Network


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

Hotspot


Dive into the research topics where Angelo Lombardo is active.

Publication


Featured researches published by Angelo Lombardo.


Nature Biotechnology | 2011

An unbiased genome-wide analysis of zinc-finger nuclease specificity

Richard Gabriel; Angelo Lombardo; Anne Arens; Jeffrey C. Miller; Pietro Genovese; Christine Kaeppel; Ali Nowrouzi; Cynthia C. Bartholomae; Jianbin Wang; Geoffrey Friedman; Michael C. Holmes; Philip D. Gregory; Hanno Glimm; Manfred Schmidt; Luigi Naldini; Christof von Kalle

Zinc-finger nucleases (ZFNs) allow gene editing in live cells by inducing a targeted DNA double-strand break (DSB) at a specific genomic locus. However, strategies for characterizing the genome-wide specificity of ZFNs remain limited. We show that nonhomologous end-joining captures integrase-defective lentiviral vectors at DSBs, tagging these transient events. Genome-wide integration site analysis mapped the actual in vivo cleavage activity of four ZFN pairs targeting CCR5 or IL2RG. Ranking loci with repeatedly detectable nuclease activity by deep-sequencing allowed us to monitor the degree of ZFN specificity in vivo at these positions. Cleavage required binding of ZFNs in specific spatial arrangements on DNA bearing high homology to the intended target site and only tolerated mismatches at individual positions of the ZFN binding sites. Whereas the consensus binding sequence derived in vivo closely matched that obtained in biochemical experiments, the ranking of in vivo cleavage sites could not be predicted in silico. Comprehensive mapping of ZFN activity in vivo will facilitate the broad application of these reagents in translational research.


Nature | 2014

Targeted genome editing in human repopulating haematopoietic stem cells

Pietro Genovese; Giulia Schiroli; Giulia Escobar; Tiziano Di Tomaso; Claudia Firrito; Andrea Calabria; Davide Moi; Roberta Mazzieri; Chiara Bonini; Michael C. Holmes; Philip D. Gregory; Mirjam van der Burg; Bernhard Gentner; Eugenio Montini; Angelo Lombardo; Luigi Naldini

Targeted genome editing by artificial nucleases has brought the goal of site-specific transgene integration and gene correction within the reach of gene therapy. However, its application to long-term repopulating haematopoietic stem cells (HSCs) has remained elusive. Here we show that poor permissiveness to gene transfer and limited proficiency of the homology-directed DNA repair pathway constrain gene targeting in human HSCs. By tailoring delivery platforms and culture conditions we overcame these barriers and provide stringent evidence of targeted integration in human HSCs by long-term multilineage repopulation of transplanted mice. We demonstrate the therapeutic potential of our strategy by targeting a corrective complementary DNA into the IL2RG gene of HSCs from healthy donors and a subject with X-linked severe combined immunodeficiency (SCID-X1). Gene-edited HSCs sustained normal haematopoiesis and gave rise to functional lymphoid cells that possess a selective growth advantage over those carrying disruptive IL2RG mutations. These results open up new avenues for treating SCID-X1 and other diseases.


Nature Medicine | 2012

Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer

Elena Provasi; Pietro Genovese; Angelo Lombardo; Zulma Magnani; Pei Qi Liu; Andreas Reik; Victoria Chu; David Paschon; Lei Zhang; Jürgen Kuball; Barbara Camisa; Attilio Bondanza; Giulia Casorati; Maurilio Ponzoni; Fabio Ciceri; Claudio Bordignon; Philip D. Greenberg; Michael C. Holmes; Philip D. Gregory; Luigi Naldini; Chiara Bonini

The transfer of high-avidity T cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal T cells is an attractive cancer immunotherapy strategy. However, TCR gene transfer results in competition for surface expression and inappropriate pairing between the exogenous and endogenous TCR chains, resulting in suboptimal activity and potentially harmful unpredicted antigen specificities of the resultant TCRs. We designed zinc-finger nucleases (ZFNs) that promoted the disruption of endogenous TCR β- and α-chain genes. Lymphocytes treated with ZFNs lacked surface expression of CD3-TCR and expanded with the addition of interleukin-7 (IL-7) and IL-15. After lentiviral transfer of a TCR specific for the Wilms tumor 1 (WT1) antigen, these TCR-edited cells expressed the new TCR at high levels, were easily expanded to near purity and were superior at specific antigen recognition compared to donor-matched, unedited TCR-transferred cells. In contrast to unedited TCR-transferred cells, the TCR-edited lymphocytes did not mediate off-target reactivity while maintaining their anti-tumor activity in vivo, thus showing that complete editing of T cell specificity generates tumor-specific lymphocytes with improved biosafety profiles.


Nature Methods | 2011

Site-specific integration and tailoring of cassette design for sustainable gene transfer

Angelo Lombardo; Daniela Cesana; Pietro Genovese; Bruno Di Stefano; Elena Provasi; Daniele F Colombo; Margherita Neri; Zulma Magnani; Alessio Cantore; Pietro Lo Riso; Martina Damo; Oscar M Pello; Michael C. Holmes; Philip D. Gregory; Angela Gritti; Vania Broccoli; Chiara Bonini; Luigi Naldini

Integrative gene transfer methods are limited by variable transgene expression and by the consequences of random insertional mutagenesis that confound interpretation in gene-function studies and may cause adverse events in gene therapy. Site-specific integration may overcome these hurdles. Toward this goal, we studied the transcriptional and epigenetic impact of different transgene expression cassettes, targeted by engineered zinc-finger nucleases to the CCR5 and AAVS1 genomic loci of human cells. Analyses performed before and after integration defined features of the locus and cassette design that together allow robust transgene expression without detectable transcriptional perturbation of the targeted locus and its flanking genes in many cell types, including primary human lymphocytes. We thus provide a framework for sustainable gene transfer in AAVS1 that can be used for dependable genetic manipulation, neutral marking of the cell and improved safety of therapeutic applications, and demonstrate its feasibility by rapidly generating human lymphocytes and stem cells carrying targeted and benign transgene insertions.


Cell Death & Differentiation | 2011

Genomic instability in induced stem cells

Cristina E. Pasi; A Dereli-Öz; S Negrini; Marc Friedli; G Fragola; Angelo Lombardo; G. Van Houwe; Luigi Naldini; S Casola; Giuseppe Testa; Didier Trono; Pier Giuseppe Pelicci; Thanos D. Halazonetis

The ability to reprogram adult cells into stem cells has raised hopes for novel therapies for many human diseases. Typical stem cell reprogramming protocols involve expression of a small number of genes in differentiated somatic cells with the c-Myc and Klf4 proto-oncogenes typically included in this mix. We have previously shown that expression of oncogenes leads to DNA replication stress and genomic instability, explaining the high frequency of p53 mutations in human cancers. Consequently, we wondered whether stem cell reprogramming also leads to genomic instability. To test this hypothesis, we examined stem cells induced by a variety of protocols. The first protocol, developed specifically for this study, reprogrammed primary mouse mammary cells into mammary stem cells by expressing c-Myc. Two other previously established protocols reprogrammed mouse embryo fibroblasts into induced pluripotent stem cells by expressing either three genes, Oct4, Sox2 and Klf4, or four genes, OSK plus c-Myc. Comparative genomic hybridization analysis of stem cells derived by these protocols revealed the presence of genomic deletions and amplifications, whose signature was suggestive of oncogene-induced DNA replication stress. The genomic aberrations were to a significant degree dependent on c-Myc expression and their presence could explain why p53 inactivation facilitates stem cell reprogramming.


Neurobiology of Disease | 2012

The first reported generation of several induced pluripotent stem cell lines from homozygous and heterozygous Huntington's disease patients demonstrates mutation related enhanced lysosomal activity

Stefano Camnasio; Alessia Delli Carri; Angelo Lombardo; Iwona Grad; Caterina Mariotti; Alessia Castucci; Björn Rozell; Pietro Lo Riso; Valentina Castiglioni; Chiara Zuccato; Christelle Rochon; Yasuhiro Takashima; Giuseppe R. Diaferia; Ida Biunno; C. Gellera; Marisa Jaconi; Austin Smith; Outi Hovatta; Luigi Naldini; Stefano Di Donato; Anis Feki

Neuronal disorders, like Huntingtons disease (HD), are difficult to study, due to limited cell accessibility, late onset manifestations, and low availability of material. The establishment of an in vitro model that recapitulates features of the disease may help understanding the cellular and molecular events that trigger disease manifestations. Here, we describe the generation and characterization of a series of induced pluripotent stem (iPS) cells derived from patients with HD, including two rare homozygous genotypes and one heterozygous genotype. We used lentiviral technology to transfer key genes for inducing reprogramming. To confirm pluripotency and differentiation of iPS cells, we used PCR amplification and immunocytochemistry to measure the expression of marker genes in embryoid bodies and neurons. We also analyzed teratomas that formed in iPS cell-injected mice. We found that the length of the pathological CAG repeat did not increase during reprogramming, after long term growth in vitro, and after differentiation into neurons. In addition, we observed no differences between normal and mutant genotypes in reprogramming, growth rate, caspase activation or neuronal differentiation. However, we observed a significant increase in lysosomal activity in HD-iPS cells compared to control iPS cells, both during self-renewal and in iPS-derived neurons. In conclusion, we have established stable HD-iPS cell lines that can be used for investigating disease mechanisms that underlie HD. The CAG stability and lysosomal activity represent novel observations in HD-iPS cells. In the future, these cells may provide the basis for a powerful platform for drug screening and target identification in HD.


Cell | 2016

Inheritable silencing of endogenous genes by hit-and-run targeted epigenetic editing

Angelo Amabile; Alessandro Migliara; Paola Capasso; Mauro Biffi; Davide Cittaro; Luigi Naldini; Angelo Lombardo

Summary Gene silencing is instrumental to interrogate gene function and holds promise for therapeutic applications. Here, we repurpose the endogenous retroviruses’ silencing machinery of embryonic stem cells to stably silence three highly expressed genes in somatic cells by epigenetics. This was achieved by transiently expressing combinations of engineered transcriptional repressors that bind to and synergize at the target locus to instruct repressive histone marks and de novo DNA methylation, thus ensuring long-term memory of the repressive epigenetic state. Silencing was highly specific, as shown by genome-wide analyses, sharply confined to the targeted locus without spreading to nearby genes, resistant to activation induced by cytokine stimulation, and relieved only by targeted DNA demethylation. We demonstrate the portability of this technology by multiplex gene silencing, adopting different DNA binding platforms and interrogating thousands of genomic loci in different cell types, including primary T lymphocytes. Targeted epigenome editing might have broad application in research and medicine.


Nature Communications | 2013

iPSC-derived neural precursors exert a neuroprotective role in immune-mediated demyelination via the secretion of LIF

Cecilia Laterza; Arianna Merlini; Donatella De Feo; Francesca Ruffini; Ramesh Menon; Marco Onorati; Evelien Fredrickx; Luca Muzio; Angelo Lombardo; Giancarlo Comi; Angelo Quattrini; Carla Taveggia; Cinthia Farina; Gianvito Martino

The possibility of generating neural stem/precursor cells (NPCs) from induced pluripotent stem cells (iPSCs) has opened a new avenue of research that might nurture bench-to-bedside translation of cell transplantation protocols in central nervous system myelin disorders. Here we show that mouse iPSC-derived NPCs (miPSC-NPCs)-when intrathecally transplanted after disease onset-ameliorate clinical and pathological features of experimental autoimmune encephalomyelitis, an animal model of multiple sclerosis. Transplanted miPSC-NPCs exert the neuroprotective effect not through cell replacement, but through the secretion of leukaemia inhibitory factor that promotes survival, differentiation and the remyelination capacity of both endogenous oligodendrocyte precursors and mature oligodendrocytes. The early preservation of tissue integrity limits blood-brain barrier damage and central nervous system infiltration of blood-borne encephalitogenic leukocytes, ultimately responsible for demyelination and axonal damage. While proposing a novel mechanism of action, our results further expand the therapeutic potential of NPCs derived from iPSCs in myelin disorders.


Genome Research | 2014

Loss of transcriptional control over endogenous retroelements during reprogramming to pluripotency

Marc Friedli; Priscilla Turelli; Adamandia Kapopoulou; Benjamin Rauwel; Nathaly Castro-Diaz; Helen M. Rowe; Gabriela Ecco; Carmen Unzu; Evarist Planet; Angelo Lombardo; Bastien Mangeat; Barbara Wildhaber; Luigi Naldini; Didier Trono

Endogenous retroelements (EREs) account for about half of the mouse or human genome, and their potential as insertional mutagens and transcriptional perturbators is suppressed by early embryonic epigenetic silencing. Here, we asked how ERE control is maintained during the generation of induced pluripotent stem cells (iPSCs), as this procedure involves profound epigenetic remodeling. We found that all EREs tested were markedly up-regulated during the reprogramming of either mouse embryonic fibroblasts, human CD34(+) cells, or human primary hepatocytes. At the iPSC stage, EREs of some classes were repressed, whereas others remained highly expressed, yielding a pattern somewhat reminiscent of that recorded in embryonic stem cells. However, variability persisted between individual iPSC clones in the control of specific ERE integrants. Both during reprogramming and in iPS cells, the up-regulation of specific EREs significantly impacted on the transcription of nearby cellular genes. While transcription triggered by specific ERE integrants at highly precise developmental stages may be an essential step toward obtaining pluripotent cells, the broad and unspecific unleashing of the repetitive genome observed here may contribute to the inefficiency of the reprogramming process and to the phenotypic heterogeneity of iPSCs.


Embo Molecular Medicine | 2014

Targeted gene therapy and cell reprogramming in Fanconi anemia

Paula Río; Rocío Baños; Angelo Lombardo; Oscar Quintana-Bustamante; Lara Álvarez; Zita Garate; Pietro Genovese; Elena Almarza; Antonio Valeri; Begoña Díez; Susana Navarro; Yaima Torres; Juan P. Trujillo; Rodolfo Murillas; José Segovia; Enrique Samper; Jordi Surrallés; Philip D. Gregory; Michael C. Holmes; Luigi Naldini; Juan A. Bueren

Gene targeting is progressively becoming a realistic therapeutic alternative in clinics. It is unknown, however, whether this technology will be suitable for the treatment of DNA repair deficiency syndromes such as Fanconi anemia (FA), with defects in homology‐directed DNA repair. In this study, we used zinc finger nucleases and integrase‐defective lentiviral vectors to demonstrate for the first time that FANCA can be efficiently and specifically targeted into the AAVS1 safe harbor locus in fibroblasts from FA‐A patients. Strikingly, up to 40% of FA fibroblasts showed gene targeting 42 days after gene editing. Given the low number of hematopoietic precursors in the bone marrow of FA patients, gene‐edited FA fibroblasts were then reprogrammed and re‐differentiated toward the hematopoietic lineage. Analyses of gene‐edited FA‐iPSCs confirmed the specific integration of FANCA in the AAVS1 locus in all tested clones. Moreover, the hematopoietic differentiation of these iPSCs efficiently generated disease‐free hematopoietic progenitors. Taken together, our results demonstrate for the first time the feasibility of correcting the phenotype of a DNA repair deficiency syndrome using gene‐targeting and cell reprogramming strategies.

Collaboration


Dive into the Angelo Lombardo's collaboration.

Top Co-Authors

Avatar

Luigi Naldini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pietro Genovese

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chiara Bonini

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Elena Provasi

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Giulia Schiroli

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Zulma Magnani

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Andrea Annoni

Vita-Salute San Raffaele University

View shared research outputs
Top Co-Authors

Avatar

Fabio Ciceri

Vita-Salute San Raffaele University

View shared research outputs
Researchain Logo
Decentralizing Knowledge