Network


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

Hotspot


Dive into the research topics where Brian DeCant is active.

Publication


Featured researches published by Brian DeCant.


Cancer Research | 2016

TGFβ signaling in the pancreatic tumor microenvironment promotes fibrosis and immune evasion to facilitate tumorigenesis

Daniel R. Principe; Brian DeCant; Emman Mascariñas; Elizabeth A. Wayne; Andrew M. Diaz; Naomi Akagi; Rosa F. Hwang; Boris Pasche; David W. Dawson; Deyu Fang; David J. Bentrem; Hidayatullah G. Munshi; Barbara Jung; Paul J. Grippo

In early pancreatic carcinogenesis, TGFβ acts as a tumor suppressor due to its growth-inhibitory effects in epithelial cells. However, in advanced disease, TGFβ appears to promote tumor progression. Therefore, to better understand the contributions of TGFβ signaling to pancreatic carcinogenesis, we generated mouse models of pancreatic cancer with either epithelial or systemic TGFBR deficiency. We found that epithelial suppression of TGFβ signals facilitated pancreatic tumorigenesis, whereas global loss of TGFβ signaling protected against tumor development via inhibition of tumor-associated fibrosis, stromal TGFβ1 production, and the resultant restoration of antitumor immune function. Similarly, TGFBR-deficient T cells resisted TGFβ-induced inactivation ex vivo, and adoptive transfer of TGFBR-deficient CD8(+) T cells led to enhanced infiltration and granzyme B-mediated destruction of developing tumors. These findings paralleled our observations in human patients, where TGFβ expression correlated with increased fibrosis and associated negatively with expression of granzyme B. Collectively, our findings suggest that, despite opposing the proliferation of some epithelial cells, TGFβ may promote pancreatic cancer development by affecting stromal and hematopoietic cell function. Therefore, the use of TGFBR inhibition to target components of the tumor microenvironment warrants consideration as a potential therapy for pancreatic cancer, particularly in patients who have already lost tumor-suppressive TGFβ signals in the epithelium. Cancer Res; 76(9); 2525-39. ©2016 AACR.


Oncotarget | 2017

Loss of TGFβ signaling promotes colon cancer progression and tumor-associated inflammation.

Daniel R. Principe; Brian DeCant; Jonas J. Staudacher; Dominic Vitello; Riley J. Mangan; Elizabeth A. Wayne; Emman Mascariñas; Andrew M. Diaz; Jessica Bauer; Ronald D. McKinney; Khashayarsha Khazaie; Boris Pasche; David W. Dawson; Hidayatullah G. Munshi; Paul J. Grippo; Barbara Jung

TGFβ has both tumor suppressive and tumor promoting effects in colon cancer. Also, TGFβ can affect the extent and composition of inflammatory cells present in tumors, contextually promoting and inhibiting inflammation. While colon tumors display intratumoral inflammation, the contributions of TGFβ to this process are poorly understood. In human patients, we found that epithelial loss of TGFβ signaling was associated with increased inflammatory burden; yet overexpression of TGFβ was also associated with increased inflammation. These findings were recapitulated in mutant APC models of murine tumorigenesis, where epithelial truncation of TGFBR2 led to lethal inflammatory disease and invasive colon cancer, mediated by IL8 and TGFβ1. Interestingly, mutant APC mice with global suppression of TGFβ signals displayed an intermediate phenotype, presenting with an overall increase in IL8-mediated inflammation and accelerated tumor formation, yet with a longer latency to the onset of disease observed in mice with epithelial TGFBR-deficiency. These results suggest that the loss of TGFβ signaling, particularly in colon epithelial cells, elicits a strong inflammatory response and promotes tumor progression. This implies that treating colon cancer patients with TGFβ inhibitors may result in a worse outcome by enhancing inflammatory responses.


Pancreas | 2015

Characterization of Mouse Models of Early Pancreatic Lesions Induced by Alcohol and Chronic Pancreatitis.

Shiping Xu; Chintan Chheda; Yassine Ouhaddi; Hajar Benhaddou; Mouloud Bourhim; Paul J. Grippo; Daniel R. Principe; Emman Mascariñas; Brian DeCant; Hidekazu Tsukamoto; Stephen J. Pandol; Mouad Edderkaoui

Objective We describe the first mouse model of pancreatic intraepithelial neoplasia (PanIN) lesions induced by alcohol in the presence and absence of chronic pancreatitis. Methods Pdx1-Cre;LSL-K-ras mice were exposed to Lieber-DeCarli alcohol diet for 6 weeks with cerulein injections. The PanIN lesions and markers of fibrosis, inflammation, histone deacetylation, epithelial-to-mesenchymal transition (EMT), and cancer stemness were measured by immunohistochemistry and Western. Results Exposure of Pdx1-Cre;LSL-K-ras mice to an alcohol diet significantly stimulated fibrosis and slightly but not significantly increased the level of PanIN lesions associated with an increase in tumor-promoting M2 macrophages. Importantly, the alcohol diet did not increase activation of stellate cells. Alcohol diet and cerulein injections resulted in synergistic and additive effects on PanIN lesion and M2 macrophage phenotype induction, respectively. Cerulein pancreatitis caused stellate cell activation, EMT, and cancer stemness in the pancreas. Pancreatitis caused histone deacetylation, which was promoted by the alcohol diet. Pancreatitis increased EMT and cancer stemness markers, which were not further affected by the alcohol diet. Conclusions The results suggest that alcohol has independent effects on promotion of PDAC associated with fibrosis formed through a stellate cell-independent mechanism and that it further promotes early PDAC and M2 macrophage induction in the context of chronic pancreatitis.


Oncotarget | 2016

PEDF inhibits pancreatic tumorigenesis by attenuating the fibroinflammatory reaction

Daniel R. Principe; Brian DeCant; Andrew M. Diaz; Riley J. Mangan; Rosa F. Hwang; Andrew M. Lowy; Brandon B. Shetuni; Bharath K. Sreekumar; Chuhan Chung; David J. Bentrem; Hidayatullah G. Munshi; Barbara Jung; Paul J. Grippo; Faraz Bishehsari

Pancreatic cancer is characterized by a pronounced fibro-inflammatory reaction that has been shown to contribute to cancer progression. Previous reports have demonstrated that pigment epithelium-derived factor (PEDF) has potent tumor suppressive effects in pancreatic cancer, though little is known about the mechanisms by which PEDF limits pancreatic tumorigenesis. We therefore employed human specimens, as well as mouse and in vitro models, to explore the effects of PEDF upon the pancreatic microenvironment. We found that PEDF expression is decreased in human pancreatic cancer samples compared to non-malignant tissue. Furthermore, PEDF-deficient patients displayed increased intratumoral inflammation/fibrosis. In mice, genetic ablation of PEDF increased cerulein-induced inflammation and fibrosis, and similarly enhanced these events in the background of oncogenic KRAS. In vitro, recombinant PEDF neutralized macrophage migration as well as inhibited macrophage-induced proliferation of tumor cells. Additionally, recombinant PEDF suppressed the synthesis of pro-inflammatory/pro-fibrotic cytokines both in vivo and in vitro, and reduced collagen I deposition and TGFβ synthesis by pancreatic stellate cells, consistent with reduced fibrosis. Combined, our results demonstrate that PEDF limits pancreatic cancer progression by attenuating the fibro-inflammatory reaction, and makes restoration of PEDF signaling a potential therapeutic approach to study in pancreatic cancer.


Frontiers in Physiology | 2014

Utilizing past and present mouse systems to engineer more relevant pancreatic cancer models.

Brian DeCant; Daniel R. Principe; Carmen Guerra; Marina Pasca di Magliano; Paul J. Grippo

The study of pancreatic cancer has prompted the development of numerous mouse models that aim to recapitulate the phenotypic and mechanistic features of this deadly malignancy. This review accomplishes two tasks. First, it provides an overview of the models that have been used as representations of both the neoplastic and carcinoma phenotypes. Second, it presents new modeling schemes that ultimately will serve to more faithfully capture the temporal and spatial progression of the human disease, providing platforms for improved understanding of the role of non-epithelial compartments in disease etiology as well as evaluating therapeutic approaches.


JCI insight | 2017

BET inhibitors block pancreatic stellate cell collagen I production and attenuate fibrosis in vivo

Krishan Kumar; Brian DeCant; Paul J. Grippo; Rosa F. Hwang; David J. Bentrem; Kazumi Ebine; Hidayatullah G. Munshi

The fibrotic reaction, which can account for over 70%-80% of the tumor mass, is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of collagen I production and fibrosis in vivo. In this report, we show that members of the bromodomain and extraterminal (BET) family of proteins are expressed in primary PSCs isolated from human PDAC tumors, with BRD4 positively regulating, and BRD2 and BRD3 negatively regulating, collagen I expression in primary cancer-associated PSCs. We show that the inhibitory effect of pan-BET inhibitors on collagen I expression in primary cancer-associated PSCs is through blocking of BRD4 function. Importantly, we show that FOSL1 is repressed by BRD4 in primary cancer-associated PSCs and negatively regulates collagen I expression. While BET inhibitors do not affect viability or induce PSC apoptosis or senescence, BET inhibitors induce primary cancer-associated PSCs to become quiescent. Finally, we show that BET inhibitors attenuate stellate cell activation, fibrosis, and collagen I production in the EL-KrasG12D transgenic mouse model of pancreatic tumorigenesis. Our results demonstrate that BET inhibitors regulate fibrosis by modulating the activation and function of cancer-associated PSCs.


Oncotarget | 2017

Thioredoxin system-mediated regulation of mutant Kras associated pancreatic neoplasia and cancer

Michelle Schultz; Andrew M. Diaz; Sharon Smite; Anna R. Lay; Brian DeCant; Ronald D. McKinney; Windel E. Mascarinas; Yinglin Xia; Carola A. Neumann; David J. Bentrem; David W. Dawson; Paul J. Grippo

Peroxiredoxin-1 (Prdx1), a member of the thioredoxin (Txn) system, is overexpressed and correlates with poor prognosis in pancreatic cancer patients and can suppress Kras signaling through redox-mediated inhibition of ERK and AKT in lung and breast cancer. Its redox function is maintained by Txn and sulfiredoxin (Srxn), and its tumor promoting functions are activated by post-translational modification. We studied the role of the Txn system in pancreatic neoplasia and cancer by determining how it regulates the phosphorylation of Kras effectors and by determining its association with patient survival. We found that elevated Prdx1 nuclear localization significantly correlated with better patient survival. Our data also demonstrate that the expression of the Txn system is dysregulated, with elevated Prdx1 expression and significantly decreased Txn and Srxn expression in pancreatic lesions of targeted mutant Kras mouse models. This correlated with distinct differences in the interconversion of Prdx1 oligomers that affect its ability to regulate ERK and AKT phosphorylation. Our data also suggest that Prdx1 post-translational modification and oligomerization suppress Prdx1 mediated redox regulation of ERK phosphorylation. We observed distinct differences in Txn expression and in the ability of pTyr-Prdx1 to bind to pERK in a PanIN model of pancreatic neoplasia as compared to an IPMN model, indicating a distinct difference in the function of post-translationally modified Prdx1 in cells with less Txn expression. Modified Txn system function and post-translational regulation may therefore play a significant role in pancreatic tumorigenesis by altering Kras effector phosphorylation and inhibiting the tumor suppressive redox functions of Prdx1.


Scientific Reports | 2016

Slug inhibits pancreatic cancer initiation by blocking Kras-induced acinar-ductal metaplasia.

Kazumi Ebine; Christina R. Chow; Brian DeCant; Holly Z. Hattaway; Paul J. Grippo; Krishan Kumar; Hidayatullah G. Munshi

Cells in the pancreas that have undergone acinar-ductal metaplasia (ADM) can transform into premalignant cells that can eventually become cancerous. Although the epithelial-mesenchymal transition regulator Snail (Snai1) can cooperate with Kras in acinar cells to enhance ADM development, the contribution of Snail-related protein Slug (Snai2) to ADM development is not known. Thus, transgenic mice expressing Slug and Kras in acinar cells were generated. Surprisingly, Slug attenuated Kras-induced ADM development, ERK1/2 phosphorylation and proliferation. Co-expression of Slug with Kras also attenuated chronic pancreatitis-induced changes in ADM development and fibrosis. In addition, Slug attenuated TGF-α-induced acinar cell metaplasia to ductal structures and TGF-α-induced expression of ductal markers in ex vivo acinar explant cultures. Significantly, blocking the Rho-associated protein kinase ROCK1/2 in the ex vivo cultures induced expression of ductal markers and reversed the effects of Slug by inducing ductal structures. In addition, blocking ROCK1/2 activity in Slug-expressing Kras mice reversed the inhibitory effects of Slug on ADM, ERK1/2 phosphorylation, proliferation and fibrosis. Overall, these results increase our understanding of the role of Slug in ADM, an early event that can eventually lead to pancreatic cancer development.


Cancer Research | 2016

Abstract 4178: Pigs as a new weapon against cancer: Modeling solid tumors in porcine

Andrew M. Diaz; Daniel R. Principe; Brian DeCant; Paul J. Grippo; Laurie A. Rund; Lawrence B. Schook

Widely-used genetically modified mice have revolutionized the field of cancer research by providing reliable in vivo systems with similar, if not identical, molecular alterations. Furthermore, Cre/Lox technology has afforded greater tissue specificity, with many of the developing cancer phenotypes recapitulating those observed in humans. However, despite these strengths, there are a number of notable limitations when comparing mice to humans including small sample sizes, dramatic differences in physiology, and often dissimilar drug responses. Pig models would alleviate these shortcomings if they were to have a repertoire of gene signatures observed in human cancer, like those engineered in mice. Therefore, to achieve this goal, we have engineered a pig model with the Cre-responsive transgene encoding KRASG12D and TP53R167H. Hence, tissue-specific targeted Cre would generate simultaneous expression of mutant KRAS and p53 alleles to induce cellular changes leading to cancer. Pig pancreases were injected with adenovirus containing a cre expression vector (adenoCre) in an attempt to generate a model of pancreatic cancer. Initial injections of adenoCre into the parenchymal body of the pancreas led to the development of acinar-ductal metaplasia (ADM), though the clear predominant histotype was leiomyosarcoma immediately adjacent to the pancreas. Pancreatic ADM was characterized by concomitant localization of amylase (acinar cell marker) and CK19 (ductal cell marker) in the same pancreatic acinus, with an occasional few cells expressing both cell markers. These lesions were further characterized and exhibited increased proliferation via PCNA staining, as well the development of surrounding desmoplastic stroma (enhanced trichrome, vimentin, and aSMA staining; a marker of pancreatic stellate cells) with a robust inflammatory component (strong CD11b and CAE staining). Based on the lack of pancreas specificity, the experiment was repeated by injecting adenoCre into the main pancreatic duct. This 10-month-old pig appears relatively healthy after, which was expected considering this was a fully adult pig prior to manipulation. We will fully access this pig in 2 months to determine if strict pancreatic duct injection of adenoCre induces ADM, neoplastic disease, and/or ductal adenocarcinoma. Based on these encouraging findings, we anticipate having a robust model of pancreatic cancer in the pig, and plan to extend this study to generate models of other solid tumors with high rates of KRAS and p53 mutations. These porcine models may significantly impact preclinical studies, as diseases induced in pigs are likely more physiologically relevant to those in humans, with a potentially similar pharmacokinetic profile following drug treatment. Indeed, due to the size of the organs and volume of blood, samples can be more readily shared among multiple groups of investigators facilitating greater collaboration and expanding future research. Citation Format: Andrew Diaz, Daniel Principe, Brian DeCant, Paul J. Grippo, Laurie Rund, Lawrence Schook. Pigs as a new weapon against cancer: Modeling solid tumors in porcine. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4178.


Scientific Reports | 2018

Interplay between interferon regulatory factor 1 and BRD4 in the regulation of PD-L1 in pancreatic stellate cells

Kazumi Ebine; Krishan Kumar; Thao N. Pham; Mario A. Shields; Katharine A. Collier; Meng Shang; Brian DeCant; Raul Urrutia; Rosa F. Hwang; Sam Grimaldo; Daniel R. Principe; Paul J. Grippo; David J. Bentrem; Hidayatullah G. Munshi

The fibrotic reaction is a characteristic feature of human pancreatic ductal adenocarcinoma (PDAC) tumors. It is associated with activation and proliferation of pancreatic stellate cells (PSCs), which are key regulators of fibrosis in vivo. While there is increasing interest in the regulation of PD-L1 expression in cancer and immune cells, the expression and regulation of PD-L1 in other stromal cells, such as PSCs, has not been fully evaluated. Here we show that PSCs in vitro express higher PD-L1 mRNA and protein levels compared to the levels present in PDAC cells. We show that inhibitors targeting bromodomain and extra-terminal (BET) proteins and BRD4 knockdown decrease interferon-γ (IFN-γ)-induced PD-L1 expression in PSCs. We also show that c-MYC, one of the well-established targets of BET inhibitors, does not mediate IFN-γ-regulated PD-L1 expression in PSCs. Instead we show that interferon regulatory factor 1 (IRF1) mediates IFN-γ-induced PD-L1 expression in PSCs. Finally, while we show that BET inhibitors do not regulate IFN-γ-induced IRF1 expression in PSCs, BET inhibitors decrease binding of IRF1 and BRD4 to the PD-L1 promoter. Together, these results demonstrate the interplay between IRF1 and BRD4 in the regulation of PD-L1 in PSCs.

Collaboration


Dive into the Brian DeCant's collaboration.

Top Co-Authors

Avatar

Paul J. Grippo

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel R. Principe

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Andrew M. Diaz

University of Illinois at Chicago

View shared research outputs
Top Co-Authors

Avatar

Barbara Jung

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kazumi Ebine

Northwestern University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge