Network


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

Hotspot


Dive into the research topics where Ashok P. Bidwai is active.

Publication


Featured researches published by Ashok P. Bidwai.


Molecular and Cellular Biology | 2005

An eh1-like motif in odd-skipped mediates recruitment of groucho and repression in vivo

Robert Goldstein; Orna Cook; Tama Dinur; Anne Pisanté; Umesh Karandikar; Ashok P. Bidwai; Ze’ev Paroush

ABSTRACT Drosophila Groucho, like its vertebrate Transducin-like Enhancer-of-split homologues, is a corepressor that silences gene expression in numerous developmental settings. Groucho itself does not bind DNA but is recruited to target promoters by associating with a large number of DNA-binding negative transcriptional regulators. These repressors tether Groucho via short conserved polypeptide sequences, of which two have been defined. First, WRPW and related tetrapeptide motifs have been well characterized in several repressors. Second, a motif termed Engrailed homology 1 (eh1) has been found predominantly in homeodomain-containing transcription factors. Here we describe a yeast two-hybrid screen that uncovered physical interactions between Groucho and transcription factors, containing eh1 motifs, with different types of DNA-binding domains. We show that one of these, the zinc finger protein Odd-skipped, requires its eh1-like sequence for repressing specific target genes in segmentation. Comparison between diverse eh1 motifs reveals a bias for the phosphoacceptor amino acids serine and threonine at a fixed position, and a mutational analysis of Odd-skipped indicates that these residues are critical for efficient interactions with Groucho and for repression in vivo. Our data suggest that phosphorylation of these phosphomeric residues, if it occurs, will down-regulate Groucho binding and therefore repression, providing a mechanism for posttranslational control of Groucho-mediated repression.


Genesis | 2009

Functional dissection of Timekeeper (Tik) implicates opposite roles for CK2 and PP2A during Drosophila neurogenesis

Ezgi Kunttas-Tatli; Anasua Bose; Bhaskar Kahali; Clifton P. Bishop; Ashok P. Bidwai

Repression by E(spl)M8 during inhibitory Notch (N) signaling (lateral inhibition) is regulated, in part, by protein kinase CK2, but the involvement of a phosphatase has been unclear. The studies we report here employ Tik, a unique dominant‐negative (DN) mutation in the catalytic subunit of CK2, in a Gal4‐UAS based assay for impaired lateral inhibition. Specifically, overexpression of Tik elicits ectopic bristles in N+ flies and suppresses the retinal defects of the gain‐of‐function allele Nspl. Functional dissection of the two substitutions in Tik (M161K and E165D), suggests that both mutations contribute to its DN effects. While the former replacement compromises CK2 activity by impairing ATP‐binding, the latter affects a conserved motif implicated in binding the phosphatase PP2A. Accordingly, overexpression of microtubule star (mts), the PP2A catalytic subunit closely mimics the phenotypic effects of loss of CK2 functions in N+ or Nspl flies, and elicits notched wings, a characteristic of N mutations. Our findings suggest antagonistic roles for CK2 and PP2A during inhibitory N signaling. genesis 47:647–658, 2009.


Biochemical and Biophysical Research Communications | 2002

Interaction of casein kinase II with ribosomal protein L22 of Drosophila melanogaster

Wenfan Zhao; Ashok P. Bidwai; Claiborne V.C. Glover

The ubiquitous eukaryotic protein kinase CKII (casein kinase II) has been found to interact with a number of cellular proteins, either through the catalytic subunit or the regulatory subunit. Using the yeast two-hybrid screening method, we found that the catalytic subunit of Drosophila melanogaster CKII (DmCKII) interacts with Drosophila ribosomal protein L22 (rpL22). This interaction was also observed in vitro with a glutathione-S-transferase (GST)-rpL22 fusion protein. The predicted full-length Drosophila rpL22 protein has an N-terminal extension rich in alanine, lysine, and proline that appears to be unique to Drosophila. Deletion mapping revealed that the conserved core of rpL22 is responsible for the interaction with CKII. Moreover, purified DmCKII can phosphorylate a GST-L22 fusion protein at the C-terminal end, suggesting that this protein may be a substrate of CKII in Drosophila.


Molecular and Cellular Biochemistry | 2009

Drosophila protein kinase CK2 is rendered temperature-sensitive by mutations of highly conserved residues flanking the activation segment

Pallavi P. Kuntamalla; Ezgi Kunttas-Tatli; Umesh Karandikar; Clifton P. Bishop; Ashok P. Bidwai

CK2 is a Ser/Thr protein kinase essential for animal development. Although null alleles for CK2 are available in the mouse and Drosophila models, they are lethal when homozygous, thus necessitating conditional alleles for analysis of its developmental roles. We describe the isolation of temperature-sensitive (ts) alleles of Drosophila CK2α (dCK2α). These alleles efficiently rescue lethality of yeast lacking endogenous CK2 at 29°C, but this ability is lost at higher temperatures in an allele-specific manner. These ts-variants exhibit properties akin to the wild type protein, and interact robustly with dCK2β. Modeling of these ts-variants using the crystal structure of human CK2α indicates that the affected residues are in close proximity to the active site. We find that substitution of Asp212 elicits potent ts-behavior, an important finding because this residue contributes to stability of the activation segment and is invariant in other Ser/Thr protein kinases.


Genesis | 2009

On the mechanism underlying the divergent retinal and bristle defects of M8* (E(spl)D) in Drosophila

Bhaskar Kahali; Anasua Bose; Umesh Karandikar; Clifton P. Bishop; Ashok P. Bidwai

Our results, using endogenous mutants and Gal4‐UAS driven transgenes, implicate multisite phosphorylation in repression by E(spl)M8. We propose that these phosphorylations occur in the morphogenetic furrow (MF) to reverse an auto‐inhibited state of M8, enabling repression of Atonal during R8 specification. Our studies address the paradoxical behavior of M8*, the truncated protein encoded by E(spl)D. We suggest that differences in N signaling in the bristle versus the eye underlie the antimorphic activity of M8* in N+ (ectopic bristles) and hypermorphic activity in Nspl (reduced eye). Ectopic M8* impairs eye development (in Nspl) only during establishment of the atonal feedback loop (anterior to the MF), but is ineffective after this time point. In contrast, a CK2 phosphomimetic M8 lacking Groucho (Gro) binding, M8SDΔGro, acts antimorphic in N+ and suppresses the eye/R8 and bristle defects of Nspl, as does reduced dosage of E(spl) or CK2. Multisite phosphorylation could serve as a checkpoint to enable a precise onset of repression, and this is bypassed in M8*. Additional implications are discussed. genesis 47:456–468, 2009.


Biochemical and Biophysical Research Communications | 2008

Drosophila CK2 phosphorylates Hairy and regulates its activity in vivo

Bhaskar Kahali; Regina L. Trott; Ze’ev Paroush; Ravi Allada; Clifton P. Bishop; Ashok P. Bidwai

Hairy is a repressor that regulates bristle patterning, and its loss elicits ectopic bristles (neural hyperplasia). However, it has remained unknown whether Hairy is regulated by phosphorylation. We describe here the interaction of protein kinase CK2 and Hairy. Hairy is robustly phosphorylated by the CK2-holoenzyme (CK2-HoloE) purified from Drosophila embryos, but weakly by the catalytic CK2alpha-subunit alone, suggesting that this interaction requires the regulatory CK2beta-subunit. Consistent with this, Hairy preferentially forms a direct complex with CK2-HoloE. Importantly, we demonstrate genetic interactions between CK2 and hairy (h). Thus, flies trans-heterozygous for alleles of CK2alpha and h display neural hyperplasia akin to homozygous hypomorphic h alleles. In addition, we show that similar phenotypes are elicited in wild-type flies upon expression of RNAi constructs against CK2alpha/beta, and that these defects are sensitive to h gene dosage. Together, these studies suggest that CK2 contributes to repression by Hairy.


Genesis | 2009

Evidence that the C-Terminal Domain (CtD) Autoinhibits Neural Repression by Drosophila E(spl)M8

Bhaskar Kahali; Jee-Eun Kim; Umesh Karandikar; Clifton P. Bishop; Ashok P. Bidwai

Analysis of the retinal defects of a CK2 phosphomimetic variant of E(spl)M8 (M8S159D) and the truncated protein M8* encoded by the E(spl)D allele, suggest that the nonphosphorylated CtD “autoinhibits” repression. We have investigated this model by testing for inhibition (in “trans”) by the CtD fragment in its nonphosphorylated (M8‐CtD) and phosphomimetic (M8SD‐CtD) states. In N+ flies, ectopic M8‐CtD compromises lateral inhibition, i.e., elicits supernumerary bristles as with loss of N signaling. This antimorphic activity of M8‐CtD strongly rescues the reduced eye and/or bristle loss phenotypes that are elicited by ectopic M8SD or wild type M8. Additionally, the severely reduced eye of Nspl/Y; E(spl)D/+ flies is also rescued by M8‐CtD. Rescue is specific to the time and place, the morphogenetic furrow, where “founding” R8 photoreceptors are specified. In contrast, the phosphomimetic M8SD‐CtD that is predicted to be deficient for autoinhibition, exhibits significantly attenuated or negligible activity. These studies provide evidence that autoinhibition by the CtD regulates M8 activity in a phosphorylation‐dependent manner. genesis 48:44–55, 2010.


Molecular and Cellular Biochemistry | 2001

Identification and characterization of proteins that interact with Drosophila melanogaster protein kinase CK2

Regina L. Trott; Madhavi Kalive; Urnesh Karandikar; Rebecca Rummer; Clifton P. Bishop; Ashok P. Bidwai

D. melanogaster CK2 (DmCK2) is a highly conserved protein kinase that is composed of catalytic, α, and regulatory, β, subunits associated as an α2β2 heterotetramer. In order to analyze the functions of CK2 in this metazoan model, we have used the two hybrid approach to identify interacting proteins. One of these cDNAs, DmA24, encodes a novel polypeptide with no homologs in GenBank, and is notable in that it contains a bipartite nuclear localization signal and two sites for phosphorylation by CK2. In situ hybridization to polytene chromosomes indicates that the DmA24 gene is located at the 61D interval of chromosome II a region that also harbors 3 additional genes with similar structure. DmA24p interacts with DmCK2α, but not with DmCK2β, demonstrating that this interaction is specific for the catalytic subunit of CK2. In addition, the protein is phosphorylated by the holoenzyme purified from Drosophila embryos. These studies identify DmA24p as a potentially new physiological partner of DmCK2. In addition, we also report the results of a large-scale screen that has identified a new set of DmCK2-interacting proteins. Most notable among these are Surf6, a nucleolar protein involved in RNA processing, and Spalt, a homeotic protein. (Mol Cell Biochem 227: 91-98, 2001)


PLOS ONE | 2014

The Ser/Thr phosphatase PP2A regulatory subunit widerborst inhibits notch signaling.

Anasua Bose; Adam T. Majot; Ashok P. Bidwai

Drosophila Enhancer of split M8, an effector of Notch signaling, is regulated by protein kinase CK2. The phosphatase PP2A is thought to play an opposing (inhibitory) role, but the identity of the regulatory subunit was unknown. The studies described here reveal a role for the PP2A regulatory subunit widerborst (wdb) in three developmental contexts; the bristle, wing and the R8 photoreceptors of the eye. wdb overexpression elicits bristle and wing defects akin to reduced Notch signaling, whereas hypomorphic mutations in this PP2A subunit elicit opposite effects. We have also evaluated wdb functions using mutations in Notch and E(spl) that affect the eye. We find that the eye and R8 defects of the well-known Nspl mutation are enhanced by a hypomorphic allele of wdb, whereas they are strongly rescued by wdb overexpression. Similarly, ectopic wdb rescues the eye and R8 defects of the E(spl)D mutation, which affects the m8 gene. In addition, wdb overexpression also rescues the bristle defects of ectopically expressed M8, or the eye and R8 defects of its CK2 phosphomimetic variant M8-S159D. The latter finding suggests that PP2A may target M8 at highly conserved residues in the vicinity of the CK2 site, whose phosphorylation controls repression of Atonal and the R8 fate. Together, the studies identify PP2A-Wdb as a participant in Notch signaling, and suggest that M8 activity is controlled by phosphorylation and dephosphorylation. The conservation of the phosphorylation sites between Drosophila E(spl) and the HES/HER proteins from mammals, reptiles, amphibians, birds and fish raises the prospect that this mode of regulation is widespread.


Molecular and Cellular Biochemistry | 2005

Drosophila CK2 phosphorylates Deadpan, a member of the HES family of basic-helix-loop-helix (bHLH) repressors

Umesh Karandikar; Jonathan Shaffer; Clifton P. Bishop; Ashok P. Bidwai

In Drosophila, protein kinase CK2 regulates a diverse array of developmental processes. One of these is cell-fate specification (neurogenesis) wherein CK2 regulates basic-helix-loop-helix (bHLH) repressors encoded by the Enhancer of Split Complex (E(spl)C). Specifically, CK2 phosphorylates and activates repressor functions of E(spl)M8 during eye development. In this study we describe the interaction of CK2 with an E(spl)-related bHLH repressor, Deadpan (Dpn). Unlike E(spl)-repressors which are expressed in cells destined for a non-neural cell fate, Dpn is expressed in the neuronal cells and is thought to control the activity of proneural genes. Dpn also regulates sex-determination by repressing sxl, the primary gene involved in sex differentiation. We demonstrate that Dpn is weakly phosphorylated by monomeric CK2α, whereas it is robustly phosphorylated by the embryo-holoenzyme, suggesting a positive role for CK2β. The weak phosphorylation by CK2α is markedly stimulated by the activator polylysine to levels comparable to those with the holoenzyme. In addition, pull down assays indicate a direct interaction between Dpn and CK2. This is the first demonstration that Dpn is a partner and target of CK2, and raises the possibility that its repressor functions might also be regulated by phosphorylation.

Collaboration


Dive into the Ashok P. Bidwai's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Bhaskar Kahali

West Virginia University

View shared research outputs
Top Co-Authors

Avatar

Anasua Bose

West Virginia University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adam T. Majot

West Virginia University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Madhavi Kalive

Arizona State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge