Network


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

Hotspot


Dive into the research topics where Ashish Yeri is active.

Publication


Featured researches published by Ashish Yeri.


Scientific Reports | 2017

Total Extracellular Small RNA Profiles from Plasma, Saliva, and Urine of Healthy Subjects

Ashish Yeri; Amanda Courtright; Rebecca Reiman; Elizabeth Carlson; Taylor Beecroft; Alex Janss; Ashley L. Siniard; Ryan Richholt; Chris Balak; Joel Rozowsky; Robert R. Kitchen; Elizabeth D. Hutchins; Joseph Winarta; Roger McCoy; Matthew Anastasi; Seungchan Kim; Matthew J. Huentelman; Kendall Van Keuren-Jensen

Interest in circulating RNAs for monitoring and diagnosing human health has grown significantly. There are few datasets describing baseline expression levels for total cell-free circulating RNA from healthy control subjects. In this study, total extracellular RNA (exRNA) was isolated and sequenced from 183 plasma samples, 204 urine samples and 46 saliva samples from 55 male college athletes ages 18–25 years. Many participants provided more than one sample, allowing us to investigate variability in an individual’s exRNA expression levels over time. Here we provide a systematic analysis of small exRNAs present in each biofluid, as well as an analysis of exogenous RNAs. The small RNA profile of each biofluid is distinct. We find that a large number of RNA fragments in plasma (63%) and urine (54%) have sequences that are assigned to YRNA and tRNA fragments respectively. Surprisingly, while many miRNAs can be detected, there are few miRNAs that are consistently detected in all samples from a single biofluid, and profiles of miRNA are different for each biofluid. Not unexpectedly, saliva samples have high levels of exogenous sequence that can be traced to bacteria. These data significantly contribute to the current number of sequenced exRNA samples from normal healthy individuals.


RNA Biology | 2017

Two distinct extracellular RNA signatures released by a single cell type identified by microarray and next-generation sequencing.

Cecilia Lässer; Ganesh V. Shelke; Ashish Yeri; Dae-Kyum Kim; Rossella Crescitelli; Stefania Raimondo; Margareta Sjöstrand; Yong Song Gho; Kendall Van Keuren Jensen; Jan Lötvall

ABSTRACT Cells secrete extracellular RNA (exRNA) to their surrounding environment and exRNA has been found in many body fluids such as blood, breast milk and cerebrospinal fluid. However, there are conflicting results regarding the nature of exRNA. Here, we have separated 2 distinct exRNA profiles released by mast cells, here termed high-density (HD) and low-density (LD) exRNA. The exRNA in both fractions was characterized by microarray and next-generation sequencing. Both exRNA fractions contained mRNA and miRNA, and the mRNAs in the LD exRNA correlated closely with the cellular mRNA, whereas the HD mRNA did not. Furthermore, the HD exRNA was enriched in lincRNA, antisense RNA, vault RNA, snoRNA, and snRNA with little or no evidence of full-length 18S and 28S rRNA. The LD exRNA was enriched in mitochondrial rRNA, mitochondrial tRNA, tRNA, piRNA, Y RNA, and full-length 18S and 28S rRNA. The proteomes of the HD and LD exRNA-containing fractions were determined with LC-MS/MS and analyzed with Gene Ontology term finder, which showed that both proteomes were associated with the term extracellular vesicles and electron microscopy suggests that at least a part of the exRNA is associated with exosome-like extracellular vesicles. Additionally, the proteins in the HD fractions tended to be associated with the nucleus and ribosomes, whereas the LD fraction proteome tended to be associated with the mitochondrion. We show that the 2 exRNA signatures released by a single cell type can be separated by floatation on a density gradient. These results show that cells can release multiple types of exRNA with substantial differences in RNA species content. This is important for any future studies determining the nature and function of exRNA released from different cells under different conditions.


Journal of extracellular vesicles | 2015

Meeting report: Discussions and preliminary findings on extracellular RNA measurement methods from laboratories in the NIH Extracellular RNA Communication Consortium

Louise C. Laurent; Asim B. Abdel-Mageed; P. David Adelson; Jorge Arango; Leonora Balaj; Xandra O. Breakefield; Elizabeth Carlson; Bob S. Carter; Blanca Majem; Clark C. Chen; Emanuele Cocucci; Kirsty Danielson; Amanda Courtright; Saumya Das; Zakaria Y. Abd Elmageed; Daniel Enderle; Alan Ezrin; Marc Ferrer; Jane E. Freedman; David J. Galas; Roopali Gandhi; Matthew J. Huentelman; Kendall Van Keuren-Jensen; Yashar Kalani; Yong Hoon Kim; Anna M. Krichevsky; Charles P. Lai; Madhu Lal-Nag; Clara D. Laurent; Trevor R. Leonardo

Extracellular RNAs (exRNAs) have been identified in all tested biofluids and have been associated with a variety of extracellular vesicles, ribonucleoprotein complexes and lipoprotein complexes. Much of the interest in exRNAs lies in the fact that they may serve as signalling molecules between cells, their potential to serve as biomarkers for prediction and diagnosis of disease and the possibility that exRNAs or the extracellular particles that carry them might be used for therapeutic purposes. Among the most significant bottlenecks to progress in this field is the lack of robust and standardized methods for collection and processing of biofluids, separation of different types of exRNA-containing particles and isolation and analysis of exRNAs. The Sample and Assay Standards Working Group of the Extracellular RNA Communication Consortium is a group of laboratories funded by the U.S. National Institutes of Health to develop such methods. In our first joint endeavour, we held a series of conference calls and in-person meetings to survey the methods used among our members, placed them in the context of the current literature and used our findings to identify areas in which the identification of robust methodologies would promote rapid advancements in the exRNA field.


Journal of Physical Chemistry B | 2010

Mutation Screening Based on the Mechanical Properties of DNA Molecules Tethered to a Solid Surface

Ashish Yeri; Lizeng Gao; Di Gao

We report a rapid gene mutation screening method by making use of the mechanical properties of single-strand DNA (ssDNA) tethered to a solid surface. With proper temperature control, ssDNA in solution undergoes intrabase pairing and forms a specific complex 3D structure. By tethering such ssDNA strands to a solid surface, a DNA film can be formed. The mechanical properties of such DNA films, probed by devices such as a quartz crystal resonator, are directly related to the specific structure of the ssDNA which is characteristic of its base sequence and thus can be used as the basis for mutation screening. Using this approach, we have detected a single base mutation among 545 bases in the P53 gene. This result suggests promising potential of employing the mechanical properties of DNA strands for rapid screening of mutations.


Oncotarget | 2016

Isolation, identification, and characterization of novel nanovesicles.

Huang-Ge Zhang; Pengxiao Cao; Yun Teng; Xin Hu; Qilong Wang; Ashish Yeri; Xiaoying Zhuang; Abhilash Samykutty; Jingyao Mu; Zhong-Bin Deng; Lifeng Zhang; James A. Mobley; Jun Yan; Kendall Van Keuren-Jensen; Donald M. Miller

Extracellular microvesicles (EVs) have been recognized for many potential clinical applications including biomarkers for disease diagnosis. In this study, we identified a major population of EVs by simply screening fluid samples with a nanosizer. Unlike other EVs, this extracellular nanovesicle (named HG-NV, HG-NV stands for HomoGenous nanovesicle as well as for Huang-Ge- nanovesicle) can be detected with a nanosizer with minimal in vitro manipulation and are much more homogenous in size (8–12 nm) than other EVs. A simple filtration platform is capable of separating HG-NVs from peripheral blood or cell culture supernatants. In comparison with corresponding exosome profiles, HG-NVs released from both mouse and human breast tumor cells are enriched with RNAs. Tumor derived HG-NVs are more potent in promoting tumor progression than exosomes. In summary, we identified a major subset of EVs as a previously unrecognized nanovesicle. Tumor cell derived HG-NVs promote tumor progression. Molecules predominantly present in breast tumor HG-NVs have been identified and characterized. This discovery may have implications in advancing both microvesicle biology research and clinical management including potential used as a biomarker.


Circulation-heart Failure | 2018

MicroRNAs Associated With Reverse Left Ventricular Remodeling in Humans Identify Pathways of Heart Failure Progression

Ravi V. Shah; Olivia Ziegler; Ashish Yeri; Xiaojun Liu; Venkatesh L. Murthy; Dustin J. Rabideau; Chun Yang Xiao; Kristina Hanspers; Arianna M. Belcher; Michael Tackett; Anthony Rosenzweig; Alexander R. Pico; James L. Januzzi; Saumya Das

Background: Plasma extracellular RNAs have recently garnered interest as biomarkers in heart failure (HF). Most studies in HF focus on single extracellular RNAs related to phenotypes and outcomes, and few describe their functional roles. We hypothesized that clusters of plasma microRNAs (miRNAs) associated with left ventricular (LV) remodeling in human HF would identify novel subsets of genes involved in HF in animal models. Methods and Results: We prospectively measured circulating miRNAs in 64 patients with systolic HF (mean age, 64.8 years; 91% men; median LV ejection fraction, 26%) with serial echocardiography (10 months apart) during medical therapy. We defined LV reverse remodeling as a 15% reduction in LV end-systolic volume index. Using principal components analysis, we identified a component associated with LV reverse remodeling (odds ratio=3.99; P=0.01) that provided risk discrimination for LV reverse remodeling superior to a clinical model (C statistic, 0.58 for a clinical model versus 0.71 for RNA-based model). Using network bioinformatics, we uncovered genes not previously widely described in HF regulated simultaneously by >2 miRNAs. We observed increased myocardial expression of these miRNAs during HF development in animals, with downregulation of target gene expression, suggesting coordinate miRNA–mRNA regulation. Target mRNAs were involved in autophagy, metabolism, and inflammation. Conclusions: Plasma miRNAs associated with LV reverse remodeling in humans are dysregulated in animal HF and target clusters of genes involved in mechanisms implicated in HF. A translational approach integrating human HF, bioinformatics, and model systems may uncover novel pathways involved in HF. CLINICAL TRIAL REGISTRATION: URL: https://www.clinicaltrials.gov. Unique identifier: NCT00351390


American Journal of Physiology-heart and Circulatory Physiology | 2017

Small RNA-seq during acute maximal exercise reveal RNAs involved in vascular inflammation and cardiometabolic health: brief report

Ravi V. Shah; Ashish Yeri; Avash Das; Amanda Courtright-Lim; Olivia Ziegler; Ernest V. Gervino; J. Ocel; Pablo Quintero Pinzon; Luke Wooster; Cole S. Bailey; Lea M. Beaulieu; Jane E. Freedman; Ionita Ghiran; Gregory D. Lewis; Kendall Van Keuren-Jensen; Saumya Das

Exercise improves cardiometabolic and vascular function, although the mechanisms remain unclear. Our objective was to demonstrate the diversity of circulating extracellular RNA (ex-RNA) release during acute exercise in humans and its relevance to exercise-mediated benefits on vascular inflammation. We performed plasma small RNA sequencing in 26 individuals undergoing symptom-limited maximal treadmill exercise, with replication of our top candidate miRNA in a separate cohort of 59 individuals undergoing bicycle ergometry. We found changes in miRNAs and other ex-RNAs with exercise (e.g., Y RNAs and tRNAs) implicated in cardiovascular disease. In two independent cohorts of acute maximal exercise, we identified miR-181b-5p as a key ex-RNA increased in plasma after exercise, with validation in a separate cohort. In a mouse model of acute exercise, we found significant increases in miR-181b-5p expression in skeletal muscle after acute exercise in young (but not older) mice. Previous work revealed a strong role for miR-181b-5p in vascular inflammation in obesity, insulin resistance, sepsis, and cardiovascular disease. We conclude that circulating ex-RNAs were altered in plasma after acute exercise target pathways involved in inflammation, including miR-181b-5p. Further investigation into the role of known (e.g., miRNA) and novel (e.g., Y RNAs) RNAs is warranted to uncover new mechanisms of vascular inflammation on exercise-mediated benefits on health.NEW & NOTEWORTHY How exercise provides benefits to cardiometabolic health remains unclear. We performed RNA sequencing in plasma during exercise to identify the landscape of small noncoding circulating transcriptional changes. Our results suggest a link between inflammation and exercise, providing rich data on circulating noncoding RNAs for future studies by the scientific community.


Journal of extracellular vesicles | 2017

Analysis of extracellular RNA in cerebrospinal fluid

Julie A. Saugstad; Theresa A. Lusardi; Kendall Van Keuren-Jensen; Jay I. Phillips; Babett Lind; Christina A. Harrington; T.J. McFarland; Amanda Courtright; Rebecca Reiman; Ashish Yeri; M. Yashar S. Kalani; P. David Adelson; Jorge Arango; John P. Nolan; Erika Duggan; Karen Messer; Johnny C. Akers; Douglas Galasko; Joseph F. Quinn; Bob S. Carter; Fred H. Hochberg

ABSTRACT We examined the extracellular vesicle (EV) and RNA composition of pooled normal cerebrospinal fluid (CSF) samples and CSF from five major neurological disorders: Alzheimer’s disease (AD), Parkinson’s disease (PD), low-grade glioma (LGG), glioblastoma multiforme (GBM), and subarachnoid haemorrhage (SAH), representing neurodegenerative disease, cancer, and severe acute brain injury. We evaluated: (I) size and quantity of EVs by nanoparticle tracking analysis (NTA) and vesicle flow cytometry (VFC), (II) RNA yield and purity using four RNA isolation kits, (III) replication of RNA yields within and between laboratories, and (IV) composition of total and EV RNAs by reverse transcription–quantitative polymerase chain reaction (RT-qPCR) and RNA sequencing (RNASeq). The CSF contained ~106 EVs/μL by NTA and VFC. Brain tumour and SAH CSF contained more EVs and RNA relative to normal, AD, and PD. RT-qPCR and RNASeq identified disease-related populations of microRNAs and messenger RNAs (mRNAs) relative to normal CSF, in both total and EV fractions. This work presents relevant measures selected to inform the design of subsequent replicative CSF studies. The range of neurological diseases highlights variations in total and EV RNA content due to disease or collection site, revealing critical considerations guiding the selection of appropriate approaches and controls for CSF studies.


JAMA Cardiology | 2017

Association of Multiorgan Computed Tomographic Phenomap With Adverse Cardiovascular Health Outcomes: The Framingham Heart Study

Ravi V. Shah; Ashish Yeri; Venkatesh L. Murthy; Joe Massaro; Ralph B. D’Agostino; Jane E. Freedman; Michelle T. Long; Caroline S. Fox; Saumya Das; Emelia J. Benjamin; Christopher J. O’Donnell; Udo Hoffmann

Importance Increased ability to quantify anatomical phenotypes across multiple organs provides the opportunity to assess their cumulative ability to identify individuals at greatest susceptibility for adverse outcomes. Objective To apply unsupervised machine learning to define the distribution and prognostic importance of computed tomography–based multiorgan phenotypes associated with adverse health outcomes. Design, Setting, and Participants This asymptomatic community-based cohort study included 2924 Framingham Heart Study participants between July 2002 and April 2005 undergoing computed tomographic imaging of the chest and abdomen. Participants are from the offspring and third-generation cohorts. Exposures Eleven computed tomography–based measures of valvular/vascular calcification, adiposity, and muscle attenuation. Main Outcomes and Measures All-cause mortality and cardiovascular disease (myocardial infarction, stroke, or cardiovascular death). Results The median age of the participants was 50 years (interquartile range, 43-60 years), and 1422 (48.6%) were men. Principal component analysis identified 3 major anatomic axes: (1) global calcification (defined by aortic, thoracic, coronary, and valvular calcification); (2) adiposity (defined by pericardial, visceral, hepatic, and intrathoracic fat); and (3) muscle attenuation that explained 65.7% of the population variation. Principal components showed different evolution with age (continuous increase in global calcification, decrease in muscle attenuation, and U-shaped association with adiposity) but similar patterns in men and women. Using unsupervised clustering approaches in the offspring cohort (n = 1150), we identified a cohort (n = 232; 20.2%) with an unfavorable multiorgan phenotype across all 3 anatomic axes as compared with a favorable multiorgan phenotype. Membership in the unfavorable phenotypic cluster was associated with a greater prevalence of cardiovascular disease risk factors and with increased all-cause mortality (hazard ratio, 2.61; 95% CI, 1.74-3.92; P < .001), independent of coronary artery calcium score, visceral adipose tissue, and 10-year global cardiovascular disease Framingham risk, and it provided improvement in metrics of discrimination and reclassification. Conclusions and Relevance This proof-of-concept analysis demonstrates that unsupervised machine learning, in an asymptomatic community cohort, identifies an unfavorable multiorgan phenotype associated with adverse health outcomes, especially in elderly American adults. Future investigations in larger populations are required not only to validate the present results, but also to harness clinical, biochemical, imaging, and genetic markers to increase our understanding of healthy cardiovascular aging.


Methods of Molecular Biology | 2011

Biosensing using nanoelectromechanical systems.

Ashish Yeri; Di Gao

Nanoelectromechanical systems (NEMS) correlate analyte-binding events with the mechanical motions of devices in nanometer scales, which in turn are converted into detectable electrical or optical signals. Biosensors based on NEMS have the potential to achieve ultimate sensitivity down to the single-molecule level, provide rapid and real-time detection signals, be operated with extremely low power consumption, and be mass produced with low cost and high reproducibility. This chapter reviews fundamental concepts in NEMS fabrication, actuation and detection, and device characterization, with examples of using NEMS for sensing DNA, proteins, viruses, and bacteria.

Collaboration


Dive into the Ashish Yeri's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kendall Van Keuren-Jensen

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jane E. Freedman

University of Massachusetts Medical School

View shared research outputs
Top Co-Authors

Avatar

Amanda Courtright

Translational Genomics Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Elizabeth Carlson

Translational Genomics Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge