Network


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

Hotspot


Dive into the research topics where Sudhanshu Shukla is active.

Publication


Featured researches published by Sudhanshu Shukla.


Journal of the National Cancer Institute | 2017

Development of a RNA-Seq Based Prognostic Signature in Lung Adenocarcinoma.

Sudhanshu Shukla; Joseph R. Evans; Rohit Malik; Felix Y. Feng; Saravana M. Dhanasekaran; Xuhong Cao; Guoan Chen; David G. Beer; Hui Jiang; Arul M. Chinnaiyan

Background: Precision therapy for lung cancer will require comprehensive genomic testing to identify actionable targets as well as ascertain disease prognosis. RNA-seq is a robust platform that meets these requirements, but microarray-derived prognostic signatures are not optimal for RNA-seq data. Thus, we undertook the first prognostic analysis of lung adenocarcinoma RNA-seq data and generated a prognostic signature. Methods: Lung adenocarcinoma RNA-seq and clinical data from The Cancer Genome Atlas (TCGA) were divided chronologically into training (n = 255) and validation (n = 157) cohorts. In the training cohort, prognostic association was assessed by univariate Cox analysis. A prognostic signature was built with stepwise multivariable Cox analysis. Outcomes by risk group, stage, and mutation status were analyzed with Kaplan-Meier and multivariable Cox analyses. All the statistical tests were two-sided. Results: In the training cohort, 96 genes had prognostic association with P values of less than or equal to 1.00x10-4, including five long noncoding RNAs (lncRNAs). Stepwise regression generated a four-gene signature, including one lncRNA. Signature high-risk cases had worse overall survival (OS) in the TCGA validation cohort (hazard ratio [HR] = 3.07, 95% confidence interval [CI] = 2.00 to 14.62) and a University of Michigan institutional cohort (n = 67; HR = 2.05, 95% CI = 1.18 to 4.55), and worse metastasis-free survival in the TCGA validation cohort (HR = 3.05, 95% CI = 2.31 to 13.37). The four-gene prognostic signature also statistically significantly stratified overall survival in important clinical subsets, including stage I (HR = 2.78, 95% CI = 1.91 to 11.13), EGFR wild-type (HR = 3.01, 95% CI = 1.73 to 14.98), and EGFR mutant (HR = 8.99, 95% CI = 62.23 to 141.44). The four-gene prognostic signature also stood out on top when compared with other prognostic signatures. Conclusions: Here, we present the first RNA-seq prognostic signature for lung adenocarcinoma that can provide a powerful prognostic tool for precision oncology as part of an integrated RNA-seq clinical sequencing program.


Neoplasia | 2016

Identification and Validation of PCAT14 as Prognostic Biomarker in Prostate Cancer

Sudhanshu Shukla; Xiang Zhang; Yashar S. Niknafs; Lanbo Xiao; Rohit Mehra; Marcin Cieślik; Ashley E. Ross; Edward M. Schaeffer; Bhavna Malik; Shuling Guo; Susan M. Freier; Huynh Hoa Bui; Javed Siddiqui; Xiaojun Jing; Xuhong Cao; Saravana M. Dhanasekaran; Felix Y. Feng; Arul M. Chinnaiyan; Rohit Malik

Rapid advances in the discovery of long noncoding RNAs (lncRNAs) have identified lineage- and cancer-specific biomarkers that may be relevant in the clinical management of prostate cancer (PCa). Here we assembled and analyzed a large RNA-seq dataset, from 585 patient samples, including benign prostate tissue and both localized and metastatic PCa to discover and validate differentially expressed genes associated with disease aggressiveness. We performed Sample Set Enrichment Analysis (SSEA) and identified genes associated with low versus high Gleason score in the RNA-seq database. Comparing Gleason 6 versus 9+ PCa samples, we identified 99 differentially expressed genes with variable association to Gleason grade as well as robust expression in prostate cancer. The top-ranked novel lncRNA PCAT14, exhibits both cancer and lineage specificity. On multivariate analysis, low PCAT14 expression independently predicts for BPFS (P = .00126), PSS (P = .0385), and MFS (P = .000609), with trends for OS as well (P = .056). An RNA in-situ hybridization (ISH) assay for PCAT14 distinguished benign vs malignant cases, as well as high vs low Gleason disease. PCAT14 is transcriptionally regulated by AR, and endogenous PCAT14 overexpression suppresses cell invasion. Thus, Using RNA-sequencing data we identify PCAT14, a novel prostate cancer and lineage-specific lncRNA. PCAT14 is highly expressed in low grade disease and loss of PCAT14 predicts for disease aggressiveness and recurrence.


Cancer Discovery | 2016

Biallelic Alteration and Dysregulation of the Hippo Pathway in Mucinous Tubular and Spindle Cell Carcinoma of the Kidney

Rohit Mehra; Pankaj Vats; Marcin Cieslik; Xuhong Cao; Fengyun Su; Sudhanshu Shukla; Aaron M. Udager; Rui Wang; Jincheng Pan; Katayoon Kasaian; Robert J. Lonigro; Javed Siddiqui; Kumpati Premkumar; Ganesh S. Palapattu; Alon Z. Weizer; Khaled S. Hafez; J. Stuart Wolf; Ankur R. Sangoi; Kiril Trpkov; Adeboye O. Osunkoya; Ming Zhou; Giovanna Giannico; Jesse K. McKenney; Saravana M. Dhanasekaran; Arul M. Chinnaiyan

Mucinous tubular and spindle cell carcinoma (MTSCC) is a relatively rare subtype of renal cell carcinoma (RCC) with distinctive morphologic and cytogenetic features. Here, we carry out whole-exome and transcriptome sequencing of a multi-institutional cohort of MTSCC (n = 22). We demonstrate the presence of either biallelic loss of Hippo pathway tumor suppressor genes (TSG) and/or evidence of alteration of Hippo pathway genes in 85% of samples. PTPN14 (31%) and NF2 (22%) were the most commonly implicated Hippo pathway genes, whereas other genes such as SAV1 and HIPK2 were also involved in a mutually exclusive fashion. Mutations in the context of recurrent chromosomal losses amounted to biallelic alterations in these TSGs. As a readout of Hippo pathway inactivation, a majority of cases (90%) exhibited increased nuclear YAP1 protein expression. Taken together, nearly all cases of MTSCC exhibit some evidence of Hippo pathway dysregulation. SIGNIFICANCE MTSCC is a rare and relatively recently described subtype of RCC. Next-generation sequencing of a multi-institutional MTSCC cohort revealed recurrent chromosomal losses and somatic mutations in the Hippo signaling pathway genes leading to potential YAP1 activation. In virtually all cases of MTSCC, there was evidence of Hippo pathway dysregulation, suggesting a common mechanistic basis for this disease. Cancer Discov; 6(11); 1258-66. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 1197.


The American Journal of Surgical Pathology | 2016

Renal Cell Carcinoma Occurring in Patients With Prior Neuroblastoma: A Heterogenous Group of Neoplasms

Sara M. Falzarano; Jesse K. McKenney; Rodolfo Montironi; John N. Eble; Adeboye O. Osunkoya; Juan Guo; Shengmei Zhou; Hong Xiao; Saravana M. Dhanasekaran; Sudhanshu Shukla; Rohit Mehra; Cristina Magi-Galluzzi

Renal cell carcinoma (RCC) associated with neuroblastoma (NB) was included as a distinct entity in the 2004 World Health Organization classification of kidney tumors. A spectrum of RCC subtypes has been reported in NB survivors. We herein describe a series of 8 RCCs diagnosed in 7 patients with a history of NB. Microscopic evaluation, immunohistochemical staining for PAX8, cathepsin K, and succinate dehydrogenase subunit B (SDHB), and fluorescence in situ hybridization (FISH) for TFE3 and TFEB were performed. Four distinct morphologic subtypes were identified: 3 tumors were characterized by cells with abundant oncocytoid cytoplasm and irregular nuclei; 3 showed features of microphthalmia transcription factor family translocation RCC (MiTF-RCC); 1 had features of hybrid oncocytic-chromophobe tumor; 1 had papillary RCC histology. All RCCs expressed PAX8 and retained SDHB expression. Cathepsin K was positive in 2 MiTF-RCCs, 1 was TFEB FISH positive, and the other was indeterminate. Cathepsin K was negative in a third MiTF-RCC with TFE3 rearrangement. TFE3 FISH was negative in 4 and insufficient in 1 of the other 5 RCCs. While a subset of RCCs associated with NB is characterized by cells with prominent oncocytoid cytoplasm, other RCC subtypes also occur in post-NB patients. Renal neoplasms occurring in patients with a history of NB do not represent a single entity but a heterogenous group of RCCs. SDHB mutations do not explain the subset of nontranslocation RCCs with oncocytoid features; therefore, further studies are needed to clarify whether they may represent a distinct entity with unique molecular abnormalities or may belong to other emerging RCC subtypes.


Oncotarget | 2016

Non-coding RNA LINC00857 is predictive of poor patient survival and promotes tumor progression via cell cycle regulation in lung cancer

Lihui Wang; Yanli He; Weijun Liu; Shengbin Bai; Lei Xiao; Jie Zhang; Saravana M. Dhanasekaran; Zhuwen Wang; Shanker Kalyana-Sundaram; O. Alejandro Balbin; Sudhanshu Shukla; Yi Lu; Jules Lin; Rishindra M. Reddy; Philip W. Carrott; William R. Lynch; Andrew C. Chang; Arul M. Chinnaiyan; David G. Beer; Jian Zhang; Guoan Chen

We employed next generation RNA sequencing analysis to reveal dysregulated long non-coding RNAs (lncRNAs) in lung cancer utilizing 461 lung adenocarcinomas (LUAD) and 156 normal lung tissues from 3 separate institutions. We identified 281 lncRNAs with significant differential-expression between LUAD and normal lung tissue. LINC00857, a top deregulated lncRNAs, was overexpressed in tumors and significantly associated with poor survival in LUAD. knockdown of LINC00857 with siRNAs decreased tumor cell proliferation, colony formation, migration and invasion in vitro, as well as tumor growth in vivo. Overexpression of LINC00857 increased cancer cell proliferation, colony formation and invasion. Mechanistic analyses indicated that LINC00857 mediates tumor progression via cell cycle regulation. Our study highlights the diagnostic/prognostic potential of LINC00857 in LUAD besides delineating the functional and mechanistic aspects of its aberrant disease specific expression and potentially using as a new therapeutic target.


Cancer Cell | 2017

ACK1/TNK2 Regulates Histone H4 Tyr88-phosphorylation and AR Gene Expression in Castration-Resistant Prostate Cancer

Kiran Mahajan; Pavani Malla; Harshani R. Lawrence; Zhihua Chen; Chandan Kumar-Sinha; Rohit Malik; Sudhanshu Shukla; Jongphil Kim; Domenico Coppola; Nicholas J. Lawrence; Nupam P. Mahajan

The androgen receptor (AR) is critical for the progression of prostate cancer to a castration-resistant (CRPC) state. AR antagonists are ineffective due to their inability to repress the expression of AR or its splice variant, AR-V7. Here, we report that the tyrosine kinase ACK1 (TNK2) phosphorylates histone H4 at tyrosine 88 upstream of the AR transcription start site. The WDR5/MLL2 complex reads the H4-Y88-phosphorylation marks and deposits the transcriptionally activating H3K4-trimethyl marks promoting AR transcription. Reversal of the pY88-H4 epigenetic marks by the ACK1 inhibitor (R)-9bMS-sensitized naive and enzalutamide-resistant prostate cancer cells and reduced AR and AR-V7 levels to mitigate CRPC tumor growth. Thus, a feedforward ACK1/pY88-H4/WDR5/MLL2/AR epigenetic circuit drives CRPC and is necessary for maintenance of the malignant state.


Nature Genetics | 2018

Analysis of the androgen receptor–regulated lncRNA landscape identifies a role for ARLNC1 in prostate cancer progression

Yajia Zhang; Sethuramasundaram Pitchiaya; Marcin Cieślik; Yashar S. Niknafs; Jean C.-Y. Tien; Yasuyuki Hosono; Matthew K. Iyer; Sahr Yazdani; Shruthi Subramaniam; Sudhanshu Shukla; Xia Jiang; Lisha Wang; Tzu-Ying Liu; Michael Uhl; Alexander Gawronski; Yuanyuan Qiao; Lanbo Xiao; Saravana M. Dhanasekaran; Kristin M. Juckette; Lakshmi P. Kunju; Xuhong Cao; Utsav Patel; Mona Batish; Girish C. Shukla; Michelle T. Paulsen; Mats Ljungman; Hui Jiang; Rohit Mehra; Rolf Backofen; Cenk Sahinalp

The androgen receptor (AR) plays a critical role in the development of the normal prostate as well as prostate cancer. Using an integrative transcriptomic analysis of prostate cancer cell lines and tissues, we identified ARLNC1 (AR-regulated long noncoding RNA 1) as an important long noncoding RNA that is strongly associated with AR signaling in prostate cancer progression. Not only was ARLNC1 induced by the AR protein, but ARLNC1 stabilized the AR transcript via RNA–RNA interaction. ARLNC1 knockdown suppressed AR expression, global AR signaling and prostate cancer growth in vitro and in vivo. Taken together, these data support a role for ARLNC1 in maintaining a positive feedback loop that potentiates AR signaling during prostate cancer progression and identify ARLNC1 as a novel therapeutic target.ARLNC1 is a newly discovered lncRNA that is induced by androgen receptor (AR) and maintains AR signaling by stabilizing the AR transcript. Knockdown of ARLNC1 suppresses AR expression, AR signaling and prostate cancer growth in vitro and in vivo.


Bioinformatics | 2015

Two-pass alignment improves novel splice junction quantification

Brendan Veeneman; Sudhanshu Shukla; Saravana M. Dhanasekaran; Arul M. Chinnaiyan; Alexey I. Nesvizhskii

MOTIVATION Discovery of novel splicing from RNA sequence data remains a critical and exciting focus of transcriptomics, but reduced alignment power impedes expression quantification of novel splice junctions. RESULTS Here, we profile performance characteristics of two-pass alignment, which separates splice junction discovery from quantification. Per sample, across a variety of transcriptome sequencing datasets, two-pass alignment improved quantification of at least 94% of simulated novel splice junctions, and provided as much as 1.7-fold deeper median read depth over those splice junctions. We further demonstrate that two-pass alignment works by increasing alignment of reads to splice junctions by short lengths, and that potential alignment errors are readily identifiable by simple classification. Taken together, two-pass alignment promises to advance quantification and discovery of novel splicing events. CONTACT [email protected], [email protected] AVAILABILITY AND IMPLEMENTATION Two-pass alignment was implemented here as sequential alignment, genome indexing, and re-alignment steps with STAR. Full parameters are provided in Supplementary Table 2. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.


Clinical Cancer Research | 2017

Age and Gender Associations of Virus Positivity in Merkel Cell Carcinoma Characterized Using a Novel RNA In Situ Hybridization Assay

Lisha Wang; Paul W. Harms; Nallasivam Palanisamy; Shannon Carskadon; Xuhong Cao; Javed Siddiqui; Rajiv M. Patel; Sylvia Zelenka-Wang; Alison B. Durham; Douglas R. Fullen; Kelly L. Harms; Fengyun Su; Sudhanshu Shukla; Rohit Mehra; Arul M. Chinnaiyan

Purpose: Merkel cell carcinoma (MCC) is a highly aggressive neuroendocrine tumor of the skin. Merkel cell polyomavirus (MCPyV) plays an oncogenic role in the majority of MCCs. Detection of MCPyV in MCCs has diagnostic utility and prognostic potential. We investigated whether RNAscope, an RNA in situ hybridization (ISH) assay for detection of RNA transcripts in tissues, is useful for MCPyV detection. Experimental Design: We applied an RNAscope probe targeting MCPyV T antigen transcripts on tissue microarrays (TMA) and whole-tissue sections encompassing 87 MCCs from 75 patients, 14 carcinomas of other types, and benign tissues. For comparison, qPCR was performed on 57 cases of MCC from 52 patients. Results: RNA-ISH demonstrated the presence of MCPyV in 37 of 75 cases (49.3%). Notably, tumors from younger patients (<73 years) had a significantly higher virus positivity than those from elderly patients (≥73 years; 64.9% vs. 34.2%, P = 0.011). Female patients had a higher positive rate of MCPyV than male patients (66.7% vs. 39.6%, P = 0.032). Data from both RNA-ISH and qPCR were available for 57 samples. Considering MCPyV qPCR as the gold standard for determining MCPyV status, RNAscope had 100% sensitivity and 100% specificity. There was a strong correlation between qPCR copy number and RNA-ISH product score (Spearman correlation coefficient R2 = 0.932, P < 0.0001). Conclusions: RNA-ISH is comparably sensitive to qPCR for detection of MCPyV and allows for correlation with tissue morphology. This study also reveals a significant association between age, gender, and MCPyV positivity. Clin Cancer Res; 23(18); 5622–30. ©2017 AACR.


Cancer Research | 2016

Abstract B39: Long noncoding RNA PCAT14/PRCAT104; A prognostic biomarker in prostate cancer

Rohit Malik; Xiang Zhang; Sudhanshu Shukla; Yashar Y. Niknafs; Shuang Zhao; Felix Y. Feng; Arul M. Chinnaiyan

Prostate cancer is the second most common epithelial cancer and the second leading cause of cancer-related death for men in the United States. While majority of prostate cancer cases are indolent and cause minimal morbidity and mortality, a subset of men progress to a hormone-refractory aggressive disease with high mortality. Markers such as PSA and PCA3 perform well in diagnosis of disease; however these biomarkers are unable to predict disease progression. Therefore, there is an urgent need to identify clinical predictors of disease progression. Long non-coding RNAs (lncRNAs) are emerging as an important class of biomolecules that exhibit significant lineage- and cancer-specificity making them ideal biomarker candidates. Using RNA-Seq data from The Cancer Genome Atlas (TCGA) and as well as from libraries generated in our laboratory, we identified PCAT14/PRCAT104 as a marker of low Gleason disease. PCAT14 was shown to be highly expressed in prostate cancer compared to benign tissue; however, its expression in prostate cancer was limited to low Gleason disease. To directly evaluate the relationship between PCAT14 levels and clinical outcome, we assessed its expression in more that 1600 Prostate cancer samples by a high-density Affymetrix GeneChip platform. In all 5 cohorts analyzed, PCAT14 was shown to be a strong prognostic marker in its ability to predict biochemical recurrence, clinical progression to systemic disease and prostate cancer–specific mortality. Furthermore, in a multivariate analysis, PCAT14 expression also predict resistance to androgen deprivation therapy (ADT) (p=0.012). More interestingly, PCAT14 was able to add to prognostic value of SCHLAP1, a lncRNA that we previously showed to be an excellent prognostic marker in prostate cancer. PCAT14 in combination with SCHLAP1 was able better in predicting 10-year metastasis free survival (AUC: 0.64) than SCHLAP1 alone (AUC=0.58). Taken together, we identified a novel marker of low grade prostate cancer that in combination with SCHALP1, a marker of aggressive cancer, can predict disease progression and hence can be of immense value for treatment individualization in prostate cancer Citation Format: Rohit Malik, Xiang Zhang, Sudhanshu Shukla, Yashar Y. Niknafs, Shuang Zhao, Felix Y. Feng, Arul M. Chinnaiyan. Long noncoding RNA PCAT14/PRCAT104; A prognostic biomarker in prostate cancer. [abstract]. In: Proceedings of the AACR Special Conference on Noncoding RNAs and Cancer: Mechanisms to Medicines ; 2015 Dec 4-7; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2016;76(6 Suppl):Abstract nr B39.

Collaboration


Dive into the Sudhanshu Shukla'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 Mehra

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Rohit Malik

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Felix Y. Feng

University of California

View shared research outputs
Top Co-Authors

Avatar

Fengyun Su

University of Michigan

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lanbo Xiao

University of Michigan

View shared research outputs
Top Co-Authors

Avatar

Lisha Wang

University of Michigan

View shared research outputs
Researchain Logo
Decentralizing Knowledge