Network


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

Hotspot


Dive into the research topics where Vivek Kadiyala is active.

Publication


Featured researches published by Vivek Kadiyala.


Journal of Proteome Research | 2012

Proteomic analysis (GeLC-MS/MS) of ePFT-collected pancreatic fluid in chronic pancreatitis.

Joao A. Paulo; Vivek Kadiyala; Linda S. Lee; Peter A. Banks; Darwin L. Conwell; Hanno Steen

Chronic pancreatitis is characterized by inflammation, fibrosis, pain, and loss of exocrine function of the pancreas. We aimed to identify differentially expressed proteins in the ePFT-collected pancreatic fluid from individuals with chronic pancreatitis (CP; n = 9) and controls with chronic abdominal pain not associated with the pancreas (NP; n = 9). Using GeLC-MS/MS techniques, we identified a total of 1391 different proteins in 18 pancreatic fluid samples. Of these proteins, 257 and 413 were identified exclusively in the control and chronic pancreatitis cohorts, respectively, and 721 were identified in both cohorts. Spectral counting and statistical analysis thereof revealed an additional 38 and 77 proteins that were up- or down-regulated, respectively, in the pancreatic fluid from individuals with chronic pancreatitis. As expected, gene ontology analysis illustrated that the largest percentage of differentially regulated proteins was secreted/extracellular in origin. In addition, proteins that were down-regulated with statistical significance in the chronic pancreatitis cohort were determined to have biological function of proteases, corresponding to the canonical pancreatic insufficiency associated with chronic pancreatitis. Proteins enriched in the pancreatic fluid of chronic pancreatitis patients had roles in fibrosis, inflammation, and pain, whereas digestive enzymes were significantly less abundant. Our workflow provided a mass spectrometry-based approach for the further study of the pancreatic fluid proteome, which may lead to the discovery potential biomarkers of chronic pancreatitis.


Clinical and translational gastroenterology | 2014

Investigating MicroRNA Expression Profiles in Pancreatic Cystic Neoplasms

Linda S. Lee; Anna E. Szafranska-Schwarzbach; Dennis Wylie; Leona A. Doyle; Andrew M. Bellizzi; Vivek Kadiyala; Shadeah Suleiman; Peter A. Banks; Bernard F. Andruss; Darwin L. Conwell

OBJECTIVES:Current diagnostic tools for pancreatic cysts fail to reliably differentiate mucinous from nonmucinous cysts. Reliable biomarkers are needed. MicroRNAs (miRNA) may offer insights into pancreatic cysts. Our aims were to (1) identify miRNAs that distinguish benign from both premalignant cysts and malignant pancreatic lesions using formalin-fixed, paraffin-embedded (FFPE) pathology specimens; (2) identify miRNAs that distinguish mucinous cystic neoplasm (MCN) from branch duct-intraductal papillary mucinous neoplasm (BD-IPMN).METHODS:A total of 69 FFPE pancreatic specimens were identified: (1) benign (20 serous cystadenoma (SCA)), (2) premalignant (10 MCN, 10 BD-IPMN, 10 main duct IPMN (MD-IPMN)), and (3) malignant (19 pancreatic ductal adenocarcinoma (PDAC)). Total nucleic acid extraction was performed followed by miRNA expression profiling of 378 miRNAs interrogated using TaqMan MicroRNA Arrays Pool A and verification of candidate miRNAs. Bioinformatics was used to generate classifiers.RESULTS:MiRNA profiling of 69 FFPE specimens yielded 35 differentially expressed miRNA candidates. Four different 4-miRNA panels differentiated among the lesions: one panel separated SCA from MCN, BD-IPMN, MD-IPMN, and PDAC with sensitivity 85% (62, 97), specificity 100% (93, 100), a second panel distinguished MCN from SCA, BD-IPMN, MD-IPMN, and PDAC with sensitivity and specificity 100% (100, 100), a third panel differentiated PDAC from IPMN with sensitivity 95% (76, 100) and specificity 85% (72, 96), and the final panel diagnosed MCN from BD-IPMN with sensitivity and specificity approaching 100%.CONCLUSIONS:MiRNA profiling of surgical pathology specimens differentiates serous cystadenoma from both premalignant pancreatic cystic neoplasms and PDAC and MCN from BD-IPMN.


Proteomics | 2013

Cross‐species analysis of nicotine‐induced proteomic alterations in pancreatic cells

Joao A. Paulo; Raul Urrutia; Vivek Kadiyala; Peter A. Banks; Darwin L. Conwell; Hanno Steen

Toxic compounds in tobacco, such as nicotine, may adversely affect pancreatic function. We aim to determine nicotine‐induced protein alterations in pancreatic cells, thereby revealing links between nicotine exposure and pancreatic disease. We compared the proteomic alterations induced by nicotine treatment in cultured pancreatic cells (mouse, rat, and human stellate cells and human duct cells) using MS‐based techniques, specifically SDS‐PAGE (gel) coupled with LC‐MS/MS and spectral counting. We identified thousands of proteins in pancreatic cells, hundreds of which were identified exclusively or in higher abundance in either nicotine‐treated or untreated cells. Interspecies comparisons of stellate cell proteins revealed several differentially abundant proteins (in nicotine treated versus untreated cells) common among the three species. Proteins appearing in all nicotine‐treated stellate cells include amyloid beta (A4), procollagen type VI alpha 1, integral membrane protein 2B, and toll‐interacting protein. Proteins that were differentially expressed upon nicotine treatment across cell lines were enriched in certain pathways, including nicotinic acetylcholine receptor, cytokine, and integrin signaling. At this analytical depth, we conclude that similar pathways are affected by nicotine, but alterations at the protein level among stellate cells of different species vary. Further interrogation of such pathways will lead to insights into the potential effect of nicotine on pancreatic cells at the biomolecular level and the extension of this concept to the effect of nicotine on pancreatic disease.


Gastroenterology Research and Practice | 2012

Interdisciplinary Management of Cystic Neoplasms of the Pancreas

Linda S. Lee; Thomas E. Clancy; Vivek Kadiyala; Shadeah Suleiman; Darwin L. Conwell

Cystic neoplasms of the pancreas are increasingly recognized due to the frequent use of abdominal imaging. It is reported that up to 20% of abdominal cross-sectional scans identify incidental asymptomatic pancreatic cysts. Proper characterization of pancreatic cystic neoplasms is important not only to recognize premalignant lesions that will require surgical resection, but also to allow nonoperative management of many cystic lesions that will not require resection with its inherent morbidity. Though reliable biomarkers are lacking, a wide spectrum of diagnostic modalities are available to evaluate pancreatic cystic neoplasms, including radiologic, endoscopic, laboratory, and pathologic analysis. An interdisciplinary approach to management of these lesions which incorporates recent, specialty-specific advances in the medical literature is herein suggested.


Journal of Immunological Methods | 2012

Inflammatory protein profiling of pancreatic cyst fluid using EUS-FNA in tandem with cytokine microarray differentiates between branch duct IPMN and inflammatory cysts

Linda S. Lee; Peter A. Banks; Andrew M. Bellizzi; Nisha I. Sainani; Vivek Kadiyala; Shadeah Suleiman; Darwin L. Conwell; Joao A. Paulo

BACKGROUND Diagnosis of pancreatic cystic neoplasms remains problematic. We hypothesize that inflammatory mediator proteins in pancreatic cyst fluid can differentiate branch duct intraductal papillary mucinous neoplasms (BD-IPMNs) and pancreatic inflammatory cysts. We aim to 1) detect inflammatory mediator proteins (IMPs) using a multiplexed IMP-targeted microarray in pancreatic cyst fluid obtained during endoscopic ultrasound fine needle aspiration (EUS-FNA) and 2) compare IMP profiles in pancreatic cyst fluid from BD-IPMNs and inflammatory cysts. Pancreatic cyst fluid from ten patients (5 BD-IPMN and 5 inflammatory cysts) was obtained by EUS-FNA and analyzed directly with a multiplexed microarray assay to determine concentrations of 89 IMPs. Statistical analysis was performed using non-parametric methods. RESULTS Eighty-three of the 89 assayed IMPs were detected in at least one of the 10 patient samples. Seven IMPs were detected in BD-IPMN but not inflammatory cysts, while eleven IMPs were identified in inflammatory cysts but not BD-IPMN. Notably, granulocyte-macrophage colony-stimulating factor (GM-CSF) expression was present in all five inflammatory cyst samples. Hepatocyte growth factor (HGF) was present in significantly higher concentrations in inflammatory cysts compared to BD-IPMN. CONCLUSION Our exploratory analysis reveals that GM-CSF and HGF in EUS-FNA-collected pancreatic cyst fluid can distinguish between BD-IPMN and inflammatory cyst. Coupling microarray molecular techniques to EUS-FNA may represent a major step forward to our understanding complex pancreatic disease.


Genomics, Proteomics & Bioinformatics | 2013

Mass spectrometry-based quantitative proteomic profiling of human pancreatic and hepatic stellate cell lines.

Joao A. Paulo; Vivek Kadiyala; Peter A. Banks; Darwin L. Conwell; Hanno Steen

The functions of the liver and the pancreas differ; however, chronic inflammation in both organs is associated with fibrosis. Evidence suggests that fibrosis in both organs is partially regulated by organ-specific stellate cells. We explore the proteome of human hepatic stellate cells (hHSC) and human pancreatic stellate cells (hPaSC) using mass spectrometry (MS)-based quantitative proteomics to investigate pathophysiologic mechanisms. Proteins were isolated from whole cell lysates of immortalized hHSC and hPaSC. These proteins were tryptically digested, labeled with tandem mass tags (TMT), fractionated by OFFGEL, and subjected to MS. Proteins significantly different in abundance (P < 0.05) were classified via gene ontology (GO) analysis. We identified 1223 proteins and among them, 1222 proteins were quantifiable. Statistical analysis determined that 177 proteins were of higher abundance in hHSC, while 157 were of higher abundance in hPaSC. GO classification revealed that proteins of relatively higher abundance in hHSC were associated with protein production, while those of relatively higher abundance in hPaSC were involved in cell structure. Future studies using the methodologies established herein, but with further upstream fractionation and/or use of enhanced MS instrumentation will allow greater proteome coverage, achieving a comprehensive proteomic analysis of hHSC and hPaSC.


Pancreas | 2016

The Atlanta Classification, Revised Atlanta Classification, and Determinant-Based Classification of Acute Pancreatitis: Which Is Best at Stratifying Outcomes?

Vivek Kadiyala; Shadeah Suleiman; Julia McNabb-Baltar; Bechien U. Wu; Peter A. Banks; Vikesh K. Singh

Objectives To determine which classification is more accurate in stratifying severity. Methods The study used a retrospective analysis of a prospective acute pancreatitis database (June 2005–December 2007). Acute pancreatitis severity was stratified according to the Atlanta classification (AC) 1992, the revised Atlanta classification (RAC) 2012, and the determinant-based classification (DBC) 2012. Receiver operating characteristic analysis (area under the curve) compared the accuracy of each classification. Logistic regression identified predictors of mortality. Results 338 patients were analyzed: 13% had persistent organ failure (POF) (>48 hours), of whom 37% had multisystem POF, and 11% had pancreatic necrosis, of whom 19% had infected necrosis. Mortality was 4.1%. For predicting mortality (area under the curve), the RAC (0.91) and DBC (0.92) were comparable (P = 0.404); both outperformed the AC (0.81) (P < 0.001). For intensive care unit admission, the RAC (0.85) and DBC (0.85) were comparable (P = 0.949); both outperformed the AC (0.79) (P < 0.05). There were 2 patients in the critical category of the DBC. Multisystem POF was an independent predictor of mortality (odds ratio, 75.0; 95% confidence interval, 13.7–410.6; P < 0.001), whereas single-system POF, sterile necrosis, and infected necrosis were not. Conclusion The RAC and DBC were generally comparable in stratifying severity. The paucity of patients in the critical category in the DBC limits its utility. Neither classification accounts for the impact of multisystem POF, which was the strongest predictor of mortality.


Journal of the Pancreas | 2013

Utility of Commercial DNA Analysis in Detecting Malignancy within Pancreatic Cysts

Linda S. Lee; Bechien U. Wu; Peter A. Banks; Vivek Kadiyala; Shivani Mehta; John R. Saltzman; Christopher C. Thompson; Andrew M. Bellizzi

CONTEXT Pancreatic cysts raise concern because of their malignant potential. Our aims were to assess accuracy of DNA analysis in detecting malignant pancreatic cysts at EUS-FNA and to determine whether DNA analysis added to imaging and cyst fluid studies enhanced International Association of Pancreatology (IAP) guidelines for resection of pancreatic cysts. METHODS This is a retrospective study including pancreatic cysts undergoing EUS-FNA and DNA analysis with k-ras and loss of heterozygosity testing. Diagnostic models of 2006 and 2012 IAP guidelines, DNA analysis alone, and DNA combined with 2012 IAP guidelines were developed, and area under receiver operator characteristic curves (AUC) compared to determine the added value of DNA for detecting malignant cysts at the time of EUS-FNA. RESULTS Two-hundreds and fifty-seven patients were included with 8 (3.1%) malignant cysts. Solid component (P<0.001), main pancreatic duct dilation (P=0.012), suspicious or malignant cytology (P=0.001), and high DNA quantity (P<0.001) were associated with malignancy. Concurrent high amplitude k-ras with loss of heterozygosity mutations was highly specific (98.4%) though insensitive (12.5%) for malignancy. The 2012 IAP guideline (AUC=0.87; 95% CI: 0.82-0.91) was superior to 2006 IAP guideline (AUC=0.54; 95% CI: 0.47-0.60) and DNA analysis alone (AUC=0.60; 95% CI: 0.53-0.66) for detecting malignant cysts (P=0.004 and P=0.002, respectively). Addition of DNA did not improve performance of the 2012 IAP guideline (AUC=0.84; 95% CI: 0.79-0.88). CONCLUSIONS Commercial DNA analysis does not add useful information beyond imaging and cytology for detection of malignant pancreatic cysts. The 2012 IAP guideline better predicted malignant cysts than the 2006 IAP guideline.


Clinical and translational gastroenterology | 2012

Mass Spectrometry-Based (GeLC-MS/MS) Comparative Proteomic Analysis of Endoscopically (ePFT) Collected Pancreatic and Gastroduodenal Fluids

Joao A. Paulo; Vivek Kadiyala; Peter A. Banks; Hanno Steen; Darwin L. Conwell

OBJECTIVES:The secretin-stimulated endoscopic pancreatic function test (ePFT) allows for the safe collection of gastroduodenal and pancreatic fluid from the duodenum. We test the hypothesis that these endoscopically collected fluids have different proteomes. As such, we aim to show that the ePFT method can be used to collect fluid enriched in pancreatic proteins to test for pancreatic function.METHODS:Gastroduodenal and pancreatic fluid were collected sequentially from chronic pancreatitis patients undergoing an ePFT. Proteins from each fluid type were extracted using previously published optimized methods and subjected to GeLC-MS/MS analysis for protein identification and bioinformatics analysis.RESULTS:Mass spectrometry analysis identified proteins that were exclusive in either gastroduodenal (46) or pancreatic fluid (234). Subsequent quantitative analysis revealed proteins that were differentially abundant with statistical significance. As expected, proteolytic enzymes and protease inhibitors were among the differentially detected proteins. The proteases pepsinogens and gastrin were enriched in gastroduodenal fluid, while common pancreatic enzymes (e.g., aminopeptidase N, chymotrypsin C, elastase-3A, trypsin, and carboxypeptidase A1, and elastase 2B) were found in greater abundance in pancreatic fluid. Similarly for protease inhibitors, members of the cystatin family were exclusive to gastroduodenal fluid, while serpins A11, B4, and D1 were exclusive to pancreatic fluid.CONCLUSIONS:We have shown that ePFT collection coupled with mass spectrometry can be used to identify differentially detected proteins in gastroduodenal and pancreatic fluids. The data obtained using GeLC-MS/MS techniques provide further evidence supporting the feasibility of using ePFT-collected fluid to study specific diseases of the upper gastrointestinal tract, such as chronic pancreatitis.


Digestive Endoscopy | 2015

Randomized Noninferiority Trial Comparing Diagnostic Yield of Cytopathologist-guided versus 7 passes for EUS-FNA of Pancreatic Masses.

Linda S. Lee; Jose Nieto; Rabindra R. Watson; Allen L. Hwang; Venkatara R. Muthusamy; Laura Walter; Kunal Jajoo; Marvin Ryou; John R. Saltzman; Michael D. Saunders; Shadeah Suleiman; Vivek Kadiyala

To improve diagnostic yield of endoscopic ultrasound‐guided fine‐needle aspiration (EUS‐FNA) in solid pancreatic lesions, on‐site cytology review has been recommended. Because this is not widely available throughout the world, the aim of the present study was to compare the diagnostic yield of EUS‐FNA carried out with rapid on‐site evaluation (ROSE) versus seven FNA passes without ROSE in pancreatic masses.

Collaboration


Dive into the Vivek Kadiyala's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Shadeah Suleiman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hanno Steen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John R. Saltzman

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

Nisha I. Sainani

Brigham and Women's Hospital

View shared research outputs
Top Co-Authors

Avatar

S.J. Burton

Brigham and Women's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge