Network


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

Hotspot


Dive into the research topics where Neil Cooch is active.

Publication


Featured researches published by Neil Cooch.


Nature | 2004

The Microprocessor complex mediates the genesis of microRNAs

Richard I. Gregory; Kai Ping Yan; Govindasamy Amuthan; Thimmalah Chendrimada; Behzad Doratotaj; Neil Cooch; Ramin Shiekhattar

MicroRNAs (miRNAs) are a growing family of small non-protein-coding regulatory genes that regulate the expression of homologous target-gene transcripts. They have been implicated in the control of cell death and proliferation in flies, haematopoietic lineage differentiation in mammals, neuronal patterning in nematodes and leaf and flower development in plants. miRNAs are processed by the RNA-mediated interference machinery. Drosha is an RNase III enzyme that was recently implicated in miRNA processing. Here we show that human Drosha is a component of two multi-protein complexes. The larger complex contains multiple classes of RNA-associated proteins including RNA helicases, proteins that bind double-stranded RNA, novel heterogeneous nuclear ribonucleoproteins and the Ewings sarcoma family of proteins. The smaller complex is composed of Drosha and the double-stranded-RNA-binding protein, DGCR8, the product of a gene deleted in DiGeorge syndrome. In vivo knock-down and in vitro reconstitution studies revealed that both components of this smaller complex, termed Microprocessor, are necessary and sufficient in mediating the genesis of miRNAs from the primary miRNA transcript.


Nature | 2005

TRBP recruits the Dicer complex to Ago2 for microRNA processing and gene silencing

Thimmaiah P. Chendrimada; Richard I. Gregory; Easwari Kumaraswamy; Jessica Norman; Neil Cooch; Kazuko Nishikura; Ramin Shiekhattar

MicroRNAs (miRNAs) are generated by a two-step processing pathway to yield RNA molecules of approximately 22 nucleotides that negatively regulate target gene expression at the post-transcriptional level. Primary miRNAs are processed to precursor miRNAs (pre-miRNAs) by the Microprocessor complex. These pre-miRNAs are cleaved by the RNase III Dicer to generate mature miRNAs that direct the RNA-induced silencing complex (RISC) to messenger RNAs with complementary sequence. Here we show that TRBP (the human immunodeficiency virus transactivating response RNA-binding protein), which contains three double-stranded, RNA-binding domains, is an integral component of a Dicer-containing complex. Biochemical analysis of TRBP-containing complexes revealed the association of Dicer–TRBP with Argonaute 2 (Ago2), the catalytic engine of RISC. The physical association of Dicer–TRBP and Ago2 was confirmed after the isolation of the ternary complex using Flag-tagged Ago2 cell lines. In vitro reconstitution assays demonstrated that TRBP is required for the recruitment of Ago2 to the small interfering RNA (siRNA) bound by Dicer. Knockdown of TRBP results in destabilization of Dicer and a consequent loss of miRNA biogenesis. Finally, depletion of the Dicer–TRBP complex via exogenously introduced siRNAs diminished RISC-mediated reporter gene silencing. These results support a role of the Dicer–TRBP complex not only in miRNA processing but also as a platform for RISC assembly.


Cell | 2005

Human RISC Couples MicroRNA Biogenesis and Posttranscriptional Gene Silencing

Richard I. Gregory; Thimmaiah P. Chendrimada; Neil Cooch; Ramin Shiekhattar

RNA interference is implemented through the action of the RNA-induced silencing complex (RISC). Although Argonaute2 has been identified as the catalytic center of RISC, the RISC polypeptide composition and assembly using short interfering RNA (siRNA) duplexes has remained elusive. Here we show that RISC is composed of Dicer, the double-stranded RNA binding protein TRBP, and Argonaute2. We demonstrate that this complex can cleave target RNA using precursor microRNA (pre-miRNA) hairpin as the source of siRNA. Although RISC can also utilize duplex siRNA, it displays a nearly 10-fold greater activity using the pre-miRNA Dicer substrate. RISC distinguishes the guide strand of the siRNA from the passenger strand and specifically incorporates the guide strand. Importantly, ATP is not required for miRNA processing, RISC assembly, or multiple rounds of target-RNA cleavage. These results define the composition of RISC and demonstrate that miRNA processing and target-RNA cleavage are coupled.


Nature | 2005

An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation

Min Gyu Lee; Christopher Wynder; Neil Cooch; Ramin Shiekhattar

We have previously described a multiprotein complex termed the BHC or BRAF–HDAC complex, which is required for the repression of neuronal-specific genes. We have shown that the BHC complex is recruited by a neuronal silencer, REST (RE1-silencing transcription factor), and mediates the repression of REST-responsive genes. BHC is a multiprotein complex consisting of two enzymatic activities: a histone deacetylase (HDAC1 or 2) and a recently described histone demethylase (BHC110, also known as LSD1 or AOF2). Here we show that BHC110-containing complexes show a nearly fivefold increase in demethylation of histone H3 lysine 4 (H3K4) compared to recombinant BHC110. Furthermore, recombinant BHC110 is unable to demethylate H3K4 on nucleosomes, but BHC110-containing complexes readily demethylate nucleosomes. In vitro reconstitution of the BHC complex using recombinant subunits reveals an essential role for the REST corepressor CoREST, not only in stimulating demethylation on core histones but also promoting demethylation of nucleosomal substrates. We find that nucleosomal demethylation is the result of CoREST enhancing the association between BHC110 and nucleosomes. Depletion of CoREST in in vivo cell culture results in de-repression of REST-responsive gene expression and increased methylation of H3K4. Together, these results highlight an essential role for CoREST in demethylation of H3K4 both in vitro and in vivo.


Molecular Cell | 2003

Regulation of BRCC, a holoenzyme complex containing BRCA1 and BRCA2, by a signalosome-like subunit and its role in DNA repair

Yuanshu Dong; Mohamed-Ali Hakimi; Xiaowei Chen; Easwari Kumaraswamy; Neil Cooch; Andrew K. Godwin; Ramin Shiekhattar

We have isolated a holoenzyme complex termed BRCC containing BRCA1, BRCA2, and RAD51. BRCC not only displays increased association with p53 following DNA damage but also ubiquitinates p53 in vitro. BRCC36 and BRCC45 are novel components of the complex with sequence homology to a subunit of the signalosome and proteasome complexes. Reconstitution of a recombinant four-subunit complex containing BRCA1/BARD1/BRCC45/BRCC36 revealed an enhanced E3 ligase activity compared to that of BRCA1/BARD1 heterodimer. In vivo, depletion of BRCC36 and BRCC45 by the small interfering RNAs (siRNAs) resulted in increased sensitivity to ionizing radiation and defects in G2/M checkpoint. BRCC36 shows aberrant expression in sporadic breast tumors. These findings identify BRCC as a ubiquitin E3 ligase complex that enhances cellular survival following DNA damage.


Molecular and Cellular Biology | 2006

Functional interplay between histone demethylase and deacetylase enzymes

Min Gyu Lee; Christopher Wynder; Daniel A. Bochar; Mohamed-Ali Hakimi; Neil Cooch; Ramin Shiekhattar

ABSTRACT Histone deacetylase (HDAC) inhibitors are a promising class of anticancer agents for the treatment of solid and hematological malignancies. The precise mechanism by which HDAC inhibitors mediate their effects on tumor cell growth, differentiation, and/or apoptosis is the subject of intense research. Previously we described a family of multiprotein complexes that contain histone deacetylase 1/2 (HDAC1/2) and the histone demethylase BHC110 (LSD1). Here we show that HDAC inhibitors diminish histone H3 lysine 4 (H3K4) demethylation by BHC110 in vitro. In vivo analysis revealed an increased H3K4 methylation concomitant with inhibition of nucleosomal deacetylation by HDAC inhibitors. Reconstitution of recombinant complexes revealed a functional connection between HDAC1 and BHC110 only when nucleosomal substrates were used. Importantly, while the enzymatic activity of BHC110 is required to achieve optimal deacetylation in vitro, in vivo analysis following ectopic expression of an enzymatically dead mutant of BHC110 (K661A) confirmed the functional cross talk between the demethylase and deacetylase enzymes. Our studies not only reveal an intimate link between the histone demethylase and deacetylase enzymes but also identify histone demethylation as a secondary target of HDAC inhibitors.


Molecular and Cellular Biology | 2007

Phosphorylation of MNAR Promotes Estrogen Activation of Phosphatidylinositol 3-Kinase

James G. Greger; Natalie Fursov; Neil Cooch; Sean McLarney; Leonard P. Freedman; Dean P. Edwards; Boris J. Cheskis

ABSTRACT Estrogen actions are mediated by a complex interface of direct control of gene expression (the so-called “genomic action”) and by regulation of cell signaling/phosphorylation cascades, referred to as the “nongenomic,” or extranuclear, action. We have previously described the identification of MNAR (modulator of nongenomic action of estrogen receptor) as a novel scaffold protein that regulates estrogen receptor alpha (ERα) activation of cSrc. In this study, we have investigated the role of MNAR in 17β-estradiol (E2)-induced activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pathway. Consistent with our previous results, a direct correlation was established between MNAR expression levels and E2-induced activation of PI3 and Akt kinases. Endogenous MNAR, ERα, cSrc, and p85, the regulatory subunit of PI3 kinase, interacted in MCF7 cells treated with E2. The interaction between p85 and MNAR required activation of cSrc and MNAR phosphorylation on Tyr 920. Consequently, the mutation of this tyrosine to alanine (Y920A) abrogated the interaction between MNAR and p85 and the E2-induced activation of the PI3K/Akt pathway, which was required for the E2-induced protection of MCF7 cells from apoptosis. Nonetheless, the Y920A mutant potentiated the E2-induced activation of the Src/MAPK pathway and MCF7 cell proliferation, as observed with the wild-type MNAR. These results provide new and important insights into the molecular mechanisms of E2-induced regulation of cell proliferation and apoptosis.


Cancer Research | 2005

Linking Transcriptional Elongation and Messenger RNA Export to Metastatic Breast Cancers

Shanchun Guo; Mohamed-Ali Hakimi; David Baillat; Xiaowei Chen; Michele J. Farber; Andres J. Klein-Szanto; Neil Cooch; Andrew K. Godwin; Ramin Shiekhattar

The biochemical pathways that are disrupted in the genesis of sporadic breast cancers remain unclear. Moreover, the present prognosticating markers used to determine the prognosis of node-negative patient leads to probabilistic results, and the eventual clinical course is far from certain. Here we identified the human TREX complex, a multiprotein complex that links transcription elongation to mRNA transport, as culprit of aggressive human breast cancers. We show that whereas p84N5 (called hTREX84) is expressed at very low levels in normal breast epithelial cells, it is highly expressed in breast tumors. Importantly, hTREX84 expression correlates with tumor size and the metastatic state of the tumor progression. Reduction of hTREX84 levels in breast cancer cell lines by small interfering RNA result in inhibition of cellular proliferation and abrogation of mRNA export. These results not only identify hTREX84 as a prognosticator of breast cancer but also delineate human TREX complex as a target for therapeutic drugs against breast cancer.


Molecular and Cellular Biology | 2010

Phosphorylation of MNAR promotes estrogen activation of phosphatidylinositol 3-kinase. Retraction.

James G. Greger; Natalie Fursov; Neil Cooch; Sean McLarney; Leonard P. Freedman; Dean P. Edwards; Boris J. Cheskis

Volume 27, no. 5, p. 1904–1913, 2007. The publisher hereby retracts this article. Although the data showing an estrogendependent ternary complex of MNAR, estrogen receptor alpha (ER ), and the p85 subunit of phosphatidylinositol 3-kinase (PI3K) and the requirement for the activation of c-Src and ER for the MNAR-p85 interaction are accurate, Wyeth Research has determined that the data demonstrating the phosphorylation of the Y920 residue of MNAR by c-Src and the requirement of Y920 phosphorylation of MNAR for estrogen-mediated activation of PI3K are unreliable.


Cell | 2005

Integrator, a Multiprotein Mediator of Small Nuclear RNA Processing, Associates with the C-Terminal Repeat of RNA Polymerase II

David Baillat; Mohamed-Ali Hakimi; Anders M. Näär; Ali Shilatifard; Neil Cooch; Ramin Shiekhattar

Collaboration


Dive into the Neil Cooch's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Boris J. Cheskis

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Leonard P. Freedman

Memorial Sloan Kettering Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Dean P. Edwards

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

David Baillat

University of Texas Medical Branch

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge