Network


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

Hotspot


Dive into the research topics where Federica Casiraghi is active.

Publication


Featured researches published by Federica Casiraghi.


Journal of Immunology | 2008

Pretransplant Infusion of Mesenchymal Stem Cells Prolongs the Survival of a Semiallogeneic Heart Transplant through the Generation of Regulatory T Cells

Federica Casiraghi; Nadia Azzollini; Paola Cassis; Barbara Imberti; Marina Morigi; Daniela Cugini; Regiane Aparecida Cavinato; Marta Todeschini; Samantha Solini; Aurelio Sonzogni; Norberto Perico; Giuseppe Remuzzi; Marina Noris

In this study, we investigated whether mesenchymal stem cells (MSC) had immunomodulatory properties in solid organ allotransplantation, using a semiallogeneic heart transplant mouse model, and studied the mechanism(s) underlying MSC tolerogenic effects. Either single (portal vein, day −7) or double (portal vein, day −7 and tail vein, day −1) pretransplant infusions of donor-derived B6C3 MSC in B6 recipients induced a profound T cell hyporesponsiveness and prolonged B6C3 cardiac allograft survival. The protolerogenic effect was abrogated when donor-derived MSC were injected together with B6C3 hematopoietic stem cells (HSC), suggesting that HSC negatively impact MSC immunomodulatory properties. Both the induction (pretransplant) and the maintenance phase (>100 days posttransplant) of donor-derived MSC-induced tolerance were associated with CD4+CD25+Foxp3+ Treg expansion and impaired anti-donor Th1 activity. MSC-induced regulatory T cells (Treg) were donor-specific since adoptive transfer of splenocytes from tolerant mice prevented the rejection of fully MHC-mismatched donor-specific secondary allografts but not of third-party grafts. In addition, infusion of recipient-derived B6 MSC tolerized a semiallogeneic B6C3 cardiac allograft, but not a fully MHC-mismatched BALB/c graft, and expanded Treg. A double i.v. pretransplant infusion of recipient-derived MSC had the same tolerogenic effect as the combined intraportal/i.v. MSC infusions, which makes the tolerogenic protocol applicable in a clinical setting. In contrast, single MSC infusions given either peritransplant or 1 day after transplant were less effective. Altogether these findings indicate that MSC immunomodulatory properties require HSC removal, partial sharing of MHC Ags between the donor and the recipient and pretransplant infusion, and are associated with expansion of donor-specific Treg.


Clinical Journal of The American Society of Nephrology | 2011

Autologous Mesenchymal Stromal Cells and Kidney Transplantation: A Pilot Study of Safety and Clinical Feasibility

Norberto Perico; Federica Casiraghi; Martino Introna; Eliana Gotti; Marta Todeschini; Regiane Aparecida Cavinato; Chiara Capelli; Alessandro Rambaldi; Paola Cassis; Paola Rizzo; Monica Cortinovis; Maddalena Marasà; Josée Golay; Marina Noris; Giuseppe Remuzzi

BACKGROUND AND OBJECTIVES Mesenchymal stromal cells (MSCs) abrogate alloimmune response in vitro, suggesting a novel cell-based approach in transplantation. Moving this concept toward clinical application in organ transplantation should be critically assessed. DESIGN, SETTING, PARTICIPANTS & MEASUREMENTS A safety and clinical feasibility study (ClinicalTrials.gov, NCT00752479) of autologous MSC infusion was conducted in two recipients of kidneys from living-related donors. Patients were given T cell-depleting induction therapy and maintenance immunosuppression with cyclosporine and mycophenolate mofetil. On day 7 posttransplant, MSCs were administered intravenously. Clinical and immunomonitoring of MSC-treated patients was performed up to day 360 postsurgery. RESULTS Serum creatinine levels increased 7 to 14 days after cell infusion in both MSC-treated patients. A graft biopsy in patient 2 excluded acute graft rejection, but showed a focal inflammatory infiltrate, mostly granulocytes. In patient 1 protocol biopsy at 1-year posttransplant showed a normal graft. Both MSC-treated patients are in good health with stable graft function. A progressive increase of the percentage of CD4+CD25highFoxP3+CD127- Treg and a marked inhibition of memory CD45RO+RA-CD8+ T cell expansion were observed posttransplant. Patient T cells showed a profound reduction of CD8+ T cell activity. CONCLUSIONS Findings from this study in the two patients show that MSC infusion in kidney transplant recipients is feasible, allows enlargement of Treg in the peripheral blood, and controls memory CD8+ T cell function. Future clinical trials with MSCs to look with the greatest care for unwanted side effects is advised.


American Journal of Transplantation | 2012

Localization of Mesenchymal Stromal Cells Dictates Their Immune or Proinflammatory Effects in Kidney Transplantation

Federica Casiraghi; Nadia Azzollini; Marta Todeschini; Regiane Aparecida Cavinato; Paola Cassis; Samantha Solini; Cinzia Rota; Marina Morigi; M. Introna; R. Maranta; Norberto Perico; G. Remuzzi; Marina Noris

Multipotent mesenchymal stromal cells (MSC) have recently emerged as promising candidates for cell‐based immunotherapy in solid‐organ transplantation. However, optimal conditions and settings for fully harnessing MSC tolerogenic properties need to be defined. We recently reported that autologous MSC given posttransplant in kidney transplant patients was associated with transient renal insufficiency associated with intragraft recruitment of neutrophils and complement C3 deposition. Here, we moved back to a murine kidney transplant model with the aim to define the best timing of MSC infusion capable of promoting immune tolerance without negative effects on early graft function. We also investigated the mechanisms of the immunomodulatory and/or proinflammatory activities of MSC according to whether cells were given before or after transplant. Posttransplant MSC infusion in mice caused premature graft dysfunction and failed to prolong graft survival. In this setting, infused MSC localized mainly into the graft and associated with neutrophils and complement C3 deposition. By contrast, pretransplant MSC infusion induced a significant prolongation of kidney graft survival by a Treg‐dependent mechanism. MSC‐infused pretransplant localized into lymphoid organs where they promoted early expansion of Tregs. Thus, pretransplant MSC infusion may be a useful approach to fully exploit their immunomodulatory properties in kidney transplantation.


Transplant International | 2013

Mesenchymal stromal cells and kidney transplantation: Pretransplant infusion protects from graft dysfunction while fostering immunoregulation

Norberto Perico; Federica Casiraghi; Eliana Gotti; Martino Introna; Marta Todeschini; Regiane Aparecida Cavinato; Chiara Capelli; Alessandro Rambaldi; Paola Cassis; Paola Rizzo; Monica Cortinovis; Marina Noris; Giuseppe Remuzzi

Bone marrow‐derived mesenchymal stromal cells (MSC) have emerged as useful cell population for immunomodulation therapy in transplantation. Moving this concept towards clinical application, however, should be critically assessed by a tailor‐made step‐wise approach. Here, we report results of the second step of the multistep MSC‐based clinical protocol in kidney transplantation. We examined in two living‐related kidney transplant recipients whether: (i) pre‐transplant (DAY‐1) infusion of autologous MSC protected from the development of acute graft dysfunction previously reported in patients given MSC post‐transplant, (ii) avoiding basiliximab in the induction regimen improved the MSC‐induced Treg expansion previously reported with therapy including this anti‐CD25‐antibody. In patient 3, MSC treatment was uneventful and graft function remained normal during 1 year follow‐up. In patient 4, acute cellular rejection occurred 2 weeks post‐transplant. Both patients had excellent graft function at the last observation. Circulating memory CD8+ T cells and donor‐specific CD8+ T‐cell cytolytic response were reduced in MSC‐treated patients, not in transplant controls not given MSC. CD4+FoxP3+Treg expansion was comparable in MSC‐treated patients with or without basiliximab induction. Thus, pre‐transplant MSC no longer negatively affect kidney graft at least to the point of impairing graft function, and maintained MSC‐immunomodulatory properties. Induction therapy without basiliximab does not offer any advantage on CD4+FoxP3+Treg expansion (ClinicalTrials.gov number: NCT 00752479).


Transplantation | 2007

Sirolimus versus cyclosporine therapy increases circulating regulatory T cells, but does not protect renal transplant patients given alemtuzumab induction from chronic allograft injury.

Piero Ruggenenti; Norberto Perico; Eliana Gotti; Paolo Cravedi; Elena Gagliardini; Mauro Abbate; Flavio Gaspari; Dario Cattaneo; Marina Noris; Federica Casiraghi; Marta Todeschini; Daniela Cugini; Sara Conti; Giuseppe Remuzzi

Background. In kidney transplant recipients with alemtuzumab induction maintained on mycophenolate mofetil (MMF) immunosuppression, sirolimus (SRL) promotes significant expansion of circulating CD4+CD25high regulatory T cells (Treg). This might translate into more effective protection against chronic graft injury compared to cyclosporin A (CsA), which, in the same clinical setting, does not affect Treg. Methods. To assess this hypothesis, in the extension of a single-center, prospective, randomized, open, blind endpoint study aimed to assess the effect of low-dose SRL or CsA on circulating Treg, we compared the outcomes of renal transplant recipients on SRL (n=11) or CsA (n=10) by per-protocol biopsies and serial measurements of glomerular filtration rate (GFR), renal plasma flow (RPF), and 24-hour proteinuria over 30 months posttransplant. Results. Despite 4-fold higher CD4+CD25high Treg counts (22.1±12.2% vs. 5.7±4.2% of CD3+CD4+ T cells), SRL-treated patients, compared to CsA-treated patients, had a significantly higher tubular C4d staining score (1.1±0.6 vs. 0.2±0.3, P<0.01), with nonsignificant trends to higher chronic allograft damage index score (5.6±2.4 vs. 3.7±3.3), faster GFR (−2.92±0.33 vs. −0.28±0.44 ml/min/1.73m2 per year), and RPF (−10.80±5.45 vs. −1.86±3.09 ml/min/1.73 m2 per year) decline, and more clinical proteinuria (n=6 vs. 4). There was no significant correlation between Treg counts and any considered outcome variable in the study group as a whole and within each cohort. Conclusions. These data suggest that, despite enhanced Treg expression, low-dose SRL combined to alemtuzumab induction and MMF-based steroid-free maintenance therapy, does not appreciably protect renal transplant recipients from chronic allograft injury and dysfunction.


Stem Cell Reviews and Reports | 2013

Multipotent Mesenchymal Stromal Cell Therapy and Risk of Malignancies

Federica Casiraghi; Giuseppe Remuzzi; Mauro Abbate; Norberto Perico

Cell therapy with Multipotent Mesenchymal Stromal Cells (MSC) holds enormous promise for the treatment of a large number of degenerative and immune/inflammatory diseases. Their multilineage differentiation potential, immunoprivilege and capacity of promoting recovery of damaged tissues coupled with anti-inflammatory and immunosuppressive properties are the focus of a multitude of clinical studies currently underway. The recognized clinical potential of MSC repairing/immunomodulatory effects now encompasses graft-versus-host disease, hematologic malignancies, cardiovascular diseases, neurologic and inherited diseases, autoimmune diseases, organ transplantation, refractory wounds, and bone/cartilage defects among others. However, it has been suggested that both the need of extensive ex vivo culture for MSC clinical use, and their proangiogenic, anti-apoptotic and immunomodulatory properties may act together as tumor promoters, raising significant safety concerns. This paper will review the available data on in vitro MSC maldifferentiation and the ability of MSC to sustain tumor growth in vivo, with the aim to clarify whether MSC-based therapeutic approaches may carry actual risk of malignancies.


Thrombosis and Haemostasis | 2005

Complement activation: the missing link between ADAMTS-13 deficiency and microvascular thrombosis of thrombotic microangiopathies.

María P. Ruiz-Torres; Federica Casiraghi; Miriam Galbusera; Daniela Macconi; Sara Gastoldi; Marta Todeschini; Francesca Porrati; Daniela Belotti; Enrico Maria Pogliani; Marina Noris; Giuseppe Remuzzi

Endothelial injury is the central factor in the events leading to thrombotic microangiopathy (TMA); however, the mechanisms involved are not fully understood. Here we investigate the role of neutrophils (PMNs) and of complement activation in inducing microvascular damage and loss of thromboresistance in TMA associated with ADAMTS-13 deficiency. PMNs isolated during the acute phase of the disease released excessive amounts of reactive-oxygen species (ROS), N-derived oxidants and proteinases and induced damage and thromboresistance loss in human microvascular endothelial cell line (HMEC-1) ex vivo. Endothelial cytotoxicity and thromboresistance loss was also induced by TMA serum. Complement-derived products were responsible for the above effects: in fact, TMA serum caused C3 and Membrane Attack Complex (MAC) deposition on HMEC-1 and its cytotoxic effect was abolished by complement inhibition. TMA serum caused surface expression of P-selectin on HMEC-1 which may promote PMN adhesion and resulted in increased PMN cytotoxicity, indicating that complement may have a role in PMN activation. In addition, TMA serum stimulated control PMNs to release ROS and proteinases, and to cause endothelial cell cytotoxicity. All of the above effects were abrogated by complement inactivation. These data document for the first time that complement-initiated PMN activation and endothelial injury may have a crucial role in microvascular thrombosis of TMA associated with ADAMTS-13 deficiency.


Journal of Immunology | 2000

Thymic Dendritic Cells Express Inducible Nitric Oxide Synthase and Generate Nitric Oxide in Response to Self- and Alloantigens

Sistiana Aiello; Marina Noris; Giampiero Piccinini; Susanna Tomasoni; Federica Casiraghi; Samantha Bonazzola; Marilena Mister; Mohamed H. Sayegh; Giuseppe Remuzzi

Thymocytes maturing in the thymus undergo clonal deletion/apoptosis when they encounter self- or allo-Ags presented by dendritic cells (DCs). How this occurs is a matter of debate, but NO may play a role given its ability of inducing apoptosis of these cells. APC (a mixed population of macrophages (Mφ) and DCs) from rat thymus expressed high levels of inducible NO synthase (iNOS) and produced large amounts of NO in basal conditions whereas iNOS expression and NO production were very low in thymocytes. Analysis by FACS and by double labeling of cytocentrifuged preparations showed that DCs and MΦ both express iNOS within APC. Analysis of a purified preparation of DCs confirmed that these cells express high levels of iNOS and produce large amounts of NO in basal conditions. The capacity of DCs to generate NO was enhanced by exposure to rat albumin, a self-protein, and required a fully expressed process of Ag internalization, processing, and presentation. Peptides derived from portions of class II MHC molecules up-regulate iNOS expression and NO production by DCs as well, both in self and allogeneic combinations, suggesting a role of NO in both self and acquired tolerance. We also found that NO induced apoptosis of rat double-positive thymocytes, the effect being more evident in anti-CD3-stimulated cells. Altogether, the present findings might suggest that DC-derived NO is at least one of the soluble factors regulating events, in the thymus, that follow recognition of self- and allo-Ags.


Transplantation | 2009

Toward MSC in Solid Organ Transplantation: 2008 Position Paper of the MISOT Study Group

Marc H. Dahlke; Martin J. Hoogduijn; Elke Eggenhofer; Felix C. Popp; Philipp Renner; Przemyslaw Slowik; Agnes Rosenauer; Pompiliu Piso; Edward K. Geissler; Claudia Lange; Dominique Chabannes; Benedetta Mazzanti; Sinda Bigenzahn; Patrick Bertolino; Uta Kunter; Martino Introna; Alessandro Rambaldi; Chiara Capelli; Norberto Perico; Federica Casiraghi; Marina Noris; Eliana Gotti; Martina Seifert; Riccardo Saccardi; Hein W. Verspaget; Bart van Hoek; Amelia Bartholomew; Thomas Wekerle; H.-D. Volk; Giuseppe Remuzzi

The following position paper summarizes the recommendations for early clinical trials and ongoing basic research in the field of mesenchymal stem cell-induced solid organ graft acceptance—agreed upon on the first meeting of the Mesenchymal Stem Cells In Solid Organ Transplantation (MISOT) study group in late 2008.


Transplantation | 2013

Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Fourth Meeting: Lessons Learned from First Clinical Trials

Marcella Franquesa; Martin J. Hoogduijn; Marlies E.J. Reinders; Elke Eggenhofer; Anja U. Engela; F. Mensah; Joan Torras; Antonello Pileggi; Cees van Kooten; Bernard P. Mahon; Olivier Detry; Felix C. Popp; Volker Benseler; Federica Casiraghi; Christian L. Johnson; Janis Ancans; Barbara Fillenberg; Olga DelaRosa; Josep M. Aran; Marieke Roemeling-Vanrhijn; Jef Pinxteren; Norberto Perico; Eliana Gotti; B Christ; James L. Reading; Martino Introna; Robert Deans; Murat Shagidulin; Ramon Farre; Alessandro Rambaldi

The Fourth Expert Meeting of the Mesenchymal Stem Cells in Solid Organ Transplantation (MiSOT) Consortium took place in Barcelona on October 19 and 20, 2012. This meeting focused on the translation of preclinical data into early clinical settings. This position paper highlights the main topics explored on the safety and efficacy of mesenchymal stem cells as a therapeutic agent in solid organ transplantation and emphasizes the issues (proper timing, concomitant immunossupression, source and immunogenicity of mesenchymal stem cells, and oncogenicity) that have been addressed and will be followed up by the MiSOT Consortium in future studies.

Collaboration


Dive into the Federica Casiraghi's collaboration.

Top Co-Authors

Avatar

Giuseppe Remuzzi

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Marina Noris

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Norberto Perico

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Marta Todeschini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Nadia Azzollini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Eliana Gotti

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Paola Cassis

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Daniela Cugini

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar

Marilena Mister

Mario Negri Institute for Pharmacological Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge