274 results on '"Holmes MC"'
Search Results
2. Editing Human Lymphocyte Specificity for Safe and Effective Adoptive Immunotherapy of Leukemia
- Author
-
Genovese P, Provasi E, Lombardo A, Magnani Z, Liu PQ, Reik A, Chu V, Paschon DE, Zhang L, Kuball J, Camisa B, Bondanza A, Casorati G, Ciceri F, Bordignon C, Greenberg PD, Holmes MC, Gregory PD, Naldini L, Bonini C, Genovese, P, Provasi, E, Lombardo, A, Magnani, Z, Liu, Pq, Reik, A, Chu, V, Paschon, De, Zhang, L, Kuball, J, Camisa, B, Bondanza, A, Casorati, G, Ciceri, F, Bordignon, C, Greenberg, Pd, Holmes, Mc, Gregory, Pd, Naldini, L, and Bonini, C
- Published
- 2011
3. Editing human lumphocyte specificity for safe and effective adoptive immunotherapy
- Author
-
Provasi E, Genovese P, Magnani Z, Lombardo A, Reik A, Pei Qi L, Muniz Pello O, Kuball J, Casorati G, Gregory PD, BORDIGNON, CLAUDIO, Holmes MC, Greenberg PD, Naldini L, Bonini C., BONDANZA , ATTILIO, Provasi, E, Genovese, P, Magnani, Z, Lombardo, A, Reik, A, Pei Qi, L, Muniz Pello, O, Kuball, J, Bondanza, Attilio, Casorati, G, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, L, and Bonini, C.
- Published
- 2010
4. Editing human lymphocyte specificity for safe and effective adoptive immunotherapy of leukemia
- Author
-
Provasi E, Genovese P, Magnani Z, Lombardo A, Reik A, Pei Qi L, Muniz Pello O, Kuball J, Bondanza A, Casorati G, Gregory PD, BORDIGNON, CLAUDIO, Holmes MC, Greenberg PD, Naldini L, BONINI , MARIA CHIARA, Provasi, E, Genovese, P, Magnani, Z, Lombardo, A, Reik, A, Pei Qi, L, Muniz Pello, O, Kuball, J, Bondanza, A, Casorati, G, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, L, and Bonini, MARIA CHIARA
- Published
- 2010
5. Combining Site-Specific Integration and Cassette Design to Achieve Robust Expression Without Impacting Endogenous Gene Expression
- Author
-
Lombardo A, Cesana D, Genovese P, Provasi, Di Stefano B, Neri M, Broccoli V, Gritti A, Holmes MC, Gregory PD, BONINI , MARIA CHIARA, Naldini L., Lombardo, A, Cesana, D, Genovese, P, Provasi, Di Stefano, B, Neri, M, Broccoli, V, Gritti, A, Holmes, Mc, Gregory, Pd, Bonini, MARIA CHIARA, and Naldini, L.
- Published
- 2010
6. Characterization of potential genomic 'safe harbor' for efficient targeted gene addition with zinc finger nucleases
- Author
-
Lombardo A, Cesana D, Genovese P, Maruggi G, Provasi E, Mavilio F, Holmes MC, Gregory PD, Montini E, Naldini L., BONINI , MARIA CHIARA, Lombardo, A, Cesana, D, Genovese, P, Maruggi, G, Provasi, E, Bonini, MARIA CHIARA, Mavilio, F, Holmes, Mc, Gregory, Pd, Montini, E, and Naldini, L.
- Published
- 2009
7. From TCR Gene Transfer to TCR Gene Editing of Central Memory T Lymphocytes for Immunotherapy of Leukemia
- Author
-
Provasi E, Genovese P, Magnani Z, Pello OM, Kuball J, Lombardo A, Bondanza A, Gregory PD, Bordignon C, Holmes MC, Greenberg PD, Naldini L, Bonini C, Provasi, E, Genovese, P, Magnani, Z, Pello, Om, Kuball, J, Lombardo, A, Bondanza, A, Gregory, Pd, Bordignon, C, Holmes, Mc, Greenberg, Pd, Naldini, L, and Bonini, C
- Published
- 2009
8. T Cell Receptor Gene Transfer into Early Differentiated Lymphocytes by Lentiviral Vectors for Safe and Effective Adoptive Immune Therapy of Leukemia
- Author
-
Provasi E, Genovese P, Magnani Z, Pello OM, Kuball J, Lombardo A, BONDANZA , ATTILIO, Gregory PD, BORDIGNON, CLAUDIO, Holmes MC, Greenberg PD, Naldini L, BONINI , MARIA CHIARA, Provasi, E, Genovese, P, Magnani, Z, Pello, Om, Kuball, J, Lombardo, A, Bondanza, Attilio, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, L, and Bonini, MARIA CHIARA
- Published
- 2009
9. Gene editing of naive and central memory T lymphocyte specificities for adoptive immune therapy of leukemia
- Author
-
Provasi E, Pello OM, Magnani Z, Kuball J, Lombardo A, BONDANZA , ATTILIO, Gregory PD, BORDIGNON, CLAUDIO, Holmes MC, Greenberg PD, Naldini L, BONINI , MARIA CHIARA, Provasi, E, Pello, Om, Magnani, Z, Kuball, J, Lombardo, A, Bondanza, Attilio, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, L, and Bonini, MARIA CHIARA
- Published
- 2008
10. Morphological transitions in model membrane systems by the addition of anesthetics
- Author
-
Holmes, MC, Bacui, M, Leaver, MS, Holmes, MC, Bacui, M, and Leaver, MS
- Published
- 2007
11. Mesh Phases in a Ternary Nonionic Surfactant, Oil, and Water System
- Author
-
Holmes, MC, Fogden, A, Leaver, MS, Wang, Y, Holmes, MC, Fogden, A, Leaver, MS, and Wang, Y
- Published
- 2006
12. H-2 NMR evidence for the formation of random mesh phases in nonionic surfactant-water systems
- Author
-
Holmes, MC, Baciu, M, Leaver, MS, Olsson, U, Holmes, MC, Baciu, M, Leaver, MS, and Olsson, U
- Published
- 2006
13. Structural models of the R(3) over-bar m intermediate mesh phase in non-ionic surfactant water mixtures.
- Author
-
Holmes, MC, Fogden, A, Fairhurst, C, Leaver, MS, Holmes, MC, Fogden, A, Fairhurst, C, and Leaver, MS
- Published
- 2001
14. Diagnosis by ultrasonic doppler flowmeter
- Author
-
Holmes Mc
- Subjects
Materials science ,Ultrasonic doppler ,Humans ,Mass Screening ,General Medicine ,Diagnostic Errors ,Thrombophlebitis ,Flow measurement ,Blood Flow Velocity ,Biomedical engineering ,Ultrasonography - Published
- 1973
15. Genome editing for scalable production of alloantigen‐free lentiviral vectors for in vivo gene therapy
- Author
-
Sara Bartolaccini, Andrea Annoni, Tiziano Di Tomaso, Mauro Biffi, Friedrich Scheiflinger, Michela Milani, Luigi Naldini, Alessio Cantore, Michael C. Holmes, Fabio Russo, Andrea Raimondi, Angelo Lombardo, Johannes Lengler, Milani, M, Annoni, A, Bartolaccini, S, Biffi, M, Russo, F, Nullt, Nulldi Tomaso, Raimondi, A, Lengler, J, Holmes, Mc, Scheiflinger, F, Lombardo, A, Cantore, A, and Naldini, L.
- Subjects
0301 basic medicine ,Isoantigens ,Genetic enhancement ,Genetic Vectors ,MHC‐I ,lentiviral vectors ,Transfection ,Hemophilia B ,Cell Line ,Factor IX ,Membrane Cofactor Protein ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Genome editing ,stable producer cell line ,hemophilia ,MHC class I ,Animals ,Humans ,Vector (molecular biology) ,Gene ,Research Articles ,Gene Editing ,Mice, Knockout ,biology ,CD55 Antigens ,HEK 293 cells ,Lentivirus ,Genetic Therapy ,biology.organism_classification ,Virology ,gene therapy ,Cell biology ,Mice, Inbred C57BL ,030104 developmental biology ,HEK293 Cells ,Vesicular stomatitis virus ,030220 oncology & carcinogenesis ,biology.protein ,Receptors, Complement 3b ,Molecular Medicine ,Genetics, Gene Therapy & Genetic Disease ,Research Article - Abstract
Lentiviral vectors (LV) are powerful and versatile vehicles for gene therapy. However, their complex biological composition challenges large‐scale manufacturing and raises concerns for in vivo applications, because particle components and contaminants may trigger immune responses. Here, we show that producer cell‐derived polymorphic class‐I major histocompatibility complexes (MHC‐I) are incorporated into the LV surface and trigger allogeneic T‐cell responses. By disrupting the beta‐2 microglobulin gene in producer cells, we obtained MHC‐free LV with substantially reduced immunogenicity. We introduce this targeted editing into a novel stable LV packaging cell line, carrying single‐copy inducible vector components, which can be reproducibly converted into high‐yield LV producers upon site‐specific integration of the LV genome of interest. These LV efficiently transfer genes into relevant targets and are more resistant to complement‐mediated inactivation, because of reduced content of the vesicular stomatitis virus envelope glycoprotein G compared to vectors produced by transient transfection. Altogether, these advances support scalable manufacturing of alloantigen‐free LV with higher purity and increased complement resistance that are better suited for in vivo gene therapy.
- Published
- 2017
16. Therapeutic gene editing in CD34+ hematopoietic progenitors from Fanconi anemia patients
- Author
-
Cristina Díaz de Heredia, Francisco J Roman-Rodriguez, Giulia Schiroli, Luigi Naldini, Lara Álvarez, Begoña Díez, Michael C. Holmes, Juan A. Bueren, Laura Ugalde, Paula Río, Angelo Lombardo, Julián Sevilla, Pietro Genovese, Sandra Rodriguez-Perales, Ministerio de Sanidad y Consumo (España), Ministerio de Economía y Competitividad (España), Instituto de Salud Carlos III, European Research Council, Diez, B, Genovese, P, Roman rodriguez, Fj, Alvarez, L, Schiroli, G, Ugalde, L, Rodriguez perales, S, Sevilla, J, Nullc, nullDiaz De Heredia, Holmes, Mc, Lombardo, ANGELO LEONE, Naldini, Luigi, Bueren, Ja, and Rio, P.
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Genetic enhancement ,hematopoietic stem and progenitor cells ,Genetic Vectors ,CD34 ,Antigens, CD34 ,Mice, Transgenic ,Hematopoietic stem cell transplantation ,Mice, SCID ,Biology ,03 medical and health sciences ,Mice ,Fanconi anemia ,Mice, Inbred NOD ,hemic and lymphatic diseases ,medicine ,Animals ,Humans ,Cells, Cultured ,Gene Editing ,Base Sequence ,Fanconi Anemia Complementation Group A Protein ,Hematopoietic Stem Cell Transplantation ,Gene targeting ,Dependovirus ,medicine.disease ,Fetal Blood ,Hematopoietic Stem Cells ,CD34+ cells ,FANCA ,Zinc Finger Nucleases ,3. Good health ,030104 developmental biology ,Fanconi Anemia ,Cord blood ,Cancer research ,Molecular Medicine ,Stem cell ,Reactive Oxygen Species - Abstract
Gene targeting constitutes a new step in the development of gene therapy for inherited diseases. Although previous studies have shown the feasibility of editing fibroblasts from Fanconi anemia (FA) patients, here we aimed at conducting therapeutic gene editing in clinically relevant cells, such as hematopoietic stem cells (HSCs). In our first experiments, we showed that zinc finger nuclease (ZFN)-mediated insertion of a non-therapeutic EGFP-reporter donor in the AAVS1 "safe harbor" locus of FA-A lymphoblastic cell lines (LCLs), indicating that FANCA is not essential for the editing of human cells. When the same approach was conducted with therapeutic FANCA donors, an efficient phenotypic correction of FA-A LCLs was obtained. Using primary cord blood CD34+ cells from healthy donors, gene targeting was confirmed not only in in vitro cultured cells, but also in hematopoietic precursors responsible for the repopulation of primary and secondary immunodeficient mice. Moreover, when similar experiments were conducted with mobilized peripheral blood CD34+ cells from FA-A patients, we could demonstrate for the first time that gene targeting in primary hematopoietic precursors from FA patients is feasible and compatible with the phenotypic correction of these clinically relevant cells. The authors would like to thank Aurora de la Cal for her assistance with thecoordination in the delivery of the samples from the patients. RebecaSánchez-Domínguez for her expertise in flow cytometry and Centro de Trans-fusiones de la Comunidad de Madrid for providing cord blood samples. Theauthors are also indebted to the FA patients, families, and clinicians from theFA spanish network. This work was supported by grants from the“7th Frame-work Program European Commission (HEALTH-F5-2012-305421; EUROFAN-COLEN)”,“Ministerio de Sanidad, Servicios Sociales e Igualdad”(EC11/060andEC11/550),“Ministerio de Economía, Comercio y Competitividad y FondoEuropeo de Desarrollo Regional (FEDER)”(SAF2015-68073-R), and“Fondo deInvestigaciones Sanitarias, Instituto de Salud Carlos III”(RD12/0019/0023). Theauthors also thank the Fundación Marcelino Botín for promoting translationalresearch at the Hematopoietic Innovative Therapies Division of the CIEMAT.CIBERER is an initiative of the Instituto de Salud Carlos III, Spain. Sí
- Published
- 2017
17. Targeted Genome Editing in Human Repopulating Hematopoietic Stem Cells
- Author
-
Mirjam van der Burg, Giulia Schiroli, Bernhard Gentner, Angelo Lombardo, Giulia Escobar, Luigi Naldini, Michael C. Holmes, Roberta Mazzieri, Davide Moi, Chiara Bonini, Pietro Genovese, Philip D. Gregory, Claudia Firrito, Eugenio Montini, Andrea Calabria, Tiziano Di Tomaso, Immunology, Genovese, P, Schiroli, G, Escobar, G, Di Tomaso, T, Firrito, C, Calabria, A, Moi, D, Mazzieri, R, Bonini, MARIA CHIARA, Holmes, Mc, Gregory, Pd, van der Burg, M, Gentner, B, Montini, E, Lombardo, ANGELO LEONE, and Naldini, Luigi
- Subjects
Male ,DNA, Complementary ,Genetic enhancement ,Transgene ,Antigens, CD34 ,Biology ,Gene delivery ,X-Linked Combined Immunodeficiency Diseases ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,SDG 3 - Good Health and Well-being ,Genome editing ,Animals ,Humans ,030304 developmental biology ,Genetics ,0303 health sciences ,Transcription activator-like effector nuclease ,Multidisciplinary ,Genome, Human ,Hematopoietic Stem Cell Transplantation ,Gene targeting ,Endonucleases ,Fetal Blood ,Hematopoietic Stem Cells ,Hematopoiesis ,Cell biology ,Haematopoiesis ,030220 oncology & carcinogenesis ,Mutation ,Gene Targeting ,Stem cell ,Interleukin Receptor Common gamma Subunit ,Targeted Gene Repair - Abstract
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.
- Published
- 2014
18. Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer
- Author
-
Giulia Casorati, Pietro Genovese, Luigi Naldini, Philip D. Gregory, Angelo Lombardo, Maurilio Ponzoni, Chiara Bonini, Claudio Bordignon, Jürgen Kuball, Philip D. Greenberg, David Paschon, Victoria Chu, Andreas Reik, Michael C. Holmes, Elena Provasi, Zulma Magnani, Attilio Bondanza, Pei-Qi Liu, Lei Zhang, Fabio Ciceri, Barbara Camisa, Provasi, E, Genovese, P, Lombardo, ANGELO LEONE, Magnani, Z, Liu Pei, Q, Reik, A, Chu, V, Paschon, D. E., Zhang, L, Kuball, J, Camisa, B, Bondanza, Attilio, Casorati, G, Ponzoni, Maurilio, Ciceri, Fabio, Bordignon, Claudio, Greenberg, P. D., Holmes, Mc, Gregory, Pd, Naldini, Luigi, and Bonini, MARIA CHIARA
- Subjects
T-Lymphocytes ,medicine.medical_treatment ,CD3 ,Molecular Sequence Data ,Receptors, Antigen, T-Cell ,T-Cell Antigen Receptor Specificity ,chemical and pharmacologic phenomena ,Endogeny ,Gene transfer ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Jurkat Cells ,03 medical and health sciences ,0302 clinical medicine ,Cancer immunotherapy ,medicine ,Humans ,Deoxyribonucleases, Type II Site-Specific ,WT1 Proteins ,030304 developmental biology ,0303 health sciences ,Leukemia ,Base Sequence ,Lentivirus ,T-cell receptor ,Gene Transfer Techniques ,food and beverages ,Zinc Fingers ,hemic and immune systems ,General Medicine ,medicine.disease ,Zinc finger nuclease ,Molecular biology ,Tumor antigen ,3. Good health ,030220 oncology & carcinogenesis ,biology.protein - Abstract
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 beta- and alpha-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.
- Published
- 2012
19. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR
- Author
-
Dean A. Lee, Helen Huls, Michael C. Holmes, Pei-Qi Liu, Chiara Bonini, Laurence J.N. Cooper, Partow Kebriaei, Yuanyue Zhou, Sourindra Maiti, Richard E. Champlin, Edward J. Rebar, Luigi Naldini, Brian Rabinovitch, Hiroki Torikai, Philip D. Gregory, Ling Zhang, Andreas Reik, Jeffrey C. Miller, Torikai, H1, Reik, A, Liu, Pq, Zhou, Y, Zhang, L, Maiti, S, Huls, H, Miller, Jc, Kebriaei, P, Rabinovitch, B, Lee, Da, Champlin, Re, Bonini, MARIA CHIARA, Naldini, Luigi, Rebar, Ej, Gregory, Pd, Holmes, Mc, and Cooper, Lj
- Subjects
Adult ,CD3 Complex ,T-Lymphocytes ,T cell ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Antigen-Presenting Cells ,Streptamer ,Biology ,Lymphocyte Activation ,Biochemistry ,Epitopes ,Gene Knockout Techniques ,03 medical and health sciences ,0302 clinical medicine ,CD28 Antigens ,Antigens, Neoplasm ,medicine ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Cells, Cultured ,030304 developmental biology ,0303 health sciences ,T-cell receptor ,Zinc Fingers ,Gene Therapy ,Cell Biology ,Hematology ,Endonucleases ,Molecular biology ,Recombinant Proteins ,Chimeric antigen receptor ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Immunotherapy ,Genetic Engineering ,K562 Cells ,CD8 ,030215 immunology - Abstract
Clinical-grade T cells are genetically modified ex vivo to express a chimeric antigen receptor (CAR) to redirect specificity to a tumor associated antigen (TAA) thereby conferring antitumor activity in vivo. T cells expressing a CD19-specific CAR recognize B-cell malignancies in multiple recipients independent of major histocompatibility complex (MHC) because the specificity domains are cloned from the variable chains of a CD19 monoclonal antibody. We now report a major step toward eliminating the need to generate patient-specific T cells by generating universal allogeneic TAA-specific T cells from one donor that might be administered to multiple recipients. This was achieved by genetically editing CD19-specific CAR+ T cells to eliminate expression of the endogenous αβ T-cell receptor (TCR) to prevent a graft-versus-host response without compromising CAR-dependent effector functions. Genetically modified T cells were generated using the Sleeping Beauty system to stably introduce the CD19-specific CAR with subsequent permanent deletion of α or β TCR chains with designer zinc finger nucleases. We show that these engineered T cells display the expected property of having redirected specificity for CD19 without responding to TCR stimulation. CAR+TCRneg T cells of this type may potentially have efficacy as an off-the-shelf therapy for investigational treatment of B-lineage malignancies.
- Published
- 2012
- Full Text
- View/download PDF
20. Targeted gene therapy and cell reprogramming in Fanconi anemia
- Author
-
Oscar Quintana-Bustamante, Antonio Valeri, Rocío Baños, Elena Almarza, Juan A. Bueren, Michael C. Holmes, Paula Río, Enrique Samper, Pietro Genovese, Philip D. Gregory, Begoña Díez, Zita Garate, Yaima Torres, José C. Segovia, Angelo Lombardo, Susana Navarro, Rodolfo Murillas, Luigi Naldini, Juan P. Trujillo, Jordi Surrallés, Lara Álvarez, Rio, P, Baños, R, Lombardo, ANGELO LEONE, Quintana Bustamante, O, Alvarez, L, Garate, Z, Genovese, P, Almarza, E, Valeri, A, Díez, B, Navarro, S, Torres, Y, Trujillo, Jp, Murillas, R, Segovia, Jc, Samper, E, Surralles, J, Gregory, Pd, Holmes, Mc, Naldini, Luigi, and Bueren, Ja
- Subjects
Gene-targeting ,DNA repair ,Genetic enhancement ,Cell reprogramming ,Induced Pluripotent Stem Cells ,iPSCs ,Biology ,Genome editing ,Fanconi anemia ,zinc finger nucleases ,medicine ,Zinc finger nucleases ,Humans ,gene-targeting ,Research Articles ,Cells, Cultured ,Fanconi Anemia Complementation Group A Protein ,cell reprogramming ,Gene targeting ,Genetic Therapy ,Fibroblasts ,medicine.disease ,Cellular Reprogramming ,Zinc finger nuclease ,FANCA ,3. Good health ,Hematopoiesis ,Fanconi Anemia ,Ipscs ,Gene Targeting ,Cancer research ,Molecular Medicine ,Reprogramming - Abstract
Altres ajuts: European Regional Development FEDER Funds, Italian Ministry of Health, Fondo de Investigaciones Sanitarias, Dirección General de Investigación de la Comunidad de Madrid S2010/BMD-2420, La Fundació Privada La Marató de TV3 121430/31/32, Marató de TV3 464/C/2012 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.
- Published
- 2014
21. An unbiased genome-wide analysis of zinc-finger nuclease specificity
- Author
-
Jianbin Wang, Ali Nowrouzi, Manfred Schmidt, Christine Kaeppel, Jeffrey C. Miller, Hanno Glimm, Michael C. Holmes, Angelo Lombardo, Richard Gabriel, Luigi Naldini, Cynthia C. Bartholomae, Geoffrey Friedman, Anne Arens, Pietro Genovese, Philip D. Gregory, Christof von Kalle, Gabriel, R, Lombardo, ANGELO LEONE, Arens, A, Miller, Jc, Genovese, P, Kaeppel, C, Nowrouzi, A, Bartholomae, Cc, Wang, J, Friedman, G, Holmes, Mc, Gregory, Pd, Glimm, H, Schmidt, M, Naldini, Luigi, and von Kalle, C.
- Subjects
In silico ,Genetic Vectors ,Biomedical Engineering ,Bioengineering ,Locus (genetics) ,HIV Integrase ,Biology ,Applied Microbiology and Biotechnology ,Substrate Specificity ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Genome editing ,Cell Line, Tumor ,Cluster Analysis ,Humans ,DNA Breaks, Double-Stranded ,Binding site ,030304 developmental biology ,Genetics ,0303 health sciences ,Nuclease ,Binding Sites ,Endodeoxyribonucleases ,fungi ,Lentivirus ,Gene targeting ,Zinc Fingers ,Sequence Analysis, DNA ,Zinc finger nuclease ,chemistry ,Genetic Loci ,030220 oncology & carcinogenesis ,Gene Targeting ,biology.protein ,Molecular Medicine ,DNA ,Biotechnology ,Protein Binding - Abstract
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.
- Published
- 2011
22. Editing central memory T-lymphocyte specificity for safe and effective adoptive immunotherapy of leukaemia
- Author
-
Genovese P, Provasi E, Magnani Z, Lombardo A, Pello OM, Kuball J, BONDANZA , ATTILIO, Gregory P, BORDIGNON, CLAUDIO, Holmes M, Greenberg P, Naldini L, BONINI , MARIA CHIARA, Provasi, E, Genovese, P, Magnani, Z, Lombardo, A, Pello, Om, Kuball, J, Bondanza, Attilio, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, L, Bonini, MARIA CHIARA, Gregory, P, Holmes, M, and Greenberg, P
- Published
- 2010
23. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery
- Author
-
Ya Li Lee, Cesare Galli, Luigi Naldini, Kenneth Kim, Christian Beauséjour, Michael C. Holmes, Silvia Colleoni, Angelo Lombardo, Pietro Genovese, Philip D. Gregory, Fyodor D. Urnov, Dale Ando, Lombardo, ANGELO LEONE, Genovese, P, Beausejour, Cm, Colleoni, S, Lee, Yl, Kim, Ka, Ando, D, Urnov, Fd, Galli, C, Gregory, Pd, Holmes, Mc, and Naldini, Luigi
- Subjects
DNA Repair ,Virus Integration ,Genetic enhancement ,Genetic Vectors ,Biomedical Engineering ,Bioengineering ,Biology ,Gene delivery ,Applied Microbiology and Biotechnology ,Genome engineering ,Viral vector ,Genome editing ,Humans ,Point Mutation ,Transgenes ,Deoxyribonucleases, Type II Site-Specific ,Embryonic Stem Cells ,Genetics ,Zinc finger ,Integrases ,Lentivirus ,fungi ,Genetic transfer ,Gene Transfer Techniques ,Zinc Fingers ,Templates, Genetic ,Zinc finger nuclease ,Cell biology ,Molecular Medicine ,Genetic Engineering ,Interleukin Receptor Common gamma Subunit ,Biotechnology - Abstract
Achieving the full potential of zinc-finger nucleases (ZFNs) for genome engineering in human cells requires their efficient delivery to the relevant cell types. Here we exploited the infectivity of integrase-defective lentiviral vectors (IDLV) to express ZFNs and provide the template DNA for gene correction in different cell types. IDLV-mediated delivery supported high rates (13-39%) of editing at the IL-2 receptor common gamma-chain gene (IL2RG) across different cell types. IDLVs also mediated site-specific gene addition by a process that required ZFN cleavage and homologous template DNA, thus establishing a platform that can target the insertion of transgenes into a predetermined genomic site. Using IDLV delivery and ZFNs targeting distinct loci, we observed high levels of gene addition (up to 50%) in a panel of human cell lines, as well as human embryonic stem cells (5%), allowing rapid, selection-free isolation of clonogenic cells with the desired genetic modification.
- Published
- 2007
24. Gene editing and site-specific gene additino in human stem cells using designed zinc finger nucleases and integrase detective lentiviral vector delivery
- Author
-
Lombardo A., Beausejour C.M., Genovese P., Colleoni S., Lee Y. L., Kim KA, Ando D., Urnov F., Gregory P.D., Holmes M.C., Naldini L., GALLI, CESARE, Lombardo A., Beausejour CM., Genovese P., Colleoni S., Lee Y-L., Kim KA, Ando D., Urnov F., Galli C., Gregory PD., Holmes MC., and Naldini L.
- Subjects
HOMOLOGUS RECOMBINATION ,fungi ,ZINC FINGER NUCLEASES ,STEM CELLS - Abstract
Achieving the full potential of zinc-finger nucleases (ZFNs) for genome engineering in human cells requires their efficient delivery to the relevant cell types. Here we exploited the infectivity of integrase-defective lentiviral vectors (IDLV) to express ZFNs and provide the template DNA for gene correction in different cell types. IDLV-mediated delivery supported high rates (13-39%) of editing at the IL-2 receptor common gamma-chain gene (IL2RG) across different cell types. IDLVs also mediated site-specific gene addition by a process that required ZFN cleavage and homologous template DNA, thus establishing a platform that can target the insertion of transgenes into a predetermined genomic site. Using IDLV delivery and ZFNs targeting distinct loci, we observed high levels of gene addition (up to 50%) in a panel of human cell lines, as well as human embryonic stem cells (5%), allowing rapid, selection-free isolation of clonogenic cells with the desired genetic modification.
- Published
- 2007
25. TCR Gene Editing Results in Effective Immunotherapy of Leukemia without the Development of GvHD
- Author
-
Maurilio Ponzoni, Giulia Casorati, Andreas Reik, Philip D. Greenberg, Barbara Camisa, Luigi Naldini, Michael C. Holmes, Angelo Lombardo, Fabio Ciceri, Claudio Bordignon, Pei-Qi Liu, Pietro Genovese, Philip D. Gregory, Chiara Bonini, Lei Zhang, J. Kuball, David Paschon, Elena Provasi, Victoria Chu, Zulma Magnani, Attilio Bondanza, Provasi, E, Genovese, P, Lombardo, A, Magnani, Z, Liu, Pq, Reik, A, Chu, V, Paschon, De, Zhang, L, Kuball, J, Camisa, B, Bondanza, Attilio, Casorati, G, Ponzoni, Maurilio, Ciceri, Fabio, Bordignon, Claudio, Greenberg, Pd, Holmes, Mc, Gregory, Pd, Naldini, L, Bonini, MARIA CHIARA, Liu, P, and Paschon, D. E.
- Subjects
T cell receptor complex ,biology ,CD3 ,T cell ,medicine.medical_treatment ,Immunology ,T-cell receptor ,CD28 ,Cell Biology ,Hematology ,Immunotherapy ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,Antigen ,biology.protein ,medicine ,CD8 - Abstract
Abstract 667 Transfer of high-avidity T-cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal CD8+ T cells is an attractive strategy for targeted cancer immunotherapy. However, the successful implementation of this approach is limited by technical and safety issues, including inefficient gene transfer, unstable transgene expression, exhaustion of gene-modified cells and most importantly, the unpredictable results of mispairing between the endogenous and exogenous TCR chains. Indeed, co-expression of the endogenous and exogenous TCR in the same cell not only reduces cell-surface expression of the introduced tumor-specific TCR, but also drives the potential for these gene-modified T cells to acquire autoreactive specificities. Such TCR mispairing has been shown to result in autoreactive T cells in animal models [Bendle et al., Nat Med. 2010]. To partially overcome these limitations, we have developed a novel strategy based on zinc finger nucleases (ZFNs) that permits editing of T cell specificity at the DNA level, combining the disruption of the endogenous TCR chain genes with the transfer of a tumor-specific TCR. Two sets of ZFNs were designed targeting the constant regions of the α (TRAC-ZFN) and β (TRBC-ZFN) TCR chain genes, respectively. We transiently delivered these ZFNs into primary T lymphocytes activated with anti-CD3 and anti-CD28 antibody-conjugated beads and cultured with low doses of IL-7 and IL-15, to promote the survival and expansion of the ZFN-modified cells. ZFN delivery into activated T lymphocytes abrogated expression of the CD3/TCR complex on the cell surface (% of CD3neg cells with TRAC-ZFN: 34%±11 and with TRBC-ZFN: 30%±9). No phenotypic differences were observed in CD3pos and CD3neg lymphocytes, which displayed a similar CD4/CD8 ratio while displaying an early T-cell differentiation phenotype, as evidenced by high expression of CD62L, CD27, CD28 and IL-7Rα markers. Sorted CD3neg cells proved stable in culture (demonstrating that ZFN exposure was well tolerated), and did not respond to TCR-dependent stimulation with the mitogen PHA, as expected for cells carrying a disrupted TCR gene. CD3neg cells were efficiently transduced with a lentiviral vector encoding a tumor-specific exogenous TCR chain, resulting in the restoration of cell surface translocation of CD3, thus facilitating the selective expansion of TCR-transduced cells by polyclonal stimulation. To demonstrate the antitumor activity of these modified cells we selected an HLA-A2 restricted, codon-optimized cysteine-modified TCR specific for the Wilms' tumor antigen 1 (WT1). To achieve complete editing of T cell specificity, we established a protocol that sequentially disrupted the endogenous TCR α and β chains with high efficiency (averages: 36% and 18%), followed by lentiviral transfer of the tumor-specific TCR α and β chains (average efficiencies: 65% and 25%). This procedure resulted in a population of TCR-edited lymphocytes encoding only the tumor-specific TCR that, in the absence of competition from the endogenous receptor, was expressed at high physiological levels. Accordingly, TCR-edited lymphocytes were superior to conventional TCR-transferred cells in promoting specific recognition of WT1-expressing targets, including primary leukemias, and most importantly, were devoid of residual endogenous TCR reactivity including alloreactivity. Finally, in a humanized GvHD model, we showed that, at a variance with 100% of mice infused with unmanipulated T cells, and 80% of mice receiving TCR-transferred lymphocytes developing lethal GvHD, no GvHD was observed upon infusion of matched TCR-edited cells, despite robust T cell engraftment rates across all groups. These data demonstrate that the successful genetic re-programming of T cell specificity in primary lymphocytes results in a functionally superior target specific killing activity and thus has the potential to greatly improve the safety and therapeutic benefit of cancer immunotherapy, without triggering its potentially negative effects. (Provasi and Genovese: equal contribution). Disclosures: Liu: Sangamo Biosciences: Employment. Reik:Sangamo Biosciences: Employment. Chu:Sangamo Biosciences: Employment. Paschon:Sangamo Biosciences: Employment. Zhang:Sangamo Biosciences: Employment. Bordignon:Molmed: Employment. Holmes:Sangamo Biosciences: Employment. Gregory:Sangamo Biosciences: Employment. Bonini:Molmed: Consultancy.
- Published
- 2011
26. T Cell Receptor Gene Transfer into Naive and Central Memory Lymphocytes by Lentiviral Vectors for a Safe and Effective Adoptive Immune Therapy of Leukemia
- Author
-
Oscar M. Pello, Angelo Lombardo, Zulma Magnani, Jurgen Kuball, Philip D. Gregory, Chiara Bonini, Claudio Bordignon, Elena Provasi, Attilio Bondanza, Michael C. Holmes, Phil Greenberg, Luigi Naldini, Provasi, E, Pello, Om, Magnani, Z, Kuball, J, Lombardo, ANGELO LEONE, Bondanza, Attilio, Gregory, Pd, Bordignon, Claudio, Holmes, Mc, Greenberg, Pd, Naldini, Luigi, and Bonini, MARIA CHIARA
- Subjects
Adoptive cell transfer ,T cell ,Immunology ,T-cell receptor ,CD28 ,hemic and immune systems ,chemical and pharmacologic phenomena ,Cell Biology ,Hematology ,Biology ,Biochemistry ,Cell biology ,Viral vector ,Transplantation ,medicine.anatomical_structure ,Antigen ,medicine ,Cytotoxic T cell - Abstract
The potency of cellular adoptive immunotherapy against cancer has been revealed by persistent and complete clinical responses obtained with allogeneic hemopoietic cell transplantation (allo-HSCT) followed by the adoptive transfer of donor T lymphocytes and by initial clinical responses observed with the adoptive transfer of tumor specific cytotoxic T lymphocytes (CTLs) in cancer patients. Major hurdles limiting adoptive T cell therapy relate to toxicity (i.e. graft-versus-host disease-GvHD in allo-HSCT) and efficacy (i.e. difficulty in expanding rare, high-avidity tumor-specific CTLs in conditions that can preserve their function and prevent exhaustion). The transfer of the T cell receptor (TCR) from high-avidity tumor-specific CTLs to polyclonal lymphocytes may overcome these difficulties, but is still limited by low and transient transgene expression, unpredictable pairing of the exogenous and endogenous TCR chains and poor survival and expansion potential of gene-modified effector lymphocytes. To overcome these limitations, we cloned genes encoding a high-avidity TCR specific for an HLA-A2-restricted peptide from the oncogenic Wilms tumor antigen 1 (WT1126-135), in a third generation lentiviral vector under the control of a bi-directional PGK or a bi-directional EF1α promoter. To increase TCR expression and facilitate appropriate TCR pairing, we used a codon-optimized TCR, modified with point mutations to introduce cysteines into the constant regions of the α and 7β chains. Human T lymphocytes were efficiently transduced by both vectors, following activation with anti-CD3 and anti-CD28 antibody-conjugated beads (bCD3/CD28) and culture with low doses of IL-7/IL-15. However, the PGK promoter was superior to EF1α in sustaining stochiometric expression of WT1-specific TCR chains, at levels appropriate for efficient HLA-A2/WT1 pentamer binding (16%), for up to 50 days, in the absence of further T cell stimulation. Additionally, we observed that a phenotype consistent with early (naïve and central memory) T cell differentiation (CD45RA−/+CD62L+, CD28+CD27+, IL7Ra+, IL-2+ γIFN±) was preserved in TCR-modified lymphocytes generated in these culture conditions. Sorted naïve (CD45RA+/CD62L+) and central memory (CD45RA−/CD62L+) lymphocytes were efficiently transduced by TCR-LV and maintained the original T cell phenotype. Accordingly, TCR-modified lymphocytes showed excellent survival and expansion capacity, and, upon antigenic stimulation mediated high WT1-specific γIFN production and cytotoxic activity. To improve the safety of the strategy, we attempted sitespecific integration of transgenes using of the ZFN technology. Adenoviral transfer of a set of ZFN specific for the putative safe-harbor locus CCR5 coupled with integrase defective lentiviral vectors carrying the donor DNA flanked transgene, enables efficient site-specific integration in human lymphocytes resulting in stable transgene expression. Analyses of sorted gene-modified cells is currently ongoing. Site-specific integration of an optimized, leukemia-specific TCR into both naïve and central memory lymphocytes may represent an effective method for the generation of robust engineered tumor-specific CTLs.
- Published
- 2008
27. Site-specific integration and tailoring of cassette design for sustainable gene transfer
- Author
-
Bruno Di Stefano, Daniela Cesana, Zulma Magnani, Michael C. Holmes, Luigi Naldini, Vania Broccoli, Martina Damo, Alessio Cantore, Margherita Neri, Pietro Lo Riso, Chiara Bonini, Oscar M Pello, Angela Gritti, Elena Provasi, Angelo Lombardo, Daniele F Colombo, Pietro Genovese, Philip D. Gregory, Lombardo, ANGELO LEONE, Cesana, D, Genovese, P, Nullb, nullDi Stefano, Provasi, E, Colombo, Df, Neri, M, Magnani, Z, Cantore, A, Nullp, nullLo Riso, Damo, M, Pello, Om, Holmes, Mc, Gregory, Pd, Gritti, A, Broccoli, V, Bonini, MARIA CHIARA, and Naldini, Luigi
- Subjects
Receptors, CCR5 ,Virus Integration ,Transgene ,Genetic enhancement ,Locus (genetics) ,Biology ,Biochemistry ,Insertional mutagenesis ,03 medical and health sciences ,0302 clinical medicine ,Humans ,Epigenetics ,Molecular Biology ,Gene ,030304 developmental biology ,Genetics ,0303 health sciences ,Gene Transfer Techniques ,Cell Biology ,Dependovirus ,Mutagenesis, Insertional ,Mutagenesis, Site-Directed ,Stem cell ,030217 neurology & neurosurgery ,Biotechnology - Abstract
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.
28. Sleeping Beauty mRNA-LNP enables stable rAAV transgene expression in mouse and NHP hepatocytes and improves vector potency.
- Author
-
Zakas PM, Cunningham SC, Doherty A, van Dijk EB, Ibraheim R, Yu S, Mekonnen BD, Lang B, English EJ, Sun G, Duncan MC, Benczkowski MS, Altshuler RC, Singh MJ, Kibbler ES, Tonga GY, Wang ZJ, Wang ZJ, Li G, An D, Rottman JB, Bhavsar Y, Purcell C, Jain R, Alberry R, Roquet N, Fu Y, Citorik RJ, Rubens JR, Holmes MC, Cotta-Ramusino C, Querbes W, Alexander IE, and Salomon WE
- Subjects
- Animals, Mice, Genetic Therapy methods, Humans, Gene Expression, Lipids chemistry, Disease Models, Animal, Gene Transfer Techniques, Ornithine Carbamoyltransferase genetics, Ornithine Carbamoyltransferase metabolism, Liposomes, Dependovirus genetics, Genetic Vectors genetics, Genetic Vectors administration & dosage, Hepatocytes metabolism, Transgenes, Transposases genetics, Transposases metabolism, Nanoparticles chemistry, RNA, Messenger genetics, RNA, Messenger metabolism
- Abstract
Recombinant adeno-associated virus (rAAV) vector gene delivery systems have demonstrated great promise in clinical trials but continue to face durability and dose-related challenges. Unlike rAAV gene therapy, integrating gene addition approaches can provide curative expression in mitotically active cells and pediatric populations. We explored a novel in vivo delivery approach based on an engineered transposase, Sleeping Beauty (SB100X), delivered as an mRNA within a lipid nanoparticle (LNP), in combination with an rAAV-delivered transposable transgene. This combinatorial approach achieved correction of ornithine transcarbamylase deficiency in the neonatal Spf
ash mouse model following a single delivery to dividing hepatocytes in the newborn liver. Correction remained stable into adulthood, while a conventional rAAV approach resulted in a return to the disease state. In non-human primates, integration by transposition, mediated by this technology, improved gene expression 10-fold over conventional rAAV-mediated gene transfer while requiring 5-fold less vector. Additionally, integration site analysis confirmed a random profile while specifically targeting TA dinucleotides across the genome. Together, these findings demonstrate that transposable elements can improve rAAV-delivered therapies by lowering the vector dose requirement and associated toxicity while expanding target cell types., Competing Interests: Declaration of interests All authors except for S.C.C., E.B.v.D., and I.E.A. are or were employees of Tessera Therapeutics and currently receive or previously received salary along with stock options as compensation for their employment. I.E.A. is a consultant of Tessera Therapeutics, and this work was performed under a Tessera Therapeutics-sponsored research agreement., (Copyright © 2024 The American Society of Gene and Cell Therapy. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
29. Formaldehyde regulates S -adenosylmethionine biosynthesis and one-carbon metabolism.
- Author
-
Pham VN, Bruemmer KJ, Toh JDW, Ge EJ, Tenney L, Ward CC, Dingler FA, Millington CL, Garcia-Prieto CA, Pulos-Holmes MC, Ingolia NT, Pontel LB, Esteller M, Patel KJ, Nomura DK, and Chang CJ
- Subjects
- Animals, Mice, Protein Isoforms antagonists & inhibitors, Protein Isoforms metabolism, Environmental Exposure, Humans, Hep G2 Cells, Carbon metabolism, Epigenesis, Genetic drug effects, S-Adenosylmethionine antagonists & inhibitors, S-Adenosylmethionine metabolism, Formaldehyde metabolism, Formaldehyde toxicity, Methionine Adenosyltransferase antagonists & inhibitors, Methionine Adenosyltransferase genetics, Methionine Adenosyltransferase metabolism, Cysteine metabolism
- Abstract
One-carbon metabolism is an essential branch of cellular metabolism that intersects with epigenetic regulation. In this work, we show how formaldehyde (FA), a one-carbon unit derived from both endogenous sources and environmental exposure, regulates one-carbon metabolism by inhibiting the biosynthesis of S -adenosylmethionine (SAM), the major methyl donor in cells. FA reacts with privileged, hyperreactive cysteine sites in the proteome, including Cys120 in S-adenosylmethionine synthase isoform type-1 (MAT1A). FA exposure inhibited MAT1A activity and decreased SAM production with MAT-isoform specificity. A genetic mouse model of chronic FA overload showed a decrease n SAM and in methylation on selected histones and genes. Epigenetic and transcriptional regulation of Mat1a and related genes function as compensatory mechanisms for FA-dependent SAM depletion, revealing a biochemical feedback cycle between FA and SAM one-carbon units.
- Published
- 2023
- Full Text
- View/download PDF
30. High salt intake activates the hypothalamic-pituitary-adrenal axis, amplifies the stress response, and alters tissue glucocorticoid exposure in mice.
- Author
-
Costello HM, Krilis G, Grenier C, Severs D, Czopek A, Ivy JR, Nixon M, Holmes MC, Livingstone DEW, Hoorn EJ, Dhaun N, and Bailey MA
- Subjects
- Humans, Mice, Animals, Male, Sodium Chloride, Dietary, Pituitary-Adrenal System metabolism, Mice, Inbred C57BL, Vasopressins genetics, Vasopressins metabolism, RNA, Messenger genetics, RNA, Messenger metabolism, Glucocorticoids metabolism, Hypothalamo-Hypophyseal System metabolism
- Abstract
Aims: High salt intake is common and contributes to poor cardiovascular health. Urinary sodium excretion correlates directly with glucocorticoid excretion in humans and experimental animals. We hypothesized that high salt intake activates the hypothalamic-pituitary-adrenal axis activation and leads to sustained glucocorticoid excess., Methods and Results: In male C57BL/6 mice, high salt intake for 2-8 weeks caused an increase in diurnal peak levels of plasma corticosterone. After 2 weeks, high salt increased Crh and Pomc mRNA abundance in the hypothalamus and anterior pituitary, consistent with basal hypothalamic-pituitary-adrenal axis activation. Additionally, high salt intake amplified glucocorticoid response to restraint stress, indicative of enhanced axis sensitivity. The binding capacity of Corticosteroid-Binding Globulin was reduced and its encoding mRNA downregulated in the liver. In the hippocampus and anterior pituitary, Fkbp5 mRNA levels were increased, indicating increased glucocorticoid exposure. The mRNA expression of the glucocorticoid-regenerating enzyme, 11β-hydroxysteroid dehydrogenase Type 1, was increased in these brain areas and in the liver. Sustained high salt intake activated a water conservation response by the kidney, increasing plasma levels of the vasopressin surrogate, copeptin. Increased mRNA abundance of Tonebp and Avpr1b in the anterior pituitary suggested that vasopressin signalling contributes to hypothalamic-pituitary-adrenal axis activation by high salt diet., Conclusion: Chronic high salt intake amplifies basal and stress-induced glucocorticoid levels and resets glucocorticoid biology centrally, peripherally and within cells., Competing Interests: Conflict of interest: N.D. has acted as a consultant for Retrophin. M.B. has consulted for River 2 Renal Corp., (© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
31. Off-the-shelf, steroid-resistant, IL13Rα2-specific CAR T cells for treatment of glioblastoma.
- Author
-
Brown CE, Rodriguez A, Palmer J, Ostberg JR, Naranjo A, Wagner JR, Aguilar B, Starr R, Weng L, Synold TW, Tran V, Wang S, Reik A, D'Apuzzo M, Ressler JA, Zhou Y, Mendel M, Gregory PD, Holmes MC, Tang WW, Forman SJ, Jensen MC, and Badie B
- Subjects
- Dexamethasone, Glucocorticoids, Humans, Immunotherapy, Adoptive, Steroids, T-Lymphocytes, Xenograft Model Antitumor Assays, Glioblastoma pathology, Interleukin-13 Receptor alpha2 Subunit
- Abstract
Background: Wide-spread application of chimeric antigen receptor (CAR) T cell therapy for cancer is limited by the current use of autologous CAR T cells necessitating the manufacture of individualized therapeutic products for each patient. To address this challenge, we have generated an off-the-shelf, allogeneic CAR T cell product for the treatment of glioblastoma (GBM), and present here the feasibility, safety, and therapeutic potential of this approach., Methods: We generated for clinical use a healthy-donor derived IL13Rα2-targeted CAR+ (IL13-zetakine+) cytolytic T-lymphocyte (CTL) product genetically engineered using zinc finger nucleases (ZFNs) to permanently disrupt the glucocorticoid receptor (GR) (GRm13Z40-2) and endow resistance to glucocorticoid treatment. In a phase I safety and feasibility trial we evaluated these allogeneic GRm13Z40-2 T cells in combination with intracranial administration of recombinant human IL-2 (rhIL-2; aldesleukin) in six patients with unresectable recurrent GBM that were maintained on systemic dexamethasone (4-12 mg/day)., Results: The GRm13Z40-2 product displayed dexamethasone-resistant effector activity without evidence for in vitro alloreactivity. Intracranial administration of GRm13Z40-2 in four doses of 108 cells over a two-week period with aldesleukin (9 infusions ranging from 2500-5000 IU) was well tolerated, with indications of transient tumor reduction and/or tumor necrosis at the site of T cell infusion in four of the six treated research subjects. Antibody reactivity against GRm13Z40-2 cells was detected in the serum of only one of the four tested subjects., Conclusions: This first-in-human experience establishes a foundation for future adoptive therapy studies using off-the-shelf, zinc-finger modified, and/or glucocorticoid resistant CAR T cells., (Published by Oxford University Press on behalf of the Society for Neuro-Oncology 2022.)
- Published
- 2022
- Full Text
- View/download PDF
32. In vivo partial cellular reprogramming enhances liver plasticity and regeneration.
- Author
-
Hishida T, Yamamoto M, Hishida-Nozaki Y, Shao C, Huang L, Wang C, Shojima K, Xue Y, Hang Y, Shokhirev M, Memczak S, Sahu SK, Hatanaka F, Ros RR, Maxwell MB, Chavez J, Shao Y, Liao HK, Martinez-Redondo P, Guillen-Guillen I, Hernandez-Benitez R, Esteban CR, Qu J, Holmes MC, Yi F, Hickey RD, Garcia PG, Delicado EN, Castells A, Campistol JM, Yu Y, Hargreaves DC, Asai A, Reddy P, Liu GH, and Izpisua Belmonte JC
- Subjects
- Animals, Cell Dedifferentiation, Hepatocytes metabolism, Liver Regeneration, Mammals, Mice, Cellular Reprogramming, Liver metabolism
- Abstract
Mammals have limited regenerative capacity, whereas some vertebrates, like fish and salamanders, are able to regenerate their organs efficiently. The regeneration in these species depends on cell dedifferentiation followed by proliferation. We generate a mouse model that enables the inducible expression of the four Yamanaka factors (Oct-3/4, Sox2, Klf4, and c-Myc, or 4F) specifically in hepatocytes. Transient in vivo 4F expression induces partial reprogramming of adult hepatocytes to a progenitor state and concomitantly increases cell proliferation. This is indicated by reduced expression of differentiated hepatic-lineage markers, an increase in markers of proliferation and chromatin modifiers, global changes in DNA accessibility, and an acquisition of liver stem and progenitor cell markers. Functionally, short-term expression of 4F enhances liver regenerative capacity through topoisomerase2-mediated partial reprogramming. Our results reveal that liver-specific 4F expression in vivo induces cellular plasticity and counteracts liver failure, suggesting that partial reprogramming may represent an avenue for enhancing tissue regeneration., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
33. Corticosteroid Receptors in Cardiac Health and Disease.
- Author
-
Ivy JR, Gray GA, Holmes MC, Denvir MA, and Chapman KE
- Subjects
- Animals, Myocytes, Cardiac metabolism, Receptors, Glucocorticoid physiology, Receptors, Thyroid Hormone metabolism, Heart Failure metabolism, Receptors, Mineralocorticoid
- Abstract
Nuclear receptors play a central role in both energy metabolism and cardiomyocyte death and survival in the heart. Recent evidence suggests they may also influence cardiomyocyte endowment. Although several members of the nuclear receptor family play key roles in heart maturation (including thyroid hormone receptors) and cardiac metabolism, here, the focus will be on the corticosteroid receptors, the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). The heart is an important target for the actions of corticosteroids, yet the homeostatic role of GR and MR in the healthy heart has been elusive. However, MR antagonists are important in the treatment of heart failure, a condition associated with mitochondrial dysfunction and energy failure in cardiomyocytes leading to mitochondria-initiated cardiomyocyte death (Ingwall and Weiss, Circ Res 95:135-145, 2014; Ingwall , Cardiovasc Res 81:412-419, 2009; Zhou and Tian , J Clin Invest 128:3716-3726, 2018). In contrast, animal studies suggest GR activation in cardiomyocytes has a cardioprotective role, including in heart failure., (© 2022. The Author(s), under exclusive license to Springer Nature Switzerland AG.)
- Published
- 2022
- Full Text
- View/download PDF
34. ZFN-mediated in vivo gene editing in hepatocytes leads to supraphysiologic α-Gal A activity and effective substrate reduction in Fabry mice.
- Author
-
Pagant S, Huston MW, Moreira L, Gan L, St Martin S, Sproul S, Holmes MC, Meyer K, Wechsler T, Desnick RJ, and Yasuda M
- Subjects
- Animals, Dependovirus genetics, Disease Models, Animal, Enzyme Activation, Gene Expression, Gene Transfer Techniques, Genetic Engineering, Genetic Therapy, Genetic Vectors genetics, Humans, Mice, Transgenes, Fabry Disease genetics, Fabry Disease therapy, Gene Editing, Hepatocytes metabolism, Zinc Finger Nucleases metabolism, alpha-Galactosidase genetics, alpha-Galactosidase metabolism
- Abstract
Fabry disease, a lysosomal storage disorder resulting from the deficient activity of α-galactosidase A (α-Gal A), is characterized by cardiac, renal, and/or cerebrovascular disease due to progressive accumulation of the enzyme's substrates, globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3). We report here the preclinical evaluation of liver-targeted in vivo genome editing using zinc-finger nuclease (ZFN) technology to insert the human α-galactosidase A (hGLA) cDNA into the albumin "safe harbor" locus of Fabry mice, thereby generating an albumin-α-Gal A fusion protein. The mature α-Gal A protein is secreted into the circulation for subsequent mannose-6-phosphate receptor-mediated tissue uptake. Donor vector optimization studies showed that replacing the hGLA cDNA signal peptide sequence with that of human iduronate 2-sulfatase (IDS) achieved higher transgene expression. Intravenous adeno-associated virus (AAV) 2/8-mediated co-delivery of the IDS-hGLA donor and ZFNs targeting the albumin locus resulted in continuous, supraphysiological plasma and tissue α-Gal A activities, which essentially normalized Gb3 and Lyso-Gb3 levels in key tissues of pathology. Notably, this was achieved with <10% of hepatocytes being edited to express hGLA, occurring mostly via non-homologous end joining (NHEJ) rather than homology-directed repair (HDR). These studies indicate that ZFN-mediated in vivo genome editing has the potential to be an effective one-time therapy for Fabry disease., Competing Interests: Declaration of interests R.J.D. is a consultant to Genzyme-Sanofi and Sangamo Therapeutics, Inc. He owns founder stock in Amicus Therapeutics and options for Sangamo Therapeutics, Inc. and receives royalties from Genzyme-Sanofi. R.J.D. and M.Y. received a research grant from Sangamo Therapeutics, Inc. to perform these studies. M.W.H., S.S.M., S.S., K.M., M.C.H., and T.W. are full-time employees and/or shareholders of Sangamo Therapeutics, Inc., (Copyright © 2021 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
35. Altered hypothalamic DNA methylation and stress-induced hyperactivity following early life stress.
- Author
-
Fitzgerald E, Sinton MC, Wernig-Zorc S, Morton NM, Holmes MC, Boardman JP, and Drake AJ
- Subjects
- Adult, Animals, Humans, Hypothalamus, Infant, Newborn, Infant, Premature, Maternal Deprivation, Mice, Young Adult, Adverse Childhood Experiences, DNA Methylation
- Abstract
Exposure to early life stress (ELS) during childhood or prenatally increases the risk of future psychiatric disorders. The effect of stress exposure during the neonatal period is less well understood. In preterm infants, exposure to invasive procedures is associated with altered brain development and future stress responses suggesting that the neonatal period could be a key time for the programming of mental health. Previous studies suggest that ELS affects the hypothalamic epigenome, making it a good candidate to mediate these effects. In this study, we used a mouse model of early life stress (modified maternal separation; MMS). We hypothesised MMS would affect the hypothalamic transcriptome and DNA methylome, and impact on adult behaviour. MMS involved repeated stimulation of pups for 1.5 h/day, whilst separated from their mother, from postnatal day (P) 4-6. 3'mRNA sequencing and DNA methylation immunoprecipitation (meDIP) sequencing were performed on hypothalamic tissue at P6. Behaviour was assessed with the elevated plus, open field mazes and in-cage monitoring at 3-4 months of age. MMS was only associated with subtle changes in gene expression, but there were widespread alterations in DNA methylation. Notably, differentially methylated regions were enriched for synapse-associated loci. MMS resulted in hyperactivity in the elevated plus and open field mazes, but in-cage monitoring revealed that this was not representative of habitual hyperactivity. ELS has marked effects on DNA methylation in the hypothalamus in early life and results in stress-specific hyperactivity in young adulthood. These results have implications for the understanding of ELS-mediated effects on brain development.
- Published
- 2021
- Full Text
- View/download PDF
36. Vortioxetine ameliorates anhedonic-like behaviour and promotes strategic cognitive performance in a rodent touchscreen task.
- Author
-
Martis LS, Højgaard K, Holmes MC, Elfving B, and Wiborg O
- Subjects
- Anhedonia drug effects, Animals, Cerebral Cortex drug effects, Cerebral Cortex physiology, Gene Expression drug effects, Hippocampus drug effects, Hippocampus physiology, Learning, Male, Rats, Long-Evans, Stress, Physiological, Rats, Antidepressive Agents pharmacology, Cognition drug effects, Vortioxetine pharmacology
- Abstract
Depression-associated cognitive impairments are among the most prevalent and persistent symptoms during remission from a depressive episode and a major risk factor for relapse. Consequently, development of antidepressant drugs, which also alleviate cognitive impairments, is vital. One such potential antidepressant is vortioxetine that has been postulated to exhibit both antidepressant and pro-cognitive effects. Hence, we tested vortioxetine for combined antidepressant and pro-cognitive effects in male Long-Evans rats exposed to the chronic mild stress (CMS) paradigm. This well-established CMS paradigm evokes cognitive deficits in addition to anhedonia, a core symptom of depression. Learning and memory performance was assessed in the translational touchscreen version of the paired-associates learning task. To identify the mechanistic underpinning of the neurobehavioural results, transcriptional profiling of genes involved in the stress response, neuronal plasticity and genes of broad relevance in neuropsychiatric pathologies were assessed. Vortioxetine substantially relieved the anhedonic-like state in the CMS rats and promoted acquisition of the cognitive test independent of hedonic phenotype, potentially due to an altered cognitive strategy. Minor alterations in gene expression profiling in prefrontal cortex and hippocampus were found. In summary, our findings suggest that vortioxetine exhibits an antidepressant effect as well as behavioural changes in a translational learning task.
- Published
- 2021
- Full Text
- View/download PDF
37. Persistent repression of tau in the brain using engineered zinc finger protein transcription factors.
- Author
-
Wegmann S, DeVos SL, Zeitler B, Marlen K, Bennett RE, Perez-Rando M, MacKenzie D, Yu Q, Commins C, Bannon RN, Corjuc BT, Chase A, Diez L, Nguyen HB, Hinkley S, Zhang L, Goodwin A, Ledeboer A, Lam S, Ankoudinova I, Tran H, Scarlott N, Amora R, Surosky R, Miller JC, Robbins AB, Rebar EJ, Urnov FD, Holmes MC, Pooler AM, Riley B, Zhang HS, and Hyman BT
- Subjects
- Amyloid beta-Peptides metabolism, Animals, Brain metabolism, Dependovirus genetics, Dependovirus metabolism, Disease Models, Animal, Mice, Plaque, Amyloid pathology, Zinc Fingers genetics, tau Proteins genetics, tau Proteins metabolism, Alzheimer Disease genetics, Alzheimer Disease pathology, Alzheimer Disease therapy, Transcription Factors genetics, Transcription Factors metabolism
- Abstract
Neuronal tau reduction confers resilience against β-amyloid and tau-related neurotoxicity in vitro and in vivo. Here, we introduce a novel translational approach to lower expression of the tau gene MAPT at the transcriptional level using gene-silencing zinc finger protein transcription factors (ZFP-TFs). Following a single administration of adeno-associated virus (AAV), either locally into the hippocampus or intravenously to enable whole-brain transduction, we selectively reduced tau messenger RNA and protein by 50 to 80% out to 11 months, the longest time point studied. Sustained tau lowering was achieved without detectable off-target effects, overt histopathological changes, or molecular alterations. Tau reduction with AAV ZFP-TFs was able to rescue neuronal damage around amyloid plaques in a mouse model of Alzheimer's disease (APP/PS1 line). The highly specific, durable, and controlled knockdown of endogenous tau makes AAV-delivered ZFP-TFs a promising approach for the treatment of tau-related human brain diseases., (Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works. Distributed under a Creative Commons Attribution NonCommercial License 4.0 (CC BY-NC).)
- Published
- 2021
- Full Text
- View/download PDF
38. AAV2/6 Gene Therapy in a Murine Model of Fabry Disease Results in Supraphysiological Enzyme Activity and Effective Substrate Reduction.
- Author
-
Yasuda M, Huston MW, Pagant S, Gan L, St Martin S, Sproul S, Richards D, Ballaron S, Hettini K, Ledeboer A, Falese L, Cao L, Lu Y, Holmes MC, Meyer K, Desnick RJ, and Wechsler T
- Abstract
Fabry disease is an X-linked lysosomal storage disorder caused by mutations in the alpha-galactosidase A ( GLA ) gene, which encodes the exogalactosyl hydrolase, alpha-galactosidase A (α-Gal A). Deficient α-Gal A activity results in the progressive, systemic accumulation of its substrates, globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), leading to renal, cardiac, and/or cerebrovascular disease and early demise. The current standard treatment for Fabry disease is enzyme replacement therapy, which necessitates lifelong biweekly infusions of recombinant enzyme. A more long-lasting treatment would benefit Fabry patients. Here, a gene therapy approach using an episomal adeno-associated viral 2/6 (AAV2/6) vector that encodes the human GLA cDNA driven by a liver-specific expression cassette was evaluated in a Fabry mouse model that lacks α-Gal A activity and progressively accumulates Gb3 and Lyso-Gb3 in plasma and tissues. A detailed 3-month pharmacology and toxicology study showed that administration of a clinical-scale-manufactured AAV2/6 vector resulted in markedly increased plasma and tissue α-Gal A activities, and essentially normalized Gb3 and Lyso-Gb3 at key sites of pathology. Further optimization of vector design identified the clinical lead vector, ST-920, which produced several-fold higher plasma and tissue α-Gal A activity levels with a good safety profile. Together, these studies provide the basis for the clinical development of ST-920., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
39. PTBP1 mRNA isoforms and regulation of their translation.
- Author
-
Arake de Tacca LM, Pulos-Holmes MC, Floor SN, and Cate JHD
- Subjects
- 3' Untranslated Regions, 5' Untranslated Regions, HEK293 Cells, Humans, Heterogeneous-Nuclear Ribonucleoproteins genetics, Polypyrimidine Tract-Binding Protein genetics, Protein Biosynthesis, RNA Isoforms genetics, RNA, Messenger genetics
- Abstract
Polypyrimidine tract-binding proteins (PTBPs) are RNA binding proteins that regulate a number of posttranscriptional events. Human PTBP1 transits between the nucleus and cytoplasm and is thought to regulate RNA processes in both. However, information about PTBP1 mRNA isoforms and regulation of PTPB1 expression remains incomplete. Here we mapped the major PTBP1 mRNA isoforms in HEK293T cells and identified alternative 5' and 3' untranslated regions (5'-UTRs, 3'-UTRs), as well as alternative splicing patterns in the protein coding region. We also assessed how the observed PTBP1 mRNA isoforms contribute to PTBP1 expression in different phases of the cell cycle. Previously, PTBP1 mRNAs were shown to crosslink to eukaryotic translation initiation factor 3 (eIF3). We find that eIF3 binds differently to each PTBP1 mRNA isoform in a cell cycle dependent manner. We also observe a strong correlation between eIF3 binding to PTBP1 mRNAs and repression of PTBP1 levels during the S phase of the cell cycle. Our results provide evidence of translational regulation of PTBP1 protein levels during the cell cycle, which may affect downstream regulation of alternative splicing and translation mediated by PTBP1 protein isoforms., (© 2019 Arake de Tacca et al.; Published by Cold Spring Harbor Laboratory Press for the RNA Society.)
- Published
- 2019
- Full Text
- View/download PDF
40. Repression of ferritin light chain translation by human eIF3.
- Author
-
Pulos-Holmes MC, Srole DN, Juarez MG, Lee AS, McSwiggen DT, Ingolia NT, and Cate JH
- Subjects
- 5' Untranslated Regions, Cell Line, Humans, Polymorphism, Single Nucleotide, Apoferritins biosynthesis, Down-Regulation, Eukaryotic Initiation Factor-3 metabolism, Protein Biosynthesis
- Abstract
A central problem in human biology remains the discovery of causal molecular links between mutations identified in genome-wide association studies (GWAS) and their corresponding disease traits. This challenge is magnified for variants residing in non-coding regions of the genome. Single-nucleotide polymorphisms (SNPs) in the 5' untranslated region (5'-UTR) of the ferritin light chain ( FTL ) gene that cause hyperferritinemia are reported to disrupt translation repression by altering iron regulatory protein (IRP) interactions with the FTL mRNA 5'-UTR. Here, we show that human eukaryotic translation initiation factor 3 (eIF3) acts as a distinct repressor of FTL mRNA translation, and eIF3-mediated FTL repression is disrupted by a subset of SNPs in FTL that cause hyperferritinemia. These results identify a direct role for eIF3-mediated translational control in a specific human disease., Competing Interests: MP, DS, MJ, AL, DM, NI, JC No competing interests declared, (© 2019, Pulos-Holmes et al.)
- Published
- 2019
- Full Text
- View/download PDF
41. Humanized Mouse Model of HIV-1 Latency with Enrichment of Latent Virus in PD-1 + and TIGIT + CD4 T Cells.
- Author
-
Llewellyn GN, Seclén E, Wietgrefe S, Liu S, Chateau M, Pei H, Perkey K, Marsden MD, Hinkley SJ, Paschon DE, Holmes MC, Zack JA, Louie SG, Haase AT, and Cannon PM
- Subjects
- Animals, Anti-Retroviral Agents pharmacology, CD4-Positive T-Lymphocytes metabolism, CD4-Positive T-Lymphocytes virology, Disease Models, Animal, HIV Infections virology, HIV Seropositivity drug therapy, HIV-1 pathogenicity, Humans, Mice, Programmed Cell Death 1 Receptor immunology, Receptors, Immunologic immunology, Transcription Activator-Like Effector Nucleases immunology, Virus Activation, Virus Replication, HIV-1 immunology, Virus Latency immunology, Virus Latency physiology
- Abstract
Combination anti-retroviral drug therapy (ART) potently suppresses HIV-1 replication but does not result in virus eradication or a cure. A major contributing factor is the long-term persistence of a reservoir of latently infected cells. To study this reservoir, we established a humanized mouse model of HIV-1 infection and ART suppression based on an oral ART regimen. Similar to humans, HIV-1 levels in the blood of ART-treated animals were frequently suppressed below the limits of detection. However, the limited timeframe of the mouse model and the small volume of available samples makes it a challenging model with which to achieve full viral suppression and to investigate the latent reservoir. We therefore used an ex vivo latency reactivation assay that allows a semiquantitative measure of the latent reservoir that establishes in individual animals, regardless of whether they are treated with ART. Using this assay, we found that latently infected human CD4 T cells can be readily detected in mouse lymphoid tissues and that latent HIV-1 was enriched in populations expressing markers of T cell exhaustion, PD-1 and TIGIT. In addition, we were able to use the ex vivo latency reactivation assay to demonstrate that HIV-specific TALENs can reduce the fraction of reactivatable virus in the latently infected cell population that establishes in vivo , supporting the use of targeted nuclease-based approaches for an HIV-1 cure. IMPORTANCE HIV-1 can establish latent infections that are not cleared by current antiretroviral drugs or the body's immune responses and therefore represent a major barrier to curing HIV-infected individuals. However, the lack of expression of viral antigens on latently infected cells makes them difficult to identify or study. Here, we describe a humanized mouse model that can be used to detect latent but reactivatable HIV-1 in both untreated mice and those on ART and therefore provides a simple system with which to study the latent HIV-1 reservoir and the impact of interventions aimed at reducing it., (Copyright © 2019 American Society for Microbiology.)
- Published
- 2019
- Full Text
- View/download PDF
42. Non-viral Delivery of Zinc Finger Nuclease mRNA Enables Highly Efficient In Vivo Genome Editing of Multiple Therapeutic Gene Targets.
- Author
-
Conway A, Mendel M, Kim K, McGovern K, Boyko A, Zhang L, Miller JC, DeKelver RC, Paschon DE, Mui BL, Lin PJC, Tam YK, Barbosa C, Redelmeier T, Holmes MC, and Lee G
- Subjects
- Animals, Cells, Cultured, Dependovirus genetics, Female, Gene Knockout Techniques, Genetic Vectors, Hepatocytes metabolism, Introns genetics, Lipids chemistry, Male, Mice, Mice, Inbred C57BL, Prealbumin genetics, Proprotein Convertase 9 genetics, RNA, Messenger genetics, Transgenes genetics, Zinc Finger Nucleases pharmacology, Drug Delivery Systems methods, Gene Editing methods, Nanoparticles administration & dosage, RNA, Messenger administration & dosage, Zinc Finger Nucleases administration & dosage
- Abstract
It has previously been shown that engineered zinc finger nucleases (ZFNs) can be packaged into adeno-associated viruses (AAVs) and delivered intravenously into mice, non-human primates, and most recently, humans to induce highly efficient therapeutic genome editing in the liver. Lipid nanoparticles (LNPs) are synthetic delivery vehicles that enable repeat administration and are not limited by the presence of preexisting neutralizing antibodies in patients. Here, we show that mRNA encoding ZFNs formulated into LNP can enable >90% knockout of gene expression in mice by targeting the TTR or PCSK9 gene, at mRNA doses 10-fold lower than has ever been reported. Additionally, co-delivering mRNA-LNP containing ZFNs targeted to intron 1 of the ALB locus with AAV packaged with a promoterless human IDS or FIX therapeutic transgene can result in high levels of targeted integration and subsequent therapeutically relevant levels of protein expression in mice. Finally, we show repeat administration of ZFN mRNA-LNP after a single AAV donor dose results in significantly increased levels of genome editing and transgene expression compared to a single dose. These results demonstrate LNP-mediated ZFN mRNA delivery can drive highly efficient levels of in vivo genome editing and can potentially offer a new treatment modality for a variety of diseases., (Copyright © 2019 The American Society of Gene and Cell Therapy. Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
43. Diversifying the structure of zinc finger nucleases for high-precision genome editing.
- Author
-
Paschon DE, Lussier S, Wangzor T, Xia DF, Li PW, Hinkley SJ, Scarlott NA, Lam SC, Waite AJ, Truong LN, Gandhi N, Kadam BN, Patil DP, Shivak DA, Lee GK, Holmes MC, Zhang L, Miller JC, and Rebar EJ
- Subjects
- Base Pairing, Base Sequence, DNA-Directed RNA Polymerases genetics, DNA-Directed RNA Polymerases metabolism, Deoxyribonucleases, Type II Site-Specific metabolism, Escherichia coli genetics, Escherichia coli metabolism, Genetic Loci, Genomic Library, Humans, INDEL Mutation, K562 Cells, Peptide Library, Plasmids chemistry, Plasmids metabolism, Transformation, Genetic, Viral Proteins genetics, Viral Proteins metabolism, Zinc Finger Nucleases metabolism, Deoxyribonucleases, Type II Site-Specific genetics, Gene Editing methods, Genome, Human, Protein Engineering methods, Zinc Finger Nucleases genetics
- Abstract
Genome editing for therapeutic applications often requires cleavage within a narrow sequence window. Here, to enable such high-precision targeting with zinc-finger nucleases (ZFNs), we have developed an expanded set of architectures that collectively increase the configurational options available for design by a factor of 64. These new architectures feature the functional attachment of the FokI cleavage domain to the amino terminus of one or both zinc-finger proteins (ZFPs) in the ZFN dimer, as well as the option to skip bases between the target triplets of otherwise adjacent fingers in each zinc-finger array. Using our new architectures, we demonstrate targeting of an arbitrarily chosen 28 bp genomic locus at a density that approaches 1.0 (i.e., efficient ZFNs available for targeting almost every base step). We show that these new architectures may be used for targeting three loci of therapeutic significance with a high degree of precision, efficiency, and specificity.
- Published
- 2019
- Full Text
- View/download PDF
44. BDNF +/- rats exhibit depressive phenotype and altered expression of genes relevant in mood disorders.
- Author
-
Martis LS, Wiborg O, Holmes MC, and Harris AP
- Subjects
- Animals, Depressive Disorder, Major metabolism, Female, Heterozygote, Male, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Neuregulin-1 genetics, Neuregulin-1 metabolism, Rats, Rats, Sprague-Dawley, Receptors, Glucocorticoid genetics, Receptors, Glucocorticoid metabolism, Tacrolimus Binding Proteins genetics, Tacrolimus Binding Proteins metabolism, Brain metabolism, Brain-Derived Neurotrophic Factor genetics, Depressive Disorder, Major genetics, Phenotype
- Abstract
Major depressive disorder (MDD) is a leading contributor to the global burden of disease. However, the causal relationship of risk factors, such as genetic predisposition or experience of augmented stress, remain unknown. Numerous studies in humans and rodents have implicated brain-derived neurotrophic factor (BDNF) in MDD pathology, as a genetic risk factor and a factor regulated by stress. Until now, the majority of preclinical studies have employed genetically modified mice as their model of choice. However, mice display a limited behavioural repertoire and lack expression of circulating BDNF, which is present in rats and humans. Therefore, heterozygous BDNF (BDNF
+/- ) rats were tested for affective behaviours and accompanying expression of key genes associated with affective disorders in the brain. We found that BDNF+/- rats, which have reduced BDNF levels in brain and plasma, displayed symptoms of anhedonia, a core symptom of MDD, and anxiety-like behaviour, but no behavioural despair or cognitive impairments. This was accompanied by changes in the expression of genes that are implicated in modulation of the stress response and affective disorders. Hence, glucocorticoid receptor, neuregulin 1 and disrupted-in-schizophrenia 1 gene expression were upregulated in the prefrontal cortex of BDFN+/- rats, whereas FK506 binding protein 5 levels were decreased in the hippocampus. We conclude that a reduction in BDNF levels alters expression of genes associated with affective disorders, which may contribute to the development of depressive-like symptoms., (© 2018 John Wiley & Sons Ltd and International Behavioural and Neural Genetics Society.)- Published
- 2019
- Full Text
- View/download PDF
45. ZFN-Mediated In Vivo Genome Editing Corrects Murine Hurler Syndrome.
- Author
-
Ou L, DeKelver RC, Rohde M, Tom S, Radeke R, St Martin SJ, Santiago Y, Sproul S, Przybilla MJ, Koniar BL, Podetz-Pedersen KM, Laoharawee K, Cooksley RD, Meyer KE, Holmes MC, McIvor RS, Wechsler T, and Whitley CB
- Subjects
- Animals, Disease Models, Animal, Enzyme Replacement Therapy, Female, Glycosaminoglycans metabolism, Iduronidase metabolism, Lysosomal Storage Diseases drug therapy, Lysosomal Storage Diseases metabolism, Lysosomal Storage Diseases therapy, Male, Mice, Mucopolysaccharidosis I drug therapy, Mucopolysaccharidosis I metabolism, Zinc Finger Nucleases genetics, Gene Editing methods, Genetic Therapy methods, Mucopolysaccharidosis I therapy, Zinc Finger Nucleases metabolism
- Abstract
Mucopolysaccharidosis type I (MPS I) is a severe disease due to deficiency of the lysosomal hydrolase α-L-iduronidase (IDUA) and the subsequent accumulation of the glycosaminoglycans (GAG), leading to progressive, systemic disease and a shortened lifespan. Current treatment options consist of hematopoietic stem cell transplantation, which carries significant mortality and morbidity risk, and enzyme replacement therapy, which requires lifelong infusions of replacement enzyme; neither provides adequate therapy, even in combination. A novel in vivo genome-editing approach is described in the murine model of Hurler syndrome. A corrective copy of the IDUA gene is inserted at the albumin locus in hepatocytes, leading to sustained enzyme expression, secretion from the liver into circulation, and subsequent uptake systemically at levels sufficient for correction of metabolic disease (GAG substrate accumulation) and prevention of neurobehavioral deficits in MPS I mice. This study serves as a proof-of-concept for this platform-based approach that should be broadly applicable to the treatment of a wide array of monogenic diseases., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
46. The Role of 11β-Hydroxy Steroid Dehydrogenase Type 2 in Glucocorticoid Programming of Affective and Cognitive Behaviours.
- Author
-
Shearer FJG, Wyrwoll CS, and Holmes MC
- Subjects
- Animals, Female, Humans, Pregnancy, Prenatal Exposure Delayed Effects, 11-beta-Hydroxysteroid Dehydrogenase Type 2 metabolism, Brain growth & development, Brain metabolism, Cognition physiology, Emotions physiology, Glucocorticoids metabolism
- Abstract
Developmental exposure to stress hormones, i.e. glucocorticoids, is central to the process of prenatal programming of later-life health. Glucocorticoid overexposure, through stress or exogenous glucocorticoids, results in a reduced birthweight, as well as affective and neuropsychiatric outcomes in adults, combined with altered hypothalamus-pituitary-adrenal (HPA) axis activity. As such, glucocorticoids are tightly regulated during development through the presence of the metabolizing enzyme 11β-hydroxysteroid dehydrogenase type 2 (HSD2). HSD2 is highly expressed in 2 hubs during development, i.e. the placenta and the fetus itself, protecting the fetus from inappropriate glucocorticoid exposure early in gestation. Through manipulation of HSD2 expression in the mouse placenta and fetal tissues, we are able to determine the relative contribution of glucocorticoid exposure in each compartment. Feto-placental HSD2 deletion resulted in a reduced birthweight and the development of anxiety- and depression-like behaviours in adult mice. The placenta itself is altered by glucocorticoid overexposure, which causes reduced placental weight and vascular arborisation. Furthermore, altered flow and resistance in the umbilical vessels and modification of fetal heart function and development are observed. However, brain-specific HSD2 removal (HSD2BKO) also generated adult phenotypes of depressive-like behaviour and memory deficits, demonstrating the importance of fetal brain HSD2 expression in development. In this review we will discuss potential mechanisms underpinning early-life programming of adult neuropsychiatric disorders and the novel therapeutic potential of statins., (© 2019 S. Karger AG, Basel.)
- Published
- 2019
- Full Text
- View/download PDF
47. Resilient and depressive-like rats show distinct cognitive impairments in the touchscreen paired-associates learning (PAL) task.
- Author
-
Martis LS, Brision C, Holmes MC, and Wiborg O
- Subjects
- Anhedonia, Animals, Cognitive Dysfunction etiology, Conditioning, Operant, Depression complications, Depressive Disorder, Major complications, Depressive Disorder, Major psychology, Disease Models, Animal, Male, Rats, Long-Evans, Stress, Psychological complications, Cognitive Dysfunction psychology, Depression psychology, Paired-Associate Learning, Resilience, Psychological, Stress, Psychological psychology
- Abstract
Depression-associated cognitive impairments persist after remission from affective symptoms of major depressive disorder (MDD), decreasing quality of life and increasing risk of relapse in patients. Conventional antidepressants are ineffective in restoring cognitive functions. Therefore, novel antidepressants with improved efficacy for ameliorating cognitive symptoms are required. For tailoring such antidepressants, translational animal models are in demand. The chronic mild stress (CMS) model is a well-validated preclinical model of depression and known for eliciting the MDD core symptom "anhedonia" in stress-susceptible rats. Thus, cognitive performance was assessed in rats susceptible (depressive-like) or resilient to CMS and in unchallenged controls. The rodent analogue of the human touchscreen Paired-Associates Learning (PAL) task was used for cognitive assessment. Both stress groups exhibited a lack of response inhibition compared to controls while only the depressive-like group was impaired in task acquisition. The results indicate that cognitive deficits specifically associate with the anhedonic-like state rather than being a general consequence of stress exposure. Hence, we propose that the application of a translational touchscreen task on the etiologically valid CMS model, displaying depression-associated cognitive impairments, provides a novel platform for pro-cognitive and clinically pertinent antidepressant drug screening., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
48. Disruption of the BCL11A Erythroid Enhancer Reactivates Fetal Hemoglobin in Erythroid Cells of Patients with β-Thalassemia Major.
- Author
-
Psatha N, Reik A, Phelps S, Zhou Y, Dalas D, Yannaki E, Levasseur DN, Urnov FD, Holmes MC, and Papayannopoulou T
- Abstract
In the present report, we carried out clinical-scale editing in adult mobilized CD34+ hematopoietic stem and progenitor cells (HSPCs) using zinc-finger nuclease-mediated disruption of BCL11a to upregulate the expression of γ-globin (fetal hemoglobin). In these cells, disruption of the erythroid-specific enhancer of the BCL11A gene increased endogenous γ-globin expression to levels that reached or exceeded those observed following knockout of the BCL11A coding region without negatively affecting survival or in vivo long-term proliferation of edited HSPCs and other lineages. In addition, BCL11A enhancer modification in mobilized CD34+ cells from patients with β-thalassemia major resulted in a readily detectable γ-globin increase with a preferential increase in G-gamma, leading to an improved phenotype and, likely, a survival advantage for maturing erythroid cells after editing. Furthermore, we documented that both normal and β-thalassemia HSPCs not only can be efficiently expanded ex vivo after editing but can also be successfully edited post-expansion, resulting in enhanced early in vivo engraftment compared with unexpanded cells. Overall, this work highlights a novel and effective treatment strategy for correcting the β-thalassemia phenotype by genome editing.
- Published
- 2018
- Full Text
- View/download PDF
49. Molecular Evidence of Genome Editing in a Mouse Model of Immunodeficiency.
- Author
-
Abdul-Razak HH, Rocca CJ, Howe SJ, Alonso-Ferrero ME, Wang J, Gabriel R, Bartholomae CC, Gan CHV, Garín MI, Roberts A, Blundell MP, Prakash V, Molina-Estevez FJ, Pantoglou J, Guenechea G, Holmes MC, Gregory PD, Kinnon C, von Kalle C, Schmidt M, Bueren JA, Thrasher AJ, and Yáñez-Muñoz RJ
- Subjects
- Animals, DNA-Activated Protein Kinase genetics, Disease Models, Animal, Humans, Mice, Mice, SCID, Nuclear Proteins genetics, Gene Editing, Severe Combined Immunodeficiency genetics
- Abstract
Genome editing is the introduction of directed modifications in the genome, a process boosted to therapeutic levels by designer nucleases. Building on the experience of ex vivo gene therapy for severe combined immunodeficiencies, it is likely that genome editing of haematopoietic stem/progenitor cells (HSPC) for correction of inherited blood diseases will be an early clinical application. We show molecular evidence of gene correction in a mouse model of primary immunodeficiency. In vitro experiments in DNA-dependent protein kinase catalytic subunit severe combined immunodeficiency (Prkdc scid) fibroblasts using designed zinc finger nucleases (ZFN) and a repair template demonstrated molecular and functional correction of the defect. Following transplantation of ex vivo gene-edited Prkdc scid HSPC, some of the recipient animals carried the expected genomic signature of ZFN-driven gene correction. In some primary and secondary transplant recipients we detected double-positive CD4/CD8 T-cells in thymus and single-positive T-cells in blood, but no other evidence of immune reconstitution. However, the leakiness of this model is a confounding factor for the interpretation of the possible T-cell reconstitution. Our results provide support for the feasibility of rescuing inherited blood disease by ex vivo genome editing followed by transplantation, and highlight some of the challenges.
- Published
- 2018
- Full Text
- View/download PDF
50. Differential impact of transplantation on peripheral and tissue-associated viral reservoirs: Implications for HIV gene therapy.
- Author
-
Peterson CW, Wang J, Deleage C, Reddy S, Kaur J, Polacino P, Reik A, Huang ML, Jerome KR, Hu SL, Holmes MC, Estes JD, and Kiem HP
- Subjects
- Animals, Anti-Retroviral Agents therapeutic use, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Macaca nemestrina, Male, Simian Acquired Immunodeficiency Syndrome genetics, Simian Acquired Immunodeficiency Syndrome virology, Transplantation, Autologous, Virus Latency, Virus Replication, Genetic Therapy, Hematopoietic Stem Cell Transplantation, Receptors, CCR5 genetics, Simian Acquired Immunodeficiency Syndrome therapy, Simian Immunodeficiency Virus physiology, Viral Load physiology
- Abstract
Autologous transplantation and engraftment of HIV-resistant cells in sufficient numbers should recapitulate the functional cure of the Berlin Patient, with applicability to a greater number of infected individuals and with a superior safety profile. A robust preclinical model of suppressed HIV infection is critical in order to test such gene therapy-based cure strategies, both alone and in combination with other cure strategies. Here, we present a nonhuman primate (NHP) model of latent infection using simian/human immunodeficiency virus (SHIV) and combination antiretroviral therapy (cART) in pigtail macaques. We demonstrate that transplantation of CCR5 gene-edited hematopoietic stem/progenitor cells (HSPCs) persist in infected and suppressed animals, and that protected cells expand through virus-dependent positive selection. CCR5 gene-edited cells are readily detectable in tissues, namely those closely associated with viral reservoirs such as lymph nodes and gastrointestinal tract. Following autologous transplantation, tissue-associated SHIV DNA and RNA levels in suppressed animals are significantly reduced (p ≤ 0.05), relative to suppressed, untransplanted control animals. In contrast, the size of the peripheral reservoir, measured by QVOA, is variably impacted by transplantation. Our studies demonstrate that CCR5 gene editing is equally feasible in infected and uninfected animals, that edited cells persist, traffic to, and engraft in tissue reservoirs, and that this approach significantly reduces secondary lymphoid tissue viral reservoir size. Our robust NHP model of HIV gene therapy and viral persistence can be immediately applied to the investigation of combinatorial approaches that incorporate anti-HIV gene therapy, immune modulators, therapeutic vaccination, and latency reversing agents.
- 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.