Network


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

Hotspot


Dive into the research topics where Rossana Tonlorenzi is active.

Publication


Featured researches published by Rossana Tonlorenzi.


Nature | 2006

Mesoangioblast stem cells ameliorate muscle function in dystrophic dogs.

Maurilio Sampaolesi; Stéphane Blot; Giuseppe D'Antona; Nicolas Granger; Rossana Tonlorenzi; Anna Innocenzi; Paolo Mognol; Jean-Laurent Thibaud; Beatriz G. Gálvez; Inès Barthélémy; Laura Perani; Sara Mantero; Maria Guttinger; Orietta Pansarasa; Chiara Rinaldi; M. Gabriella Cusella De Angelis; Yvan Torrente; Claudio Bordignon; Roberto Bottinelli; Giulio Cossu

Duchenne muscular dystrophy remains an untreatable genetic disease that severely limits motility and life expectancy in affected children. The only animal model specifically reproducing the alterations in the dystrophin gene and the full spectrum of human pathology is the golden retriever dog model. Affected animals present a single mutation in intron 6, resulting in complete absence of the dystrophin protein, and early and severe muscle degeneration with nearly complete loss of motility and walking ability. Death usually occurs at about 1 year of age as a result of failure of respiratory muscles. Here we report that intra-arterial delivery of wild-type canine mesoangioblasts (vessel-associated stem cells) results in an extensive recovery of dystrophin expression, normal muscle morphology and function (confirmed by measurement of contraction force on single fibres). The outcome is a remarkable clinical amelioration and preservation of active motility. These data qualify mesoangioblasts as candidates for future stem cell therapy for Duchenne patients.


Journal of Cell Biology | 2004

Extracellular HMGB1, a signal of tissue damage, induces mesoangioblast migration and proliferation

Roberta Palumbo; Maurilio Sampaolesi; Francesco De Marchis; Rossana Tonlorenzi; Sara Colombetti; Anna Mondino; Giulio Cossu; Marco Bianchi

High mobility group box 1 (HMGB1) is an abundant chromatin protein that acts as a cytokine when released in the extracellular milieu by necrotic and inflammatory cells. Here, we show that extracellular HMGB1 and its receptor for advanced glycation end products (RAGE) induce both migration and proliferation of vessel-associated stem cells (mesoangioblasts), and thus may play a role in muscle tissue regeneration. In vitro, HMGB1 induces migration and proliferation of both adult and embryonic mesoangioblasts, and disrupts the barrier function of endothelial monolayers. In living mice, mesoangioblasts injected into the femoral artery migrate close to HMGB1-loaded heparin-Sepharose beads implanted in healthy muscle, but are unresponsive to control beads. Interestingly, α-sarcoglycan null dystrophic muscle contains elevated levels of HMGB1; however, mesoangioblasts migrate into dystrophic muscle even if their RAGE receptor is disabled. This implies that the HMGB1–RAGE interaction is sufficient, but not necessary, for mesoangioblast homing; a different pathway might coexist. Although the role of endogenous HMGB1 in the reconstruction of dystrophic muscle remains to be clarified, injected HMGB1 may be used to promote tissue regeneration.


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.


Science Translational Medicine | 2012

Transplantation of Genetically Corrected Human iPSC-Derived Progenitors in Mice with Limb-Girdle Muscular Dystrophy

Francesco Saverio Tedesco; Mattia F M Gerli; Laura Perani; Sara Benedetti; Federica Ungaro; Marco Cassano; Stefania Antonini; Enrico Tagliafico; Valentina Artusi; Emanuela Longa; Rossana Tonlorenzi; Martina Ragazzi; Giorgia Calderazzi; Hidetoshi Hoshiya; Ornella Cappellari; Marina Mora; Benedikt Schoser; Peter Schneiderat; Mitsuo Oshimura; Roberto Bottinelli; Maurilio Sampaolesi; Yvan Torrente; Vania Broccoli; Giulio Cossu

Genetically corrected mesoangioblasts from human iPSCs derived from limb-girdle muscular dystrophy patients produce muscle fibers expressing the therapeutic gene in a mouse model of the disease. Muscle Progenitors Find Their Way Home Muscular dystrophies are genetic disorders primarily affecting skeletal muscle that result in greatly impaired mobility and, in severe cases, respiratory and cardiac dysfunction. There is no effective treatment, although several new approaches are entering clinical testing including cell therapy. Cell therapy aims to replace lost muscle fibers by transplanting healthy donor muscle progenitor cells or cells from dystrophic patients that have been genetically corrected in vitro. Mesoangioblasts are progenitor cells from blood vessel walls that have shown potential as a cell therapy in animal models of muscular dystrophy. In a new study, Tedesco et al. explore whether genetically corrected mesoangioblasts from patients with limb-girdle muscular dystrophy 2D (LGMD2D) have potential as an autologous cell therapy to treat this disease. The authors quickly found that they could not derive a sufficient number of mesoangioblasts from LGMD2D patients because the muscles of the patients were depleted of these progenitor cells. To overcome this problem, the authors reprogrammed fibroblasts or myoblasts from the LGMD2D patients to obtain human induced pluripotent stem cells (iPSCs) and induced them to differentiate into mesoangioblast-like cells that were then genetically corrected in vitro using a viral vector expressing the defective gene SGCA, which encodes α-sarcoglycan. After intramuscular or intra-arterial injection of these genetically corrected, iPSC-derived mesoangioblasts into mice with LGMD2D (immune-deficient Sgca-null mice), the cells homed to damaged mouse skeletal muscle, engrafted, and formed muscle fibers expressing α-sarcoglycan. Using mouse iPSC-derived mesoangioblasts, the researchers showed that the transplanted engrafted cells imbued muscle with greater strength and enabled the dystrophic mice to run for longer on a treadmill than dystrophic mice that did not receive the cells. This strategy offers the advantage of being able to produce unlimited numbers of genetically corrected progenitor cells, which perhaps could be used in the future as cell therapy for treating LGMD2D and other forms of muscular dystrophy. Mesoangioblasts are stem/progenitor cells derived from a subset of pericytes found in muscle that express alkaline phosphatase. They have been shown to ameliorate the disease phenotypes of different animal models of muscular dystrophy and are now undergoing clinical testing in children affected by Duchenne’s muscular dystrophy. Here, we show that patients with a related disease, limb-girdle muscular dystrophy 2D (LGMD2D), which is caused by mutations in the gene encoding α-sarcoglycan, have reduced numbers of this pericyte subset and thus produce too few mesoangioblasts for use in autologous cell therapy. Hence, we reprogrammed fibroblasts and myoblasts from LGMD2D patients to generate human induced pluripotent stem cells (iPSCs) and developed a protocol for the derivation of mesoangioblast-like cells from these iPSCs. The iPSC-derived mesoangioblasts were expanded and genetically corrected in vitro with a lentiviral vector carrying the gene encoding human α-sarcoglycan and a promoter that would ensure expression only in striated muscle. When these genetically corrected human iPSC-derived mesoangioblasts were transplanted into α-sarcoglycan–null immunodeficient mice, they generated muscle fibers that expressed α-sarcoglycan. Finally, transplantation of mouse iPSC-derived mesoangioblasts into α-sarcoglycan–null immunodeficient mice resulted in functional amelioration of the dystrophic phenotype and restoration of the depleted progenitors. These findings suggest that transplantation of genetically corrected mesoangioblast-like cells generated from iPSCs from LGMD2D patients may be useful for treating this type of muscular dystrophy and perhaps other forms of muscular dystrophy as well.


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 Leukocyte Biology | 2009

Inflammatory and alternatively activated human macrophages attract vessel-associated stem cells, relying on separate HMGB1- and MMP-9-dependent pathways.

Karine Lolmede; Lara Campana; Michela Vezzoli; Lidia Bosurgi; Rossana Tonlorenzi; Emilio Clementi; Marco Bianchi; Giulio Cossu; Angelo A. Manfredi; Silvia Brunelli; Patrizia Rovere-Querini

Inflammatory macrophages recruited at the site of damaged muscles progressively acquire an alternative activation profile. Inflammatory (M1) and alternatively activated (M2) macrophages exert various and even opposite functions. M1 cells amplify tissue damage, and M2 cells dispose of necrotic fibers and deliver survival signals to myogenic precursors, finally supporting healing. A critical step in muscle healing is the recruitment of myogenic stem cells, including vessel‐associated stem cells (mesoangioblasts), which have been demonstrated to home to damaged skeletal muscle selectively and preferentially. Little information is available about the signals involved and the role played by infiltrating macrophages. Here, we report that the polarization of macrophages dramatically skews the secretion of high mobility group box 1 (HMGB1), TNF‐α, vascular endothelial growth factor, and metalloproteinase 9 (MMP‐9), molecules involved in the regulation of cell diapedesis and migration. All polarized macrophage populations were strikingly effective at inducing mesoangioblast migration. By means of specific inhibitors, we verified that the recruitment of mesoangioblasts requires the secretion of HMGB1 and TNF‐α by M1 cells and of MMP‐9 by M2 cells. Together, these data demonstrate a feature, unrecognized previously, of macrophages: their ability to attract stem cells, which is conserved throughout their polarization. Moreover, they open the possibility of novel strategies, aimed at interfering selectively with signals that recruit blood‐derived stem cells toward pro‐ or anti‐inflammatory macrophages.


The EMBO Journal | 1997

Rox, a novel bHLHZip protein expressed in quiescent cells that heterodimerizes with Max, binds a non‐canonical E box and acts as a transcriptional repressor

Germana Meroni; Alexandre Reymond; Myriam Alcalay; Giuseppe Borsani; Akira Tanigami; Rossana Tonlorenzi; Cristiana Lo Nigro; Silvia Messali; Massimo Zollo; David H. Ledbetter; Roger Brent; Andrea Ballabio; Romeo Carrozzo

Proteins of the Myc and Mad family are involved in transcriptional regulation and mediate cell differentiation and proliferation. These molecules share a basic‐helix–loop–helix leucine zipper domain (bHLHZip) and bind DNA at the E box (CANNTG) consensus by forming heterodimers with Max. We report the isolation, characterization and mapping of a human gene and its mouse homolog encoding a new member of this family of proteins, named Rox. Through interaction mating and immunoprecipitation techniques, we demonstrate that Rox heterodimerizes with Max and weakly homodimerizes. Interestingly, bandshift assays demonstrate that the Rox–Max heterodimer shows a novel DNA binding specificity, having a higher affinity for the CACGCG site compared with the canonical E box CACGTG site. Transcriptional studies indicate that Rox represses transcription in both human HEK293 cells and yeast. We demonstrate that repression in yeast is through interaction between the N–terminus of the protein and the Sin3 co‐repressor, as previously shown for the other Mad family members. ROX is highly expressed in quiescent fibroblasts and expression markedly decreases when cells enter the cell cycle. Moreover, ROX expression appears to be induced in U937 myeloid leukemia cells stimulated to differentiate with 12‐O‐tetradecanoylphorbol‐13‐acetate. The identification of a novel Max‐interacting protein adds an important piece to the puzzle of Myc/Max/Mad coordinated action and function in normal and pathological situations. Furthermore, mapping of the human gene to chromosome 17p13.3 in a region that frequently undergoes loss of heterozygosity in a number of malignancies, together with the biochemical and expression features, suggest involvement of ROX in human neoplasia.


Science Translational Medicine | 2011

Stem Cell–Mediated Transfer of a Human Artificial Chromosome Ameliorates Muscular Dystrophy

Francesco Saverio Tedesco; Hidetoshi Hoshiya; Giuseppe D'Antona; Mattia F M Gerli; Graziella Messina; Stefania Antonini; Rossana Tonlorenzi; Sara Benedetti; Libera Berghella; Yvan Torrente; Yasuhiro Kazuki; Roberto Bottinelli; Mitsuo Oshimura; Giulio Cossu

Combining gene delivery using a human artificial chromosome with stem cell transplantation ameliorates muscular dystrophy in a mouse model. Stem Cells Muscle in on the Action The progressive muscle loss that is the hallmark of Duchenne muscular dystrophy (DMD) has proved very difficult to halt or reverse. Although the causative mutations of DMD were identified in the X-linked gene encoding dystrophin (a structural muscle protein) several decades ago, translating this genetic discovery into new treatments has been challenging. Most therapeutic strategies aim to use gene therapy to deliver the normal dystrophin gene to the dystrophic muscles of DMD patients. However, the dystrophin gene is too large to be carried by the viral vectors usually used in gene therapy and all muscles in the body would have to be injected with the vector and replacement gene. Now, Tedesco and colleagues combine stem cell therapy with a human artificial chromosome vector to overcome these two challenges in the mdx mouse model of DMD. This team had previously identified a blood vessel stem cell called “mesoangioblast” that has the dual talents of being able to cross blood vessel walls and to differentiate into a variety of mesodermal cell types including muscle cells. Would these stem cells be able to deliver a replacement dystrophin gene to dystrophic muscles in the mdx mouse? Predicting that they would, Tedesco and colleagues used a human artificial chromosome vector engineered to carry the entire normal human dystrophin gene including the regulatory regions. They transferred the vector and its large cargo into mesoangioblasts isolated from mdx mice; then they injected the corrected mesoangioblasts directly into the dystrophic skeletal muscles of recipient immune-deficient mdx mice (to prevent reaction against the human protein). The authors showed that the transplanted mesoangioblasts were able to engraft in dystrophic muscles, express normal dystrophin, and produce functional muscle fibers with amelioration of dystrophic pathology. They also found that the transplanted mesoangioblasts contributed to the muscle satellite cell pool, which produces new muscle cells under normal conditions. Next, the authors showed that if they injected the corrected mesoangioblasts into the arterial circulation of mdx mice, the cells were able to cross blood vessel walls, home to dystrophic muscles and graft contribute to the formation of new dystrophin-expressing myofibers. The authors then showed that mice receiving the mesoangioblast transplants showed reduced fiber fragility, increased force, and greater motor capacity on treadmill and freewheel tests. Although there are still technical and regulatory hurdles to be overcome before this strategy can be used in DMD patients, stem cell–mediated transfer of the normal dystrophin gene using a human artificial chromosome shows promise as a treatment for this tragic and ultimately fatal disease. In contrast to conventional gene therapy vectors, human artificial chromosomes (HACs) are episomal vectors that can carry large regions of the genome containing regulatory elements. So far, HACs have not been used as vectors in gene therapy for treating genetic disorders. Here, we report the amelioration of the dystrophic phenotype in the mdx mouse model of Duchenne muscular dystrophy (DMD) using a combination of HAC-mediated gene replacement and transplantation with blood vessel–associated stem cells (mesoangioblasts). We first genetically corrected mesoangioblasts from dystrophic mdx mice with a HAC vector containing the entire (2.4 Mb) human dystrophin genetic locus. Genetically corrected mesoangioblasts engrafted robustly and gave rise to many dystrophin-positive muscle fibers and muscle satellite cells in dystrophic mice, leading to morphological and functional amelioration of the phenotype that lasted for up to 8 months after transplantation. Thus, HAC-mediated gene transfer shows efficacy in a preclinical model of DMD and offers potential for future clinical translation.


Circulation Research | 2004

Msx2 and Necdin Combined Activities Are Required for Smooth Muscle Differentiation in Mesoangioblast Stem Cells

Silvia Brunelli; Enrico Tagliafico; Fernanda Gabriella De Angelis; Rossana Tonlorenzi; Silvia Baesso; Sergio Ferrari; Michio Niinobe; Kazuaki Yoshikawa; Robert J. Schwartz; Irene Bozzoni; Stefano Ferrari; Giulio Cossu

Little is known about the molecular mechanism underlying specification and differentiation of smooth muscle (SM), and this is, at least in part, because of the few cellular systems available to study the acquisition of a SM phenotype in vitro. Mesoangioblasts are vessel-derived stem cells that can be induced to differentiate into different cell types of the mesoderm, including SM. We performed a DNA microarray analysis of a mesoangioblast clone that spontaneously expresses an immature SM phenotype and compared it with a sister clone mainly composed of undifferentiated progenitor cells. This study allowed us to define a gene expression profile for “stem” cells versus smooth muscle cells (SMCs) in the absence of differentiation inducers such as transforming growth factor β. Two transcription factors, msx2 and necdin, are expressed at least 100 times more in SMCs than in stem cells, are coexpressed in all SMCs and tissues, are induced by transforming growth factor β, and, when coexpressed, induce a number of SM markers in mesoangioblast, fibroblast, and endothelial cell lines. Conversely, their downregulation through RNA interference results in a decreased expression of SM markers. These data support the hypothesis that Msx2 and necdin act as master genes regulating SM differentiation in at least a subset of SMCs.


Current protocols in stem cell biology | 2007

Isolation and Characterization of Mesoangioblasts from Mouse, Dog, and Human Tissues

Rossana Tonlorenzi; Arianna Dellavalle; Esther Schnapp; Giulio Cossu; Maurilio Sampaolesi

Mesoangioblasts are recently identified stem/progenitor cells, associated with small vessels of the mesoderm in mammals. Originally described in the mouse embryonic dorsal aorta, similar though not identical cells have been later identified and characterized from postnatal small vessels of skeletal muscle and heart (not described in this unit). They have in common the anatomical location, the expression of endothelial and/or pericyte markers, the ability to proliferate in culture, and the ability to undergo differentiation into various types of mesoderm cells upon proper culture conditions. Currently, the developmental origin of mesoangioblasts, their phenotypic heterogeneity, and the relationship with other mesoderm stem cells are not understood in detail and are the subject of active research. However, from a practical point of view, these cells have been successfully used in cell transplantation protocols that have yielded a significant rescue of structure and function in skeletal muscle of dystrophic mice and dogs. Since the corresponding human cells have been recently isolated and characterized, a clinical trial with these cells is planned in the near future. This unit provides detailed methods for isolation, culture, and characterization of mesoangioblasts.

Collaboration


Dive into the Rossana Tonlorenzi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yvan Torrente

Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico

View shared research outputs
Top Co-Authors

Avatar

Maurilio Sampaolesi

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Innocenzi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Laura Perani

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge