Network


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

Hotspot


Dive into the research topics where Diego Alignani is active.

Publication


Featured researches published by Diego Alignani.


Nature Genetics | 2015

Whole-genome fingerprint of the DNA methylome during human B cell differentiation.

Marta Kulis; Angelika Merkel; Simon Heath; Ana C. Queirós; Ronald Schuyler; Giancarlo Castellano; Renée Beekman; Emanuele Raineri; Anna Esteve; Guillem Clot; Néria Verdaguer-Dot; Martí Duran-Ferrer; Nuria Russiñol; Roser Vilarrasa-Blasi; Simone Ecker; Vera Pancaldi; Daniel Rico; Lidia Agueda; Julie Blanc; David C. Richardson; Laura Clarke; Avik Datta; Marien Pascual; Xabier Agirre; Felipe Prosper; Diego Alignani; Bruno Paiva; Gersende Caron; Thierry Fest; Marcus O. Muench

We analyzed the DNA methylome of ten subpopulations spanning the entire B cell differentiation program by whole-genome bisulfite sequencing and high-density microarrays. We observed that non-CpG methylation disappeared upon B cell commitment, whereas CpG methylation changed extensively during B cell maturation, showing an accumulative pattern and affecting around 30% of all measured CpG sites. Early differentiation stages mainly displayed enhancer demethylation, which was associated with upregulation of key B cell transcription factors and affected multiple genes involved in B cell biology. Late differentiation stages, in contrast, showed extensive demethylation of heterochromatin and methylation gain at Polycomb-repressed areas, and genes with apparent functional impact in B cells were not affected. This signature, which has previously been linked to aging and cancer, was particularly widespread in mature cells with an extended lifespan. Comparing B cell neoplasms with their normal counterparts, we determined that they frequently acquire methylation changes in regions already undergoing dynamic methylation during normal B cell differentiation.


Clinical Cancer Research | 2016

Tumor-produced interleukin-8 attracts human myeloid-derived suppressor cells and elicits extrusion of neutrophil extracellular traps (NETs)

Carlos Alfaro; Alvaro Teijeira; Carmen Oñate; Guiomar Perez; Miguel F. Sanmamed; Maria Pilar Andueza; Diego Alignani; Sara Labiano; Arantza Azpilikueta; Alfonso Rodriguez-Paulete; Saray Garasa; Juan Pablo Fusco; María Ángela Aznar; Susana Inoges; José Medina-Echeverz; Pedro Berraondo; Jose Luis Perez-Gracia; Ignacio Melero

Purpose: Myeloid-derived suppressor cells (MDSC) are considered an important T-cell immunosuppressive component in cancer-bearing hosts. The factors that attract these cells to the tumor microenvironment are poorly understood. IL8 (CXCL8) is a potent chemotactic factor for neutrophils and monocytes. Experimental Design: MDSC were characterized and sorted by multicolor flow cytometry on ficoll-gradient isolated blood leucokytes from healthy volunteers (n = 10) and advanced cancer patients (n = 28). In chemotaxis assays, sorted granulocytic and monocytic MDSC were tested in response to recombinant IL8, IL8 derived from cancer cell lines, and patient sera. Neutrophil extracellular traps (NETs) formation was assessed by confocal microscopy, fluorimetry, and time-lapse fluorescence confocal microscopy on short-term MDSC cultures. Results: IL8 chemoattracts both granulocytic (GrMDSC) and monocytic (MoMDSC) human MDSC. Monocytic but not granulocytic MDSC exerted a suppressor activity on the proliferation of autologous T cells isolated from the circulation of cancer patients. IL8 did not modify the T-cell suppressor activity of human MDSC. However, IL8 induced the formation of NETs in the GrMDSC subset. Conclusions: IL8 derived from tumors contributes to the chemotactic recruitment of MDSC and to their functional control. Clin Cancer Res; 22(15); 3924–36. ©2016 AACR.


Blood | 2015

The cellular origin and malignant transformation of Waldenström macroglobulinemia

Bruno Paiva; Luis A. Corchete; María-Belén Vidriales; Ramón García-Sanz; Jose J. Perez; Irene Aires-Mejia; María-Luz Sanchez; Paloma Bárcena; Diego Alignani; Cristina Jiménez; María Eugenia Sarasquete; Maria-Victoria Mateos; Enrique M. Ocio; Noemi Puig; Escalante F; Hernández Jm; Cuello R; García de Coca A; Sierra M; Montes Mc; González-López Tj; Galende J; Bárez A; Alonso J; Pardal E; Alberto Orfao; Norma C. Gutiérrez; San Miguel Jf

Although information about the molecular pathogenesis of Waldenström macroglobulinemia (WM) has significantly advanced, the precise cell of origin and the mechanisms behind WM transformation from immunoglobulin-M (IgM) monoclonal gammopathy of undetermined significance (MGUS) remain undetermined. Here, we undertook an integrative phenotypic, molecular, and genomic approach to study clonal B cells from newly diagnosed patients with IgM MGUS (n = 22), smoldering (n = 16), and symptomatic WM (n = 11). Through principal component analysis of multidimensional flow cytometry data, we demonstrated highly overlapping phenotypic profiles for clonal B cells from IgM MGUS, smoldering, and symptomatic WM patients. Similarly, virtually no genes were significantly deregulated between fluorescence-activated cell sorter-sorted clonal B cells from the 3 disease groups. Interestingly, the transcriptome of the Waldenström B-cell clone was highly different than that of normal CD25(-)CD22(+) B cells, whereas significantly less genes were differentially expressed and specific WM pathways normalized once the transcriptome of the Waldenström B-cell clone was compared with its normal phenotypic (CD25(+)CD22(+low)) B-cell counterpart. The frequency of specific copy number abnormalities [+4, del(6q23.3-6q25.3), +12, and +18q11-18q23] progressively increased from IgM MGUS and smoldering WM vs symptomatic WM (18% vs 20% and 73%, respectively; P = .008), suggesting a multistep transformation of clonal B cells that, albeit benign (ie, IgM MGUS and smoldering WM), already harbor the phenotypic and molecular signatures of the malignant Waldenström clone.


Journal of Immunology | 2012

CD8 T Cell Priming in the Presence of IFN-α Renders CTLs with Improved Responsiveness to Homeostatic Cytokines and Recall Antigens: Important Traits for Adoptive T Cell Therapy

Sandra Hervas-Stubbs; Uxua Mancheño; J.I. Riezu-Boj; Ana Larraga; Maria C. Ochoa; Diego Alignani; Carlos Alfaro; Aizea Morales-Kastresana; Iranzu González; Esther Larrea; Hanspeter Pircher; Agnes Le Bon; José María López-Picazo; Salvador Martín-Algarra; Jesús Prieto; Ignacio Melero

Previous mouse and human studies have demonstrated that direct IFN-α/β signaling on naive CD8 T cells is critical to support their expansion and acquisition of effector functions. In this study, we show that human naive CD8 T cells primed in the presence of IFN-α possess a heightened ability to respond to homeostatic cytokines and to secondary Ag stimulation, but rather than differentiating to effector or memory CTLs, they preserve nature-like phenotypic features. These are qualities associated with greater efficacy in adoptive immunotherapy. In a mouse model of adoptive transfer, CD8 T cells primed in the presence of IFN-α are able to persist and to mediate a robust recall response even after a long period of naturally driven homeostatic maintenance. The long-lasting persistence of IFN-α–primed CD8 T cells is favored by their enhanced responsiveness to IL-15 and IL-7, as demonstrated in IL-15−/− and IL-7−/− recipient mice. In humans, exposure to IFN-α during in vitro priming of naive HLA-A2+ CD8 T cells with autologous dendritic cells loaded with MART126–35 peptide renders CD8 T cells with an improved capacity to respond to homeostatic cytokines and to specifically lyse MART1-expressing melanoma cells. Furthermore, in a mouse model of melanoma, adoptive transfer of tumor-specific CD8 T cells primed ex vivo in the presence of IFN-α exhibits an improved ability to contain tumor progression. Therefore, exposure to IFN-α during priming of naive CD8 T cells imprints decisive information on the expanded cells that can be exploited to improve the efficacy of adoptive T cell therapy.


Blood | 2016

Phenotypic and genomic analysis of multiple myeloma minimal residual disease tumor cells: a new model to understand chemoresistance

Bruno Paiva; Luis A. Corchete; María-Belén Vidriales; Noemi Puig; Patricia Maiso; Idoia Rodriguez; Diego Alignani; Leire Burgos; María-Luz Sanchez; Paloma Bárcena; María-Asunción Echeveste; Miguel T. Hernandez; Ramón García-Sanz; Enrique M. Ocio; Albert Oriol; Mercedes Gironella; Luis Palomera; Felipe de Arriba; Yolanda Gonzalez; Sarah K. Johnson; Joshua Epstein; Bart Barlogie; Juan José Lahuerta; Joan Bladé; Alberto Orfao; Maria-Victoria Mateos; Jesús F. San Miguel

Persistence of chemoresistant minimal residual disease (MRD) plasma cells (PCs) is associated with inferior survival in multiple myeloma (MM). Thus, characterization of the minor MRD subclone may represent a unique model to understand chemoresistance, but to our knowledge, the phenotypic and genetic features of the MRD subclone have never been investigated. Here, we compared the antigenic profile of MRD vs diagnostic clonal PCs in 40 elderly MM patients enrolled in the GEM2010MAS65 study and showed that the MRD subclone is enriched in cells overexpressing integrins (CD11a/CD11c/CD29/CD49d/CD49e), chemokine receptors (CXCR4), and adhesion molecules (CD44/CD54). Genetic profiling of MRD vs diagnostic PCs was performed in 12 patients; 3 of them showed identical copy number alterations (CNAs), in another 3 cases, MRD clonal PCs displayed all genetic alterations detected at diagnosis plus additional CNAs that emerged at the MRD stage, whereas in the remaining 6 patients, there were CNAs present at diagnosis that were undetectable in MRD clonal PCs, but also a selected number of genetic alterations that became apparent only at the MRD stage. The MRD subclone showed significant downregulation of genes related to protein processing in endoplasmic reticulum, as well as novel deregulated genes such as ALCAM that is prognostically relevant in MM and may identify chemoresistant PCs in vitro. Altogether, our results suggest that therapy-induced clonal selection could be already present at the MRD stage, where chemoresistant PCs show a singular phenotypic signature that may result from the persistence of clones with different genetic and gene expression profiles. This trial was registered atwww.clinicaltrials.gov as #NCT01237249.


Leukemia | 2017

Differentiation stage of myeloma plasma cells: biological and clinical significance.

Bruno Paiva; Noemi Puig; Maria-Teresa Cedena; B G de Jong; Y Ruiz; Inmaculada Rapado; Joaquin Martinez-Lopez; Lourdes Cordón; Diego Alignani; Jose Antonio Delgado; M C van Zelm; J J M van Dongen; Marien Pascual; Xabier Agirre; Felipe Prosper; José I. Martín-Subero; M B Vidriales; Norma C. Gutiérrez; M. Hernández; Albert Oriol; María-Asunción Echeveste; Yolanda Gonzalez; Sarah K. Johnson; Joshua Epstein; Bart Barlogie; Gareth J. Morgan; Alberto Orfao; J. Bladé; M.V. Mateos; Juan-José Lahuerta

The notion that plasma cells (PCs) are terminally differentiated has prevented intensive research in multiple myeloma (MM) about their phenotypic plasticity and differentiation. Here, we demonstrated in healthy individuals (n=20) that the CD19−CD81 expression axis identifies three bone marrow (BM)PC subsets with distinct age-prevalence, proliferation, replication-history, immunoglobulin-production, and phenotype, consistent with progressively increased differentiation from CD19+CD81+ into CD19−CD81+ and CD19−CD81− BMPCs. Afterwards, we demonstrated in 225 newly diagnosed MM patients that, comparing to normal BMPC counterparts, 59% had fully differentiated (CD19−CD81−) clones, 38% intermediate-differentiated (CD19−CD81+) and 3% less-differentiated (CD19+CD81+) clones. The latter patients had dismal outcome, and PC differentiation emerged as an independent prognostic marker for progression-free (HR: 1.7; P=0.005) and overall survival (HR: 2.1; P=0.006). Longitudinal comparison of diagnostic vs minimal-residual-disease samples (n=40) unraveled that in 20% of patients, less-differentiated PCs subclones become enriched after therapy-induced pressure. We also revealed that CD81 expression is epigenetically regulated, that less-differentiated clonal PCs retain high expression of genes related to preceding B-cell stages (for example: PAX5), and show distinct mutation profile vs fully differentiated PC clones within individual patients. Together, we shed new light into PC plasticity and demonstrated that MM patients harbouring less-differentiated PCs have dismal survival, which might be related to higher chemoresistant potential plus different molecular and genomic profiles.


Blood | 2016

Phenotypic, transcriptomic, and genomic features of clonal plasma cells in light-chain amyloidosis

Bruno Paiva; Joaquin Martinez-Lopez; Luis A. Corchete; Beatriz Sanchez-Vega; Inmaculada Rapado; Noemi Puig; Santiago Barrio; Maria Luz Sanchez; Diego Alignani; Marta Lasa; Alfonso García de Coca; Emilia Pardal; Albert Oriol; Maria-Esther Gonzalez Garcia; Fernando Escalante; Tomás J. González-López; Luis Palomera; José María Alonso; Felipe Prosper; Alberto Orfao; Maria Belen Vidriales; Maria Victoria Mateos; Juan José Lahuerta; Norma C. Gutiérrez; Jesús F. San Miguel

Immunoglobulin light-chain amyloidosis (AL) and multiple myeloma (MM) are 2 distinct monoclonal gammopathies that involve the same cellular compartment: clonal plasma cells (PCs). Despite the fact that knowledge about MM PC biology has significantly increased in the last decade, the same does not apply for AL. Here, we used an integrative phenotypic, molecular, and genomic approach to study clonal PCs from 24 newly diagnosed patients with AL. Through principal-component-analysis, we demonstrated highly overlapping phenotypic profiles between AL and both monoclonal gammopathy of undetermined significance and MM PCs. However, in contrast to MM, highly purified fluorescence-activated cell-sorted clonal PCs from AL (n = 9) showed almost normal transcriptome, with only 38 deregulated genes vs normal PCs; these included a few tumor-suppressor (CDH1, RCAN) and proapoptotic (GLIPR1, FAS) genes. Notwithstanding, clonal PCs in AL (n = 11) were genomically unstable, with a median of 9 copy number alterations (CNAs) per case, many of such CNAs being similar to those found in MM. Whole-exome sequencing (WES) performed in 5 AL patients revealed a median of 15 nonrecurrent mutations per case. Altogether, our results show that in the absence of a unifying mutation by WES, clonal PCs in AL display phenotypic and CNA profiles similar to MM, but their transcriptome is remarkably similar to that of normal PCs.


Journal of Immunology | 2014

Conventional but Not Plasmacytoid Dendritic Cells Foster the Systemic Virus–Induced Type I IFN Response Needed for Efficient CD8 T Cell Priming

Sandra Hervas-Stubbs; J.I. Riezu-Boj; Uxua Mancheño; Paloma Rueda; Lissette López; Diego Alignani; Estefania Rodríguez-Garcia; Nathalie Thieblemont; Claude Leclerc

Plasmacytoid dendritic cells (pDCs) are considered to be the principal type-I IFN (IFN-I) source in response to viruses, whereas the contribution of conventional DCs (cDCs) has been underestimated because, on a per-cell basis, they are not considered professional IFN-I–producing cells. We have investigated their respective roles in the IFN-I response required for CTL activation. Using a nonreplicative virus, baculovirus, we show that despite the high IFN-I–producing abilities of pDCs, in vivo cDCs but not pDCs are the pivotal IFN-I producers upon viral injection, as demonstrated by selective pDC or cDC depletion. The pathway involved in the virus-triggered IFN-I response is dependent on TLR9/MyD88 in pDCs and on stimulator of IFN genes (STING) in cDCs. Importantly, STING is the key molecule for the systemic baculovirus-induced IFN-I response required for CTL priming. The supremacy of cDCs over pDCs in fostering the IFN-I response required for CTL activation was also verified in the lymphocytic choriomeningitis virus model, in which IFN-β promoter stimulator 1 plays the role of STING. However, when the TLR-independent virus-triggered IFN-I production is impaired, the pDC-induced IFNs-I have a primary impact on CTL activation, as shown by the detrimental effect of pDC depletion and IFN-I signaling blockade on the residual lymphocytic choriomeningitis virus–triggered CTL response detected in IFN-β promoter stimulator 1−/− mice. Our findings reveal that cDCs play a major role in the TLR-independent virus-triggered IFN-I production required for CTL priming, whereas pDC-induced IFNs-I are dispensable but become relevant when the TLR-independent IFN-I response is impaired.


Oncotarget | 2017

IL-10 expression defines an immunosuppressive dendritic cell population induced by antitumor therapeutic vaccination

Diana Llopiz; Marta Ruiz; Stefany Infante; Lorea Villanueva; Leyre Silva; Sandra Hervas-Stubbs; Diego Alignani; Elizabeth Guruceaga; Juan J. Lasarte; Pablo Sarobe

Vaccination induces immunostimulatory signals that are often accompanied by regulatory mechanisms such as IL-10, which control T-cell activation and inhibit vaccine-dependent antitumor therapeutic effect. Here we characterized IL-10-producing cells in different tumor models treated with therapeutic vaccines. Although several cell subsets produced IL-10 irrespective of treatment, an early vaccine-dependent induction of IL-10 was detected in dendritic cells (DC). IL-10 production defined a DC population characterized by a poorly mature phenotype, lower expression of T-cell stimulating molecules and upregulation of PD-L1. These IL-10+ DC showed impaired in vitro T-cell stimulatory capacity, which was rescued by incubation with IL-10R and PD-L1-inhibiting antibodies. In vivo IL-10 blockade during vaccination decreased the proportion of IL-10+ DC and improved their maturation, without modifying PD-L1 expression. Similarly, PD-L1 blockade did not affect IL- 10 expression. Interestingly, vaccination combined with simultaneous blockade of IL-10 and PD-L1 induced stronger immune responses, resulting in a higher therapeutic efficacy in tumor-bearing mice. These results show that vaccine-induced immunoregulatory IL- 10+ DC impair priming of antitumor immunity, suggesting that therapeutic vaccination protocols may benefit from combined targeting of inhibitory molecules expressed by this DC subset.


Haematologica | 2015

Chronic exposure to IFNα drives medullar lymphopoiesis towards T-cell differentiation in mice

Marianna Di Scala; Irene Gil-Fariña; Lucia Vanrell; Rodrigo Sánchez-Bayona; Diego Alignani; Cristina Olagüe; Africa Vales; Pedro Berraondo; Jesús Prieto; Gloria González-Aseguinolaza

Interferon-α is a potent antiviral agent and a vigorous adjuvant in the induction of T-cell responses but its use is limited by hematologic toxicity. Interferon-α alters hematopoietic stem cell dormancy and impairs myelocytic and erythrocytic/megakaryocytic differentiation from hematopoietic progenitors. However, the effect of chronic interferon-α exposure on hematopoietic precursors has still not been well characterized. Here, we transduced the liver of mice with an adenoassociated vector encoding interferon-α to achieve sustained high serum levels of the cytokine. The bone marrow of these animals showed diminished long-term and short-term hematopoietic stem cells, reduction of multipotent progenitor cells, and marked decrease of B cells, but significant increase in the proportion of CD8+ and CD4+CD8+ T cells. Upon adoptive transfer to RAG−/− mice, bone marrow cells from interferon-α-treated animals generated CD4+ and CD8+ T cells while CD19+, CD11b+ and NK1.1+ lineages failed to develop. These effects are associated with the transcriptional downregulation of transcription factors involved in B-cell differentiation and modulation of key factors for T-cell development. Thus, sustained interferon-α exposure causes hematopoietic stem cells exhaustion and drives common lymphoid progenitors towards T-cell generation.

Collaboration


Dive into the Diego Alignani's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alberto Orfao

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Noemi Puig

Spanish National Research Council

View shared research outputs
Top Co-Authors

Avatar

Albert Oriol

Autonomous University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Joan Bladé

University of Barcelona

View shared research outputs
Top Co-Authors

Avatar

Joaquin Martinez-Lopez

Complutense University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Juan José Lahuerta

Complutense University of Madrid

View shared research outputs
Researchain Logo
Decentralizing Knowledge