Network


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

Hotspot


Dive into the research topics where Madhavi N. L. Nalam is active.

Publication


Featured researches published by Madhavi N. L. Nalam.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Programming peptidomimetic syntheses by translating genetic codes designed de novo

Anthony C. Forster; Zhongping Tan; Madhavi N. L. Nalam; Hening Lin; Hui Qu; Virginia W. Cornish; Stephen C. Blacklow

Although the universal genetic code exhibits only minor variations in nature, Francis Crick proposed in 1955 that “the adaptor hypothesis allows one to construct, in theory, codes of bewildering variety.” The existing code has been expanded to enable incorporation of a variety of unnatural amino acids at one or two nonadjacent sites within a protein by using nonsense or frameshift suppressor aminoacyl-tRNAs (aa-tRNAs) as adaptors. However, the suppressor strategy is inherently limited by compatibility with only a small subset of codons, by the ways such codons can be combined, and by variation in the efficiency of incorporation. Here, by preventing competing reactions with aa-tRNA synthetases, aa-tRNAs, and release factors during translation and by using nonsuppressor aa-tRNA substrates, we realize a potentially generalizable approach for template-encoded polymer synthesis that unmasks the substantially broader versatility of the core translation apparatus as a catalyst. We show that several adjacent, arbitrarily chosen sense codons can be completely reassigned to various unnatural amino acids according to de novo genetic codes by translating mRNAs into specific peptide analog polymers (peptidomimetics). Unnatural aa-tRNA substrates do not uniformly function as well as natural substrates, revealing important recognition elements for the translation apparatus. Genetic programming of peptidomimetic synthesis should facilitate mechanistic studies of translation and may ultimately enable the directed evolution of small molecules with desirable catalytic or pharmacological properties.


Structure | 2010

Crystal Structure of the APOBEC3G Catalytic Domain Reveals Potential Oligomerization Interfaces

Shivender M.D. Shandilya; Madhavi N. L. Nalam; Ellen A. Nalivaika; Phillip J. Gross; Johnathan C. Valesano; Keisuke Shindo; Ming Li; Mary Munson; William E. Royer; Takahide Kono; Hiroshi Matsuo; Reuben S. Harris; Mohan Somasundaran; Celia A. Schiffer

APOBEC3G is a DNA cytidine deaminase that has antiviral activity against HIV-1 and other pathogenic viruses. In this study the crystal structure of the catalytically active C-terminal domain was determined to 2.25 A. This structure corroborates features previously observed in nuclear magnetic resonance (NMR) studies, a bulge in the second beta strand and a lengthening of the second alpha helix. Oligomerization is postulated to be critical for the function of APOBEC3G. In this structure, four extensive intermolecular interfaces are observed, suggesting potential models for APOBEC3G oligomerization. The structural and functional significance of these interfaces was probed by solution NMR and disruptive variants were designed and tested for DNA deaminase and anti-HIV activities. The variant designed to disrupt the most extensive interface lost both activities. NMR solution data provides evidence that another interface, which coordinates a novel zinc site, also exists. Thus, the observed crystallographic interfaces of APOBEC3G may be important for both oligomerization and function.


Journal of the American Chemical Society | 2008

HIV-1 Protease Inhibitors from Inverse Design in the Substrate Envelope Exhibit Subnanomolar Binding to Drug-Resistant Variants

Michael D. Altman; Akbar Ali; G. S. Kiran Kumar Reddy; Madhavi N. L. Nalam; Saima Ghafoor Anjum; Hong Cao; Sripriya Chellappan; Visvaldas Kairys; Miguel X. Fernandes; Michael K. Gilson; Celia A. Schiffer; Tariq M. Rana; Bruce Tidor

The acquisition of drug-resistant mutations by infectious pathogens remains a pressing health concern, and the development of strategies to combat this threat is a priority. Here we have applied a general strategy, inverse design using the substrate envelope, to develop inhibitors of HIV-1 protease. Structure-based computation was used to design inhibitors predicted to stay within a consensus substrate volume in the binding site. Two rounds of design, synthesis, experimental testing, and structural analysis were carried out, resulting in a total of 51 compounds. Improvements in design methodology led to a roughly 1000-fold affinity enhancement to a wild-type protease for the best binders, from a Ki of 30-50 nM in round one to below 100 pM in round two. Crystal structures of a subset of complexes revealed a binding mode similar to each design that respected the substrate envelope in nearly all cases. All four best binders from round one exhibited broad specificity against a clinically relevant panel of drug-resistant HIV-1 protease variants, losing no more than 6-13-fold affinity relative to wild type. Testing a subset of second-round compounds against the panel of resistant variants revealed three classes of inhibitors: robust binders (maximum affinity loss of 14-16-fold), moderate binders (35-80-fold), and susceptible binders (greater than 100-fold). Although for especially high-affinity inhibitors additional factors may also be important, overall, these results suggest that designing inhibitors using the substrate envelope may be a useful strategy in the development of therapeutics with low susceptibility to resistance.


Viruses | 2010

Molecular Basis for Drug Resistance in HIV-1 Protease

Akbar Ali; Rajintha M. Bandaranayake; Yufeng Cai; Nancy M. King; Madhavi Kolli; Seema Mittal; Jennifer F. Murzycki; Madhavi N. L. Nalam; Ellen A. Nalivaika; Ayşegül Özen; Moses Prabu-Jeyabalan; Kelly Thayer; Celia A. Schiffer

HIV-1 protease is one of the major antiviral targets in the treatment of patients infected with HIV-1. The nine FDA approved HIV-1 protease inhibitors were developed with extensive use of structure-based drug design, thus the atomic details of how the inhibitors bind are well characterized. From this structural understanding the molecular basis for drug resistance in HIV-1 protease can be elucidated. Selected mutations in response to therapy and diversity between clades in HIV-1 protease have altered the shape of the active site, potentially altered the dynamics and even altered the sequence of the cleavage sites in the Gag polyprotein. All of these interdependent changes act in synergy to confer drug resistance while simultaneously maintaining the fitness of the virus. New strategies, such as incorporation of the substrate envelope constraint to design robust inhibitors that incorporate details of HIV-1 protease’s function and decrease the probability of drug resistance, are necessary to continue to effectively target this key protein in HIV-1 life cycle.


Journal of Virology | 2010

Evaluating the substrate-envelope hypothesis: structural analysis of novel HIV-1 protease inhibitors designed to be robust against drug resistance.

Madhavi N. L. Nalam; Akbar Ali; Michael D. Altman; G. S. Kiran Kumar Reddy; Sripriya Chellappan; Visvaldas Kairys; Ayşegül Özen; Hong Cao; Michael K. Gilson; Bruce Tidor; Tariq M. Rana; Celia A. Schiffer

ABSTRACT Drug resistance mutations in HIV-1 protease selectively alter inhibitor binding without significantly affecting substrate recognition and cleavage. This alteration in molecular recognition led us to develop the substrate-envelope hypothesis which predicts that HIV-1 protease inhibitors that fit within the overlapping consensus volume of the substrates are less likely to be susceptible to drug-resistant mutations, as a mutation impacting such inhibitors would simultaneously impact the processing of substrates. To evaluate this hypothesis, over 130 HIV-1 protease inhibitors were designed and synthesized using three different approaches with and without substrate-envelope constraints. A subset of 16 representative inhibitors with binding affinities to wild-type protease ranging from 58 nM to 0.8 pM was chosen for crystallographic analysis. The inhibitor-protease complexes revealed that tightly binding inhibitors (at the picomolar level of affinity) appear to “lock” into the protease active site by forming hydrogen bonds to particular active-site residues. Both this hydrogen bonding pattern and subtle variations in protein-ligand van der Waals interactions distinguish nanomolar from picomolar inhibitors. In general, inhibitors that fit within the substrate envelope, regardless of whether they are picomolar or nanomolar, have flatter profiles with respect to drug-resistant protease variants than inhibitors that protrude beyond the substrate envelope; this provides a strong rationale for incorporating substrate-envelope constraints into structure-based design strategies to develop new HIV-1 protease inhibitors.


Chemical Biology & Drug Design | 2007

Design of Mutation‐resistant HIV Protease Inhibitors with the Substrate Envelope Hypothesis

Sripriya Chellappan; G. S. Kiran Kumar Reddy; Akbar Ali; Madhavi N. L. Nalam; Saima Ghafoor Anjum; Hong Cao; Visvaldas Kairys; Miguel X. Fernandes; Michael D. Altman; Bruce Tidor; Tariq M. Rana; Celia A. Schiffer; Michael K. Gilson

There is a clinical need for HIV protease inhibitors that can evade resistance mutations. One possible approach to designing such inhibitors relies upon the crystallographic observation that the substrates of HIV protease occupy a rather constant region within the binding site. In particular, it has been hypothesized that inhibitors which lie within this region will tend to resist clinically relevant mutations. The present study offers the first prospective evaluation of this hypothesis, via computational design of inhibitors predicted to conform to the substrate envelope, followed by synthesis and evaluation against wild‐type and mutant proteases, as well as structural studies of complexes of the designed inhibitors with HIV protease. The results support the utility of the substrate envelope hypothesis as a guide to the design of robust protease inhibitors.


Journal of the American Chemical Society | 2012

Hydrophobic core flexibility modulates enzyme activity in HIV-1 protease.

Seema Mittal; Yufeng Cai; Madhavi N. L. Nalam; Daniel N. Bolon; Celia A. Schiffer

Human immunodeficiency virus Type-1 (HIV-1) protease is crucial for viral maturation and infectivity. Studies of protease dynamics suggest that the rearrangement of the hydrophobic core is essential for enzyme activity. Many mutations in the hydrophobic core are also associated with drug resistance and may modulate the core flexibility. To test the role of flexibility in protease activity, pairs of cysteines were introduced at the interfaces of flexible regions remote from the active site. Disulfide bond formation was confirmed by crystal structures and by alkylation of free cysteines and mass spectrometry. Oxidized and reduced crystal structures of these variants show the overall structure of the protease is retained. However, cross-linking the cysteines led to drastic loss in enzyme activity, which was regained upon reducing the disulfide cross-links. Molecular dynamics simulations showed that altered dynamics propagated throughout the enzyme from the engineered disulfide. Thus, altered flexibility within the hydrophobic core can modulate HIV-1 protease activity, supporting the hypothesis that drug resistant mutations distal from the active site can alter the balance between substrate turnover and inhibitor binding by modulating enzyme activity.


Current Opinion in Hiv and Aids | 2008

New approaches to HIV protease inhibitor drug design II: testing the substrate envelope hypothesis to avoid drug resistance and discover robust inhibitors

Madhavi N. L. Nalam; Celia A. Schiffer

Purpose of reviewDrug resistance results when the balance between the binding of inhibitors and the turnover of substrates is perturbed in favor of the substrates. Resistance is quite widespread to the HIV-1 protease inhibitors permitting the protease to process its 10 different substrates. This processing of the substrates permits the virus HIV-1 to mature and become infectious. The design of HIV-1 protease inhibitors that closely fit within the substrate-binding region is proposed to be a strategy to avoid drug resistance. Recent findingsCocrystal structures of HIV-1 protease with its substrates define an overlapping substrate-binding region or substrate envelope. Novel HIV-1 protease inhibitors that were designed to fit within this substrate envelope were found to retain high binding affinity and have a flat binding profile against a panel of drug-resistant HIV-1 proteases. SummaryThe avoidance of drug resistance needs to be considered in the initial design of inhibitors to quickly evolving targets such as HIV-1 protease. Using a detailed knowledge of substrate binding appears to be a promising strategy for achieving this goal to obtain robust HIV-1 protease inhibitors.


Journal of Virology | 2007

Crystal Structure of Lysine Sulfonamide Inhibitor Reveals the Displacement of the Conserved Flap Water Molecule in Human Immunodeficiency Virus Type 1 Protease

Madhavi N. L. Nalam; Anik Peeters; Tim Hugo Maria Jonckers; Inge Dierynck; Celia A. Schiffer

ABSTRACT Human immunodeficiency virus type 1 (HIV-1) protease has been continuously evolving and developing resistance to all of the protease inhibitors. This requires the development of new inhibitors that bind to the protease in a novel fashion. Most of the inhibitors that are on the market are peptidomimetics, where a conserved water molecule mediates hydrogen bonding interactions between the inhibitors and the flaps of the protease. Recently a new class of inhibitors, lysine sulfonamides, was developed to combat the resistant variants of HIV protease. Here we report the crystal structure of a lysine sulfonamide. This inhibitor binds to the active site of HIV-1 protease in a novel manner, displacing the conserved water and making extensive hydrogen bonds with every region of the active site.


Journal of the American Chemical Society | 2014

Drug Resistance Conferred by Mutations Outside the Active Site through Alterations in the Dynamic and Structural Ensemble of HIV-1 Protease

Debra A. Ragland; Ellen A. Nalivaika; Madhavi N. L. Nalam; Kristina L. Prachanronarong; Hong Cao; Rajintha M. Bandaranayake; Yufeng Cai; Nese Kurt-Yilmaz; Celia A. Schiffer

HIV-1 protease inhibitors are part of the highly active antiretroviral therapy effectively used in the treatment of HIV infection and AIDS. Darunavir (DRV) is the most potent of these inhibitors, soliciting drug resistance only when a complex combination of mutations occur both inside and outside the protease active site. With few exceptions, the role of mutations outside the active site in conferring resistance remains largely elusive. Through a series of DRV–protease complex crystal structures, inhibition assays, and molecular dynamics simulations, we find that single and double site mutations outside the active site often associated with DRV resistance alter the structure and dynamic ensemble of HIV-1 protease active site. These alterations correlate with the observed inhibitor binding affinities for the mutants, and suggest a network hypothesis on how the effect of distal mutations are propagated to pivotal residues at the active site and may contribute to conferring drug resistance.

Collaboration


Dive into the Madhavi N. L. Nalam's collaboration.

Top Co-Authors

Avatar

Celia A. Schiffer

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Akbar Ali

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Hong Cao

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

G. S. Kiran Kumar Reddy

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Bruce Tidor

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Saima Ghafoor Anjum

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Michael D. Altman

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Ellen A. Nalivaika

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Moses Prabu-Jeyabalan

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Nancy M. King

University of Massachusetts Medical School

View shared research outputs
Researchain Logo
Decentralizing Knowledge