Network


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

Hotspot


Dive into the research topics where Marzia Belicchi is active.

Publication


Featured researches published by Marzia Belicchi.


Journal of Clinical Investigation | 2004

Human circulating AC133 + stem cells restore dystrophin expression and ameliorate function in dystrophic skeletal muscle

Yvan Torrente; Marzia Belicchi; Maurilio Sampaolesi; Federica Pisati; Mirella Meregalli; Giuseppe D’Antona; Rossana Tonlorenzi; Laura Porretti; Manuela Gavina; Kamel Mamchaoui; Denis Furling; Vincent Mouly; Gillian Butler-Browne; Roberto Bottinelli; Giulio Cossu; Nereo Bresolin

Duchenne muscular dystrophy (DMD) is a common X-linked disease characterized by widespread muscle damage that invariably leads to paralysis and death. There is currently no therapy for this disease. Here we report that a subpopulation of circulating cells expressing AC133, a well-characterized marker of hematopoietic stem cells, also expresses early myogenic markers. Freshly isolated, circulating AC133(+) cells were induced to undergo myogenesis when cocultured with myogenic cells or exposed to Wnt-producing cells in vitro and when delivered in vivo through the arterial circulation or directly into the muscles of transgenic scid/mdx mice (which allow survival of human cells). Injected cells also localized under the basal lamina of host muscle fibers and expressed satellite cell markers such as M-cadherin and MYF5. Furthermore, functional tests of injected muscles revealed a substantial recovery of force after treatment. As these cells can be isolated from the blood, manipulated in vitro, and delivered through the circulation, they represent a possible tool for future cell therapy applications in DMD disease or other muscular dystrophies.


Cell Stem Cell | 2007

Restoration of Human Dystrophin Following Transplantation of Exon-Skipping-Engineered DMD Patient Stem Cells into Dystrophic Mice

Rachid Benchaouir; Mirella Meregalli; Andrea Farini; Giuseppe D'Antona; Marzia Belicchi; Aurélie Goyenvalle; M. Battistelli; Nereo Bresolin; Roberto Bottinelli; Luis Garcia; Yvan Torrente

Duchenne muscular dystrophy (DMD) is a hereditary disease caused by mutations that disrupt the dystrophin mRNA reading frame. In some cases, forced exclusion (skipping) of a single exon can restore the reading frame, giving rise to a shorter, but still functional, protein. In this study, we constructed lentiviral vectors expressing antisense oligonucleotides in order to induce an efficient exon skipping and to correct the initial frameshift caused by the DMD deletion of CD133+ stem cells. The intramuscular and intra-arterial delivery of genetically corrected CD133 expressing myogenic progenitors isolated from the blood and muscle of DMD patients results in a significant recovery of muscle morphology, function, and dystrophin expression in scid/mdx mice. These data demonstrate that autologous engrafting of blood or muscle-derived CD133+ cells, previously genetically modified to reexpress a functional dystrophin, represents a promising approach for DMD.


Cell Transplantation | 2007

Autologous transplantation of muscle-derived CD133(+) stem cells in Duchenne muscle patients

Yvan Torrente; Marzia Belicchi; C. Marchesi; Giuseppe D'Antona; Filippo Cogiamanian; Federica Pisati; Manuela Gavina; Giordano R; Rossana Tonlorenzi; Gigliola Fagiolari; Costanza Lamperti; Porretti L; Lopa R; Maurilio Sampaolesi; Vicentini L; N. Grimoldi; Tiberio F; Songa; Baratta P; Alessandro Prelle; Forzenigo L; Michela Guglieri; Orietta Pansarasa; Chiara Rinaldi; Mouly; Gillian Butler-Browne; Giacomo P. Comi; Biondetti P; Maurizio Moggio; S.M. Gaini

Duchenne muscular dystrophy (DMD) is a lethal X-linked recessive muscle disease due to defect on the gene encoding dystrophin. The lack of a functional dystrophin in muscles results in the fragility of the muscle fiber membrane with progressive muscle weakness and premature death. There is no cure for DMD and current treatment options focus primarily on respiratory assistance, comfort care, and delaying the loss of ambulation. Recent works support the idea that stem cells can contribute to muscle repair as well as to replenishment of the satellite cell pool. Here we tested the safety of autologous transplantation of muscle-derived CD133+ cells in eight boys with Duchenne muscular dystrophy in a 7-month, double-blind phase I clinical trial. Stem cell safety was tested by measuring muscle strength and evaluating muscle structures with MRI and histological analysis. Timed cardiac and pulmonary function tests were secondary outcome measures. No local or systemic side effects were observed in all treated DMD patients. Treated patients had an increased ratio of capillary per muscle fibers with a switch from slow to fast myosin-positive myofibers.


Journal of Neuroscience Research | 2004

Human skin-derived stem cells migrate throughout forebrain and differentiate into astrocytes after injection into adult mouse brain.

Marzia Belicchi; Federica Pisati; Raffaella Lopa; Laura Porretti; Francesco Fortunato; Manuela Sironi; Mario Scalamogna; Eugenio Parati; Nereo Bresolin; Yvan Torrente

Recent evidence indicates that neural stem cell properties can be found among a mammalian skin‐derived multipotent population. A major barrier in the further characterization of the human skin‐derived neural progenitors is the inability to isolate this population based on expression of cell surface markers. Our work has been devoted to purified human skin‐derived stem cells that are capable of neural differentiation, based on the presence or absence of the AC133 cell surface marker. The enriched skin‐derived AC133+ cells express the CD34 and Thy‐1 antigens. These cells cultured in a growth medium containing epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF) proliferate, forming spheres, and differentiate in vitro into neurons, astrocytes, and rarely into oligodendrocytes. Single cells from sphere cultures initiated from human purified AC133+ cells were replated as single cells and were able to generate new spheres, demonstrating the self‐renewing ability of these stem cell populations. Brain engraftment of cells obtained from human purified AC133+‐derived spheres generated different neural phenotypes: immature neurons and a most abundant population of well differentiated astrocytes. The AC133‐derived astrocytes assumed perivascular locations in the frontal cortex. No donor‐derived oligodendrocytes were found in the transplanted mouse brains. Several donor small, rounded cells that expressed endothelial markers were found close to the host vessel and near the subventricular zone. Thus, mammalian skin AC133‐derived cells behave as a multipotent population with the capacity to differentiate into neural lineages in vitro and, prevalently, endothelium and astrocytes in vivo, demonstrating the great plasticity of these cells and suggesting potential clinical application.


Molecular Therapy | 2009

In vivo myogenic potential of human CD133+ muscle-derived stem cells: a quantitative study.

Elisa Negroni; Ingo Riederer; Soraya Chaouch; Marzia Belicchi; Paola Razini; James P. Di Santo; Yvan Torrente; Gillian Butler-Browne; Vincent Mouly

In recent years, numerous reports have identified in mouse different sources of myogenic cells distinct from satellite cells that exhibited a variable myogenic potential in vivo. Myogenic stem cells have also been described in humans, although their regenerative potential has rarely been quantified. In this study, we have investigated the myogenic potential of human muscle-derived cells based on the expression of the stem cell marker CD133 as compared to bona fide satellite cells already used in clinical trials. The efficiency of these cells to participate in muscle regeneration and contribute to the renewal of the satellite cell pool, when injected intramuscularly, has been evaluated in the Rag2(-/-) gammaC(-/-) C5(-/-) mouse in which muscle degeneration is induced by cryoinjury. We demonstrate that human muscle-derived CD133+ cells showed a much greater regenerative capacity when compared to human myoblasts. The number of fibers expressing human proteins and the number of human cells in a satellite cell position are all dramatically increased when compared to those observed after injection of human myoblasts. In addition, CD133+/CD34+ cells exhibited a better dispersion in the host muscle when compared to human myoblasts. We propose that muscle-derived CD133+ cells could be an attractive candidate for cellular therapy.


Cell Transplantation | 2007

Induction of neurotrophin expression via human adult mesenchymal stem cells: implication for cell therapy in neurodegenerative diseases.

Federica Pisati; Patrizia Bossolasco; Mirella Meregalli; Lidia Cova; Marzia Belicchi; Manuela Gavina; C. Marchesi; Cinzia Calzarossa; Davide Soligo; Giorgio Lambertenghi-Deliliers; Nereo Bresolin; Vincenzo Silani; Yvan Torrente; Elio Polli

In animal models of neurological disorders for cerebral ischemia, Parkinsons disease, and spinal cord lesions, transplantation of mesenchymal stem cells (MSCs) has been reported to improve functional outcome. Three mechanisms have been suggested for the effects of the MSCs: transdifferentiation of the grafted cells with replacement of degenerating neural cells, cell fusion, and neuroprotection of the dying cells. Here we demonstrate that a restricted number of cells with differentiated astroglial features can be obtained from human adult MSCs (hMSCs) both in vitro using different induction protocols and in vivo after transplantation into the developing mouse brain. We then examined the in vitro differentiation capacity of the hMSCs in coculture with slices of neonatal brain cortex. In this condition the hMSCs did not show any neuronal transdifferentiation but expressed neurotrophin low-affinity (NGFRp75) and high-affinity (trkC) receptors and released nerve growth factor (NGF) and neurotrophin-3 (NT-3). The same neurotrophins expression was demonstrated 45 days after the intracerebral transplantation of hMSCs into nude mice with surviving astroglial cells. These data further confirm the limited capability of adult hMSC to differentiate into neurons whereas they differentiated in astroglial cells. Moreover, the secretion of neurotrophic factors combined with activation of the specific receptors of transplanted hMSCs demonstrated an alternative mechanism for neuroprotection of degenerating neurons. hMSCs are further defined in their transplantation potential for treating neurological disorders.


Cell Transplantation | 2003

Tumor necrosis factor-alpha (TNF-alpha) stimulates chemotactic response in mouse myogenic cells.

Yvan Torrente; El Fahime E; Nicolas Caron; Del Bo R; Marzia Belicchi; Federica Pisati; Jacques P. Tremblay; Nereo Bresolin

Migration of transplanted myogenic cells occurs during both embryogenesis and regeneration of skeletal muscles and is important for successful myoblast transplantation, but little is known about factors that promote chemotaxis of these cells. Tumor necrosis factor-α (TNF-α) is known to induce chemotactic effect on several cell types. In this study, we investigated its influence on the in vitro and in vivo motility of C2C12 and primary myoblasts. In the in vitro test performed in the blind-well Boyden chambers, we showed that TNF-α (50–400 U/ml) significantly enhanced the ability of myogenic cells to migrate. The dose–response curve for this factor was bell shaped, with maximum activity in the 200 U/ml range. In the in vivo test, intramuscular administration of TNF-α was performed by an Alzet pump connected to a perforated polyethylene microtube inserted in the tibialis anterior (TA) of CD1 mice. In these experiments, myoblasts were injected under the muscle epimysium. The recipient mice were immunosuppressed with FK506. Our results showed that, 5 days after myoblast transplantation, cells migrated further in the muscles infused with TNF-α than in the muscles not exposed to TNF-α. TNF-α not only has a chemotactic activity but may also modify cell migration via its action on matrix metalloproteinase (MMP) expression. The proteolytic activities of the MMPs secreted in the muscles were thus also assessed by gelatin zymography. The results showed an increased of MMP-2 and MMP-9 transcripts in the TNF-α-infused muscles injected with myogenic cells. Myoblast migration during transplantation may be enhanced by overlapping gradients of several effector molecules such as TNF-α, interferon-γ (INF-γ), and interleukins, released at the site of muscle injury. We propose that TNF-α may promote myoblast migration directly through chemotactic activity and indirectly by enhancing MMP activity at the site of muscle injury.


Journal of Cell Biology | 2003

Identification of a putative pathway for the muscle homing of stem cells in a muscular dystrophy model

Yvan Torrente; Geoffrey Camirand; Federica Pisati; Marzia Belicchi; Barbara Rossi; Fabio Colombo; Mosthapha El Fahime; Nicolas Caron; Andrew C. Issekutz; Gabriela Constantin; Jacques P. Tremblay; Nereo Bresolin

Attempts to repair muscle damage in Duchenne muscular dystrophy (DMD) by transplanting skeletal myoblasts directly into muscles are faced with the problem of the limited migration of these cells in the muscles. The delivery of myogenic stem cells to the sites of muscle lesions via the systemic circulation is a potential alternative approach to treat this disease. Muscle-derived stem cells (MDSCs) were obtained by a MACS® multisort method. Clones of MDSCs, which were Sca-1+/CD34−/L-selectin+, were found to adhere firmly to the endothelium of mdx dystrophic muscles after i.v. or i.m. injections. The subpopulation of Sca-1+/CD34− MDSCs expressing L-selectin was called homing MDSCs (HMDSCs). Treatment of HMDSCs with antibodies against L-selectin prevented adhesion to the muscle endothelium. Importantly, we found that vascular endothelium from striate muscle of young mdx mice expresses mucosal addressin cell adhesion molecule-1 (MAdCAM-1), a ligand for L-selectin. Our results showed for the first time that the expression of the adhesion molecule L-selectin is important for muscle homing of MDSCs. This discovery will aid in the improvement of a potential therapy for muscular dystrophy based on the systemic delivery of MDSCs.


Glia | 2007

Skin-derived stem cells transplanted into resorbable guides provide functional nerve regeneration after sciatic nerve resection

C. Marchesi; Mauro Pluderi; Federica Colleoni; Marzia Belicchi; Mirella Meregalli; Andrea Farini; D. Parolini; L. Draghi; M. E. Fruguglietti; Manuela Gavina; Laura Porretti; Alessandra Cattaneo; M. Battistelli; Alessandro Prelle; Maurizio Moggio; S. Borsa; Luca Bello; Diego Spagnoli; S. M. Gaini; Maria Cristina Tanzi; Nereo Bresolin; Nadia Grimoldi; Yvan Torrente

The regeneration in the peripheral nervous system is often incomplete and the treatment of severe lesions with nerve tissue loss is primarily aimed at recreating nerve continuity. Guide tubes of various types, filled with Schwann cells, stem cells, or nerve growth factors are attractive as an alternative therapy to nerve grafts. In this study, we evaluated whether skin‐derived stem cells (SDSCs) can improve peripheral nerve regeneration after transplantation into nerve guides. We compared peripheral nerve regeneration in adult rats with sciatic nerve gaps of 16 mm after autologous transplantation of GFP‐labeled SDSCs into two different types of guides: a synthetic guide, obtained by dip coating with a L‐lactide and trimethylene carbonate (PLA‐TMC) copolymer and a collagen‐based guide. The sciatic function index and the recovery rates of the compound muscle action potential were significantly higher in the animals that received SDSCs transplantation, in particular, into the collagen guide, compared to the control guides filled only with PBS. For these guides the morphological and immunohistochemical analysis demonstrated an increased number of myelinated axons expressing S100 and Neurofilament 70, suggesting the presence of regenerating nerve fibers along the gap. GFP positive cells were found around regenerating nerve fibers and few of them were positive for the expression of glial markers as S‐100 and glial fibrillary acidic protein. RT‐PCR analysis confirmed the expression of S100 and myelin basic protein in the animals treated with the collagen guide filled with SDSCs. These data support the hypothesis that SDSCs could represent a tool for future cell therapy applications in peripheral nerve regeneration.


The Journal of Pathology | 2007

T and B lymphocyte depletion has a marked effect on the fibrosis of dystrophic skeletal muscles in the scid/mdx mouse

Andrea Farini; Mirella Meregalli; Marzia Belicchi; M. Battistelli; D. Parolini; Giuseppe D'Antona; Manuela Gavina; Linda Ottoboni; Gabriela Constantin; Roberto Bottinelli; Yvan Torrente

Abnormal connective tissue proliferation following muscle degeneration is a major pathological feature of Duchenne muscular dystrophy (DMD), a genetic myopathy due to lack of the sarcolemmal dystrophin protein. Since this fibrotic proliferation is likely to be a major obstacle to the efficacy of future therapies, research is needed to understand and prevent the fibrotic process in order to develop an effective treatment. Murine muscular dystrophy (mdx) is genetically homologous to DMD, and histopatological alterations are comparable to those of the muscles of patients with DMD. To investigate the development of fibrosis, we bred the mdx mouse with the scid immunodepressed mouse and analysed fibrosis histologically; we used ELISA analysis to determine TGF‐β1 expression. Significant reduction of fibrosis and TGF‐β1 expression was found in the muscles of the scid/mdx mice. However, we observed similar centrally located nuclei, necrosis, muscle degeneration and muscle force compared to the mdx animals. These data demonstrate a correlation between the absence of B and T lymphocytes and loss of fibrosis accompanied by reduction of TGF‐β1, suggesting the importance of modulation of the immune system in DMD. Copyright

Collaboration


Dive into the Marzia Belicchi's collaboration.

Top Co-Authors

Avatar

Yvan Torrente

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Mirella Meregalli

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrea Farini

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Paola Razini

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvia Erratico

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Chiara Villa

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Researchain Logo
Decentralizing Knowledge