Network


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

Hotspot


Dive into the research topics where Noel Lenny is active.

Publication


Featured researches published by Noel Lenny.


Molecular and Cellular Biology | 1995

The t(8;21) fusion protein interferes with AML-1B-dependent transcriptional activation.

Shari Meyers; Noel Lenny; Scott W. Hiebert

The AML-1/CBF beta transcription factor complex is targeted by both the t(8;21) and the inv(16) chromosomal alterations, which are frequently observed in acute myelogenous leukemia. AML-1 is a site-specific DNA-binding protein that recognizes the enhancer core motif TGTGGT. The t(8;21) translocation fuses the first 177 amino acids of AML-1 to MTG8 (also known as ETO), generating a chimeric protein that retains the DNA-binding domain of AML-1. Analysis of endogenous AML-1 DNA-binding complexes suggested the presence of at least two AML-1 isoforms. Accordingly, we screened a human B-cell cDNA library and isolated a larger, potentially alternatively spliced, form of AML1, termed AML1B. AML-1B is a protein of 53 kDa that binds to a consensus AML-1-binding site and complexes with CBF beta. Subcellular fractionation experiments demonstrated that both AML-1 and AML-1/ETO are efficiently extracted from the nucleus under ionic conditions but that AML-1B is localized to a salt-resistant nuclear compartment. Analysis of the transcriptional activities of AML-1, AML-1B, and AML-1/ETO demonstrated that only AML-1B activates transcription from the T-cell receptor beta enhancer. Mixing experiments indicated that AML-1/ETO can efficiently block AML-1B-dependent transcriptional activation, suggesting that the t(8;21) translocation creates a dominant interfering protein.


Molecular and Cellular Biology | 2001

ETO, a Target of t(8;21) in Acute Leukemia, Makes Distinct Contacts with Multiple Histone Deacetylases and Binds mSin3A through Its Oligomerization Domain

Joseph M. Amann; John Nip; David K. Strom; Bart Lutterbach; Hironori Harada; Noel Lenny; James R. Downing; Shari Meyers; Scott W. Hiebert

ABSTRACT t(8;21) and t(16;21) create two fusion proteins, AML-1–ETO and AML-1–MTG16, respectively, which fuse the AML-1 DNA binding domain to putative transcriptional corepressors, ETO and MTG16. Here, we show that distinct domains of ETO contact the mSin3A and N-CoR corepressors and define two binding sites within ETO for each of these corepressors. In addition, of eight histone deacetylases (HDACs) tested, only the class I HDACs HDAC-1, HDAC-2, and HDAC-3 bind ETO. However, these HDACs bind ETO through different domains. We also show that the murine homologue of MTG16, ETO-2, is also a transcriptional corepressor that works through a similar but distinct mechanism. Like ETO, ETO-2 interacts with N-CoR, but ETO-2 fails to bind mSin3A. Furthermore, ETO-2 binds HDAC-1, HDAC-2, and HDAC-3 but also interacts with HDAC-6 and HDAC-8. In addition, we show that expression of AML-1–ETO causes disruption of the cell cycle in the G1 phase. Disruption of the cell cycle required the ability of AML-1–ETO to repress transcription because a mutant of AML-1–ETO, Δ469, which removes the majority of the corepressor binding sites, had no phenotype. Moreover, treatment of AML-1–ETO-expressing cells with trichostatin A, an HDAC inhibitor, restored cell cycle control. Thus, AML-1–ETO makes distinct contacts with multiple HDACs and an HDAC inhibitor biologically inactivates this fusion protein.


Molecular and Cellular Biology | 1998

The t(8;21) Fusion Product, AML-1–ETO, Associates with C/EBP-α, Inhibits C/EBP-α-Dependent Transcription, and Blocks Granulocytic Differentiation

Jennifer J. Westendorf; Cindy M. Yamamoto; Noel Lenny; James R. Downing; Michael E. Selsted; Scott W. Hiebert

ABSTRACT AML-1B is a hematopoietic transcription factor that is functionally inactivated by multiple chromosomal translocations in human acute myeloblastic and B-cell lymphocytic leukemias. The t(8;21)(q22;q22) translocation replaces the C terminus, including the transactivation domain of AML-1B, with ETO, a nuclear protein of unknown function. We previously showed that AML-1–ETO is a dominant inhibitor of AML-1B-dependent transcriptional activation. Here we demonstrate that AML-1–ETO also inhibits C/EBP-α-dependent activation of the myeloid cell-specific, rat defensin NP-3 promoter. AML-1B bound the core enhancer motifs present in the NP-3 promoter and activated transcription approximately sixfold. Similarly, C/EBP-α bound NP-3 promoter sequences and activated transcription approximately sixfold. Coexpression of C/EBP-α with AML-1B or its family members, AML-2 and murine AML-3, synergistically activated the NP-3 promoter up to 60-fold. The t(8;21) product, AML-1–ETO, repressed AML-1B-dependent activation of NP-3 and completely inhibited C/EBP-α-dependent activity as well as the synergistic activation. In contrast, theinv(16) product, which indirectly targets AML family members by fusing their heterodimeric DNA binding partner, CBF-β, to the myosin heavy chain, inhibited AML-1B but not C/EBP-α activation or the synergistic activation. AML-1–ETO and C/EBP-α were coimmunoprecipitated and thus physically interact in vivo. Deletion mutants demonstrated that the C terminus of ETO was required for AML-1–ETO-mediated repression of the synergistic activation but not for association with C/EBP-α. Finally, overexpression of AML-1–ETO in myeloid progenitor cells prevented granulocyte colony-stimulating factor-induced differentiation. Thus, AML-1–ETO may contribute to leukemogenesis by specifically inhibiting C/EBP-α- and AML-1B-dependent activation of myeloid promoters and blocking differentiation.


Molecular and Cellular Biology | 1996

The t (12;21) translocation converts AML-1B from an activator to a repressor of transcription

Scott W. Hiebert; W Sun; J N Davis; Todd R. Golub; Sheila A. Shurtleff; A Buijs; James R. Downing; Gerard Grosveld; M F Roussell; D G Gilliland; Noel Lenny; Shari Meyers

The t(12;21) translocation is present in up to 30% of childhood B-cell acute lymphoblastic and fuses a potential dimerization motif from the ets-related factor TEL to the N terminus of AML1. The t(12;21) translocation encodes a 93-kDa fusion protein that localizes to a high-salt- and detergent-resistant nuclear compartment. This protein binds the enhancer core motif, TGTGGT, and interacts with the AML-1-binding protein, core-binding factor beta. Although TEL/AML-1B retains the C-terminal domain of AML-1B that is required for transactivation of the T-cell receptor beta enhancer, it fails to activate transcription but rather inhibits the basal activity of this enhancer. TEL/AML-1B efficiently interferes with AML-1B dependent transactivation of the T-cell receptor beta enhancer, and coexpression of wild-type TEL does not reverse this inhibition. The N-terminal TEL helix-loop-helix domain is essential for TEL/AML-1B-mediated repression. Thus, the t(12;21) fusion protein dominantly interferes with AML-1B-dependent transcription, suggesting that the inhibition of expression of AML-1 genes is critical for B-cell leukemogenesis.


Molecular Biology Reports | 1997

Transcriptional regulation during myelopoiesis.

Noel Lenny; Jennifer J. Westendorf; Scott W. Hiebert

The coordinated production of all blood cells from a common stem cell is a highly regulated process involving successive stages of commitment and differentiation. From analyses of mice deficient in transcription factor genes and from the characterizations of chromosome breakpoints in human leukemias, it has become evident that transcription factors are important regulators of hematopoiesis. During myelopoiesis, which includes the development of granulocytic and monocytic lineages, transcription factors from several families are active, including AML1/CBFβ, C/EBP, Ets, c-Myb, HOX, and MZF-1. Few of these factors are expressed exclusively in myeloid cells; instead it appears that they cooperatively regulate transcription of myeloid-specific genes. Here we discuss recent advances in transcriptional regulation during myelopoiesis.


Molecular and Cellular Biology | 2001

The E2A-HLF oncoprotein activates Groucho-related genes and suppresses Runx1.

Jinjun Dang; Takeshi Inukai; Hidemitsu Kurosawa; Kumiko Goi; Toshiya Inaba; Noel Lenny; James R. Downing; Stefano Stifani; A. Thomas Look

ABSTRACT The E2A-HLF fusion gene, formed by the t(17;19)(q22;p13) chromosomal translocation in leukemic pro-B cells, encodes a chimeric transcription factor consisting of the transactivation domain of E2A linked to the bZIP DNA-binding and protein dimerization domain of hepatic leukemia factor (HLF). This oncoprotein blocks apoptosis induced by growth factor deprivation or irradiation, but the mechanism for this effect remains unclear. We therefore performed representational difference analysis (RDA) to identify downstream genetic targets of E2A-HLF, using a murine FL5.12 pro-B cell line that had been stably transfected with E2A-HLF cDNA under the control of a zinc-regulated metallothionein promoter. Two RDA clones, designated RDA1 and RDA3, were differentially upregulated in E2A-HLF-positive cells after zinc induction. The corresponding cDNAs encoded two WD40 repeat-containing proteins, Grg2 and Grg6. Both are related to the Drosophila protein Groucho, a transcriptional corepressor that lacks DNA-binding activity on its own but can act in concert with other proteins to regulate embryologic development of the fly. Expression of both Grg2 and Grg6 was upregulated 10- to 50-fold by E2A-HLF. Immunoblot analysis detected increased amounts of two additional Groucho-related proteins, Grg1 and Grg4, in cells expressing E2A-HLF. A mutant E2A-HLF protein with a disabled DNA-binding region also mediated pro-B cell survival and activated Groucho-related genes. Among the transcription factors known to interact with Groucho-related protein, only RUNX1 was appreciably downregulated by E2A-HLF. Our results identify a highly conserved family of transcriptional corepressors that are activated by E2A-HLF, and they suggest that downregulation of RUNX1 may contribute to E2A-HLF-mediated leukemogenesis.


Current Topics in Microbiology and Immunology | 1996

Transcriptional Regulation by the t(8;21 )Fusion Protein, AML-1/ETO

Scott W. Hiebert; James R. Downing; Noel Lenny; Shari Meyers

Certain chromosomal translocations are characteristically associated with acute myeloid leukemia (AML), suggesting that alteration of specific genes at the translocation breakpoints contribute to the genesis or maintenance of leukemia. The t(8;21) is the second most frequent chromosomal abnormality associated with AML, occurring in 12–15% of cases (Downing et al. 1993). The t(8;21) breakpoint was cloned and the disrupted gene on chromosome 21 was termed AML1 (Miyoshi et al. 1991). Cytogenetic evidence from t(8;21)-containing leukemia cells with complex translocations indicates that the der(8) chromosome is conserved in each case; tightly linking this chromosomal aberration to the pathogenesis of AML (Rowley 1982). In addition, AML1 is interrupted in the less commonly observed (3;21) and (12;21) translocations found in chronic myeloid leukemia and acute lymphocytic leukemia, respectively (Nucifora et al. 1993; Golub et al. 1995), suggesting that AML-l may be a principal regulatory protein.


Leukemia | 1997

CBFA2, frequently rearranged in leukemia, is not responsible for a familial leukemia syndrome.

Rd Legare; Lu D; M Gallagher; C Ho; X Tan; G. F. Barker; K Shimizu; M Ohki; Noel Lenny; Scott W. Hiebert; Gilliland Dg

We have identified a family with an autosomal dominant platelet disorder with a predisposition for developing myeloid malignancies and have previously demonstrated linkage of this trait to chromosome 21q22.1-22.2. The nearest flanking markers, D21S1265 and D21S167, define the familial platelet disorder (FPD) critical region at a genetic distance of approximately 15.2 centimorgans and physical distance of approximately 6 megabases. This locus is of particular interest as it has previously been implicated in the pathogenesis of acute myelogenous leukemia (AML) and acute lymphoblastic leukemia (ALL) through the (8;21), (3;21) and (12;21) chromosomal translocations. In each of these cases, the CBFA2 gene is rearranged. As well, there is a potential association of this locus with the hematologic abnormalities seen in Down syndrome (trisomy 21). To identify the mutant gene in this pedigree, a positional cloning strategy has been undertaken. Several candidate genes map to this locus including: CBFA2, IFNAR1, IFNAR2, CRFB4, GART, SON, KCNE1, SCL5A3 and ATP50. CBFA2, as well as IFNAR1 and CRFB4, were the focus of initial mutational analysis efforts. In this report, we exclude CBFA2 as a candidate by Northern and Southern blotting, RNase protection, single-strand conformational polymorphism (SSCP), direct sequencing and gel-shift analysis. Exons of the IFNAR1 and CRFB4 genes were also analyzed by SSCP and demonstrated no evidence of mutation. SSCP analysis identified a new polymorphism in the second exon of the CRFB4 gene and confirmed a previously described polymorphism in the fourth exon of IFNAR1. Efforts are currently underway to delimit further the FPD critical region and to analyze the other known candidate genes, as well as novel candidate genes, which map to this locus.


Clinical Microbiology: Open Access | 2018

Traumatic Lumbar Puncture is Unlikely to Reduce Modern Molecular Detection of HSV Encephalitis (HSVE)

Kacy Ramirez; Rebecca Thompson; Ishita Pareek; Carson House; Noel Lenny; Anami Patel; John P. DeVincenzo

Polymerase chain reaction (PCR) is the gold-standard for diagnosing HSVE, but may be inhibited by blood contamination. We mimicked traumatic lumbar puncture and measured its effect on the molecular detection of HSV-1 in CSF. Clinically meaningful reductions in sensitivity were not observed. Pre-PCR processing allows sensitive detection of HSVE despite traumatic lumbar puncture.


Blood | 2003

Classification of pediatric acute lymphoblastic leukemia by gene expression profiling

Mary E. Ross; Xiaodong Zhou; Guangchun Song; Sheila A. Shurtleff; Kevin Girtman; W. Kent Williams; Hsi Che Liu; Rami Mahfouz; Susana C. Raimondi; Noel Lenny; Anami Patel; James R. Downing

Collaboration


Dive into the Noel Lenny's collaboration.

Top Co-Authors

Avatar

James R. Downing

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shari Meyers

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Anami Patel

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sheila A. Shurtleff

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bart Lutterbach

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Charles G. Mullighan

St. Jude Children's Research Hospital

View shared research outputs
Top Co-Authors

Avatar

Christopher B. Miller

St. Jude Children's Research Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge