Network


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

Hotspot


Dive into the research topics where Kamaleldin E. Elagib is active.

Publication


Featured researches published by Kamaleldin E. Elagib.


Nature Chemical Biology | 2009

Discovering chemical modifiers of oncogene-regulated hematopoietic differentiation

Jing-Ruey J. Yeh; Kathleen M. Munson; Kamaleldin E. Elagib; Adam N. Goldfarb; David A. Sweetser; Randall T. Peterson

It has been proposed that inhibitors of an oncogenes effects on multipotent hematopoietic progenitor cell differentiation may change the properties of the leukemic stem cells and complement the clinical use of cytotoxic drugs. Using zebrafish, we developed a robust in vivo hematopoietic differentiation assay that reflects the activity of the oncogene AML1-ETO. Screening for modifiers of AML1-ETO-mediated hematopoietic dysregulation uncovered unexpected roles of COX-2 and β-catenin-dependent pathways in AML1-ETO function. This approach may open doors for developing therapeutics targeting oncogene function within leukemic stem cells.


Molecular and Cellular Biology | 2004

Jun Blockade of Erythropoiesis: Role for Repression of GATA-1 by HERP2

Kamaleldin E. Elagib; Mang Xiao; Isa M. Hussaini; Lorrie L. Delehanty; Lisa A. Palmer; Frederick Racke; Michael J. Birrer; Ganapath Shanmugasundaram; Michael A. McDevitt; Adam N. Goldfarb

ABSTRACT Although Jun upregulation and activation have been established as critical to oncogenesis, the relevant downstream pathways remain incompletely characterized. In this study, we found that c-Jun blocks erythroid differentiation in primary human hematopoietic progenitors and, correspondingly, that Jun factors block transcriptional activation by GATA-1, the central regulator of erythroid differentiation. Mutagenesis of c-Jun suggested that its repression of GATA-1 occurs through a transcriptional mechanism involving activation of downstream genes. We identified the hairy-enhancer-of-split-related factor HERP2 as a novel gene upregulated by c-Jun. HERP2 showed physical interaction with GATA-1 and repressed GATA-1 transcriptional activation. Furthermore, transduction of HERP2 into primary human hematopoietic progenitors inhibited erythroid differentiation. These results thus define a novel regulatory pathway linking the transcription factors c-Jun, HERP2, and GATA-1. Furthermore, these results establish a connection between the Notch signaling pathway, of which the HERP factors are a critical component, and the GATA family, which participates in programming of cellular differentiation.


Cancer Research | 2006

Erythroid Inhibition by the Leukemic Fusion AML1-ETO Is Associated with Impaired Acetylation of the Major Erythroid Transcription Factor GATA-1

Young Jin Choi; Kamaleldin E. Elagib; Lorrie L. Delehanty; Adam N. Goldfarb

Human acute myeloid leukemias with the t(8;21) translocation express the AML1-ETO fusion protein in the hematopoietic stem cell compartment and show impairment in erythroid differentiation. This clinical finding is reproduced in multiple murine and cell culture model systems in which AML1-ETO specifically interferes with erythroid maturation. Using purified normal human early hematopoietic progenitor cells, we find that AML1-ETO impedes the earliest discernable steps of erythroid lineage commitment. Correspondingly, GATA-1, a central transcriptional regulator of erythroid differentiation, undergoes repression by AML1-ETO in a nonconventional histone deacetylase-independent manner. In particular, GATA-1 acetylation by its transcriptional coactivator, p300/CBP, a critical regulatory step in programming erythroid development, is efficiently blocked by AML1-ETO. Fusion of a heterologous E1A coactivator recruitment module to GATA-1 overrides the inhibitory effects of AML1-ETO on GATA-1 acetylation and transactivation. Furthermore, the E1A-GATA-1 fusion, but not wild-type GATA-1, rescues erythroid lineage commitment in primary human progenitors expressing AML1-ETO. These results ascribe a novel repressive mechanism to AML1-ETO, blockade of GATA-1 acetylation, which correlates with its inhibitory effects on primary erythroid lineage commitment.


Blood | 2008

Cross-talk of GATA-1 and P-TEFb in megakaryocyte differentiation.

Kamaleldin E. Elagib; Ivailo S. Mihaylov; Lorrie L. Delehanty; Grant C. Bullock; Kevin D. Ouma; Jill F. Caronia; Sara L. Gonias; Adam N. Goldfarb

The transcription factor GATA-1 participates in programming the differentiation of multiple hematopoietic lineages. In megakaryopoiesis, loss of GATA-1 function produces complex developmental abnormalities and underlies the pathogenesis of megakaryocytic leukemia in Down syndrome. Its distinct functions in megakaryocyte and erythroid maturation remain incompletely understood. In this study, we identified functional and physical interaction of GATA-1 with components of the positive transcriptional elongation factor P-TEFb, a complex containing cyclin T1 and the cyclin-dependent kinase 9 (Cdk9). Megakaryocytic induction was associated with dynamic changes in endogenous P-TEFb composition, including recruitment of GATA-1 and dissociation of HEXIM1, a Cdk9 inhibitor. shRNA knockdowns and pharmacologic inhibition both confirmed contribution of Cdk9 activity to megakaryocytic differentiation. In mice with megakaryocytic GATA-1 deficiency, Cdk9 inhibition produced a fulminant but reversible megakaryoblastic disorder reminiscent of the transient myeloproliferative disorder of Down syndrome. P-TEFb has previously been implicated in promoting elongation of paused RNA polymerase II and in programming hypertrophic differentiation of cardiomyocytes. Our results offer evidence for P-TEFb cross-talk with GATA-1 in megakaryocytic differentiation, a program with parallels to cardiomyocyte hypertrophy.


Journal of Clinical Investigation | 2017

Neonatal expression of RNA-binding protein IGF2BP3 regulates the human fetal-adult megakaryocyte transition

Kamaleldin E. Elagib; Chih Huan Lu; Goar Mosoyan; Shadi Khalil; Ewelina Zasadzińska; Daniel R. Foltz; Peter Balogh; Alejandro A. Gru; Deborah A. Fuchs; Lisa M. Rimsza; Els Verhoeyen; Miriam Sansó; Robert P. Fisher; Camelia Iancu-Rubin; Adam N. Goldfarb

Hematopoietic transitions that accompany fetal development, such as erythroid globin chain switching, play important roles in normal physiology and disease development. In the megakaryocyte lineage, human fetal progenitors do not execute the adult morphogenesis program of enlargement, polyploidization, and proplatelet formation. Although these defects decline with gestational stage, they remain sufficiently severe at birth to predispose newborns to thrombocytopenia. These defects may also contribute to inferior platelet recovery after cord blood stem cell transplantation and may underlie inefficient platelet production by megakaryocytes derived from pluripotent stem cells. In this study, comparison of neonatal versus adult human progenitors has identified a blockade in the specialized positive transcription elongation factor b (P-TEFb) activation mechanism that is known to drive adult megakaryocyte morphogenesis. This blockade resulted from neonatal-specific expression of an oncofetal RNA-binding protein, IGF2BP3, which prevented the destabilization of the nuclear RNA 7SK, a process normally associated with adult megakaryocytic P-TEFb activation. Knockdown of IGF2BP3 sufficed to confer both phenotypic and molecular features of adult-type cells on neonatal megakaryocytes. Pharmacologic inhibition of IGF2BP3 expression via bromodomain and extraterminal domain (BET) inhibition also elicited adult features in neonatal megakaryocytes. These results identify IGF2BP3 as a human ontogenic master switch that restricts megakaryocyte development by modulating a lineage-specific P-TEFb activation mechanism, revealing potential strategies toward enhancing platelet production.


Journal of Biological Chemistry | 2012

Cyclic AMP Signaling Inhibits Megakaryocytic Differentiation by Targeting Transcription Factor 3 (E2A) Cyclin-dependent Kinase Inhibitor 1A (CDKN1A) Transcriptional Axis

Jeremy D. Rubinstein; Kamaleldin E. Elagib; Adam N. Goldfarb

Background: Cyclic AMP signaling impairs megakaryopoiesis through an unknown mechanism. Results: cAMP transcriptionally represses the transcription factor E2A with resultant impairment in the expression of its target CDKN1A. Conclusion: In discovering a mechanism for cAMP-induced inhibition, E2A is identified clearly as critical megakaryocytic factor. Significance: Elucidating the repertoire of necessary transcription factors is crucial for understanding control of megakaryocytic lineage. Signaling via the intracellular second messenger cyclic AMP (cAMP) has long been implicated in the repression of megakaryocytic differentiation. However, the mechanisms by which cAMP signaling impairs megakaryopoiesis have never been elucidated. In a human CD34+ cell culture model, we show that the adenylyl cyclase agonist forskolin inhibits megakaryocytic differentiation in a protein kinase A-dependent manner. Using this system to screen for downstream effectors, we identified the transcription factor E2A as a key target in a novel repressive signaling pathway. Specifically, forskolin acting through protein kinase A-induced E2A down-regulation and enforced expression of E2A overrode the inhibitory effects of forskolin on megakaryopoiesis. The dependence of megakaryopoiesis on critical thresholds of E2A expression was confirmed in vivo in haploinsufficient mice and ex vivo using shRNA knockdown in human progenitors. Using a variety of approaches, we further identified p21 (encoded by CDKN1A) as a functionally important megakaryopoietic regulator residing downstream of E2A. These results thus implicate the E2A-CDKN1A transcriptional axis in the control of megakaryopoiesis and reveal the lineage-selective inhibition of this axis as a likely mechanistic basis for the inhibitory effects of cAMP signaling.


Experimental Hematology | 2018

Megakaryocyte ontogeny: Clinical and molecular significance

Kamaleldin E. Elagib; Ashton T. Brock; Adam N. Goldfarb

Fetal megakaryocytes (Mks) differ from adult Mks in key parameters that affect their capacity for platelet production. However, despite being smaller, more proliferative, and less polyploid, fetal Mks generally mature in the same manner as adult Mks. The phenotypic features unique to fetal Mks predispose patients to several disease conditions, including infantile thrombocytopenia, infantile megakaryoblastic leukemias, and poor platelet recovery after umbilical cord blood stem cell transplantations. Ontogenic Mk differences also affect new strategies being developed to address global shortages of platelet transfusion units. These donor-independent, ex vivo production platforms are hampered by the limited proliferative capacity of adult-type Mks and the inferior platelet production by fetal-type Mks. Understanding the molecular programs that distinguish fetal versus adult megakaryopoiesis will help in improving approaches to these clinical problems. This review summarizes the phenotypic differences between fetal and adult Mks, the disease states associated with fetal megakaryopoiesis, and recent advances in the understanding of mechanisms that determine ontogenic Mk transitions.


Molecular and Cellular Biology | 2015

Erratum for Elagib et al., Jun Blockade of Erythropoiesis: Role for Repression of GATA-1 by HERP2

Kamaleldin E. Elagib; Mang Xiao; Isa M. Hussaini; Lorrie L. Delehanty; Lisa A. Palmer; Frederick Racke; Michael J. Birrer; Shanmugasundaram Ganapathy-Kanniappan; Michael A. McDevitt; Adam N. Goldfarb

Kamaleldin E. Elagib, Mang Xiao, Isa M. Hussaini, Lorrie L. Delehanty, Lisa A. Palmer, Frederick K. Racke, Michael J. Birrer, Shanmugasundaram Ganapathy-Kanniappan, Michael A. McDevitt, and Adam N. Goldfarb Department of Pathology and Department of Pediatrics, University of Virginia School of Medicine, Charlottesville, Virginia, USA; Department of Pathology and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, and Molecular Mechanism Section, National Cancer Institute, Rockville, Maryland, USA


Blood | 2003

RUNX1 and GATA-1 coexpression and cooperation in megakaryocytic differentiation

Kamaleldin E. Elagib; Frederick K. Racke; Michael Mogass; Rina Khetawat; Lorrie L. Delehanty; Adam N. Goldfarb


Cancer Letters | 2007

Oncogenic pathways of AML1-ETO in acute myeloid leukemia: multifaceted manipulation of marrow maturation

Kamaleldin E. Elagib; Adam N. Goldfarb

Collaboration


Dive into the Kamaleldin E. Elagib's collaboration.

Top Co-Authors

Avatar

Adam N. Goldfarb

Case Western Reserve University

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
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge