Network


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

Hotspot


Dive into the research topics where Sethuramasundaram Pitchiaya is active.

Publication


Featured researches published by Sethuramasundaram Pitchiaya.


Journal of Biological Chemistry | 2009

Purification and Functional Reconstitution of Monomeric μ-Opioid Receptors ALLOSTERIC MODULATION OF AGONIST BINDING BY Gi2

Adam J. Kuszak; Sethuramasundaram Pitchiaya; Jessica P. Anand; Henry I. Mosberg; Nils G. Walter; Roger K. Sunahara

Despite extensive characterization of the μ-opioid receptor (MOR), the biochemical properties of the isolated receptor remain unclear. In light of recent reports, we proposed that the monomeric form of MOR can activate G proteins and be subject to allosteric regulation. A μ-opioid receptor fused to yellow fluorescent protein (YMOR) was constructed and expressed in insect cells. YMOR binds ligands with high affinity, displays agonist-stimulated [35S]guanosine 5′-(γ-thio)triphosphate binding to Gαi, and is allosterically regulated by coupled Gi protein heterotrimer both in insect cell membranes and as purified protein reconstituted into a phospholipid bilayer in the form of high density lipoprotein particles. Single-particle imaging of fluorescently labeled receptor indicates that the reconstituted YMOR is monomeric. Moreover, single-molecule imaging of a Cy3-labeled agonist, [Lys7, Cys8]dermorphin, illustrates a novel method for studying G protein-coupled receptor-ligand binding and suggests that one molecule of agonist binds per monomeric YMOR. Together these data support the notion that oligomerization of the μ-opioid receptor is not required for agonist and antagonist binding and that the monomeric receptor is the minimal functional unit in regard to G protein activation and strong allosteric regulation of agonist binding by G proteins.


Nature Communications | 2013

High-resolution three-dimensional mapping of mRNA export through the nuclear pore

Jiong Ma; Zhen Liu; Nicole Michelotti; Sethuramasundaram Pitchiaya; Ram Veerapaneni; John R. Androsavich; Nils G. Walter; Weidong Yang

The flow of genetic information is regulated by selective nucleocytoplasmic transport of messenger RNA:protein complexes (mRNPs) through the nuclear pore complexes (NPCs) of eukaryotic cells. However, the three-dimensional pathway taken by mRNPs as they transit through the NPC, and the kinetics and selectivity of transport, remain obscure. Here we employ single-molecule fluorescence microscopy with an unprecedented spatiotemporal accuracy of 8 nm and 2 ms to provide new insights into the mechanism of nuclear mRNP export in live human cells. We find that mRNPs exiting the nucleus are decelerated and selected at the centre of the NPC, and adopt a fast-slow-fast diffusion pattern during their brief, ~12 ms interaction with the NPC. A 3D reconstruction of the export route indicates that mRNPs primarily interact with the periphery on the nucleoplasmic side and in the center of the NPC, without entering the central axial conduit utilized for passive diffusion of small molecules, and eventually dissociate on the cytoplasmic side.


Chemical Reviews | 2014

Single Molecule Fluorescence Approaches Shed Light on Intracellular RNAs

Sethuramasundaram Pitchiaya; Laurie A. Heinicke; Thomas C. Custer; Nils G. Walter

The eukaryotic cell is highly complex. Ever since Robert Hooke discovered “cells” in 1665 when training his comparably primitive microscope on a sliver of cork, scientists have aimed to identify and characterize all functional components of the cell. Around the turn of the millennium, the Human Genome Project laid open our entire cellular catalogue, but shockingly discovered that less than 21,000 protein-coding genes – just ~5-times the number of a bacterium such as Escherichia coli – span only ~1.2% of the over 3 billion base pairs of the human genome.1-4 This lack of proteomic inventory initially perplexed the scientific community, but then spurred debates of possible underlying RNA contributions to cellular complexity.5,6 The Encyclopedia Of DNA Elements (ENCODE) project, an international collaborative research effort, was initiated to provide a comprehensive picture of all functional elements within the human genome through unbiased, transcriptome-wide coverage by RNA deep-sequencing (RNA-seq).7 Particularly striking are the discoveries that at least 75% of the genome is transcribed and that by far most of these transcripts do not code for proteins, but rather “non-coding” RNAs (ncRNAs), many of which are still uncharacterized in terms of their structure and function.7,8 Currently, more than 80,000 distinct ncRNAs have been identified in human cells, which reveals an unexpected and exciting RNA landscape in our body (with excerpts highlighted in Figure 1).9 Many RNA elements have been found to originate from overlapping loci, suggesting that similar RNA sequences can be distinctly generated or processed to perform different biological functions.10,11 In an effort to understand the complex functional networks these RNAs are involved in, systems biology approaches are beginning to be implemented. Abetting such holistic approaches are single molecule methods that promise to provide quantitative mechanistic details for individual biomolecules within living cells. Figure 1 Survey of the RNA biology in a eukaryotic cell While RNA-seq has proven powerful for discovering novel cellular RNAs, the approach is limited by the ensemble averaging and loss of spatiotemporal information caused by the isolation of cellular RNA. It thus remains unclear whether, for example, functionally important ncRNAs are expressed in low quantities across all cells of a sample or selectively expressed only in a few cells, which feigns low expression by dilution within the averaged measurement. Single molecule approaches have emerged as an unparalleled means to resolve complex cellular processes that are otherwise masked by such ensemble averaging. The recent implementation of single molecule fluorescence tools to characterize of mRNA expression rates and levels, mRNA and microRNA localization, and ribonucleoprotein complex (RNP) association in living cells, together with the emergence of super-resolution imaging techniques such as PALM and STORM,12 endows single molecule techniques with the potential to broadly dissect the functions and mechanisms of ncRNAs. In this review, we begin with an overview of the different classes of RNAs in eukaryotic cells, in terms of their biogenesis, function and localization (Figure 1). Given the extraordinary amount of literature on these subjects, where appropriate we guide the reader to pertinent reviews for further detail. Next, we summarize recent technical achievements of single molecule fluorescence microscopy in visualizing RNA and RNA-protein complexes in vivo. Finally, we highlight some applications of single molecule tools over the last 15 years that investigate RNA function within cells. Throughout the text, we will promote a vision of uniquely resolving the still shrouded multitude of functional mechanisms of RNAs, especially ncRNAs, through single molecule approaches.


Nature Communications | 2016

The lncRNA landscape of breast cancer reveals a role for DSCAM-AS1 in breast cancer progression

Yashar S. Niknafs; Sumin Han; Teng Ma; Chao Zhang; Kari Wilder-Romans; Matthew K. Iyer; Sethuramasundaram Pitchiaya; Rohit Malik; Yasuyuki Hosono; John R. Prensner; Anton Poliakov; Udit Singhal; Lanbo Xiao; Steven Kregel; Ronald F. Siebenaler; Shuang G. Zhao; Michael Uhl; Alexander Gawronski; Daniel F. Hayes; Lori J. Pierce; Xuhong Cao; Colin Collins; Rolf Backofen; Cenk Sahinalp; James M. Rae; Arul M. Chinnaiyan; Felix Y. Feng

Molecular classification of cancers into subtypes has resulted in an advance in our understanding of tumour biology and treatment response across multiple tumour types. However, to date, cancer profiling has largely focused on protein-coding genes, which comprise <1% of the genome. Here we leverage a compendium of 58,648 long noncoding RNAs (lncRNAs) to subtype 947 breast cancer samples. We show that lncRNA-based profiling categorizes breast tumours by their known molecular subtypes in breast cancer. We identify a cohort of breast cancer-associated and oestrogen-regulated lncRNAs, and investigate the role of the top prioritized oestrogen receptor (ER)-regulated lncRNA, DSCAM-AS1. We demonstrate that DSCAM-AS1 mediates tumour progression and tamoxifen resistance and identify hnRNPL as an interacting protein involved in the mechanism of DSCAM-AS1 action. By highlighting the role of DSCAM-AS1 in breast cancer biology and treatment resistance, this study provides insight into the potential clinical implications of lncRNAs in breast cancer.


EMBO Reports | 2012

Intracellular single molecule microscopy reveals two kinetically distinct pathways for microRNA assembly.

Sethuramasundaram Pitchiaya; John R. Androsavich; Nils G. Walter

MicroRNAs (miRNAs) associate with components of the RNA‐induced silencing complex (RISC) to assemble on mRNA targets and regulate protein expression in higher eukaryotes. Here we describe a method for the intracellular single‐molecule, high‐resolution localization and counting (iSHiRLoC) of miRNAs. Microinjected, singly fluorophore‐labelled, functional miRNAs were tracked within diffusing particles, a majority of which contained single such miRNA molecules. Mobility and mRNA‐dependent assembly changes suggest the existence of two kinetically distinct pathways for miRNA assembly, revealing the dynamic nature of this important gene regulatory pathway. iSHiRLOC achieves an unprecedented resolution in the visualization of functional miRNAs, paving the way to understanding RNA silencing through single‐molecule systems biology.


Science Signaling | 2015

The kinase activity of the Ser/Thr kinase BUB1 promotes TGF-β signaling.

Shyam Nyati; Katrina Schinske-Sebolt; Sethuramasundaram Pitchiaya; Katerina Chekhovskiy; Areeb Chator; Nauman Chaudhry; Joseph Dosch; Marcian E. Van Dort; Sooryanarayana Varambally; Chandan Kumar-Sinha; Mukesh K. Nyati; Dipankar Ray; Nils G. Walter; Hongtao Yu; Brian D. Ross; Alnawaz Rehemtulla

A kinase that controls cell division also promotes the activity of the transforming growth factor–β pathway. Placing BUB1 in the TGF-β Pathway The transforming growth factor–β (TGF-β) pathway regulates cell proliferation and migration, processes involved in development, regeneration, and tumorigenesis. The kinase BUB1, which promotes proper chromosome alignment as cells prepare to divide, also regulates cell proliferation. Nyati et al. connected BUB1 to TGF-β signaling. They found that knocking down BUB1 impaired TGF-β–mediated proliferation of tumor cells—but not by acting at chromosomes. Instead, cytoplasmic BUB1 interacted with TGF-β receptor subunits at the cell surface, promoting the interaction between receptor subunits and between the receptor and downstream signaling proteins. Inhibiting the kinase activity of BUB1 suppressed TGF-β pathway activity in cells in culture and in xenografts. The findings suggest a possible point of crosstalk between the mitotic checkpoint and TGF-β signaling. Transforming growth factor–β (TGF-β) signaling regulates cell proliferation and differentiation, which contributes to development and disease. Upon binding TGF-β, the type I receptor (TGFBRI) binds TGFBRII, leading to the activation of the transcription factors SMAD2 and SMAD3. Using an RNA interference screen of the human kinome and a live-cell reporter for TGFBR activity, we identified the kinase BUB1 (budding uninhibited by benzimidazoles-1) as a key mediator of TGF-β signaling. BUB1 interacted with TGFBRI in the presence of TGF-β and promoted the heterodimerization of TGFBRI and TGFBRII. Additionally, BUB1 interacted with TGFBRII, suggesting the formation of a ternary complex. Knocking down BUB1 prevented the recruitment of SMAD3 to the receptor complex, the phosphorylation of SMAD2 and SMAD3 and their interaction with SMAD4, SMAD-dependent transcription, and TGF-β–mediated changes in cellular phenotype including epithelial-mesenchymal transition (EMT), migration, and invasion. Knockdown of BUB1 also impaired noncanonical TGF-β signaling mediated by the kinases AKT and p38 MAPK (mitogen-activated protein kinase). The ability of BUB1 to promote TGF-β signaling depended on the kinase activity of BUB1. A small-molecule inhibitor of the kinase activity of BUB1 (2OH-BNPP1) and a kinase-deficient mutant of BUB1 suppressed TGF-β signaling and formation of the ternary complex in various normal and cancer cell lines. 2OH-BNPP1 administration to mice bearing lung carcinoma xenografts reduced the amount of phosphorylated SMAD2 in tumor tissue. These findings indicated that BUB1 functions as a kinase in the TGF-β pathway in a role beyond its established function in cell cycle regulation and chromosome cohesion.


Cancer Cell | 2017

Development of Peptidomimetic Inhibitors of the ERG Gene Fusion Product in Prostate Cancer

Xiaoju Wang; Yuanyuan Qiao; Irfan A. Asangani; Bushra Ateeq; Anton Poliakov; Marcin Cieślik; Sethuramasundaram Pitchiaya; Balabhadrapatruni V. S. K. Chakravarthi; Xuhong Cao; Xiaojun Jing; Cynthia X. Wang; Ingrid J. Apel; Rui Wang; Jean Ching Yi Tien; Kristin M. Juckette; Wei Yan; Hui Jiang; Shaomeng Wang; Sooryanarayana Varambally; Arul M. Chinnaiyan

Transcription factors play a key role in the development of diverse cancers, and therapeutically targeting them has remained a challenge. In prostate cancer, the gene encoding the transcription factor ERG is recurrently rearranged and plays a critical role in prostate oncogenesis. Here, we identified a series of peptides that interact specifically with the DNA binding domain of ERG. ERG inhibitory peptides (EIPs) and derived peptidomimetics bound ERG with high affinity and specificity, leading to proteolytic degradation of the ERG protein. The EIPs attenuated ERG-mediated transcription, chromatin recruitment, protein-protein interactions, cell invasion and proliferation, and tumor growth. Thus, peptidomimetic targeting of transcription factor fusion products may provide a promising therapeutic strategy for prostate cancer as well as other malignancies.


Methods | 2013

Dissecting non-coding RNA mechanisms in cellulo by Single-molecule High-Resolution Localization and Counting

Sethuramasundaram Pitchiaya; Vishalakshi Krishnan; Thomas C. Custer; Nils G. Walter

Non-coding RNAs (ncRNAs) recently were discovered to outnumber their protein-coding counterparts, yet their diverse functions are still poorly understood. Here we report on a method for the intracellular Single-molecule High-Resolution Localization and Counting (iSHiRLoC) of microRNAs (miRNAs), a conserved, ubiquitous class of regulatory ncRNAs that controls the expression of over 60% of all mammalian protein coding genes post-transcriptionally, by a mechanism shrouded by seemingly contradictory observations. We present protocols to execute single particle tracking (SPT) and single-molecule counting of functional microinjected, fluorophore-labeled miRNAs and thereby extract diffusion coefficients and molecular stoichiometries of micro-ribonucleoprotein (miRNP) complexes from living and fixed cells, respectively. This probing of miRNAs at the single molecule level sheds new light on the intracellular assembly/disassembly of miRNPs, thus beginning to unravel the dynamic nature of this important gene regulatory pathway and facilitating the development of a parsimonious model for their obscured mechanism of action.


Nature Communications | 2017

DNA damage response inhibition at dysfunctional telomeres by modulation of telomeric DNA damage response RNAs

Francesca Rossiello; Julio Aguado; Sara Sepe; Fabio Iannelli; Quan Nguyen; Sethuramasundaram Pitchiaya; Piero Carninci; Fabrizio d'Adda di Fagagna

The DNA damage response (DDR) is a set of cellular events that follows the generation of DNA damage. Recently, site-specific small non-coding RNAs, also termed DNA damage response RNAs (DDRNAs), have been shown to play a role in DDR signalling and DNA repair. Dysfunctional telomeres activate DDR in ageing, cancer and an increasing number of identified pathological conditions. Here we show that, in mammals, telomere dysfunction induces the transcription of telomeric DDRNAs (tDDRNAs) and their longer precursors from both DNA strands. DDR activation and maintenance at telomeres depend on the biogenesis and functions of tDDRNAs. Their functional inhibition by sequence-specific antisense oligonucleotides allows the unprecedented telomere-specific DDR inactivation in cultured cells and in vivo in mouse tissues. In summary, these results demonstrate that tDDRNAs are induced at dysfunctional telomeres and are necessary for DDR activation and they validate the viability of locus-specific DDR inhibition by targeting DDRNAs.


Nature Cell Biology | 2017

Damage-induced lncRNAs control the DNA damage response through interaction with DDRNAs at individual double-strand breaks

Flavia Michelini; Sethuramasundaram Pitchiaya; Valerio Vitelli; Sheetal Sharma; Ubaldo Gioia; Fabio Pessina; Matteo Cabrini; Yejun Wang; Ilaria Capozzo; Fabio Iannelli; Valentina Matti; Sofia Francia; G. V. Shivashankar; Nils G. Walter; Fabrizio d'Adda di Fagagna

The DNA damage response (DDR) preserves genomic integrity. Small non-coding RNAs termed DDRNAs are generated at DNA double-strand breaks (DSBs) and are critical for DDR activation. Here we show that active DDRNAs specifically localize to their damaged homologous genomic sites in a transcription-dependent manner. Following DNA damage, RNA polymerase II (RNAPII) binds to the MRE11–RAD50–NBS1 complex, is recruited to DSBs and synthesizes damage-induced long non-coding RNAs (dilncRNAs) from and towards DNA ends. DilncRNAs act both as DDRNA precursors and by recruiting DDRNAs through RNA–RNA pairing. Together, dilncRNAs and DDRNAs fuel DDR focus formation and associate with 53BP1. Accordingly, inhibition of RNAPII prevents DDRNA recruitment, DDR activation and DNA repair. Antisense oligonucleotides matching dilncRNAs and DDRNAs impair site-specific DDR focus formation and DNA repair. We propose that DDR signalling sites, in addition to sharing a common pool of proteins, individually host a unique set of site-specific RNAs necessary for DDR activation.

Collaboration


Dive into the Sethuramasundaram Pitchiaya's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xuhong Cao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Rohit Malik

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge