Network


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

Hotspot


Dive into the research topics where Manuela Gavina is active.

Publication


Featured researches published by Manuela Gavina.


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 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.


Stem Cells | 2006

Galectin-1 induces skeletal muscle differentiation in human fetal mesenchymal stem cells and increases muscle regeneration

Jerry Chan; Keelin O'Donoghue; Manuela Gavina; Yvan Torrente; Nigel L. Kennea; Huseyin Mehmet; Helen J. S. Stewart; Diana J. Watt; Jennifer E. Morgan; Nicholas M. Fisk

Cell therapy for degenerative muscle diseases such as the muscular dystrophies requires a source of cells with the capacity to participate in the formation of new muscle fibers. We investigated the myogenic potential of human fetal mesenchymal stem cells (hfMSCs) using a variety of stimuli. The use of 5‐azacytidine or steroids did not produce skeletal muscle differentiation, whereas myoblast‐conditioned medium resulted in only 1%–2% of hfMSCs undergoing muscle differentiation. However, in the presence of galectin‐1, 66.1% ± 5.7% of hfMSCs, but not adult bone marrow‐derived mesenchymal stem cells, assumed a muscle phenotype, forming long, multinucleated fibers expressing both desmin and sarcomeric myosin via activation of muscle regulatory factors. Continuous exposure to galectin‐1 resulted in more efficient muscle differentiation than pulsed exposure (62.3% vs. 39.1%; p < .001). When transplanted into regenerating murine muscle, galectin‐1‐exposed hfMSCs formed fourfold more human muscle fibers than nonstimulated hfMSCs (p = .008), with similar results obtained in a scid/mdx dystrophic mouse model. These data suggest that hfMSCs readily undergo muscle differentiation in response to galectin‐1 through a stepwise progression similar to that which occurs during embryonic myogenesis. The high degree of myogenic conversion achieved by this method has relevance for the development of therapies for muscular dystrophies.


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.


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


Cancer Research | 2007

Effect of Human Skin-Derived Stem Cells on Vessel Architecture, Tumor Growth, and Tumor Invasion in Brain Tumor Animal Models

Federica Pisati; Marzia Belicchi; Francesco Acerbi; C. Marchesi; Carlo Giussani; Manuela Gavina; Sophie Javerzat; Martin Hagedorn; Giorgio Carrabba; Valeria Lucini; S. M. Gaini; Nereo Bresolin; Lorenzo Bello; Andreas Bikfalvi; Yvan Torrente

Glioblastomas represent an important cause of cancer-related mortality with poor survival. Despite many advances, the mean survival time has not significantly improved in the last decades. New experimental approaches have shown tumor regression after the grafting of neural stem cells and human mesenchymal stem cells into experimental intracranial gliomas of adult rodents. However, the cell source seems to be an important limitation for autologous transplantation in glioblastoma. In the present study, we evaluated the tumor targeting and antitumor activity of human skin-derived stem cells (hSDSCs) in human brain tumor models. The hSDSCs exhibit tumor targeting characteristics in vivo when injected into the controlateral hemisphere or into the tail vein of mice. When implanted directly into glioblastomas, hSDSCs distributed themselves extensively throughout the tumor mass, reduced tumor vessel density, and decreased angiogenic sprouts. In addition, transplanted hSDSCs differentiate into pericyte cell and release high amounts of human transforming growth factor-beta1 with low expression of vascular endothelial growth factor, which may contribute to the decreased tumor cell invasion and number of tumor vessels. In long-term experiments, the hSDSCs were also able to significantly inhibit tumor growth and to prolong animal survival. Similar behavior was seen when hSDSCs were implanted into two different tumor models, the chicken embryo experimental glioma model and the transgenic Tyrp1-Tag mice. Taken together, these data validate the use of hSDSCs for targeting human brain tumors. They may represent therapeutically effective cells for the treatment of intracranial tumors after autologous transplantation.


FEBS Letters | 2006

High-resolution X-ray microtomography for three-dimensional visualization of human stem cell muscle homing

Yvan Torrente; Manuela Gavina; Marzia Belicchi; F. Fiori; V. S. Komlev; Nereo Bresolin; Franco Rustichelli

In the perspective of clinical translation of stem cell research, it would be advantageous to develop new techniques to detect donor cells after transplantation to track their fate and thus better understand their role in regeneration of damaged and diseased tissues. In this study we use X‐ray computed microtomography for three‐dimensional visualization of stem cells that were labeled with magnetic nanoparticles and transplanted via intra‐arterial infusion. We show that X‐ray computed microtomography offers the possibility to detect with high definition and resolution human cells after transplantation, and opens new possibilities for both experimental stem cell research.


PLOS ONE | 2008

Correlation of circulating CD133+ progenitor subclasses with a mild phenotype in Duchenne muscular dystrophy patients.

C. Marchesi; Marzia Belicchi; Mirella Meregalli; Andrea Farini; Alessandra Cattaneo; Daniele Parolini; Manuela Gavina; Laura Porretti; Maria Grazia D'Angelo; Nereo Bresolin; Giulio Cossu; Yvan Torrente

Background Various prognostic serum and cellular markers have been identified for many diseases, such as cardiovascular diseases and tumor pathologies. Here we assessed whether the levels of certain stem cells may predict the progression of Duchenne muscular dystrophy (DMD). Methods and Findings The levels of several subpopulations of circulating stem cells expressing the CD133 antigen were determined by flow cytometry in 70 DMD patients. The correlation between the levels and clinical status was assessed by statistical analysis. The median (±SD) age of the population was 10.66±3.81 (range 3 to 20 years). The levels of CD133+CXCR4+CD34- stem cells were significantly higher in DMD patients compared to healthy controls (mean±standard deviation: 17.38±1.38 vs. 11.0±1.70; P = 0.03) with a tendency towards decreased levels in older patients. Moreover, the levels of this subpopulation of cells correlated with the clinical condition. In a subgroup of 19 DMD patients after 24 months of follow-up, increased levels of CD133+CXCR4+CD34- cells was shown to be associated with a phenotype characterised by slower disease progression. The circulating CD133+CXCR4+CD34- cells in patients from different ages did not exhibit significant differences in their myogenic and endothelial in vitro differentiation capacity. Conclusions Our results suggest that levels of CD133+CXCR4+CD34- could function as a new prognostic clinical marker for the progression of DMD.


Journal of Cell Biology | 2006

Correction 174, p. 231

Beatriz G. Gálvez; Maurilio Sampaolesi; Silvia Brunelli; Diego Covarello; Manuela Gavina; Barbara Rossi; Gabriela Constantin; Yvan Torrente; Giulio Cossu

Vol. 174 No. 2, July 17, 2006. Pages [231–243][1]. In the original version of this article, an authors name was misspelled. The correct spelling is Gabriela Constantin. The name has been corrected in the online version of the article. [1]: /lookup/volpage/174/231?iss=2

Collaboration


Dive into the Manuela Gavina's collaboration.

Top Co-Authors

Avatar

Yvan Torrente

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Marzia Belicchi

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
Top Co-Authors

Avatar

Andrea Farini

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maurilio Sampaolesi

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar

Barbara Rossi

Elettra Sincrotrone Trieste

View shared research outputs
Researchain Logo
Decentralizing Knowledge