Network


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

Hotspot


Dive into the research topics where Jaira F. de Vasconcellos is active.

Publication


Featured researches published by Jaira F. de Vasconcellos.


Blood | 2013

LIN28B-mediated expression of fetal hemoglobin and production of fetal-like erythrocytes from adult human erythroblasts ex vivo

Y. Terry Lee; Jaira F. de Vasconcellos; Joan Yuan; Colleen Byrnes; Seung-Jae Noh; Emily Riehm Meier; Ki Soon Kim; Antoinette Rabel; Megha Kaushal; Stefan A. Muljo; Jeffery L. Miller

Reactivation of fetal hemoglobin (HbF) holds therapeutic potential for sickle cell disease and β-thalassemias. In human erythroid cells and hematopoietic organs, LIN28B and its targeted let-7 microRNA family, demonstrate regulated expression during the fetal-to-adult developmental transition. To explore the effects of LIN28B in human erythroid cell development, lentiviral transduction was used to knockdown LIN28B expression in erythroblasts cultured from human umbilical cord CD34+ cells. The subsequent reduction in LIN28B expression caused increased expression of let-7 and significantly reduced HbF expression. Conversely, LIN28B overexpression in cultured adult erythroblasts reduced the expression of let-7 and significantly increased HbF expression. Cellular maturation was maintained including enucleation. LIN28B expression in adult erythroblasts increased the expression of γ-globin, and the HbF content of the cells rose to levels >30% of their hemoglobin. Expression of carbonic anhydrase I, glucosaminyl (N-acetyl) transferase 2, and miR-96 (three additional genes marking the transition from fetal-to-adult erythropoiesis) were reduced by LIN28B expression. The transcription factor BCL11A, a well-characterized repressor of γ-globin expression, was significantly down-regulated. Independent of LIN28B, experimental suppression of let-7 also reduced BCL11A expression and significantly increased HbF expression. LIN28B expression regulates HbF levels and causes adult human erythroblasts to differentiate with a more fetal-like phenotype.


Blood | 2015

Inhibition of G9a methyltransferase stimulates fetal hemoglobin production by facilitating LCR/γ-globin looping

Ivan Krivega; Colleen Byrnes; Jaira F. de Vasconcellos; Y. Terry Lee; Megha Kaushal; Ann Dean; Jeffery L. Miller

Induction of fetal hemoglobin (HbF) production in adult erythrocytes can reduce the severity of sickle cell disease and β-thalassemia. Transcription of β-globin genes is regulated by the distant locus control region (LCR), which is brought into direct gene contact by the LDB1/GATA-1/TAL1/LMO2-containing complex. Inhibition of G9a H3K9 methyltransferase by the chemical compound UNC0638 activates fetal and represses adult β-globin gene expression in adult human hematopoietic precursor cells, but the underlying mechanisms are unclear. Here we studied UNC0638 effects on β-globin gene expression using ex vivo differentiation of CD34(+) erythroid progenitor cells from peripheral blood of healthy adult donors. UNC0638 inhibition of G9a caused dosed accumulation of HbF up to 30% of total hemoglobin in differentiated cells. Elevation of HbF was associated with significant activation of fetal γ-globin and repression of adult β-globin transcription. Changes in gene expression were associated with widespread loss of H3K9me2 in the locus and gain of LDB1 complex occupancy at the γ-globin promoters as well as de novo formation of LCR/γ-globin contacts. Our findings demonstrate that G9a establishes epigenetic conditions preventing activation of γ-globin genes during differentiation of adult erythroid progenitor cells. In this view, manipulation of G9a represents a promising epigenetic approach for treatment of β-hemoglobinopathies.


PLOS ONE | 2015

Erythroid-Specific Expression of LIN28A Is Sufficient for Robust Gamma-Globin Gene and Protein Expression in Adult Erythroblasts.

Y. Terry Lee; Jaira F. de Vasconcellos; Colleen Byrnes; Megha Kaushal; Antoinette Rabel; Laxminath Tumburu; Joshua M. Allwardt; Jeffery L. Miller

Increasing fetal hemoglobin (HbF) levels in adult humans remains an active area in hematologic research. Here we explored erythroid-specific LIN28A expression for its effect in regulating gamma-globin gene expression and HbF levels in cultured adult erythroblasts. For this purpose, lentiviral transduction vectors were produced with LIN28A expression driven by erythroid-specific gene promoter regions of the human KLF1 or SPTA1 genes. Transgene expression of LIN28A with a linked puromycin resistance marker was restricted to the erythroid lineage as demonstrated by selective survival of erythroid colonies (greater than 95% of all colonies). Erythroblast LIN28A over-expression (LIN28A-OE) did not significantly affect proliferation or inhibit differentiation. Greater than 70% suppression of total let-7 microRNA levels was confirmed in LIN28A-OE cells. Increases in gamma-globin mRNA and protein expression with HbF levels reaching 30–40% were achieved. These data suggest that erythroblast targeting of LIN28A expression is sufficient for increasing fetal hemoglobin expression in adult human erythroblasts.


PLOS ONE | 2014

LIN28A Expression Reduces Sickling of Cultured Human Erythrocytes

Jaira F. de Vasconcellos; Ross M. Fasano; Y. Terry Lee; Megha Kaushal; Colleen Byrnes; Emily Riehm Meier; Molly Anderson; Antoinette Rabel; Raul C. Braylan; David F. Stroncek; Jeffery L. Miller

Induction of fetal hemoglobin (HbF) has therapeutic importance for patients with sickle cell disease (SCD) and the beta-thalassemias. It was recently reported that increased expression of LIN28 proteins or decreased expression of its target let-7 miRNAs enhances HbF levels in cultured primary human erythroblasts from adult healthy donors. Here LIN28A effects were studied further using erythrocytes cultured from peripheral blood progenitor cells of pediatric subjects with SCD. Transgenic expression of LIN28A was accomplished by lentiviral transduction in CD34(+) sickle cells cultivated ex vivo in serum-free medium. LIN28A over-expression (LIN28A-OE) increased HbF, reduced beta (sickle)-globin, and strongly suppressed all members of the let-7 family of miRNAs. LIN28A-OE did not affect erythroblast differentiation or prevent enucleation, but it significantly reduced or ameliorated the sickling morphologies of the enucleated erythrocytes.


PLOS ONE | 2013

A synthetic model of human beta-thalassemia erythropoiesis using CD34+ cells from healthy adult donors

Y. Terry Lee; Ki Soon Kim; Colleen Byrnes; Jaira F. de Vasconcellos; Seung-Jae Noh; Antoinette Rabel; Emily Riehm Meier; Jeffery L. Miller

Based upon the lack of clinical samples available for research in many laboratories worldwide, a significant gap exists between basic and clinical studies of beta-thalassemia major. To bridge this gap, we developed an artificially engineered model for human beta thalassemia by knocking down beta-globin gene and protein expression in cultured CD34+ cells obtained from healthy adults. Lentiviral-mediated transduction of beta-globin shRNA (beta-KD) caused imbalanced globin chain production. Beta-globin mRNA was reduced by 90% compared to controls, while alpha-globin mRNA levels were maintained. HPLC analyses revealed a 96% reduction in HbA with only a minor increase in HbF. During the terminal phases of differentiation (culture days 14–21), beta-KD cells demonstrated increased levels of insoluble alpha-globin, as well as activated caspase-3. The majority of the beta-KD cells underwent apoptosis around the polychromatophilic stage of maturation. GDF15, a marker of ineffective erythropoiesis in humans with thalassemia, was significantly increased in the culture supernatants from the beta-KD cells. Knockdown of beta-globin expression in cultured primary human erythroblasts provides a robust ex vivo model for beta-thalassemia.


Proceedings of the National Academy of Sciences of the United States of America | 2017

IGF2BP1 overexpression causes fetal-like hemoglobin expression patterns in cultured human adult erythroblasts

Jaira F. de Vasconcellos; Laxminath Tumburu; Colleen Byrnes; Y. Terry Lee; Pauline C. Xu; May Li; Antoinette Rabel; Benjamin A. Clarke; Nicholas R. Guydosh; Richard L. Proia; Jeffery L. Miller

Significance Fetal hemoglobin (HbF) expression is a tissue- and stage-specific marker of ontogeny in large mammals, which also has therapeutic importance for beta hemoglobinopathies. The heterochronic let-7 miRNAs, which regulate the time and sequence of stage-specific developmental events, have also been shown to regulate HbF in adult human erythroblasts. Here we provide a focused investigation of a let-7 target named “insulin-like growth-factor 2 mRNA-binding protein 1” (IGF2BP1), for its potential role in reactivating HbF in adult cells. IGF2BP1 overexpression caused robust increases of HbF and a reversal from the adult toward a fetal-like globin phenotype. IGF2BP1 effects are partially mediated by posttranscriptional regulation of the known HbF regulator BCL11A. These results suggest a novel mechanism for the regulation of BCL11A and HbF in humans. Here we investigated in primary human erythroid tissues a downstream element of the heterochronic let-7 miRNA pathway, the insulin-like growth factor 2 mRNA-binding protein 1 (IGF2BP1), for its potential to affect the hemoglobin profiles in human erythroblasts. Comparison of adult bone marrow to fetal liver lysates demonstrated developmental silencing in IGF2BP1. Erythroid-specific overexpression of IGF2BP1 caused a nearly complete and pancellular reversal of the adult pattern of hemoglobin expression toward a more fetal-like phenotype. The reprogramming of hemoglobin expression was achieved at the transcriptional level by increased gamma-globin combined with decreased beta-globin transcripts resulting in gamma-globin rising to 90% of total beta-like mRNA. Delta-globin mRNA was reduced to barely detectable levels. Alpha-globin levels were not significantly changed. Fetal hemoglobin achieved levels of 68.6 ± 3.9% in the IGF2BP1 overexpression samples compared with 5.0 ± 1.8% in donor matched transduction controls. In part, these changes were mediated by reduced protein expression of the transcription factor BCL11A. mRNA stability and polysome studies suggest IGF2BP1 mediates posttranscriptional loss of BCL11A. These results suggest a mechanism for chronoregulation of fetal and adult hemoglobin expression in humans.


PLOS ONE | 2016

HMGA2 Moderately Increases Fetal Hemoglobin Expression in Human Adult Erythroblasts

Jaira F. de Vasconcellos; Y. Terry Lee; Colleen Byrnes; Laxminath Tumburu; Antoinette Rabel; Jeffery L. Miller

Induction of fetal hemoglobin (HbF) has therapeutic importance for patients with beta-hemoglobin disorders. Previous studies showed that let-7 microRNAs (miRNAs) are highly regulated in erythroid cells during the fetal-to-adult developmental transition, and that targeting let-7 mediated the up-regulation of HbF to greater than 30% of the total globin levels in human adult cultured erythroblasts. HMGA2 is a member of the high-mobility group A family of proteins and a validated target of the let-7 family of miRNAs. Here we investigate whether expression of HMGA2 directly regulates fetal hemoglobin in adult erythroblasts. Let-7 resistant HMGA2 expression was studied after lentiviral transduction of CD34(+) cells. The transgene was regulated by the erythroid-specific gene promoter region of the human SPTA1 gene (HMGA2-OE). HMGA2-OE caused significant increases in gamma-globin mRNA expression and HbF to around 16% of the total hemoglobin levels compared to matched control transductions. Interestingly, no significant changes in KLF1, SOX6, GATA1, ZBTB7A and BCL11A mRNA levels were observed. Overall, our data suggest that expression of HMGA2, a downstream target of let-7 miRNAs, causes moderately increased gamma-globin gene and protein expression in adult human erythroblasts.


Journal of Translational Medicine | 2017

Tough decoy targeting of predominant let - 7 miRNA species in adult human hematopoietic cells

Jaira F. de Vasconcellos; Colleen Byrnes; Y. Terry Lee; Joshua M. Allwardt; Megha Kaushal; Antoinette Rabel; Jeffery L. Miller


Blood | 2015

IGF2BP1 Reverses Hemoglobin Switching in Adult Erythroblasts

Laxminath Tumburu; Colleen Byrnes; Y. Terry Lee; Jaira F. de Vasconcellos; Antoinette Rabel; Jeffery L. Miller


Blood | 2016

IGF2BP1 Protein Binding of Erythroblast Transcription Factor mRNA: A Cytoplasm-Localized Mechanism for Globin Gene Regulation

Jaira F. de Vasconcellos; Y. Terry Lee; Colleen Byrnes; Benjamin A. Clarke; Pauline C. Xu; Antoinette Rabel; Richard L. Proia; Jeffery L. Miller

Collaboration


Dive into the Jaira F. de Vasconcellos's collaboration.

Top Co-Authors

Avatar

Colleen Byrnes

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Jeffery L. Miller

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Y. Terry Lee

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Antoinette Rabel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Megha Kaushal

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Laxminath Tumburu

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Emily Riehm Meier

George Washington University

View shared research outputs
Top Co-Authors

Avatar

Joshua M. Allwardt

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Ki Soon Kim

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Seung-Jae Noh

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge