73 results on '"I, Rooman"'
Search Results
2. Setting up a PDXO platform of pancreatic cancer with spatial -omics characterization
- Author
-
E. Michiels, N. Messaoudi, Y. Heremans, P. Giron, T. Janssens, K. Frederix, S. Aerts, H. Reynaert, and I. Rooman
- Subjects
Hepatology ,Endocrinology, Diabetes and Metabolism ,Gastroenterology - Published
- 2021
- Full Text
- View/download PDF
3. PCN224 - TOWARDS CLINICAL ADOPTION OF REPURPOSED DRUGS IN ONCOLOGY: OVERCOMING REGULATORY AND MARKET ACCESS BARRIERS IN EUROPE
- Author
-
Isabelle Huys, L. Meheus, I. Rooman, and C. Verbaanderd
- Subjects
business.industry ,Health Policy ,Public Health, Environmental and Occupational Health ,Market access ,Public relations ,business - Published
- 2018
- Full Text
- View/download PDF
4. Proliferation and differentiation of adult rat pancreatic exocrine cells
- Author
-
I, Rooman
- Subjects
Vascular Endothelial Growth Factor A ,Lymphokines ,Cell Transplantation ,Vascular Endothelial Growth Factors ,Cell Differentiation ,Endothelial Growth Factors ,Rats ,Embryonic and Fetal Development ,Islets of Langerhans ,Gastrins ,Models, Animal ,Diabetes Mellitus ,Animals ,Humans ,Intercellular Signaling Peptides and Proteins ,Regeneration ,Pancreas ,Cell Division ,Cells, Cultured - Published
- 2003
5. Effect of vascular endothelial growth factor on growth and differentiation of pancreatic ductal epithelium
- Author
-
I, Rooman, F, Schuit, and L, Bouwens
- Subjects
Male ,Vascular Endothelial Growth Factor A ,Lymphokines ,Vascular Endothelial Growth Factors ,Pancreatic Ducts ,Epithelial Cells ,Endothelial Growth Factors ,Immunohistochemistry ,Epithelium ,Rats ,Islets of Langerhans ,Pancreatitis ,Animals ,Rats, Wistar ,Ligation ,Cell Division ,Cells, Cultured - Abstract
Endocrine and exocrine pancreatic morphogenesis is known to occur from ductal epithelium, but the factors that regulate this process are unknown. Vascular endothelial growth factor (VEGF)/vascular permeability factor has recently been reported to affect fetal islet ontogenesis. VEGF is an angiogenic factor with a growth-promoting effect that is thought to be restricted to vascular endothelial cells. We demonstrated that VEGF is also a mitogen for adult rat pancreatic duct epithelial cells in primary culture. VEGF supplementation to a serum-free culture medium increased the 5-bromo-2'-deoxyuridine-pulse labeling index of ductal cells more than 2-fold. Immunohistochemical staining and protein blots revealed that pancreatic duct cells express fetal liver kinase-1 high-affinity receptors for VEGF. In pancreatic tissue, immunohistochemistry shows that VEGF peptide is expressed in normal pancreatic islet cells. In duct ligation-induced acute pancreatitis, numerous inflammatory leukocytes containing VEGF were seen to infiltrate between hyperplastic ducts. In the latter model, islet neogenesis has previously been observed. Our data indicate the possibility that VEGF plays a role in the paracrine regulation of ductal growth and differentiation in vivo, eg, in pancreatitis. In vitro, however, VEGF did not induce endocrine differentiation of ductal cells, indicating that it is not the only factor required for the activation of islet neogenesis.
- Published
- 1997
6. A Single-Cell Atlas of the Murine Pancreatic Ductal Tree Identifies Novel Cell Populations With Potential Implications in Pancreas Regeneration and Exocrine Pathogenesis.
- Author
-
Fernández Á, Casamitjana J, Holguín-Horcajo A, Coolens K, Mularoni L, Guo L, Hartwig O, Düking T, Vidal N, Strickland LN, Pasquali L, Bailey-Lundberg JM, Rooman I, Wang YJ, and Rovira M
- Subjects
- Animals, Mice, Organoids, Pancreas, Exocrine pathology, Pancreas, Exocrine metabolism, Pancreatic Neoplasms pathology, Pancreatic Neoplasms metabolism, Cell Differentiation, Humans, RNA-Seq, Disease Models, Animal, Cell Lineage, Ceruletide, Pancreatic Ducts pathology, Pancreatic Ducts cytology, Pancreatic Ducts metabolism, Single-Cell Analysis, Regeneration
- Abstract
Background & Aims: Pancreatic ducts form an intricate network of tubules that secrete bicarbonate and drive acinar secretions into the duodenum. This network is formed by centroacinar cells, terminal, intercalated, intracalated ducts, and the main pancreatic duct. Ductal heterogeneity at the single-cell level has been poorly characterized; therefore, our understanding of the role of ductal cells in pancreas regeneration and exocrine pathogenesis has been hampered by the limited knowledge and unexplained diversity within the ductal network., Methods: We used single cell RNA sequencing to comprehensively characterize mouse ductal heterogeneity at single-cell resolution of the entire ductal epithelium from centroacinar cells to the main duct. Moreover, we used organoid cultures, injury models, and pancreatic tumor samples to interrogate the role of novel ductal populations in pancreas regeneration and exocrine pathogenesis., Results: We have identified the coexistence of 15 ductal populations within the healthy pancreas and characterized their organoid formation capacity and endocrine differentiation potential. Cluster isolation and subsequent culturing let us identify ductal cell populations with high organoid formation capacity and endocrine and exocrine differentiation potential in vitro, including a Wnt-responsive population, a ciliated population, and Flrt3
+ cells. Moreover, we have characterized the location of these novel ductal populations in healthy pancreas, chronic pancreatitis, and tumor samples. The expression of Wnt-responsive, interferon-responsive, and epithelial-to-mesenchymal transition population markers increases in chronic pancreatitis and tumor samples., Conclusions: In light of our discovery of previously unidentified ductal populations, we unmask potential roles of specific ductal populations in pancreas regeneration and exocrine pathogenesis. Thus, novel lineage-tracing models are needed to investigate ductal-specific populations in vivo., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
7. Resolution of Acinar Dedifferentiation Regulates Tissue Remodeling in Pancreatic Injury and Cancer Initiation.
- Author
-
Baldan J, Camacho-Roda J, Ballester M, Høj K, Kurilla A, Maurer HC, Arcila-Barrera S, Lin X, Pan Z, Castro JL, Mayorca-Guiliani AE, Rift CV, Hasselby J, Bouwens L, Lefebvre V, David CJ, Parnas O, DelGiorno KE, Erler JT, Rooman I, and Arnes L
- Subjects
- Animals, Mice, Humans, Pancreatitis pathology, Pancreatitis genetics, Pancreatitis metabolism, SOXC Transcription Factors genetics, SOXC Transcription Factors metabolism, Disease Models, Animal, Pancreas pathology, Pancreas metabolism, Cell Transformation, Neoplastic pathology, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Gene Expression Regulation, Neoplastic, Gene Expression Profiling, Carcinoma in Situ pathology, Carcinoma in Situ genetics, Carcinoma in Situ metabolism, Transcriptome, Pancreatic Neoplasms pathology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Acinar Cells pathology, Acinar Cells metabolism, Carcinoma, Pancreatic Ductal pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Metaplasia genetics, Metaplasia pathology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Cell Dedifferentiation, Ceruletide
- Abstract
Background & Aims: Acinar-to-ductal metaplasia (ADM) is crucial in the development of pancreatic ductal adenocarcinoma. However, our understanding of the induction and resolution of ADM remains limited. We conducted comparative transcriptome analyses to identify conserved mechanisms of ADM in mouse and human., Methods: We identified Sox4 among the top up-regulated genes. We validated the analysis by RNA in situ hybridization. We performed experiments in mice with acinar-specific deletion of Sox4 (Ptf1a: CreER; Rosa26
-LSL-YFPLSL-YFP ; Sox4fl/fl ) with and without an activating mutation in Kras (KrasLSL-G12D/+ ). Mice were given caerulein to induce pancreatitis. We performed phenotypic analysis by immunohistochemistry, tissue decellularization, and single-cell RNA sequencing., Results: We demonstrated that Sox4 is reactivated in ADM and pancreatic intraepithelial neoplasias. Contrary to findings in other tissues, Sox4 actually counteracts cellular dedifferentiation and helps maintain tissue homeostasis. Moreover, our investigations unveiled the indispensable role of Sox4 in the specification of mucin-producing cells and tuft-like cells from acinar cells. We identified Sox4-dependent non-cell-autonomous mechanisms regulating the stromal reaction during disease progression. Notably, Sox4-inferred targets are activated upon KRAS inactivation and tumor regression., Conclusions: Our results indicate that our transcriptome analysis can be used to investigate conserved mechanisms of tissue injury. We demonstrate that Sox4 restrains acinar dedifferentiation and is necessary for the specification of acinar-derived metaplastic cells in pancreatic injury and cancer initiation and is activated upon Kras ablation and tumor regression in mice. By uncovering novel potential strategies to promote tissue homeostasis, our findings offer new avenues for preventing the development of pancreatic ductal adenocarcinoma., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
8. Compartmentalized role of xCT in supporting pancreatic tumor growth, inflammation and mood disturbance in mice.
- Author
-
Lara O, Janssen P, Mambretti M, De Pauw L, Ates G, Mackens L, De Munck J, Walckiers J, Pan Z, Beckers P, Espinet E, Sato H, De Ridder M, Marks DL, Barbé K, Aerts JL, Hermans E, Rooman I, and Massie A
- Subjects
- Mice, Animals, Brain, Inflammation, Hippocampus, Neuroinflammatory Diseases, Pancreatic Neoplasms
- Abstract
xCT (Slc7a11), the specific subunit of the cystine/glutamate antiporter system x
c - , is present in the brain and on immune cells, where it is known to modulate behavior and inflammatory responses. In a variety of cancers -including pancreatic ductal adenocarcinoma (PDAC)-, xCT is upregulated by tumor cells to support their growth and spread. Therefore, we studied the impact of xCT deletion in pancreatic tumor cells (Panc02) and/or the host (xCT-/- mice) on tumor burden, inflammation, cachexia and mood disturbances. Deletion of xCT in the tumor strongly reduced tumor growth. Targeting xCT in the host and not the tumor resulted only in a partial reduction of tumor burden, while it did attenuate tumor-related systemic inflammation and prevented an increase in immunosuppressive regulatory T cells. The latter effect could be replicated by specific xCT deletion in immune cells. xCT deletion in the host or the tumor differentially modulated neuroinflammation. When mice were grafted with xCT-deleted tumor cells, hypothalamic inflammation was reduced and, accordingly, food intake improved. Tumor bearing xCT-/- mice showed a trend of reduced hippocampal neuroinflammation with less anxiety- and depressive-like behavior. Taken together, targeting xCT may have beneficial effects on pancreatic cancer-related comorbidities, beyond reducing tumor burden. The search for novel and specific xCT inhibitors is warranted as they may represent a holistic therapy in pancreatic cancer., Competing Interests: Declaration of Competing Interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2024
- Full Text
- View/download PDF
9. High-resolution and quantitative spatial analysis reveal intra-ductal phenotypic and functional diversification in pancreatic cancer.
- Author
-
Michiels E, Madhloum H, Van Lint S, Messaoudi N, Kunda R, Martens S, Giron P, Olsen C, Lefesvre P, Dusetti N, El Mohajer L, Tomasini R, Hawinkels LJ, Ahsayni F, Nicolle R, Arsenijevic T, Bouchart C, Van Laethem JL, and Rooman I
- Subjects
- Humans, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Phenotype, RNA, Messenger, Pancreatic Neoplasms pathology, Carcinoma, Pancreatic Ductal pathology
- Abstract
A 'classical' and a 'basal-like' subtype of pancreatic cancer have been reported, with differential expression of GATA6 and different dosages of mutant KRAS. We established in situ detection of KRAS point mutations and mRNA panels for the consensus subtypes aiming to project these findings to paraffin-embedded clinical tumour samples for spatial quantitative analysis. We unveiled that, next to inter-patient and intra-patient inter-ductal heterogeneity, intraductal spatial phenotypes exist with anti-correlating expression levels of GATA6 and KRAS
G12D . The basal-like mRNA panel better captured the basal-like cell states than widely used protein markers. The panels corroborated the co-existence of the classical and basal-like cell states in a single tumour duct with functional diversification, i.e. proliferation and epithelial-to-mesenchymal transition respectively. Mutant KRASG12D detection ascertained an epithelial origin of vimentin-positive cells in the tumour. Uneven spatial distribution of cancer-associated fibroblasts could recreate similar intra-organoid diversification. This extensive heterogeneity with functional cooperation of plastic tumour cells poses extra challenges to therapeutic approaches. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland., (© 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.)- Published
- 2024
- Full Text
- View/download PDF
10. Characterization of the long-term effects of lethal total body irradiation followed by bone marrow transplantation on the brain of C57BL/6 mice.
- Author
-
Janssen P, De Pauw L, Mambretti M, Lara O, Walckiers J, Mackens L, Rooman I, Guillaume B, De Ridder M, Ates G, and Massie A
- Subjects
- Humans, Adult, Mice, Animals, Aged, Neuroinflammatory Diseases, Mice, Inbred C57BL, Brain, Bone Marrow Transplantation, Whole-Body Irradiation adverse effects
- Abstract
Purpose: Total body irradiation (TBI) followed by bone marrow transplantation (BMT) is used in pre-clinical research to generate mouse chimeras that allow to study the function of a protein specifically on immune cells. Adverse consequences of irradiation on the juvenile body and brain are well described and include general fatigue, neuroinflammation, neurodegeneration and cognitive impairment. Yet, the long-term consequences of TBI/BMT performed on healthy adult mice have been poorly investigated., Material and Methods: We developed a robust protocol to achieve near complete bone marrow replacement in mice using 2x550cGy TBI and evaluated the impact of the procedure on their general health, mood disturbances, memory, brain atrophy, neurogenesis, neuroinflammation and blood-brain barrier (BBB) permeability 2 and/or 16 months post-BMT., Results: We found a persistent decrease in weight along with long-term impact on locomotion after TBI and BMT. Although the TBI/BMT procedure did not lead to anxiety- or depressive-like behavior 2- or 16-months post-BMT, long-term spatial memory of the irradiated mice was impaired. We also observed radiation-induced impaired neurogenesis and cortical microglia activation 2 months post-BMT. Moreover, higher levels of hippocampal IgG in aged BMT mice suggest an enhanced age-related increase in BBB permeability that could potentially contribute to the observed memory deficit., Conclusions: Overall health of the mice did not seem to be majorly impacted by TBI followed by BMT during adulthood. Yet, TBI-induced alterations in the brain and behavior could lead to erroneous conclusions on the function of a protein on immune cells when comparing mouse chimeras with different genetic backgrounds that might display altered susceptibility to radiation-induced damage. Ultimately, the BMT model we here present could also be used to study the related long-term consequences of TBI and BMT seen in patients.
- Published
- 2024
- Full Text
- View/download PDF
11. Heterotopia of salivary gland tissue in the pancreas.
- Author
-
Martens S, Coolens K, Olsen C, Lefesvre P, and Rooman I
- Subjects
- Male, Humans, Adult, Epithelial Cells, Mucins, Pancreas, Choristoma
- Abstract
Heterotopia of the salivary gland occurs mainly in the head and neck region of the human body, rarely in regions such as the rectum, but has never been demonstrated in the pancreas. Within a screening effort of pancreatic samples for detecting ΔNp63 expression, we discovered two pancreatic samples from a 35-year-old male showing salivary gland heterotopia. Immunohistochemical stainings were done for markers of healthy and neoplastic salivary glands and showed expression of calponin, CD142 and KRT14 but not of S100p, GFAP or CD117. A PAS-staining and Alcian Blue staining showed the presence of acid mucins. These staining patterns were consistent with non-neoplastic submandibular gland tissue comprised of abundant seromucous glands, basal cells and myoepithelial cells, all features typically absent in the pancreas. Also, no pancreatic islets of Langerhans were detected. We show for the first time that salivary gland heterotopia can occur at the location of the pancreas., (© 2023. BioMed Central Ltd., part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
12. Pancreatic acinar cell fate relies on system x C - to prevent ferroptosis during stress.
- Author
-
Pan Z, Van den Bossche JL, Rodriguez-Aznar E, Janssen P, Lara O, Ates G, Massie A, De Paep DL, Houbracken I, Mambretti M, and Rooman I
- Subjects
- Humans, Animals, Mice, Acinar Cells, Acute Disease, Reactive Oxygen Species, Glutamic Acid, Ferroptosis genetics, Pancreatitis genetics
- Abstract
Acinar cell dedifferentiation is one of the most notable features of acute and chronic pancreatitis. It can also be the initial step that facilitates pancreatic cancer development. In the present study, we further decipher the precise mechanisms and regulation using primary human cells and murine experimental models. Our RNAseq analysis indicates that, in both species, early acinar cell dedifferentiation is accompanied by multiple pathways related to cell survival that are highly enriched, and where SLC7A11 (xCT) is transiently upregulated. xCT is the specific subunit of the cystine/glutamate antiporter system x
C - . To decipher its role, gene silencing, pharmacological inhibition and a knock-out mouse model were used. Acinar cells with depleted or reduced xCT function show an increase in ferroptosis relating to lipid peroxidation. Lower glutathione levels and more lipid ROS accumulation could be rescued by the antioxidant N-acetylcysteine or the ferroptosis inhibitor ferrostatin-1. In caerulein-induced acute pancreatitis in mice, xCT also prevents lipid peroxidation in acinar cells. In conclusion, during stress, acinar cell fate seems to be poised for avoiding several forms of cell death. xCT specifically prevents acinar cell ferroptosis by fueling the glutathione pool and maintaining ROS balance. The data suggest that xCT offers a druggable tipping point to steer the acinar cell fate in stress conditions., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
13. Axon guidance genes control hepatic artery development.
- Author
-
Gannoun L, De Schrevel C, Belle M, Dauguet N, Achouri Y, Loriot A, Vanderaa C, Cordi S, Dili A, Heremans Y, Rooman I, Leclercq IA, Jacquemin P, Gatto L, and Lemaigre FP
- Subjects
- Animals, Mice, Bile Ducts, Morphogenesis, Gene Silencing, Hepatic Artery, Axon Guidance
- Abstract
Earlier data on liver development demonstrated that morphogenesis of the bile duct, portal mesenchyme and hepatic artery is interdependent, yet how this interdependency is orchestrated remains unknown. Here, using 2D and 3D imaging, we first describe how portal mesenchymal cells become organised to form hepatic arteries. Next, we examined intercellular signalling active during portal area development and found that axon guidance genes are dynamically expressed in developing bile ducts and portal mesenchyme. Using tissue-specific gene inactivation in mice, we show that the repulsive guidance molecule BMP co-receptor A (RGMA)/neogenin (NEO1) receptor/ligand pair is dispensable for portal area development, but that deficient roundabout 2 (ROBO2)/SLIT2 signalling in the portal mesenchyme causes reduced maturation of the vascular smooth muscle cells that form the tunica media of the hepatic artery. This arterial anomaly does not impact liver function in homeostatic conditions, but is associated with significant tissular damage following partial hepatectomy. In conclusion, our work identifies new players in development of the liver vasculature in health and liver regeneration., Competing Interests: Competing interests The authors declare no competing or financial interests., (© 2023. Published by The Company of Biologists Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
14. Multicellular Modelling of Difficult-to-Treat Gastrointestinal Cancers: Current Possibilities and Challenges.
- Author
-
Hakuno SK, Michiels E, Kuhlemaijer EB, Rooman I, Hawinkels LJAC, and Slingerland M
- Subjects
- Animals, Disease Models, Animal, Humans, Organoids pathology, Carcinoma, Pancreatic Ductal pathology, Gastrointestinal Neoplasms pathology, Gastrointestinal Neoplasms therapy, Pancreatic Neoplasms pathology
- Abstract
Cancers affecting the gastrointestinal system are highly prevalent and their incidence is still increasing. Among them, gastric and pancreatic cancers have a dismal prognosis (survival of 5-20%) and are defined as difficult-to-treat cancers. This reflects the urge for novel therapeutic targets and aims for personalised therapies. As a prerequisite for identifying targets and test therapeutic interventions, the development of well-established, translational and reliable preclinical research models is instrumental. This review discusses the development, advantages and limitations of both patient-derived organoids (PDO) and patient-derived xenografts (PDX) for gastric and pancreatic ductal adenocarcinoma (PDAC). First and next generation multicellular PDO/PDX models are believed to faithfully generate a patient-specific avatar in a preclinical setting, opening novel therapeutic directions for these difficult-to-treat cancers. Excitingly, future opportunities such as PDO co-cultures with immune or stromal cells, organoid-on-a-chip models and humanised PDXs are the basis of a completely new area, offering close-to-human models. These tools can be exploited to understand cancer heterogeneity, which is indispensable to pave the way towards more tumour-specific therapies and, with that, better survival for patients.
- Published
- 2022
- Full Text
- View/download PDF
15. On the Origin of Pancreatic Cancer: Molecular Tumor Subtypes in Perspective of Exocrine Cell Plasticity.
- Author
-
Backx E, Coolens K, Van den Bossche JL, Houbracken I, Espinet E, and Rooman I
- Subjects
- Acinar Cells pathology, Cell Plasticity, Humans, Carcinoma, Pancreatic Ductal pathology, Pancreatic Neoplasms pathology
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating type of cancer. While many studies have shed light into the pathobiology of PDAC, the nature of PDAC's cell of origin remains under debate. Studies in adult pancreatic tissue have unveiled a remarkable exocrine cell plasticity including transitional states, mostly exemplified by acinar to ductal cell metaplasia, but also with recent evidence hinting at duct to basal cell transitions. Single-cell RNA sequencing has further revealed intrapopulation heterogeneity among acinar and duct cells. Transcriptomic and epigenomic relationships between these exocrine cell differentiation states and PDAC molecular subtypes have started to emerge, suggesting different ontogenies for different tumor subtypes. This review sheds light on these diverse aspects with particular focus on studies with human cells. Understanding the "masked ball" of exocrine cells at origin of PDAC and leaving behind the binary acinar vs duct cell classification may significantly advance our insights in PDAC biology., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
16. MECOM permits pancreatic acinar cell dedifferentiation avoiding cell death under stress conditions.
- Author
-
Backx E, Wauters E, Baldan J, Van Bulck M, Michiels E, Heremans Y, De Paep DL, Kurokawa M, Goyama S, Bouwens L, Jacquemin P, Houbracken I, and Rooman I
- Subjects
- Animals, Cell Dedifferentiation, Disease Models, Animal, Humans, Mice, Signal Transduction, Acinar Cells metabolism, Cell Death genetics, MDS1 and EVI1 Complex Locus Protein metabolism, Oncogenes genetics
- Abstract
Maintenance of the pancreatic acinar cell phenotype suppresses tumor formation. Hence, repetitive acute or chronic pancreatitis, stress conditions in which the acinar cells dedifferentiate, predispose for cancer formation in the pancreas. Dedifferentiated acinar cells acquire a large panel of duct cell-specific markers. However, it remains unclear to what extent dedifferentiated acini differ from native duct cells and which genes are uniquely regulating acinar cell dedifferentiation. Moreover, most studies have been performed on mice since the availability of human cells is scarce. Here, we applied a non-genetic lineage tracing method of human pancreatic exocrine acinar and duct cells that allowed cell-type-specific gene expression profiling by RNA sequencing. Subsequent to this discovery analysis, one transcription factor that was unique for dedifferentiated acinar cells was functionally characterized. RNA sequencing analysis showed that human dedifferentiated acinar cells expressed genes in "Pathways of cancer" with a prominence of MECOM (EVI-1), a transcription factor that was not expressed by duct cells. During mouse embryonic development, pre-acinar cells also transiently expressed MECOM and in the adult mouse pancreas, MECOM was re-expressed when mice were subjected to acute and chronic pancreatitis, conditions in which acinar cells dedifferentiate. In human cells and in mice, MECOM expression correlated with and was directly regulated by SOX9. Mouse acinar cells that, by genetic manipulation, lose the ability to upregulate MECOM showed impaired cell adhesion, more prominent acinar cell death, and suppressed acinar cell dedifferentiation by limited ERK signaling. In conclusion, we transcriptionally profiled the two major human pancreatic exocrine cell types, acinar and duct cells, during experimental stress conditions. We provide insights that in dedifferentiated acinar cells, cancer pathways are upregulated in which MECOM is a critical regulator that suppresses acinar cell death by permitting cellular dedifferentiation., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
17. Discovery and 3D imaging of a novel ΔNp63-expressing basal cell type in human pancreatic ducts with implications in disease.
- Author
-
Martens S, Coolens K, Van Bulck M, Arsenijevic T, Casamitjana J, Fernandez Ruiz A, El Kaoutari A, Martinez de Villareal J, Madhloum H, Esni F, Heremans Y, Leuckx G, Heimberg H, Bouwens L, Jacquemin P, De Paep DL, In't Veld P, D'Haene N, Bouchart C, Dusetti N, Van Laethem JL, Waelput W, Lefesvre P, Real FX, Rovira M, and Rooman I
- Abstract
Objective: The aggressive basal-like molecular subtype of pancreatic ductal adenocarcinoma (PDAC) harbours a ΔNp63 (p40) gene expression signature reminiscent of a basal cell type. Distinct from other epithelia with basal tumours, ΔNp63
+ basal cells reportedly do not exist in the normal pancreas., Design: We evaluated ΔNp63 expression in human pancreas, chronic pancreatitis (CP) and PDAC. We further studied in depth the non-cancerous tissue and developed a three-dimensional (3D) imaging protocol (FLIP-IT, Fluorescence Light sheet microscopic Imaging of Paraffin-embedded or Intact Tissue) to study formalin-fixed paraffin-embedded samples at single cell resolution. Pertinent mouse models and HPDE cells were analysed., Results: In normal human pancreas, rare ΔNp63+ cells exist in ducts while their prevalence increases in CP and in a subset of PDAC. In non-cancer tissue, ΔNp63+ cells are atypical KRT19+ duct cells that overall lack SOX9 expression while they do express canonical basal markers and pertain to a niche of cells expressing gastrointestinal stem cell markers. 3D views show that the basal cells anchor on the basal membrane of normal medium to large ducts while in CP they exist in multilayer dome-like structures. In mice, ΔNp63 is not found in adult pancreas nor in selected models of CP or PDAC, but it is induced in organoids from larger Sox9low ducts. In HPDE, ΔNp63 supports a basal cell phenotype at the expense of a classical duct cell differentiation programme., Conclusion: In larger human pancreatic ducts, basal cells exist. ΔNp63 suppresses duct cell identity. These cells may play an important role in pancreatic disease, including PDAC ontogeny, but are not present in mouse models., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2021
- Full Text
- View/download PDF
18. Cystine-glutamate antiporter deletion accelerates motor recovery and improves histological outcomes following spinal cord injury in mice.
- Author
-
Sprimont L, Janssen P, De Swert K, Van Bulck M, Rooman I, Gilloteaux J, Massie A, and Nicaise C
- Subjects
- Animals, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Motor Neurons cytology, Amino Acid Transport System y+ physiology, Cystine metabolism, Glutamic Acid metabolism, Motor Neurons physiology, Recovery of Function, Spinal Cord Injuries physiopathology
- Abstract
xCT is the specific subunit of System xc-, an antiporter importing cystine while releasing glutamate. Although xCT expression has been found in the spinal cord, its expression and role after spinal cord injury (SCI) remain unknown. The aim of this study was to characterize the role of xCT on functional and histological outcomes following SCI induced in wild-type (xCT+/+) and in xCT-deficient mice (xCT-/-). In the normal mouse spinal cord, slc7a11/xCT mRNA was detected in meningeal fibroblasts, vascular mural cells, astrocytes, motor neurons and to a lesser extent in microglia. slc7a11/xCT gene and protein were upregulated within two weeks post-SCI. xCT-/- mice recovered muscular grip strength as well as pre-SCI weight faster than xCT+/+ mice. Histology of xCT-/- spinal cords revealed significantly more spared motor neurons and a higher number of quiescent microglia. In xCT-/- mice, inflammatory polarization shifted towards higher mRNA expression of ym1 and igf1 (anti-inflammatory) while lower levels of nox2 and tnf-a (pro-inflammatory). Although astrocyte polarization did not differ, we quantified an increased expression of lcn2 mRNA. Our results show that slc7a11/xCT is overexpressed early following SCI and is detrimental to motor neuron survival. xCT deletion modulates intraspinal glial activation by shifting towards an anti-inflammatory profile.
- Published
- 2021
- Full Text
- View/download PDF
19. Exploring new uses for existing drugs: innovative mechanisms to fund independent clinical research.
- Author
-
Verbaanderd C, Rooman I, and Huys I
- Subjects
- Humans, Public-Private Sector Partnerships, Financial Management, Pharmaceutical Preparations
- Abstract
Background: Finding new therapeutic uses for existing medicines could lead to safe, affordable and timely new treatment options for patients with high medical needs. However, due to a lack of economic incentives, pharmaceutical developers are rarely interested to invest in research with approved medicines, especially when they are out of basic patent or regulatory protection. Consequently, potential new uses for these medicines are mainly studied in independent clinical trials initiated and led by researchers from academia, research institutes, or collaborative groups. Yet, additional financial support is needed to conduct expensive phase III clinical trials to confirm the results from exploratory research., Methods: In this study, scientific and grey literature was searched to identify and evaluate new mechanisms for funding clinical trials with repurposed medicines. Semi-structured interviews were conducted with 16 European stakeholders with expertise in clinical research, funding mechanisms and/or drug repurposing between November 2018 and February 2019 to consider the future perspectives of applying new funding mechanisms., Results: Traditional grant funding awarded by government and philanthropic organisations or companies is well known and widely implemented in all research fields. In contrast, only little research has focused on the application potential of newer mechanisms to fund independent clinical research, such as social impact bonds, crowdfunding or public-private partnerships. Interviewees stated that there is a substantial need for additional financial support in health research, especially in areas where there is limited commercial interest. However, the implementation of new funding mechanisms is facing several practical and financial challenges, such as a lack of expertise and guidelines, high transaction costs and difficulties to measure health outcomes. Furthermore, interviewees highlighted the need for increased collaboration and centralisation at a European and international level to make clinical research more efficient and reduce the need for additional funding., Conclusions: New funding mechanisms to support clinical research may become more important in the future but the unresolved issues identified in the current study warrant further exploration.
- Published
- 2021
- Full Text
- View/download PDF
20. Aggressive PDACs Show Hypomethylation of Repetitive Elements and the Execution of an Intrinsic IFN Program Linked to a Ductal Cell of Origin.
- Author
-
Espinet E, Gu Z, Imbusch CD, Giese NA, Büscher M, Safavi M, Weisenburger S, Klein C, Vogel V, Falcone M, Insua-Rodríguez J, Reitberger M, Thiel V, Kossi SO, Muckenhuber A, Sarai K, Lee AYL, Backx E, Zarei S, Gaida MM, Rodríguez-Paredes M, Donato E, Yen HY, Eils R, Schlesner M, Pfarr N, Hackert T, Plass C, Brors B, Steiger K, Weichenhan D, Arda HE, Rooman I, Kopp JL, Strobel O, Weichert W, Sprick MR, and Trumpp A
- Subjects
- Carcinoma, Pancreatic Ductal mortality, Carcinoma, Pancreatic Ductal pathology, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, CpG Islands, Disease Progression, Disease Susceptibility, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Humans, Models, Biological, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Prognosis, Reproducibility of Results, Signal Transduction, Transcriptome, Tumor Microenvironment genetics, Carcinoma, Pancreatic Ductal etiology, Carcinoma, Pancreatic Ductal metabolism, DNA Methylation, Interferons metabolism, Pancreatic Neoplasms etiology, Pancreatic Neoplasms metabolism, Repetitive Sequences, Nucleic Acid
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by extensive desmoplasia, which challenges the molecular analyses of bulk tumor samples. Here we FACS-purified epithelial cells from human PDAC and normal pancreas and derived their genome-wide transcriptome and DNA methylome landscapes. Clustering based on DNA methylation revealed two distinct PDAC groups displaying different methylation patterns at regions encoding repeat elements. Methylation
low tumors are characterized by higher expression of endogenous retroviral transcripts and double-stranded RNA sensors, which lead to a cell-intrinsic activation of an interferon signature (IFNsign). This results in a protumorigenic microenvironment and poor patient outcome. Methylationlow /IFNsignhigh and Methylationhigh /IFNsignlow PDAC cells preserve lineage traits, respective of normal ductal or acinar pancreatic cells. Moreover, ductal-derived KrasG12D / Trp53-/- mouse PDACs show higher expression of IFNsign compared with acinar-derived counterparts. Collectively, our data point to two different origins and etiologies of human PDACs, with the aggressive Methylationlow /IFNsignhigh subtype potentially targetable by agents blocking intrinsic IFN signaling. SIGNIFICANCE: The mutational landscapes of PDAC alone cannot explain the observed interpatient heterogeneity. We identified two PDAC subtypes characterized by differential DNA methylation, preserving traits from normal ductal/acinar cells associated with IFN signaling. Our work suggests that epigenetic traits and the cell of origin contribute to PDAC heterogeneity. This article is highlighted in the In This Issue feature, p. 521 ., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
21. Nicotinamide combined with gemcitabine is an immunomodulatory therapy that restrains pancreatic cancer in mice.
- Author
-
Selvanesan BC, Meena K, Beck A, Meheus L, Lara O, Rooman I, and Gravekamp C
- Subjects
- Animals, Antimetabolites, Antineoplastic pharmacology, Deoxycytidine pharmacology, Deoxycytidine therapeutic use, Disease Models, Animal, Female, Humans, Mice, Niacinamide pharmacology, Vitamin B Complex pharmacology, Gemcitabine, Antimetabolites, Antineoplastic therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Deoxycytidine analogs & derivatives, Immunomodulation drug effects, Niacinamide therapeutic use, Vitamin B Complex therapeutic use
- Abstract
Background: Treatments for pancreatic ductal adenocarcinoma are poorly effective, at least partly due to the tumor's immune-suppressive stromal compartment. New evidence of positive effects on immune responses in the tumor microenvironment (TME), compelled us to test the combination of gemcitabine (GEM), a standard chemotherapeutic for pancreatic cancer, with nicotinamide (NAM), the amide form of niacin (vitamin B
3 ), in mice with pancreatic cancer., Methods: Various mouse tumor models of pancreatic cancer, that is, orthotopic Panc-02 and KPC (KrasG12D , p53R172H , Pdx1-Cre) grafts, were treated alternately with NAM and GEM for 2 weeks, and the effects on efficacy, survival, stromal architecture and tumor-infiltrating immune cells was examined by immunohistochemistry (IHC), flow cytometry, Enzyme-linked immunospot (ELISPOT), T cell depletions in vivo, Nanostring analysis and RNAscope., Results: A significant reduction in tumor weight and number of metastases was found, as well as a significant improved survival of the NAM+GEM group compared with all control groups. IHC and flow cytometry showed a significant decrease in tumor-associated macrophages and myeloid-derived suppressor cells in the tumors of NAM+GEM-treated mice. This correlated with a significant increase in the number of CD4 and CD8 T cells of NAM+GEM-treated tumors, and CD4 and CD8 T cell responses to tumor-associated antigen survivin, most likely through epitope spreading. In vivo depletions of T cells demonstrated the involvement of CD4 T cells in the eradication of the tumor by NAM+GEM treatment. In addition, remodeling of the tumor stroma was observed with decreased collagen I and lower expression of hyaluronic acid binding protein, reorganization of the immune cells into lymph node like structures and CD31 positive vessels. Expression profiling for a panel of immuno-oncology genes revealed significant changes in genes involved in migration and activation of T cells, attraction of dendritic cells and epitope spreading., Conclusion: This study highlights the potential of NAM+GEM as immunotherapy for advanced pancreatic cancer., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)- Published
- 2020
- Full Text
- View/download PDF
22. Pancreatic resection with perioperative drug repurposing of propranolol and etodolac: trial protocol of the phase-II randomised placebo controlled PROSPER trial.
- Author
-
Hüttner FJ, Rooman I, Bouche G, Knebel P, Hüsing J, Mihaljevic AL, Hackert T, Strobel O, Büchler MW, and Diener MK
- Subjects
- Adult, Clinical Trials, Phase II as Topic, Drug Repositioning, Humans, Pancreatectomy, Prospective Studies, Randomized Controlled Trials as Topic, Etodolac therapeutic use, Propranolol therapeutic use
- Abstract
Introduction: Pancreatic cancer is the fourth-leading cause of cancer-related death in developed countries. Despite advances in systemic chemotherapy, the mainstay of curative therapy for non-metastatic disease is surgical resection. However, the perioperative period is characterised by stress and inflammatory reactions that can contribute to metastatic spread and disease recurrence. Catecholamines and prostaglandins play a crucial role in these reactions. Therefore, a drug repurposing of betablockers and cyclooxygenase inhibitors seems reasonable to attenuate tumour-associated inflammation by inhibiting psychological, surgical and inflammatory stress responses. This may cause a relevant antitumourigenic and antimetastatic effect during the perioperative period, a window for cancer-directed therapy that is currently largely unexploited., Methods and Analysis: This is a prospective, single-centre, two-arm randomised, patient and observer blinded, placebo-controlled, phase-II trial evaluating safety and feasibility of combined perioperative treatment with propranolol and etodolac in adult patients with non-metastatic cancer of the pancreatic head undergoing elective pancreatoduodenectomy. 100 patients fulfilling the eligibility criteria will be randomised to perioperative treatment for 25 days perioperatively with a combination of propranolol and etodolac or placebo. Primary outcome of interest will be safety in terms of serious adverse events and reactions within 3 months. Furthermore, adherence to trial medication will be assessed as feasibility outcomes. Preliminary efficacy data will be evaluated for the purpose of power calculation for a potential subsequent phase-III trial. The clinical trial is accompanied by a translational study investigating the mechanisms of action of the combined therapy on a molecular basis., Ethics and Dissemination: The PROSPER-trial has been approved by the German Federal Institute for Drugs and Medical Devices (reference number 4042875) and the Ethics Committee of the Medical Faculty of the University of Heidelberg (reference number AFmo-385/2018). The final trial results will be published in a peer-reviewed journal and will be presented at appropriate national and international conferences., Trial Registration Numbers: DRKS00014054; EudraCT number: 2018-000415-25., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2020. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2020
- Full Text
- View/download PDF
23. On-Label or Off-Label? Overcoming Regulatory and Financial Barriers to Bring Repurposed Medicines to Cancer Patients.
- Author
-
Verbaanderd C, Rooman I, Meheus L, and Huys I
- Abstract
Repurposing of medicines has gained a lot of interest from the research community in recent years as it could offer safe, timely, and affordable new treatment options for cancer patients with high unmet needs. Increasingly, questions arise on how new uses will be translated into clinical practice, especially in case of marketed medicinal products that are out of basic patent or regulatory protection. The aim of this study was to portray the regulatory framework relevant for making repurposed medicines available to cancer patients in Europe and propose specific policy recommendations to address the current regulatory and financial barriers. We outlined two routes relevant to the clinical adoption of a repurposed medicine. First, a new indication can be approved, and thus brought on-label, via the marketing authorization procedures established in European and national legislation. Such procedures initiate a detailed and independent assessment of the quality and the benefit-risk balance of a medicinal product in a specific indication, benefiting both prescribers and patients as it reassures them that the scientific evidence is robust. However, the process of marketing authorization for new therapeutic indications entails a high administrative burden and significant costs while the return-on-investment for the pharmaceutical industry is expected to be low or absent for medicines that are out of basic patent and regulatory protection. Moreover, most of the repurposing research is conducted by independent or academic researchers who do not have the expertise or resources to get involved in regulatory procedures. A second option is to prescribe a medicine off-label for the new indication, which is managed at the national level in Europe. While off-label use could provide timely access to treatments for patients with urgent medical needs, it also entails important safety, liability and financial risks for patients, physicians, and society at large. In view of that, we recommend finding solutions to facilitate bringing new uses on-label, for example by developing a collaborative framework between not-for-profit and academic organizations, pharmaceutical industry, health technology assessment bodies, payers, and regulators., (Copyright © 2020 Verbaanderd, Rooman, Meheus and Huys.)
- Published
- 2020
- Full Text
- View/download PDF
24. Different shades of pancreatic ductal adenocarcinoma, different paths towards precision therapeutic applications.
- Author
-
Martens S, Lefesvre P, Nicolle R, Biankin AV, Puleo F, Van Laethem JL, and Rooman I
- Subjects
- Adenocarcinoma classification, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal classification, Carcinoma, Pancreatic Ductal pathology, Gene Expression Regulation, Neoplastic genetics, Humans, Precision Medicine trends, Prognosis, Adenocarcinoma genetics, Biomarkers, Tumor genetics, Carcinoma, Pancreatic Ductal genetics, Transcriptome genetics
- Abstract
Background: Different histological and molecular subtypes of pancreatic ductal adenocarcinoma (PDAC), with different molecular composition and survival statistics, have recently been recognised., Materials and Methods: This review describes the currently available studies regarding molecular and histological subtypes in PDAC. Studies from major cohorts such as International Cancer Genome Consortium as well as smaller cohorts are reviewed. We discuss where the described subtypes overlap, where the discrepancies are and which paths forward could be taken regarding diagnosis, ontogeny and therapy., Results: Four molecular subtypes with strong overlap among the different studies can be found, next to a list of mixed findings. Two of the four subtypes (epithelial classical and mesenchymal basal-like) were represented in every study and were often discriminated in other solid tumours as well. These two subtypes differ substantially in prognosis. One biomarker has been discovered, only discriminating these two subtypes, and insights into subtype-specific therapeutic vulnerabilities are scarce., Conclusion: Subtypes can be reproducibly detected in cohorts of PDAC patients and two of them directly relate with prognosis. A consensus on the subtypes is warranted. Further discovery and validation studies are needed to identify strong biomarkers, to comprehend subtype ontogeny and to define strategies for precision medicine., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society for Medical Oncology. All rights reserved. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2019
- Full Text
- View/download PDF
25. Adult human pancreatic acinar cells dedifferentiate into an embryonic progenitor-like state in 3D suspension culture.
- Author
-
Baldan J, Houbracken I, Rooman I, and Bouwens L
- Subjects
- Acinar Cells metabolism, Adult, Antigens, Tumor-Associated, Carbohydrate metabolism, Biomarkers metabolism, Cell Plasticity, Cells, Cultured, GPI-Linked Proteins metabolism, Homeodomain Proteins metabolism, Humans, Pancreas, Exocrine metabolism, Plant Lectins chemistry, SOX9 Transcription Factor metabolism, Thromboplastin metabolism, Trans-Activators metabolism, Acinar Cells cytology, Cell Lineage, Pancreas, Exocrine cytology
- Abstract
Human pancreatic exocrine cells were cultured in 3D suspension and formed pancreatospheres composed of acinar-derived and duct-like cells. We investigated, up to 6 days, the fate of human pancreatic acinar cells using fluorescein-conjugated Ulex Europaeus Agglutinin 1 lectin, a previously published acinar-specific non-genetic lineage tracing strategy. At day 4, fluorescence-activated cell sort for the intracellularly incorporated FITC-conjugated UEA1 lectin and the duct-specific CA19.9 surface marker, distinguished acinar-derived cells (UEA1
+ CA19.9- ) from duct-like cells (UEA1- CA19.9+ ) and acinar-to-duct-like transdifferentiated cells (UEA1+ CA19.9+ ). mRNA expression analysis of the acinar-derived (UEA1+ CA19.9- ) and duct-like (UEA1- CA19.9+ ) cell fractions with concomitant immunocytochemical analysis of the pancreatospheres revealed acquisition of an embryonic signature in the UEA1+ CA19.9- acinar-derived cells characterized by de novo expression of SOX9 and CD142, robust expression of PDX1 and surface expression of GP2. The colocalisation of CD142, a multipotent pancreatic progenitor surface marker, PDX1, SOX9 and GP2 is reminiscent of a cellular state present during human embryonic development. Addition of TGF-beta signalling inhibitor Alk5iII, induced a 28-fold increased KI67-labeling in pancreatospheres, more pronounced in the CD142+ GP2+ acinar-derived cells. These findings with human cells underscore the remarkable plasticity of pancreatic exocrine acinar cells, previously described in rodents, and could find applications in the field of regenerative medicine.- Published
- 2019
- Full Text
- View/download PDF
26. ReDO_DB: the repurposing drugs in oncology database.
- Author
-
Pantziarka P, Verbaanderd C, Sukhatme V, Rica Capistrano I, Crispino S, Gyawali B, Rooman I, Van Nuffel AM, Meheus L, Sukhatme VP, and Bouche G
- Abstract
Repurposing is a drug development strategy that seeks to use existing medications for new indications. In oncology, there is an increased level of activity looking at the use of non-cancer drugs as possible cancer treatments. The Repurposing Drugs in Oncology (ReDO) project has used a literature-based approach to identify licensed non-cancer drugs with published evidence of anticancer activity. Data from 268 drugs have been included in a database (ReDO_DB) developed by the ReDO project. Summary results are outlined and an assessment of clinical trial activity also described. The database has been made available as an online open-access resource (http://www.redo-project.org/db/).
- Published
- 2018
- Full Text
- View/download PDF
27. ROBO2 is a stroma suppressor gene in the pancreas and acts via TGF-β signalling.
- Author
-
Pinho AV, Van Bulck M, Chantrill L, Arshi M, Sklyarova T, Herrmann D, Vennin C, Gallego-Ortega D, Mawson A, Giry-Laterriere M, Magenau A, Leuckx G, Baeyens L, Gill AJ, Phillips P, Timpson P, Biankin AV, Wu J, and Rooman I
- Subjects
- Animals, Blotting, Western, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Cells, Cultured, Female, Flow Cytometry, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, In Situ Hybridization, In Vitro Techniques, Male, Mice, Nerve Tissue Proteins genetics, Nerve Tissue Proteins metabolism, Pancreatitis genetics, Pancreatitis metabolism, Receptors, Immunologic genetics, Signal Transduction genetics, Signal Transduction physiology, Trans-Activators genetics, Trans-Activators metabolism, Roundabout Proteins, Pancreas metabolism, Pancreas pathology, Receptors, Immunologic metabolism, Transforming Growth Factor beta metabolism
- Abstract
Whereas genomic aberrations in the SLIT-ROBO pathway are frequent in pancreatic ductal adenocarcinoma (PDAC), their function in the pancreas is unclear. Here we report that in pancreatitis and PDAC mouse models, epithelial Robo2 expression is lost while Robo1 expression becomes most prominent in the stroma. Cell cultures of mice with loss of epithelial Robo2 (Pdx1
Cre ;Robo2F/F ) show increased activation of Robo1+ myofibroblasts and induction of TGF-β and Wnt pathways. During pancreatitis, Pdx1Cre ;Robo2F/F mice present enhanced myofibroblast activation, collagen crosslinking, T-cell infiltration and tumorigenic immune markers. The TGF-β inhibitor galunisertib suppresses these effects. In PDAC patients, ROBO2 expression is overall low while ROBO1 is variably expressed in epithelium and high in stroma. ROBO2low ;ROBO1high patients present the poorest survival. In conclusion, Robo2 acts non-autonomously as a stroma suppressor gene by restraining myofibroblast activation and T-cell infiltration. ROBO1/2 expression in PDAC patients may guide therapy with TGF-β inhibitors or other stroma /immune modulating agents.- Published
- 2018
- Full Text
- View/download PDF
28. Young GI angle: My biggest (career) mistake.
- Author
-
Fichna J, Vergnolle N, and Rooman I
- Published
- 2018
- Full Text
- View/download PDF
29. Corrigendum: Whole-genome landscape of pancreatic neuroendocrine tumours.
- Author
-
Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, Rusev B, Scardoni M, Antonello D, Barbi S, Sikora KO, Cingarlini S, Vicentini C, McKay S, Quinn MCJ, Bruxner TJC, Christ AN, Harliwong I, Idrisoglu S, McLean S, Nourse C, Nourbakhsh E, Wilson PJ, Anderson MJ, Fink JL, Newell F, Waddell N, Holmes O, Kazakoff SH, Leonard C, Wood S, Xu Q, Hiriyur Nagaraj S, Amato E, Dalai I, Bersani S, Cataldo I, Dei Tos AP, Capelli P, Vittoria Davì M, Landoni L, Malpaga A, Miotto M, Whitehall VLJ, Leggett BA, Harris JL, Harris J, Jones MD, Humphris J, Chantrill LA, Chin V, Nagrial AM, Pajic M, Scarlett CJ, Pinho A, Rooman I, Toon C, Wu J, Pinese M, Cowley M, Barbour A, Mawson A, Humphrey ES, Colvin EK, Chou A, Lovell JA, Jamieson NB, Duthie F, Gingras MC, Fisher WE, Dagg RA, Lau LMS, Lee M, Pickett HA, Reddel RR, Samra JS, Kench JG, Merrett ND, Epari K, Nguyen NQ, Zeps N, Falconi M, Simbolo M, Butturini G, Van Buren G, Partelli S, Fassan M, Khanna KK, Gill AJ, Wheeler DA, Gibbs RA, Musgrove EA, Bassi C, Tortora G, Pederzoli P, Pearson JV, Waddell N, Biankin AV, and Grimmond SM
- Abstract
This corrects the article DOI: 10.1038/nature21063.
- Published
- 2017
- Full Text
- View/download PDF
30. Whole-genome landscape of pancreatic neuroendocrine tumours.
- Author
-
Scarpa A, Chang DK, Nones K, Corbo V, Patch AM, Bailey P, Lawlor RT, Johns AL, Miller DK, Mafficini A, Rusev B, Scardoni M, Antonello D, Barbi S, Sikora KO, Cingarlini S, Vicentini C, McKay S, Quinn MC, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, McLean S, Nourse C, Nourbakhsh E, Wilson PJ, Anderson MJ, Fink JL, Newell F, Waddell N, Holmes O, Kazakoff SH, Leonard C, Wood S, Xu Q, Nagaraj SH, Amato E, Dalai I, Bersani S, Cataldo I, Dei Tos AP, Capelli P, Davì MV, Landoni L, Malpaga A, Miotto M, Whitehall VL, Leggett BA, Harris JL, Harris J, Jones MD, Humphris J, Chantrill LA, Chin V, Nagrial AM, Pajic M, Scarlett CJ, Pinho A, Rooman I, Toon C, Wu J, Pinese M, Cowley M, Barbour A, Mawson A, Humphrey ES, Colvin EK, Chou A, Lovell JA, Jamieson NB, Duthie F, Gingras MC, Fisher WE, Dagg RA, Lau LM, Lee M, Pickett HA, Reddel RR, Samra JS, Kench JG, Merrett ND, Epari K, Nguyen NQ, Zeps N, Falconi M, Simbolo M, Butturini G, Van Buren G, Partelli S, Fassan M, Khanna KK, Gill AJ, Wheeler DA, Gibbs RA, Musgrove EA, Bassi C, Tortora G, Pederzoli P, Pearson JV, Waddell N, Biankin AV, and Grimmond SM
- Subjects
- Base Sequence, Calmodulin-Binding Proteins genetics, Chromatin Assembly and Disassembly genetics, Chromosome Aberrations, DNA Copy Number Variations genetics, DNA Glycosylases genetics, DNA Mutational Analysis, DNA Repair genetics, Female, Germ-Line Mutation genetics, Humans, Male, RNA-Binding Protein EWS, RNA-Binding Proteins genetics, TOR Serine-Threonine Kinases metabolism, Telomere genetics, Telomere metabolism, Carcinoma, Neuroendocrine genetics, Genome, Human genetics, Genomics, Pancreatic Neoplasms genetics
- Abstract
The diagnosis of pancreatic neuroendocrine tumours (PanNETs) is increasing owing to more sensitive detection methods, and this increase is creating challenges for clinical management. We performed whole-genome sequencing of 102 primary PanNETs and defined the genomic events that characterize their pathogenesis. Here we describe the mutational signatures they harbour, including a deficiency in G:C > T:A base excision repair due to inactivation of MUTYH, which encodes a DNA glycosylase. Clinically sporadic PanNETs contain a larger-than-expected proportion of germline mutations, including previously unreported mutations in the DNA repair genes MUTYH, CHEK2 and BRCA2. Together with mutations in MEN1 and VHL, these mutations occur in 17% of patients. Somatic mutations, including point mutations and gene fusions, were commonly found in genes involved in four main pathways: chromatin remodelling, DNA damage repair, activation of mTOR signalling (including previously undescribed EWSR1 gene fusions), and telomere maintenance. In addition, our gene expression analyses identified a subgroup of tumours associated with hypoxia and HIF signalling.
- Published
- 2017
- Full Text
- View/download PDF
31. ROCK signaling promotes collagen remodeling to facilitate invasive pancreatic ductal adenocarcinoma tumor cell growth.
- Author
-
Rath N, Morton JP, Julian L, Helbig L, Kadir S, McGhee EJ, Anderson KI, Kalna G, Mullin M, Pinho AV, Rooman I, Samuel MS, and Olson MF
- Subjects
- Animals, Gene Expression Profiling, Gene Regulatory Networks, Humans, Mice, Survival Analysis, Adenocarcinoma pathology, Carcinoma, Pancreatic Ductal pathology, Collagen metabolism, Signal Transduction, rho-Associated Kinases metabolism
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a major cause of cancer death; identifying PDAC enablers may reveal potential therapeutic targets. Expression of the actomyosin regulatory ROCK1 and ROCK2 kinases increased with tumor progression in human and mouse pancreatic tumors, while elevated ROCK1/ROCK2 expression in human patients, or conditional ROCK2 activation in a Kras
G12D /p53R172H mouse PDAC model, was associated with reduced survival. Conditional ROCK1 or ROCK2 activation promoted invasive growth of mouse PDAC cells into three-dimensional collagen matrices by increasing matrix remodeling activities. RNA sequencing revealed a coordinated program of ROCK-induced genes that facilitate extracellular matrix remodeling, with greatest fold-changes for matrix metalloproteinases (MMPs) Mmp10 and Mmp13 MMP inhibition not only decreased collagen degradation and invasion, but also reduced proliferation in three-dimensional contexts. Treatment of KrasG12D /p53R172H PDAC mice with a ROCK inhibitor prolonged survival, which was associated with increased tumor-associated collagen. These findings reveal an ancillary role for increased ROCK signaling in pancreatic cancer progression to promote extracellular matrix remodeling that facilitates proliferation and invasive tumor growth., (© 2016 Cancer Research UK Beatson Institute. Published under the terms of the CC BY 4.0 license.)- Published
- 2017
- Full Text
- View/download PDF
32. Hypermutation In Pancreatic Cancer.
- Author
-
Humphris JL, Patch AM, Nones K, Bailey PJ, Johns AL, McKay S, Chang DK, Miller DK, Pajic M, Kassahn KS, Quinn MC, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Stone A, Wilson PJ, Anderson M, Fink JL, Holmes O, Kazakoff S, Leonard C, Newell F, Waddell N, Wood S, Mead RS, Xu Q, Wu J, Pinese M, Cowley MJ, Jones MD, Nagrial AM, Chin VT, Chantrill LA, Mawson A, Chou A, Scarlett CJ, Pinho AV, Rooman I, Giry-Laterriere M, Samra JS, Kench JG, Merrett ND, Toon CW, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, McKay CJ, Carter CR, Dickson EJ, Graham JS, Duthie F, Oien K, Hair J, Morton JP, Sansom OJ, Grützmann R, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Rusev B, Corbo V, Salvia R, Cataldo I, Tortora G, Tempero MA, Hofmann O, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Gill AJ, Pearson JV, Grimmond SM, Waddell N, and Biankin AV
- Subjects
- Adult, Aged, Aged, 80 and over, DNA Mutational Analysis, Female, Genome, Humans, Male, Middle Aged, MutL Protein Homolog 1 genetics, MutS Homolog 2 Protein genetics, Proto-Oncogene Proteins p21(ras) genetics, Carcinoma, Pancreatic Ductal genetics, DNA Mismatch Repair genetics, Mutation, Pancreatic Neoplasms genetics, Transcriptome
- Abstract
Pancreatic cancer is molecularly diverse, with few effective therapies. Increased mutation burden and defective DNA repair are associated with response to immune checkpoint inhibitors in several other cancer types. We interrogated 385 pancreatic cancer genomes to define hypermutation and its causes. Mutational signatures inferring defects in DNA repair were enriched in those with the highest mutation burdens. Mismatch repair deficiency was identified in 1% of tumors harboring different mechanisms of somatic inactivation of MLH1 and MSH2. Defining mutation load in individual pancreatic cancers and the optimal assay for patient selection may inform clinical trial design for immunotherapy in pancreatic cancer., (Copyright © 2017 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
33. Sirtuin 1 stimulates the proliferation and the expression of glycolysis genes in pancreatic neoplastic lesions.
- Author
-
Pinho AV, Mawson A, Gill A, Arshi M, Warmerdam M, Giry-Laterriere M, Eling N, Lie T, Kuster E, Camargo S, Biankin AV, Wu J, and Rooman I
- Subjects
- Animals, Carcinoma in Situ, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation, Cell Transformation, Neoplastic genetics, Cell Transformation, Neoplastic metabolism, Disease Models, Animal, Glycolysis genetics, Humans, Mice, Mice, Transgenic, Pancreatic Neoplasms mortality, Pancreatic Neoplasms pathology, Prognosis, Gene Expression Regulation, Neoplastic, Glucose metabolism, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Sirtuin 1 metabolism
- Abstract
Metabolic reprogramming is a feature of neoplasia and tumor growth. Sirtuin 1 (SIRT1) is a lysine deacetylase of multiple targets including metabolic regulators such as p53. SIRT1 regulates metaplasia in the pancreas. Nevertheless, it is unclear if SIRT1 affects the development of neoplastic lesions and whether metabolic gene expression is altered.To assess neoplastic lesion development, mice with a pancreas-specific loss of Sirt1 (Pdx1-Cre;Sirt1-lox) were bred into a KrasG12D mutant background (KC) that predisposes to the development of pancreatic intra-epithelial neoplasia (PanIN) and ductal adenocarcinoma (PDAC). Similar grade PanIN lesions developed in KC and KC;Sirt1-lox mice but specifically early mucinous PanINs occupied 40% less area in the KC;Sirt1-lox line, attributed to reduced proliferation. This was accompanied by reduced expression of proteins in the glycolysis pathway, such as GLUT1 and GAPDH.The stimulatory effect of SIRT1 on proliferation and glycolysis gene expression was confirmed in a human PDAC cell line. In resected PDAC samples, higher proliferation and expression of glycolysis genes correlated with poor patient survival. SIRT1 expression per se was not prognostic but low expression of Cell Cycle and Apoptosis Regulator 2 (CCAR2), a reported SIRT1 inhibitor, corresponded to poor patient survival.These findings open perspectives for novel targeted therapies in pancreatic cancer.
- Published
- 2016
- Full Text
- View/download PDF
34. Repurposing Drugs in Oncology (ReDO)-Propranolol as an anti-cancer agent.
- Author
-
Pantziarka P, Bouche G, Sukhatme V, Meheus L, Rooman I, and Sukhatme VP
- Abstract
Propranolol (PRO) is a well-known and widely used non-selective beta-adrenergic receptor antagonist (beta-blocker), with a range of actions which are of interest in an oncological context. PRO displays effects on cellular proliferation and invasion, on the immune system, on the angiogenic cascade, and on tumour cell sensitivity to existing treatments. Both pre-clinical and clinical evidence of these effects, in multiple cancer types, is assessed and summarised and relevant mechanisms of action outlined. In particular there is evidence that PRO is effective at multiple points in the metastatic cascade, particularly in the context of the post-surgical wound response. Based on this evidence the case is made for further clinical investigation of the anticancer effects of PRO, particularly in combination with other agents. A number of trials are on-going, in different treatment settings for various cancers.
- Published
- 2016
- Full Text
- View/download PDF
35. JRK is a positive regulator of β-catenin transcriptional activity commonly overexpressed in colon, breast and ovarian cancer.
- Author
-
Pangon L, Ng I, Giry-Laterriere M, Currey N, Morgan A, Benthani F, Tran PN, Al-Sohaily S, Segelov E, Parker BL, Cowley MJ, Wright DC, St Heaps L, Carey L, Rooman I, and Kohonen-Corish MR
- Subjects
- Active Transport, Cell Nucleus, Base Sequence, Breast Neoplasms pathology, Cell Line, Tumor, Cell Nucleus metabolism, Cell Proliferation, Colonic Neoplasms pathology, Computer Simulation, DNA-Binding Proteins, Female, Humans, Mutation, Nuclear Proteins chemistry, Nuclear Proteins genetics, Oncogenes genetics, Ovarian Neoplasms pathology, Protein Domains, RNA-Binding Proteins, Up-Regulation, Wnt Signaling Pathway genetics, beta Catenin metabolism, Breast Neoplasms genetics, Colonic Neoplasms genetics, Gene Expression Regulation, Neoplastic, Nuclear Proteins metabolism, Ovarian Neoplasms genetics, Transcription, Genetic, beta Catenin genetics
- Abstract
The loss of β-catenin inhibitory components is a well-established mechanism of carcinogenesis but β-catenin hyperactivity can also be enhanced through its coactivators. Here we first interrogated a highly validated genomic screen and the largest repository of cancer genomics data and identified JRK as a potential new oncogene and therapeutic target of the β-catenin pathway. We proceeded to validate the oncogenic role of JRK in colon cancer cells and primary tumors. Consistent with a β-catenin activator function, depletion of JRK in several cancer cell lines repressed β-catenin transcriptional activity and reduced cell proliferation. Importantly, JRK expression was aberrantly elevated in 21% of colorectal cancers, 15% of breast and ovarian cancers and was associated with increased expression of β-catenin target genes and increased cell proliferation. This study shows that JRK is required for β-catenin hyperactivity regardless of the adenomatous polyposis coli/β-catenin mutation status and targeting JRK presents new opportunities for therapeutic intervention in cancer.
- Published
- 2016
- Full Text
- View/download PDF
36. Genomic analyses identify molecular subtypes of pancreatic cancer.
- Author
-
Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, Miller DK, Christ AN, Bruxner TJ, Quinn MC, Nourse C, Murtaugh LC, Harliwong I, Idrisoglu S, Manning S, Nourbakhsh E, Wani S, Fink L, Holmes O, Chin V, Anderson MJ, Kazakoff S, Leonard C, Newell F, Waddell N, Wood S, Xu Q, Wilson PJ, Cloonan N, Kassahn KS, Taylor D, Quek K, Robertson A, Pantano L, Mincarelli L, Sanchez LN, Evers L, Wu J, Pinese M, Cowley MJ, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chantrill LA, Mawson A, Humphris J, Chou A, Pajic M, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Lovell JA, Merrett ND, Toon CW, Epari K, Nguyen NQ, Barbour A, Zeps N, Moran-Jones K, Jamieson NB, Graham JS, Duthie F, Oien K, Hair J, Grützmann R, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Rusev B, Capelli P, Salvia R, Tortora G, Mukhopadhyay D, Petersen GM, Munzy DM, Fisher WE, Karim SA, Eshleman JR, Hruban RH, Pilarsky C, Morton JP, Sansom OJ, Scarpa A, Musgrove EA, Bailey UM, Hofmann O, Sutherland RL, Wheeler DA, Gill AJ, Gibbs RA, Pearson JV, Waddell N, Biankin AV, and Grimmond SM
- Subjects
- Animals, Basic Helix-Loop-Helix Transcription Factors genetics, Carcinoma, Pancreatic Ductal classification, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, DNA Methylation, DNA-Binding Proteins genetics, Gene Expression Regulation, Neoplastic, Gene Regulatory Networks, Hepatocyte Nuclear Factor 3-beta genetics, Hepatocyte Nuclear Factor 3-gamma genetics, Histone Demethylases genetics, Homeobox Protein Nkx-2.2, Homeodomain Proteins genetics, Humans, Mice, Nuclear Proteins genetics, Pancreatic Neoplasms immunology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Prognosis, Receptors, Cytoplasmic and Nuclear genetics, Survival Analysis, Trans-Activators genetics, Transcription Factors genetics, Transcription, Genetic, Transcriptome, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Proteins genetics, Zebrafish Proteins, Genes, Neoplasm genetics, Genome, Human genetics, Genomics, Mutation genetics, Pancreatic Neoplasms classification, Pancreatic Neoplasms genetics
- Abstract
Integrated genomic analysis of 456 pancreatic ductal adenocarcinomas identified 32 recurrently mutated genes that aggregate into 10 pathways: KRAS, TGF-β, WNT, NOTCH, ROBO/SLIT signalling, G1/S transition, SWI-SNF, chromatin modification, DNA repair and RNA processing. Expression analysis defined 4 subtypes: (1) squamous; (2) pancreatic progenitor; (3) immunogenic; and (4) aberrantly differentiated endocrine exocrine (ADEX) that correlate with histopathological characteristics. Squamous tumours are enriched for TP53 and KDM6A mutations, upregulation of the TP63∆N transcriptional network, hypermethylation of pancreatic endodermal cell-fate determining genes and have a poor prognosis. Pancreatic progenitor tumours preferentially express genes involved in early pancreatic development (FOXA2/3, PDX1 and MNX1). ADEX tumours displayed upregulation of genes that regulate networks involved in KRAS activation, exocrine (NR5A2 and RBPJL), and endocrine differentiation (NEUROD1 and NKX2-2). Immunogenic tumours contained upregulated immune networks including pathways involved in acquired immune suppression. These data infer differences in the molecular evolution of pancreatic cancer subtypes and identify opportunities for therapeutic development.
- Published
- 2016
- Full Text
- View/download PDF
37. SOX9 regulates ERBB signalling in pancreatic cancer development.
- Author
-
Grimont A, Pinho AV, Cowley MJ, Augereau C, Mawson A, Giry-Laterrière M, Van den Steen G, Waddell N, Pajic M, Sempoux C, Wu J, Grimmond SM, Biankin AV, Lemaigre FP, Rooman I, and Jacquemin P
- Subjects
- Adenocarcinoma genetics, Animals, Carcinoma, Pancreatic Ductal genetics, Cell Transformation, Neoplastic, Gene Expression Regulation, Neoplastic, Humans, Mice, Pancreatic Neoplasms genetics, SOX9 Transcription Factor genetics, Signal Transduction, Adenocarcinoma etiology, Carcinoma, Pancreatic Ductal etiology, ErbB Receptors physiology, Pancreatic Neoplasms etiology, SOX9 Transcription Factor physiology
- Abstract
Objective: The transcription factor SOX9 was recently shown to stimulate ductal gene expression in pancreatic acinar-to-ductal metaplasia and to accelerate development of premalignant lesions preceding pancreatic ductal adenocarcinoma (PDAC). Here, we investigate how SOX9 operates in pancreatic tumourigenesis., Design: We analysed genomic and transcriptomic data from surgically resected PDAC and extended the expression analysis to xenografts from PDAC samples and to PDAC cell lines. SOX9 expression was manipulated in human cell lines and mouse models developing PDAC., Results: We found genetic aberrations in the SOX9 gene in about 15% of patient tumours. Most PDAC samples strongly express SOX9 protein, and SOX9 levels are higher in classical PDAC. This tumour subtype is associated with better patient outcome, and cell lines of this subtype respond to therapy targeting epidermal growth factor receptor (EGFR/ERBB1) signalling, a pathway essential for pancreatic tumourigenesis. In human PDAC, high expression of SOX9 correlates with expression of genes belonging to the ERBB pathway. In particular, ERBB2 expression in PDAC cell lines is stimulated by SOX9. Inactivating Sox9 expression in mice confirmed its role in PDAC initiation; it demonstrated that Sox9 stimulates expression of several members of the ERBB pathway and is required for ERBB signalling activity., Conclusions: By integrating data from patient samples and mouse models, we found that SOX9 regulates the ERBB pathway throughout pancreatic tumourigenesis. Our work opens perspectives for therapy targeting tumourigenic mechanisms., (Published by the BMJ Publishing Group Limited. For permission to use (where not already granted under a licence) please go to http://group.bmj.com/group/rights-licensing/permissions.)
- Published
- 2015
- Full Text
- View/download PDF
38. Pancreas-Specific Sirt1-Deficiency in Mice Compromises Beta-Cell Function without Development of Hyperglycemia.
- Author
-
Pinho AV, Bensellam M, Wauters E, Rees M, Giry-Laterriere M, Mawson A, Ly le Q, Biankin AV, Wu J, Laybutt DR, and Rooman I
- Subjects
- Animals, Blood Glucose analysis, Down-Regulation, Endoplasmic Reticulum metabolism, Glucagon-Like Peptide-1 Receptor genetics, Glucagon-Like Peptide-1 Receptor metabolism, Glucose Transporter Type 2 genetics, Glucose Transporter Type 2 metabolism, Homeodomain Proteins genetics, Hyperglycemia metabolism, Hyperglycemia pathology, Islets of Langerhans pathology, Mice, Mice, Knockout, Microscopy, Fluorescence, Molecular Chaperones genetics, Molecular Chaperones metabolism, Oligonucleotide Array Sequence Analysis, Real-Time Polymerase Chain Reaction, Sirtuin 1 deficiency, Sirtuin 1 genetics, Trans-Activators genetics, Unfolded Protein Response, Insulin-Secreting Cells metabolism, Islets of Langerhans metabolism, Sirtuin 1 metabolism
- Abstract
Aims/hypothesis: Sirtuin 1 (Sirt1) has been reported to be a critical positive regulator of glucose-stimulated insulin secretion in pancreatic beta-cells. The effects on islet cells and blood glucose levels when Sirt1 is deleted specifically in the pancreas are still unclear., Methods: This study examined islet glucose responsiveness, blood glucose levels, pancreatic islet histology and gene expression in Pdx1Cre; Sirt1ex4F/F mice that have loss of function and loss of expression of Sirt1 specifically in the pancreas., Results: We found that in the Pdx1Cre; Sirt1ex4F/F mice, the relative insulin positive area and the islet size distribution were unchanged. However, beta-cells were functionally impaired, presenting with lower glucose-stimulated insulin secretion. This defect was not due to a reduced expression of insulin but was associated with a decreased expression of the glucose transporter Slc2a2/Glut2 and of the Glucagon like peptide-1 receptor (Glp1r) as well as a marked down regulation of endoplasmic reticulum (ER) chaperones that participate in the Unfolded Protein Response (UPR) pathway. Counter intuitively, the Sirt1-deficient mice did not develop hyperglycemia. Pancreatic polypeptide (PP) cells were the only other islet cells affected, with reduced numbers in the Sirt1-deficient pancreas., Conclusions/interpretation: This study provides new mechanistic insights showing that beta-cell function in Sirt1-deficient pancreas is affected due to altered glucose sensing and deregulation of the UPR pathway. Interestingly, we uncovered a context in which impaired beta-cell function is not accompanied by increased glycemia. This points to a unique compensatory mechanism. Given the reduction in PP, investigation of its role in the control of blood glucose is warranted.
- Published
- 2015
- Full Text
- View/download PDF
39. Whole genomes redefine the mutational landscape of pancreatic cancer.
- Author
-
Waddell N, Pajic M, Patch AM, Chang DK, Kassahn KS, Bailey P, Johns AL, Miller D, Nones K, Quek K, Quinn MC, Robertson AJ, Fadlullah MZ, Bruxner TJ, Christ AN, Harliwong I, Idrisoglu S, Manning S, Nourse C, Nourbakhsh E, Wani S, Wilson PJ, Markham E, Cloonan N, Anderson MJ, Fink JL, Holmes O, Kazakoff SH, Leonard C, Newell F, Poudel B, Song S, Taylor D, Waddell N, Wood S, Xu Q, Wu J, Pinese M, Cowley MJ, Lee HC, Jones MD, Nagrial AM, Humphris J, Chantrill LA, Chin V, Steinmann AM, Mawson A, Humphrey ES, Colvin EK, Chou A, Scarlett CJ, Pinho AV, Giry-Laterriere M, Rooman I, Samra JS, Kench JG, Pettitt JA, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Jamieson NB, Graham JS, Niclou SP, Bjerkvig R, Grützmann R, Aust D, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Wolfgang CL, Morgan RA, Lawlor RT, Corbo V, Bassi C, Falconi M, Zamboni G, Tortora G, Tempero MA, Gill AJ, Eshleman JR, Pilarsky C, Scarpa A, Musgrove EA, Pearson JV, Biankin AV, and Grimmond SM
- Subjects
- Adenocarcinoma drug therapy, Adenocarcinoma genetics, Animals, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal genetics, DNA Repair genetics, Female, Genes, BRCA1, Genes, BRCA2, Genetic Markers genetics, Genomic Instability genetics, Genotype, Humans, Mice, Pancreatic Neoplasms classification, Pancreatic Neoplasms drug therapy, Platinum pharmacology, Point Mutation genetics, Poly(ADP-ribose) Polymerase Inhibitors, Xenograft Model Antitumor Assays, DNA Mutational Analysis, Genome, Human genetics, Genomics, Mutation genetics, Pancreatic Neoplasms genetics
- Abstract
Pancreatic cancer remains one of the most lethal of malignancies and a major health burden. We performed whole-genome sequencing and copy number variation (CNV) analysis of 100 pancreatic ductal adenocarcinomas (PDACs). Chromosomal rearrangements leading to gene disruption were prevalent, affecting genes known to be important in pancreatic cancer (TP53, SMAD4, CDKN2A, ARID1A and ROBO2) and new candidate drivers of pancreatic carcinogenesis (KDM6A and PREX2). Patterns of structural variation (variation in chromosomal structure) classified PDACs into 4 subtypes with potential clinical utility: the subtypes were termed stable, locally rearranged, scattered and unstable. A significant proportion harboured focal amplifications, many of which contained druggable oncogenes (ERBB2, MET, FGFR1, CDK6, PIK3R3 and PIK3CA), but at low individual patient prevalence. Genomic instability co-segregated with inactivation of DNA maintenance genes (BRCA1, BRCA2 or PALB2) and a mutational signature of DNA damage repair deficiency. Of 8 patients who received platinum therapy, 4 of 5 individuals with these measures of defective DNA maintenance responded.
- Published
- 2015
- Full Text
- View/download PDF
40. Emerging Drug Target In Pancreatic Cancer: Placing Sirtuin 1 on the Canvas.
- Author
-
Giry-Laterriere M, Pinho AV, Eling N, Chantrill L, and Rooman I
- Subjects
- Animals, Antineoplastic Agents adverse effects, Carcinoma, Pancreatic Ductal enzymology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Gene Expression Regulation, Neoplastic, Histone Deacetylase Inhibitors adverse effects, Humans, Pancreatic Neoplasms enzymology, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology, Signal Transduction drug effects, Sirtuin 1 genetics, Sirtuin 1 metabolism, Treatment Outcome, Antineoplastic Agents therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Drug Discovery methods, Histone Deacetylase Inhibitors therapeutic use, Molecular Targeted Therapy, Pancreatic Neoplasms drug therapy, Sirtuin 1 antagonists & inhibitors
- Abstract
Sirtuin 1 is a protein deacetylase that regulates a large number of proteins often functionally implicated in tumor development and progression. Its pleiotropic function has turned SIRT1 into an attractive chemotherapeutic target, underscored by very promising preclinical results with SIRT1 inhibitors in the treatment of chronic myeloid leukemia. Here, we revisit the studies on SIRT1 as an emerging target for therapy in pancreatic cancer, a tumor with dismal outcomes for which currently few therapeutic options are available. We highlight those potential SIRT1 target genes that are commonly affected in pancreatic cancer according to recent genomic analyses.
- Published
- 2015
- Full Text
- View/download PDF
41. Genome-wide DNA methylation patterns in pancreatic ductal adenocarcinoma reveal epigenetic deregulation of SLIT-ROBO, ITGA2 and MET signaling.
- Author
-
Nones K, Waddell N, Song S, Patch AM, Miller D, Johns A, Wu J, Kassahn KS, Wood D, Bailey P, Fink L, Manning S, Christ AN, Nourse C, Kazakoff S, Taylor D, Leonard C, Chang DK, Jones MD, Thomas M, Watson C, Pinese M, Cowley M, Rooman I, Pajic M, Butturini G, Malpaga A, Corbo V, Crippa S, Falconi M, Zamboni G, Castelli P, Lawlor RT, Gill AJ, Scarpa A, Pearson JV, Biankin AV, and Grimmond SM
- Subjects
- Adult, Aged, Aged, 80 and over, Base Sequence, Cell Adhesion genetics, Female, Gene Expression Profiling, Humans, Integrin alpha2 genetics, Integrins metabolism, Intercellular Signaling Peptides and Proteins genetics, Male, Membrane Proteins genetics, Middle Aged, Nerve Tissue Proteins genetics, Pancreatic Ducts pathology, Pancreatic Stellate Cells pathology, Promoter Regions, Genetic genetics, Proto-Oncogene Proteins c-met genetics, RNA, Messenger biosynthesis, Receptors, Immunologic genetics, Sequence Analysis, DNA, Signal Transduction genetics, Transforming Growth Factor beta genetics, Wnt Proteins genetics, Roundabout Proteins, Slit Homolog 2 Protein, Carcinoma, Pancreatic Ductal genetics, DNA Methylation, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Pancreatic Neoplasms genetics
- Abstract
The importance of epigenetic modifications such as DNA methylation in tumorigenesis is increasingly being appreciated. To define the genome-wide pattern of DNA methylation in pancreatic ductal adenocarcinomas (PDAC), we captured the methylation profiles of 167 untreated resected PDACs and compared them to a panel of 29 adjacent nontransformed pancreata using high-density arrays. A total of 11,634 CpG sites associated with 3,522 genes were significantly differentially methylated (DM) in PDAC and were capable of segregating PDAC from non-malignant pancreas, regardless of tumor cellularity. As expected, PDAC hypermethylation was most prevalent in the 5' region of genes (including the proximal promoter, 5'UTR and CpG islands). Approximately 33% DM genes showed significant inverse correlation with mRNA expression levels. Pathway analysis revealed an enrichment of aberrantly methylated genes involved in key molecular mechanisms important to PDAC: TGF-β, WNT, integrin signaling, cell adhesion, stellate cell activation and axon guidance. Given the recent discovery that SLIT-ROBO mutations play a clinically important role in PDAC, the role of epigenetic perturbation of axon guidance was pursued in more detail. Bisulfite amplicon deep sequencing and qRT-PCR expression analyses confirmed recurrent perturbation of axon guidance pathway genes SLIT2, SLIT3, ROBO1, ROBO3, ITGA2 and MET and suggests epigenetic suppression of SLIT-ROBO signaling and up-regulation of MET and ITGA2 expression. Hypomethylation of MET and ITGA2 correlated with high gene expression, which was associated with poor survival. These data suggest that aberrant methylation plays an important role in pancreatic carcinogenesis affecting core signaling pathways with potential implications for the disease pathophysiology and therapy., (The Authors. Published by Wiley Periodicals, Inc. on behalf of UICC.)
- Published
- 2014
- Full Text
- View/download PDF
42. Early epigenetic downregulation of WNK2 kinase during pancreatic ductal adenocarcinoma development.
- Author
-
Dutruel C, Bergmann F, Rooman I, Zucknick M, Weichenhan D, Geiselhart L, Kaffenberger T, Rachakonda PS, Bauer A, Giese N, Hong C, Xie H, Costello JF, Hoheisel J, Kumar R, Rehli M, Schirmacher P, Werner J, Plass C, Popanda O, and Schmezer P
- Subjects
- Animals, Carcinoma, Pancreatic Ductal pathology, Cell Line, Tumor, Cell Proliferation, CpG Islands genetics, Down-Regulation, Extracellular Signal-Regulated MAP Kinases biosynthesis, Female, Humans, Male, Mice, Mice, Transgenic, Oligonucleotide Array Sequence Analysis, Pancreatic Neoplasms pathology, Pancreatitis, Chronic genetics, Promoter Regions, Genetic, Protein Serine-Threonine Kinases biosynthesis, RNA, Messenger biosynthesis, Carcinoma, Pancreatic Ductal genetics, DNA Methylation genetics, Epigenesis, Genetic, Gene Expression Regulation, Neoplastic, Pancreatic Neoplasms genetics, Protein Serine-Threonine Kinases genetics
- Abstract
Pancreatic ductal adenocarcinoma (PDAC) is usually incurable. Contrary to genetic mechanisms involved in PDAC pathogenesis, epigenetic alterations are ill defined. Here, we determine the contribution of epigenetically silenced genes to the development of PDAC. We analyzed enriched, highly methylated DNAs from PDACs, chronic pancreatitis (CP) and normal tissues using CpG island microarrays and identified WNK2 as a prominent candidate tumor suppressor gene being downregulated early in PDAC development. WNK2 was further investigated in tissue microarrays, methylation analysis of early pancreatic intraepithelial neoplasia (PanIN), mouse models for PDAC and pancreatitis, re-expression studies after demethylation, and cell growth assays using WNK2 overexpression. Demethylation assays confirmed the link between methylation and expression. WNK2 hypermethylation was higher in tumor than in surrounding inflamed tissues and was observed in PanIN lesions as well as in a PDAC mouse model. WNK2 mRNA and protein expressions were lower in PDAC and CP compared with normal tissues both in patients and mouse models. Overexpression of WNK2 led to reduced cell growth, and WNK2 expression in tissues correlated negatively with pERK1/2 expression, a downstream target of WNK2 responsible for cell proliferation. Downregulation of WNK2 by promoter hypermethylation occurs early in PDAC pathogenesis and may support tumor cell growth via the ERK-MAPK pathway.
- Published
- 2014
- Full Text
- View/download PDF
43. Chronic pancreatitis: a path to pancreatic cancer.
- Author
-
Pinho AV, Chantrill L, and Rooman I
- Subjects
- Animals, Anti-Inflammatory Agents therapeutic use, Anticarcinogenic Agents therapeutic use, Antineoplastic Agents therapeutic use, Carcinoma, Pancreatic Ductal drug therapy, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal prevention & control, Cell Transformation, Neoplastic immunology, Cell Transformation, Neoplastic metabolism, Humans, Inflammation Mediators metabolism, Pancreatic Neoplasms drug therapy, Pancreatic Neoplasms immunology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms prevention & control, Pancreatitis, Chronic drug therapy, Pancreatitis, Chronic immunology, Pancreatitis, Chronic metabolism, Risk Factors, Signal Transduction, Carcinoma, Pancreatic Ductal etiology, Pancreatic Neoplasms etiology, Pancreatitis, Chronic complications
- Abstract
Chronic pancreatitis predisposes to pancreatic cancer development and both diseases share a common etiology. A central role has been proposed for the digestive enzyme-secreting acinar cell that can undergo ductal metaplasia in the inflammatory environment of pancreatitis. This metaplastic change is now a recognised precursor of pancreatic cancer. Inflammatory molecules also foster tumour growth through autocrine and paracrine effects in the epithelium and the stroma. These insights have raised new opportunities such as the manipulation of inflammation as a preventive and/or therapeutic strategy for pancreatic cancer. Finally, we address the need for an in-depth study of the pancreatic acinar cells., (Copyright © 2013 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
44. Sirtainties in pancreatic cancer?
- Author
-
Rooman I
- Published
- 2014
- Full Text
- View/download PDF
45. Amino acid transporters expression in acinar cells is changed during acute pancreatitis.
- Author
-
Rooman I, Lutz C, Pinho AV, Huggel K, Reding T, Lahoutte T, Verrey F, Graf R, and Camargo SM
- Subjects
- Acute Disease, Amino Acid Transport System y+ biosynthesis, Amino Acid Transport Systems, Neutral biosynthesis, Animals, Cells, Cultured, Fusion Regulatory Protein 1, Light Chains biosynthesis, Large Neutral Amino Acid-Transporter 1 biosynthesis, Male, Mice, Pancreas physiopathology, Acinar Cells metabolism, Amino Acid Transport Systems biosynthesis, Pancreas physiology, Pancreatitis physiopathology
- Abstract
Pancreatic acinar cells accumulate amino acids against a marked concentration gradient to synthesize digestive enzymes. Thus, the function of acinar cells depends on amino acid uptake mediated by active transport. Despite the importance of this process, pancreatic amino acid transporter expression and cellular localization is still unclear. We screened mouse pancreas for the expression of genes encoding amino acid transporters. We showed that the most highly expressed transporters, namely sodium dependent SNAT3 (Slc38a3) and SNAT5 (Slc38a5) and sodium independent neutral amino acids transporters LAT1 (Slc7a5) and LAT2 (Slc7a8), are expressed in the basolateral membrane of acinar cells. SNAT3 and SNAT5, LAT1 and LAT2 are expressed in acinar cells. Additional evidence that these transporters are expressed in mature acinar cells was gained using acinar cell culture and acute pancreatitis models. In the acute phase of pancreatic injury, when acinar cell loss occurs, and in an acinar cell culture model, which mimics changes occurring during pancreatitis, SNAT3 and SNAT5 are strongly down-regulated. LAT1 and LAT2 were down-regulated only in the in vitro model. At protein level, SNAT3 and SNAT5 expression was also reduced during pancreatitis. Expression of other amino acid transporters was also modified in both models of pancreatitis. The subset of transporters with differential expression patterns during acute pancreatitis might be involved in the injury/regeneration phases. Further expression, localization and functional studies will follow to better understand changes occurring during acute pancreatitis. These findings provide insight into pancreatic amino acid transport in healthy pancreas and during acute pancreatitis injury., (Copyright © 2013 IAP and EPC. Published by Elsevier B.V. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
46. Clinical and molecular characterization of HER2 amplified-pancreatic cancer.
- Author
-
Chou A, Waddell N, Cowley MJ, Gill AJ, Chang DK, Patch AM, Nones K, Wu J, Pinese M, Johns AL, Miller DK, Kassahn KS, Nagrial AM, Wasan H, Goldstein D, Toon CW, Chin V, Chantrill L, Humphris J, Mead RS, Rooman I, Samra JS, Pajic M, Musgrove EA, Pearson JV, Morey AL, Grimmond SM, and Biankin AV
- Abstract
Background: Pancreatic cancer is one of the most lethal and molecularly diverse malignancies. Repurposing of therapeutics that target specific molecular mechanisms in different disease types offers potential for rapid improvements in outcome. Although HER2 amplification occurs in pancreatic cancer, it is inadequately characterized to exploit the potential of anti-HER2 therapies., Methods: HER2 amplification was detected and further analyzed using multiple genomic sequencing approaches. Standardized reference laboratory assays defined HER2 amplification in a large cohort of patients (n = 469) with pancreatic ductal adenocarcinoma (PDAC)., Results: An amplified inversion event (1 MB) was identified at the HER2 locus in a patient with PDAC. Using standardized laboratory assays, we established diagnostic criteria for HER2 amplification in PDAC, and observed a prevalence of 2%. Clinically, HER2- amplified PDAC was characterized by a lack of liver metastases, and a preponderance of lung and brain metastases. Excluding breast and gastric cancer, the incidence of HER2-amplified cancers in the USA is >22,000 per annum., Conclusions: HER2 amplification occurs in 2% of PDAC, and has distinct features with implications for clinical practice. The molecular heterogeneity of PDAC implies that even an incidence of 2% represents an attractive target for anti-HER2 therapies, as options for PDAC are limited. Recruiting patients based on HER2 amplification, rather than organ of origin, could make trials of anti-HER2 therapies feasible in less common cancer types.
- Published
- 2013
- Full Text
- View/download PDF
47. Histomolecular phenotypes and outcome in adenocarcinoma of the ampulla of vater.
- Author
-
Chang DK, Jamieson NB, Johns AL, Scarlett CJ, Pajic M, Chou A, Pinese M, Humphris JL, Jones MD, Toon C, Nagrial AM, Chantrill LA, Chin VT, Pinho AV, Rooman I, Cowley MJ, Wu J, Mead RS, Colvin EK, Samra JS, Corbo V, Bassi C, Falconi M, Lawlor RT, Crippa S, Sperandio N, Bersani S, Dickson EJ, Mohamed MA, Oien KA, Foulis AK, Musgrove EA, Sutherland RL, Kench JG, Carter CR, Gill AJ, Scarpa A, McKay CJ, and Biankin AV
- Subjects
- Adenocarcinoma pathology, Adult, Aged, Aged, 80 and over, Ampulla of Vater pathology, CDX2 Transcription Factor, Cohort Studies, Common Bile Duct Neoplasms pathology, Female, Humans, Immunohistochemistry, Kaplan-Meier Estimate, Keratin-20 biosynthesis, Keratin-7 biosynthesis, Male, Middle Aged, Mucin-2 biosynthesis, Multivariate Analysis, Neoplasm Staging, Prognosis, Adenocarcinoma metabolism, Ampulla of Vater metabolism, Common Bile Duct Neoplasms metabolism, Homeodomain Proteins biosynthesis, Mucin-1 biosynthesis
- Abstract
Purpose: Individuals with adenocarcinoma of the ampulla of Vater demonstrate a broad range of outcomes, presumably because these cancers may arise from any one of the three epithelia that converge at that location. This variability poses challenges for clinical decision making and the development of novel therapeutic strategies., Patients and Methods: We assessed the potential clinical utility of histomolecular phenotypes defined using a combination of histopathology and protein expression (CDX2 and MUC1) in 208 patients from three independent cohorts who underwent surgical resection for adenocarcinoma of the ampulla of Vater., Results: Histologic subtype and CDX2 and MUC1 expression were significant prognostic variables. Patients with a histomolecular pancreaticobiliary phenotype (CDX2 negative, MUC1 positive) segregated into a poor prognostic group in the training (hazard ratio [HR], 3.34; 95% CI, 1.69 to 6.62; P < .001) and both validation cohorts (HR, 5.65; 95% CI, 2.77 to 11.5; P < .001 and HR, 2.78; 95% CI, 1.25 to 7.17; P = .0119) compared with histomolecular nonpancreaticobiliary carcinomas. Further stratification by lymph node (LN) status defined three clinically relevant subgroups: one, patients with histomolecular nonpancreaticobiliary (intestinal) carcinoma without LN metastases who had an excellent prognosis; two, those with histomolecular pancreaticobiliary carcinoma with LN metastases who had a poor outcome; and three, the remainder of patients (nonpancreaticobiliary, LN positive or pancreaticobiliary, LN negative) who had an intermediate outcome., Conclusion: Histopathologic and molecular criteria combine to define clinically relevant histomolecular phenotypes of adenocarcinoma of the ampulla of Vater and potentially represent distinct diseases with significant implications for current therapeutic strategies, the ability to interpret past clinical trials, and future trial design.
- Published
- 2013
- Full Text
- View/download PDF
48. Sirtuin-1 regulates acinar-to-ductal metaplasia and supports cancer cell viability in pancreatic cancer.
- Author
-
Wauters E, Sanchez-Arévalo Lobo VJ, Pinho AV, Mawson A, Herranz D, Wu J, Cowley MJ, Colvin EK, Njicop EN, Sutherland RL, Liu T, Serrano M, Bouwens L, Real FX, Biankin AV, and Rooman I
- Subjects
- Acinar Cells metabolism, Animals, Biomarkers, Tumor genetics, Blotting, Western, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal metabolism, Cell Differentiation, Cell Nucleus metabolism, Cell Survival, Cells, Cultured, Cytoplasm metabolism, Humans, Immunoenzyme Techniques, Metaplasia genetics, Metaplasia metabolism, Mice, Pancreas, Pancreatic Neoplasms genetics, Pancreatic Neoplasms metabolism, Pancreatitis genetics, Pancreatitis metabolism, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Sirtuin 1 genetics, Acinar Cells cytology, Biomarkers, Tumor metabolism, Carcinoma, Pancreatic Ductal pathology, Metaplasia pathology, Pancreatic Neoplasms pathology, Pancreatitis pathology, Sirtuin 1 metabolism
- Abstract
The exocrine pancreas can undergo acinar-to-ductal metaplasia (ADM), as in the case of pancreatitis where precursor lesions of pancreatic ductal adenocarcinoma (PDAC) can arise. The NAD(+)-dependent protein deacetylase Sirtuin-1 (Sirt1) has been implicated in carcinogenesis with dual roles depending on its subcellular localization. In this study, we examined the expression and the role of Sirt1 in different stages of pancreatic carcinogenesis, i.e. ADM models and established PDAC. In addition, we analyzed the expression of KIAA1967, a key mediator of Sirt1 function, along with potential Sirt1 downstream targets. Sirt1 was co-expressed with KIAA1967 in the nuclei of normal pancreatic acinar cells. In ADM, Sirt1 underwent a transient nuclear-to-cytoplasmic shuttling. Experiments where during ADM, we enforced repression of Sirt1 shuttling, inhibition of Sirt1 activity or modulation of its expression, all underscore that the temporary decrease of nuclear and increase of cytoplasmic Sirt1 stimulate ADM. Our results further underscore that important transcriptional regulators of acinar differentiation, that is, Pancreatic transcription factor-1a and β-catenin can be deacetylated by Sirt1. Inhibition of Sirt1 is effective in suppression of ADM and in reducing cell viability in established PDAC tumors. KIAA1967 expression is differentially downregulated in PDAC and impacts on the sensitivity of PDAC cells to the Sirt1/2 inhibitor Tenovin-6. In PDAC, acetylation of β-catenin is not affected, unlike p53, a well-characterized Sirt1-regulated protein in tumor cells. Our results reveal that Sirt1 is an important regulator and potential therapeutic target in pancreatic carcinogenesis., (©2012 AACR.)
- Published
- 2013
- Full Text
- View/download PDF
49. The histone deacetylase SIRT2 stabilizes Myc oncoproteins.
- Author
-
Liu PY, Xu N, Malyukova A, Scarlett CJ, Sun YT, Zhang XD, Ling D, Su SP, Nelson C, Chang DK, Koach J, Tee AE, Haber M, Norris MD, Toon C, Rooman I, Xue C, Cheung BB, Kumar S, Marshall GM, Biankin AV, and Liu T
- Subjects
- Aurora Kinases, Cell Line, Tumor, Cell Proliferation, Endosomal Sorting Complexes Required for Transport, Gene Expression, Humans, Naphthols pharmacology, Nedd4 Ubiquitin Protein Ligases, Phenylpropionates pharmacology, Phosphorylation, Protein Binding, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Protein Stability, Proto-Oncogene Proteins c-myc antagonists & inhibitors, Proto-Oncogene Proteins c-myc genetics, RNA Interference, RNA, Messenger metabolism, RNA, Small Interfering metabolism, Sirtuin 2 antagonists & inhibitors, Sirtuin 2 genetics, Ubiquitin-Protein Ligases, Ubiquitination, Up-Regulation drug effects, Proto-Oncogene Proteins c-myc metabolism, Sirtuin 2 metabolism
- Abstract
Myc oncoproteins are commonly upregulated in human cancers of different organ origins, stabilized by Aurora A, degraded through ubiquitin-proteasome pathway-mediated proteolysis, and exert oncogenic effects by modulating gene and protein expression. Histone deacetylases are emerging as targets for cancer therapy. Here we demonstrated that the class III histone deacetylase SIRT2 was upregulated by N-Myc in neuroblastoma cells and by c-Myc in pancreatic cancer cells, and that SIRT2 enhanced N-Myc and c-Myc protein stability and promoted cancer cell proliferation. Affymetrix gene array studies revealed that the gene most significantly repressed by SIRT2 was the ubiquitin-protein ligase NEDD4. Consistent with this finding, SIRT2 repressed NEDD4 gene expression by directly binding to the NEDD4 gene core promoter and deacetylating histone H4 lysine 16. Importantly, NEDD4 directly bound to Myc oncoproteins and targeted Myc oncoproteins for ubiquitination and degradation, and small-molecule SIRT2 inhibitors reactivated NEDD4 gene expression, reduced N-Myc and c-Myc protein expression, and suppressed neuroblastoma and pancreatic cancer cell proliferation. Additionally, SIRT2 upregulated and small-molecule SIRT2 inhibitors decreased Aurora A expression. Our data reveal a novel pathway critical for Myc oncoprotein stability, and provide important evidences for potential application of SIRT2 inhibitors for the prevention and therapy of Myc-induced malignancies.
- Published
- 2013
- Full Text
- View/download PDF
50. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes.
- Author
-
Biankin AV, Waddell N, Kassahn KS, Gingras MC, Muthuswamy LB, Johns AL, Miller DK, Wilson PJ, Patch AM, Wu J, Chang DK, Cowley MJ, Gardiner BB, Song S, Harliwong I, Idrisoglu S, Nourse C, Nourbakhsh E, Manning S, Wani S, Gongora M, Pajic M, Scarlett CJ, Gill AJ, Pinho AV, Rooman I, Anderson M, Holmes O, Leonard C, Taylor D, Wood S, Xu Q, Nones K, Fink JL, Christ A, Bruxner T, Cloonan N, Kolle G, Newell F, Pinese M, Mead RS, Humphris JL, Kaplan W, Jones MD, Colvin EK, Nagrial AM, Humphrey ES, Chou A, Chin VT, Chantrill LA, Mawson A, Samra JS, Kench JG, Lovell JA, Daly RJ, Merrett ND, Toon C, Epari K, Nguyen NQ, Barbour A, Zeps N, Kakkar N, Zhao F, Wu YQ, Wang M, Muzny DM, Fisher WE, Brunicardi FC, Hodges SE, Reid JG, Drummond J, Chang K, Han Y, Lewis LR, Dinh H, Buhay CJ, Beck T, Timms L, Sam M, Begley K, Brown A, Pai D, Panchal A, Buchner N, De Borja R, Denroche RE, Yung CK, Serra S, Onetto N, Mukhopadhyay D, Tsao MS, Shaw PA, Petersen GM, Gallinger S, Hruban RH, Maitra A, Iacobuzio-Donahue CA, Schulick RD, Wolfgang CL, Morgan RA, Lawlor RT, Capelli P, Corbo V, Scardoni M, Tortora G, Tempero MA, Mann KM, Jenkins NA, Perez-Mancera PA, Adams DJ, Largaespada DA, Wessels LF, Rust AG, Stein LD, Tuveson DA, Copeland NG, Musgrove EA, Scarpa A, Eshleman JR, Hudson TJ, Sutherland RL, Wheeler DA, Pearson JV, McPherson JD, Gibbs RA, and Grimmond SM
- Subjects
- Animals, Gene Dosage, Gene Expression Regulation, Neoplastic, Humans, Kaplan-Meier Estimate, Mice, Mutation, Proteins genetics, Signal Transduction, Axons metabolism, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal pathology, Genome genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms pathology
- Abstract
Pancreatic cancer is a highly lethal malignancy with few effective therapies. We performed exome sequencing and copy number analysis to define genomic aberrations in a prospectively accrued clinical cohort (n = 142) of early (stage I and II) sporadic pancreatic ductal adenocarcinoma. Detailed analysis of 99 informative tumours identified substantial heterogeneity with 2,016 non-silent mutations and 1,628 copy-number variations. We define 16 significantly mutated genes, reaffirming known mutations (KRAS, TP53, CDKN2A, SMAD4, MLL3, TGFBR2, ARID1A and SF3B1), and uncover novel mutated genes including additional genes involved in chromatin modification (EPC1 and ARID2), DNA damage repair (ATM) and other mechanisms (ZIM2, MAP2K4, NALCN, SLC16A4 and MAGEA6). Integrative analysis with in vitro functional data and animal models provided supportive evidence for potential roles for these genetic aberrations in carcinogenesis. Pathway-based analysis of recurrently mutated genes recapitulated clustering in core signalling pathways in pancreatic ductal adenocarcinoma, and identified new mutated genes in each pathway. We also identified frequent and diverse somatic aberrations in genes described traditionally as embryonic regulators of axon guidance, particularly SLIT/ROBO signalling, which was also evident in murine Sleeping Beauty transposon-mediated somatic mutagenesis models of pancreatic cancer, providing further supportive evidence for the potential involvement of axon guidance genes in pancreatic carcinogenesis.
- Published
- 2012
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.