Network


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

Hotspot


Dive into the research topics where Laura Passerini is active.

Publication


Featured researches published by Laura Passerini.


Journal of Clinical Investigation | 2005

The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs

Sarah E. Allan; Laura Passerini; Rosa Bacchetta; Natasha K. Crellin; Minyue Dai; Paul C. Orban; Steven F. Ziegler; Maria Grazia Roncarolo; Megan K. Levings

Little is known about the molecules that control the development and function of CD4+ CD25+ Tregs. Recently, it was shown that the transcription factor FOXP3 is necessary and sufficient for the generation of CD4+ CD25+ Tregs in mice. We investigated the capacity of FOXP3 to drive the generation of suppressive CD4+ CD25+ Tregs in humans. Surprisingly, although ectopic expression of FOXP3 in human CD4+ T cells resulted in induction of hyporesponsiveness and suppression of IL-2 production, it did not lead to acquisition of significant suppressor activity in vitro. Similarly, ectopic expression of FOXP3delta2, an isoform found in human CD4+ CD25+ Tregs that lacks exon 2, also failed to induce the development of suppressor T cells. Moreover, when FOXP3 and FOXP3delta2 were simultaneously overexpressed, although the expression of several Treg-associated cell surface markers was significantly increased, only a modest suppressive activity was induced. These data indicate that in humans, overexpression of FOXP3 alone or together with FOXP3delta2 is not an effective method to generate potent suppressor T cells in vitro and suggest that factors in addition to FOXP3 are required during the process of activation and/or differentiation for the development of bona fide Tregs.


Frontiers in Immunology | 2012

Immune dysregulation, polyendocrinopathy, enteropathy, x-linked syndrome: a paradigm of immunodeficiency with autoimmunity.

Federica Barzaghi; Laura Passerini; Rosa Bacchetta

Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a rare monogenic primary immunodeficiency (PID) due to mutations of FOXP3, a key transcription factor for naturally occurring (n) regulatory T (Treg) cells. The dysfunction of Treg cells is the main pathogenic event leading to the multi-organ autoimmunity that characterizes IPEX syndrome, a paradigm of genetically determined PID with autoimmunity. IPEX has a severe early onset and can become rapidly fatal within the first year of life regardless of the type and site of the mutation. The initial presenting symptoms are severe enteritis and/or type-1 diabetes mellitus, alone or in combination with eczema and elevated serum IgE. Other autoimmune symptoms, such as hypothyroidism, cytopenia, hepatitis, nephropathy, arthritis, and alopecia can develop in patients who survive the initial acute phase. The current therapeutic options for IPEX patients are limited. Supportive and replacement therapies combined with pharmacological immunosuppression are required to control symptoms at onset. However, these procedures can allow only a reduction of the clinical manifestations without a permanent control of the disease. The only known effective cure for IPEX syndrome is hematopoietic stem cell transplantation, but it is always limited by the availability of a suitable donor and the lack of specific guidelines for bone marrow transplant in the context of this disease. This review aims to summarize the clinical histories and genomic mutations of the IPEX patients described in the literature to date. We will focus on the clinical and immunological features that allow differential diagnosis of IPEX syndrome and distinguish it from other PID with autoimmunity. The efficacy of the current therapies will be reviewed, and possible innovative approaches, based on the latest highlights of the pathogenesis to treat this severe primary autoimmune disease of childhood, will be discussed.


The Journal of Allergy and Clinical Immunology | 2008

Clinical and molecular profile of a new series of patients with immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome: Inconsistent correlation between forkhead box protein 3 expression and disease severity

Eleonora Gambineri; Lucia Perroni; Laura Passerini; Lucia Bianchi; Claudio Doglioni; Franco Meschi; Riccardo Bonfanti; Yves Sznajer; Alberto Tommasini; Anita Lawitschka; Anne K. Junker; Desiree Dunstheimer; Peter H. Heidemann; Giantonio Cazzola; Marco Cipolli; Wilhelm Friedrich; Dragana Janic; Nadira Azzi; Erick Richmond; Silvia Vignola; Arrigo Barabino; Giuseppe Chiumello; Chiara Azzari; Maria Grazia Roncarolo; Rosa Bacchetta

BACKGROUND Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is an autoimmune genetic disorder caused by mutation of the forkhead box protein 3 gene (FOXP3), a key regulator of immune tolerance. OBJECTIVE We sought to provide clinical and molecular indicators that facilitate the understanding and diagnosis of IPEX syndrome. METHODS In 14 unrelated affected male subjects who were given diagnoses of IPEX syndrome based on FOXP3 gene sequencing, we determined whether particular FOXP3 mutations affected FOXP3 protein expression and correlated the molecular and clinical data. RESULTS Molecular analysis of FOXP3 in the 14 subjects revealed 13 missense and splice-site mutations, including 7 novel mutations. Enteropathy, generally associated with endocrinopathy and eczema, was reported in all patients, particularly in those carrying mutations within FOXP3 functional domains or mutations that altered protein expression. However, similar genotypes did not always result in similar phenotypes in terms of disease presentation and severity. In addition, FOXP3 protein expression did not correlate with disease severity. CONCLUSION Severe autoimmune enteropathy, which is often associated with increased IgE levels and eosinophilia, is the most prominent early manifestation of IPEX syndrome. Nevertheless, the disease course is variable and somewhat unpredictable. Therefore genetic analysis of FOXP3 should always be performed to ensure an accurate diagnosis, and FOXP3 protein expression analysis should not be the only diagnostic tool for IPEX syndrome.


International Immunology | 2008

STAT5-signaling cytokines regulate the expression of FOXP3 in CD4+CD25+ regulatory T cells and CD4+CD25-effector T cells

Laura Passerini; Sarah E. Allan; Manuela Battaglia; Sara Di Nunzio; Alicia N. Alstad; Megan K. Levings; Maria Grazia Roncarolo; Rosa Bacchetta

Forkhead box P3 (FOXP3) is considered a specific marker for CD4(+)CD25(+) regulatory T (Treg) cells, but increasing evidence suggests that human CD4(+)CD25(-) effector T (Teff) cells can transiently express FOXP3 upon activation. We demonstrate that the signal transducer and activator of transcription 5 (STAT5)-signaling cytokines, IL-2, IL-15 and to a lesser extent IL-7, induce FOXP3 up-regulation in vitro in activated human Teff cells. In contrast, cytokines which do not activate STAT5, such as IL-4 or transforming growth factor-beta alone, do not directly induce FOXP3 expression in activated Teff cells. Moreover, expression of a constitutively active form of STAT5a is sufficient to induce FOXP3 expression in Teff cells. Expression of FOXP3 in activated Teff cells requires both TCR-mediated activation and endogenous IL-2, but is not dependent on cell division and does not induce suppressive function. The presence of STAT5-activating cytokines is also required to maintain high FOXP3 expression and suppressive activity of Treg cells in vitro. These data indicate that activation of STAT5 sustains FOXP3 expression in both Treg and Teff cells and contribute to our understanding of how cytokines affect the expression of FOXP3.


Blood | 2013

Accumulation of peripheral autoreactive B cells in the absence of functional human regulatory T cells

Tuure Kinnunen; Nicolas Chamberlain; Henner Morbach; Jin-Young Choi; Sangtaek Kim; Joe Craft; Lloyd Mayer; Caterina Cancrini; Laura Passerini; Rosa Bacchetta; Hans D. Ochs; Troy R. Torgerson; Eric Meffre

Regulatory T cells (Tregs) play an essential role in preventing autoimmunity. Mutations in the forkhead box protein 3 (FOXP3) gene, which encodes a transcription factor critical for Treg function, result in a severe autoimmune disorder and the production of various autoantibodies in mice and in IPEX (immune dysregulation, polyendocrinopathy, enteropathy, X-linked) patients. However, it is unknown whether Tregs normally suppress autoreactive B cells. To investigate a role for Tregs in maintaining human B-cell tolerance, we tested the reactivity of recombinant antibodies isolated from single B cells isolated from IPEX patients. Characteristics and reactivity of antibodies expressed by new emigrant/transitional B cells from IPEX patients were similar to those from healthy donors, demonstrating that defective Treg function does not impact central B-cell tolerance. In contrast, mature naive B cells from IPEX patients often expressed autoreactive antibodies, suggesting an important role for Tregs in maintaining peripheral B-cell tolerance. T cells displayed an activated phenotype in IPEX patients, including their Treg-like cells, and showed up-regulation of CD40L, PD-1, and inducibl T-cell costimulator (ICOS), which may favor the accumulation of autoreactive mature naive B cells in these patients. Hence, our data demonstrate an essential role for Tregs in the establishment and the maintenance of peripheral B-cell tolerance in humans.


Molecular Therapy | 2009

Regulated and multiple miRNA and siRNA delivery into primary cells by a lentiviral platform.

Mario Amendola; Laura Passerini; Ferdinando Pucci; Bernhard Gentner; Rosa Bacchetta; Luigi Naldini

RNA interference (RNAi) has tremendous potential for investigating gene function and developing new therapies. However, the design and validation of proficient vehicles for stable and safe microRNA (miR) and small interfering RNA (siRNA) delivery into relevant target cells remains an active area of investigation. Here, we developed a lentiviral platform to efficiently coexpress one or more natural/artificial miR together with a gene of interest from constitutive or regulated polymerase-II (Pol-II) promoters. By swapping the stem-loop (sl) sequence of a selected primary transcript (pri-miR) with that of other miR or replacing the stem with an siRNA of choice, we consistently obtained robust expression of the chimeric/artificial miR in several cell types. We validated our platform transducing a panel of engineered cells stably expressing sensitive reporters for miR activity and on a natural target. This approach allowed us to quantitatively assess at steady state the target suppression activity and expression level of each delivered miR and to compare it to those of endogenous miR. Exogenous/artificial miR reached the concentration and activity typical of highly expressed natural miR without perturbing endogenous miR maturation or regulation. Finally, we demonstrate the robust performance of the platform reversing the anergic/suppressive phenotype of human primary regulatory T cells (Treg) by knocking-down their master gene Forkhead Transcription Factor P3 (FOXP3).


European Journal of Immunology | 2011

Functional type 1 regulatory T cells develop regardless of FOXP3 mutations in patients with IPEX syndrome

Laura Passerini; Sara Di Nunzio; Silvia Gregori; Eleonora Gambineri; Massimiliano Cecconi; Markus G. Seidel; Giantonio Cazzola; Lucia Perroni; Alberto Tommasini; Silvia Vignola; Luisa Guidi; Maria Grazia Roncarolo; Rosa Bacchetta

Mutations of forkhead box p3 (FOXP3), the master gene for naturally occurring regulatory T cells (nTregs), are responsible for the impaired function of nTregs, resulting in an autoimmune disease known as the immune dysregulation, polyendocrinopathy, enteropathy, X‐linked (IPEX) syndrome. The relevance of other peripheral tolerance mechanisms, such as the presence and function of type 1 regulatory T (Tr1) cells, the major adaptive IL‐10‐producing Treg subset, in patients with IPEX syndrome remains to be clarified. FOXP3mutated Tr1‐polarized cells, differentiated in vitro from CD4+ T cells of four IPEX patients, were enriched in IL‐10+IL‐4−IFN‐γ+ T cells, a cytokine production profile specific for Tr1 cells, and expressed low levels of FOXP3 and high levels of Granzyme‐B. IPEX Tr1 cells were hypoproliferative and suppressive, thus indicating that FOXP3 mutations did not impair their function. Furthermore, we isolated Tr1 cell clones from the peripheral blood of one FOXP3null patient, demonstrating that Tr1 cells are present in vivo and they can be expanded in vitro in the absence of WT FOXP3. Overall, our results (i) show that functional Tr1 cells differentiate independently of FOXP3, (ii) confirm that human Tr1 and nTregs are distinct T‐cell lineages, and (iii) suggest that under favorable conditions Tr1 cells could exert regulatory functions in IPEX patients.


Journal of Autoimmunity | 2012

Demethylation analysis of the FOXP3 locus shows quantitative defects of regulatory T cells in IPEX-like syndrome

Federica Barzaghi; Laura Passerini; Eleonora Gambineri; S. Ciullini Mannurita; T. Cornu; E.S. Kang; Y.H. Choe; Caterina Cancrini; S Corrente; Rachele Ciccocioppo; Massimiliano Cecconi; G. Zuin; V. Discepolo; Claudia Sartirana; J. Schmidtko; Aydan Ikinciogullari; Alessandro Ambrosi; Maria Grazia Roncarolo; Sven Olek; Rosa Bacchetta

Immune dysregulation, Polyendocrinopathy, Enteropathy X-linked (IPEX) syndrome is a unique example of primary immunodeficiency characterized by autoimmune manifestations due to defective regulatory T (Treg) cells, in the presence of FOXP3 mutations. However, autoimmune symptoms phenotypically resembling IPEX often occur in the absence of detectable FOXP3 mutations. The cause of this “IPEX-like” syndrome presently remains unclear. To investigate whether a defect in Treg cells sustains the immunological dysregulation in IPEX-like patients, we measured the amount of peripheral Treg cells within the CD3+ T cells by analysing demethylation of the Treg cell-Specific-Demethylated-Region (TSDR) in the FOXP3 locus and demethylation of the T cell-Specific-Demethylated-Region (TLSDR) in the CD3 locus, highly specific markers for stable Treg cells and overall T cells, respectively. TSDR demethylation analysis, alone or normalized for the total T cells, showed that the amount of peripheral Treg cells in a cohort of IPEX-like patients was significantly reduced, as compared to both healthy subjects and unrelated disease controls. This reduction could not be displayed by flow cytometric analysis, showing highly variable percentages of FOXP3+ and CD25+FOXP3+ T cells. These data provide evidence that a quantitative defect of Treg cells could be considered a common biological hallmark of IPEX-like syndrome. Since Treg cell suppressive function was not impaired, we propose that this reduction per se could sustain autoimmunity.


Science Translational Medicine | 2013

CD4+ T Cells from IPEX Patients Convert into Functional and Stable Regulatory T Cells by FOXP3 Gene Transfer

Laura Passerini; Eva Rossi Mel; Claudia Sartirana; Georgia Fousteri; Attilio Bondanza; Luigi Naldini; Maria Grazia Roncarolo; Rosa Bacchetta

FOXP3 gene transfer in FOXP3-mutated CD4+ T cells is feasible and converts IPEX conventional T cells into regulatory T cells. FOXy Genes Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a severe genetic autoimmune disease linked to dysfunction of FOXP3 in regulatory T (Treg) cells. FOXP3 is thought to be a master transcriptional regulator in these cells, which suppress immune responses in other immune cells. The only current way to cure IPEX syndrome in affected patients is hematopoietic stem cell transplantation. Now, Passerini et al. use FOXP3 gene therapy to stabilize Treg cells and restore tolerance in IPEX syndrome. The authors transfer nonmutated FOXP3 into CD4+ T cells and demonstrate that these cells become Treg cells both phenotypically and functionally. These cells are stable in inflammatory conditions both in vitro and in vivo in a model of graft-versus-host disease. Transfer of nonmutated FOXP3 into CD4+ T cells from IPEX patients resulted in similarly suppressive cells. These data suggest that gene transfer of FOXP3 could be a new therapeutic approach for patients with IPEX syndrome. In humans, mutations in the gene encoding for forkhead box P3 (FOXP3), a critically important transcription factor for CD4+CD25+ regulatory T (Treg) cell function, lead to a life-threatening systemic poly-autoimmune disease, known as immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome. Severe autoimmunity results from the inborn dysfunction and instability of FOXP3-mutated Treg cells. Hematopoietic stem cell transplantation is the only current curative option for affected patients. We show here that when CD4+ T cells are converted into Treg cells after lentivirus-mediated FOXP3 gene transfer, the resulting CD4FOXP3 T cell population displays stable phenotype and suppressive function, especially when naïve T cells are converted. We further demonstrate that CD4FOXP3 T cells are stable in inflammatory conditions not only in vitro but also in vivo in a model of xenogeneic graft-versus-host disease. We therefore applied this FOXP3 gene transfer strategy for the development of a Treg cell–based therapeutic approach to restore tolerance in IPEX syndrome. IPEX-derived CD4FOXP3 T cells mirrored Treg cells from healthy donors in terms of cellular markers, anergic phenotype, cytokine production, and suppressive function. These findings pave the way for the treatment of IPEX patients by adoptive cell therapy with genetically engineered Treg cells and are seminal for future potential application in patients with autoimmune disorders of different origin.


The Journal of Allergy and Clinical Immunology | 2010

Point mutants of forkhead box P3 that cause immune dysregulation, polyendocrinopathy, enteropathy, X-linked have diverse abilities to reprogram T cells into regulatory T cells

Alicia N. McMurchy; Jana Gillies; Sarah E. Allan; Laura Passerini; Eleonora Gambineri; Maria Grazia Roncarolo; Rosa Bacchetta; Megan K. Levings

BACKGROUND Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) is a primary immunodeficiency with autoimmunity caused by mutations in forkhead box P3 (FOXP3), which encodes a transcription factor involved in regulatory T (Treg) cell function. The mechanistic basis for how different mutations in FOXP3 cause distinct manifestations of IPEX remains unclear. OBJECTIVE To determine whether 3 different point mutants of FOXP3 that cause severe or mild IPEX differ in their ability to reprogram conventional T cells into Treg cells. METHODS Human CD4(+) T cells were transduced with wild-type or point mutant forms of FOXP3, and changes in cell surface marker expression, cytokine production, proliferation and suppressive capacity were assessed. Ex vivo T(H)17 cells were also transduced with different forms of FOXP3 to monitor changes in IL-17 production. RESULTS The forkhead mutant F373A failed to upregulate CD25 and CCR4, did not suppress cytokine production, and induced suppressive activity less effectively than wild-type FOXP3. In contrast, although the forkhead mutant R347H was also defective in upregulation of CD25, it suppressed the production of cytokines, conferred suppressive capacity on CD4(+) T cells, and suppressed IL-17 production. F324L, a mutant outside the forkhead domain associated with mild IPEX, was equivalent to wild-type FOXP3 in all aspects tested. CONCLUSION Mutations in FOXP3 that cause IPEX do not uniformly abrogate the ability of FOXP3 to regulate transcription and drive the development of Treg cells. These data support the notion that factors in addition to functional changes in Treg cells, such as alterations in conventional T cells, are involved in the pathogenesis of IPEX.

Collaboration


Dive into the Laura Passerini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Megan K. Levings

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah E. Allan

University of British Columbia

View shared research outputs
Top Co-Authors

Avatar

Federica Barzaghi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Silvia Gregori

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Sara Di Nunzio

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge