Network


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

Hotspot


Dive into the research topics where Kevin N. Dalby is active.

Publication


Featured researches published by Kevin N. Dalby.


Autophagy | 2010

Targeting the prodeath and prosurvival functions of autophagy as novel therapeutic strategies in cancer

Kevin N. Dalby; Ibrahim Tekedereli; Gabriel Lopez-Berestein; Bulent Ozpolat

Autophagy is an evolutionarily conserved lysosomal pathway for degrading cytoplasmic proteins, macromolecules, and organelles. While autophagy has become one of the most attractive topics in cancer research the current autophagy literature is often viewed as confusing, because of its dual role and association with apparently contradictory roles, such as survival and cell death. Autophagy can serve as a tumor suppressor, as a partial reduction in autophagic capacity or defective autophagy (e.g. heterozygous knockdown beclin-1 (+/-) in mice) provides an oncogenic stimulus, causing malignant transformation and spontaneous tumors. In addition, autophagy seems to function as a protective cell survival mechanism against environmental and cellular stress (e.g., nutrient deprivation, hypoxia and therapeutic stress) and causes resistance to antineoplastic therapies. Recent studies have demonstrated that the inhibition of autophagy in cancer cells may be therapeutically beneficial in some circumstances, as it can sensitize cancer cells to different therapies, including DNA-damaging agents, antihormone therapies (e.g., tamoxifen), and radiation therapy. This supports the hypothesis that inhibiting autophagy can negatively influence cancer cell survival and increase cell death when combined with anticancer agents, providing a therapeutic advantage against cancer. On the other hand, the induction of autophagy by the inhibition of anti-autophagic proteins, such as Bcl-2, PKCδ, and tissue transglutaminase 2 (TG2), may lead to autophagic cell death in some apoptosis-resistant cancers (i.e., breast and pancreatic cancers), indicating that the induction of autophagy alone may also be used as a potential therapy. Overall, the data suggest that, depending on the cellular features, either the induction or the inhibition of autophagy can provide therapeutic benefits to patients and that the design and synthesis of the first-generation modulators of autophagy may provide the tools for proof of concept experiments and the impetus for translational studies that may ultimately lead to new therapeutic strategies in cancer.


Molecular Systems Biology | 2014

Construction of human activity-based phosphorylation networks.

Robert H. Newman; Jianfei Hu; Hee-Sool Rho; Zhi Xie; Crystal Woodard; John Neiswinger; Christopher D.O. Cooper; Matthew D. Shirley; Hillary M. Clark; Shaohui Hu; Woochang Hwang; Jun Seop Jeong; George Wu; Jimmy Lin; Xinxin Gao; Qiang Ni; Renu Goel; Shuli Xia; Hongkai Ji; Kevin N. Dalby; Morris J. Birnbaum; Philip A. Cole; Stefan Knapp; Alexey G. Ryazanov; Donald J. Zack; Seth Blackshaw; Tony Pawson; Anne-Claude Gingras; Stephen Desiderio; Akhilesh Pandey

The landscape of human phosphorylation networks has not been systematically explored, representing vast, unchartered territories within cellular signaling networks. Although a large number of in vivo phosphorylated residues have been identified by mass spectrometry (MS)‐based approaches, assigning the upstream kinases to these residues requires biochemical analysis of kinase‐substrate relationships (KSRs). Here, we developed a new strategy, called CEASAR, based on functional protein microarrays and bioinformatics to experimentally identify substrates for 289 unique kinases, resulting in 3656 high‐quality KSRs. We then generated consensus phosphorylation motifs for each of the kinases and integrated this information, along with information about in vivo phosphorylation sites determined by MS, to construct a high‐resolution map of phosphorylation networks that connects 230 kinases to 2591 in vivo phosphorylation sites in 652 substrates. The value of this data set is demonstrated through the discovery of a new role for PKA downstream of Btk (Brutons tyrosine kinase) during B‐cell receptor signaling. Overall, these studies provide global insights into kinase‐mediated signaling pathways and promise to advance our understanding of cellular signaling processes in humans.


Journal of Biological Chemistry | 2008

Substrate Discrimination among Mitogen-activated Protein Kinases through Distinct Docking Sequence Motifs

Douglas L. Sheridan; Yong Kong; Sirlester A. Parker; Kevin N. Dalby; Benjamin E. Turk

Mitogen-activated protein kinases (MAPKs) mediate cellular responses to a wide variety of extracellular stimuli. MAPK signal transduction cascades are tightly regulated, and individual MAPKs display exquisite specificity in recognition of their target substrates. All MAPK family members share a common phosphorylation site motif, raising questions as to how substrate specificity is achieved. Here we describe a peptide library screen to identify sequence requirements of the DEF site (docking site for ERK FXF), a docking motif separate from the phosphorylation site. We show that MAPK isoforms recognize DEF sites with unique sequences and identify two key residues on the MAPK that largely dictate sequence specificity. Based on these observations and computational docking studies, we propose a revised model for MAPK interaction with substrates containing DEF sites. Variations in DEF site sequence requirements provide one possible mechanism for encoding complex target specificity among MAPK isoforms.


Proteins | 2011

Virtual screening using molecular simulations

Tianyi Yang; Johnny C. Wu; Chunli Yan; Yuanfeng Wang; Ray Luo; Michael B. Gonzales; Kevin N. Dalby; Pengyu Ren

Effective virtual screening relies on our ability to make accurate prediction of protein‐ligand binding, which remains a great challenge. In this work, utilizing the molecular‐mechanics Poisson‐Boltzmann (or Generalized Born) surface area approach, we have evaluated the binding affinity of a set of 156 ligands to seven families of proteins, trypsin β, thrombin α, cyclin‐dependent kinase (CDK), cAMP‐dependent kinase (PKA), urokinase‐type plasminogen activator, β‐glucosidase A, and coagulation factor Xa. The effect of protein dielectric constant in the implicit‐solvent model on the binding free energy calculation is shown to be important. The statistical correlations between the binding energy calculated from the implicit‐solvent approach and experimental free energy are in the range of 0.56–0.79 across all the families. This performance is better than that of typical docking programs especially given that the latter is directly trained using known binding data whereas the molecular mechanics is based on general physical parameters. Estimation of entropic contribution remains the barrier to accurate free energy calculation. We show that the traditional rigid rotor harmonic oscillator approximation is unable to improve the binding free energy prediction. Inclusion of conformational restriction seems to be promising but requires further investigation. On the other hand, our preliminary study suggests that implicit‐solvent based alchemical perturbation, which offers explicit sampling of configuration entropy, can be a viable approach to significantly improve the prediction of binding free energy. Overall, the molecular mechanics approach has the potential for medium to high‐throughput computational drug discovery. Proteins 2011;


Journal of Neurochemistry | 2007

Dopamine depletion and subsequent treatment with l-DOPA, but not the long-lived dopamine agonist pergolide, enhances activity of the Akt pathway in the rat striatum

Evgeny Bychkov; M. Rafiuddin Ahmed; Kevin N. Dalby; Eugenia V. Gurevich

Dysregulation of signaling pathways is believed to contribute to Parkinson’s disease pathology and l‐DOPA‐induced motor complications. Long‐lived dopamine (DA) agonists are less likely to cause motor complications by virtue of continuous stimulation of DA receptors. In this study, we compared the effects of the unilateral 6‐hydroxydopamine lesion and subsequent treatment with l‐DOPA and DA agonist pergolide on signaling pathways in rats. Pergolide caused less pronounced behavioral sensitization than l‐DOPA (25 mg/kg, i.p., 10 days), particularly at lower dose (0.5 and 0.25 mg/kg, i.p.). Pergolide, but not l‐DOPA, reversed lesion‐induced up‐regulation of preproenkephalin and did not up‐regulate preprodynorphine or DA D3 receptor in the lesioned hemisphere. Pergolide was as effective as l‐DOPA in reversing the lesion‐induced elevation of ERK2 phosphorylation in response to acute apomorphine administration (0.05 mg/kg, s.c.). Chronic l‐DOPA significantly elevated the level of Akt phosphorylation at both Thr308 and Ser473 and concentration of phosphorylated GSK3α, whereas pergolide suppressed the lesion‐ and/or challenge‐induced supersensitive Akt responses. The data indicate that l‐DOPA, unlike pergolide, exacerbates imbalances in the Akt pathway caused by the loss of DA. The results support the hypothesis that the Akt pathway is involved in long‐term actions of l‐DOPA and may be linked to l‐DOPA‐induced dyskinesia.


PLOS ONE | 2012

Targeted silencing of elongation factor 2 kinase suppresses growth and sensitizes tumors to doxorubicin in an orthotopic model of breast cancer.

Ibrahim Tekedereli; S. Neslihan Alpay; Clint D. J. Tavares; Zehra E. Cobanoglu; Tamer S. Kaoud; Ibrahim Halil Sahin; Anil K. Sood; Gabriel Lopez-Berestein; Kevin N. Dalby; Bulent Ozpolat

Eukaryotic elongation factor 2 kinase (eEF-2K), through its phosphorylation of elongation factor 2 (eEF2), provides a mechanism by which cells can control the rate of the elongation phase of protein synthesis. The activity of eEF-2K is increased in rapidly proliferating malignant cells, is inhibited during mitosis, and may contribute to the promotion of autophagy in response to anti-cancer therapies. The purpose of this study was to examine the therapeutic potential of targeting eEF-2K in breast cancer tumors. Through the systemic administration of liposomal eEF-2K siRNA (twice a week, i.v. 150 µg/kg), the expression of eEF-2K was down-regulated in vivo in an orthotopic xenograft mouse model of a highly aggressive triple negative MDA-MB-231 tumor. This targeting resulted in a substantial decrease in eEF2 phosphorylation in the tumors, and led to the inhibition of tumor growth, the induction of apoptosis and the sensitization of tumors to the chemotherapy agent doxorubicin. eEF-2K down-modulation in vitro resulted in a decrease in the expression of c-Myc and cyclin D1 with a concomitant increase in the expression of p27Kip1. A decrease in the basal activity of c-Src (phospho-Tyr-416), focal adhesion kinase (phospho-Tyr-397), and Akt (phospho-Ser-473) was also detected following eEF-2K down-regulation in MDA-MB-231 cells, as determined by Western blotting. Where tested, similar results were seen in ER-positive MCF-7 cells. These effects were also accompanied by a decrease in the observed invasive phenotype of the MDA-MB-231 cells. These data support the notion that the disruption of eEF-2K expression in breast cancer cells results in the down-regulation of signaling pathways affecting growth, survival and resistance and has potential as a therapeutic approach for the treatment of breast cancer.


Proteomics | 2011

193‐nm photodissociation of singly and multiply charged peptide anions for acidic proteome characterization

James A. Madsen; Tamer S. Kaoud; Kevin N. Dalby; Jennifer S. Brodbelt

193‐nm ultraviolet photodissociation (UVPD) was implemented to sequence singly and multiply charged peptide anions. Upon dissociation by this method, a‐/x‐type, followed by d and w side‐chain loss ions, were the most prolific and abundant sequence ions, often yielding 100% sequence coverage. The dissociation behavior of singly and multiply charged anions was significantly different with higher charged precursors yielding more sequence ions; however, all charge states investigated (1− through 3−) produced rich diagnostic information. UVPD at 193 nm was also shown to successfully differentiate and pinpoint labile phosphorylation modifications. The sequence ions were produced with high abundances, requiring limited averaging for satisfactory spectral quality. The intact, charge‐reduced radical products generated by UV photoexcitation were also subjected to collision‐induced dissociation (termed, activated‐electron photodetachment dissociation (a‐EPD)), but UVPD alone yielded more predictable and higher abundance sequence ions. With the use of a basic (pH∼11.5), piperidine‐modified mobile phase, LC‐MS/UVPD was implemented and resulted in the successful analysis of mitogen‐activated pathway kinases (MAPKs) using ultrafast activation times (5 ns).


PLOS ONE | 2011

The Effect of Arrestin Conformation on the Recruitment of c-Raf1, MEK1, and ERK1/2 Activation

Sergio Coffa; Maya Breitman; Susan M. Hanson; Kari Callaway; Seunghyi Kook; Kevin N. Dalby; Vsevolod V. Gurevich

Arrestins are multifunctional signaling adaptors originally discovered as proteins that “arrest” G protein activation by G protein-coupled receptors (GPCRs). Recently GPCR complexes with arrestins have been proposed to activate G protein-independent signaling pathways. In particular, arrestin-dependent activation of extracellular signal-regulated kinase 1/2 (ERK1/2) has been demonstrated. Here we have performed in vitro binding assays with pure proteins to demonstrate for the first time that ERK2 directly binds free arrestin-2 and -3, as well as receptor-associated arrestins-1, -2, and -3. In addition, we showed that in COS-7 cells arrestin-2 and -3 association with β2-adrenergic receptor (β2AR) significantly enhanced ERK2 binding, but showed little effect on arrestin interactions with the upstream kinases c-Raf1 and MEK1. Arrestins exist in three conformational states: free, receptor-bound, and microtubule-associated. Using conformationally biased arrestin mutants we found that ERK2 preferentially binds two of these: the “constitutively inactive” arrestin-Δ7 mimicking microtubule-bound state and arrestin-3A, a mimic of the receptor-bound conformation. Both rescue arrestin-mediated ERK1/2/activation in arrestin-2/3 double knockout fibroblasts. We also found that arrestin-2-c-Raf1 interaction is enhanced by receptor binding, whereas arrestin-3-c-Raf1 interaction is not.


Journal of Pharmacology and Experimental Therapeutics | 2008

Haloperidol and Clozapine Differentially Affect the Expression of Arrestins, Receptor Kinases, and Extracellular Signal-Regulated Kinase Activation

Mohamed R. Ahmed; Vsevolod V. Gurevich; Kevin N. Dalby; Jeffrey L. Benovic; Eugenia V. Gurevich

Dopamine and other G protein-coupled receptors (GPCRs) represent the major target of antipsychotic drugs. GPCRs undergo desensitization via activation-dependent phosphorylation by G protein-coupled receptor kinases (GRKs) followed by arrestin binding. Arrestins and GRKs are major regulators of GPCR signaling. We elucidated changes in expression of two arrestins and four GRKs following chronic (21 days) treatment with haloperidol (1 mg/kg i.p.) or clozapine (20 mg/kg i.p.) 2 or 24 h after the last injection in 11 brain regions. Haloperidol decreased GRK3 in ventrolateral caudate-putamen and transiently down-regulated GRK5 in globus pallidus and caudal caudate-putamen. Clozapine also caused a short-term suppression of the GRK5 expression in the caudal caudate-putamen and globus pallidus, but, unlike haloperidol, elevated GRK5 in the caudal caudate-putamen after 24 h. Unlike haloperidol, clozapine decreased arrestin2 and GRK3 in hippocampus and GRK3 in globus pallidus but increased arrestin2 in the core of nucleus accumbens and ventrolateral caudate-putamen and GRK2 in prefrontal cortex. Clozapine, but not haloperidol, induced long-term activation of extracellular signal-regulated kinase (ERK) 2 in ventrolateral caudate-putamen and transient in prefrontal cortex. The data demonstrate that haloperidol and clozapine differentially affect the expression of arrestins and GRKs and ERK activity, which may play a role in determining their clinical profile.


Biochemistry | 2012

Calcium/calmodulin stimulates the autophosphorylation of elongation factor 2 kinase on Thr-348 and Ser-500 to regulate its activity and calcium dependence

Clint D. J. Tavares; John P. O'Brien; Olga Abramczyk; Ashwini K. Devkota; Kevin S. Shores; Scarlett B. Ferguson; Tamer S. Kaoud; Mangalika Warthaka; Kyle D. Marshall; Karin M. Keller; Yan Zhang; Jennifer S. Brodbelt; Bulent Ozpolat; Kevin N. Dalby

Eukaryotic elongation factor 2 kinase (eEF-2K) is an atypical protein kinase regulated by Ca(2+) and calmodulin (CaM). Its only known substrate is eukaryotic elongation factor 2 (eEF-2), whose phosphorylation by eEF-2K impedes global protein synthesis. To date, the mechanism of eEF-2K autophosphorylation has not been fully elucidated. To investigate the mechanism of autophosphorylation, human eEF-2K was coexpressed with λ-phosphatase and purified from bacteria in a three-step protocol using a CaM affinity column. Purified eEF-2K was induced to autophosphorylate by incubation with Ca(2+)/CaM in the presence of MgATP. Analyzing tryptic or chymotryptic peptides by mass spectrometry monitored the autophosphorylation over 0-180 min. The following five major autophosphorylation sites were identified: Thr-348, Thr-353, Ser-445, Ser-474, and Ser-500. In the presence of Ca(2+)/CaM, robust phosphorylation of Thr-348 occurs within seconds of addition of MgATP. Mutagenesis studies suggest that phosphorylation of Thr-348 is required for substrate (eEF-2 or a peptide substrate) phosphorylation, but not self-phosphorylation. Phosphorylation of Ser-500 lags behind the phosphorylation of Thr-348 and is associated with the Ca(2+)-independent activity of eEF-2K. Mutation of Ser-500 to Asp, but not Ala, renders eEF-2K Ca(2+)-independent. Surprisingly, this Ca(2+)-independent activity requires the presence of CaM.

Collaboration


Dive into the Kevin N. Dalby's collaboration.

Top Co-Authors

Avatar

Tamer S. Kaoud

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Pengyu Ren

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Ashwini K. Devkota

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Clint D. J. Tavares

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Eun Jeong Cho

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar

Mangalika Warthaka

University of Texas at Austin

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Chandra Bartholomeusz

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Ranajeet Ghose

City University of New York

View shared research outputs
Top Co-Authors

Avatar

Bulent Ozpolat

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge