Network


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

Hotspot


Dive into the research topics where Channabasavaiah B. Gurumurthy is active.

Publication


Featured researches published by Channabasavaiah B. Gurumurthy.


Journal of Biological Chemistry | 2007

The Notch Regulator MAML1 Interacts with p53 and Functions as a Coactivator

Yongtong Zhao; Rebecca B. Katzman; Laurie M. Delmolino; Ishfaq Bhat; Ying Zhang; Channabasavaiah B. Gurumurthy; Aleksandra Germaniuk-Kurowska; Honey V. Reddi; Aharon Solomon; Mu Sheng Zeng; Aisha Kung; Hui Ma; Qingshen Gao; Goberdhan P. Dimri; Adina Stanculescu; Lucio Miele; Lizi Wu; James D. Griffin; David E. Wazer; Hamid Band; Vimla Band

Members of the evolutionarily conserved Mastermind (MAM) protein family, including the three related mammalian Mastermind-like (MAML) proteins MAML1–3, function as crucial coactivators of Notch-mediated transcriptional activation. Given the recent evidence of cross-talk between the p53 and Notch signal transduction pathways, we have investigated whether MAML1 may also be a transcriptional coactivator of p53. Indeed, we show here that MAML1 is able to interact with p53. We show that MAML1-p53 interaction involves the N-terminal region of MAML1 and the DNA-binding domain of p53, and we use a chromatin immunoprecipitation assay to show that MAML1 is part of the activator complex that binds to native p53-response elements within the promoter of the p53 target genes. Overexpression of wild-type MAML1 as well as a mutant, defective in Notch signaling, enhanced the p53-dependent gene induction in mammalian cells, whereas MAML1 knockdown reduced the p53-dependent gene expression. MAML1 increases the half-life of p53 protein and enhances its phosphorylation/acetylation upon DNA damage of cells. Finally, RNA interference-mediated knockdown of the single Caenorhabditis elegans MAML homolog, Lag-3, led to substantial abrogation of p53-mediated germ-cell apoptotic response to DNA damage and markedly reduced the expression of Ced-13 and Egl-1, downstream pro-apoptotic targets of the C. elegans p53 homolog Cep-1. Thus, we present evidence for a novel coactivator function of MAML1 for p53, independent of its function as a coactivator of Notch signaling pathway.


Genome Biology | 2017

Easi-CRISPR: a robust method for one-step generation of mice carrying conditional and insertion alleles using long ssDNA donors and CRISPR ribonucleoproteins

Rolen M. Quadros; Hiromi Miura; Donald W. Harms; Hisako Akatsuka; Takehito Sato; Tomomi Aida; Ronald Redder; Guy P. Richardson; Yutaka Inagaki; Daisuke Sakai; Shannon M. Buckley; Parthasarathy Seshacharyulu; Surinder K. Batra; Mark A. Behlke; Sarah A. Zeiner; Ashley M. Jacobi; Yayoi Izu; Wallace B. Thoreson; Lisa D. Urness; Suzanne L. Mansour; Masato Ohtsuka; Channabasavaiah B. Gurumurthy

BackgroundConditional knockout mice and transgenic mice expressing recombinases, reporters, and inducible transcriptional activators are key for many genetic studies and comprise over 90% of mouse models created. Conditional knockout mice are generated using labor-intensive methods of homologous recombination in embryonic stem cells and are available for only ~25% of all mouse genes. Transgenic mice generated by random genomic insertion approaches pose problems of unreliable expression, and thus there is a need for targeted-insertion models. Although CRISPR-based strategies were reported to create conditional and targeted-insertion alleles via one-step delivery of targeting components directly to zygotes, these strategies are quite inefficient.ResultsHere we describe Easi-CRISPR (Efficient additions with ssDNA inserts-CRISPR), a targeting strategy in which long single-stranded DNA donors are injected with pre-assembled crRNA + tracrRNA + Cas9 ribonucleoprotein (ctRNP) complexes into mouse zygotes. We show for over a dozen loci that Easi-CRISPR generates correctly targeted conditional and insertion alleles in 8.5–100% of the resulting live offspring.ConclusionsEasi-CRISPR solves the major problem of animal genome engineering, namely the inefficiency of targeted DNA cassette insertion. The approach is robust, succeeding for all tested loci. It is versatile, generating both conditional and targeted insertion alleles. Finally, it is highly efficient, as treating an average of only 50 zygotes is sufficient to produce a correctly targeted allele in up to 100% of live offspring. Thus, Easi-CRISPR offers a comprehensive means of building large-scale Cre-LoxP animal resources.


Scientific Reports | 2015

CRISPR/Cas9-based generation of knockdown mice by intronic insertion of artificial microRNA using longer single-stranded DNA

Hiromi Miura; Channabasavaiah B. Gurumurthy; Takehito Sato; Masahiro Sato; Masato Ohtsuka

Knockdown mouse models, where gene dosages can be modulated, provide valuable insights into gene function. Typically, such models are generated by embryonic stem (ES) cell-based targeted insertion, or pronuclear injection, of the knockdown expression cassette. However, these methods are associated with laborious and time-consuming steps, such as the generation of large constructs with elements needed for expression of a functional RNAi-cassette, ES-cell handling, or screening for mice with the desired knockdown effect. Here, we demonstrate that reliable knockdown models can be generated by targeted insertion of artificial microRNA (amiRNA) sequences into a specific locus in the genome [such as intronic regions of endogenous eukaryotic translation elongation factor 2 (eEF-2) gene] using the Clustered Regularly Interspaced Short Palindromic Repeats/Crispr associated 9 (CRISPR/Cas9) system. We used in vitro synthesized single-stranded DNAs (about 0.5-kb long) that code for amiRNA sequences as repair templates in CRISPR/Cas9 mutagenesis. Using this approach we demonstrate that amiRNA cassettes against exogenous (eGFP) or endogenous [orthodenticle homeobox 2 (Otx2)] genes can be efficiently targeted to a predetermined locus in the genome and result in knockdown of gene expression. We also provide a strategy to establish conditional knockdown models with this method.


Scientific Reports | 2015

GONAD: Genome-editing via Oviductal Nucleic Acids Delivery system: a novel microinjection independent genome engineering method in mice.

Gou Takahashi; Channabasavaiah B. Gurumurthy; Kenta Wada; Hiromi Miura; Masahiro Sato; Masato Ohtsuka

Microinjection is considered the gold standard technique for delivery of nucleic acids (NAs; transgenes or genome editing tools such as CRISPR/Cas9 systems) into embryos, for creating genetically modified organisms. It requires sophisticated equipment as wel as well-trained and highly skilled personnel to perform the micro-injection technique. Here, we describe a novel and simple microinjection-independent technique, called Genome-editing via Oviductal Nucleic Acids Delivery (GONAD). Using GONAD, we show that NAs (e.g., eGFP mRNA or Cas9 mRNA/sgRNAs) can be effectively delivered to pre-implantation embryos within the intact mouse oviduct by a simple electroporation method, and result in the desired genetic modification in the embryos. Thus GONAD can bypass many complex steps in transgenic technology such as isolation of zygotes, microinjection of NAs into them, and their subsequent transfer to pseudo-pregnant animals. Furthermore, this method can potentially be used for genome editing in species other than mice.


Current protocols in human genetics | 2014

Mouse Genome Editing Using the CRISPR/Cas System

Donald W. Harms; Rolen M. Quadros; Davide Seruggia; Masato Ohtsuka; Gou Takahashi; Lluís Montoliu; Channabasavaiah B. Gurumurthy

The availability of techniques to create desired genetic mutations has enabled the laboratory mouse as an extensively used model organism in biomedical research including human genetics. A new addition to this existing technical repertoire is the CRISPR/Cas system. Specifically, this system allows editing of the mouse genome much more quickly than the previously used techniques, and, more importantly, multiple mutations can be created in a single experiment. Here we provide protocols for preparation of CRISPR/Cas reagents and microinjection into one‐cell mouse embryos to create knockout or knock‐in mouse models. Curr. Protoc. Hum. Genet. 83:15.7.1‐15.7.27.


PLOS ONE | 2011

Renal Thrombotic Microangiopathy in Mice with Combined Deletion of Endocytic Recycling Regulators EHD3 and EHD4

Manju George; Mark A. Rainey; Mayumi Naramura; Kirk W. Foster; Melissa S. Holzapfel; Laura L. Willoughby; Guo Guang Ying; Rasna Goswami; Channabasavaiah B. Gurumurthy; Vimla Band; Simon C. Satchell; Hamid Band

Eps15 Homology Domain-containing 3 (EHD3), a member of the EHD protein family that regulates endocytic recycling, is the first protein reported to be specifically expressed in the glomerular endothelium in the kidney; therefore we generated Ehd3 –/– mice and assessed renal development and pathology. Ehd3 –/– animals showed no overt defects, and exhibited no proteinuria or glomerular pathology. However, as the expression of EHD4, a related family member, was elevated in the glomerular endothelium of Ehd3 –/– mice and suggested functional compensation, we generated and analyzed Ehd3 –/–; Ehd4 –/– mice. These mice were smaller, possessed smaller and paler kidneys, were proteinuric and died between 3–24 weeks of age. Detailed analyses of Ehd3 –/–; Ehd4 –/– kidneys demonstrated thrombotic microangiopathy (TMA)-like glomerular lesions including thickening and duplication of glomerular basement membrane, endothelial swelling and loss of fenestrations. Other changes included segmental podocyte foot process effacement, mesangial interposition, and abnormal podocytic and mesangial marker expression. The glomerular lesions observed were strikingly similar to those seen in human pre-eclampsia and mouse models of reduced VEGF expression. As altered glomerular endothelial VEGFR2 expression and localization and increased apoptosis was observed in the absence of EHD3 and EHD4, we propose that EHD-mediated endocytic traffic of key surface receptors such as VEGFR2 is essential for physiological control of glomerular function. Furthermore, Ehd3 –/–; Ehd4 –/– mice provide a unique model to elucidate mechanisms of glomerular endothelial injury which is observed in a wide variety of human renal and extra-renal diseases.


Journal of Biological Chemistry | 2016

Cleavage by caspase 8 and mitochondrial membrane association activate the BH3-only protein bid during TRAIL-induced apoptosis

Kai Huang; Jingjing Zhang; Katelyn L. O'Neill; Channabasavaiah B. Gurumurthy; Rolen M. Quadros; Yaping Tu; Xu Luo

The BH3-only protein Bid is known as a critical mediator of the mitochondrial pathway of apoptosis following death receptor activation. However, since full-length Bid possesses potent apoptotic activity, the role of a caspase-mediated Bid cleavage is not established in vivo. In addition, due to the fact that multiple caspases cleave Bid at the same site in vitro, the identity of the Bid-cleaving caspase during death receptor signaling remains uncertain. Moreover, as Bid maintains its overall structure following its cleavage by caspase 8, it remains unclear how Bid is activated upon cleavage. Here, Bid-deficient (Bid KO) colon cancer cells were generated by gene editing, and were reconstituted with wild-type or mutants of Bid. While the loss of Bid blocked apoptosis following treatment by TNF-related apoptosis inducing ligand (TRAIL), this blockade was relieved by re-introduction of the wild-type Bid. In contrast, the caspase-resistant mutant BidD60E and a BH3 defective mutant BidG94E failed to restore TRAIL-induced apoptosis. By generating Bid/Bax/Bak-deficient (TKO) cells, we demonstrated that Bid is primarily cleaved by caspase 8, not by effector caspases, to give rise to truncated Bid (tBid) upon TRAIL treatment. Importantly, despite the presence of an intact BH3 domain, a tBid mutant lacking the mitochondrial targeting helices (α6 and α7) showed diminished apoptotic activity. Together, these results for the first time establish that cleavage by caspase 8 and the subsequent association with the outer mitochondrial membrane are two critical events that activate Bid during death receptor-mediated apoptosis.


Journal of Biological Chemistry | 2012

Mammalian Alteration/Deficiency in Activation 3 (Ada3) Is Essential for Embryonic Development and Cell Cycle Progression

Shakur Mohibi; Channabasavaiah B. Gurumurthy; Alo Nag; Jun Wang; Sameer Mirza; Yousaf Mian; Meghan Quinn; Bryan J. Katafiasz; James D. Eudy; Sanjit Pandey; Chittibabu Guda; Mayumi Naramura; Hamid Band; Vimla Band

Background: Ada3 is a core component of HAT containing coactivator complexes. Results: Germline deletion of Ada3 is embryonic lethal, and cell deletion leads to abnormal cell cycle progression. Conclusion: Ada3 is a critical protein at organismic and cellular level. Significance: This study describes a novel role of Ada3, a component of HAT complexes, as a critical regulator of cell survival. Ada3 protein is an essential component of histone acetyl transferase containing coactivator complexes conserved from yeast to human. We show here that germline deletion of Ada3 in mouse is embryonic lethal, and adenovirus-Cre mediated conditional deletion of Ada3 in Ada3FL/FL mouse embryonic fibroblasts leads to a severe proliferation defect which was rescued by ectopic expression of human Ada3. A delay in G1 to S phase of cell cycle was also seen that was due to accumulation of Cdk inhibitor p27 which was an indirect effect of c-myc gene transcription control by Ada3. We further showed that this defect could be partially reverted by knocking down p27. Additionally, drastic changes in global histone acetylation and changes in global gene expression were observed in microarray analyses upon loss of Ada3. Lastly, formation of abnormal nuclei, mitotic defects and delay in G2/M to G1 transition was seen in Ada3 deleted cells. Taken together, we provide evidence for a critical role of Ada3 in embryogenesis and cell cycle progression as an essential component of HAT complex.


Methods | 2017

Simplified CRISPR tools for efficient genome editing and streamlined protocols for their delivery into mammalian cells and mouse zygotes

Ashley M. Jacobi; Garrett Richard Rettig; Rolf Turk; Michael A. Collingwood; Sarah A. Zeiner; Rolen M. Quadros; Donald W. Harms; Paul J. Bonthuis; Christopher Gregg; Masato Ohtsuka; Channabasavaiah B. Gurumurthy; Mark A. Behlke

Genome editing using the CRISPR/Cas9 system requires the presence of guide RNAs bound to the Cas9 endonuclease as a ribonucleoprotein (RNP) complex in cells, which cleaves the host cell genome at sites specified by the guide RNAs. New genetic material may be introduced during repair of the double-stranded break via homology dependent repair (HDR) if suitable DNA templates are delivered with the CRISPR components. Early methods used plasmid or viral vectors to make these components in the host cell, however newer approaches using recombinant Cas9 protein with synthetic guide RNAs introduced directly as an RNP complex into cells shows faster onset of action with fewer off-target effects. This approach also enables use of chemically modified synthetic guide RNAs that have improved nuclease stability and reduces the risk of triggering an innate immune response in the host cell. This article provides detailed methods for genome editing using the RNP approach with synthetic guide RNAs using lipofection or electroporation in mammalian cells or using microinjection in murine zygotes, with or without addition of a single-stranded HDR template DNA.


Journal of Cellular Physiology | 2015

TGFβ3 Regulates Periderm Removal Through ΔNp63 in the Developing Palate

Lihua Hu; Zhi Li; Ferhat Ozturk; Channabasavaiah B. Gurumurthy; Rose-Anne Romano; Satrajit Sinha; Ali Nawshad

The periderm is a flat layer of epithelium created during embryonic development. During palatogenesis, the periderm forms a protective layer against premature adhesion of the oral epithelia, including the palate. However, the periderm must be removed in order for the medial edge epithelia (MEE) to properly adhere and form a palatal seam. Improper periderm removal results in a cleft palate. Although the timing of transforming growth factor β3 (TGFβ3) expression in the MEE coincides with periderm degeneration, its role in periderm desquamation is not known. Interestingly, murine models of knockout (−/−) TGFβ3, interferon regulatory factor 6 (IRF6) (−/−), and truncated p63 (ΔNp63) (−/−) are born with palatal clefts because of failure of the palatal shelves to adhere, suggesting that these genes regulate palatal epithelial differentiation. However, despite having similar phenotypes in null mouse models, no studies have analyzed the possible association between the TGFβ3 signaling cascade and the IRF6/ΔNp63 genes during palate development. Recent studies indicate that regulation of ΔNp63, which depends on IRF6, facilitates epithelial differentiation. We performed biochemical analysis, gene activity and protein expression assays with palatal sections of TGFβ3 (−/−), ΔNp63 (−/−), and wild‐type (WT) embryos, and primary MEE cells from WT palates to analyze the association between TGFβ3 and IRF6/ΔNp63. Our results suggest that periderm degeneration depends on functional TGFβ3 signaling to repress ΔNp63, thereby coordinating periderm desquamation. Cleft palate occurs in TGFβ3 (−/−) because of inadequate periderm removal that impedes palatal seam formation, while cleft palate occurs in ΔNp63 (−/−) palates because of premature fusion. J. Cell. Physiol. 230: 1212–1225, 2015.

Collaboration


Dive into the Channabasavaiah B. Gurumurthy's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rolen M. Quadros

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Vimla Band

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hamid Band

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Donald W. Harms

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Sameer Mirza

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Shakur Mohibi

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Aditya Bele

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge