Network


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

Hotspot


Dive into the research topics where Priya Chatterji is active.

Publication


Featured researches published by Priya Chatterji.


Molecular and Cellular Biology | 2015

The Poly(C) Binding Protein Pcbp2 and Its Retrotransposed Derivative Pcbp1 Are Independently Essential to Mouse Development

Louis R. Ghanem; Andrew Kromer; Ian M. Silverman; Priya Chatterji; Elizabeth A. Traxler; Alfredo I. Penzo-Méndez; Mitchell J. Weiss; Ben Z. Stanger; Stephen A. Liebhaber

ABSTRACT RNA-binding proteins participate in a complex array of posttranscriptional controls essential to cell type specification and somatic development. Despite their detailed biochemical characterizations, the degree to which each RNA-binding protein impacts mammalian embryonic development remains incompletely defined, and the level of functional redundancy among subsets of these proteins remains open to question. The poly(C) binding proteins, PCBPs (αCPs and hnRNP E proteins), are encoded by a highly conserved and broadly expressed gene family. The two major Pcbp isoforms, Pcbp2 and Pcbp1, are robustly expressed in a wide range of tissues and exert both nuclear and cytoplasmic controls over gene expression. Here, we report that Pcbp1-null embryos are rendered nonviable in the peri-implantation stage. In contrast, Pcbp2-null embryos undergo normal development until midgestation (12.5 to 13.5 days postcoitum), at which time they undergo a dramatic loss in viability associated with combined cardiovascular and hematopoietic abnormalities. Mice heterozygous for either Pcbp1 or Pcbp2 null alleles display a mild and nondisruptive defect in initial postpartum weight gain. These data reveal that Pcbp1 and Pcbp2 are individually essential for mouse embryonic development and have distinct impacts on embryonic viability and that Pcpb2 has a nonredundant in vivo role in hematopoiesis. These data further provide direct evidence that Pcbp1, a retrotransposed derivative of Pcpb2, has evolved an essential function(s) in the mammalian genome.


Molecular Cancer Research | 2015

Loss of Stromal IMP1 Promotes a Tumorigenic Microenvironment in the Colon.

Kathryn E. Hamilton; Priya Chatterji; Emma Lundsmith; Sarah F. Andres; Veronique Giroux; Philip D. Hicks; Felicite K. Noubissi; Vladimir S. Spiegelman; Anil K. Rustgi

The colon tumor microenvironment is becoming increasingly recognized as a complex but central player in the development of many cancers. Previously, we identified an oncogenic role for the mRNA-binding protein IMP1 (IGF2BP1) in the epithelium during colon tumorigenesis. In the current study, we reveal the contribution of stromal IMP1 in the context of colitis-associated colon tumorigenesis. Interestingly, stromal deletion of Imp1 (Dermo1Cre;Imp1LoxP/LoxP, or Imp1ΔMes) in the azoxymethane/dextran sodium sulfate (AOM/DSS) model of colitis-associated cancer resulted in increased tumor numbers of larger size and more advanced histologic grade than controls. In addition, Imp1ΔMes mice exhibited a global increase in protumorigenic microenvironment factors, including enhanced inflammation and stromal components. Evaluation of purified mesenchyme from AOM/DSS-treated Imp1ΔMes mice demonstrated an increase in hepatocyte growth factor (HGF), which has not been associated with regulation via IMP1. Genetic knockdown of Imp1 in human primary fibroblasts confirmed an increase in HGF with Imp1 loss, demonstrating a specific, cell-autonomous role for Imp1 loss to increase HGF expression. Taken together, these data demonstrate a novel tumor-suppressive role for IMP1 in colon stromal cells and underscore an exquisite, context-specific function for mRNA-binding proteins, such as IMP1, in disease states. Implications: The tumor-suppressive role of stromal IMP1 and its ability to modulate protumorigenic factors suggest that IMP1 status is important for the initiation and growth of epithelial tumors. Mol Cancer Res; 13(11); 1478–86. ©2015 AACR. See related article by Koltsova and Grivennikov, p. 1452


Gene Expression Patterns | 2014

Specific enrichment of the RNA-binding proteins PCBP1 and PCBP2 in chief cells of the murine gastric mucosa.

Louis R. Ghanem; Priya Chatterji; Stephen A. Liebhaber

RNA-binding proteins and corresponding post-transcriptional controls play critical roles in gene expression. The poly-(C) binding proteins, PCBPs (αCPs, hnRNPEs), comprise a well-characterized family of abundant RNA-binding proteins that impact on RNA processing in the nucleus as well as mRNA stability and translation in the cytoplasm. Here we demonstrate that PCBP1 and PCBP2 are abundantly expressed in the gastric epithelium with prominent enrichment in specific cell types within the gastric glandular mucosa. The spatial and intracellular patterns of PCBP1 and PCBP2 expression in these regions are highly correlated. Remarkably, we observe that these proteins are present in the nuclear and cytoplasmic compartments of zymogenic chief cells while they are restricted to the nuclear compartment in acid-secreting parietal cells and poorly expressed in pit cells that line the gland exit. This specificity of expression patterns and subcellular localization of PCBP1 and PCBP2, along with their appearance in the precursor tissues of the gastric epithelium during early postnatal development, suggests these RNA-binding proteins play specific roles in cell differentiation and organismal development within the gastric glandular epithelium.


Trends in Molecular Medicine | 2018

RNA Binding Proteins in Intestinal Epithelial Biology and Colorectal Cancer

Priya Chatterji; Anil K. Rustgi

The intestinal epithelium is highly proliferative and consists of crypt invaginations that house stem cells and villus projections with differentiated cells. There exists a dynamic equilibrium between proliferation, migration, differentiation, and senescence that is regulated by several factors. Among these are RNA binding proteins (RBPs) that bind their targets in a both context dependent and independent manner. RBP-RNA complexes act as rheostats by regulating expression of RNAs both co- and post-transcriptionally. This is important, especially in response to intestinal injury, to fuel regeneration. The manner in which these RBPs function in the intestine and their interactions with other pivotal pathways in colorectal cancer may provide a framework for new insights and potential therapeutic applications.


Molecular Cancer Research | 2018

Differential Regulation of LET-7 by LIN28B Isoform–Specific Functions

Rei Mizuno; Priya Chatterji; Sarah F. Andres; Kathryn E. Hamilton; Lauren Simon; Shawn W. Foley; Arjun N. Jeganathan; Brian D. Gregory; Blair B. Madison; Anil K. Rustgi

The RNA-binding protein LIN28B plays an important role in development, stem cell biology, and tumorigenesis. LIN28B has two isoforms: the LIN28B-long and -short isoforms. Although studies have revealed the functions of the LIN28B-long isoform in tumorigenesis, the role of the LIN28B-short isoform remains unclear and represents a major gap in the field. The LIN28B-long and -short isoforms are expressed in a subset of human colorectal cancers and adjacent normal colonic mucosa, respectively. To elucidate the functional and mechanistic aspects of these isoforms, colorectal cancer cells (Caco-2 and LoVo) were generated to either express no LIN28B or the -short or -long isoform. Interestingly, the long isoform suppressed LET-7 expression and activated canonical RAS/ERK signaling, whereas the short isoform did not. The LIN28B-long isoform–expressing cells demonstrated increased drug resistance to 5-fluorouracil and cisplatin through the upregulation of ERCC1, a DNA repair gene, in a LET-7–dependent manner. The LIN28B-short isoform preserved its ability to bind pre-let-7, without inhibiting the maturation of LET-7, and competed with the LIN28B-long isoform for binding to pre-let-7. Coexpression of the short isoform in the LIN28B-long isoform–expressing cells rescued the phenotypes induced by the LIN28B-long isoform. Implications: This study demonstrates the differential antagonistic functions of the LIN28B-short isoform against the LIN28B-long isoform through an inability to degrade LET-7, which leads to the novel premise that the short isoform may serve to counterbalance the long isoform during normal colonic epithelial homeostasis, but its downregulation during colonic carcinogenesis may reveal the protumorigenic effects of the long isoform. Mol Cancer Res; 16(3); 403–16. ©2018 AACR.


Cellular and molecular gastroenterology and hepatology | 2017

Pancreas 3D Organoids: Current and Future Aspects as a Research Platform for Personalized Medicine in Pancreatic Cancer

Leticia Moreira; Basil Bakir; Priya Chatterji; Zahra Dantes; Maximilian Reichert; Anil K. Rustgi

Pancreatic ductal adenocarcinoma is one of the most aggressive forms of cancer, and the third leading cause of cancer-related mortality in the United States. Although important advances have been made in the last decade, the mortality rate of pancreatic ductal adenocarcinoma has not changed appreciably. This review summarizes a rapidly emerging model of pancreatic cancer research, focusing on 3-dimensional organoids as a powerful tool for several applications, but above all, representing a step toward personalized medicine.


bioRxiv | 2018

Posttranscriptional regulation of intestinal epithelial cell repair by RNA binding protein IMP1

Priya Chatterji; Kelly A. Whelan; Sarah F. Andres; Fernando Cid Samper; Lauren A Simon; Rei Mizuno; Emma T. Lundsmith; David S.M. Lee; Shun Liang; H.R. Sagara Wijeratne; Stefanie Marti; Lillian Chau; Patrick A Williams; Veronique Giroux; Benjamin J. Wilkins; Gary D. Wu; Premal Shah; Gian Gaetano Tartaglia; Kathryn E Hamilton

RNA binding proteins, such as IMP1, are emerging as essential regulators of intestinal development and cancer. IMP1 hypomorphic mice exhibit severe intestinal growth defects, yet it’s role in adult intestinal epithelium is unclear. We employed ribosome profiling to test the effect of IMP1 loss on the “translatome” in colon cancer cell lines. In parallel, we evaluated mice with intestinal epithelial-specific Imp1 deletion (Imp1ΔIEC) following irradiation or colitis models. Ribosome-profiling revealed translation efficiency changes for multiple pathways important for intestinal homeostasis, including autophagy, in IMP1 knockout cells. We found increased autophagy flux in Imp1ΔIEC mice, reinforced through in silico and biochemical analyses revealing direct binding of IMP1 to autophagy transcripts MAP1LC3B and ATG3. We found that Imp1ΔIEC mice exhibit enhanced recovery following irradiation, which is attenuated with genetic deletion of autophagy gene Atg7. Finally, we demonstrated that IMP1 is upregulated in Crohn’s disease patients and Imp1 loss lessened colitis severity in mice. These studies demonstrate that IMP1 acts as a posttranscriptional regulator of gut epithelial repair post-irradiation and colitis, in part through modulation of autophagy.


Stem cell reports | 2018

Mouse Intestinal Krt15+ Crypt Cells Are Radio-Resistant and Tumor Initiating

Veronique Giroux; Julien Stephan; Priya Chatterji; Ben Rhoades; E. Paul Wileyto; Andres J. Klein-Szanto; Christopher J. Lengner; Kathryn E. Hamilton; Anil K. Rustgi

Summary Two principal stem cell pools orchestrate the rapid cell turnover in the intestinal epithelium. Rapidly cycling Lgr5+ stem cells are intercalated between the Paneth cells at the crypt base (CBCs) and injury-resistant reserve stem cells reside above the crypt base. The intermediate filament Keratin 15 (Krt15) marks either stem cells or long-lived progenitor cells that contribute to tissue repair in the hair follicle or the esophageal epithelium. Herein, we demonstrate that Krt15 labels long-lived and multipotent cells in the small intestinal crypt by lineage tracing. Krt15+ crypt cells display self-renewal potential in vivo and in 3D organoid cultures. Krt15+ crypt cells are resistant to high-dose radiation and contribute to epithelial regeneration following injury. Notably, loss of the tumor suppressor Apc in Krt15+ cells leads to adenoma and adenocarcinoma formation. These results indicate that Krt15 marks long-lived, multipotent, and injury-resistant crypt cells that may function as a cell of origin in intestinal cancer.


Genes & Development | 2018

The LIN28B–IMP1 post-transcriptional regulon has opposing effects on oncogenic signaling in the intestine

Priya Chatterji; Kathryn E. Hamilton; Shun Liang; Sarah F. Andres; H.R. Sagara Wijeratne; Rei Mizuno; Lauren Simon; Philip D. Hicks; Shawn W. Foley; Jason R. Pitarresi; Andres J. Klein-Szanto; Amanda T. Mah; Laurianne Van Landeghem; Brian D. Gregory; Christopher J. Lengner; Blair B. Madison; Premal Shah; Anil K. Rustgi

RNA-binding proteins (RBPs) are expressed broadly during both development and malignant transformation, yet their mechanistic roles in epithelial homeostasis or as drivers of tumor initiation and progression are incompletely understood. Here we describe a novel interplay between RBPs LIN28B and IMP1 in intestinal epithelial cells. Ribosome profiling and RNA sequencing identified IMP1 as a principle node for gene expression regulation downstream from LIN28B In vitro and in vivo data demonstrate that epithelial IMP1 loss increases expression of WNT target genes and enhances LIN28B-mediated intestinal tumorigenesis, which was reversed when we overexpressed IMP1 independently in vivo. Furthermore, IMP1 loss in wild-type or LIN28B-overexpressing mice enhances the regenerative response to irradiation. Together, our data provide new evidence for the opposing effects of the LIN28B-IMP1 axis on post-transcriptional regulation of canonical WNT signaling, with implications in intestinal homeostasis, regeneration and tumorigenesis.


Inflammatory Bowel Diseases | 2016

O-010 Novel Regulation of Autophagy and Intestinal Homeostasis Via mRNA Binding Protein IMP1.

Kathryn E. Hamilton; Priya Chatterji; Sarah F. Andres; Emma Lundsmith; Kelly A. Whelan; Rei Mizuno; Veronique Giroux; Amanda Mah; Lillian Chua; Philip D. Hicks; Laurianne Van Landeghem; Pauline Kay Lund; Gary D. Wu; Anil K. Rustgi

Background:IMP1 (Insulin-like growth factor-2 mRNA binding protein 1) is essential for normal gut development and aberrant overexpression promotes colorectal tumors; however, the role of IMP1 in epithelial homeostasis in the adult intestine remains unclear. Our preliminary findings suggest that Imp1 loss may alter autophagy in intestinal epithelium during homeostasis and response to injury. Recent studies have linked aberrations in autophagy to Crohns disease. We therefore sought to determine if Imp1-mediated changes in autophagy may affect response to injury in the intestine epithelium and whether Imp1 expression is altered in Crohns disease patients. Methods:Mice with intestine-epithelial specific Imp1 deletion (VillinCre;Imp1fl/fl) were used to evaluate autophagy flux and response to irradiation or Heligmosomoides polygyrus infection via gene expression analyses, flow cytometry, and IHC/IF. Crypt enteroid assays were utilized to evaluate stem cell growth. Imp1 expression in Crohns disease patients was evaluated via qRT-PCR. Results:Imp1 expression is enriched in the crypt region of the small intestine, and VillinCre;Imp1fl/fl mice exhibit an increase in autophagy flux and enhanced expression of Paneth and stem cell markers in isolated crypts. Following challenge with irradiation or Heligmosomoides polygyrus infection, VillinCre;Imp1fl/fl mice exhibit robust crypt enteroid growth and improved clinical parameters consistent with Imp1 loss being protective in these contexts. Analysis of tissue biopsies from Crohns disease patients reveal a significant upregulation of Imp1 compared to unaffected patients, suggesting the possibility that overexpression of Imp1 may contribute to autophagy-related pathogenesis in Crohns disease. Conclusions:Our data demonstrate in 2 independent models that intestinal epithelial deletion of Imp1 promotes enhanced recovery from injury, possibly due to upregulation of stem cell gene expression and autophagy. Furthermore, our data reveal for the first time that Imp1 expression is increased in tissue from Crohns disease patients. Taken together, our findings may suggest a novel mechanism for IMP1 to promote pathogenesis of Crohns disease via negative regulation of autophagy.

Collaboration


Dive into the Priya Chatterji's collaboration.

Top Co-Authors

Avatar

Anil K. Rustgi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sarah F. Andres

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Rei Mizuno

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Philip D. Hicks

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Emma Lundsmith

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Laurianne Van Landeghem

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Amanda Mah

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Kelly A. Whelan

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Lauren Simon

University of Pennsylvania

View shared research outputs
Researchain Logo
Decentralizing Knowledge