Network


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

Hotspot


Dive into the research topics where Nabeel R. Yaseen is active.

Publication


Featured researches published by Nabeel R. Yaseen.


Cancer Research | 2006

NUP98-HOXA9 Induces Long-term Proliferation and Blocks Differentiation of Primary Human CD34+ Hematopoietic Cells

Akiko Takeda; Charles L. Goolsby; Nabeel R. Yaseen

NUP98-HOXA9, the chimeric protein resulting from the t(7;11)(p15;p15) chromosomal translocation, is a prototype of several NUP98 fusions that occur in myelodysplastic syndromes and acute myeloid leukemia. We examined its effect on differentiation, proliferation, and gene expression in primary human CD34+ hematopoietic cells. Colony-forming cell (CFC) assays in semisolid medium combined with morphologic examination and flow cytometric immunophenotyping revealed that NUP98-HOXA9 increased the numbers of erythroid precursors and impaired both myeloid and erythroid differentiation. In continuous liquid culture, cells transduced with NUP98-HOXA9 exhibited a biphasic growth curve with initial growth inhibition followed by enhanced long-term proliferation, suggesting an increase in the numbers of primitive self-renewing cells. This was confirmed by a dramatic increase in the numbers of long-term culture-initiating cells, the most primitive hematopoietic cells detectable in vitro. To understand the molecular mechanisms underlying the effects of NUP98-HOXA9 on hematopoietic cell proliferation and differentiation, oligonucleotide microarray analysis was done at several time points over 16 days, starting at 6 hours posttransduction. The early growth suppression was preceded by up-regulation of IFNbeta1 and accompanied by marked up-regulation of IFN-induced genes, peaking at 3 days posttransduction. In contrast, oncogenes such as homeobox transcription factors, FLT3, KIT, and WT1 peaked at 8 days or beyond, coinciding with increased proliferation. In addition, several putative tumor suppressors and genes associated with hematopoietic differentiation were repressed at later time points. These findings provide a comprehensive picture of the changes in proliferation, differentiation, and global gene expression that underlie the leukemic transformation of human hematopoietic cells by NUP98-HOXA9.


Journal of Biological Chemistry | 2000

The Nucleoporin Nup98 Is a Site for GDP/GTP Exchange on Ran and Termination of Karyopherin β2-mediated Nuclear Import

Beatriz M. A. Fontoura; Günter Blobel; Nabeel R. Yaseen

Karyopherin β2 (Kapβ2, transportin) binds the M9 sequence of human ribonucleoprotein A1 and mediates its nuclear import. Here we show a role for the nucleoporin Nup98 in the disassembly of Kapβ2 import complexes at the nuclear side of the nuclear pore complex (NPC). Kapβ2 bound to a region at the N terminus of Nup98 that contains an M9-like sequence. The human ribonucleoprotein A1 M9 sequence competed with Nup98 for binding to Kapβ2, indicating that Nup98 can dissociate Kapβ2 from its substrate. Binding of Kapβ2 to Nup98 was inhibited by Ran loaded with guanylyl imidophosphate, suggesting that RanGTP dissociates Kapβ2 from Nup98. RanGTP is produced from RanGDP through nucleotide exchange mediated by RanGEF (RCC1). Immunoelectron microscopy and nucleotide exchange assays revealed functional RanGEF on both sides of the NPC. On the nuclear side, the localization of RanGEF coincided with that of Nup98. RanGEF bound to Nup98 at a region adjacent to the Kapβ2-binding site. These findings suggest a model where 1) import substrate is released from Kapβ2 at the nucleoplasmic side of the NPC by competition with the Nup98 M9-like site, 2) Nup98-bound RanGEF catalyzes the formation of RanGTP, and 3) RanGTP dissociates Kapβ2 from Nup98 allowing repeated cycles of import.


Journal of Biological Chemistry | 1999

GTP Hydrolysis Links Initiation and Termination of Nuclear Import on the Nucleoporin Nup358

Nabeel R. Yaseen; Günter Blobel

Binding of GTP-bound Ran (RanGTP) to karyopherin β1 (Kapβ1) releases import cargo into the nucleus. Using an ultrastructural, biochemical, and functional approach, we have studied the mechanism by which Kapβ1·RanGTP is recycled at the nuclear pore complex for repeated rounds of import. In vitro, Kapβ1 bound to the RanBP1-homologous (RBH) domains of Nup358 in the presence of either RanGTP or RanGDP, forming trimeric complexes. The Kapβ1·RanGTP·RBH complex resisted dissociation by RanBP1 and GTP hydrolysis by Ran GTPase activating protein 1. Ran-dependent binding of gold-conjugated Kapβ1 to the cytoplasmic fibers of the nuclear pore complex in digitonin-permeabilized cells and RanBP1 competition confirmed thein vitro binding data. Interaction of karyopherin α and a classical nuclear localization sequence peptide with the Kapβ1·RanGTP·RBH complex stimulated GTP hydrolysis by Ran GTPase activating protein 1 both in vitro and in permeabilized cells. This GTP hydrolysis was required for reinitiation of import of a nuclear localization sequence-bearing substrate in permeabilized cells. These data suggest that GTP hydrolysis on the RBH domains of Nup358 couples the termination of one cycle of nuclear import with the initiation of the next.


Journal of Biological Chemistry | 2010

Inhibition of CRM1-mediated Nuclear Export of Transcription Factors by Leukemogenic NUP98 Fusion Proteins

Akiko Takeda; Nayan J. Sarma; Anmaar M. Abdul-Nabi; Nabeel R. Yaseen

NUP98 is a nucleoporin that plays complex roles in the nucleocytoplasmic trafficking of macromolecules. Rearrangements of the NUP98 gene in human leukemia result in the expression of numerous fusion oncoproteins whose effect on nucleocytoplasmic trafficking is poorly understood. The present study was undertaken to determine the effects of leukemogenic NUP98 fusion proteins on CRM1-mediated nuclear export. NUP98-HOXA9, a prototypic NUP98 fusion, inhibited the nuclear export of two known CRM1 substrates: mutated cytoplasmic nucleophosmin and HIV-1 Rev. In vitro binding assays revealed that NUP98-HOXA9 binds CRM1 through the FG repeat motif in a Ran-GTP-dependent manner similar to but stronger than the interaction between CRM1 and its export substrates. Two NUP98 fusions, NUP98-HOXA9 and NUP98-DDX10, whose fusion partners are structurally and functionally unrelated, interacted with endogenous CRM1 in myeloid cells as shown by co-immunoprecipitation. These leukemogenic NUP98 fusion proteins interacted with CRM1, Ran, and the nucleoporin NUP214 in a manner fundamentally different from that of wild-type NUP98. NUP98-HOXA9 and NUP98-DDX10 formed characteristic aggregates within the nuclei of a myeloid cell line and primary human CD34+ cells and caused aberrant localization of CRM1 to these aggregates. These NUP98 fusions caused nuclear accumulation of two transcription factors, NFAT and NFκB, that are regulated by CRM1-mediated export. The nuclear entrapment of NFAT and NFκB correlated with enhanced transcription from promoters responsive to these transcription factors. Taken together, the results suggest a new mechanism by which NUP98 fusions dysregulate transcription and cause leukemia, namely, inhibition of CRM1-mediated nuclear export with aberrant nuclear retention of transcriptional regulators.


Journal of Biological Chemistry | 2005

Carrier-independent Nuclear Import of the Transcription Factor PU.1 via RanGTP-stimulated Binding to Nup153

Hualin Zhong; Akiko Takeda; Reza Nazari; Helen Shio; Günter Blobel; Nabeel R. Yaseen

PU.1 is a transcription factor of the Ets family with important functions in hematopoietic cell differentiation. Using green fluorescent protein-PU.1 fusions, we show that the Ets DNA binding domain of PU.1 is necessary and sufficient for its nuclear localization. Fluorescence and ultrastructural nuclear import assays showed that PU.1 nuclear import requires energy but not soluble carriers. PU.1 interacted directly with two nucleoporins, Nup62 and Nup153. The binding of PU.1 to Nup153, but not to Nup62, increased dramatically in the presence of RanGMPPNP, indicating the formation of a PU.1·RanGTP·Nup153 complex. The Ets domain accounted for the bulk of the interaction of PU.1 with Nup153 and RanGMPPNP. Because Nup62 is located close to the midplane of the nuclear pore complex whereas Nup153 is at its nuclear side, these findings suggest a model whereby RanGTP propels PU.1 toward the nuclear side of the nuclear pore complex by increasing its affinity for Nup153. This notion was confirmed by ultrastructural studies using gold-labeled PU.1 in permeabilized cells.


Molecular and Cellular Biology | 1994

A CENTRAL ROLE FOR FOS IN HUMAN B- AND T-CELL NFAT (NUCLEAR FACTOR OF ACTIVATED T CELLS) : AN ACIDIC REGION IS REQUIRED FOR IN VITRO ASSEMBLY

Nabeel R. Yaseen; Jungchan Park; Tom Kerppola; Tom Curran; Surendra Sharma

Nuclear factor of activated T cells (NFAT) is a multicomponent transcription factor that contains Fos and Jun family proteins in addition to a constitutively expressed factor(s). It is important for the production of interleukin 2 (IL-2) by T cells and is also expressed in B cells. Here we show that NFAT complexes in B- and T-cell nuclear extracts can be supershifted prominently with Fos antibodies and to a variable extent with Jun family protein antibodies. Fos and Jun proteins appear to participate in NFAT complexes as heterodimers, since efficient in vitro reconstitution of NFAT in unstimulated B- or T-cell nuclear extracts required both Fos and Jun. Using Fos and Jun deletion derivatives, we found that an acidic Fos region (amino acids 118 to 138), outside the DNA binding and dimerization domains, was necessary for the in vitro reconstitution of the NFAT complex in both B- and T-lymphocyte extracts although it was not required for binding to an AP-1 site. Fos-Jun heterodimers exhibited low-affinity direct binding to the NFAT site in the absence of nuclear extracts. This binding also required the Fos acidic region, amino acids 118 to 138. Mutating a variant AP-1 site in the NFAT oligonucleotide abolished both direct binding of Fos-Jun heterodimers and in vitro reconstitution of NFAT. These results demonstrate a central role of Fos in NFAT complex formation in both B and T lymphocytes and show that NFAT assembly involves direct binding of Fos-Jun heterodimers to a variant AP-1 site within the human NFAT recognition site.


Leukemia | 2010

Effects of the NUP98-DDX10 oncogene on primary human CD34+ cells: role of a conserved helicase motif.

Enas R. Yassin; Anmaar M. Abdul-Nabi; Akiko Takeda; Nabeel R. Yaseen

NUP98 gene rearrangements occur in acute myeloid leukemia and result in the expression of fusion proteins. One of the most frequent is NUP98–DDX10 that fuses a portion of NUP98 to a portion of DDX10, a putative DEAD-box RNA helicase. Here, we show that NUP98–DDX10 dramatically increases proliferation and self-renewal of primary human CD34+ cells, and disrupts their erythroid and myeloid differentiation. It localizes to their nuclei and extensively deregulates gene expression. Comparison to another leukemogenic NUP98 fusion, NUP98–HOXA9, reveals a number of genes deregulated by both oncoproteins, including HOX genes, COX-2, MYCN, ANGPT1, REN, HEY1, SOX4 and others. These genes may account for the similar leukemogenic properties of NUP98 fusion oncogenes. The YIHRAGRTAR sequence in the DDX10 portion of NUP98–DDX10 represents a major motif shared by DEAD-box RNA helicases that is required for ATP binding, RNA-binding and helicase functions. Mutating this motif diminished the in vitro transforming ability of NUP98–DDX10, indicating that it has a role in leukemogenesis. These data show for the first time the in vitro transforming ability of NUP98–DDX10 and show that it is partially dependent on one of the consensus helicase motifs of DDX10. They also point to common pathways that may underlie leukemogenesis by different NUP98 fusions.


PLOS ONE | 2009

Dissection of the transformation of primary human hematopoietic cells by the oncogene NUP98-HOXA9

Enas R. Yassin; Nayan J. Sarma; Anmaar M. Abdul-Nabi; James Dombrowski; Ye Han; Akiko Takeda; Nabeel R. Yaseen

NUP98-HOXA9 is the prototype of a group of oncoproteins associated with acute myeloid leukemia. It consists of an N-terminal portion of NUP98 fused to the homeodomain of HOXA9 and is believed to act as an aberrant transcription factor that binds DNA through the homeodomain. Here we show that NUP98-HOXA9 can regulate transcription without binding to DNA. In order to determine the relative contributions of the NUP98 and HOXA9 portions to the transforming ability of NUP98-HOXA9, the effects of NUP98-HOXA9 on primary human CD34+ cells were dissected and compared to those of wild-type HOXA9. In contrast to previous findings in mouse cells, HOXA9 had only mild effects on the differentiation and proliferation of primary human hematopoietic cells. The ability of NUP98-HOXA9 to disrupt the differentiation of primary human CD34+ cells was found to depend primarily on the NUP98 portion, whereas induction of long-term proliferation required both the NUP98 moiety and an intact homeodomain. Using oligonucleotide microarrays in primary human CD34+ cells, a group of genes was identified whose dysregulation by NUP98-HOXA9 is attributable primarily to the NUP98 portion. These include RAP1A, HEY1, and PTGS2 (COX-2). Their functions may reflect the contribution of the NUP98 moiety of NUP98-HOXA9 to leukemic transformation. Taken together, these results suggest that the effects of NUP98-HOXA9 on gene transcription and cell transformation are mediated by at least two distinct mechanisms: one that involves promoter binding through the homeodomain with direct transcriptional activation, and another that depends predominantly on the NUP98 moiety and does not involve direct DNA binding.


Seminars in Cancer Biology | 2014

Nucleoporins and nucleocytoplasmic transport in hematologic malignancies

Akiko Takeda; Nabeel R. Yaseen

Hematologic malignancies are often associated with chromosomal rearrangements that lead to the expression of chimeric fusion proteins. Rearrangements of the genes encoding two nucleoporins, NUP98 and NUP214, have been implicated in the pathogenesis of several types of hematologic malignancies, particularly acute myeloid leukemia. NUP98 rearrangements result in fusion of an N-terminal portion of NUP98 to one of numerous proteins. These rearrangements often follow treatment with topoisomerase II inhibitors and tend to occur in younger patients. They have been shown to induce leukemia in mice and to enhance proliferation and disrupt differentiation in primary human hematopoietic precursors. NUP214 has only a few fusion partners. DEK-NUP214 is the most common NUP214 fusion in AML; it tends to occur in younger patients and is usually associated with FLT3 internal tandem duplications. The leukemogenic activity of NUP214 fusions is less well characterized. Normal nucleoporins, including NUP98 and NUP214, have important functions in nucleocytoplasmic transport, transcription, and mitosis. These functions and their disruptions by oncogenic nucleoporin fusions are discussed.


PLOS ONE | 2010

In vitro transformation of primary human CD34+ cells by AML fusion oncogenes: Early gene expression profiling reveals possible drug target in AML

Anmaar M. Abdul-Nabi; Enas R. Yassin; Nobish Varghese; Hrishikesh Deshmukh; Nabeel R. Yaseen

Different fusion oncogenes in acute myeloid leukemia (AML) have distinct clinical and laboratory features suggesting different modes of malignant transformation. Here we compare the in vitro effects of representatives of 4 major groups of AML fusion oncogenes on primary human CD34+ cells. As expected from their clinical similarities, MLL-AF9 and NUP98-HOXA9 had very similar effects in vitro. They both caused erythroid hyperplasia and a clear block in erythroid and myeloid maturation. On the other hand, AML1-ETO and PML-RARA had only modest effects on myeloid and erythroid differentiation. All oncogenes except PML-RARA caused a dramatic increase in long-term proliferation and self-renewal. Gene expression profiling revealed two distinct temporal patterns of gene deregulation. Gene deregulation by MLL-AF9 and NUP98-HOXA9 peaked 3 days after transduction. In contrast, the vast majority of gene deregulation by AML1-ETO and PML-RARA occurred within 6 hours, followed by a dramatic drop in the numbers of deregulated genes. Interestingly, the p53 inhibitor MDM2 was upregulated by AML1-ETO at 6 hours. Nutlin-3, an inhibitor of the interaction between MDM2 and p53, specifically inhibited the proliferation and self-renewal of primary human CD34+ cells transduced with AML1-ETO, suggesting that MDM2 upregulation plays a role in cell transformation by AML1-ETO. These data show that differences among AML fusion oncogenes can be recapitulated in vitro using primary human CD34+ cells and that early gene expression profiling in these cells can reveal potential drug targets in AML.

Collaboration


Dive into the Nabeel R. Yaseen's collaboration.

Top Co-Authors

Avatar

Akiko Takeda

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Anmaar M. Abdul-Nabi

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Günter Blobel

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Enas R. Yassin

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar

Nayan J. Sarma

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Helen Shio

Rockefeller University

View shared research outputs
Top Co-Authors

Avatar

Hualin Zhong

City University of New York

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge