2019
DOI: 10.1101/835793
|View full text |Cite
Preprint
|
Sign up to set email alerts
|

Paradoxical role of AT-rich interactive domain 1A in restraining pancreatic carcinogenesis

Abstract: Background & AimsARID1A is postulated to be a tumor suppressor gene owing to loss-of-function mutations in human pancreatic ductal adenocarcinomas (PDAC). However, its role in pancreatic pathogenesis is not clear despite recent studies using genetically engineered mouse (GEM) models. We aimed at further understanding of its direct functional role in PDAC, using a combination of GEM model, PDAC cell lines.MethodsPancreas-specific mutant Arid1a-driven GEM model (Ptf1a-Cre;KrasG12D;Arid1af/f or “KAC”) was gen… Show more

Help me understand this report
View published versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

0
6
0

Year Published

2020
2020
2022
2022

Publication Types

Select...
2
1
1

Relationship

0
4

Authors

Journals

citations
Cited by 4 publications
(6 citation statements)
references
References 67 publications
0
6
0
Order By: Relevance
“…Pancreatic ductal adenocarcinoma (PDAC), generally known as pancreatic cancer (PC), ranks the fourth leading cause of cancer-related deaths in the Western world ( 57 ). Recent integrated multiplatform sequencing analyses of PDAC have revealed ARID1A mutations in 6% of the cases ( 58 , 59 ). ARID1A may function as a tumor-suppressor gene in pancreatic carcinogenesis, because its expression levels are associated with tumor differentiation and tumor stage, but not to lymph node metastasis, distant metastasis, sex, or age ( 60 ).…”
Section: Tumor Suppression By Arid1amentioning
confidence: 99%
See 1 more Smart Citation
“…Pancreatic ductal adenocarcinoma (PDAC), generally known as pancreatic cancer (PC), ranks the fourth leading cause of cancer-related deaths in the Western world ( 57 ). Recent integrated multiplatform sequencing analyses of PDAC have revealed ARID1A mutations in 6% of the cases ( 58 , 59 ). ARID1A may function as a tumor-suppressor gene in pancreatic carcinogenesis, because its expression levels are associated with tumor differentiation and tumor stage, but not to lymph node metastasis, distant metastasis, sex, or age ( 60 ).…”
Section: Tumor Suppression By Arid1amentioning
confidence: 99%
“…ARID1A may function as a tumor-suppressor gene in pancreatic carcinogenesis, because its expression levels are associated with tumor differentiation and tumor stage, but not to lymph node metastasis, distant metastasis, sex, or age ( 60 ). In a pancreas-specific mutant Arid1a -driven mouse model ( Ptf1a-Cre;Kras G12D ;Arid1a f/f ), despite early development of relatively indolent cystic precursor lesions called intraductal papillary mucinous neoplasms (IPMNs), deficiency of Arid1a in mice pancreas develops aggressive PDAC in later ages with extensive parenchymal replacement by mucinous cysts resembling low-grade branched duct gastric type IPMN (LG-IPMN) in humans ( 59 , 61 ), suggesting Arid1a loss accelerates PDAC formation.…”
Section: Tumor Suppression By Arid1amentioning
confidence: 99%
“…However, even with long-term (60 day) 17b-estradiol treatment, endometrial cancer was not discovered, suggesting that something molecular may be restraining cancer in the endometrium rather than the vagina. Similarly, in the pancreatic cancer field, oncogenic Kras was found to be insufficient to drive pancreatic adenocarcinoma due to the protective effects of ARID1A [42][43][44][45][46][47]. For example, oncogenic expression of Kras G12D alone leads to pancreatic intraepithelial lesions but not malignancy.…”
Section: Discussionmentioning
confidence: 99%
“…These studies all support the hypothesis that ARID1A restrains malignant transformation from benign lesions driven by oncogenic Kras G12D . Work in pancreatic adenocarcinoma [42][43][44][45][46][47] has identified escape mechanisms that need to be explored in the endometrium.…”
Section: Discussionmentioning
confidence: 99%
“…While the functional implications of Arid1a deficiency in Kras-driven pancreatic carcinogenesis have been studied intensively, findings characterizing the consequences of sole Arid1a deficiency in the absence of oncogenic Kras are conflicting (Wang S.C et al 2019;Livshits et al 2018;Ferri-Borgogno et al 2019). We propose, that Arid1a deficiency critically impacts on acinar reprogramming and hypothesize that the chromatin remodeling protein´s function and activity is critically influenced by the activity of signaling pathways which also play a pivotal role in acinar cell plasticity and pancreatic carcinogenesis.…”
Section: Aims Of the Studymentioning
confidence: 99%