Network


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

Hotspot


Dive into the research topics where Priya Pai is active.

Publication


Featured researches published by Priya Pai.


Cancer Letters | 2013

Guggulsterone decreases proliferation and metastatic behavior of pancreatic cancer cells by modulating JAK/STAT and Src/FAK signaling.

Muzafar A. Macha; Satyanarayana Rachagani; Suprit Gupta; Priya Pai; Moorthy P. Ponnusamy; Surinder K. Batra; Maneesh Jain

Inadequate efficacy, high toxicity and drug resistance associated with existing chemotherapeutic agents mandate a need for novel therapeutic strategies for highly aggressive Pancreatic Cancer (PC). Guggulsterone (GS) exhibits potent anti-proliferative effects against various cancer cells and has emerged as an attractive candidate for use in complementary or preventive cancer therapies. However, the knowledge regarding the therapeutic potential of GS in PC is still limited and needs to be explored. We studied the effect of GS on PC cell growth, motility and invasion and elucidated the molecular mechanisms associated with its anti-tumor effects. Treatment of Capan1 and CD18/HPAF PC cells with GS resulted in dose- and time-dependent growth inhibition and decreased colony formation. Further, GS treatment induced apoptosis and cell cycle arrest as assessed by Annexin-V assay and FACS analysis. Increased apoptosis following GS treatment was accompanied with Bad dephosphorylation and its translocation to the mitochondria, increased Caspase-3 activation, decreased Cyclin D1, Bcl-2 and xIAP expression. Additionally, GS treatment decreased motility and invasion of PC cells by disrupting cytoskeletal organization, inhibiting activation of FAK and Src signaling and decreased MMP9 expression. More importantly, GS treatment decreased mucin MUC4 expression in Capan1 and CD18/HPAF cells through transcriptional regulation by inhibiting Jak/STAT pathway. In conclusion, our results support the utility of GS as a potential therapeutic agent for lethal PC.


PLOS ONE | 2013

MUC4 Overexpression Augments Cell Migration and Metastasis through EGFR Family Proteins in Triple Negative Breast Cancer Cells

Partha Mukhopadhyay; Imayavaramban Lakshmanan; Moorthy P. Ponnusamy; Subhankar Chakraborty; Maneesh Jain; Priya Pai; Lynette M. Smith; Subodh M. Lele; Surinder K. Batra

Introduction Current studies indicate that triple negative breast cancer (TNBC), an aggressive breast cancer subtype, is associated with poor prognosis and an early pattern of metastasis. Emerging evidence suggests that MUC4 mucin is associated with metastasis of various cancers, including breast cancer. However, the functional role of MUC4 remains unclear in breast cancers, especially in TNBCs. Method In the present study, we investigated the functional and mechanistic roles of MUC4 in potentiating pathogenic signals including EGFR family proteins to promote TNBC aggressiveness using in vitro and in vivo studies. Further, we studied the expression of MUC4 in invasive TNBC tissue and normal breast tissue by immunostaining. Results MUC4 promotes proliferation, anchorage-dependent and-independent growth of TNBC cells, augments TNBC cell migratory and invasive potential in vitro, and enhances tumorigenicity and metastasis in vivo. In addition, our studies demonstrated that MUC4 up-regulates the EGFR family of proteins, and augments downstream Erk1/2, PKC-γ, and FAK mediated oncogenic signaling. Moreover, our studies also showed that knockdown of MUC4 in TNBC cells induced molecular changes suggestive of mesenchymal to epithelial transition. We also demonstrated in this study, for the first time, that knockdown of MUC4 was associated with reduced expression of EGFR and ErbB3 (EGFR family proteins) in TNBC cells, suggesting that MUC4 uses an alternative to ErbB2 mechanism to promote aggressiveness. We further demonstrate that MUC4 is differentially over-expressed in invasive TNBC tissues compared to normal breast tissue. Conclusions MUC4 mucin expression is associated with TNBC pathobiology, and its knockdown reduced aggressiveness in vitro, and tumorigenesis and metastasis in vivo. Overall, our findings suggest that MUC4 mucin promotes invasive activities of TNBC cells by altering the expression of EGFR, ErbB2, and ErbB3 molecules and their downstream signaling.


Current Pharmaceutical Design | 2014

MicroRNAs (miRNAs) as Biomarker(s) for Prognosis and Diagnosis of Gastrointestinal (GI) Cancers

Muzafar A. Macha; Parthasarathy Seshacharyulu; Shiv Ram Krishn; Priya Pai; Satyanarayana Rachagani; Maneesh Jain; Surinder K. Batra

Gastrointestinal (GI) cancers remain one of the most common malignancies and are the second common cause of cancer deaths worldwide. The limited effectiveness of therapy for patients with advanced stage and recurrent disease is a reflection of an incomplete understanding of the molecular basis of GI carcinogenesis. Major advancements have improved our understanding of pathology and pathogenesis of GI cancers, but high mortality rates, unfavorable prognosis and lack of clinical predictive biomarkers provide an impetus to investigate new sensitive and specific diagnostic and prognostic markers for GI cancers. MicroRNAs (miRNAs) are short (19-24 nucleotides) noncoding RNA molecules that regulate gene expression at the posttranscriptional level thus playing an important role in modulating various biological processes including, but not limited to developmental processes, proliferation, apoptosis, metabolism, differentiation, epithelial-mechenchymal transition and are involved in the initiation and progression of various human cancers. Unique miRNA expression profiles have been observed in various cancer types at different stages, suggesting their potential as diagnostic and prognostic biomarkers. Due to their tumor-specific and tissue-specific expression profiles, stability, robust clinical assays for detection in serum as well as in formalin-fixed tissue samples, miRNAs have emerged as attractive candidates for diagnostic and prognostic applications. This review summarizes recent research supporting the utility of miRNAs as novel diagnostic and prognostic tools for GI cancers.


Current Drug Targets | 2013

Prospects of miRNA-Based Therapy for Pancreatic Cancer

Priya Pai; Satyanarayana Rachagani; Chandrakanth Are; Surinder K. Batra

Pancreatic cancer (PC) is the fourth leading cause of cancer related deaths in the U.S., with a less than 6% five-year survival rate. Treatment is confounded by advanced stage of disease at presentation, frequent metastasis to distant organs at the time of diagnosis and resistance to conventional chemotherapy. In addition, the molecular pathogenesis of the disease is unclear. The extensive study of miRNAs over the past several years has revealed that miRNAs are frequently de-regulated in pancreatic cancer and contribute to the pathogenesis and aggressiveness of the disease. Several studies have tackled the practical difficulties in the application of miRNAs as viable therapeutic and diagnostic tools. Given that a single miRNA can affect a myriad of cellular processes, successful targeting of miRNAs as therapeutic agents could likely yield dramatic results. The current review attempts to summarize the advances in the field and assesses the prospects for miRNA profiling and targeting in aiding PC treatment.


Oncotarget | 2015

Changes in microRNA (miRNA) expression during pancreatic cancer development and progression in a genetically engineered KrasG12D;Pdx1-Cre mouse (KC) model

Satyanarayana Rachagani; Muzafar A. Macha; Melanie Menning; Parama Dey; Priya Pai; Lynette M. Smith; Yin Yuan Mo; Surinder K. Batra

Differential expression of microRNAs (miRNAs) has been demonstrated in various cancers, including pancreatic cancer (PC). Due to the lack of tissue samples from early-stages of PC, the stage-specific alteration of miRNAs during PC initiation and progression is largely unknown. In this study, we investigated the global miRNA expression profile and their processing machinery during PC progression using the KrasG12D;Pdx1-Cre (KC) mouse model. At 25 weeks, the miRNA microarray analysis revealed significant downregulation of miR-150, miR-494, miR-138, miR-148a, miR-216a, and miR-217 and upregulation of miR-146b, miR-205, miR-31, miR-192, and miR-21 in KC mice compared to controls. Further, expression of miRNA biosynthetic machinery including Dicer, Exportin-5, TRKRA, and TARBP2 were downregulated, while DGCR8 and Ago2 were upregulated in KC mice. In addition, from 10 to 50 weeks of age, stage-specific expression profiling of miRNA in KC mice revealed downregulation of miR-216, miR-217, miR-100, miR-345, miR-141, miR-483-3p, miR-26b, miR-150, miR-195, Let-7b and Let-96 and upregulation of miR-21, miR-205, miR-146b, miR-34c, miR-1273, miR-223 and miR-195 compared to control mice. Interestingly, the differential expression of miRNA in mice also corroborated with the miRNA expression in human PC cell lines and tissue samples; ectopic expression of Let-7b in CD18/HPAF and Capan1 cells resulted in downregulation of KRAS and MSST1 expression. Overall, the present study aids an understanding of miRNA expression patterns during PC pathogenesis and helps to facilitate the identification of promising and novel early diagnostic/prognostic markers and therapeutic targets.


Carcinogenesis | 2016

Mucins and Wnt/β-catenin signaling in gastrointestinal cancers: an unholy nexus

Priya Pai; Satyanarayana Rachagani; Punita Dhawan; Surinder K. Batra

The Wnt/β-catenin signaling pathway is indispensable for embryonic development, maintenance of adult tissue homeostasis and repair of epithelial injury. Unsurprisingly, aberrations in this pathway occur frequently in many cancers and often result in increased nuclear β-catenin. While mutations in key pathway members, such as β-catenin and adenomatous polyposis coli, are early and frequent occurrences in most colorectal cancers (CRC), mutations in canonical pathway members are rare in pancreatic ductal adenocarcinoma (PDAC). Instead, in the majority of PDACs, indirect mechanisms such as promoter methylation, increased ligand secretion and decreased pathway inhibitor secretion work in concert to promote aberrant cytosolic/nuclear localization of β-catenin. Concomitant with alterations in β-catenin localization, changes in mucin expression and localization have been documented in multiple malignancies. Indeed, numerous studies over the years suggest an intricate and mutually regulatory relationship between mucins (MUCs) and β-catenin. In the current review, we summarize several studies that describe the relationship between mucins and β-catenin in gastrointestinal malignancies, with particular emphasis upon colorectal and pancreatic cancer.


Oncotarget | 2017

Mucins and associated O-glycans based immunoprofile for stratification of colorectal polyps: clinical implication for improved colon surveillance

Shiv Ram Krishn; Sukhwinder Kaur; Yuri Sheinin; Lynette M. Smith; Shailendra K. Gautam; Asish Patel; Maneesh Jain; Vasthala Juvvigunta; Priya Pai; Audrey J. Lazenby; Hemant K. Roy; Surinder K. Batra

Sessile serrated adenoma/polyps (SSA/P) are premalignant lesions of colorectal cancer that are difficult to distinguish histologically from hyperplastic polyps (HP) of minimal to no malignant potential. Specific markers for differentiating SSA/P from HP can aid clinicians for optimizing colon surveillance intervals. The present study investigates the potential of mucins and associated O-glycans to distinguish SSA/P from HP. Expression of colonic mucins (MUC1, MUC4, MUC17, MUC2, and MUC5AC) and O-glycans [Sialyl LewisA (CA19-9) and Tn/Sialyl-Tn on MUC1] were analyzed in HP (n=33), SSA/P (n=39), and tubular adenoma (TA) (n=36) samples by immunohistochemistry. A significantly reduced expression of MUC4 (p=0.0066), elevated expression of MUC17 (p=0.0002), and MUC5AC (p<0.0001) was observed in SSA/P cases in comparison to HP cases. Interestingly, significantly higher number of SSA/P cases (p<0.0001) exhibited MUC5AC expression in the goblet cells as well as filled the crypt lumen compared to only goblet cells in majority of the HP cases. Improved diagnostic potential was revealed by multivariate logistic regression analysis where combinatorial panel of MUC5AC/MUC17 discriminated SSA/P from HP (SN/SP=85/82%). Finally, the decision tree model based marker panel (CA19-9/MUC17/MUC5AC) predicted HP, SSA/P and TA with SN/SP of 58%/95%, 79%/90% and 97%/83%, respectively. Overall, the mucin and associated O-glycan based panel defined in the present study could aid in discriminating SSA/P from HP to devise better colon surveillance strategies.


Molecular Oncology | 2016

The canonical Wnt pathway regulates the metastasis-promoting mucin MUC4 in pancreatic ductal adenocarcinoma.

Priya Pai; Satyanarayana Rachagani; Imayavaramban Lakshmanan; Muzafar A. Macha; Yuri Sheinin; Lynette M. Smith; Moorthy P. Ponnusamy; Surinder K. Batra

Aberrant Wnt signaling frequently occurs in pancreatic cancer (PC) and contributes to disease progression/metastases. Likewise, the transmembrane‐mucin MUC4 is expressed de novo in early pancreatic intraepithelial neoplasia (PanINs) and incrementally increases with PC progression, contributing to metastasis. To determine the mechanism of MUC4 upregulation in PC, we examined factors deregulated in early PC progression, such as Wnt/β‐catenin signaling. MUC4 promoter analysis revealed the presence of three putative TCF/LEF‐binding sites, leading us to hypothesize that MUC4 can be regulated by β‐catenin. Immunohistochemical (IHC) analysis of rapid autopsy PC tissues showed a correlation between MUC4 and cytosolic/nuclear β‐catenin expression. Knock down (KD) of β‐catenin in CD18/HPAF and T3M4 cell lines resulted in decreased MUC4 transcript and protein. Three MUC4 promoter luciferase constructs, p3778, p3000, and p2700, were generated. The construct p3778, encompassing the entire MUC4 promoter, elicited increased luciferase activity in the presence of stabilized β‐catenin. Mutation of the TCF/LEF site closest to the transcription start site (i.e., −2629/−2612) and furthest from the start site (i.e., −3425/−3408) reduced MUC4 promoter luciferase activity. Transfection with dominant negative TCF4 decreased MUC4 transcript and protein levels. Chromatin immunoprecipitation confirmed enrichment of β‐catenin on −2629/−2612 and −3425/−3408 of the MUC4 promoter in CD18/HPAF. Functionally, CD18/HPAF and T3M4 β‐catenin KD cells showed decreased migration and decreased Vimentin, N‐cadherin, and pERK1/2 expression. Tumorigenicity studies in athymic nude mice showed CD18/HPAF β‐catenin KD cells significantly reduced primary tumor sizes and metastases compared to scrambled control cells. We show for the first time that β‐catenin directly governs MUC4 in PC.


Genes & Cancer | 2016

MUC4 is negatively regulated through the Wnt/β-catenin pathway via the Notch effector Hath1 in colorectal cancer.

Priya Pai; Satyanarayana Rachagani; Punita Dhawan; Yuri Sheinin; Muzafar A. Macha; Asif Khurshid Qazi; Seema Chugh; Moorthy P. Ponnusamy; Kavita Mallya; Ramesh Pothuraju; Surinder K. Batra

MUC4 is a transmembrane mucin lining the normal colonic epithelium. The aberrant/de novo over-expression of MUC4 is well documented in malignancies of the pancreas, ovary and breast. However, studies have reported the loss of MUC4 expression in the majority of colorectal cancers (CRCs). A MUC4 promoter analysis showed the presence of three putative TCF/LEF sites, implying a possible regulation by the Wnt/β-catenin pathway, which has been shown to drive CRC progression. Thus, the objective of our study was to determine whether MUC4 is regulated by β-catenin in CRC. We first knocked down (KD) β-catenin in three CRC cell lines; LS180, HCT-8 and HCT116, which resulted in increased MUC4 transcript and MUC4 protein. Additionally, the overexpression of stabilized mutant β-catenin in LS180 and HCT-8 resulted in a decrease in MUC4 expression. Immunohistochemistry (IHC) of mouse colon tissue harboring tubular adenomas and high grade dysplasia showed dramatically reduced Muc4 in lesions relative to adjacent normal tissue, with increased cytosolic/nuclear β-catenin. Luciferase assays with the complete MUC4 promoter construct p3778 showed increased MUC4 promoter luciferase activity in the absence of β-catenin (KD). Mutation of all three putative TCF/LEF sites showed that MUC4 promoter luciferase activity was increased relative to the un-mutated promoter. Interestingly, it was observed that MUC4 expressing CRC cell lines also expressed high levels of Hath1, a transcription factor repressed by both active Wnt/β-catenin and Notch signaling. The KD of β-catenin and/or treatment with a Notch γ-secretase inhibitor, Dibenzazepine (DBZ) resulted in increased Hath1 and MUC4 in LS180, HCT-8 and HCT116. Furthermore, overexpression of Hath1 in HCT-8 and LS180 caused increased MUC4 transcript and MUC4 protein. Taken together, our results indicate that the Wnt/β-catenin pathway suppresses the Notch pathway effector Hath1, resulting in reduced MUC4 in CRC.


Oncogene | 2015

Erratum: MUC4 regulates cellular senescence in head and neck squamous cell carcinoma through p16/Rb pathway (Oncogene (2015) 34:2814 DOI:10.1038/onc.2015.116)

Macha Ma; Satyanarayana Rachagani; Priya Pai; Suprit Gupta; W. M. Lydiatt; Russell B. Smith; Sonny L. Johansson; Subodh M. Lele; Sham S. Kakar; Hanan Farghaly; John H. Lee; Jane L. Meza; Ganti Ak; Maneesh Jain; Surinder K. Batra

Correction to: Oncogene (2015) 34, 1698–1708; doi:10.1038/onc.2014.102; published online 21 April 2014 The authors wish to add H Farghaly, originally included in the Acknowledgements, to the authorship of this article, published online 21 April 2014. Dr Farghaly was omitted from the byline at final submission due to communication impediments outside the authors’ control during the manuscript review period which were shortly thereafter resolved.

Collaboration


Dive into the Priya Pai's collaboration.

Top Co-Authors

Avatar

Surinder K. Batra

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Satyanarayana Rachagani

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Maneesh Jain

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Muzafar A. Macha

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lynette M. Smith

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Moorthy P. Ponnusamy

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Subodh M. Lele

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

Suprit Gupta

University of Nebraska Medical Center

View shared research outputs
Top Co-Authors

Avatar

John H. Lee

University of South Dakota

View shared research outputs
Top Co-Authors

Avatar

Russell B. Smith

University of Nebraska Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge