Network


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

Hotspot


Dive into the research topics where Alessandro Mariani is active.

Publication


Featured researches published by Alessandro Mariani.


ACS Nano | 2013

Selective Nanovector Mediated Treatment of Activated Proinflammatory Microglia/Macrophages in Spinal Cord Injury

Simonetta Papa; Filippo Rossi; Raffaele Ferrari; Alessandro Mariani; Massimiliano De Paola; Ilaria Caron; Fabio Fiordaliso; Cinzia Bisighini; Eliana Sammali; Claudio Colombo; Marco Gobbi; Mara Canovi; Jacopo Lucchetti; Marco Peviani; Massimo Morbidelli; Gianluigi Forloni; Giuseppe Perale; Davide Moscatelli; Pietro Veglianese

Much evidence shows that acute and chronic inflammation in spinal cord injury (SCI), characterized by immune cell infiltration and release of inflammatory mediators, is implicated in development of the secondary injury phase that occurs after spinal cord trauma and in the worsening of damage. Activation of microglia/macrophages and the associated inflammatory response appears to be a self-propelling mechanism that leads to progressive neurodegeneration and development of persisting pain state. Recent advances in polymer science have provided a huge amount of innovations leading to increased interest for polymeric nanoparticles (NPs) as drug delivery tools to treat SCI. In this study, we tested and evaluated in vitro and in vivo a new drug delivery nanocarrier: minocycline loaded in NPs composed by a polymer based on poly-ε-caprolactone and polyethylene glycol. These NPs are able to selectively target and modulate, specifically, the activated proinflammatory microglia/macrophages in subacute progression of the secondary injury in SCI mouse model. After minocycline-NPs treatment, we demonstrate a reduced activation and proliferation of microglia/macrophages around the lesion site and a reduction of cells with round shape phagocytic-like phenotype in favor of a more arborized resting-like phenotype with low CD68 staining. Treatment here proposed limits, up to 15 days tested, the proinflammatory stimulus associated with microglia/macrophage activation. This was demonstrated by reduced expression of proinflammatory cytokine IL-6 and persistent reduced expression of CD68 in traumatized site. The nanocarrier drug delivery tool developed here shows potential advantages over the conventionally administered anti-inflammatory therapy, maximizing therapeutic efficiency and reducing side effects.


Journal of Controlled Release | 2014

Polymeric nanoparticle system to target activated microglia/macrophages in spinal cord injury

Simonetta Papa; Raffaele Ferrari; Massimiliano De Paola; Filippo Rossi; Alessandro Mariani; Ilaria Caron; Eliana Sammali; Marco Peviani; Valentina Dell'Oro; Claudio Colombo; Massimo Morbidelli; Gianluigi Forloni; Giuseppe Perale; Davide Moscatelli; Pietro Veglianese

The possibility to control the fate of the cells responsible for secondary mechanisms following spinal cord injury (SCI) is one of the most relevant challenges to reduce the post traumatic degeneration of the spinal cord. In particular, microglia/macrophages associated inflammation appears to be a self-propelling mechanism which leads to progressive neurodegeneration and development of persisting pain state. In this study we analyzed the interactions between poly(methyl methacrylate) nanoparticles (PMMA-NPs) and microglia/macrophages in vitro and in vivo, characterizing the features that influence their internalization and ability to deliver drugs. The uptake mechanisms of PMMA-NPs were in-depth investigated, together with their possible toxic effects on microglia/macrophages. In addition, the possibility to deliver a mimetic drug within microglia/macrophages was characterized in vitro and in vivo. Drug-loaded polymeric NPs resulted to be a promising tool for the selective administration of pharmacological compounds in activated microglia/macrophages and thus potentially able to counteract relevant secondary inflammatory events in SCI.


Molecular Medicine | 2012

Neuroprotective effects of toll-like receptor 4 antagonism in spinal cord cultures and in a mouse model of motor neuron degeneration.

Massimiliano De Paola; Alessandro Mariani; Paolo Bigini; Marco Peviani; Giovanni Ferrara; Monica Molteni; Sabrina Gemma; Pietro Veglianese; Valeria Castellaneta; Valentina Boldrin; Carlo Rossetti; Chiara Chiabrando; Gianluigi Forloni; Tiziana Mennini; Roberto Fanelli

Sustained inflammatory reactions are common pathological events associated with neuron loss in neurodegenerative diseases. Reported evidence suggests that Toll-like receptor 4 (TLR4) is a key player of neuroinflammation in several neurodegenerative diseases. However, the mechanisms by which TLR4 mediates neurotoxic signals remain poorly understood. We investigated the role of TLR4 in in vitro and in vivosettings of motor neuron degeneration. Using primary cultures from mouse spinal cords, we characterized both the proinflammatory and neurotoxic effects of TLR4 activation with lipopolysaccharide (activation of microglial cells, release of proinflammatory cytokines and motor neuron death) and the protective effects of a cyanobacteriaderived TLR4 antagonist (VB3323). With the use of TLR4-deficient cells, a critical role of the microglial component with functionally active TLR4 emerged in this setting. The in vivo experiments were carried out in a mouse model of spontaneous motor neuron degeneration, the wobbler mouse, where we preliminarily confirmed a protective effect of TLR4 antagonism. Compared with vehicle- and riluzole-treated mice, those chronically treated with VB3323 showed a decrease in microglial activation and morphological alterations of spinal cord neurons and a better performance in the paw abnormality and grip-strength tests. Taken together, our data add new understanding of the role of TLR4 in mediating neurotoxicity in the spinal cord and suggest that TLR4 antagonists could be considered in future studies as candidate protective agents for motor neurons in degenerative diseases.


Biomaterials | 2016

Early modulation of pro-inflammatory microglia by minocycline loaded nanoparticles confers long lasting protection after spinal cord injury.

Simonetta Papa; Ilaria Caron; Eugenio Erba; Nicolò Panini; Massimiliano De Paola; Alessandro Mariani; Claudio Colombo; Raffaele Ferrari; Diego Pozzer; Elisa R. Zanier; Francesca Pischiutta; Jacopo Lucchetti; Andrea Bassi; Gianluca Valentini; Giulio Alfredo Simonutti; Filippo Rossi; Davide Moscatelli; Gianluigi Forloni; Pietro Veglianese

Many efforts have been performed in order to understand the role of recruited macrophages in the progression of spinal cord injury (SCI). Different studies revealed a pleiotropic effect played by these cells associated to distinct phenotypes (M1 and M2), showing a predictable spatial and temporal distribution in the injured site after SCI. Differently, the role of activated microglia in injury progression has been poorly investigated, mainly because of the challenges to target and selectively modulate them in situ. A delivery nanovector tool (poly-ε-caprolactone-based nanoparticles) able to selectively treat/target microglia has been developed and used here to clarify the temporal and spatial involvement of the pro-inflammatory response associated to microglial cells in SCI. We show that a treatment with nanoparticles loaded with minocycline, the latter a well-known anti-inflammatory drug, when administered acutely in a SCI mouse model is able to efficiently modulate the resident microglial cells reducing the pro-inflammatory response, maintaining a pro-regenerative milieu and ameliorating the behavioral outcome up to 63 days post injury. Furthermore, by using this selective delivery tool we demonstrate a mechanistic link between early microglia activation and M1 macrophages recruitment to the injured site via CCL2 chemokine, revealing a detrimental contribution of pro-inflammatory macrophages to injury progression after SCI.


Pharmacological Research | 2016

Synthetic and natural small molecule TLR4 antagonists inhibit motoneuron death in cultures from ALS mouse model

Massimiliano De Paola; Stefania E. Sestito; Alessandro Mariani; Christian Memo; Roberto Fanelli; Mattia Freschi; Caterina Bendotti; Valentina Calabrese; Francesco Peri

Increasing evidence indicates that inflammatory responses could play a critical role in the pathogenesis of motor neuron injury in amyotrophic lateral sclerosis (ALS). Recent findings have underlined the role of Toll-like receptors (TLRs) and the involvement of both the innate and adaptive immune responses in ALS pathogenesis. In particular, abnormal TLR4 signaling in pro-inflammatory microglia cells has been related to motoneuron degeneration leading to ALS. In this study the effect of small molecule TLR4 antagonists on in vitro ALS models has been investigated. Two different types of synthetic glycolipids and the phenol fraction extracted from commercial extra-virgin olive oil (EVOO) were selected since they efficiently inhibit TLR4 stimulus in HEK cells by interacting with the TLR4·MD-2 complex and CD14 co-receptor. Here, TLR4 antagonists efficiently protected motoneurons from LPS-induced lethality in spinal cord cultures, and inhibited the interleukine-1β production by LPS-stimulated microglia. In motoneurons/glia cocultures obtained from wild type or SOD1 G93A mice, motoneuron death induced by SOD1mut glia was counteracted by TLR4 antagonists. The release of nitric oxide by LPS treatment or SOD1mut glia was also inhibited by EVOO, suggesting that the action of this natural extract could be mainly related to the modulation of this inflammatory mediator.


Journal of Proteomics | 2012

Insight into the neuroproteomics effects of the food-contaminant non-dioxin like polychlorinated biphenyls

Laura Brunelli; Marta Llansola; Vicente Felipo; Roberta Campagna; Luisa Airoldi; Massimiliano De Paola; Roberto Fanelli; Alessandro Mariani; Marco Mazzoletti; Roberta Pastorelli

Recent studies showed that food-contaminant non-dioxin-like polychlorinated biphenyls (NDL-PCBs) congeners (PCB52, PCB138, PCB180) have neurotoxic potential, but the cellular and molecular mechanisms underlying neuronal damage are not entirely known. The aim of this study was to assess whether in-vitro exposure to NDL-PCBs may alter the proteome profile of primary cerebellar neurons in order to expand our knowledge on NDL-PCBs neurotoxicity. Comparison of proteome from unexposed and exposed rat cerebellar neurons was performed using state-of-the-art label-free semi-quantitative mass-spectrometry method. We observed significant changes in the abundance of several proteins, that fall into two main classes: (i) novel targets for both PCB138 and 180, mediating the dysregulation of CREB pathways and ubiquitin-proteasome system; (ii) different congeners-specific targets (alpha-actinin-1 for PCB138; microtubule-associated-protein-2 for PCB180) that might lead to similar deleterious consequences on neurons cytoskeleton organization. Interference of the PCB congeners with synaptic formation was supported by the increased expression of pre- and post-synaptic proteins quantified by western blot and immunocytochemistry. Expression alteration of synaptic markers was confirmed in the cerebellum of rats developmentally exposed to these congeners, suggesting an adaptive response to neurodevelopmental toxicity on brain structures. As such, our work is expected to lead to new insights into the mechanisms of NDL-PCBs neurotoxicity.


Developmental Neurobiology | 2015

Decabrominated diphenyl ether and methylmercury impair fetal nervous system development in mice at documented human exposure levels.

Alessandro Mariani; Roberto Fanelli; Andrea Re Depaolini; Massimiliano De Paola

The central nervous system (CNS) is extremely vulnerable to the toxic effects of environmental pollutants during development. Polybrominated diphenyl ethers (PBDEs) are persistent contaminants, increasingly present in the environment and in human tissues. Recent investigations identified a correlation between maternal exposure to PBDEs and impairment in fetal neurobehavioral development, suggesting that these contaminants pose a potential risk for children. We investigated on the potential effects of environmental decabrominated diphenyl ether (decaBDE, the fully brominated congener) on key neurodevelopmental molecules (e.g., synaptic proteins and immature neuron markers) in fetal mouse neurons. Methylmercury was used as reference neurotoxic contaminant and to evaluate its possible synergism with decaBDE. The neurotoxic effects of decaBDE and methylmercury were determined in developing cultured neurons from mouse fetal hippocampus and cerebellum. Neuron death, dendritic branching, synaptic protein expression, markers of immature neurons, and microglia activation were evaluated by immunocytochemistry. Brain samples from prenatally treated embryos were also examined for neurotoxicity signs by immunoblotting and histochemistry. DecaBDE significantly affected (down to 0.4 nM) the number of dendritic branches, and the levels of synaptic proteins and doublecortin in cultured neurons. Prenatal exposure to decaBDE decreased the synaptic proteins and increased the expression of the immature neuron and microglial markers in mouse fetuses. In conclusion, prenatal exposure to realistic (relevant for human exposure) concentrations of decaBDE induces impairment of fetal CNS development in mice, suggesting a potential risk of fetotoxicity in humans.


Scientific Reports | 2015

Doxycycline hinders phenylalanine fibril assemblies revealing a potential novel therapeutic approach in phenylketonuria

Ada De Luigi; Alessandro Mariani; Massimiliano De Paola; Andrea Re Depaolini; Laura Colombo; Luca Russo; Valeria Rondelli; Paola Brocca; Lihi Adler-Abramovich; Ehud Gazit; Elena Del Favero; Laura Cantù; Mario Salmona

A new paradigm for the aetiopathology of phenylketonuria suggests the presence of amyloid-like assemblies in the brains of transgenic mouse models and patients with phenylketonuria, possibly shedding light on the selective cognitive deficit associated with this disease. Paralleling the amyloidogenic route that identifies different stages of peptide aggregation, corresponding to different levels of toxicity, we experimentally address for the first time, the physico-chemical properties of phenylalanine aggregates via Small Angle, Wide Angle X-ray Scattering and Atomic Force Microscopy. Results are consistent with the presence of well-structured, aligned fibres generated by milliMolar concentrations of phenylalanine. Moreover, the amyloid-modulating doxycycline agent affects the local structure of phenylalanine aggregates, preventing the formation of well-ordered crystalline structures. Phenylalanine assemblies prove toxic in vitro to immortalized cell lines and primary neuronal cells. Furthermore, these assemblies also cause dendritic sprouting alterations and synaptic protein impairment in neurons. Doxycycline counteracts these toxic effects, suggesting an approach for the development of future innovative non-dietary preventive therapies.


Journal of Controlled Release | 2018

Mesenchymal stem cells encapsulated into biomimetic hydrogel scaffold gradually release CCL2 chemokine in situ preserving cytoarchitecture and promoting functional recovery in spinal cord injury

Simonetta Papa; Irma Vismara; Alessandro Mariani; M. Barilani; Stefano Rimondo; M. De Paola; Nicolò Panini; Eugenio Erba; Emanuele Mauri; Filippo Rossi; Gianluigi Forloni; Lorenza Lazzari; Pietro Veglianese

ABSTRACT Spinal cord injury (SCI) is an acute neurodegenerative disorder caused by traumatic damage of the spinal cord. The neuropathological evolution of the primary trauma involves multifactorial processes that exacerbate the pathology, worsening the neurodegeneration and limiting neuroregeneration. This complexity suggests that multi‐therapeutic approaches, rather than any single treatment, might be more effective. Encouraging preclinical results indicate that stem cell‐based treatments may improve the disease outcome due to their multi‐therapeutic ability. Mesenchymal Stem Cells (MSCs) are currently considered one of the most promising approaches. Significant improvement in the behavioral outcome after MSC treatment sustained by hydrogel has been demonstrated. However, it is still not known how hydrogel contribute to the delivery of factors secreted from MSCs and what factors are released in situ. Among different mediators secreted by MSCs after seeding into hydrogel, we have found CCL2 chemokine, which could account for the neuroprotective mechanisms of these cells. CCL2 secreted from human MSCs is delivered efficaciously in the lesioned spinal cord acting not only on recruitment of macrophages, but driving also their conversion to an M2 neuroprotective phenotype. Surprisingly, human CCL2 delivered also plays a key role in preventing motor neuron degeneration in vitro and after spinal cord trauma in vivo, with a significant improvement of the motor performance of the rodent SCI models.


RSC Advances | 2017

Double conjugated nanogels for selective intracellular drug delivery

Emanuele Mauri; Pietro Veglianese; Simonetta Papa; Alessandro Mariani; Massimiliano De Paola; Riccardo Rigamonti; Giulia M.F. Chincarini; Irma Vismara; Stefano Rimondo; Alessandro Sacchetti; Filippo Rossi

One of the most important drawbacks of nanomedicine is related to the unwanted rapid diffusion of drugs loaded within nanocarriers towards the external biological environment, according to the high clearance of body fluids. Therefore, colloids can carry only a small amount of their initial content in the target district, limiting their pharmacological activity and then the therapy. To overcome this limitation, we synthesized double conjugated nanogels: the first click strategy (1,3 Huisgen cycloaddition) guarantees the traceability of nanogels while the second one (disulfide bond) links drug molecules to polymeric chains. In this study, we proposed the above-mentioned double strategy and we validated the synthesized colloids and the selective release kinetics in microglia cells, dramatically involved in several diseases of the central nervous system. Cleavable linked drugs prove to be a promising tool for the selective administration of pharmacological compounds in microglia cells and potentially in many others counteracting some relevant events.

Collaboration


Dive into the Alessandro Mariani's collaboration.

Top Co-Authors

Avatar

Massimiliano De Paola

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Pietro Veglianese

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Simonetta Papa

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Gianluigi Forloni

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Eugenio Erba

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Ilaria Caron

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Roberto Fanelli

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Jacopo Lucchetti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Roberto Fanelli

Mario Negri Institute for Pharmacological Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge