Network


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

Hotspot


Dive into the research topics where Sreenivas Kanugula is active.

Publication


Featured researches published by Sreenivas Kanugula.


The EMBO Journal | 2000

Active and alkylated human AGT structures: a novel zinc site, inhibitor and extrahelical base binding.

Douglas S. Daniels; Clifford D. Mol; Andrew S. Arvai; Sreenivas Kanugula; Anthony E. Pegg; John A. Tainer

Human O6‐alkylguanine‐DNA alkyltransferase (AGT), which directly reverses endogenous alkylation at the O6‐position of guanine, confers resistance to alkylation chemotherapies and is therefore an active anticancer drug target. Crystal structures of active human AGT and its biologically and therapeutically relevant methylated and benzylated product complexes reveal an unexpected zinc‐stabilized helical bridge joining a two‐domain α/β structure. An asparagine hinge couples the active site motif to a helix–turn–helix (HTH) motif implicated in DNA binding. The reactive cysteine environment, its position within a groove adjacent to the alkyl‐binding cavity and mutational analyses characterize DNA‐damage recognition and inhibitor specificity, support a structure‐based dealkylation mechanism and suggest a molecular basis for destabilization of the alkylated protein. These results support damaged nucleotide flipping facilitated by an arginine finger within the HTH motif to stabilize the extrahelical O6‐alkylguanine without the protein conformational change originally proposed from the empty Ada structure. Cysteine alkylation sterically shifts the HTH recognition helix to evidently mechanistically couple release of repaired DNA to an opening of the protein fold to promote the biological turnover of the alkylated protein.


Nature | 2009

Flipping of alkylated DNA damage bridges base and nucleotide excision repair

Julie L. Tubbs; Vitaly Latypov; Sreenivas Kanugula; Amna Butt; Manana Melikishvili; Rolf Kraehenbuehl; Oliver Fleck; Andrew S. Marriott; Amanda J. Watson; Barbara Verbeek; Gail McGown; Mary Thorncroft; Mauro Santibanez-Koref; Christopher L. Millington; Andrew S. Arvai; Matthew D Kroeger; Lisa A. Peterson; David M. Williams; Michael Fried; Geoffrey P. Margison; Anthony E. Pegg; John A. Tainer

Alkyltransferase-like proteins (ATLs) share functional motifs with the cancer chemotherapy target O6-alkylguanine-DNA alkyltransferase (AGT) and paradoxically protect cells from the biological effects of DNA alkylation damage, despite lacking the reactive cysteine and alkyltransferase activity of AGT. Here we determine Schizosaccharomyces pombe ATL structures without and with damaged DNA containing the endogenous lesion O6-methylguanine or cigarette-smoke-derived O6-4-(3-pyridyl)-4-oxobutylguanine. These results reveal non-enzymatic DNA nucleotide flipping plus increased DNA distortion and binding pocket size compared to AGT. Our analysis of lesion-binding site conservation identifies new ATLs in sea anemone and ancestral archaea, indicating that ATL interactions are ancestral to present-day repair pathways in all domains of life. Genetic connections to mammalian XPG (also known as ERCC5) and ERCC1 in S. pombe homologues Rad13 and Swi10 and biochemical interactions with Escherichia coli UvrA and UvrC combined with structural results reveal that ATLs sculpt alkylated DNA to create a genetic and structural intersection of base damage processing with nucleotide excision repair.


Journal of Biological Chemistry | 1997

Repair of O6-Benzylguanine by the Escherichia coli Ada and Ogt and the Human O6-Alkylguanine-DNA Alkyltransferases

Karina Goodtzova; Sreenivas Kanugula; Suvarchala Edara; Gary T. Pauly; Robert C. Moschel; Anthony E. Pegg

O6-Methylguanine is removed from DNA via the transfer of the methyl group to a cysteine acceptor site present in the DNA repair protein O6-alkylguanine-DNA alkyltransferase. The human alkyltransferase is inactivated by the free base O6-benzylguanine, raising the possibility that substantially larger alkyl groups could also be accepted as substrates. However, the Escherichia coli alkyltransferase, Ada-C, is not inactivated by O6-benzylguanine. The Ada-C protein was rendered capable of reaction by the incorporation of two site-directed mutations converting Ala316 to a proline (A316P) and Trp336 to alanine (W336A) or glycine (W336G). These changes increase the space at the active site of the protein where Cys321 is buried and thus permit access of the O6-benzylguanine inhibitor. Reaction of the mutant A316P/W336A-Ada-C with O6-benzylguanine was greatly stimulated by the presence of DNA, providing strong support for the concept that binding of DNA to the Ada-C protein activates the protein. The Ada-C protein was able to repair O6-benzylguanine in a 16-mer oligodeoxyribonucleotide. However, the rate of repair was very slow, whereas the E. coli Ogt, the human alkyltransferase, and the mutant A316P/W336A-Ada-C alkyltransferases reacted very rapidly with this 16-mer substrate and preferentially repaired it when incubated with a mixture of the methylated and benzylated 16-mers. These results show that benzyl groups are better substrates than methyl groups for alkyltransferases provided that steric factors do not prevent binding of the substrate in the correct orientation for alkyl group transfer.


Biochemical Pharmacology | 1999

Protection of CHO cells by mutant forms of O6-alkylguanine-DNA alkyltransferase from killing by 1,3-Bis-(2-chloroethyl)-1-nitrosourea (BCNU) plus O6-benzylguanine or O6-benzyl-8-oxoguanine

Natalia A. Loktionova; Meng Xu-Welliver; Tina M. Crone; Sreenivas Kanugula; Anthony E. Pegg

O6-Benzylguanine (BG) is an inactivator of human O6-alkylguanine-DNA alkyltransferase (AGT) currently undergoing clinical trials to enhance cancer chemotherapy by alkylating agents. Mutant forms of AGT resistant to BG in vitro were expressed in CHO cells to determine if they could impart resistance to killing by the combination of BG and 1,3-bis-(2-chloroethyl)-1-nitrosourea (BCNU). All the BG-resistant mutant proteins tested (P140A, P140K, P138M/V139L/P140K, G156A, P140A/G160R, and G160R) showed a reduced rate of reaction with methylated DNA substrates in vitro. However, when expressed in equal amounts in CHO cells, mutants P140A, P140K, P138M/V139L/P140K, and G160R gave levels of protection from the chloroethylating agent BCNU equivalent to that of wild-type AGT. This indicates that a 10-fold reduction in rate constant did not prevent their ability to repair chloroethylated DNA in the cell. AGT activity was readily lost when CHO cells expressing wild-type AGT were exposed to BG or its 8-oxo metabolite (O6-benzyl-8-oxoguanine), but cells expressing mutants P140A or G160R required 30-fold higher concentrations and cells expressing mutants P140K or P138M/V139L/P140K were totally resistant. When cells were treated with 80 microM BCNU plus BG or 8-oxo-BG, those expressing wild-type AGT were killed when inhibitor concentrations of up to 500 microM were used, whereas cells expressing P140K or P138M/V139L/P140K showed no effect, and cells expressing P140A or G160R showed an intermediate resistance. These results suggest that: (i) appearance of BG-resistant mutant AGTs may be a problem during therapy, and (ii) the P140K mutant AGT is an excellent candidate for gene therapy approaches where expression of a BG-resistant AGT in hematopoietic cells is used to reduce toxicity.


Cancer Research | 2006

DNA Sequence Context Affects Repair of the Tobacco-Specific Adduct O6-[4-Oxo-4-(3-pyridyl)butyl]guanine by Human O6-Alkylguanine-DNA Alkyltransferases

Renée S. Mijal; Sreenivas Kanugula; Choua C. Vu; Qingming Fang; Anthony E. Pegg; Lisa A. Peterson

The repair protein O(6)-alkylguanine-DNA alkyltransferase (AGT) protects cells from the mutagenic and carcinogenic effects of alkylating agents by removing O(6)-alkylguanine adducts from DNA. Recently, we established that AGT protects against the mutagenic effects of pyridyloxobutylation resulting from the metabolic activation of the tobacco-specific nitrosamines (TSNA) 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone and N-nitrosonornicotine by repairing O(6)-[4-oxo-4-(3-pyridyl)butyl]guanine (O(6)-pobG). There have been several epidemiologic studies examining the association between the I143V/K178R AGT genotype and lung cancer risk. Two studies have found positive associations, suggesting that AGT proteins differ in their repair of DNA damage caused by TSNA. However, it is not known how this genotype alters the biochemical activity of AGT. We proposed that AGT proteins may differ in their ability to remove large O(6)-alkylguanine adducts, such as O(6)-pobG, from DNA. Therefore, we examined the repair of O(6)-pobG by wild-type (WT) human, I143V/K178R, and L84F AGT proteins when contained in multiple sequence contexts, including the twelfth codon of H-ras, a mutational hotspot within this oncogene. The AGT-mediated repair of O(6)-pobG was more profoundly influenced by sequence context than that of O(6)-methylguanine. These differences are not the result of secondary structure (hairpin) formation in DNA. In addition, the I143V/K178R variant seems less sensitive to the effects of sequence context than the WT or L84F proteins. These studies indicate that the sequence dependence of O(6)-pobG repair by human AGT (hAGT) varies with subtle changes in protein structure. These data establish a novel functional difference between the I143V/K178R protein and other hAGTs in the repair of a toxicologically relevant substrate, O(6)-pobG.


Journal of Biological Chemistry | 2010

Repair of O4-alkylthymine by O6-alkylguanine-DNA alkyltransferases

Qingming Fang; Sreenivas Kanugula; Julie L. Tubbs; John A. Tainer; Anthony E. Pegg

O6-Alkylguanine-DNA alkyltransferase (AGT) plays a major role in repair of the cytotoxic and mutagenic lesion O6-methylguanine (m6G) in DNA. Unlike the Escherichia coli alkyltransferase Ogt that also repairs O4-methylthymine (m4T) efficiently, the human AGT (hAGT) acts poorly on m4T. Here we made several hAGT mutants in which residues near the cysteine acceptor site were replaced by corresponding residues from Ogt to investigate the basis for the inefficiency of hAGT in repair of m4T. Construct hAGT-03 (where hAGT sequence -V149CSSGAVGN157- was replaced with the corresponding Ogt -I143GRNGTMTG151-) exhibited enhanced m4T repair activity in vitro compared with hAGT. Three AGT proteins (hAGT, hAGT-03, and Ogt) exhibited similar protection from killing by N-methyl-N′-nitro-N-nitrosoguanidine and caused a reduction in m6G-induced G:C to A:T mutations in both nucleotide excision repair (NER)-proficient and -deficient Escherichia coli strains that lack endogenous AGTs. hAGT-03 resembled Ogt in totally reducing the m4T-induced T:A to C:G mutations in NER-proficient and -deficient strains. Surprisingly, wild type hAGT expression caused a significant but incomplete decrease in NER-deficient strains but a slight increase in T:A to C:G mutation frequency in NER-proficient strains. The T:A to C:G mutations due to O4-alkylthymine formed by ethylating and propylating agents were also efficiently reduced by either hAGT-03 or Ogt, whereas hAGT had little effect irrespective of NER status. These results show that specific alterations in the hAGT active site facilitate efficient recognition and repair of O4-alkylthymines and reveal damage-dependent interactions of base and nucleotide excision repair.


Journal of Biological Chemistry | 1998

REACTION OF O6-BENZYLGUANINE-RESISTANT MUTANTS OF HUMAN O6-ALKYLGUANINE-DNA ALKYLTRANSFERASE WITH O6-BENZYLGUANINE IN OLIGODEOXYRIBONUCLEOTIDES

Anthony E. Pegg; Sreenivas Kanugula; Suvarchala Edara; Gary T. Pauly; Robert C. Moschel; Karina Goodtzova

Inactivation of the human DNA repair protein,O 6-alkylguanine-DNA alkyltransferase (AGT), byO 6-benzylguanine renders tumor cells susceptible to killing by alkylating agents. AGT mutants resistant toO 6-benzylguanine can be made by converting Pro140 to an alanine (P140A) or Gly156 to an alanine (G156A). These mutations had a much smaller effect on the reaction with O 6-benzylguanine when it was incorporated into a short single-stranded oligodeoxyribonucleotide. Such oligodeoxyribonucleotides could form the basis for the design of improved AGT inhibitors. AGT and mutants P140A and G156A preferentially reacted with O 6-benzylguanine when incubated with a mixture of two 16-mer oligodeoxyribonucleotides, one containingO 6-benzylguanine and the other,O 6-methylguanine. When the 6 amino acids located in positions 159–164 in AGT were replaced by the equivalent sequence from the Escherichia coli Ada-C protein (mutant AGT/6ada) the preference for benzyl repair was eliminated. Further mutation incorporating the P140A change into AGT/6ada giving mutant P140A/6ada led to a protein that resembled Ada-C in preference for the repair of methyl groups, but P140A/6ada did not differ from P140A in reaction with the free base O 6-benzylguanine. Changes in the AGT active site pocket can therefore affect the preference for repair of O 6-benzyl or -methyl groups when present in an oligodeoxyribonucleotide without altering the reaction with free O 6-benzylguanine.


Biochemistry | 2013

DNA-reactive protein monoepoxides induce cell death and mutagenesis in mammalian cells.

Natalia Tretyakova; Erin D. Michaelson-Richie; Teshome B. Gherezghiher; Jamie Kurtz; Xun Ming; Susith Wickramaratne; Melissa Campion; Sreenivas Kanugula; Anthony E. Pegg; Colin Campbell

Although cytotoxic alkylating agents possessing two electrophilic reactive groups are thought to act by cross-linking cellular biomolecules, their exact mechanisms of action have not been established. In cells, these compounds form a mixture of DNA lesions, including nucleobase monoadducts, interstrand and intrastrand cross-links, and DNA-protein cross-links (DPCs). Interstrand DNA-DNA cross-links block replication and transcription by preventing DNA strand separation, contributing to toxicity and mutagenesis. In contrast, potential contributions of drug-induced DPCs are poorly understood. To gain insight into the biological consequences of DPC formation, we generated DNA-reactive protein reagents and examined their toxicity and mutagenesis in mammalian cells. Recombinant human O(6)-alkylguanine DNA alkyltransferase (AGT) protein or its variants (C145A and K125L) were treated with 1,2,3,4-diepoxybutane to yield proteins containing 2-hydroxy-3,4-epoxybutyl groups on cysteine residues. Gel shift and mass spectrometry experiments confirmed that epoxide-functionalized AGT proteins formed covalent DPC but no other types of nucleobase damage when incubated with duplex DNA. Introduction of purified AGT monoepoxides into mammalian cells via electroporation generated AGT-DNA cross-links and induced cell death and mutations at the hypoxanthine-guanine phosphoribosyltransferase gene. Smaller numbers of DPC lesions and reduced levels of cell death were observed when using protein monoepoxides generated from an AGT variant that fails to accumulate in the cell nucleus (K125L), suggesting that nuclear DNA damage is required for toxicity. Taken together, these results indicate that AGT protein monoepoxides produce cytotoxic and mutagenic DPC lesions within chromosomal DNA. More generally, these data suggest that covalent DPC lesions contribute to the cytotoxic and mutagenic effects of bis-electrophiles.


Journal of Biological Chemistry | 2010

Structural Basis of O6-Alkylguanine Recognition by a Bacterial Alkyltransferase-like DNA Repair Protein

James M. Aramini; Julie L. Tubbs; Sreenivas Kanugula; Paolo Rossi; Asli Ertekin; Melissa Maglaqui; Keith Hamilton; Colleen Ciccosanti; Mei Jiang; Rong Xiao; Ta Tsen Soong; Burkhard Rost; Thomas B. Acton; John K. Everett; Anthony E. Pegg; John A. Tainer; Gaetano T. Montelione

Alkyltransferase-like proteins (ATLs) are a novel class of DNA repair proteins related to O6-alkylguanine-DNA alkyltransferases (AGTs) that tightly bind alkylated DNA and shunt the damaged DNA into the nucleotide excision repair pathway. Here, we present the first structure of a bacterial ATL, from Vibrio parahaemolyticus (vpAtl). We demonstrate that vpAtl adopts an AGT-like fold and that the protein is capable of tightly binding to O6-methylguanine-containing DNA and disrupting its repair by human AGT, a hallmark of ATLs. Mutation of highly conserved residues Tyr23 and Arg37 demonstrate their critical roles in a conserved mechanism of ATL binding to alkylated DNA. NMR relaxation data reveal a role for conformational plasticity in the guanine-lesion recognition cavity. Our results provide further evidence for the conserved role of ATLs in this primordial mechanism of DNA repair.


Biochemical Journal | 2000

Conserved residue lysine165 is essential for the ability of O6-alkylguanine-DNA alkyltransferase to react with O6-benzylguanine.

Meng Xu-Welliver; Sreenivas Kanugula; Natalia A. Loktionova; Tina M. Crone; Anthony E. Pegg

The role of lysine(165) in the activity of the DNA repair protein, O(6)-alkylguanine-DNA alkyltransferase (AGT), and the ability of AGT to react with the pseudosubstrate inhibitor, O(6)-benzylguanine (BG), was investigated by changing this lysine to all other 19 possibilities. All of these mutants (except for K165T, which could not be tested as it was too poorly active for assay in crude cell extracts) gave BG-resistant AGTs with increases in the amount of inhibitor needed to produce a 50% loss of activity in a 30 min incubation (ED(50)) from 100-fold (K165A) to 2400-fold (K165F). Lys(165) is a completely conserved residue in AGTs from many species, and all of the mutations at this site also reduced the ability to repair methylated DNA. The least deleterious change was that to arginine, which reduced the rate constant for DNA repair by approx. 2.5-fold. Mutant K165R resembled all of the other mutants in being highly resistant to BG, with an ED(50) value for inactivation by BG>200-fold greater than wild-type. Detailed studies of purified K165A AGT showed that the rate constant for repair and the binding to methylated DNA substrates were reduced by 10-20-fold. Despite this, the K165A mutant AGT was able to protect cells from alkylating agents and this protection was not abolished by BG. These results show that, firstly, lysine at position 165 is needed for optimal activity of AGT towards methylated DNA substrates and is essential for efficient reaction with BG; and second, even if the AGT activity towards methylated DNA substrates is impaired by mutations at codon 165, such mutants can protect tumour cells from therapeutic alkylating agents. These results raise the possibility that the conservation of Lys(165) is due to the need for AGT activity towards substrates containing more bulky adducts than O(6)-methylguanine. They also suggest that alterations at Lys(165) may occur during chemotherapy with BG and alkylating agents and could limit the effectiveness of this therapy.

Collaboration


Dive into the Sreenivas Kanugula's collaboration.

Top Co-Authors

Avatar

Anthony E. Pegg

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Karina Goodtzova

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar

Gary T. Pauly

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Natalia A. Loktionova

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Robert C. Moschel

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Suvarchala Edara

Penn State Milton S. Hershey Medical Center

View shared research outputs
Top Co-Authors

Avatar

John A. Tainer

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Qingming Fang

Pennsylvania State University

View shared research outputs
Top Co-Authors

Avatar

Andrew S. Arvai

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge