130 results on '"Miccio, Annarita"'
Search Results
102. 70. Long Term Correction of beta-Thalassemia by Transplantation of Transduced Hematopoietic Stem Cells
- Author
-
Miccio, Annarita, primary, Cesari, Rossano, additional, Lotti, Francesco, additional, Rossi, Claudia, additional, Tiboni, Francesca, additional, Sanvito, Francesca, additional, Ponzoni, Maurilio, additional, Routledge, Samantha, additional, Antoniou, Michael, additional, and Ferrari, Giuliana, additional
- Published
- 2006
- Full Text
- View/download PDF
103. 734. The Influence of Vector Design on Integration Site Selection by gamma-Retroviral and Lentiviral Vectors
- Author
-
Recchia, Alessandra, primary, Cattoglio, Claudia, additional, Miccio, Annarita, additional, Antonelli, Antonella, additional, Testa, Anna, additional, Facchini, Giulia, additional, Sartori, Daniela, additional, Ferrari, Giuliana, additional, and Mavilio, Fulvio, additional
- Published
- 2006
- Full Text
- View/download PDF
104. Phosphorylation of Synapsin I by cAMP-Dependent Protein Kinase Controls Synaptic Vesicle Dynamics in Developing Neurons.
- Author
-
Bonanomi, Dario, Menegon, Andrea, Miccio, Annarita, Ferrari, Giuliana, Corradi, Anna, Hung-Teh Kao, Benfenati, Fabio, and Valtorta, Flavia
- Subjects
PHOSPHORYLATION ,PROTEIN kinases ,NEURONS ,AXONS ,EXOCYTOSIS ,DIFFUSION - Abstract
In developing neurons, synaptic vesicles (SVs) undergo cycles of exo-endocytosis along isolated axons. However, it is currently unknown whether SV exocytosis is regulated before synaptogenesis. Here, we show that cAMP-dependent pathways affect SV distribution and recycling in the axonal growth cone and that these effects are mediated by the SV-associated phosphoprotein synapsin I. The presence of synapsin I on SVs is necessary for the correct localization of the vesicles in the central portion of the growth cone. Phosphorylation of synapsin I by cAMP-dependent protein kinase (protein kinase A) causes the dissociation of the protein from the SV membrane, allowing diffusion of the vesicles to the periphery of the growth cone and enhancing their rate of recycling. These results provide new clues as to the bases of the well known activity of synapsin I in synapse maturation and indicate that molecular mechanisms similar to those operating at mature nerve terminals are active in developing neurons to regulate the SV life cycle before synaptogenesis. [ABSTRACT FROM AUTHOR]
- Published
- 2005
- Full Text
- View/download PDF
105. Hot spots of retroviral integration in human CD34+hematopoietic cells
- Author
-
Cattoglio, Claudia, Facchini, Giulia, Sartori, Daniela, Antonelli, Antonella, Miccio, Annarita, Cassani, Barbara, Schmidt, Manfred, von Kalle, Christof, Howe, Steve, Thrasher, Adrian J., Aiuti, Alessandro, Ferrari, Giuliana, Recchia, Alessandra, and Mavilio, Fulvio
- Abstract
Insertional oncogenesis is a possible consequence of the integration of gamma-retroviral (RV) or lentiviral (LV) vectors into the human genome. RV common insertion sites (CISs) have been identified in hematopoietic malignancies and in the nonmalignant progeny of transduced hematopoietic stem/progenitor cells (HSCs), possibly as a consequence of clonal selection in vivo. We have mapped a large number of RV and LV integrations in human CD34+HSCs, transduced in vitro and analyzed without selection. Recurrent insertion sites (hot spots) account for more than 21% of the RV integration events, while they are significantly less frequent in the case of LV vectors. RV but not LV hot spots are highly enriched in proto-oncogenes, cancer-associated CISs, and growth-controlling genes, indicating that at least part of the biases observed in the HSC progeny in vivo are characteristics of RV integration, already present in nontransplanted cells. Genes involved in hematopoietic and immune system development are targeted at high frequency and enriched in hot spots, suggesting that the CD34+gene expression program is instrumental in directing RV integration. The lower propensity of LV vectors for integrating in potentially dangerous regions of the human genome may be a factor determining a better safety profile for gene therapy applications.
- Published
- 2007
- Full Text
- View/download PDF
106. Novel genome-editing-based approaches to treat motor neuron diseases: Promises and challenges
- Author
-
Miccio, Annarita, Antoniou, Panagiotis, Ciura, Sorana, and Kabashi, Edor
- Full Text
- View/download PDF
107. Combination of lentiviral and genome editing technologies for the treatment of sickle cell disease
- Author
-
Ramadier, Sophie, Chalumeau, Anne, Felix, Tristan, Othman, Nadia, Aknoun, Sherazade, Casini, Antonio, Maule, Giulia, Masson, Cecile, De Cian, Anne, Frati, Giacomo, Brusson, Megane, Concordet, Jean-Paul, Cavazzana, Marina, Cereseto, Anna, El Nemer, Wassim, Amendola, Mario, Wattellier, Benoit, Meneghini, Vasco, and Miccio, Annarita
- Abstract
Sickle cell disease (SCD) is caused by a mutation in the β-globin gene leading to polymerization of the sickle hemoglobin (HbS) and deformation of red blood cells. Autologous transplantation of hematopoietic stem/progenitor cells (HSPCs) genetically modified using lentiviral vectors (LVs) to express an anti-sickling β-globin leads to some clinical benefit in SCD patients, but requires high- level transgene expression (i.e., high vector copy number [VCN]) to counteract HbS polymerization.
- Published
- 2021
- Full Text
- View/download PDF
108. Results from the Completed Hgb-205 Trial of Lentiglobin for ß-Thalassemia and Lentiglobin for Sickle Cell Disease Gene Therapy
- Author
-
Magrin, Elisa, Semeraro, Michaela, Magnani, Alessandra, Puy, Hervé, Miccio, Annarita, Hebert, Nicolas, Diana, Jean-Sebastien, Lefrere, Francois, Suarez, Felipe, Hermine, Olivier, Brousse, Valentine, Poirot, Catherine, Bourget, Philippe, El Nemer, Wassim, Guichard, Isabelle, Moshous, Despina, Neven, Benedicte, Monpoux, Fabrice, Poirée, Marilyne, Bartolucci, Pablo, Meritet, Jean-François, Grévent, David, Lefebvre, Thibaud, Asmal, Mohammed, Whitney, Erin, Gayron, Marisa, Huang, Wenmei, Funck-Brentano, Isabelle, de Montalembert, Mariane, Joseph, Laure, Ribeil, Jean-Antoine, and Cavazzana, Marina
- Abstract
Hermine: Celgene: Research Funding; Novartis: Research Funding; AB science: Consultancy, Equity Ownership, Honoraria, Research Funding. Brousse:bluebird bio, Inc: Consultancy; AddMedica: Consultancy. El Nemer:Hemanext: Other: Other. Bartolucci:Novartis: Membership on an entity's Board of Directors or advisory committees; AddMedica: Honoraria, Membership on an entity's Board of Directors or advisory committees; Global Blood Therapeutics: Membership on an entity's Board of Directors or advisory committees; Agios: Membership on an entity's Board of Directors or advisory committees; Roche: Membership on an entity's Board of Directors or advisory committees; HEMANEXT: Membership on an entity's Board of Directors or advisory committees. Asmal:bluebird bio, Inc: Employment, Equity Ownership. Whitney:bluebird bio, Inc: Employment, Equity Ownership. Gayron:bluebird bio, Inc: Employment, Equity Ownership. Huang:bluebird bio, Inc.: Employment, Equity Ownership. de Montalembert:AddMedica: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; bluebird bio, Inc: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees; Novartis: Consultancy, Honoraria, Membership on an entity's Board of Directors or advisory committees. Ribeil:bluebird bio, Inc: Employment, Equity Ownership. Cavazzana:SmartImmune: Other: Founder.
- Published
- 2019
- Full Text
- View/download PDF
109. Editing a ?-Globin Repressor Binding Site Restores Fetal Hemoglobin Synthesis and Corrects the Phenotype of Sickle Cell Disease Erythrocytes
- Author
-
Weber, Leslie, Frati, Giacomo, Felix, Tristan, Wollenschlaeger, Clara, Casini, Antonio, Meneghini, Vasco, Masson, Cecile, De Cian, Anne, Chalumeau, Anne, Mavilio, Fulvio, Amendola, Mario, André, Isabelle, Cereseto, Anna, El Nemer, Wassim, Concordet, Jean-Paul, Giovannangeli, Carine, Cavazzana, Marina, and Miccio, Annarita
- Abstract
El Nemer: Hemanext: Other: Other. Cavazzana:SmartImmune: Other: Founder.
- Published
- 2019
- Full Text
- View/download PDF
110. 83. Persistent and Position Independent Transgene Expression in Erythroid Cells Transduced by Lentiviral Vectors
- Author
-
Miccio, Annarita, Lotti, Francesco, Rossi, Claudia, and Ferrari, Giuliana
- Subjects
- *
TRANSGENE expression - Abstract
An abstract of the article "Persistent and Position Independent Transgene Expression in Erythroid Cells Transduced by Lentiviral Vectors," by Annarita Miccio, Francesco Lotti, Claudia Rossi and Giuliana Ferrari is presented.
- Published
- 2005
- Full Text
- View/download PDF
111. Nup153 and Nup98 bind the HIV-1 core and contribute to the early steps of HIV-1 replication
- Author
-
Di Nunzio, Francesca, Fricke, Thomas, Miccio, Annarita, Valle-Casuso, Jose Carlos, Perez, Patricio, Souque, Philippe, Rizzi, Ermanno, Severgnini, Marco, Mavilio, Fulvio, Charneau, Pierre, and Diaz-Griffero, Felipe
- Subjects
- *
HIV , *VIRAL replication , *VIRAL genes , *CHROMATIN , *VIRUS diseases , *NUCLEAR pore complex - Abstract
Abstract: The early steps of HIV-1 replication involve the entry of HIV-1 into the nucleus, which is characterized by viral interactions with nuclear pore components. HIV-1 developed an evolutionary strategy to usurp the nuclear pore machinery and chromatin in order to integrate and efficiently express viral genes. In the current work, we studied the role of nucleoporins 153 and 98 (Nup153 and Nup98) in infection of human Jurkat lymphocytes by HIV-1. We showed that Nup153-depleted cells exhibited a defect in nuclear import, while depletion of Nup 98 caused a slight defect in HIV integration. To explore the biochemical viral determinants for the requirement of Nup153 and Nup98 during HIV-1 infection, we tested the ability of these nucleoporins to interact with HIV-1 cores. Our findings showed that both nucleoporins bind HIV-1 cores suggesting that this interaction is important for HIV-1 nuclear import and/or integration. Distribution analysis of integration sites in Nup153-depleted cells revealed a reduced tendency of HIV-1 to integrate in intragenic sites, which in part could account for the large infectivity defect observed in Nup153-depleted cells. Our work strongly supports a role for Nup153 in HIV-1 nuclear import and integration. [Copyright &y& Elsevier]
- Published
- 2013
- Full Text
- View/download PDF
112. Transcriptional, epigenetic and retroviral signatures identify regulatory regions involved in hematopoietic lineage commitment
- Author
-
Silvio Bicciato, Fulvio Mavilio, Alessandro Ambrosi, Guidantonio Malagoli Tagliazucchi, Marco Severgnini, Oriana Romano, Gianluca De Bellis, Giuliana Ferrari, Annarita Miccio, Alessia Cavazza, Ermanno Rizzi, Valentina Poletti, Fabienne Cocchiarella, Clelia Peano, Claudia Rossi, Pasqualepaolo Pagliaro, Luca Petiti, Site de Recherche Intégrée en Cancérologie (SIRIC-ONCOLille), Université de Lille, Sciences et Technologies-Université de Lille, Sciences Humaines et Sociales-Centre Régional de Lutte contre le Cancer Oscar Lambret [Lille] (UNICANCER/Lille), Université de Lille-UNICANCER-Université de Lille-UNICANCER-Cancéropole Nord-Ouest-Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Pierantoni-Morgagni Hospital, Partenaires INRAE, San Raffaele Telethon (TIGET), Institute for Gene Therapy, Universita Vita Salute San Raffaele = Vita-Salute San Raffaele University [Milan, Italie] (UniSR), Approches génétiques intégrées et nouvelles thérapies pour les maladies rares (INTEGRARE), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université d'Évry-Val-d'Essonne (UEVE)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Généthon, Université Lille Nord de France (COMUE)-UNICANCER-Université Lille Nord de France (COMUE)-UNICANCER-Cancéropole Nord-Ouest-Université de Lille, Droit et Santé-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), École pratique des hautes études (EPHE), Romano, Oriana, Peano, Clelia, Tagliazucchi, Guidantonio Malagoli, Petiti, Luca, Poletti, Valentina, Cocchiarella, Fabienne, Rizzi, Ermanno, Severgnini, Marco, Cavazza, Alessia, Rossi, Claudia, Pagliaro, Pasqualepaolo, Ambrosi, Alessandro, Ferrari, Giuliana, Bicciato, Silvio, De Bellis, Gianluca, Mavilio, Fulvio, and Miccio, Annarita
- Subjects
0301 basic medicine ,Gene Expression Regulation, Viral ,Cellular differentiation ,[SDV]Life Sciences [q-bio] ,Enhancer RNAs ,Biology ,Regulatory Sequences, Nucleic Acid ,Article ,Epigenesis, Genetic ,Histones ,03 medical and health sciences ,Consensus Sequence ,Epigenetic Profile ,Humans ,Position-Specific Scoring Matrices ,Cell Lineage ,Epigenetics ,Enhancer ,Promoter Regions, Genetic ,Transcription Initiation, Genetic ,Genetics ,Multidisciplinary ,epigenetics ,Base Sequence ,Multipotent Stem Cells ,Cell Differentiation ,differentiation ,Hematopoietic Stem Cells ,Cap analysis gene expression ,Chromatin ,030104 developmental biology ,Enhancer Elements, Genetic ,Retroviridae ,Organ Specificity ,Transcriptome ,Adult stem cell - Abstract
Genome-wide approaches allow investigating the molecular circuitry wiring the genetic and epigenetic programs of human somatic stem cells. Hematopoietic stem/progenitor cells (HSPC) give rise to the different blood cell types; however, the molecular basis of human hematopoietic lineage commitment is poorly characterized. Here, we define the transcriptional and epigenetic profile of human HSPC and early myeloid and erythroid progenitors by a combination of Cap Analysis of Gene Expression (CAGE), ChIP-seq and Moloney leukemia virus (MLV) integration site mapping. Most promoters and transcripts were shared by HSPC and committed progenitors, while enhancers and super-enhancers consistently changed upon differentiation, indicating that lineage commitment is essentially regulated by enhancer elements. A significant fraction of CAGE promoters differentially expressed upon commitment were novel, harbored a chromatin enhancer signature, and may identify promoters and transcribed enhancers driving cell commitment. MLV-targeted genomic regions co-mapped with cell-specific active enhancers and super-enhancers. Expression analyses, together with an enhancer functional assay, indicate that MLV integration can be used to identify bona fide developmentally regulated enhancers. Overall, this study provides an overview of transcriptional and epigenetic changes associated to HSPC lineage commitment, and a novel signature for regulatory elements involved in cell identity.
- Published
- 2016
113. Genotoxicity of CRISPR-Cas9 systems
- Author
-
Cullot, Grégoire, Moreau-Gaudry, François, Giovannangeli, Carine, Arveiler, Benoît, Miccio, Annarita, Massy, Bernard de, STAR, ABES, Inserm U1035, Biotherapies des Maladies Genetiques et Cancers, Universite' de Bordeaux,CHU de Bordeaux,Pole de Biologie et Pathologie, Institut National de la Santé et de la Recherche Médicale (INSERM), Université de Bordeaux, and François Moreau-Gaudry
- Subjects
Homology Directed Repair ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,Réparation Dirigée par Homologie ,Edition génique ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,Gene editing ,Genotoxicity ,Genotoxicité ,CRISPR/Cas9 ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology - Abstract
Gene therapy is a promising therapeutic strategy for the monogenic diseases treatment. If the first approaches, called additive, have relied on the use of viral vectors, a growing share is now turning to gene editing. Less than a decade after its characterization, the CRISPR-Cas9 system has moved gene editing to a clinical stage. However, in the same period of time, several questions have been raised regarding the genotoxicity that can be induced by Cas9. An emerging literature points to the risk of genotoxicity at the targeted site. The thesis work presented here is part of this theme. The first part of the study aimed to describe the genotoxicity induced by a single double-stranded break made by Cas9. Characterization of the effects was done both at the nucleotide level, by monitoring the HDR / InDels balance, but also at the chromosome scale. The monitoring of chromosomal integrity has brought to light a new risk of genotoxicity that was not characterized. A sensitive and specific detection system for this risk has been developed to further characterize it. The second objective was to address the limitations of unwanted genotoxicity by developing a safer and more efficient gene editing method through the use of a single single-stranded breakage by Cas9D10A-nickase., La thérapie génique est une stratégie thérapeutique prometteuse pour le traitement des maladies monogéniques. Si les premières approches, dites additives, ont reposées sur l’utilisation de vecteurs viraux, une part grandissante se tourne désormais vers l’édition génique. Celle-ci est permise par la mise au point de nouvelles générations d’endonucléases, et en particulier le système CRISPR-Cas9. Moins d’une décennie après sa caractérisation, le système CRISPR-Cas9 a permis de faire passer l’édition génique à un stade clinique. Toutefois, dans le même laps de temps, plusieurs interrogations ont été soulevées vis-à-vis de la génotoxicité pouvant être induite par la Cas9. Une littérature émergente pointe le risque de génotoxicité au site ciblé. Le travail de thèse présentée ici s’inscrit dans cette thématique. La première partie de l’étude a eu pour objectif de décrire la génotoxicité induite par une unique cassure double-brin faite par la Cas9. La caractérisation des effets a été faite à la fois à l’échelle nucléotidique, par le suivi de la balance HDR / InDels, mais également à l’échelle du chromosome. Le suivi de l’intégrité chromosomique a permis de mettre en lumière un nouveau risque de génotoxicité encore non-caractérisé. Un système de détection sensible et spécifique de ce risque a été mis au point pour continuer de le caractériser. Le second objectif a été de répondre aux limites soulevées par la génotoxicité non-voulus, en mettant au point une méthode d’édition génique plus sûre et aussi efficace, via l’utilisation d’une unique cassure simple-brin par la Cas9D10A -nickase.
114. Cyclosporin H Improves the Transduction of CD34 + Cells with an Anti-Sickling Globin Vector, a Possible Therapeutic Approach for Sickle Cell Disease.
- Author
-
Mormin M, Rigonnot L, Chalumeau A, Miccio A, Fournier C, Pajanissamy S, Dewannieux M, and Galy A
- Subjects
- Humans, Hematopoietic Stem Cells metabolism, Transgenes, beta-Globins genetics, Anemia, Sickle Cell therapy, Anemia, Sickle Cell genetics, Genetic Vectors genetics, Transduction, Genetic, Genetic Therapy methods, Antigens, CD34 metabolism, Cyclosporine pharmacology, Cyclosporine therapeutic use, Lentivirus genetics
- Abstract
Sickle cell disease (SCD) is a debilitating monogenic disease originating from mutations in the hemoglobin beta chain gene producing an abnormal hemoglobin HbS. The polymerization of HbS is responsible for the sickling of erythrocytes leading to anemia and vaso-occlusive events. Gene therapy is a promising treatment of SCD, and two different gene therapy drugs, using gene editing or gene transfer, have already reached the marketing stage. There is still a need to improve the efficacy of gene therapy in SCD, particularly when using anti-sickling beta-globin gene transfer strategies, which must outcompete the pathological HbS. One possibility is to increase transduction by inhibiting lentiviral restriction factors such as interferon-induced transmembrane proteins (IFITMs). This can be achieved by the addition of cyclosporin H (CsH) during the transduction process. This strategy was applied here in CD34
+ hematopoietic progenitor and stem cells obtained from cord blood (CB). A first series of experiments with lentiviral vector coding for a green fluorescent protein (GFP) gene confirmed that the addition of CsH enhanced transgene expression levels and vector copy number per cell (VCN), while CD34+ cells remained viable and functional. Notably, the production of colony-forming cells (CFC) remained unaffected unless very high VCN values were reached. In a second step, CD34+ cells obtained from the CB of newborns with homozygous ( n = 2) or heterozygous ( n = 1) SCD mutations were transduced with the GLOBE-AS3 lentiviral vector coding for the HbAS3 anti-sickling beta globin. As with GFP, GLOBE-AS3 lentiviral transduction was clearly enhanced by CsH, leading to VCN > 2 and therapeutic levels of expression of the HbAS3. Moreover, the process did not affect the viability or functions of CFC. The combination of CB progenitors, the GLOBE-AS3 vector, and CsH is thus shown here to be a promising approach for the treatment of SCD.- Published
- 2024
- Full Text
- View/download PDF
115. Human iPSC-derived neural stem cells displaying radial glia signature exhibit long-term safety in mice.
- Author
-
Luciani M, Garsia C, Beretta S, Cifola I, Peano C, Merelli I, Petiti L, Miccio A, Meneghini V, and Gritti A
- Subjects
- Humans, Animals, Mice, Neuroglia metabolism, Neuroglia cytology, Astrocytes metabolism, Astrocytes cytology, Epigenesis, Genetic, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Neural Stem Cells metabolism, Neural Stem Cells cytology, Cell Differentiation
- Abstract
Human induced pluripotent stem cell-derived neural stem/progenitor cells (hiPSC-NSCs) hold promise for treating neurodegenerative and demyelinating disorders. However, comprehensive studies on their identity and safety remain limited. In this study, we demonstrate that hiPSC-NSCs adopt a radial glia-associated signature, sharing key epigenetic and transcriptional characteristics with human fetal neural stem cells (hfNSCs) while exhibiting divergent profiles from glioblastoma stem cells. Long-term transplantation studies in mice showed robust and stable engraftment of hiPSC-NSCs, with predominant differentiation into glial cells and no evidence of tumor formation. Additionally, we identified the Sterol Regulatory Element Binding Transcription Factor 1 (SREBF1) as a regulator of astroglial differentiation in hiPSC-NSCs. These findings provide valuable transcriptional and epigenetic reference datasets to prospectively define the maturation stage of NSCs derived from different hiPSC sources and demonstrate the long-term safety of hiPSC-NSCs, reinforcing their potential as a viable alternative to hfNSCs for clinical applications., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
116. Treating Sickle Cell Disease: Gene Therapy Approaches.
- Author
-
Cavazzana M, Corsia A, Brusson M, Miccio A, and Semeraro M
- Abstract
Sickle cell disease (SCD) is a hereditary blood disorder characterized by the presence of abnormal hemoglobin molecules and thus distortion (sickling) of the red blood cells. SCD causes chronic pain and organ damage and shortens life expectancy. Gene therapy emerges as a potentially curative approach for people with SCD who lack a matched sibling donor for hematopoietic stem cell transplantation. Here, we review recent progress in gene therapy for SCD and focus on innovative technologies that target the genetic roots of the disease. We also review the challenges associated with gene therapy, including oncogenic risks, and the need for refined delivery methods. Despite these hurdles, the rapidly evolving landscape of gene therapy for SCD raises hope for a paradigm shift in the treatment of this debilitating disease. As research progresses, a deeper understanding of the molecular mechanisms involved and continuous improvements in gene-editing technologies promise to bring gene therapy for SCD closer to mainstream clinical application, offering a transformative, curative option for patients with this genetic disorder.
- Published
- 2024
- Full Text
- View/download PDF
117. Safety and efficacy studies of CRISPR-Cas9 treatment of sickle cell disease highlights disease-specific responses.
- Author
-
Frati G, Brusson M, Sartre G, Mlayah B, Felix T, Chalumeau A, Antoniou P, Hardouin G, Concordet JP, Romano O, Turchiano G, and Miccio A
- Abstract
Fetal hemoglobin (HbF) reactivation expression through CRISPR-Cas9 is a promising strategy for the treatment of sickle cell disease (SCD). Here, we describe a genome editing strategy leading to reactivation of HbF expression by targeting the binding sites (BSs) for the lymphoma-related factor (LRF) repressor in the γ-globin promoters. CRISPR-Cas9 treatment in healthy donor (HD) and patient-derived HSPCs resulted in a high frequency of LRF BS disruption and potent HbF synthesis in their erythroid progeny. LRF BS disruption did not impair HSPC engraftment and differentiation but was more efficient in SCD than in HD cells. However, SCD HSPCs showed a reduced engraftment and a myeloid bias compared with HD cells. We detected off-target activity and chromosomal rearrangements, particularly in SCD samples (likely because of the higher overall editing efficiency) but did not impact the target gene expression and HSPC engraftment and differentiation. Transcriptomic analyses showed that the editing procedure results in the up-regulation of genes involved in DNA damage and inflammatory responses, which was more evident in SCD HSPCs. This study provides evidence of efficacy and safety for an editing strategy based on HbF reactivation and highlights the need of performing safety studies in clinically relevant conditions, i.e., in patient-derived HSPCs., Competing Interests: Declaration of interests A.M. is named as inventor on a patent describing GE approaches for hemoglobinopathies (WO/2020/053224/PCT/EP2019/074131: Methods for increasing fetal hemoglobin content in eukaryotic cells and uses thereof for the treatment of hemoglobinopathies)., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
118. Non-viral DNA delivery and TALEN editing correct the sickle cell mutation in hematopoietic stem cells.
- Author
-
Moiani A, Letort G, Lizot S, Chalumeau A, Foray C, Felix T, Le Clerre D, Temburni-Blake S, Hong P, Leduc S, Pinard N, Marechal A, Seclen E, Boyne A, Mayer L, Hong R, Pulicani S, Galetto R, Gouble A, Cavazzana M, Juillerat A, Miccio A, Duclert A, Duchateau P, and Valton J
- Subjects
- Animals, Humans, Female, Mice, Hematopoietic Stem Cell Transplantation, beta-Globins genetics, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, DNA Repair, Mutation, beta-Thalassemia therapy, beta-Thalassemia genetics, Disease Models, Animal, Gene Transfer Techniques, Anemia, Sickle Cell therapy, Anemia, Sickle Cell genetics, Gene Editing methods, Hematopoietic Stem Cells metabolism, Genetic Therapy methods, Transcription Activator-Like Effector Nucleases metabolism, Transcription Activator-Like Effector Nucleases genetics
- Abstract
Sickle cell disease is a devastating blood disorder that originates from a single point mutation in the HBB gene coding for hemoglobin. Here, we develop a GMP-compatible TALEN-mediated gene editing process enabling efficient HBB correction via a DNA repair template while minimizing risks associated with HBB inactivation. Comparing viral versus non-viral DNA repair template delivery in hematopoietic stem and progenitor cells in vitro, both strategies achieve comparable HBB correction and result in over 50% expression of normal adult hemoglobin in red blood cells without inducing β-thalassemic phenotype. In an immunodeficient female mouse model, transplanted cells edited with the non-viral strategy exhibit higher engraftment and gene correction levels compared to those edited with the viral strategy. Transcriptomic analysis reveals that non-viral DNA repair template delivery mitigates P53-mediated toxicity and preserves high levels of long-term hematopoietic stem cells. This work paves the way for TALEN-based autologous gene therapy for sickle cell disease., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
119. Revolutionising healing: Gene Editing's breakthrough against sickle cell disease.
- Author
-
Dimitrievska M, Bansal D, Vitale M, Strouboulis J, Miccio A, Nicolaides KH, El Hoss S, Shangaris P, and Jacków-Malinowska J
- Subjects
- Humans, Gene Editing methods, CRISPR-Cas Systems, Fetal Hemoglobin genetics, Anemia, Sickle Cell genetics, Anemia, Sickle Cell therapy, Hemoglobinopathies genetics
- Abstract
Recent advancements in gene editing illuminate new potential therapeutic approaches for Sickle Cell Disease (SCD), a debilitating monogenic disorder caused by a point mutation in the β-globin gene. Despite the availability of several FDA-approved medications for symptomatic relief, allogeneic hematopoietic stem cell transplantation (HSCT) remains the sole curative option, underscoring a persistent need for novel treatments. This review delves into the growing field of gene editing, particularly the extensive research focused on curing haemoglobinopathies like SCD. We examine the use of techniques such as CRISPR-Cas9 and homology-directed repair, base editing, and prime editing to either correct the pathogenic variant into a non-pathogenic or wild-type one or augment fetal haemoglobin (HbF) production. The article elucidates ways to optimize these tools for efficacious gene editing with minimal off-target effects and offers insights into their effective delivery into cells. Furthermore, we explore clinical trials involving alternative SCD treatment strategies, such as LentiGlobin therapy and autologous HSCT, distilling the current findings. This review consolidates vital information for the clinical translation of gene editing for SCD, providing strategic insights for investigators eager to further the development of gene editing for SCD., Competing Interests: Declaration of competing interest The authors have no conflicts of interest to declare., (Copyright © 2024 The Authors. Published by Elsevier Ltd.. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
120. CoCas9 is a compact nuclease from the human microbiome for efficient and precise genome editing.
- Author
-
Pedrazzoli E, Demozzi M, Visentin E, Ciciani M, Bonuzzi I, Pezzè L, Lucchetta L, Maule G, Amistadi S, Esposito F, Lupo M, Miccio A, Auricchio A, Casini A, Segata N, and Cereseto A
- Subjects
- Humans, Animals, Mice, Dependovirus genetics, CRISPR-Associated Protein 9 metabolism, CRISPR-Associated Protein 9 genetics, RNA, Guide, CRISPR-Cas Systems genetics, RNA, Guide, CRISPR-Cas Systems metabolism, Retina metabolism, Clostridiales genetics, Clostridiales enzymology, HEK293 Cells, Genetic Vectors metabolism, Genetic Vectors genetics, Gene Editing methods, CRISPR-Cas Systems, Microbiota genetics
- Abstract
The expansion of the CRISPR-Cas toolbox is highly needed to accelerate the development of therapies for genetic diseases. Here, through the interrogation of a massively expanded repository of metagenome-assembled genomes, mostly from human microbiomes, we uncover a large variety (n = 17,173) of type II CRISPR-Cas loci. Among these we identify CoCas9, a strongly active and high-fidelity nuclease with reduced molecular size (1004 amino acids) isolated from an uncultivated Collinsella species. CoCas9 is efficiently co-delivered with its sgRNA through adeno associated viral (AAV) vectors, obtaining efficient in vivo editing in the mouse retina. With this study we uncover a collection of previously uncharacterized Cas9 nucleases, including CoCas9, which enriches the genome editing toolbox., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
121. Progress and harmonization of gene editing to treat human diseases: Proceeding of COST Action CA21113 GenE-HumDi.
- Author
-
Cavazza A, Hendel A, Bak RO, Rio P, Güell M, Lainšček D, Arechavala-Gomeza V, Peng L, Hapil FZ, Harvey J, Ortega FG, Gonzalez-Martinez C, Lederer CW, Mikkelsen K, Gasiunas G, Kalter N, Gonçalves MAFV, Petersen J, Garanto A, Montoliu L, Maresca M, Seemann SE, Gorodkin J, Mazini L, Sanchez R, Rodriguez-Madoz JR, Maldonado-Pérez N, Laura T, Schmueck-Henneresse M, Maccalli C, Grünewald J, Carmona G, Kachamakova-Trojanowska N, Miccio A, Martin F, Turchiano G, Cathomen T, Luo Y, Tsai SQ, and Benabdellah K
- Abstract
The European Cooperation in Science and Technology (COST) is an intergovernmental organization dedicated to funding and coordinating scientific and technological research in Europe, fostering collaboration among researchers and institutions across countries. Recently, COST Action funded the "Genome Editing to treat Human Diseases" (GenE-HumDi) network, uniting various stakeholders such as pharmaceutical companies, academic institutions, regulatory agencies, biotech firms, and patient advocacy groups. GenE-HumDi's primary objective is to expedite the application of genome editing for therapeutic purposes in treating human diseases. To achieve this goal, GenE-HumDi is organized in several working groups, each focusing on specific aspects. These groups aim to enhance genome editing technologies, assess delivery systems, address safety concerns, promote clinical translation, and develop regulatory guidelines. The network seeks to establish standard procedures and guidelines for these areas to standardize scientific practices and facilitate knowledge sharing. Furthermore, GenE-HumDi aims to communicate its findings to the public in accessible yet rigorous language, emphasizing genome editing's potential to revolutionize the treatment of many human diseases. The inaugural GenE-HumDi meeting, held in Granada, Spain, in March 2023, featured presentations from experts in the field, discussing recent breakthroughs in delivery methods, safety measures, clinical translation, and regulatory aspects related to gene editing., Competing Interests: P.R. has licensed the PGK:FANCAWpre∗ LV medicinal product and receives funding and equity from Rocket Pharmaceuticals, Inc., patents and royalties, research & consulting funding. D.L. is an inventor on a patent National Institute of Chemistry filed (WO/2021/032759 patent application, European patent application EP 3783104, China patent application CN 114269930 with National Phase entry EP2020756868). R.O.B. holds patents related to CRISPR-Cas genome editing and has equity in Graphite Bio and is consultant for UNIKUM Tx. G.G. holds patents related to CRISPR-Cas genome editing, is an employee of CasZyme, and has equity in CasZyme. S.Q.T. is a co-inventor on patents for GUIDE-seq, CHANGE-seq, and other genome editing technologies and a member of the scientific advisory boards of Prime Medicine and Ensoma. T.C. is a co-inventor on patents for CAST-seq, Abnoba-Seq, and other genome editing technologies, and a member of the scientific advisory boards of Cimeo Therapeutics, Excision BioTherapeutics, and GenCC. A.C. and G.T. are inventors on a patent for MEGA (WO/2023/079285), G.T. is also co-inventor on a patent for CAST-seq., (© 2023 The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
122. An optimized SpCas9 high-fidelity variant for direct protein delivery.
- Author
-
Pedrazzoli E, Bianchi A, Umbach A, Amistadi S, Brusson M, Frati G, Ciciani M, Badowska KA, Arosio D, Miccio A, Cereseto A, and Casini A
- Subjects
- Gene Editing, CRISPR-Associated Protein 9 genetics, Electroporation, CRISPR-Cas Systems, Streptococcus pyogenes genetics
- Abstract
Electroporation of the Cas9 ribonucleoprotein (RNP) complex offers the advantage of preventing off-target cleavages and potential immune responses produced by long-term expression of the nuclease. Nevertheless, the majority of engineered high-fidelity Streptococcus pyogenes Cas9 (SpCas9) variants are less active than the wild-type enzyme and are not compatible with RNP delivery. Building on our previous studies on evoCas9, we developed a high-fidelity SpCas9 variant suitable for RNP delivery. The editing efficacy and precision of the recombinant high-fidelity Cas9 (rCas9HF), characterized by the K526D substitution, was compared with the R691A mutant (HiFi Cas9), which is currently the only available high-fidelity Cas9 that can be used as an RNP. The comparative analysis was extended to gene substitution experiments where the two high fidelities were used in combination with a DNA donor template, generating different ratios of non-homologous end joining (NHEJ) versus homology-directed repair (HDR) for precise editing. The analyses revealed a heterogeneous efficacy and precision indicating different targeting capabilities between the two variants throughout the genome. The development of rCas9HF, characterized by an editing profile diverse from the currently used HiFi Cas9 in RNP electroporation, increases the genome editing solutions for the highest precision and efficient applications., Competing Interests: Declaration of interests A. Casini and A. Cereseto are founders of and hold shares in Alia Therapeutics, a genome editing company. A. Casini is an employee of Alia Therapeutics, and A. Cereseto is a consultant for Alia Therapeutics. A patent application covering the technology disclosed in this manuscript has been filed, and A. Casini and A. Cereseto are listed as inventors., (Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
123. Novel lentiviral vectors for gene therapy of sickle cell disease combining gene addition and gene silencing strategies.
- Author
-
Brusson M, Chalumeau A, Martinucci P, Romano O, Felix T, Poletti V, Scaramuzza S, Ramadier S, Masson C, Ferrari G, Mavilio F, Cavazzana M, Amendola M, and Miccio A
- Abstract
Sickle cell disease (SCD) is due to a mutation in the β-globin gene causing production of the toxic sickle hemoglobin (HbS; α
2 βS 2 ). Transplantation of autologous hematopoietic stem and progenitor cells (HSPCs) transduced with lentiviral vectors (LVs) expressing an anti-sickling β-globin (βAS) is a promising treatment; however, it is only partially effective, and patients still present elevated HbS levels. Here, we developed a bifunctional LV expressing βAS3-globin and an artificial microRNA (amiRNA) specifically downregulating βS -globin expression with the aim of reducing HbS levels and favoring βAS3 incorporation into Hb tetramers. Efficient transduction of SCD HSPCs by the bifunctional LV led to a substantial decrease of βS -globin transcripts in HSPC-derived erythroid cells, a significant reduction of HbS+ red cells, and effective correction of the sickling phenotype, outperforming βAS gene addition and BCL11A gene silencing strategies. The bifunctional LV showed a standard integration profile, and neither HSPC viability, engraftment, and multilineage differentiation nor the erythroid transcriptome and miRNAome were affected by the treatment, confirming the safety of this therapeutic strategy. In conclusion, the combination of gene addition and gene silencing strategies can improve the efficacy of current LV-based therapeutic approaches without increasing the mutagenic vector load, thus representing a novel treatment for SCD., Competing Interests: M.B., F.M., M.C., M.A., and A.M. are the inventors of two patents describing bifunctional LVs for hemoglobinopathies., (© 2023 The Authors.)- Published
- 2023
- Full Text
- View/download PDF
124. Adenine base editor-mediated correction of the common and severe IVS1-110 (G>A) β-thalassemia mutation.
- Author
-
Hardouin G, Antoniou P, Martinucci P, Felix T, Manceau S, Joseph L, Masson C, Scaramuzza S, Ferrari G, Cavazzana M, and Miccio A
- Subjects
- Humans, Gene Editing, CRISPR-Cas Systems, Mutation, beta-Globins genetics, beta-Thalassemia genetics, beta-Thalassemia therapy
- Abstract
β-Thalassemia (BT) is one of the most common genetic diseases worldwide and is caused by mutations affecting β-globin production. The only curative treatment is allogenic hematopoietic stem/progenitor cells (HSPCs) transplantation, an approach limited by compatible donor availability and immunological complications. Therefore, transplantation of autologous, genetically-modified HSPCs is an attractive therapeutic option. However, current gene therapy strategies based on the use of lentiviral vectors are not equally effective in all patients and CRISPR/Cas9 nuclease-based strategies raise safety concerns. Thus, base editing strategies aiming to correct the genetic defect in patients' HSPCs could provide safe and effective treatment. Here, we developed a strategy to correct one of the most prevalent BT mutations (IVS1-110 [G>A]) using the SpRY-ABE8e base editor. RNA delivery of the base editing system was safe and led to ∼80% of gene correction in the HSPCs of patients with BT without causing dangerous double-strand DNA breaks. In HSPC-derived erythroid populations, this strategy was able to restore β-globin production and correct inefficient erythropoiesis typically observed in BT both in vitro and in vivo. In conclusion, this proof-of-concept study paves the way for the development of a safe and effective autologous gene therapy approach for BT., (© 2023 by The American Society of Hematology. Licensed under Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International (CC BY-NC-ND 4.0), permitting only noncommercial, nonderivative use with attribution. All other rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
125. Fetal hemoglobin rescues ineffective erythropoiesis in sickle cell disease.
- Author
-
El Hoss S, Cochet S, Godard A, Yan H, Dussiot M, Frati G, Boutonnat-Faucher B, Laurance S, Renaud O, Joseph L, Miccio A, Brousse V, Narla M, and El Nemer W
- Subjects
- Erythroblasts, Erythrocytes, Erythropoiesis, Humans, Anemia, Sickle Cell, Fetal Hemoglobin
- Abstract
While ineffective erythropoiesis has long been recognized as a key contributor to anemia in thalassemia, its role in anemia of sickle cell disease (SCD) has not been critically explored. Using in vitro and in vivo derived human erythroblasts we assessed the extent of ineffective erythropoiesis in SCD. Modeling the bone marrow hypoxic environment, we found that hypoxia induces death of sickle erythroblasts starting at the polychromatic stage, positively selecting cells with high levels of fetal hemoglobin (HbF). Cell death was associated with cytoplasmic sequestration of heat shock protein 70 and was rescued by induction of HbF synthesis. Importantly, we document that in the bone marrow of SCD patients similar cell loss occurs during the final stages of terminal differentiation. Our study provides evidence for ineffective erythropoiesis in SCD and highlights an anti-apoptotic role for HbF during the terminal stages of erythroid differentiation. These findings imply that the beneficial effect on anemia of increased HbF levels is not only due to the increased life span of red cells but also a consequence of decreased ineffective erythropoiesis.
- Published
- 2021
- Full Text
- View/download PDF
126. Genome editing approaches to β-hemoglobinopathies.
- Author
-
Brusson M and Miccio A
- Subjects
- Fetal Hemoglobin genetics, Gene Editing, Humans, beta-Globins genetics, Hemoglobinopathies genetics, Hemoglobinopathies therapy, beta-Thalassemia genetics, beta-Thalassemia therapy
- Abstract
β-hemoglobinopathies are the most common monogenic disorders worldwide and are caused by mutations in the β-globin locus altering the production of adult hemoglobin (HbA). Transplantation of autologous hematopoietic stem cells (HSCs) corrected by lentiviral vector-mediated addition of a functional β-like globin raised new hopes to treat sickle cell disease and β-thalassemia patients; however, the low expression of the therapeutic gene per vector copy is often not sufficient to fully correct the patients with a severe clinical phenotype. Recent advances in the genome editing field brought new possibilities to cure β-hemoglobinopathies by allowing the direct modification of specific endogenous loci. Double-strand breaks (DSBs)-inducing nucleases (i.e., ZFNs, TALENs and CRISPR-Cas9) or DSB-free tools (i.e., base and prime editing) have been used to directly correct the disease-causing mutations, restoring HbA expression, or to reactivate the expression of the fetal hemoglobin (HbF), which is known to alleviate clinical symptoms of β-hemoglobinopathy patients. Here, we describe the different genome editing tools, their application to develop therapeutic approaches to β-hemoglobinopathies and ongoing clinical trials using genome editing strategies., Competing Interests: Conflicts of interest The Authors have no conflict of interest., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
127. A Genome Editing System for Therapeutical Targeting of Stem Cells.
- Author
-
Frati G and Miccio A
- Subjects
- Animals, Heterografts, Humans, Mice, Mice, Inbred NOD, Mice, SCID, CRISPR-Cas Systems, Cell Differentiation genetics, Gene Editing, Hematopoietic Stem Cell Transplantation, Hematopoietic Stem Cells metabolism
- Abstract
The CRISPR/Cas9 system can be exploited to disrupt genes or cis-regulatory elements in the genome of human hematopoietic stem cells. Here, we describe a protocol to deliver the CRISPR/Cas9 ribonucleoprotein complexes into primary human hematopoietic stem cells and to evaluate the engraftment and multilineage differentiation of edited cells in immunodeficient mice. This procedure allows the editing of a high proportion of long-term repopulating hematopoietic stem cells.
- Published
- 2021
- Full Text
- View/download PDF
128. [CRISPR/Cas9: a patch on the double helix ?]
- Author
-
Miccio A and Munnich A
- Subjects
- Humans, Nobel Prize, Awards and Prizes, CRISPR-Cas Systems
- Abstract
Competing Interests: A. Miccio et A. Munnich déclarent n’avoir aucun lien d’intérêts.
- Published
- 2020
129. Extensive multilineage analysis in patients with mixed chimerism after allogeneic transplantation for sickle cell disease: insight into hematopoiesis and engraftment thresholds for gene therapy.
- Author
-
Magnani A, Pondarré C, Bouazza N, Magalon J, Miccio A, Six E, Roudaut C, Arnaud C, Kamdem A, Touzot F, Gabrion A, Magrin E, Couzin C, Fusaro M, André I, Vernant JP, Gluckman E, Bernaudin F, Bories D, and Cavazzana M
- Subjects
- Chimerism, Genetic Therapy, Hematopoiesis, Humans, Transplantation Chimera, Transplantation, Homologous, Anemia, Sickle Cell therapy, Hematopoietic Stem Cell Transplantation
- Abstract
Although studies of mixed chimerism following hematopoietic stem cell transplantation in patients with sickle cell disease (SCD) may provide insights into the engraftment needed to correct the disease and into immunological reconstitution, an extensive multilineage analysis is lacking. We analyzed chimerism simultaneously in peripheral erythroid and granulomonocytic precursors/progenitors, highly purified B and T lymphocytes, monocytes, granulocytes and red blood cells (RBC). Thirty-four patients with mixed chimerism and ≥12 months of follow-up were included. A selective advantage of donor RBC and their progenitors/precursors led to full chimerism in mature RBC (despite partial engraftment of other lineages), and resulted in the clinical control of the disease. Six patients with donor chimerism <50% had hemolysis (reticulocytosis) and higher HbS than their donor. Four of them had donor chimerism <30%, including a patient with AA donor (hemoglobin >10 g/dL) and three with AS donors (hemoglobin <10 g/dL). However, only one vaso-occlusive crisis occurred with 68.7% HbS. Except in the patients with the lowest chimerism, the donor engraftment was lower for T cells than for the other lineages. In a context of mixed chimerism after hematopoietic stem cell transplantation for SCD, myeloid (rather than T cell) engraftment was the key efficacy criterion. Results show that myeloid chimerism as low as 30% was sufficient to prevent a vaso-occlusive crisis in transplants from an AA donor but not constantly from an AS donor. However, the correction of hemolysis requires higher donor chimerism levels ( i.e ≥50%) in both AA and AS recipients. In the future, this group of patients may need a different therapeutic approach., (Copyright© 2020 Ferrata Storti Foundation.)
- Published
- 2020
- Full Text
- View/download PDF
130. An Optimized Lentiviral Vector Efficiently Corrects the Human Sickle Cell Disease Phenotype.
- Author
-
Weber L, Poletti V, Magrin E, Antoniani C, Martin S, Bayard C, Sadek H, Felix T, Meneghini V, Antoniou MN, El-Nemer W, Mavilio F, Cavazzana M, Andre-Schmutz I, and Miccio A
- Abstract
Autologous transplantation of hematopoietic stem cells transduced with a lentiviral vector (LV) expressing an anti-sickling HBB variant is a potential treatment for sickle cell disease (SCD). With a clinical trial as our ultimate goal, we generated LV constructs containing an anti-sickling HBB transgene ( HBBAS3 ), a minimal HBB promoter, and different combinations of DNase I hypersensitive sites (HSs) from the locus control region (LCR). Hematopoietic stem progenitor cells (HSPCs) from SCD patients were transduced with LVs containing either HS2 and HS3 (β-AS3) or HS2, HS3, and HS4 (β-AS3 HS4). The inclusion of the HS4 element drastically reduced vector titer and infectivity in HSPCs, with negligible improvement of transgene expression. Conversely, the LV containing only HS2 and HS3 was able to efficiently transduce SCD bone marrow and Plerixafor-mobilized HSPCs, with anti-sickling HBB representing up to ∼60% of the total HBB-like chains. The expression of the anti-sickling HBB and the reduced incorporation of the β
S -chain in hemoglobin tetramers allowed up to 50% reduction in the frequency of RBC sickling under hypoxic conditions. Together, these results demonstrate the ability of a high-titer LV to express elevated levels of a potent anti-sickling HBB transgene ameliorating the SCD cell phenotype.- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.