Network


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

Hotspot


Dive into the research topics where Srinivas Jayanthi is active.

Publication


Featured researches published by Srinivas Jayanthi.


Journal of Biological Chemistry | 2015

Regulation of Structural Dynamics within a Signal Recognition Particle Promotes Binding of Protein Targeting Substrates

Feng Gao; Alicia Kight; Rory Henderson; Srinivas Jayanthi; Parth Patel; Marissa Murchison; Priyanka Sharma; Robyn L. Goforth; Thallapuranam Krishnaswamy Suresh Kumar; Ralph Henry; Colin D. Heyes

Background: Targeting of proteins requires a signal recognition particle (SRP) and multiple protein interactions. Results: We observed a decrease in the structural dynamics of cpSRP43 and an increase in substrate affinity upon its binding to cpSRP54. Conclusion: Changes in domain dynamics induced by cpSRP subunit interactions mediate substrate affinity. Significance: Relating structure and dynamics of SRP proteins allows for a better understanding of vectorial targeting within cells. Protein targeting is critical in all living organisms and involves a signal recognition particle (SRP), an SRP receptor, and a translocase. In co-translational targeting, interactions among these proteins are mediated by the ribosome. In chloroplasts, the light-harvesting chlorophyll-binding protein (LHCP) in the thylakoid membrane is targeted post-translationally without a ribosome. A multidomain chloroplast-specific subunit of the SRP, cpSRP43, is proposed to take on the role of coordinating the sequence of targeting events. Here, we demonstrate that cpSRP43 exhibits significant interdomain dynamics that are reduced upon binding its SRP binding partner, cpSRP54. We showed that the affinity of cpSRP43 for the binding motif of LHCP (L18) increases when cpSRP43 is complexed to the binding motif of cpSRP54 (cpSRP54pep). These results support the conclusion that substrate binding to the chloroplast SRP is modulated by protein structural dynamics in which a major role of cpSRP54 is to improve substrate binding efficiency to the cpSRP.


Journal of Biomolecular Structure & Dynamics | 2014

Identification of antagonists to the vasotocin receptor sub-type 4 (VT4R) involved in stress by molecular modelling and verification using anterior pituitary cells

Srinivas Jayanthi; Seong W. Kang; Daniel Bingham; Brian A. Tessaro; Thallapuranam Krishnaswamy Suresh Kumar; Wayne J. Kuenzel

The vasotocin receptor family is homologous to the mammalian vasopressin G-protein coupled receptor (GPCR) family. The vasotocin receptor 2 (VT2R) and 4 (VT4R) have recently been shown to play important role(s) in the neuroendocrine regulation of stress in birds. A homology-based structural model of VT4R of the domestic chicken, Gallus gallus, was built using the sophisticated SYBYL-X suite. The structure of VT4R built with and without extra- and intracellular unstructured loops showed a seven-helix transmembrane domain, which is a characteristic feature of GPCRs. Several agonists and antagonists were screened by molecular docking to map their potential binding sites on the structure of VT4R. Interestingly, the presence of the N-terminal, intracellular and extracellular loops and C-terminal amino acid sequences emerging from the transmembrane domains during molecular docking appeared to influence the binding interface of the peptide agonists and peptide/non-peptide antagonists on the VT4R. The presence of unstructured loops, however, did not affect the relative binding affinity ranking of the peptide antagonists to VT4R. In general, the natural ligand, arginine vasotocin and the peptide/non-peptide antagonists were observed to be more deeply buried in the receptor. Results of in vitro inhibition experiments, using cultured anterior pituitary cells, showed excellent agreement with the binding affinity of the antagonists predicted by molecular docking. The results of this study provide valuable clues for the rational design of novel pharmaceutical compounds capable of blocking or attenuating the stress response.


Journal of Biological Chemistry | 2016

Novel Molecular Interactions of Acylcarnitines and Fatty Acids with Myoglobin

Sree V. Chintapalli; Srinivas Jayanthi; Prema L. Mallipeddi; Ravikumar Gundampati; Thallapuranam Krishnaswamy Suresh Kumar; Damian B. van Rossum; Andriy Anishkin; Sean H. Adams

Previous research has indicated that long-chain fatty acids can bind myoglobin (Mb) in an oxygen-dependent manner. This suggests that oxy-Mb may play an important role in fuel delivery in Mb-rich muscle fibers (e.g. type I fibers and cardiomyocytes), and raises the possibility that Mb also serves as an acylcarnitine-binding protein. We report for the first time the putative interaction and affinity characteristics for different chain lengths of both fatty acids and acylcarnitines with oxy-Mb using molecular dynamic simulations and isothermal titration calorimetry experiments. We found that short- to medium-chain fatty acids or acylcarnitines (ranging from C2:0 to C10:0) fail to achieve a stable conformation with oxy-Mb. Furthermore, our results indicate that C12:0 is the minimum chain length essential for stable binding of either fatty acids or acylcarnitines with oxy-Mb. Importantly, the empirical lipid binding studies were consistent with structural modeling. These results reveal that: (i) the lipid binding affinity for oxy-Mb increases as the chain length increases (i.e. C12:0 to C18:1), (ii) the binding affinities of acylcarnitines are higher when compared with their respective fatty acid counterparts, and (iii) both fatty acids and acylcarnitines bind to oxy-Mb in 1:1 stoichiometry. Taken together, our results support a model in which oxy-Mb is a novel regulator of long-chain acylcarnitine and fatty acid pools in Mb-rich tissues. This has important implications for physiological fuel management during exercise, and relevance to pathophysiological conditions (e.g. fatty acid oxidation disorders and cardiac ischemia) where long-chain acylcarnitine accumulation is evident.


Protein Expression and Purification | 2014

Efficient production and purification of recombinant human interleukin-12 (IL-12) overexpressed in mammalian cells without affinity tag

Srinivas Jayanthi; Bhanu prasanth Koppolu; Sean G. Smith; Rashmi Jalah; Jenifer Bear; Margherita Rosati; George N. Pavlakis; Barbara K. Felber; David A. Zaharoff; Thallapuranam Krishnaswamy Suresh Kumar

Interleukin-12 is a heterodimeric, pro-inflammatory cytokine that is a key driver of cell-mediated immunity. Clinical interest in IL-12 is significant due to its potent anti-tumor activity and efficacy in controlling certain infectious diseases such as Leishmaniasis and Listeria infection. For clinical applications, the ease of production and purification of IL-12 and the associated cost continues to be a consideration. In this context, we report a simple and effective heparin-affinity based purification of recombinant human IL-12 (hIL-12) from the serum-free supernatants of stable IL-12-transduced HEK293 cells. Fractionation of culture supernatants on heparin Sepharose columns revealed that hIL-12 elutes as a single peak in 500 mM NaCl. Coomassie staining and Western blot analysis showed that hIL-12 eluted in 500 mM NaCl is homogeneous. Purity of hIL-12 was ascertained by RP-HPLC and ESI-MS analysis, and found to be ∼98%. Western blot analysis, using monoclonal antibodies, demonstrated that the crucial inter-subunit disulfide bond linking the p35 and p40 subunits is intact in the purified hIL-12. Results of far UV circular dichroism, steady-state tryptophan fluorescence, and differential scanning calorimetry experiments suggest that purified hIL-12 is in its stable native conformation. Enzyme linked immunosorbent assays (ELISAs) and bioactivity studies demonstrate that hIL-12 is obtained in high yields (0.31±0.05 mg/mL of the culture medium) and is also fully bioactive. Isothermal titration calorimetry data show that IL-12 exhibits a moderate binding affinity (Kd(app)=69±1 μM) to heparin. The purification method described in this study is expected to provide greater impetus for research on the role of heparin in the regulation of the function of IL-12. In addition, the results of this study provide an avenue to obtain high amounts of IL-12 required for structural studies which are aimed at the development of novel IL-12-based therapeutics.


international conference on information science and applications | 2013

Identification of Consensus Glycosaminoglycan Binding Strings in Proteins

Chris Dempewolf; Jacqueline Morris; Meghna Chopra; Srinivas Jayanthi; Thallapuranam Krishnaswamy Suresh Kumar; Wing Ning Li

Heparin is a member of the negatively charged glycosaminoglycan family which is present in the extracellular matrix of all eukaryotic cells. Heparin-binding proteins (HBPs) play an important role in various biological processes like cell signaling, development, and angiogenesis. Interestingly, despite their wide array of functions, very little knowledge exists on the precise structural determinants that govern the specificity of the heparin-protein interaction(s). In this context, a critical survey of the presence of heparin-binding amino acid sequence(s) is a pre-requisite for the development of therapeutic principles against heparin protein-mediated diseases. Many proteins have been shown in vivo to bind to heparin. Based on previous structural studies on heparin-binding proteins (HBPs), nineteen consensus heparin-binding motifs have been identified. The length of these strings varies between five and eight amino acids. These heparin-binding motifs were found to contain a pattern of basic natural amino acids (B= lysine/arginine/histidine) spread between the remaining 17 natural amino acids (X). In the present study, a novel search algorithm has been developed to identify the frequency of occurrence of heparin-binding motifs in 200 proteins which have been experimentally identified to have a strong heparin-binding affinity. The list includes the 23 fibroblast growth factors that bind strongly to heparin, whose sequences were obtained from the UniProt Knowledgebase. Results reveal that the HBPs contain multiple putative heparin-binding motifs. Motifs such as, XBXBX, XBXXBX, and XBXXXBX are most preferred in the heparin-binding proteins. Lysine and arginine are the most preferred basic amino acids (B) in the HBPs. Interestingly, hydrophobic amino acids methionine and isoleucine rarely occur in the heparin-binding motifs. In our opinion, results of our study will enable the design of novel heparin-binding peptides and peptoids which can potentially inhibit heparin mediated pathogenesis. In addition, the algorithm developed in this study can also be of significant use to identify new heparin-binding proteins that have still not been annotated in the human genome databank.


Journal of Biological Chemistry | 2018

Stilbenoid prenyltransferases define key steps in the diversification of peanut phytoalexins

Tianhong Yang; Lingling Fang; Sheri Sanders; Srinivas Jayanthi; Gayathri Rajan; Ram Podicheti; Suresh Kumar Krishnaswamy Thallapuranam; Keithanne Mockaitis; Fabricio Medina-Bolivar

Defense responses of peanut (Arachis hypogaea) to biotic and abiotic stresses include the synthesis of prenylated stilbenoids. Members of this compound class show several protective activities in human disease studies, and the list of potential therapeutic targets continues to expand. Despite their medical and biological importance, the biosynthetic pathways of prenylated stilbenoids remain to be elucidated, and the genes encoding stilbenoid-specific prenyltransferases have yet to be identified in any plant species. In this study, we combined targeted transcriptomic and metabolomic analyses to discover prenyltransferase genes in elicitor-treated peanut hairy root cultures. Transcripts encoding five enzymes were identified, and two of these were functionally characterized in a transient expression system consisting of Agrobacterium-infiltrated leaves of Nicotiana benthamiana. We observed that one of these prenyltransferases, AhR4DT-1, catalyzes a key reaction in the biosynthesis of prenylated stilbenoids, in which resveratrol is prenylated at its C-4 position to form arachidin-2, whereas another, AhR3′DT-1, added the prenyl group to C-3′ of resveratrol. Each of these prenyltransferases was highly specific for stilbenoid substrates, and we confirmed their subcellular location in the plastid by fluorescence microscopy. Structural analysis of the prenylated stilbenoids suggested that these two prenyltransferase activities represent the first committed steps in the biosynthesis of a large number of prenylated stilbenoids and their derivatives in peanut. In summary, we have identified five candidate prenyltransferases in peanut and confirmed that two of them are stilbenoid-specific, advancing our understanding of this specialized enzyme family and shedding critical light onto the biosynthesis of bioactive stilbenoids.


Biomolecular Nmr Assignments | 2017

Backbone and side-chain 1H, 15N, and 13C resonance assignments of a novel Staphylococcal inhibitor of myeloperoxidase

Nicoleta T. Ploscariu; Alvaro I. Herrera; Srinivas Jayanthi; Thallapuranam Krishnaswamy Suresh Kumar; Brian V. Geisbrecht; Om Prakash

The bacterium Staphylococcus aureus produces an array of anti-inflammatory molecules that prevent the innate immune system from recognizing it as a pathogen and clearing it from the host. In the acute phase of inflammation, our immune system relies on neutrophils to clear invading bacteria. Recently, novel classes of secreted proteins from S. aureus, including the Extracellular Adherence Protein (EAP) family (Stapels et al., Proc Natl Acad Sci USA 111:13187–13192, 2014) and the Staphylococcal Peroxidase Inhibitor (SPIN), (unpublished work) have been identified as highly selective inhibitors acting on Neutrophil Serine Proteases (NSPs) and myeloperoxidase (MPO) respectively. SPIN is a protein found only in Staphylococci, with no sequence homology to any known proteins. Solution NMR structural studies of SPIN are therefore expected to provide a deeper understanding of its interaction with MPO. In this study, we report the backbone and side-chain 1H, 15N, and 13C resonance assignments of SPIN. Furthermore, using the chemical shifts of these resonances, we predicted the secondary structure of SPIN in solution via the TALOS-N server. The assignment data has been deposited in the BMRB data bank under Accession No. 27069.


Biochemistry and biophysics reports | 2018

Effect of extension of the heparin binding pocket on the structure, stability, and cell proliferation activity of the human acidic fibroblast growth factor

Julie Eberle Davis; Ravi Kumar Gundampati; Srinivas Jayanthi; Joshua D. Anderson; Abigail Pickhardt; Bhanu prasanth Koppolu; David A. Zaharoff; Thallapuranam Krishnaswamy Suresh Kumar

Acidic human fibroblast growth factor (hFGF1) plays a key role in cell growth and proliferation. Activation of the cell surface FGF receptor is believed to involve the glycosaminoglycan, heparin. However, the exact role of heparin is a subject of considerable debate. In this context, in this study, the correlation between heparin binding affinity and cell proliferation activity of hFGF1 is examined by extending the heparin binding pocket through selective engineering via charge reversal mutations (D82R, D84R and D82R/D84R). Results of biophysical experiments such as intrinsic tryptophan fluorescence and far UV circular dichroism spectroscopy suggest that the gross native structure of hFGF1 is not significantly perturbed by the engineered mutations. However, results of limited trypsin digestion and ANS binding experiments show that the backbone structure of the D82R variant is more flexible than that of the wild type hFGF1. Results of the temperature and urea-induced equilibrium unfolding experiments suggest that the stability of the charge-reversal mutations increases in the presence of heparin. Isothermal titration calorimetry (ITC) data reveal that the heparin binding affinity is significantly increased when the charge on D82 is reversed but not when the negative charge is reversed at both positions D82 and D84 (D82R/D84R). However, despite the increased affinity of D82R for heparin, the cell proliferation activity of the D82R variant is observed to be reduced compared to the wild type hFGF1. The results of this study clearly demonstrate that heparin binding affinity of hFGF1 is not strongly correlated to its cell proliferation activity.


Scientific Reports | 2017

Modulation of Interleukin-12 activity in the presence of heparin

Srinivas Jayanthi; Bhanu prasanth Koppolu; Khue G. Nguyen; Sean G. Smith; Barbara K. Felber; Thallapuranam Krishnaswamy Suresh Kumar; David A. Zaharoff

Glycosaminoglycans (GAGs), especially heparin and heparan sulfate (HS), modulate the functions of numerous cytokines. The aims of this multidisciplinary research were to characterize heparin binding to interleukin-12 (IL-12) and determine the mechanism(s) by which heparin influences IL-12 bioactivity. Heparin and HS were found to bind human IL-12 (hIL-12) with low micromolar affinity and increase hIL-12 bioactivity by more than 6-fold. Conversely, other GAGs did not demonstrate significant binding, nor did their addition affect hIL-12 bioactivity. Biophysical studies demonstrated that heparin induced only minor conformational changes while size-exclusion chromatography and small angle X-ray scattering studies indicated that heparin induced dimerization of hIL-12. Heparin modestly protected hIL-12 from proteolytic degradation, however, this was not a likely mechanism for increased cytokine activity in vitro. Flow cytometry studies revealed that heparin increased the amount of hIL-12 bound to cell surfaces. Heparin also facilitated hIL-12 binding and signaling in cells in which both hIL-12 receptor subunits were functionally deleted. Results of this study demonstrate a new role for heparin in modulating the biological activity of IL-12.


Biotechnology Progress | 2016

Production of an anti-Candida peptide via fed batch and ion exchange chromatography.

Rudra Palash Mukherjee; Robert R. Beitle; Srinivas Jayanthi; T.K.S. Kumar; David S. McNabb

Interest in peptides as diagnostic and therapeutic materials require their manufacture via either a recombinant or synthetic route. This study examined the former, where a recombinant fusion consisting of an antifungal peptide was expressed and isolated from Escherichia coli. Fed batch fermentation with E. coli harboring an arabinose‐inducible plasmid produced the 12 residue anti‐Candida peptide fused to the N‐terminal of Green Fluorescent Protein (GFPUV). The purification of the fusion protein, using ion‐exchange chromatography, was monitored by using the intrinsic fluorescence of GFPUV. The recombinant antifungal peptide was successfully released by cyanogen bromide‐induced cleavage of the fusion protein. The recombinant peptide showed the expected antifungal activity.

Collaboration


Dive into the Srinivas Jayanthi's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ralph Henry

University of Arkansas

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge