Network


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

Hotspot


Dive into the research topics where Maria Anna Zipeto is active.

Publication


Featured researches published by Maria Anna Zipeto.


Proceedings of the National Academy of Sciences of the United States of America | 2013

ADAR1 promotes malignant progenitor reprogramming in chronic myeloid leukemia

Qingfei Jiang; Leslie Crews; Christian L. Barrett; Hye Jung E Chun; Angela C. Court; Jane M. Isquith; Maria Anna Zipeto; Daniel Goff; Mark D. Minden; Anil Sadarangani; Jessica Rusert; Kim Hien T Dao; Sheldon R. Morris; Lawrence S.B. Goldstein; Marco A. Marra; Kelly A. Frazer; Catriona Jamieson

The molecular etiology of human progenitor reprogramming into self-renewing leukemia stem cells (LSC) has remained elusive. Although DNA sequencing has uncovered spliceosome gene mutations that promote alternative splicing and portend leukemic transformation, isoform diversity also may be generated by RNA editing mediated by adenosine deaminase acting on RNA (ADAR) enzymes that regulate stem cell maintenance. In this study, whole-transcriptome sequencing of normal, chronic phase, and serially transplantable blast crisis chronic myeloid leukemia (CML) progenitors revealed increased IFN-γ pathway gene expression in concert with BCR-ABL amplification, enhanced expression of the IFN-responsive ADAR1 p150 isoform, and a propensity for increased adenosine-to-inosine RNA editing during CML progression. Lentiviral overexpression experiments demonstrate that ADAR1 p150 promotes expression of the myeloid transcription factor PU.1 and induces malignant reprogramming of myeloid progenitors. Moreover, enforced ADAR1 p150 expression was associated with production of a misspliced form of GSK3β implicated in LSC self-renewal. Finally, functional serial transplantation and shRNA studies demonstrate that ADAR1 knockdown impaired in vivo self-renewal capacity of blast crisis CML progenitors. Together these data provide a compelling rationale for developing ADAR1-based LSC detection and eradication strategies.


Cell Stem Cell | 2016

ADAR1 Activation Drives Leukemia Stem Cell Self-Renewal by Impairing Let-7 Biogenesis

Maria Anna Zipeto; Angela C. Court; Anil Sadarangani; Nathaniel Delos Santos; Larisa Balaian; Hye-Jung Chun; Gabriel Pineda; Sheldon R. Morris; Cayla N. Mason; Ifat Geron; Christian L. Barrett; Daniel Goff; Russell Wall; Maurizio Pellecchia; Mark D. Minden; Kelly A. Frazer; Marco A. Marra; Leslie Crews; Qingfei Jiang; Catriona Jamieson

Post-transcriptional adenosine-to-inosine RNA editing mediated by adenosine deaminase acting on RNA1 (ADAR1) promotes cancer progression and therapeutic resistance. However, ADAR1 editase-dependent mechanisms governing leukemia stem cell (LSC) generation have not been elucidated. In blast crisis chronic myeloid leukemia (BC CML), we show that increased JAK2 signaling and BCR-ABL1 amplification activate ADAR1. In a humanized BC CML mouse model, combined JAK2 and BCR-ABL1 inhibition prevents LSC self-renewal commensurate with ADAR1 downregulation. Lentiviral ADAR1 wild-type, but not an editing-defective ADAR1(E912A) mutant, induces self-renewal gene expression and impairs biogenesis of stem cell regulatory let-7 microRNAs. Combined RNA sequencing, qRT-PCR, CLIP-ADAR1, and pri-let-7 mutagenesis data suggest that ADAR1 promotes LSC generation via let-7 pri-microRNA editing and LIN28B upregulation. A small-molecule tool compound antagonizes ADAR1s effect on LSC self-renewal in stromal co-cultures and restores let-7 biogenesis. Thus, ADAR1 activation represents a unique therapeutic vulnerability in LSCs with active JAK2 signaling.


Trends in Molecular Medicine | 2015

RNA rewriting, recoding, and rewiring in human disease.

Maria Anna Zipeto; Qingfei Jiang; Etienne Melese; Catriona Jamieson

ADAR (adenosine deAminase acting on RNA) editases catalyze the deamination of adenosine to inosine (A-to-I), a post-transcriptional modification that alters coding and non-coding RNA stability and function. ADAR editases such as ADAR1 have recently been shown to play a key role in normal stem cell maintenance. While ADAR mutations are associated with hereditary autoimmune diseases such as Aicardi-Goutières syndrome, ADAR copy-number alterations and editase activation have been associated with progression of a broad array of malignancies. In this review we discuss evidence linking aberrant A-to-I editing to cancer and other degenerative diseases, and the mechanisms that may be targeted by novel therapeutic strategies.


Cell Stem Cell | 2016

RNA Splicing Modulation Selectively Impairs Leukemia Stem Cell Maintenance in Secondary Human AML

Leslie Crews; Larisa Balaian; Nathaniel Delos Santos; Heather Leu; Angela C. Court; Elisa Lazzari; Anil Sadarangani; Maria Anna Zipeto; James J. La Clair; Reymundo Villa; Anna A. Kulidjian; Rainer Storb; Sheldon R. Morris; Edward D. Ball; Michael D. Burkart; Catriona Jamieson

Age-related human hematopoietic stem cell (HSC) exhaustion and myeloid-lineage skewing promote oncogenic transformation of hematopoietic progenitor cells into therapy-resistant leukemia stem cells (LSCs) in secondary acute myeloid leukemia (AML). While acquisition of clonal DNA mutations has been linked to increased rates of secondary AML for individuals older than 60 years, the contribution of RNA processing alterations to human hematopoietic stem and progenitor aging and LSC generation remains unclear. Comprehensive RNA sequencing and splice-isoform-specific PCR uncovered characteristic RNA splice isoform expression patterns that distinguished normal young and aged human stem and progenitor cells (HSPCs) from malignant myelodysplastic syndrome (MDS) and AML progenitors. Inxa0splicing reporter assays and pre-clinical patient-derived AMLxa0models, treatment with a pharmacologic splicing modulator, 17S-FD-895, reversed pro-survival splice isoform switching and significantly impaired LSC maintenance. Therapeutic splicing modulation, together with monitoring splice isoform biomarkers of healthy HSPC aging versus LSC generation, may be employed safely and effectively to prevent relapse, the leading cause of leukemia-related mortality.


Journal of Translational Medicine | 2015

An RNA editing fingerprint of cancer stem cell reprogramming

Leslie Crews; Qingfei Jiang; Maria Anna Zipeto; Elisa Lazzari; Angela C. Court; Shawn Ali; Christian L. Barrett; Kelly A. Frazer; Catriona Jamieson

BackgroundDeregulation of RNA editing by adenosine deaminases acting on dsRNA (ADARs) has been implicated in the progression of diverse human cancers including hematopoietic malignancies such as chronic myeloid leukemia (CML). Inflammation-associated activation of ADAR1 occurs in leukemia stem cells specifically in the advanced, often drug-resistant stage of CML known as blast crisis. However, detection of cancer stem cell-associated RNA editing by RNA sequencing in these rare cell populations can be technically challenging, costly and requires PCR validation. The objectives of this study were to validate RNA editing of a subset of cancer stem cell-associated transcripts, and to develop a quantitative RNA editing fingerprint assay for rapid detection of aberrant RNA editing in human malignancies.MethodsTo facilitate quantification of cancer stem cell-associated RNA editing in exons and intronic or 3UTR primate-specific Alu sequences using a sensitive, cost-effective method, we established an in vitro RNA editing model and developed a sensitive RNA editing fingerprint assay that employs a site-specific quantitative PCR (RESSq-PCR) strategy. This assay was validated in a stably-transduced human leukemia cell line, lentiviral-ADAR1 transduced primary hematopoietic stem and progenitor cells, and in primary human chronic myeloid leukemia stem cells.ResultsIn lentiviral ADAR1-expressing cells, increased RNA editing of MDM2, APOBEC3D, GLI1 and AZIN1 transcripts was detected by RESSq-PCR with improved sensitivity over sequencing chromatogram analysis. This method accurately detected cancer stem cell-associated RNA editing in primary chronic myeloid leukemia samples, establishing a cancer stem cell-specific RNA editing fingerprint of leukemic transformation that will support clinical development of novel diagnostic tools to predict and prevent cancer progression.ConclusionsRNA editing quantification enables rapid detection of malignant progenitors signifying cancer progression and therapeutic resistance, and will aid future RNA editing inhibitor development efforts.


Cancer Research | 2017

Abstract 299: RNA editing enzyme ADAR1 accelerates normal hematopoiesis cell cycle by regulation microRNA biogenesis

Qingfei Jiang; Maria Anna Zipeto; Nathan Delos Santos; Sheldon R. Morris; Catriona Jamieson

Compelling murine studies demonstrate that adenosine-to-inosine (A-to-I) RNA editing mediated by adenosine deaminase associated with RNA1 (ADAR1) is vital for both fetal and adult hematopoiesis. While genetic ablation of ADAR1 leads to murine embryonic lethality due to severe defects in erythropoiesis, conditional deletion in the hematopoietic system impairs maintenance indicative of cell type and context specific roles for ADAR1 in cell fate specification and self-renewal. By regulating mRNA and microRNA (miRNA) stability, ADAR1 exhibit wide-ranging effects on embryonic development and stem cell regulation. We have previously shown that inflammation-responsive ADAR1 plays important roles in both stem cell differentiation and self-renewal in CML (chronic myeloid leukemia) disease progression. Here, we describe a novel function of ADAR1 in cell cycle regulation of normal hematopoietic stem cell and progenitors (HSPC) by regulation of miRNA biogenesis. Our results demonstrated that ADAR1 induces G0 to G1 phase transition in normal cord blood HSPCs, as demonstrated by elevated expression of Ki67, reduced DiR signal, and enhanced in vivo cord blood engraftment. Cell cycle qRT-qPCR microarray of 84 cell cycle transcripts and whole transcriptome RNA-sequencing analysis of KEGG cell cycle pathway indicate that several cell cycle genes are differentially expression upon overexpression of ADAR1 WT or an A-to-I editing deficient ADAR1 mutant (ADAR1E912A). We previously demonstrated that impaired biogenesis of let-7 miRNAs by ADAR1 WT induces enhanced self-renewal in cord blood CD34+ HSPCs. To determine the miRNA targets of ADAR1-mediated RNA editing, we performed miRNome miScript PCR array of 1008 miRNA candidates in cord blood CD34+ HSPCs overexpressing ADAR1 WT or ADAR1E912A. Total of 263 miRNAs were differentially expressed (142 upregulated and 121 downregulated) by comparing ADAR1 WT to the backbone control. Interestingly, ADAR1E912A mutant also exhibit A-to-I editing independent regulation of miRNAs (307 upregulated and 59 downregulated). We found that the expression of miR-26a-5p, a miRNA frequently downregulated in leukemia, is inhibited by ADAR1-mediated RNA editing. ADAR1 directly binds and edits the DROSHA cleavage site of primary miR-26a transcript, thereby prevent miR26a-5p maturation. Moreover, lentiviral expression of mature miR26-5p reverses the effect of ADAR1 WT, including enhanced CDKN1A expression, inhibition of cord blood proliferation in vivo, as well as reduced HSC self-renewal as measured by colony-formation assay. Our finding suggests carefully regulated A-to-I editing by ADAR1 is essential for the maintenance of proper cell proliferation in HSC. For future study, it will be interesting to investigate if the elevated expression of ADAR1 in CML BC LSC contributes to false regulation of cell cycle that leads to the expansion of malignant leukemia stem cells. Citation Format: Qingfei Jiang, Maria Anna Zipeto, Nathan Delos Santos, Sheldon Morris, Catriona Jamieson. RNA editing enzyme ADAR1 accelerates normal hematopoiesis cell cycle by regulation microRNA biogenesis [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 299. doi:10.1158/1538-7445.AM2017-299


Cancer Research | 2014

Abstract 375: A novel diagnostic assay for detection of primate-specific RNA editing events in leukemia stem cells

Leslie Crews; Qingfei Jiang; Maria Anna Zipeto; Angela C. Court; Christian L. Barrett; Marco A. Marra; Kelly A. Frazer; Catriona Jamieson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CAnnIntroductionnnThe adenosine deaminase acting on RNA (ADAR) family of RNA editases has been linked to the pathogenesis of diverse malignancies, including leukemia, breast cancer and hepatocellular carcinoma. We previously showed that human leukemia stem cells (LSC) from blast crisis (BC) chronic myeloid leukemia (CML) patients harbor increased ADAR1 expression compared with normal and chronic phase (CP) progenitors. Whole transcriptome RNA sequencing (RNA-Seq) revealed increased adenosine to inosine (A-to-I) RNA editing during CML progression concentrated within primate specific Alu-containing transcripts. However, detection of RNA editing by RNA-Seq in rare cell populations can be technically challenging, costly and requires PCR validation. Thus, the objectives of this study were to validate RNA editing of a subset of these LSC-associated transcripts in the context of lentivirally enforced ADAR1 expression, and to develop an RNA editing reporter reporter assay in human leukemia cells and a qPCR-based diagnostic test to rapidly detect CSC-associated aberrant RNA editing.nnMethodsnnThe BCR-ABL+ human leukemia cell line K562 was stably transduced with lentiviral human ADAR1 or vector. FACS-purified K562-ADAR1 cells were transfected with a luciferase-based reporter vector to confirm RNA editing activity. Two genes, MDM2 and APOBEC3D, were selected from our previous RNA-Seq studies of BC progenitors (Jiang et al, 2013). Targeted sequencing was performed on high fidelity PCR products using primers flanking each of 2 editing sites in each gene. RNA editing-specific qPCR primers were designed for each editing site using an allele-specific strategy that detects cDNA containing either an A or G(I) representing an RNA editing event. Both targeted sequencing and qPCR were used to detect RNA editing in K562-ADAR1 and primary cord blood-derived hematopoietic stem cells (HSC) lentivirally transduced with ADAR1.nnResultsnnLentivirally enforced ADAR1 expression promoted RNA editing activity as measured by luciferase reporter activity. Increased A-to-I changes in MDM2 and APOBEC3D were confirmed by targeted sequencing. In independent experiments, RNA editing site-specific qRT-PCR accurately detected RNA editing in K562-ADAR1 cells (n=3) and in primary HSC overexpressing ADAR1 (n=4). Site-specific primers distinguished G(I) bases at RNA editing sites in cDNA and as predicted gave no signal in gDNA. Relative A-to-I RNA editing ratios were increased by 2 to 3 fold in ADAR1-expressing cells at all four sites.nnConclusionsnnThese results set the stage for development of primate-specific RNA editing as a novel diagnostic strategy for clinical LSC detection and identify ADAR1 as a potential therapeutic target in LSC. These data shed new light on the mechanisms of ADAR1-mediated generation of malignant progenitors that drive therapeutic resistance, disease progression and relapse in CML and may be applicable to other CSC-driven malignancies.nnCitation Format: Leslie A. Crews, Qingfei Jiang, Maria A. Zipeto, Angela C. Court, Christian L. Barrett, Marco A. Marra, Kelly A. Frazer, Catriona H. M. Jamieson. A novel diagnostic assay for detection of primate-specific RNA editing events in leukemia stem cells. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 375. doi:10.1158/1538-7445.AM2014-375


Cancer Research | 2014

Abstract 1912: ADAR1-mediated microRNA regulation and blast crisis leukemia stem cell generation in chronic myeloid leukemia

Maria Anna Zipeto; Qingfei Jiang; Leslie C. Robertson; Catriona Jamieson

Proceedings: AACR Annual Meeting 2014; April 5-9, 2014; San Diego, CAnnIntroductionnnChronic myeloid leukemia (CML) was one of the first malignancies shown to be initiated in hematopoietic stem cells by the BCR-ABL1 oncogene and sustained in blast crisis (BC) by progenitor cells that co-opt stem cell properties and behave like leukemia stem cells (BC LSC). The BCR-ABL fusion oncogene encodes a constitutively active tyrosine kinase BCR-ABL. Although tyrosine kinase inhibitor (TKI) therapy targeting BCR-ABL suppresses CML during the chronic phase (CP), progenitors undergo expansion as a consequence of subsequent genetic and epigenetic alterations that fuel blast crisis (BC) transformation, BC LSC generation and TKI resistance. We have recently shown that adenosine to insoine (A-to-I) RNA editing activity, mediated by the adenosine deaminase acting on dsRNAs ADAR1, is enhanced in BC CML and drives leukemic progression by inducing myeloid progenitor reprogramming and enhancing self-renewal. ADAR1 targets double-stranded RNA hairpin-containing loop structures, such as microRNAs (miRNAs). MiRNAs, by operating through base-pairing with complementary sequences within a mRNA molecule lead to mRNA degradation and gene silencing. Emerging evidence suggests miRNAs alterations introduced at the pri-miRNA or pre-miRNA level by A-to-I editing by ADAR1 may be responsible for broad changes in the transcriptome that drives LSC maintenance. However, the overall change in edited miRNA profiles in CP CML compared with BC progenitors is unclear. In this study we aimed to investigate the role of ADAR1 in driving malignant progression by focusing on its regulation of miRNA expression in CML.nnMethods and ResultsnnMicroRNA expression profiles were evaluated in CD34+ cells derived from CP and BC CML patient samples as well as cord blood. MiRNA PCR array (Qiagen) allowed us to identify differentially expressed miRNAs that were subsequently validated by q-PCR with specific primers targeting individual miRNA. We observed the downregulation of 13 miRNA in CML BC compared to CML CP progenitors. Lentiviral overexpression of ADAR1 in CD34+ cord blood cells led to significant downregulation of 17 miRNAs, including members of the let-7 family, and other miRNAs involved in the regulation of self-renewal. Interestingly, BC CD34+ cells showed a miRNA profile similar to that of lentiviral ADAR1 transduced cord blood progenitors. Lentiviral overexpression of ADAR1 in CD34+ chronic phase CML cells, led to a significant downregulation of 14 miRNAs compared with their lentiviral backbone transduced counterparts.nnConclusionnnOur data suggest that ADAR1 contributes to BC LSC generation by downregulating miRNAs that target stem cell regulatory gene products. A further characterization of ADAR-editing related miRNA signatures may enable BC LSC detection and early intervention with combined BCR-ABL1 and ADAR1-targeting strategies that may obviate CML relapse and progression.nnCitation Format: Maria Zipeto, Qingfei Jiang, Leslie Crews Robertson, Catriona HM Jamieson. ADAR1-mediated microRNA regulation and blast crisis leukemia stem cell generation in chronic myeloid leukemia. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1912. doi:10.1158/1538-7445.AM2014-1912


Cancer Research | 2016

Abstract 915: RNA processing signatures of normal versus malignant progenitor cell aging predict leukemia stem cell sensitivity to RNA splicing modulation

Leslie Crews; Larisa Balaian; Heather Leu; Nathaniel Delos Santos; Angela C. Court; Anil Sadarangani; Maria Anna Zipeto; James J. La Clair; Reymundo Villa; Sheldon R. Morris; Rainer Storb; Anna A. Kulidjian; Edward D. Ball; Michael D. Burkart; Catriona Jamieson


Blood | 2016

RNA Editing Enzyme ADAR1 Accelerates Normal Hematopoiesis Cell Cycle By Regulating microRNA Biogenesis

Qingfei Jiang; Maria Anna Zipeto; Nathaniel Delos Santos; Heather Leu; Jane M. Isquith; Etienne Atien; Catriona Jamieson

Collaboration


Dive into the Maria Anna Zipeto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Qingfei Jiang

University of California

View shared research outputs
Top Co-Authors

Avatar

Leslie Crews

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anil Sadarangani

Pontifical Catholic University of Chile

View shared research outputs
Top Co-Authors

Avatar

Elisa Lazzari

University of California

View shared research outputs
Researchain Logo
Decentralizing Knowledge