Network


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

Hotspot


Dive into the research topics where Stefano Pluchino is active.

Publication


Featured researches published by Stefano Pluchino.


Nature | 2003

Injection of adult neurospheres induces recovery in a chronic model of multiple sclerosis

Stefano Pluchino; Angelo Quattrini; Elena Brambilla; Angela Gritti; Giuliana Salani; Giorgia Dina; Rossella Galli; Ubaldo Del Carro; Stefano Amadio; Alessandra Bergami; Roberto Furlan; Giancarlo Comi; Angelo L. Vescovi; Gianvito Martino

Widespread demyelination and axonal loss are the pathological hallmarks of multiple sclerosis. The multifocal nature of this chronic inflammatory disease of the central nervous system complicates cellular therapy and puts emphasis on both the donor cell origin and the route of cell transplantation. We established syngenic adult neural stem cell cultures and injected them into an animal model of multiple sclerosis—experimental autoimmune encephalomyelitis (EAE) in the mouse—either intravenously or intracerebroventricularly. In both cases, significant numbers of donor cells entered into demyelinating areas of the central nervous system and differentiated into mature brain cells. Within these areas, oligodendrocyte progenitors markedly increased, with many of them being of donor origin and actively remyelinating axons. Furthermore, a significant reduction of astrogliosis and a marked decrease in the extent of demyelination and axonal loss were observed in transplanted animals. The functional impairment caused by EAE was almost abolished in transplanted mice, both clinically and neurophysiologically. Thus, adult neural precursor cells promote multifocal remyelination and functional recovery after intravenous or intrathecal injection in a chronic model of multiple sclerosis.


Nature | 2005

Neurosphere-derived multipotent precursors promote neuroprotection by an immunomodulatory mechanism

Stefano Pluchino; Lucia Zanotti; Barbara Rossi; Elena Brambilla; Linda Ottoboni; Giuliana Salani; Marianna Martinello; Alessandro Cattalini; Alessandra Bergami; Roberto Furlan; Giancarlo Comi; Gabriela Constantin; Gianvito Martino

In degenerative disorders of the central nervous system (CNS), transplantation of neural multipotent (stem) precursor cells (NPCs) is aimed at replacing damaged neural cells. Here we show that in CNS inflammation, NPCs are able to promote neuroprotection by maintaining undifferentiated features and exerting unexpected immune-like functions. In a mouse model of chronic CNS inflammation, systemically injected adult syngeneic NPCs use constitutively activated integrins and functional chemokine receptors to selectively enter the inflamed CNS. These undifferentiated cells survive repeated episodes of CNS inflammation by accumulating within perivascular areas where reactive astrocytes, inflamed endothelial cells and encephalitogenic T cells produce neurogenic and gliogenic regulators. In perivascular CNS areas, surviving adult NPCs induce apoptosis of blood-borne CNS-infiltrating encephalitogenic T cells, thus protecting against chronic neural tissue loss as well as disease-related disability. These results indicate that undifferentiated adult NPCs have relevant therapeutic potential in chronic inflammatory CNS disorders because they display immune-like functions that promote long-lasting neuroprotection.


Molecular and Cellular Neuroscience | 2006

Microglia activated by IL-4 or IFN-γ differentially induce neurogenesis and oligodendrogenesis from adult stem/progenitor cells

Oleg Butovsky; Yaniv Ziv; Adi Schwartz; Gennady Landa; Adolfo E. Talpalar; Stefano Pluchino; Gianvito Martino; Michal Schwartz

Cell renewal in the adult central nervous system (CNS) is limited, and is blocked in inflammatory brain conditions. We show that both neurogenesis and oligodendrogenesis of adult neural progenitor cells in mice are blocked by inflammation-associated (endotoxin-activated) microglia, but induced by microglia activated by cytokines (IL-4 or low level of IFN-gamma) associated with T-helper cells. Blockage was correlated with up-regulation of microglial production of tumor necrosis factor-alpha. The effect induced by IL-4-activated microglia was mediated, at least in part, by insulin-like growth factor-I. The IL-4-activated microglia showed a bias towards oligodendrogenesis whereas the IFN-gamma-activated microglia showed a bias towards neurogenesis. It thus appears that microglial phenotype critically affects their ability to support or impair cell renewal from adult stem cell.


PLOS Biology | 2012

Vesiclepedia: A Compendium for Extracellular Vesicles with Continuous Community Annotation

Hina Kalra; Richard J. Simpson; Hong Ji; Elena Aikawa; Peter Altevogt; Philip W. Askenase; Vincent C. Bond; Francesc E. Borràs; Xandra O. Breakefield; Vivian Budnik; Edit I. Buzás; Giovanni Camussi; Aled Clayton; Emanuele Cocucci; Juan M. Falcon-Perez; Susanne Gabrielsson; Yong Song Gho; Dwijendra K. Gupta; H. C. Harsha; An Hendrix; Andrew F. Hill; Jameel M. Inal; Guido Jenster; Eva-Maria Krämer-Albers; Sai Kiang Lim; Alicia Llorente; Jan Lötvall; Antonio Marcilla; Lucia Mincheva-Nilsson; Irina Nazarenko

Vesiclepedia is a community-annotated compendium of molecular data on extracellular vesicles.


PLOS Medicine | 2009

Infiltrating Blood-Derived Macrophages Are Vital Cells Playing an Anti-inflammatory Role in Recovery from Spinal Cord Injury in Mice

Ravid Shechter; Anat London; Chen Varol; Catarina Raposo; Melania Cusimano; Gili Yovel; Asya Rolls; Matthias Mack; Stefano Pluchino; Gianvito Martino; Steffen Jung; Michal Schwartz

Using a mouse model of spinal injury, Michal Schwartz and colleagues tested the effect of macrophages on the recovery process and demonstrate an important anti-inflammatory role for a subset of infiltrating monocyte-derived macrophages that is dependent upon their expression of interleukin 10.


Brain | 2009

Delayed post-ischaemic neuroprotection following systemic neural stem cell transplantation involves multiple mechanisms

Marco Bacigaluppi; Stefano Pluchino; Luca Peruzzotti Jametti; Ertugrul Kilic; Ülkan Kilic; Giuliana Salani; Elena Brambilla; Mark J. West; Giancarlo Comi; Gianvito Martino; Dirk M. Hermann

Recent evidence suggests that neural stem/precursor cells (NPCs) promote recovery in animal models with delayed neuronal death via a number of indirect bystander effects. A comprehensive knowledge of how transplanted NPCs exert their therapeutic effects is still lacking. Here, we investigated the effects of a delayed transplantation of adult syngenic NPCs--injected intravenously 72 h after transient middle cerebral artery occlusion--on neurological recovery, histopathology and gene expression. NPC-transplanted mice showed a significantly improved recovery from 18 days post-transplantation (dpt) onwards, which persisted throughout the study. A small percentage of injected NPCs accumulated in the brain, integrating mainly in the infarct boundary zone, where most of the NPCs remained undifferentiated up to 30 dpt. Histopathological analysis revealed a hitherto unreported very delayed neuroprotective effect of NPCs, becoming evident at 10 and 30 dpt. Tissue survival was associated with downregulation of markers of inflammation, glial scar formation and neuronal apoptotic death at both mRNA and protein levels. Our data highlight the relevance of very delayed degenerative processes in the stroke brain that are intimately associated with inflammatory and glial responses. These processes may efficaciously be antagonized by (stem) cell-based strategies at time-points far beyond established therapeutic windows for pharmacological neuroprotection.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Synergy between immune cells and adult neural stem/progenitor cells promotes functional recovery from spinal cord injury.

Yaniv Ziv; Hila Avidan; Stefano Pluchino; Gianvito Martino; Michal Schwartz

The well regulated activities of microglia and T cells specific to central nervous system (CNS) antigens can contribute to the protection of CNS neural cells and their renewal from adult neural stem/progenitor cells (aNPCs). Here we report that T cell-based vaccination of mice with a myelin-derived peptide, when combined with transplantation of aNPCs into the cerebrospinal fluid (CSF), synergistically promoted functional recovery after spinal cord injury. The synergistic effect was correlated with modulation of the nature and intensity of the local T cell and microglial response, expression of brain-derived neurotrophic factor and noggin protein, and appearance of newly formed neurons from endogenous precursor-cell pools. These results substantiate the contention that the local immune response plays a crucial role in recruitment of aNPCs to the lesion site, and suggest that similar immunological manipulations might also serve as a therapeutic means for controlled migration of stem/progenitor cells to other acutely injured CNS sites.


Journal of extracellular vesicles | 2015

Applying extracellular vesicles based therapeutics in clinical trials - an ISEV position paper.

Thomas Lener; Mario Gimona; Ludwig Aigner; Verena Börger; Edit I. Buzás; Giovanni Camussi; Nathalie Chaput; Devasis Chatterjee; Felipe A. Court; Hernando A. del Portillo; Lorraine O'Driscoll; Stefano Fais; Juan M. Falcon-Perez; Ursula Felderhoff-Mueser; Lorenzo Fraile; Yong Song Gho; André Görgens; Ramesh C. Gupta; An Hendrix; Dirk M. Hermann; Andrew F. Hill; Fred H. Hochberg; Peter A. Horn; Dominique P.V. de Kleijn; Lambros Kordelas; Boris W. Kramer; Eva Maria Krämer-Albers; Sandra Laner-Plamberger; Saara Laitinen; Tommaso Leonardi

Extracellular vesicles (EVs), such as exosomes and microvesicles, are released by different cell types and participate in physiological and pathophysiological processes. EVs mediate intercellular communication as cell-derived extracellular signalling organelles that transmit specific information from their cell of origin to their target cells. As a result of these properties, EVs of defined cell types may serve as novel tools for various therapeutic approaches, including (a) anti-tumour therapy, (b) pathogen vaccination, (c) immune-modulatory and regenerative therapies and (d) drug delivery. The translation of EVs into clinical therapies requires the categorization of EV-based therapeutics in compliance with existing regulatory frameworks. As the classification defines subsequent requirements for manufacturing, quality control and clinical investigation, it is of major importance to define whether EVs are considered the active drug components or primarily serve as drug delivery vehicles. For an effective and particularly safe translation of EV-based therapies into clinical practice, a high level of cooperation between researchers, clinicians and competent authorities is essential. In this position statement, basic and clinical scientists, as members of the International Society for Extracellular Vesicles (ISEV) and of the European Cooperation in Science and Technology (COST) program of the European Union, namely European Network on Microvesicles and Exosomes in Health and Disease (ME-HaD), summarize recent developments and the current knowledge of EV-based therapies. Aspects of safety and regulatory requirements that must be considered for pharmaceutical manufacturing and clinical application are highlighted. Production and quality control processes are discussed. Strategies to promote the therapeutic application of EVs in future clinical studies are addressed.


Brain | 2008

Persistent inflammation alters the function of the endogenous brain stem cell compartment.

Stefano Pluchino; Luca Muzio; Jaime Imitola; Michela Deleidi; Clara Alfaro-Cervello; Giuliana Salani; Cristina Porcheri; Elena Brambilla; Francesca Cavasinni; Andrea Bergamaschi; Jose Manuel Garcia-Verdugo; Giancarlo Comi; Samia J. Khoury; Gianvito Martino

Endogenous neural stem/precursor cells (NPCs) are considered a functional reservoir for promoting tissue homeostasis and repair after injury, therefore regenerative strategies that mobilize these cells have recently been proposed. Despite evidence of increased neurogenesis upon acute inflammatory insults (e.g. ischaemic stroke), the plasticity of the endogenous brain stem cell compartment in chronic CNS inflammatory disorders remains poorly characterized. Here we show that persistent brain inflammation, induced by immune cells targeting myelin, extensively alters the proliferative and migratory properties of subventricular zone (SVZ)-resident NPCs in vivo leading to significant accumulation of non-migratory neuroblasts within the SVZ germinal niche. In parallel, we demonstrate a quantitative reduction of the putative brain stem cells proliferation in the SVZ during persistent brain inflammation, which is completely reversed after in vitro culture of the isolated NPCs. Together, these data indicate that the inflamed brain microenvironment sustains a non cell-autonomous dysfunction of the endogenous CNS stem cell compartment and challenge the potential efficacy of proposed therapies aimed at mobilizing endogenous precursors in chronic inflammatory brain disorders.


Biochimie | 2013

The stem cell secretome and its role in brain repair

Denise Drago; Chiara Cossetti; Nunzio Iraci; Edoardo Gaude; Giovanna Musco; Angela Bachi; Stefano Pluchino

Compelling evidence exists that non-haematopoietic stem cells, including mesenchymal (MSCs) and neural/progenitor stem cells (NPCs), exert a substantial beneficial and therapeutic effect after transplantation in experimental central nervous system (CNS) disease models through the secretion of immune modulatory or neurotrophic paracrine factors. This paracrine hypothesis has inspired an alternative outlook on the use of stem cells in regenerative neurology. In this paradigm, significant repair of the injured brain may be achieved by injecting the biologics secreted by stem cells (secretome), rather than implanting stem cells themselves for direct cell replacement. The stem cell secretome (SCS) includes cytokines, chemokines and growth factors, and has gained increasing attention in recent years because of its multiple implications for the repair, restoration or regeneration of injured tissues. Thanks to recent improvements in SCS profiling and manipulation, investigators are now inspired to harness the SCS as a novel alternative therapeutic option that might ensure more efficient outcomes than current stem cell-based therapies for CNS repair. This review discusses the most recent identification of MSC- and NPC-secreted factors, including those that are trafficked within extracellular membrane vesicles (EVs), and reflects on their potential effects on brain repair. It also examines some of the most convincing advances in molecular profiling that have enabled mapping of the SCS.

Collaboration


Dive into the Stefano Pluchino's collaboration.

Top Co-Authors

Avatar

Gianvito Martino

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Luca Peruzzotti-Jametti

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Giancarlo Comi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Tommaso Leonardi

European Bioinformatics Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nunzio Iraci

University of Cambridge

View shared research outputs
Top Co-Authors

Avatar

Roberto Furlan

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Elena Brambilla

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Joshua D. Bernstock

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge