathologica | 2016

NG2, a common denominator for neuroinflammation, blood–brain barrier alteration, and oligodendrocyte precursor response in EAE, plays a role in dendritic cell activation

Giovanni Battista Ferrara; Mariella Errede; Francesco Girolamo; Sara Morando; Federico Ivaldi; Nicolò Panini; Caterina Bendotti; Roberto Perris; Roberto Furlan; Daniela Virgintino; Nicole Kerlero de Rosbo; Antonio Uccelli

In adult CNS, nerve/glial-antigen 2 (NG2) is expressed by oligodendrocyte progenitor cells (OPCs) and is an early marker of pericyte activation in pathological conditions. NG2 could, therefore, play a role in experimental autoimmune encephalomyelitis (EAE), a disease associated with increased blood–brain barrier (BBB) permeability, inflammatory infiltrates, and CNS damage. We induced EAE in NG2 knock-out (NG2KO) mice and used laser confocal microscopy immunofluorescence and morphometry to dissect the effect of NG2 KO on CNS pathology. NG2KO mice developed milder EAE than their wild-type (WT) counterparts, with less intense neuropathology associated with a significant improvement in BBB stability. In contrast to WT mice, OPC numbers did not change in NG2KO mice during EAE. Through FACS and confocal microscopy, we found that NG2 was also expressed by immune cells, including T cells, macrophages, and dendritic cells (DCs). Assessment of recall T cell responses to the encephalitogen by proliferation assays and ELISA showed that, while WT and NG2KO T cells proliferated equally to the encephalitogenic peptide MOG35-55, NG2KO T cells were skewed towards a Th2-type response. Because DCs could be responsible for this effect, we assessed their expression of IL-12 by PCR and intracellular FACS. IL-12-expressing CD11c+ cells were significantly decreased in MOG35-55-primed NG2KO lymph node cells. Importantly, in WT mice, the proportion of IL-12-expressing cells was significantly lower in CD11c+ NG2- cells than in CD11c+ NG2+ cells. To assess the relevance of NG2 at immune system and CNS levels, we induced EAE in bone-marrow chimeric mice, generated with WT recipients of NG2KO bone-marrow cells and vice versa. Regardless of their original phenotype, mice receiving NG2KO bone marrow developed milder EAE than those receiving WT bone marrow. Our data suggest that NG2 plays a role in EAE not only at CNS/BBB level, but also at immune response level, impacting on DC activation and thereby their stimulation of reactive T cells, through controlling IL-12 expression.


Journal of Neuroimmunology | 2014

Monomethyl fumarate inhibits the NFkB pathway and pro-inflammatory cytokine expression in microglia through HCA2 signaling via the AMPK/Sirt axis

Benedetta Parodi; Christian Cordano; Sara Morando; Alberto Bragoni; Debora Giunti; Cesare Usai; Diego Centonze; Nicole Kerlero de Rosbo; Robert H. Scannevin; Antonio Uccelli

injury (SCI) in rodent. However, it is still unclear that the PACAP induced by hMSCs contributes to the cellar protection. Therefore, we examined effect of hMSCs on SCI of PACAP gene deficient mice. We also determined that hMSCs partially suppressed neural inflammation through PACAP with host tissues. Under inhalation of anesthesia, mice either PACAP+/+ (wild) or +/− (KO) were subjected to spinal cord transection by a razor at level of Th9–10 intervertebral spinal cord. One day later, the mice were injected hMSCs (5 × 10 cells/0.5 μL) or vehicle one caudal the intervertebral spinal cord. The mice were scored locomotor activity detected by Basso Mouse Scale (BMS) and evaluated injury area with glial fibrillary acidic protein (GFAP) surrounded area at 7 days post operation. We also examined mouse PACAP and PACAP specific receptor (PAC1R) and human or mice proand anti-inflammatory cytokine gene expressions. WTmice implanted hMSCs into spinal cord ameliorated significantly locomotor activity and injury volume to compare with vehicle-treated one. The protections were canceled by inviable hMSCs implanted into WT mice and by viable hMSCs implanted into KO mice. Spinal cord implanted hMSCs expressed an increase of mouse PACAP gene, but not PAC1R. The hMSCs implanted WT animals were decreased mouse IL-1, TNF-alpha, IL-10 and TGF-beta, and increased IL-4 compared with vehicle treatedWT one. However, given hMSCs into KOmice, IL-1, TGFbeta and IL-4 expressed similar to vehicle-treatedWT one. These results suggest that hMSCs suppress neural damage partially mediated by PACAP of mice and decrease of IL-1 and TGF-beta and increase of IL-4 were regulated by the PACAP expression.

Collaboration


Dive into the Sara Morando'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

Roberto Furlan

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caterina Bendotti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Giovanni Ferrara

Mario Negri Institute for Pharmacological Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge