Network


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

Hotspot


Dive into the research topics where Insha Ahmad is active.

Publication


Featured researches published by Insha Ahmad.


Journal of Molecular Biology | 2010

An Enhanced System for Unnatural Amino Acid Mutagenesis in E. coli

Travis S. Young; Insha Ahmad; Jun A. Yin; Peter G. Schultz

We report a new vector, pEVOL, for the incorporation of unnatural amino acids into proteins in Escherichia coli using evolved Methanocaldococcus jannaschii aminoacyl-tRNA synthetase(s) (aaRS)/suppressor tRNA pairs. This new system affords higher yields of mutant proteins through the use of both constitutive and inducible promoters to drive the transcription of two copies of the M. jannaschii aaRS gene. Yields were further increased by coupling the dual-aaRS promoter system with a newly optimized suppressor tRNA(CUA)(opt) in a single-vector construct. The optimized suppressor tRNA(CUA)(opt) afforded increased plasmid stability compared with previously reported vectors for unnatural amino acid mutagenesis. To demonstrate the utility of this new system, we introduced 14 mutant aaRS into pEVOL and compared their ability to insert unnatural amino acids in response to three independent amber nonsense codons in sperm whale myoglobin or green fluorescent protein. When cultured in rich media in shake flasks, pEVOL was capable of producing more than 100 mg/L mutant GroEL protein. The versatility, increased yields, and increased stability of the pEVOL vector will further facilitate the expression of proteins with unnatural amino acids.


Nature | 2013

A regenerative approach to the treatment of multiple sclerosis

Vishal Deshmukh; Virginie Tardif; Costas A. Lyssiotis; Chelsea C. Green; Bilal E. Kerman; Hyung Joon Kim; Krishnan Padmanabhan; Jonathan G. Swoboda; Insha Ahmad; Toru Kondo; Fred H. Gage; Argyrios N. Theofilopoulos; Brian R. Lawson; Peter G. Schultz; Luke L. Lairson

Progressive phases of multiple sclerosis are associated with inhibited differentiation of the progenitor cell population that generates the mature oligodendrocytes required for remyelination and disease remission. To identify selective inducers of oligodendrocyte differentiation, we performed an image-based screen for myelin basic protein (MBP) expression using primary rat optic-nerve-derived progenitor cells. Here we show that among the most effective compounds identifed was benztropine, which significantly decreases clinical severity in the experimental autoimmune encephalomyelitis (EAE) model of relapsing-remitting multiple sclerosis when administered alone or in combination with approved immunosuppressive treatments for multiple sclerosis. Evidence from a cuprizone-induced model of demyelination, in vitro and in vivo T-cell assays and EAE adoptive transfer experiments indicated that the observed efficacy of this drug results directly from an enhancement of remyelination rather than immune suppression. Pharmacological studies indicate that benztropine functions by a mechanism that involves direct antagonism of M1 and/or M3 muscarinic receptors. These studies should facilitate the development of effective new therapies for the treatment of multiple sclerosis that complement established immunosuppressive approaches.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Identification of a small molecule with activity against drug-resistant and persistent tuberculosis

Feng Wang; Dhinakaran Sambandan; Rajkumar Halder; Jianing Wang; Sarah M. Batt; Brian Weinrick; Insha Ahmad; Peng-Yu Yang; Yong Zhang; John Kim; Morad Hassani; Stanislav Huszár; Claudia Trefzer; Zhenkun Ma; Takushi Kaneko; Khisi E. Mdluli; Scott G. Franzblau; Arnab K. Chatterjee; Kai Johnson; Katarína Mikušová; Gurdyal S. Besra; Klaus Fütterer; William R. Jacobs; Peter G. Schultz

Significance The global problem of TB has worsened in recent years with the emergence of drug-resistant organisms, and new drugs are clearly needed. In a cell-based high-throughput screen, a small molecule, TCA1, was discovered that has activity against replicating and nonreplicating Mycobacterium tuberculosis. It is also efficacious in acute and chronic rodent models of TB alone or combined with frontline TB drugs. TCA1 functions by a unique mechanism, inhibiting enzymes involved in cell wall and molybdenum cofactor biosynthesis. This discovery represents a significant advance in the search for new agents to treat persistent and drug-resistant TB. A cell-based phenotypic screen for inhibitors of biofilm formation in mycobacteria identified the small molecule TCA1, which has bactericidal activity against both drug-susceptible and -resistant Mycobacterium tuberculosis (Mtb) and sterilizes Mtb in vitro combined with rifampicin or isoniazid. In addition, TCA1 has bactericidal activity against nonreplicating Mtb in vitro and is efficacious in acute and chronic Mtb infection mouse models both alone and combined with rifampicin or isoniazid. Transcriptional analysis revealed that TCA1 down-regulates genes known to be involved in Mtb persistence. Genetic and affinity-based methods identified decaprenyl-phosphoryl-β-D-ribofuranose oxidoreductase DprE1 and MoeW, enzymes involved in cell wall and molybdenum cofactor biosynthesis, respectively, as targets responsible for the activity of TCA1. These in vitro and in vivo results indicate that this compound functions by a unique mechanism and suggest that TCA1 may lead to the development of a class of antituberculosis agents.


Biochemistry | 2011

An Evolved Aminoacyl-tRNA Synthetase with Atypical Polysubstrate Specificity

Douglas D. Young; Travis S. Young; Michael Jahnz; Insha Ahmad; Glen Spraggon; Peter G. Schultz

We have employed a rapid fluorescence-based screen to assess the polyspecificity of several aminoacyl-tRNA synthetases (aaRSs) against an array of unnatural amino acids. We discovered that a p-cyanophenylalanine specific aminoacyl-tRNA synthetase (pCNF-RS) has high substrate permissivity for unnatural amino acids, while maintaining its ability to discriminate against the 20 canonical amino acids. This orthogonal pCNF-RS, together with its cognate amber nonsense suppressor tRNA, is able to selectively incorporate 18 unnatural amino acids into proteins, including trifluoroketone-, alkynyl-, and halogen-substituted amino acids. In an attempt to improve our understanding of this polyspecificity, the X-ray crystal structure of the aaRS-p-cyanophenylalanine complex was determined. A comparison of this structure with those of other mutant aaRSs showed that both binding site size and other more subtle features control substrate polyspecificity.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Evolution of cyclic peptide protease inhibitors

Travis S. Young; Douglas D. Young; Insha Ahmad; John M. Louis; Stephen J. Benkovic; Peter G. Schultz

We report a bacterial system for the evolution of cyclic peptides that makes use of an expanded set of amino acid building blocks. Orthogonal aminoacyl-tRNA synthetase/tRNACUA pairs, together with a split intein system were used to biosynthesize a library of ribosomal peptides containing amino acids with unique structures and reactivities. This peptide library was subsequently used to evolve an inhibitor of HIV protease using a selection based on cellular viability. Two of three cyclic peptides isolated after two rounds of selection contained the keto amino acid p-benzoylphenylalanine (pBzF). The most potent peptide (G12: GIXVSL; X = pBzF) inhibited HIV protease through the formation of a covalent Schiff base adduct of the pBzF residue with the ϵ-amino group of Lys 14 on the protease. This result suggests that an expanded genetic code can confer an evolutionary advantage in response to selective pressure. Moreover, the combination of natural evolutionary processes with chemically biased building blocks provides another strategy for the generation of biologically active peptides using microbial systems.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Somatic hypermutation maintains antibody thermodynamic stability during affinity maturation

Feng Wang; Shiladitya Sen; Yong Zhang; Insha Ahmad; Xueyong Zhu; Ian A. Wilson; Vaughn V. Smider; Thomas J. Magliery; Peter G. Schultz

Somatic hypermutation and clonal selection lead to B cells expressing high-affinity antibodies. Here we show that somatic mutations not only play a critical role in antigen binding, they also affect the thermodynamic stability of the antibody molecule. Somatic mutations directly involved in antigen recognition by antibody 93F3, which binds a relatively small hapten, reduce the melting temperature compared with its germ-line precursor by up to 9 °C. The destabilizing effects of these mutations are compensated by additional somatic mutations located on surface loops distal to the antigen binding site. Similarly, somatic mutations enhance both the affinity and thermodynamic stability of antibody OKT3, which binds the large protein antigen CD3. Analysis of the crystal structures of 93F3 and OKT3 indicates that these somatic mutations modulate antibody stability primarily through the interface of the heavy and light chain variable domains. The historical view of antibody maturation has been that somatic hypermutation and subsequent clonal selection increase antigen–antibody specificity and binding energy. Our results suggest that this process also optimizes protein stability, and that many peripheral mutations that were considered to be neutral are required to offset deleterious effects of mutations that increase affinity. Thus, the immunological evolution of antibodies recapitulates on a much shorter timescale the natural evolution of enzymes in which function and thermodynamic stability are simultaneously enhanced through mutation and selection.


Biochemistry | 2009

Expanding the Genetic Repertoire of the Methylotrophic Yeast Pichia pastoris

Travis S. Young; Insha Ahmad; Ansgar Brock; Peter G. Schultz

To increase the utility of protein mutagenesis with unnatural amino acids, a recombinant expression system in the methylotrophic yeast Pichia pastoris was developed. Aminoacyl-tRNA synthetase/suppressor tRNA (aaRS/tRNA(CUA)) pairs previously evolved in Saccharomyces cerevisiae to be specific for unnatural amino acids were inserted between eukaryotic transcriptional control elements and stably incorporated into the P. pastoris genome. Both the Escherichia coli tyrosyl- and leucyl-RS/tRNA(CUA) pairs were shown to be orthogonal in P. pastoris and used to incorporate eight unnatural amino acids in response to an amber codon with high yields and fidelities. In one example, we show that a recombinant human serum albumin mutant containing a keto amino acid (p-acetylphenylalanine) can be efficiently expressed in this system and selectively conjugated via oxime ligation to a therapeutic peptide mimetic containing an permittivity-(2-(aminooxy)acetyl)-L-lysine residue. Moreover, unnatural amino acid expression in the methylotrophic host was systematically optimized by modulation of aaRS levels to express mutant human serum albumin in excess of 150 mg/L in shake flasks, more than an order of magnitude better than that reported in S. cerevisiae. This methodology should allow the production of high yields of complex proteins containing unnatural amino acids whose expression is not practical in existing systems.


Angewandte Chemie | 2016

Design of Switchable Chimeric Antigen Receptor T Cells Targeting Breast Cancer

Yu Cao; David T. Rodgers; Juanjuan Du; Insha Ahmad; Eric Hampton; Magdalena Mazagova; Seihyun Choi; Hwa Young Yun; Han Xiao; Peng-Yu Yang; Xiaozhou Luo; Reyna K. V. Lim; Holly Pugh; Feng Wang; Stephanie A. Kazane; Timothy M. Wright; Chan Hyuk Kim; Peter G. Schultz; Travis S. Young

Chimeric antigen receptor T (CAR-T) cells have demonstrated promising results against hematological malignancies, but have encountered significant challenges in translation to solid tumors. To overcome these hurdles, we have developed a switchable CAR-T cell platform in which the activity of the engineered cell is controlled by dosage of an antibody-based switch. Herein, we apply this approach to Her2-expressing breast cancers by engineering switch molecules through site-specific incorporation of FITC or grafting of a peptide neo-epitope (PNE) into the anti-Her2 antibody trastuzumab (clone 4D5). We demonstrate that both switch formats can be readily optimized to redirect CAR-T cells (specific for the corresponding FITC or PNE) to Her2-expressing tumor cells, and afford dose-titratable activation of CAR-T cells ex vivo and complete clearance of the tumor in rodent xenograft models. This strategy may facilitate the application of immunotherapy to solid tumors by affording comparable efficacy with improved safety owing to switch-based control of the CAR-T response.


Proceedings of the National Academy of Sciences of the United States of America | 2017

A vimentin binding small molecule leads to mitotic disruption in mesenchymal cancers

Michael Bollong; Mika Pietilä; Aaron D. Pearson; Tapasree Roy Sarkar; Insha Ahmad; Rama Soundararajan; Costas A. Lyssiotis; Sendurai A. Mani; Peter G. Schultz; Luke L. Lairson

Significance Cancer cells derived from mesenchymal tissues or induced to adopt a mesenchymal state have been demonstrated to be largely resistant to standard chemotherapies, necessitating the identification of new effective treatment strategies. From a high throughput screen, we have discovered FiVe1, a compound capable of irreversibly inhibiting the growth of mesenchymally transformed cancer cells by binding to and interfering with the organization and phosphorylation of vimentin (VIM) during mitosis. In contrast to the many naturally and synthetically derived compounds targeting microtubules, we report that chemically targeting an intermediate filament protein, VIM, promotes mitotic catastrophe. As VIM expression is restricted to mesenchymal cells, these results provide a mechanistic basis toward developing genotype-selective chemotherapeutics for the treatment of mesenchymal cancers. Expression of the transcription factor FOXC2 is induced and necessary for successful epithelial–mesenchymal transition, a developmental program that when activated in cancer endows cells with metastatic potential and the properties of stem cells. As such, identifying agents that inhibit the growth of FOXC2-transformed cells represents an attractive approach to inhibit chemotherapy resistance and metastatic dissemination. From a high throughput synthetic lethal screen, we identified a small molecule, FiVe1, which selectively and irreversibly inhibits the growth of mesenchymally transformed breast cancer cells and soft tissue sarcomas of diverse histological subtypes. FiVe1 targets the intermediate filament and mesenchymal marker vimentin (VIM) in a mode which promotes VIM disorganization and phosphorylation during metaphase, ultimately leading to mitotic catastrophe, multinucleation, and the loss of stemness. These findings illustrate a previously undescribed mechanism for interrupting faithful mitotic progression and may ultimately inform the design of therapies for a broad range of mesenchymal cancers.


Nature | 2018

A metabolite-derived protein modification integrates glycolysis with KEAP1–NRF2 signalling

Michael Bollong; Gihoon Lee; John S. Coukos; Hwayoung Yun; Claudio Zambaldo; Jae Won Chang; Emily N. Chin; Insha Ahmad; Arnab K. Chatterjee; Luke L. Lairson; Peter G. Schultz; Raymond E. Moellering

Mechanisms that integrate the metabolic state of a cell with regulatory pathways are necessary to maintain cellular homeostasis. Endogenous, intrinsically reactive metabolites can form functional, covalent modifications on proteins without the aid of enzymes1,2, and regulate cellular functions such as metabolism3–5 and transcription6. An important ‘sensor’ protein that captures specific metabolic information and transforms it into an appropriate response is KEAP1, which contains reactive cysteine residues that collectively act as an electrophile sensor tuned to respond to reactive species resulting from endogenous and xenobiotic molecules. Covalent modification of KEAP1 results in reduced ubiquitination and the accumulation of NRF27,8, which then initiates the transcription of cytoprotective genes at antioxidant-response element loci. Here we identify a small-molecule inhibitor of the glycolytic enzyme PGK1, and reveal a direct link between glycolysis and NRF2 signalling. Inhibition of PGK1 results in accumulation of the reactive metabolite methylglyoxal, which selectively modifies KEAP1 to form a methylimidazole crosslink between proximal cysteine and arginine residues (MICA). This posttranslational modification results in the dimerization of KEAP1, the accumulation of NRF2 and activation of the NRF2 transcriptional program. These results demonstrate the existence of direct inter-pathway communication between glycolysis and the KEAP1–NRF2 transcriptional axis, provide insight into the metabolic regulation of the cellular stress response, and suggest a therapeutic strategy for controlling the cytoprotective antioxidant response in several human diseases.Inhibition of the glycolytic enzyme PGK1 using a small molecular probe reveals a molecular link between glycolysis and the KEAP1–NRF2 signalling cascade.

Collaboration


Dive into the Insha Ahmad's collaboration.

Top Co-Authors

Avatar

Peter G. Schultz

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Peng-Yu Yang

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yong Zhang

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Arnab K. Chatterjee

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Feng Wang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Brian Weinrick

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar

Dhinakaran Sambandan

Albert Einstein College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Jianing Wang

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

John Kim

Albert Einstein College of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge