Network


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

Hotspot


Dive into the research topics where Lakshman Bindu is active.

Publication


Featured researches published by Lakshman Bindu.


Journal of Medicinal Chemistry | 2013

Synthesis and biological evaluation of indenoisoquinolines that inhibit both tyrosyl-DNA phosphodiesterase I (Tdp1) and topoisomerase I (Top1).

Martin Conda-Sheridan; P. V. Narasimha Reddy; Andrew Morrell; Brooklyn T. Cobb; Christophe Marchand; Keli Agama; Adel Chergui; Amelie Renaud; Andrew G. Stephen; Lakshman Bindu; Yves Pommier; Mark Cushman

Tyrosyl-DNA phosphodiesterase I (Tdp1) plays a key role in the repair of damaged DNA resulting from the topoisomerase I (Top1) inhibitor camptothecin and a variety of other DNA-damaging anticancer agents. This report documents the design, synthesis, and evaluation of new indenoisoquinolines that are dual inhibitors of both Tdp1 and Top1. Enzyme inhibitory data and cytotoxicity data from human cancer cell cultures were used to establish structure-activity relationships. The potencies of the indenoisoquinolines against Tdp1 ranged from 5 μM to 111 μM, which places the more active compounds among the most potent known inhibitors of this target. The cytotoxicity mean graph midpoints ranged from 0.02 to 2.34 μM. Dual Tdp1-Top1 inhibitors are of interest because the Top1 and Tdp1 inhibitory activities could theoretically work synergistically to create more effective anticancer agents.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Structural basis of recognition of farnesylated and methylated KRAS4b by PDEδ

Srisathiyanarayanan Dharmaiah; Lakshman Bindu; Timothy Tran; William K. Gillette; Peter Frank; Rodolfo Ghirlando; Dwight V. Nissley; Dominic Esposito; Frank McCormick; Andrew G. Stephen; Dhirendra Simanshu

Significance Despite the significant progress made in the last few years toward targeting phosphodiesterase-δ (PDEδ) for KRAS (Kirsten rat sarcoma isoform)-driven cancers, there is no structural information available on posttranslationally modified KRAS4b in complex with PDEδ. The KRAS4b–PDEδ structure reported here provides the structural details of the protein–protein interaction interface and the atomic details of the hypervariable region of KRAS4b. Structural comparison of the two crystal forms allowed identification of a 5-aa-long sequence motif in KRAS4b that could allow PDEδ to bind to both farnesylated and geranylgeranylated KRAS4b. Structural insights obtained from this study could be used to guide the development of improved and more specific inhibitors of the KRAS4b–PDEδ complex. Farnesylation and carboxymethylation of KRAS4b (Kirsten rat sarcoma isoform 4b) are essential for its interaction with the plasma membrane where KRAS-mediated signaling events occur. Phosphodiesterase-δ (PDEδ) binds to KRAS4b and plays an important role in targeting it to cellular membranes. We solved structures of human farnesylated–methylated KRAS4b in complex with PDEδ in two different crystal forms. In these structures, the interaction is driven by the C-terminal amino acids together with the farnesylated and methylated C185 of KRAS4b that binds tightly in the central hydrophobic pocket present in PDEδ. In crystal form II, we see the full-length structure of farnesylated–methylated KRAS4b, including the hypervariable region. Crystal form I reveals structural details of farnesylated–methylated KRAS4b binding to PDEδ, and crystal form II suggests the potential binding mode of geranylgeranylated–methylated KRAS4b to PDEδ. We identified a 5-aa-long sequence motif (Lys-Ser-Lys-Thr-Lys) in KRAS4b that may enable PDEδ to bind both forms of prenylated KRAS4b. Structure and sequence analysis of various prenylated proteins that have been previously tested for binding to PDEδ provides a rationale for why some prenylated proteins, such as KRAS4a, RalA, RalB, and Rac1, do not bind to PDEδ. Comparison of all four available structures of PDEδ complexed with various prenylated proteins/peptides shows the presence of additional interactions due to a larger protein–protein interaction interface in KRAS4b–PDEδ complex. This interface might be exploited for designing an inhibitor with minimal off-target effects.


ChemBioChem | 2010

HER2-and EGFR-specific Affiprobes—Novel Recombinant Optical Probes for Cell Imaging

Ilya Lyakhov; Rafal Zielinski; Monika Kuban; Gabriela Kramer-Marek; Robert J. Fisher; Oleg Chertov; Lakshman Bindu; Jacek Capala

The human epidermal growth factor receptors, EGFR and HER2, are members of the EGFR family of cell‐surface receptors/tyrosine kinases. EGFR‐ and HER2‐positive cancers represent a more aggressive disease with greater likelihood of recurrence, poorer prognosis, and decreased survival rate, compared to EGFR‐ or HER2‐negative cancers. The details of HER2 proto‐oncogenic functions are not deeply understood, partially because of a restricted availability of tools for EGFR and HER2 detection (A. Sorkin and L. K. Goh, Exp. Cell Res. 2009, 315, 683–696). We have created photostable and relatively simple‐to‐produce imaging probes for in vitro staining of EGFR and HER2. These new reagents, called affiprobes, consist of a targeting moiety, a HER2‐ or EGFR‐specific Affibody® molecule, and a fluorescent moiety, mCherry (red) or EGFP (green). Our flow cytometry and confocal microscopy experiments demonstrated high specificity and signal/background ratio of affiprobes. Affiprobes are able to stain both live cells and frozen tumor xenograph sections. This type of optical probe can easily be extended for targeting other cell‐surface antigens/ receptors.


Scientific Reports | 2015

Farnesylated and methylated KRAS4b: high yield production of protein suitable for biophysical studies of prenylated protein-lipid interactions

William K. Gillette; Dominic Esposito; Maria Abreu Blanco; Patrick J. Alexander; Lakshman Bindu; Cammi Bittner; Oleg Chertov; Peter Frank; Carissa Grose; Jane E. Jones; Zhaojing Meng; Shelley Perkins; Que N. Van; Rodolfo Ghirlando; Matthew J. Fivash; Dwight V. Nissley; Frank McCormick; Matthew Holderfield; Andrew G. Stephen

Prenylated proteins play key roles in several human diseases including cancer, atherosclerosis and Alzheimer’s disease. KRAS4b, which is frequently mutated in pancreatic, colon and lung cancers, is processed by farnesylation, proteolytic cleavage and carboxymethylation at the C-terminus. Plasma membrane localization of KRAS4b requires this processing as does KRAS4b-dependent RAF kinase activation. Previous attempts to produce modified KRAS have relied on protein engineering approaches or in vitro farnesylation of bacterially expressed KRAS protein. The proteins produced by these methods do not accurately replicate the mature KRAS protein found in mammalian cells and the protein yield is typically low. We describe a protocol that yields 5–10 mg/L highly purified, farnesylated, and methylated KRAS4b from insect cells. Farnesylated and methylated KRAS4b is fully active in hydrolyzing GTP, binds RAF-RBD on lipid Nanodiscs and interacts with the known farnesyl-binding protein PDEδ.


ChemBioChem | 2005

Ring‐Closing Metathesis of C‐Terminal Allylglycine Residues with an N‐Terminal β‐Vinyl‐Substituted Phosphotyrosyl Mimetic as an Approach to Novel Grb2 SH2 Domain‐Binding Macrocycles

Shinya Oishi; Zhen-Dan Shi; Karen M. Worthy; Lakshman Bindu; Robert J. Fisher; Terrence R. Burke

Ring‐closing metathesis (RCM) of peptides often requires insertion of allylglycines at the intended sites of ring juncture, which can result in the displacement of residues that are needed for biological activity. This type of side‐chain deletion can be avoided by appending β‐vinyl substituents onto the parent residues at the intended sites of ring juncture, thereby effectively converting them into functionalized allylglycine equivalents. Such an approach has been previously applied in modified form to growth‐factor receptor bound 2 (Grb2) SH2 domain‐binding peptides by using an N‐terminal β‐vinyl‐functionalized phosphotyrosyl mimetic and C‐terminal 2‐allyl‐3‐aryl‐1‐propanamides that lacked the α‐carboxyl portion of allylglycine residues. These C‐terminal moieties involved lengthy synthesis and once prepared, required an individual total synthesis of each final macrocycle. Work reported herein significantly enhances the versatility of the original approach through the use of C‐terminal allylglycine amides that can be prepared from commercially available L‐ and D‐allylglycines and suitable amines. This methodology could be generally useful where macrocylization is desired with maintenance of functionality at a site of ring juncture.


Bioorganic & Medicinal Chemistry Letters | 2009

Discovery of thioether-bridged cyclic pentapeptides binding to Grb2-SH2 domain with high affinity

Sheng Jiang; Chenzhong Liao; Lakshman Bindu; Biaolin Yin; Karen W. Worthy; Robert J. Fisher; Terrence R. Burke; Marc C. Nicklaus; Peter P. Roller

Blocking the interaction between phosphotyrosine (pTyr)-containing activated receptors and the Src homology 2 (SH2) domain of the growth factor receptor-bound protein 2 (Grb 2) is considered to be an effective and non-cytotoxic strategy to develop new anti-proliferate agents due to its potential to shut down the Ras activation pathway. In this study, a series of phosphotyrosine containing cyclic pentapeptides were designed and synthesized based upon the phage library derived cyclopeptide, G1TE. A comprehensive SAR study was also carried out to develop potent Grb2-SH2 domain antagonists based upon this novel template. With both the peptidomimetic optimization of the amino acid side-chains and the constraint of the backbone conformation guided by molecular modeling, we developed several potent antagonists with low micromolar range binding affinity, such as cyclic peptide 15 with an K(d)=0.359microM, which is providing a novel template for the development of Grb2-SH2 domain antagonists as potential therapeutics for certain cancers.


Organic and Biomolecular Chemistry | 2007

Utilization of achiral alkenyl amines for the preparation of high affinity Grb2 SH2 domain-binding macrocycles by ring-closing metathesis.

Fa Liu; Karen M. Worthy; Lakshman Bindu; Alessio Giubellino; Donald P. Bottaro; Robert J. Fisher; Terrence R. Burke

A family of previously reported ring-closing metathesis (RCM)-derived macrocycles that exhibit potent Grb2 SH2 domain-binding affinity is characterized by stereoselectively-introduced upper ring junctions that bear bicyclic aryl substituents. However, the synthetic complexity of these macrocycles presents a potential limit to their therapeutic application. Therefore, the current study was undertaken to simplify these macrocycles through the use of achiral 4-pentenylamides as ring-forming components. A series of macrocycles (5a-f) was prepared bearing both open and cyclic constructs at the upper ring junction. The Grb2 SH2 domain-binding affinities of these macrocycles varied, with higher affinities being obtained with cyclo-substituents. The most potent analogue (5d) contained a cyclohexyl group and exhibited Grb2 SH2 domain-binding affinity (K(D) = 1.3 nM) that was nearly equal to the parent macrocycle (2), which bore a stereoselectively-introduced naphthylmethyl substituent at the upper ring junction (K(D) = 0.9 nM). The results of this study advance design considerations that should facilitate the development of Grb2 SH2 domain-binding antagonists.


Journal of Medicinal Chemistry | 2008

Selectivity and Mechanism of Action of a Growth Factor Receptor- Bound Protein 2 Src Homology 2 Domain Binding Antagonist

Alessio Giubellino; Zhen Dan Shi; Lisa M. Miller Jenkins; Karen M. Worthy; Lakshman Bindu; Gagani Athauda; Benedetta Peruzzi; Robert J. Fisher; Ettore Appella; Terrence R. Burke; Donald P. Bottaro

We have shown previously that a potent synthetic antagonist of growth factor receptor-bound protein 2 (Grb2) Src homology 2 (SH2) domain binding (1) blocks growth factor stimulated motility, invasion, and angiogenesis in cultured cell models, as well as tumor metastasis in animals. To characterize the selectivity of 1 for the SH2 domain of Grb2 over other proteins containing similar structural binding motifs, we synthesized a biotinylated derivative (3) that retained high affinity Grb2 SH2 domain binding and potent biological activity. To investigate the selectivity of 1 and 3 for Grb2, the biotinylated antagonist 3 was used to immobilize target proteins from cell extracts for subsequent identification by mass spectrometry. Non-specific binding was identified in parallel using a biotinylated analogue that lacked a single critical binding determinant. The mechanism of action of the antagonist was further characterized by immunoprecipitation, immunoblotting, and light microscopy. This approach to defining protein binding antagonist selectivity and molecular basis of action should be widely applicable in drug development.


Science Signaling | 2018

The bacterial Ras/Rap1 site-specific endopeptidase RRSP cleaves Ras through an atypical mechanism to disrupt Ras-ERK signaling

Marco Biancucci; George Minasov; Avik Banerjee; Alfa Herrera; Patrick J. Woida; Matthew B. Kieffer; Lakshman Bindu; María T. Abreu-Blanco; Wayne F. Anderson; Vadim Gaponenko; Andrew G. Stephen; Matthew Holderfield; Karla J. F. Satchell

A bacterial toxin effector domain disrupts RAS-ERK signaling by cleaving RAS through an unusual mechanism. How Vibrio disrupts RAS signaling Many pathogenic bacteria target the small GTPase Ras because it is critical for signaling pathways that control host cell biology and innate defenses. Highly virulent strains of the opportunistic pathogen Vibrio vulnificus produce a toxin that includes the Ras/Rap1-specific endopeptidase (RRSP) effector domain, which cleaves Ras but has no homology to known proteases. Biancucci et al. solved the crystal structure of RRSP, which showed similarity to a family of hydrolytic enzymes, and identified the catalytic residues required for RRSP to cleave KRAS. Cleavage by RRSP did not release any fragments or bound nucleotides from KRAS but altered the structure of KRAS so that nucleotide exchange was inhibited and it could not bind to its downstream effector RAF. These findings provide insight into the mechanism of an unusual protease and may prove useful for developing new strategies for targeting cancer-associated activated RAS proteins. The Ras–extracellular signal–regulated kinase pathway is critical for controlling cell proliferation, and its aberrant activation drives the growth of various cancers. Because many pathogens produce toxins that inhibit Ras activity, efforts to develop effective Ras inhibitors to treat cancer could be informed by studies of Ras inhibition by pathogens. Vibrio vulnificus causes fatal infections in a manner that depends on multifunctional autoprocessing repeats-in-toxin, a toxin that releases bacterial effector domains into host cells. One such domain is the Ras/Rap1-specific endopeptidase (RRSP), which site-specifically cleaves the Switch I domain of the small GTPases Ras and Rap1. We solved the crystal structure of RRSP and found that its backbone shares a structural fold with the EreA/ChaN-like superfamily of enzymes. Unlike other proteases in this family, RRSP is not a metalloprotease. Through nuclear magnetic resonance analysis and nucleotide exchange assays, we determined that the processing of KRAS by RRSP did not release any fragments or cause KRAS to dissociate from its bound nucleotide but instead only locally affected its structure. However, this structural alteration of KRAS was sufficient to disable guanine nucleotide exchange factor–mediated nucleotide exchange and prevent KRAS from binding to RAF. Thus, RRSP is a bacterial effector that represents a previously unrecognized class of protease that disconnects Ras from its signaling network while inducing limited structural disturbance in its target.


Archive | 2006

Application of Phenylphosphate Mimetics to the Design and Synthesis of Olefin Metathesis-Derived Grb2 SH2 Domain-Binding Macrocycles

Sang-Uk Kang; Zhen-Dan Shi; Rajeshri Kariki; Jason Phan; Karen M. Worthy; Lakshman Bindu; Marc C. Nicklaus; David S. Waugh; Robert J. Fisher; Terrence R. Burke

Introduction The growth factor receptor bound protein 2 (Grb2) is an SH2 domain-containing component of signaling pathways associated with a variety of proliferative diseases. Grb2 SH2 domains preferentially recognize sequences of the form, “pTyr-Xxx-Asn”, with binding occurring in type-I β-bend conformations [1]. Significant research has been devoted to developing Grb2 SH2 domain-binding peptides and peptide mimetics as potential therapeutics. One aspect of these efforts has been directed toward overcoming bioavailability issues raised by the dianionic phenylphosphate moiety of pTyr. Using an open-chain display platform based on the peptide AcpTyr-Ac6c-Asn-[3-(1-naphthyl)propylamide] reported by Novartis Corp [2], we had previously examined a number of monoanionic phosphoryl mimetics that exhibited micromolar to sub-micromolar Grb2 SH2 domain-binding affinities [3]. More recently, we reported macrocyclic variants of the Novartis peptide bearing dianionic phosphoryl replacements that provide low nanomolar to sub-nanomolar binding constants (analogs 1 [4] and 2 [5], X = a and b, Fig. 1). The focus of the current study was to examine phosphoryl mimicking groups bearing monoanionic charge (X = c,d,e and f) or no charge (X = g and h) within a macrocyclic platform (Fig. 1).

Collaboration


Dive into the Lakshman Bindu's collaboration.

Top Co-Authors

Avatar

Robert J. Fisher

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Karen M. Worthy

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Terrence R. Burke

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Marc C. Nicklaus

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrew G. Stephen

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Zhen-Dan Shi

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Rajeshri G. Karki

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Dominic Esposito

Science Applications International Corporation

View shared research outputs
Top Co-Authors

Avatar

Donald P. Bottaro

National Institutes of Health

View shared research outputs
Researchain Logo
Decentralizing Knowledge