Network


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

Hotspot


Dive into the research topics where Zulema Romero is active.

Publication


Featured researches published by Zulema Romero.


Blood | 2015

Correction of the sickle-cell disease mutation in human hematopoietic stem/progenitor cells

Megan D. Hoban; Gregory J. Cost; Matthew C. Mendel; Zulema Romero; Michael L. Kaufman; Alok V. Joglekar; Michelle Ho; Dianne Lumaquin; David Gray; Georgia R. Lill; Aaron R. Cooper; Fabrizia Urbinati; Shantha Senadheera; Allen Zhu; Pei-Qi Liu; David Paschon; Lei Zhang; Edward J. Rebar; Andrew Wilber; Xiaoyan Wang; Philip D. Gregory; Michael C. Holmes; Andreas Reik; Roger P. Hollis; Donald B. Kohn

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Journal of Clinical Investigation | 2013

β-globin gene transfer to human bone marrow for sickle cell disease

Zulema Romero; Fabrizia Urbinati; Sabine Geiger; Aaron R. Cooper; Jennifer Wherley; Michael L. Kaufman; Roger P. Hollis; Rafael Ruiz de Assin; Shantha Senadheera; Arineh Sahagian; Xiangyang Jin; Alyse Gellis; Xiaoyan Wang; David W. Gjertson; Satiro deOliveira; Pamela Kempert; Sally Shupien; Hisham Abdel-Azim; Mark C. Walters; Herbert J. Meiselman; Rosalinda B. Wenby; Theresa Gruber; Victor J. Marder; Thomas D. Coates; Donald B. Kohn

Autologous hematopoietic stem cell gene therapy is an approach to treating sickle cell disease (SCD) patients that may result in lower morbidity than allogeneic transplantation. We examined the potential of a lentiviral vector (LV) (CCL-βAS3-FB) encoding a human hemoglobin (HBB) gene engineered to impede sickle hemoglobin polymerization (HBBAS3) to transduce human BM CD34+ cells from SCD donors and prevent sickling of red blood cells produced by in vitro differentiation. The CCL-βAS3-FB LV transduced BM CD34+ cells from either healthy or SCD donors at similar levels, based on quantitative PCR and colony-forming unit progenitor analysis. Consistent expression of HBBAS3 mRNA and HbAS3 protein compromised a fourth of the total β-globin-like transcripts and hemoglobin (Hb) tetramers. Upon deoxygenation, a lower percentage of HBBAS3-transduced red blood cells exhibited sickling compared with mock-transduced cells from sickle donors. Transduced BM CD34+ cells were transplanted into immunodeficient mice, and the human cells recovered after 2-3 months were cultured for erythroid differentiation, which showed levels of HBBAS3 mRNA similar to those seen in the CD34+ cells that were directly differentiated in vitro. These results demonstrate that the CCL-βAS3-FB LV is capable of efficient transfer and consistent expression of an effective anti-sickling β-globin gene in human SCD BM CD34+ progenitor cells, improving physiologic parameters of the resulting red blood cells.


Molecular Therapy | 2016

CRISPR/Cas9-Mediated Correction of the Sickle Mutation in Human CD34+ cells

Megan D. Hoban; Dianne Lumaquin; Caroline Y. Kuo; Zulema Romero; Joseph Long; Michelle Ho; Courtney S. Young; Michelle Mojadidi; Sorel Fitz-Gibbon; Aaron R. Cooper; Georgia R. Lill; Fabrizia Urbinati; Beatriz Campo-Fernandez; Carmen Flores Bjurström; Matteo Pellegrini; Roger P. Hollis; Donald B. Kohn

Targeted genome editing technology can correct the sickle cell disease mutation of the β-globin gene in hematopoietic stem cells. This correction supports production of red blood cells that synthesize normal hemoglobin proteins. Here, we demonstrate that Transcription Activator-Like Effector Nucleases (TALENs) and the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 nuclease system can target DNA sequences around the sickle-cell mutation in the β-globin gene for site-specific cleavage and facilitate precise correction when a homologous donor template is codelivered. Several pairs of TALENs and multiple CRISPR guide RNAs were evaluated for both on-target and off-target cleavage rates. Delivery of the CRISPR/Cas9 components to CD34+ cells led to over 18% gene modification in vitro. Additionally, we demonstrate the correction of the sickle cell disease mutation in bone marrow derived CD34+ hematopoietic stem and progenitor cells from sickle cell disease patients, leading to the production of wild-type hemoglobin. These results demonstrate correction of the sickle mutation in patient-derived CD34+ cells using CRISPR/Cas9 technology.


Experimental Hematology | 2015

Potentially therapeutic levels of anti-sickling globin gene expression following lentivirus-mediated gene transfer in sickle cell disease bone marrow CD34+ cells

Fabrizia Urbinati; Phillip W. Hargrove; Sabine Geiger; Zulema Romero; Jennifer Wherley; Michael L. Kaufman; Roger P. Hollis; Christopher B. Chambers; Derek A. Persons; Donald B. Kohn; Andrew Wilber

Sickle cell disease (SCD) can be cured by allogeneic hematopoietic stem cell transplant. However, this is only possible when a matched donor is available, making the development of gene therapy using autologous hematopoietic stem cells a highly desirable alternative. We used a culture model of human erythropoiesis to directly compare two insulated, self-inactivating, and erythroid-specific lentiviral vectors, encoding for γ-globin (V5m3-400) or a modified β-globin (βAS3-FB) for production of antisickling hemoglobin (Hb) and correction of red cell deformability after deoxygenation. Bone marrow CD34+ cells from three SCD patients were transduced using V5m3-400 or βAS3-FB and compared with mock-transduced SCD or healthy donor CD34+ cells. Lentiviral transduction did not impair cell growth or differentiation, as gauged by proliferation and acquisition of erythroid markers. Vector copy number averaged approximately one copy per cell, and corrective globin mRNA levels were increased more than sevenfold over mock-transduced controls. Erythroblasts derived from healthy donor and mock-transduced SCD cells produced a low level of fetal Hb that was increased to 23.6 ± 4.1% per vector copy for cells transduced with V5m3-400. Equivalent levels of modified normal adult Hb of 17.6 ± 3.8% per vector copy were detected for SCD cells transduced with βAS3-FB. These levels of antisickling Hb production were sufficient to reduce sickling of terminal-stage red blood cells upon deoxygenation. We concluded that the achieved levels of fetal Hb and modified normal adult Hb would likely prove therapeutic to SCD patients who lack matched donors.


Stem Cells | 2015

Enrichment of Human Hematopoietic Stem/Progenitor Cells Facilitates Transduction for Stem Cell Gene Therapy

Kismet M Baldwin; Fabrizia Urbinati; Zulema Romero; Beatriz Campo-Fernandez; Michael L. Kaufman; Aaron R. Cooper; Katelyn Masiuk; Roger P. Hollis; Donald B. Kohn

Autologous hematopoietic stem cell (HSC) gene therapy for sickle cell disease has the potential to treat this illness without the major immunological complications associated with allogeneic transplantation. However, transduction efficiency by β‐globin lentiviral vectors using CD34‐enriched cell populations is suboptimal and large vector production batches may be needed for clinical trials. Transducing a cell population more enriched for HSC could greatly reduce vector needs and, potentially, increase transduction efficiency. CD34+/CD38− cells, comprising ∼1%–3% of all CD34+ cells, were isolated from healthy cord blood CD34+ cells by fluorescence‐activated cell sorting and transduced with a lentiviral vector expressing an antisickling form of beta‐globin (CCL‐βAS3‐FB). Isolated CD34+/CD38− cells were able to generate progeny over an extended period of long‐term culture (LTC) compared to the CD34+ cells and required up to 40‐fold less vector for transduction compared to bulk CD34+ preparations containing an equivalent number of CD34+/CD38− cells. Transduction of isolated CD34+/CD38− cells was comparable to CD34+ cells measured by quantitative PCR at day 14 with reduced vector needs, and average vector copy/cell remained higher over time for LTC initiated from CD34+/38− cells. Following in vitro erythroid differentiation, HBBAS3 mRNA expression was similar in cultures derived from CD34+/CD38− cells or unfractionated CD34+ cells. In vivo studies showed equivalent engraftment of transduced CD34+/CD38− cells when transplanted in competition with 100‐fold more CD34+/CD38+ cells. This work provides initial evidence for the beneficial effects from isolating human CD34+/CD38− cells to use significantly less vector and potentially improve transduction for HSC gene therapy. Stem Cells 2015;33:1532–1542


Molecular therapy. Methods & clinical development | 2015

The human ankyrin 1 promoter insulator sustains gene expression in a β-globin lentiviral vector in hematopoietic stem cells.

Zulema Romero; Beatriz Campo-Fernandez; Jennifer Wherley; Michael L. Kaufman; Fabrizia Urbinati; Aaron R. Cooper; Megan D. Hoban; Kismet M Baldwin; Dianne Lumaquin; Xiaoyan Wang; Shantha Senadheera; Roger P. Hollis; Donald B. Kohn

Lentiviral vectors designed for the treatment of the hemoglobinopathies require the inclusion of regulatory and strong enhancer elements to achieve sufficient expression of the β-globin transgene. Despite the inclusion of these elements, the efficacy of these vectors may be limited by transgene silencing due to the genomic environment surrounding the integration site. Barrier insulators can be used to give more consistent expression and resist silencing even with lower vector copies. Here, the barrier activity of an insulator element from the human ankyrin-1 gene was analyzed in a lentiviral vector carrying an antisickling human β-globin gene. Inclusion of a single copy of the Ankyrin insulator did not affect viral titer, and improved the consistency of expression from the vector in murine erythroleukemia cells. The presence of the Ankyrin insulator element did not change transgene expression in human hematopoietic cells in short-term erythroid culture or in vivo in primary murine transplants. However, analysis in secondary recipients showed that the lentiviral vector with the Ankyrin element preserved transgene expression, whereas expression from the vector lacking the Ankyrin insulator decreased in secondary recipients. These studies demonstrate that the Ankyrin insulator may improve long-term β-globin expression in hematopoietic stem cells for gene therapy of hemoglobinopathies.


Current protocols in stem cell biology | 2016

Delivery of Genome Editing Reagents to Hematopoietic Stem/Progenitor Cells.

Megan D. Hoban; Zulema Romero; Gregory J. Cost; Matthew C. Mendel; Michael C. Holmes; Donald B. Kohn

This unit describes the protocol for the delivery of reagents for targeted genome editing to CD34(+) hematopoietic stem/progenitor cells (HSPCs). Specifically, this unit focuses on the process of thawing and pre-stimulating CD34(+) HSPCs, as well as the details of their electroporation with in vitro-transcribed mRNA-encoding site-specific nucleases [in this case zinc-finger nucleases (ZFNs)]. In addition, discussed is delivery of a gene editing donor template in the form of an oligonucleotide or integrase-defective lentiviral vector (IDLV). Finally, an analysis of cell survival following treatment and downstream culture conditions are presented. While optimization steps might be needed for each specific application with respect to nuclease and donor template amount, adherence to this protocol will serve as an excellent starting point for this further work.


Cytotherapy | 2017

Preclinical studies for a phase 1 clinical trial of autologous hematopoietic stem cell gene therapy for sickle cell disease

Fabrizia Urbinati; Jennifer Wherley; Sabine Geiger; Beatriz Campo Fernandez; Michael L. Kaufman; Aaron R. Cooper; Zulema Romero; Filippo Marchioni; Lilith Reeves; Elizabeth J. Read; Barbara Nowicki; Elke Grassman; Shivkumar Viswanathan; Xiaoyan Wang; Roger P. Hollis; Donald B. Kohn

BACKGROUND AIMS Gene therapy by autologous hematopoietic stem cell transplantation (HSCT) represents a new approach to treat sickle cell disease (SCD). Optimization of the manufacture, characterization and testing of the transduced hematopoietic stem cell final cell product (FCP), as well as an in depth in vivo toxicology study, are critical for advancing this approach to clinical trials. METHODS Data are shown to evaluate and establish the feasibility of isolating, transducing with the Lenti/βAS3-FB vector and cryopreserving CD34+ cells from human bone marrow (BM) at clinical scale. In vitro and in vivo characterization of the FCP was performed, showing that all the release criteria were successfully met. In vivo toxicology studies were conducted to evaluate potential toxicity of the Lenti/βAS3-FB LV in the context of a murine BM transplant. RESULTS Primary and secondary transplantation did not reveal any toxicity from the lentiviral vector. Additionally, vector integration site analysis of murine and human BM cells did not show any clonal skewing caused by insertion of the Lenti/βAS3-FB vector in cells from primary and secondary transplanted mice. CONCLUSIONS We present here a complete protocol, thoroughly optimized to manufacture, characterize and establish safety of a FCP for gene therapy of SCD.


Molecular Therapy | 2017

Characterization of Gene Alterations following Editing of the β-Globin Gene Locus in Hematopoietic Stem/Progenitor Cells

Joseph Long; Megan D. Hoban; Aaron R. Cooper; Michael L. Kaufman; Caroline Y. Kuo; Beatriz Campo-Fernandez; Dianne Lumaquin; Roger P. Hollis; Xiaoyan Wang; Donald B. Kohn; Zulema Romero

The use of engineered nucleases combined with a homologous DNA donor template can result in targeted gene correction of the sickle cell disease mutation in hematopoietic stem and progenitor cells. However, because of the high homology between the adjacent human β- and δ-globin genes, off-target cleavage is observed at δ-globin when using some endonucleases targeted to the sickle mutation in β-globin. Introduction of multiple double-stranded breaks by endonucleases has the potential to induce intergenic alterations. Using a novel droplet digital PCR assay and high-throughput sequencing, we characterized the frequency of rearrangements between the β- and δ-globin paralogs when delivering these nucleases. Pooled CD34+ cells and colony-forming units from sickle bone marrow were treated with nuclease only or including a donor template and then analyzed for potential gene rearrangements. It was observed that, in pooled CD34+ cells and colony-forming units, the intergenic β-δ-globin deletion was the most frequent rearrangement, followed by inversion of the intergenic fragment, with the inter-chromosomal translocation as the least frequent. No rearrangements were observed when endonuclease activity was restricted to on-target β-globin cleavage. These findings demonstrate the need to develop site-specific endonucleases with high specificity to avoid unwanted gene alterations.


bioRxiv | 2018

In vivo selection for corrected β-globin alleles after CRISPR/Cas9 editing in human sickle hematopoietic stem cells enhances therapeutic potential

Wendy Magis; Mark A. DeWitt; Stacia K. Wyman; Jonathan Vu; Seok-Jin Heo; Shirley J Shao; Fiona Hennig; Zulema Romero; Beatriz Campo-Fernandez; Matthew S. McNeill; Garret R. Rettig; Yongming Sun; Patrick J. Lau; Yu Wang; Mark A. Behlke; Donald B. Kohn; Dario Boffelli; Mark C. Walters; Jacob E. Corn; David I. K. Martin

Sickle Cell Disease (SCD), one of the world’s most common genetic disorders, causes anemia and progressive multiorgan damage that typically shortens lifespan by decades; currently there is no broadly applicable curative therapy. Here we show that Cas9 RNP-mediated gene editing with an ssDNA oligonucleotide donor yields more than 20% correction of the sickle mutation in long-term engrafting human HSCs. Using RNA-seq, we further find that in vivo erythroid differentiation markedly enriches for cells carrying corrected ß-globin alleles. Adoption of a high-fidelity Cas9 variant demonstrates that this approach can yield efficient editing with almost no off-target events. These findings indicate that the sickle mutation can be corrected in human HSCs at levels that are likely to be curative if translated into a therapy.ABSTRACT Sickle Cell Disease (SCD), one of the world’s most common genetic disorders, causes anemia and progressive multiorgan damage that typically shortens lifespan by decades; currently there is no broadly applicable curative therapy. Here we show that Cas9 RNP-mediated gene editing with an ssDNA oligonucleotide donor yields markerless correction of the sickle mutation in more than 30% of long-term engrafting human hematopoietic stem cells (HSCs), using a selection-free protocol that is directly applicable to a clinical setting. We further find that in vivo erythroid differentiation markedly enriches for corrected s-globin alleles. Adoption of a high-fidelity Cas9 variant demonstrates that this approach can yield efficient editing with almost no off-target events. These findings indicate that the sickle mutation can be corrected in human HSCs at curative levels with a streamlined protocol that is ready to be translated into a therapy. ONE SENTENCE SUMMARY Cas9-mediated correction of the sickle mutation in human hematopoietic stem cells can be accomplished at curative levels.

Collaboration


Dive into the Zulema Romero's collaboration.

Top Co-Authors

Avatar

Donald B. Kohn

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Megan D. Hoban

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xiaoyan Wang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge