Network


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

Hotspot


Dive into the research topics where Srinath Kasturirangan is active.

Publication


Featured researches published by Srinath Kasturirangan.


Journal of Molecular Biology | 2008

Anti-oligomeric Aβ Single-chain Variable Domain Antibody Blocks Aβ-induced Toxicity Against Human Neuroblastoma Cells

Andleeb Zameer; Srinath Kasturirangan; Sharareh Emadi; Sridevi V. Nimmagadda; Michael R. Sierks

The Amyloid-beta (Abeta) peptide is a major component of the amyloid plaques associated with Alzheimers disease (AD). Recent studies suggest that the most toxic forms of Abeta are small, soluble oligomeric aggregates. Here, we report the isolation and characterization of a single-chain variable domain (scFv) antibody isolated against oligomeric Abeta using a protocol developed in our laboratory that combines phage display technology and atomic force microscopy (AFM). Starting with a randomized, single framework phage display library, after three rounds of selection against oligomeric Abeta, we identified an scFv that bound oligomeric Abeta specifically, but not monomeric or fibrillar forms. The anti-oligomeric scFv inhibits Abeta aggregation and toxicity, and reduces the toxicity of preformed oligomeric Abeta towards human neuroblastoma cells. When used to probe samples of human brain tissue, the scFv reacted with AD tissue but not a healthy control or Parkinsons disease brain samples. The anti-oligomeric Abeta scFv therefore has potential therapeutic and diagnostic applications in specifically targeting or identifying the toxic morphologies of Abeta in AD brains.


Journal of Biological Chemistry | 2009

Detecting Morphologically Distinct Oligomeric Forms of α-Synuclein

Sharareh Emadi; Srinath Kasturirangan; Min S. Wang; Philip Schulz; Michael R. Sierks

Neuropathologic and genetics studies as well as transgenic animal models have provided strong evidence linking misfolding and aggregation of α-synuclein to the progression of Parkinson disease (PD) and other related disorders. A growing body of evidence implicates various oligomeric forms of α-synuclein as the toxic species responsible for neurodegeneration and neuronal cell death. Although numerous different oligomeric forms of α-synuclein have been identified in vitro, it is not known which forms are involved in PD or how, when, and where different forms contribute to the progression of PD. Reagents that can interact with specific aggregate forms of α-synuclein would be very useful not only as tools to study how different aggregate forms affect cell function, but also as potential diagnostic and therapeutic agents for PD. Here we show that a single chain antibody fragment (syn-10H scFv) isolated from a phage display antibody library binds to a larger, later stage oligomeric form of α-synuclein than a previously reported oligomeric specific scFv isolated in our laboratory. The scFv described here inhibits aggregation of α-synuclein in vitro, blocks extracellular α-synuclein-induced toxicity in both undifferentiated and differentiated human neuroblastoma cell lines (SH-SY5Y), and specifically recognizes naturally occurring aggregates in PD but not in healthy human brain tissue.


Integrative Biology | 2011

CSF levels of oligomeric alpha-synuclein and beta-amyloid as biomarkers for neurodegenerative disease

Michael R. Sierks; Gaurav Chatterjee; Claire McGraw; Srinath Kasturirangan; Philip Schulz; Shalini Prasad

Protein misfolding and aggregation is a critically important feature in many devastating neurodegenerative diseases, therefore characterization of the CSF concentration profiles of selected key forms and morphologies of proteins involved in these diseases, including β-amyloid (Aβ) and α-synuclein (a-syn), can be an effective diagnostic assay for these diseases. CSF levels of tau and Aβ have been shown to have great promise as biomarkers for Alzheimers disease. However since the onset and progression of many neurodegenerative diseases have been strongly correlated with the presence of soluble oligomeric aggregates of proteins including various Aβ and a-syn aggregate species, specific detection and quantification of levels of each of these different toxic protein species in CSF may provide a simple and accurate means to presymptomatically diagnose and distinguish between these diseases. Here we show that the presence of different protein morphologies in human CSF samples can be readily detected using highly selective morphology specific reagents in conjunction with a sensitive electronic biosensor. We further show that these morphology specific reagents can readily distinguish between post-mortem CSF samples from AD, PD and cognitively normal sources. These studies suggest that detection of specific oligomeric aggregate species holds great promise as sensitive biomarkers for neurodegenerative disease.


Neurobiology of Aging | 2012

Nanobody specific for oligomeric beta-amyloid stabilizes nontoxic form

Srinath Kasturirangan; Lin Li; Sharareh Emadi; Shanta Boddapati; Philip Schulz; Michael R. Sierks

While accumulation and deposition of beta amyloid (Aβ) is a primary pathological feature of Alzheimers disease (AD), increasing evidence has implicated small, soluble oligomeric aggregates of Aβ as the neurotoxic species in AD. Reagents that specifically recognize oligomeric morphologies of Aβ have potential diagnostic and therapeutic value. Using a novel biopanning technique that combines phage display technology and atomic force microscopy, we isolated the nanobody E1 against oligomeric Aβ. Here we show that E1 specifically recognizes a small oligomeric Aβ aggregate species distinct from the species recognized by the A4 nanobody previously reported by our group. While E1, like A4, blocks assembly of Aβ into larger oligomeric and fibrillar forms and prevents any Aβ induced toxicity toward neuronal cells, it does so by binding a small Aβ oligomeric species, directing its assembly toward a stable nontoxic conformation. The E1 nanobody selectively recognizes naturally occurring Aβ aggregates produced in human AD brain tissue indicating that a variety of morphologically distinct Aβ aggregate forms occur naturally and that a stable low-n nontoxic Aβ form exists that does not readily aggregate into larger forms. Because E1 catalyses the formation of a stable nontoxic low-n Aβ species it has potential value as a therapeutic reagent for AD which can be used in combination with other therapeutic approaches.


Biotechnology Progress | 2013

Isolation and characterization of antibody fragments selective for specific protein morphologies from nanogram antigen samples

Srinath Kasturirangan; Tim Reasoner; Philip Schulz; Shanta Boddapati; Sharareh Emadi; Jon Valla; Michael R. Sierks

We developed atomic force microscope (AFM)‐based protocols that enable isolation and characterization of antibody‐based reagents that selectively bind target protein variants using low nanogram amounts or less of unpurified starting material. We isolated single‐chain antibody fragments (scFvs) that specifically recognize an oligomeric beta‐amyloid (Aβ) species correlated with Alzheimers disease (AD) using only a few nanograms of an enriched but not purified sample obtained from human AD brain tissue. We used several subtractive panning steps to remove all phage binding nondesired antigens and then used a single positive panning step using minimal antigen. We also used AFM to characterize the specificity of the isolated clones, again using minimal material, selecting the C6 scFv based on expression levels. We show that C6 selectively binds cell and brain‐derived oligomeric Aβ. The protocols described are readily adapted to isolating antibody‐based reagents against other antigenic targets with limited availability.


Biochemistry | 2010

Engineered Proteolytic Nanobodies Reduce Aβ Burden and Ameliorate Aβ-Induced Cytotoxicity

Srinath Kasturirangan; Shanta Boddapati; Michael R. Sierks

Deposition of beta-amyloid (Abeta) is considered an important early event in the pathogenesis of Alzheimers disease (AD), and reduction of Abeta levels in the brain could be a viable therapeutic approach. A potentially noninflammatory route to facilitate clearance and reduce toxicity of Abeta is to degrade the peptide using proteolytic nanobodies. Here we show that a proteolytic nanobody engineered to cleave Abeta at its alpha-secretase site has potential therapeutic value. The Asec-1A proteolytic nanobody, derived from a parent catalytic light chain antibody, prevents aggregation of monomeric Abeta, inhibits further aggregation of preformed Abeta aggregates, and reduces Abeta-induced cytotoxicity toward a human neuroblastoma cell line. The nanobody also reduces toxicity induced by overexpression of the human amyloid precursor protein (APP) in a Chinese hamster ovary (CHO) cell line by cleaving APP at the alpha-secretase site which precludes formation of Abeta. Targeted proteolysis of APP and Abeta with catalytic nanobodies represents a novel therapeutic approach for treating AD where potentially harmful side effects can be minimized.


Biotechnology Progress | 2009

Promoting α-secretase cleavage of beta-amyloid with engineered proteolytic antibody fragments

Srinath Kasturirangan; Dan Brune; Michael R. Sierks

Deposition of beta‐amyloid (Aβ) is considered as an important early event in the pathogenesis of Alzheimers Disease (AD), and reduction of Aβ levels by various therapeutic approaches is actively being pursued. A potentially non‐inflammatory approach to facilitate clearance and reduce toxicity is to hydrolyze Aβ at its α‐secretase site. We have previously identified a light chain fragment, mk18, with α‐secretase‐like catalytic activity, producing the 1–16 and 17–40 amino acid fragments of Aβ40 as primary products, although hydrolysis is also observed following other lysine and arginine residues. To improve the specific activity of the recombinant antibody by affinity maturation, we constructed a single chain variable fragment (scFv) library containing a randomized CDR3 heavy chain region. A biotinylated covalently reactive analog mimicking α‐secretase site cleavage was synthesized, immobilized on streptavidin beads, and used to select yeast surface expressed scFvs with increased specificity for Aβ. After two rounds of selection against the analog, yeast cells were individually screened for proteolytic activity towards an internally quenched fluorogenic substrate that contains the α‐secretase site of Aβ. From 750 clones screened, the two clones with the highest increase in proteolytic activity compared to the parent mk18 were selected for further study. Kinetic analyses using purified soluble scFvs showed a 3‐ and 6‐fold increase in catalytic activity (kcat/KM) toward the synthetic Aβ substrate compared to the original scFv primarily due to an expected decrease in KM rather than an increase in kcat. This affinity maturation strategy can be used to select for scFvs with increased catalytic specificity for Aβ. These proteolytic scFvs have potential therapeutic applications for AD by decreasing soluble Aβ levels in vivo.


Journal of Alzheimer's Disease | 2012

Bispecific Tandem Single Chain Antibody Simultaneously Inhibits β-Secretase and Promotes α-Secretase Processing of AβPP

Shanta Boddapati; Yona Levites; Vick Suryadi; Srinath Kasturirangan; Michael R. Sierks

Misfolding and aggregation of amyloid-β (Aβ) is an important early event in the pathogenesis of Alzheimers disease. Aβ is produced by sequential proteolysis of the amyloid-β protein precursor (AβPP) by β- and γ-secretases. A third protease, α-secretase, cleaves AβPP in the middle of the Aβ sequence precluding formation of Aβ. The levels of Aβ generated from AβPP can therefore be controlled by tailoring activity of these proteases toward AβPP. We previously showed that β-secretase proteolysis of AβPP could be selectively inhibited using the single chain antibody fragment (scFv) iBSEC1, which blocks the cleavage site on AβPP, and α-secretase proteolysis of AβPP could be selectively enhanced using a proteolytic scFv (Asec1A) engineered to have α-secretase-like activity. Here we show that DIA10D, a novel tandem bispecific scFv combining iBSEC1 with the ASec1A can control amyloidogenic processing of AβPP by simultaneously inhibiting β-secretase and increasing α-secretase processing of AβPP. When expressed in H4 (neuroglioma) cells overexpressing AβPP, DIA10D potently reduces levels of extracellular Aβ by around 50% while also increasing levels of neuroprotective sAβPPα. DIA10D activity has been designed to selectively target AβPP, so this modulation of AβPP processing should not affect endogenous activity of α-and β-secretases towards other substrates.


Alzheimers & Dementia | 2008

P2-335: Supplementing α-secretase cleavage of beta-amyloid by targeted proteolytic antibody fragments using yeast display technology

Srinath Kasturirangan; Mike Sierks

Background: Deposition of beta-amyloid (A ) is considered an important early event in the pathogenesis of Alzheimer’s Disease (AD), and clearance of A represents a potential therapeutic approach. The recombinant antibody light chain fragment, mK18 was identified to have -secretase-like activity, producing the 1-16 and 17-40 amino acid fragments of A 40 as a primary product. In order to improve the targeting specificity of the proteolytic antibody for A , we have constructed a second generation library from a parent single chain antibody fragment (scFv) containing the variable domain of the proteolytic light chain and a random heavy chain. Methods: The scFv library was created by introducing random mutations in the CDR3 region of the heavy chain and subsequently expressing the library on the surface of yeast. Biotinylated covalently reactive analogs based on the -secretase site sequence were synthesized, immobilized on streptavidin beads, and used to select scFvs with increased specificity for A using magnetic bead enrichment. Collected cells were individually screened for proteolytic activity using 96-well plates and an internally quenched fluorogenic substrate that contains the -secretase site of A . Several individual clones with increased proteolytic activity were identified from 750 screened clones. Kinetic parameters were determined and compared. In-vitro studies using Thioflavin-T fluorescence, cytotoxicity using SHSY5Y cells and AFM imaging will be used to determine the effects of the second generation scFv on beta amyloid aggregation. Results: Second generation yeast display library was constructed in yeast. Two clones with highest binding affinity compared to the original clone were identified. Analysis of kinetic parameters (kcat and KM) of purified soluble scFv showed one clone had a 10-fold increase in activity (kcat/KM) toward the synthetic A substrate compared to the original scFv. Conclusions: Results on construction of the library, synthesis of analogs, the panning protocol and catalytic specificity and activity of selected clones will be presented. Catalytic activity toward A oligomers and fibrils and subsequent effects on aggregation and toxicity will be discussed.


Archive | 2011

MORPHOLOGY AND PROTEIN SPECIFIC REAGENTS AS DIAGNOSTICS FOR NEURODEGENERATIVE DISEASES

Michael R. Sierks; Srinath Kasturirangan; Sharareh Emadi; Shalini Prasad

Collaboration


Dive into the Srinath Kasturirangan's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Philip Schulz

Arizona State University

View shared research outputs
Top Co-Authors

Avatar

Sharareh Emadi

Arizona State University

View shared research outputs
Top Co-Authors

Avatar

Shalini Prasad

University of Texas at Dallas

View shared research outputs
Top Co-Authors

Avatar

Claire McGraw

Arizona State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andleeb Zameer

Arizona State University

View shared research outputs
Top Co-Authors

Avatar

Dan Brune

Arizona State University

View shared research outputs
Top Co-Authors

Avatar

Jon Valla

Barrow Neurological Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge