1
|
Frendi S, Chassac A, Veron K, Raffenne J, Nicolle R, Albuquerque M, Paradis V, Couvelard A, Cros J, Rebours V. Protective role of oleic acid against palmitic acid-induced pancreatic fibrosis. J Transl Med 2025; 23:416. [PMID: 40211240 PMCID: PMC11983757 DOI: 10.1186/s12967-025-06313-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 02/23/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Obesity has been associated with several pancreatic disorders and is an important risk factor for pancreatic cancer. Nevertheless, the role of lipids in the early steps of carcinogenesis is unknown. Although we previously identified two types of pancreatic fatty infiltration with different lipid compositions that were associated with precancerous lesions and fibrosis, their mechanisms of action have not been clarified. METHODS We hypothesized that saturated palmitic acid and mono-unsaturated oleic acid (OA and PA) could play diverse roles in the activation of pancreatic stellate cells (PSCs) during the genesis of pancreatic fibrosis and the promotion of precancerous lesions. This study explored the lipotoxic effect of OA and PA on PSCs and exocrine pancreatic tissue (acinar cells). We also explored PA-induced pyroptosis in PSCs. A three-dimensional culture system of organotypic slices from human pancreatic tissues was used as well as a two-dimensional culture of hTERT immortalized PSCs. RESULTS The results show that PA could induce the secretion of collagens and inflammatory cytokines (IL18) in PSCs (p < 0.05). We defined a standardized protocol of precision-cut pancreas slices cultured from human non-tumoral pancreatic tissue (n = 9). Both OA and PA are involved in the initiation of acinar cell transformation into ductal cells. OA was found to have a protective effect against PA-induced fibrosis (p < 0.05). CONCLUSION These results highlight the antagonistic roles of oleic and palmitic acids in the initiation of pancreatic fibrosis and show that palmitic acid has a profibrotic role.
Collapse
Affiliation(s)
- Sonia Frendi
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
| | - Anaïs Chassac
- Pathology Department, Bichat and Beaujon Hospitals, AP-HP, FHU MOSAIC, Paris-Cité University, Paris, France
| | - Killian Veron
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
| | | | - Rémy Nicolle
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149,, CNRS, ERL 8252, F-75018, Paris, France
| | - Miguel Albuquerque
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
- Pathology Department, Bichat and Beaujon Hospitals, AP-HP, FHU MOSAIC, Paris-Cité University, Paris, France
| | - Valérie Paradis
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
- Pathology Department, Bichat and Beaujon Hospitals, AP-HP, FHU MOSAIC, Paris-Cité University, Paris, France
| | - Anne Couvelard
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
- Pathology Department, Bichat and Beaujon Hospitals, AP-HP, FHU MOSAIC, Paris-Cité University, Paris, France
| | - Jérôme Cros
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France
- Pathology Department, Bichat and Beaujon Hospitals, AP-HP, FHU MOSAIC, Paris-Cité University, Paris, France
| | - Vinciane Rebours
- Paris-Cité University, Inflammation Research Center (CRI), INSERM, U1149, F-75018, Paris, France.
- Pancreatology and Digestive Oncology Department - DMU Digest, Beaujon Hospital, AP-HP, INSERM, UMR 1149, Paris-Cité University, Clichy, France.
| |
Collapse
|
2
|
Sharma R, Kumar S, Komal K, Ghosh R, Thakur S, Pal RR, Kumar M. Comprehensive insights into pancreatic cancer treatment approaches and cutting-edge nanocarrier solutions: from pathology to nanomedicine. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04094-y. [PMID: 40202672 DOI: 10.1007/s00210-025-04094-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 03/21/2025] [Indexed: 04/10/2025]
Abstract
Pancreatic cancer is one of the most lethal malignancies worldwide. It is characterized by poor prognosis, high mortality, and recurrence rates. Various modifiable and non-modifiable risk factors are associated with pancreatic cancer incidence. Available treatments for pancreatic cancer include surgery, chemotherapy, radiotherapy, photodynamic therapy, supportive care, targeted therapy, and immunotherapy. However, the survival rates for PC are very low. Regrettably, despite efforts to enhance prognosis, the survival rate of pancreatic cancer remains relatively low. Therefore, it is essential to investigate new approaches to improve pancreatic cancer treatment. By synthesizing current knowledge and identifying existing gaps, this article provides a comprehensive overview of risk factors, pathology, conventional treatments, targeted therapies, and recent advancements in nanocarriers for its treatment, along with various clinical trials and patents that justify the safety and efficacy of innovative carriers for drug delivery systems. Ultimately, this review underscores the potential of these innovative formulations to improve outcomes and contribute significantly to the advancement of Pancreatic Cancer treatment. Together, these insights highlight nano-formulations as a promising frontier for effectively treating Pancreatic Cancer.
Collapse
Affiliation(s)
- Rohit Sharma
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Sourabh Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Kumari Komal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Rashmi Ghosh
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Shubham Thakur
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Ravi Raj Pal
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India
| | - Manish Kumar
- Department of Pharmaceutics, ISF College Pharmacy, GT Road, Moga, 142001, Punjab, India.
| |
Collapse
|
3
|
Liaki V, Rosas-Perez B, Guerra C. Unlocking the Genetic Secrets of Pancreatic Cancer: KRAS Allelic Imbalances in Tumor Evolution. Cancers (Basel) 2025; 17:1226. [PMID: 40227826 PMCID: PMC11987834 DOI: 10.3390/cancers17071226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/13/2025] [Accepted: 03/31/2025] [Indexed: 04/15/2025] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDAC) belongs to the types of cancer with the highest lethality. It is also remarkably chemoresistant to the few available cytotoxic therapeutic options. PDAC is characterized by limited mutational heterogeneity of the known driver genes, KRAS, CDKN2A, TP53, and SMAD4, observed in both early-stage and advanced tumors. In this review, we summarize the two proposed models of genetic evolution of pancreatic cancer. The gradual or stepwise accumulated mutations model has been widely studied. On the contrary, less evidence exists on the more recent simultaneous model, according to which rapid tumor evolution is driven by the concurrent accumulation of genetic alterations. In both models, oncogenic KRAS mutations are the main initiating event. Here, we analyze the emerging topic of KRAS allelic imbalances and how it arises during tumor evolution, as it is often detected in advanced and metastatic PDAC. We also summarize recent evidence on how it affects tumor biology, metastasis, and response to therapy. To this extent, we highlight the necessity to include studies of KRAS allelic frequencies in the design of future therapeutic strategies against pancreatic cancer.
Collapse
Affiliation(s)
- Vasiliki Liaki
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Blanca Rosas-Perez
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
| | - Carmen Guerra
- Molecular Oncology Program, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain; (B.R.-P.); (C.G.)
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
4
|
Qin C, Xu C, Zhu Z, Song X, Wang X, Xu W, Zhu M. A study of the association between Helicobacter pylori infection type and pancreatic cancer risk: A systematic review and meta‑analysis. Oncol Lett 2025; 29:174. [PMID: 39975953 PMCID: PMC11837465 DOI: 10.3892/ol.2025.14920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Accepted: 01/16/2025] [Indexed: 02/21/2025] Open
Abstract
Pancreatic cancer is a highly invasive malignant tumor with a complex pathogenesis that makes early diagnosis challenging. The potential association between Helicobacter pylori infection and pancreatic cancer risk has been noted; however, the available results are still highly divergent. The aim of the present study was to systematically evaluate the association between different types of H. pylori infection and pancreatic cancer risk as well as to explore the possible causes. A systematic search was conducted using the PubMed, Embase and Cochrane Library databases up to August 2023. The literature quality was evaluated using the Newcastle-Ottawa Scale. All studies that met the criteria were included in the overall meta-analysis to calculate the odds ratios (ORs) and corresponding 95% confidence intervals (CIs). In addition, subgroup analyses were performed based on factors such as diagnostic criteria for H. pylori infection, study region, type of study design and CagA status. The effect of publication bias on the quantitative synthesis results was assessed using the trim-and-fill analysis, and sensitivity analyses were used to verify the robustness of the quantitative synthesis results. A total of 17 studies involving 67,910 participants, including 64,372 controls and 3,538 patients with pancreatic cancer, were included in the present study. The overall analysis showed that no significant association was observed between H. pylori infection and pancreatic cancer risk (OR, 1.15; 95% CI, 0.93-1.41). Further subgroup analyses, which did not consider the effects of study quality, diagnostic criteria, geographical distribution and the type of study design, did not produce new findings that contradicted the results of the overall analysis. CagA+ H. pylori infection did not significantly affect the risk of pancreatic cancer (OR, 0.95; 95% CI, 0.78-1.16), whereas CagA- H. pylori infection may be a possible risk factor for pancreatic cancer (OR, 1.24; 95% CI, 1.004-1.541). The H. pylori infection did not significantly increase the risk of pancreatic cancer. However, it is noteworthy that CagA- H. pylori infection could be a potential factor that elevated the risk of pancreatic cancer.
Collapse
Affiliation(s)
- Chao Qin
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Chonghe Xu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, P.R. China
| | - Zhongqi Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xixi Song
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Xin Wang
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| | - Wei Xu
- Department of Blood Transfusion, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Mei Zhu
- Department of Clinical Laboratory, The Affiliated Chaohu Hospital of Anhui Medical University, Chaohu, Anhui 238000, P.R. China
| |
Collapse
|
5
|
Moore LL, Houchen CW. Evolving insights into pancreatic tumor initiation and progression through DCLK1-expressing tuft cells. Dev Cell 2025; 60:817-818. [PMID: 40132546 DOI: 10.1016/j.devcel.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 02/19/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
In this issue of Developmental Cell, Salas-Escabillas et al. find that tuft cells undergo a tuft-to-neuroendocrine transition, switching from tumor suppressors to tumor promoters via c-Myc in pancreatic ductal adenocarcinoma (PDAC). This result offers exciting insights into PDAC's aggressiveness, therapy resistance, and high mortality.
Collapse
Affiliation(s)
- Landon L Moore
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA
| | - Courtney W Houchen
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Veterans Affairs Medical Center, Oklahoma City, OK 73104, USA; The Peggy and Charles Stephenson Cancer Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
6
|
Zhao G, Li Y, Li H, Bao M, Lubamba GP, Wang G, Han B, Tang Y, Li T, Li C. Integrating single-cell sequencing and clinical insights to explore malignant transformation in odontogenic keratocyst. Comput Struct Biotechnol J 2025; 27:1158-1172. [PMID: 40206344 PMCID: PMC11981763 DOI: 10.1016/j.csbj.2025.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025] Open
Abstract
The malignant transformation of odontogenic keratocysts (OKC) into cancerous odontogenic keratocysts (COKC) is exceedingly rare, and its mechanisms remain poorly understood. Studies exploring the cellular heterogeneity, molecular pathways, and clinical features of COKC are limited. In this study, we performed single-cell RNA sequencing (scRNA-seq) on three COKC samples and integrated the data with a public OKC dataset, identifying 22,509 single cells. Two COKC-specific epithelial subpopulations, Basal-C0-EXT1 and Basal-C3-HIST1H3B, were identified. These subpopulations exhibited enhanced stemness and invasive potential, respectively, suggesting their roles as key drivers of OKC carcinogenesis. Fibroblasts underwent phenotypic transitions, particularly from inflammation-associated fibroblasts (IFBs) to myofibroblasts (MFBs). Similarly, macrophage phenotypic transformation may also play a role in OKC carcinogenesis. Clinical observations of severe lesion-area pain in COKC patients suggest potential neuroinvasiveness, Supported by single-cell transcriptomic data, imaging findings, and histopathological evidence. A review of clinical data revealed that none of the COKC patients exhibited cervical lymph node metastasis. Single-cell transcriptomics suggests that this phenomenon may be associated with an active immune microenvironment in COKC, reduced epithelial-mesenchymal transition (EMT) activity, lower VEGFC expression, and upregulated MAST4 expression as a potential regulator of lymphatic metastasis. In conclusion, COKC exhibits distinct molecular, cellular, and clinical characteristics compared to OKC, featuring potent neuroinvasiveness and low lymph node metastatic potential. These findings provide important insights into the mechanisms underlying COKC development and may guide novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Guile Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yike Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hongling Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Mingzhe Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Grace Paka Lubamba
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Oral and Maxillofacial Surgery, University Clinics of Kinshasa, Faculty of Dental Medicine, University of Kinshasa, Kinshasa B.P.127, Congo
| | - Guanru Wang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Bo Han
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Yaling Tang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Taiwen Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
| | - Chunjie Li
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, Chengdu, Sichuan 610041, China
- Department of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Schneider AT, Koppe C, Crouchet E, Papargyriou A, Singer MT, Büttner V, Keysberg L, Szydlowska M, Jühling F, Moehlin J, Chen MC, Leone V, Mueller S, Neuß T, Castoldi M, Lesina M, Bergmann F, Hackert T, Steiger K, Knoefel WT, Zaufel A, Kather JN, Esposito I, Gaida MM, Ghallab A, Hengstler JG, Einwächter H, Unger K, Algül H, Gassler N, Schmid RM, Rad R, Baumert TF, Reichert M, Heikenwalder M, Kondylis V, Vucur M, Luedde T. A decision point between transdifferentiation and programmed cell death priming controls KRAS-dependent pancreatic cancer development. Nat Commun 2025; 16:1765. [PMID: 39971907 PMCID: PMC11839950 DOI: 10.1038/s41467-025-56493-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
KRAS-dependent acinar-to-ductal metaplasia (ADM) is a fundamental step in the development of pancreatic ductal adenocarcinoma (PDAC), but the involvement of cell death pathways remains unclear. Here, we show that key regulators of programmed cell death (PCD) become upregulated during KRAS-driven ADM, thereby priming transdifferentiated cells to death. Using transgenic mice and primary cell and organoid cultures, we show that transforming growth factor (TGF)-β-activated kinase 1 (TAK1), a kinase regulating cell survival and inflammatory pathways, prevents the elimination of transdifferentiated cells through receptor-interacting protein kinase 1 (RIPK1)-mediated apoptosis and necroptosis, enabling PDAC development. Accordingly, pharmacological inhibition of TAK1 induces PCD in patient-derived PDAC organoids. Importantly, cell death induction via TAK1 inhibition does not appear to elicit an overt injury-associated inflammatory response. Collectively, these findings suggest that TAK1 supports cellular plasticity by suppressing spontaneous PCD activation during ADM, representing a promising pharmacological target for the prevention and treatment of PDAC.
Collapse
Affiliation(s)
- Anne T Schneider
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Christiane Koppe
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Emilie Crouchet
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Aristeidis Papargyriou
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Institute of Stem Cell Research, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Michael T Singer
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Veronika Büttner
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Leonie Keysberg
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Marta Szydlowska
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Frank Jühling
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Julien Moehlin
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
| | - Min-Chun Chen
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Valentina Leone
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Research Unit Radiation Cytogenetics, Helmholtz-Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Sebastian Mueller
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, TU Munich, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
| | - Thorsten Neuß
- Lehrstuhl für Biophysik E27, Center for Protein Assemblies (CPA), Technical University Munich (TUM), Garching, Germany
| | - Mirco Castoldi
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Marina Lesina
- Comprehensive Cancer Center München, Institute for Tumor Metabolism, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Frank Bergmann
- Institut of Pathology, Heidelberg University Hospital, Heidelberg, Germany
- Clinical Pathology, Klinikum Darmstadt GmbH, Darmstadt, Germany
| | - Thilo Hackert
- Department of General, Visceral, and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Katja Steiger
- Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Wolfram T Knoefel
- Department of Surgery A, Heinrich-Heine-University Düsseldorf and University Hospital Düsseldorf, Duesseldorf, Germany
| | - Alex Zaufel
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Jakob N Kather
- Else Kroener Fresenius Center for Digital Health (EFFZ), Technical University Dresden, Dresden, Germany
- Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| | - Irene Esposito
- Institute of Pathology, University Hospital Duesseldorf, Heinrich-Heine University, Duesseldorf, Germany
| | - Matthias M Gaida
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- Research Center for Immunotherapy, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- Joint Unit Immunopathology, Institute of Pathology, University Medical Center, JGU-Mainz, Mainz, Germany
- TRON, Translational Oncology at the University Medical Center, JGU-Mainz, Mainz, Germany
| | - Ahmed Ghallab
- Leibniz Research Centre for Working Environment and Human Factors (IfADo) at the Technical University Dortmund, Dortmund, Germany
- Forensic Medicine and Toxicology Department, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Jan G Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IfADo) at the Technical University Dortmund, Dortmund, Germany
| | - Henrik Einwächter
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Kristian Unger
- Department of Radiation Oncology, University Hospital, LMU Munich, Munich, Germany
- Research Unit Translational Metabolic Oncology, Institute for Diabetes and Cancer, Helmholtz Zentrum München Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Hana Algül
- Comprehensive Cancer Center München, Institute for Tumor Metabolism, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Nikolaus Gassler
- Section Pathology of the Institute of Forensic Medicine, University Hospital Jena, Jena, Germany
| | - Roland M Schmid
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Roland Rad
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, Munich, Germany
- Department of Internal Medicine II, Klinikum Rechts der Isar, Technical University of Munich, Munich, Germany
| | - Thomas F Baumert
- University of Strasbourg, Inserm, Institute for Translational Medicine and Liver Disease (ITM), UMR_S1110, Strasbourg, France
- Pôle des Pathologies Hépatiques et Digestives, Service d'Hepato-Gastroenterologie, Strasbourg University Hospitals, Strasbourg, France
- Institut Hospitalo-Universitaire (IHU) Strasbourg, Strasbourg, France
- Institut Universitaire de France (IUF), Paris, France
| | - Maximilian Reichert
- Translational Pancreatic Cancer Research Center, Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Klinik und Poliklinik für Innere Medizin II, Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
- Center for Organoid Systems (COS), Technical University of Munich, Garching, Germany
- Munich Institute of Biomedical Engineering (MIBE), Technical University of Munich, Garching, Germany
- German Center for Translational Cancer Research (DKTK), Munich, Germany
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
- The M3 Research Institute, Karls Eberhards Universität Tübingen, Tübingen, Germany
| | - Vangelis Kondylis
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Mihael Vucur
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital Düsseldorf, Medical Faculty at Heinrich-Heine-University, Duesseldorf, Germany.
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany.
| |
Collapse
|
8
|
Manoukian P, Kuhnen LC, van Laarhoven HWM, Bijlsma MF. Association of epigenetic landscapes with heterogeneity and plasticity in pancreatic cancer. Crit Rev Oncol Hematol 2025; 206:104573. [PMID: 39581245 DOI: 10.1016/j.critrevonc.2024.104573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 11/13/2024] [Accepted: 11/17/2024] [Indexed: 11/26/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has a poor prognosis. Due to a lack of clear symptoms, patients often present with advanced disease, with limited clinical intervention options. The high mortality rate of PDAC is, however, also a result of several other factors that include a high degree of heterogeneity and treatment resistant cellular phenotypes. Molecular subtypes of PDAC have been identified that are thought to represent cellular phenotypes at the tissue level. The epigenetic landscape is an important factor that dictates these subtypes. Permissive epigenetic landscapes serve as drivers of molecular heterogeneity and cellular plasticity in developing crypts as well as metaplastic lesions. Drawing parallels with other cancers, we hypothesize that epigenetic permissiveness is a potential driver of cellular plasticity in PDAC. In this review will explore the epigenetic alterations that underlie PDAC cell states and relate them to cellular plasticity from other contexts. In doing so, we aim to highlight epigenomic drivers of PDAC heterogeneity and plasticity and, with that, offer some insight to guide pre-clinical research.
Collapse
Affiliation(s)
- Paul Manoukian
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands.
| | - Leo C Kuhnen
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands
| | - Hanneke W M van Laarhoven
- Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands; Amsterdam UMC location University of Amsterdam, Department of Medical Oncology, Amsterdam, the Netherlands
| | - Maarten F Bijlsma
- Amsterdam UMC location University of Amsterdam, Center for Experimental and Molecular Medicine, Laboratory of Experimental Oncology and Radiobiology, Amsterdam, the Netherlands; Cancer Center Amsterdam, Cancer Biology, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Zhou C, Feng Z, Qian W, Zhu Z, Cao R, Wang Q, Zhang W, Liu R, Wu S, Hao J, Wang Z, Ma Q, Wu Z, Li X. Elevated SREBP1 accelerates the initiation and growth of pancreatic cancer by targeting SOX9. Biol Direct 2025; 20:6. [PMID: 39806513 PMCID: PMC11731174 DOI: 10.1186/s13062-025-00595-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025] Open
Abstract
Pancreatic cancer is a lethal disease with an insidious onset, and little is known about its early molecular events. Here, we found that the sterol regulatory element-binding protein 1 (SREBP1) expression is gradually upregulated during the initiation of pancreatic cancer. Through in vitro 3D culture of pancreatic acinar cells and experiments in LSL-KrasG12D/+;Pdx1-Cre (KC) mice, we found that pharmacological inhibition of SREBP1 suppressed pancreatic tumorigenesis. In vitro, either knockdown or pharmacological inhibition of SREBP1 suppressed tumor proliferation but SREBP1 overexpression promoted tumor proliferation. In LSL-KrasG12D/+;Trp53fl/+;Pdx1-Cre (KPC) mice, we confirmed the tumor-promoting role of SREBP1 in pancreatic cancer progression. Mechanistically, we revealed SOX9 as a downstream target of SREPB1. SREBP1 inhibition decreased SOX9 expression in both acinar cells and pancreatic cancer cells. Indeed, we identified SREBP1 binding sites in the SOX9 promoter region and reported that SOX9 is transcriptionally regulated by SREBP1. Taken together, our findings demonstrate that SREBP1/SOX9 inhibition suppresses pancreatic cancer initiation and growth, suggesting that SREBP1 could serve as a potential target for cancer screening and treatment.
Collapse
Affiliation(s)
- Cancan Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhengyuan Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Weikun Qian
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zeen Zhu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ruiqi Cao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qiqi Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Wunai Zhang
- Department of General Surgery, Second Affiliated Hospital, Xi'an Jiaotong University, Xi'an, 710004, China
| | - Rujuan Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shuai Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Jie Hao
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Qingyong Ma
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zheng Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China.
- Pancreas Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| | - Xuqi Li
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 West Yanta Road, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
10
|
Zu F, Chen C, Geng Q, Li H, Chan B, Luo G, Wu M, Ilmer M, Renz BW, Bentum-Ennin L, Gu H, Sheng W. Smad2 Cooperating with TGIF2 Contributes to EMT and Cancer Stem Cells Properties in Pancreatic Cancer via Co-Targeting SOX2. Int J Biol Sci 2025; 21:524-543. [PMID: 39781447 PMCID: PMC11705628 DOI: 10.7150/ijbs.102381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/18/2024] [Indexed: 01/12/2025] Open
Abstract
The underlying mechanisms between cancer stem cells (CSC) and epithelial-mesenchymal transition (EMT) in pancreatic cancer (PC) remain unclear. In this study, we identified TGIF2 as a target gene of CSC using sncRNA and machine learning. TGIF2 is closely related to the expression of SOX2, EGFR, and E-cadherin, indicating poor prognosis. Mechanistically, TGIF2 promoted the EMT phenotype and CSC properties following the activation of SOX2, Slug, CD44, and ERGF/MAPK signaling, which were rescued by SOX2 silencing. TGIF2 silencing contributes to the opposite phenotype via SOX2. Notably, Smad2 cooperates with TGIF2 to co-regulate the SOX2 promoter, which in turn promotes EMT and CSC signaling by transactivating Slug and EGFR, respectively. The transactivation of EGFR/MAPK signaling by SOX2 promotes TGIF2 nuclear translocation, forming a positive feedback loop in vitro. Moreover, the interaction of TGIF2 and SOX2 with EGFR inhibitors promoted subcutaneous tumors and liver metastasis in vivo. Thus, the TGIF2/SOX2 axis contributes to CSC, EMT, and chemoresistance, providing a promising target for PC therapy.
Collapse
Affiliation(s)
- Fuqiang Zu
- Department of General Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - ChuanPing Chen
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, China
| | - Qilong Geng
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Haoyu Li
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Boyuan Chan
- Department of Clinical Medicine, Anhui Medical University, Hefei, 230022, China
| | - Guopei Luo
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Mengcheng Wu
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Matthias Ilmer
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Bernhard W Renz
- German Cancer Consortium (DKTK), Partner Site Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Lutterodt Bentum-Ennin
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Hao Gu
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Weiwei Sheng
- Department of General Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
11
|
Walker M, Morton JP. Hydrogel models of pancreatic adenocarcinoma to study cell mechanosensing. Biophys Rev 2024; 16:851-870. [PMID: 39830124 PMCID: PMC11735828 DOI: 10.1007/s12551-024-01265-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and one of the leading causes of cancer-related death worldwide, with an extremely poor prognosis after diagnosis. High mortality from PDAC arises partly due to late diagnosis resulting from a lack of early-stage biomarkers and due to chemotherapeutic drug resistance, which arises from a highly fibrotic stromal response known as desmoplasia. Desmoplasia alters tissue mechanics, which triggers changes in cell mechanosensing and leads to dysregulated transcriptional activity and disease phenotypes. Hydrogels are effective in vitro models to mimic mechanical changes in tissue mechanics during PDAC progression and to study the influence of these changes on mechanosensitive cell responses. Despite the complex biophysical changes that occur within the PDAC microenvironment, carefully designed hydrogels can very closely recapitulate these properties during PDAC progression. Hydrogels are relatively inexpensive, highly reproducible and can be designed in a humanised manner to increase their relevance for human PDAC studies. In vivo models have some limitations, including species-species differences, high variability, expense and legal/ethical considerations, which make hydrogel models a promising alternative. Here, we comprehensively review recent advancements in hydrogel bioengineering for developing our fundamental understanding of mechanobiology in PDAC, which is critical for informing advanced therapeutics.
Collapse
Affiliation(s)
- M Walker
- Centre for the Cellular Microenvironment, Advanced Research Centre, 11 Chapel Lane, James Watt School of Engineering, University of Glasgow, Glasgow, G11 6EW UK
| | - JP Morton
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Rd, Glasgow, G61 1BD UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow, G61 1QH UK
| |
Collapse
|
12
|
Wang H, Gao J, Wen L, Huang K, Liu H, Zeng L, Zeng Z, Liu Y, Mo Z. Ion channels in acinar cells in acute pancreatitis: crosstalk of calcium, iron, and copper signals. Front Immunol 2024; 15:1444272. [PMID: 39606246 PMCID: PMC11599217 DOI: 10.3389/fimmu.2024.1444272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024] Open
Abstract
The initial stages of acute pancreatitis (AP) are characterized by a significant event - acinar ductal metaplasia (ADM). This process is a crucial feature of both acute and chronic pancreatitis, serving as the first step in the development of pancreatic cancer. Ion channels are integral transmembrane proteins that play a pivotal role in numerous biological processes by modulating ion flux. In many diseases, the expression and activity of ion channels are often dysregulated. Metal ions, including calcium ions (Ca2+), ferrous ions (Fe2+), and Copper ions (Cu2+), assume a distinctive role in cellular metabolism. These ions possess specific biological properties relevant to cellular function. However, the interactions among these ions exacerbate the imbalance within the intracellular environment, resulting in cellular damage and influencing the progression of AP. A more in-depth investigation into the mechanisms by which these ions interact with acinar cells is essential for elucidating AP's pathogenesis and identifying novel therapeutic strategies. Currently, treatment for AP primarily focuses on pain relief, complications prevention, and prognosis improvement. There are limited specific treatments targeting acinous cell dedifferentiation or ion imbalance. This study aims to investigate potential therapeutic strategies by examining ion crosstalk within acinar cells in the context of acute pancreatitis.
Collapse
Affiliation(s)
- Hanli Wang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Jianhua Gao
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Lingling Wen
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Kejun Huang
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Huixian Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Linsheng Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhongyi Zeng
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Qazi S, Trieu V. TGFB2 mRNA Levels Prognostically Interact with Interferon-Alpha Receptor Activation of IRF9 and IFI27, and an Immune Checkpoint LGALS9 to Impact Overall Survival in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2024; 25:11221. [PMID: 39457004 PMCID: PMC11508538 DOI: 10.3390/ijms252011221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/13/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
The treatment of pancreatic ductal adenocarcinoma (PDAC) is an unmet challenge, with the median overall survival rate remaining less than a year, even with the use of FOLFIRINOX-based therapies. This study analyzed archived macrophage-associated mRNA expression using datasets deposited in the UCSC Xena web platform to compare normal pancreatic tissue and PDAC tumor samples. The TGFB2 gene exhibited low mRNA expression levels in normal tissue, with less than one TPM. In contrast, in tumor tissue, TGFB2 expression levels exhibited a 7.9-fold increase in mRNA expression relative to normal tissue (p < 0.0001). Additionally, components of the type-I interferon signaling pathway exhibited significant upregulation of mRNA levels in tumor tissue, including Interferon alpha/beta receptor 1 (IFNAR1; 3.4-fold increase, p < 0.0001), Interferon regulatory factor 9 (IRF9; 4.2-fold increase, p < 0.0001), Signal transducer and activator of transcription 1 (STAT1; 7.1-fold increase, p < 0.0001), and Interferon Alpha Inducible Protein 27 (IFI27; 66.3-fold increase, p < 0.0001). We also utilized TCGA datasets deposited in cBioportal and KMplotter to relate mRNA expression levels to overall survival outcomes. These increased levels of mRNA expression were found to be prognostically significant, whereby patients with high expression levels of either TGFB2, IRF9, or IFI27 showed median OS times ranging from 16 to 20 months (p < 0.01 compared to 72 months for patients with low levels of expression for both TGFB2 and either IRF9 or IFI27). Examination of the KMplotter database determined the prognostic impact of TGFB2 mRNA expression levels by comparing patients expressing high versus low levels of TGFB2 (50th percentile cut-off) in low macrophage TME. In TME with low macrophage levels, patients with high levels of TGFB2 mRNA exhibited significantly shorter OS outcomes than patients with low TGFB2 mRNA levels (Median OS of 15.3 versus 72.7 months, p < 0.0001). Furthermore, multivariate Cox regression models were applied to control for age at diagnosis. Nine genes exhibited significant increases in hazard ratios for TGFB2 mRNA expression, marker gene mRNA expression, and a significant interaction term between TGFB2 and marker gene expression (mRNA for markers: C1QA, CD74, HLA-DQB1, HLA-DRB1, HLA-F, IFI27, IRF9, LGALS9, MARCO). The results of our study suggest that a combination of pharmacological tools can be used in treating PDAC patients, targeting both TGFB2 and the components of the type-I interferon signaling pathway. The significant statistical interaction between TGFB2 and the nine marker genes suggests that TGFB2 is a negative prognostic indicator at low levels of the IFN-I activated genes and TAM marker expression, including the immune checkpoint LGALS9 (upregulated 16.5-fold in tumor tissue; p < 0.0001).
Collapse
MESH Headings
- Humans
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/mortality
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/pathology
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/mortality
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Prognosis
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Female
- Male
- Gene Expression Regulation, Neoplastic
- Transforming Growth Factor beta2/genetics
- Transforming Growth Factor beta2/metabolism
- Interferon-Stimulated Gene Factor 3, gamma Subunit/genetics
- Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism
- Middle Aged
- Aged
- Receptor, Interferon alpha-beta/genetics
- Receptor, Interferon alpha-beta/metabolism
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Galectins
Collapse
Affiliation(s)
- Sanjive Qazi
- Oncotelic Therapeutics, 29397 Agoura Road, Suite 107, Agoura Hills, CA 91301, USA;
| | | |
Collapse
|
14
|
Silva AD, Hwang J, Marciel MP, Bellis SL. The pro-inflammatory cytokines IL-1β and IL-6 promote upregulation of the ST6GAL1 sialyltransferase in pancreatic cancer cells. J Biol Chem 2024; 300:107752. [PMID: 39260693 PMCID: PMC11470512 DOI: 10.1016/j.jbc.2024.107752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024] Open
Abstract
The ST6GAL1 sialyltransferase is overexpressed in multiple cancers, including pancreatic ductal adenocarcinoma (PDAC). ST6GAL1 adds an α2-6-linked sialic acid to N-glycosylated membrane receptors, which consequently modulates receptor structure and function. While many studies have investigated the effects of ST6GAL1 on cell phenotype, there is a dearth of knowledge regarding mechanisms that regulate ST6GAL1 expression. In the current study, we evaluated the regulation of ST6GAL1 by two pro-inflammatory cytokines, IL-1β and IL-6, which are abundant within the PDAC tumor microenvironment. Cytokine activity was monitored using the Suit-2 PDAC cell line and two Suit-2-derived metastatic subclones, S2-013 and S2-LM7AA. For all three cell models, treatment with IL-1β or IL-6 increased the expression of ST6GAL1 protein and mRNA. Specifically, IL-1β and IL-6 induced expression of the ST6GAL1 YZ mRNA isoform, which is driven by the P3 promoter. The ST6GAL1 H and X isoforms were not detected. Promoter reporter assays confirmed that IL-1β and IL-6 activated transcription from the P3 promoter. We then examined downstream signaling mechanisms. IL-1β is known to signal through the NFκB transcription factor, whereas IL-6 signals through the STAT3 transcription factor. CUT&RUN experiments revealed that IL-1β promoted the binding of NFκB to the ST6GAL1 P3 promoter, and IL-6 induced the binding of STAT3 to the P3 promoter. Finally, we determined that inhibitors of NFκB and STAT3 blocked the upregulation of ST6GAL1 stimulated by IL-1β and IL-6, respectively. Together, these results highlight a novel molecular pathway by which cytokines within the tumor microenvironment stimulate the upregulation of ST6GAL1 in PDAC cells.
Collapse
Affiliation(s)
- Austin D Silva
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jihye Hwang
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Michael P Marciel
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Susan L Bellis
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
15
|
Wang L, Baek S, Prasad G, Wildenthal J, Guo K, Sturgill D, Truongvo T, Char E, Pegoraro G, McKinnon K, Hoskins JW, Amundadottir LT, Arda HE. Predictive Prioritization of Enhancers Associated with Pancreas Disease Risk. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.07.611794. [PMID: 39314336 PMCID: PMC11418953 DOI: 10.1101/2024.09.07.611794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Genetic and epigenetic variations in regulatory enhancer elements increase susceptibility to a range of pathologies. Despite recent advances, linking enhancer elements to target genes and predicting transcriptional outcomes of enhancer dysfunction remain significant challenges. Using 3D chromatin conformation assays, we generated an extensive enhancer interaction dataset for the human pancreas, encompassing more than 20 donors and five major cell types, including both exocrine and endocrine compartments. We employed a network approach to parse chromatin interactions into enhancer-promoter tree models, facilitating a quantitative, genome-wide analysis of enhancer connectivity. With these tree models, we developed a machine learning algorithm to estimate the impact of enhancer perturbations on cell type-specific gene expression in the human pancreas. Orthogonal to our computational approach, we perturbed enhancer function in primary human pancreas cells using CRISPR interference and quantified the effects at the single-cell level through RNA FISH coupled with high-throughput imaging. Our enhancer tree models enabled the annotation of common germline risk variants associated with pancreas diseases, linking them to putative target genes in specific cell types. For pancreatic ductal adenocarcinoma, we found a stronger enrichment of disease susceptibility variants within acinar cell regulatory elements, despite ductal cells historically being assumed as the primary cell-of-origin. Our integrative approach-combining cell type-specific enhancer-promoter interaction mapping, computational models, and single-cell enhancer perturbation assays-produced a robust resource for studying the genetic basis of pancreas disorders.
Collapse
Affiliation(s)
- Li Wang
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Songjoon Baek
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gauri Prasad
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - John Wildenthal
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Konnie Guo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Sturgill
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Thucnhi Truongvo
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Erin Char
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Gianluca Pegoraro
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine McKinnon
- Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | - Jason W. Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology & Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - H. Efsun Arda
- Laboratory of Receptor Biology and Gene Expression, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
16
|
Do CTP, Prochnau JY, Dominguez A, Wang P, Rao MK. The Road Ahead in Pancreatic Cancer: Emerging Trends and Therapeutic Prospects. Biomedicines 2024; 12:1979. [PMID: 39335494 PMCID: PMC11428787 DOI: 10.3390/biomedicines12091979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
This review explores the challenges and emerging trends in pancreatic cancer therapy. In particular, we focus on the tumor microenvironment and the potential of immunotherapy for pancreatic cancer. Pancreatic ductal adenocarcinoma, characterized by its dense stromal architecture, presents unique challenges for effective treatment. Recent advancements have emphasized the role of the tumor microenvironment in therapeutic resistance and disease progression. We discuss novel strategies targeting the desmoplastic barrier and immunosuppressive cells to enhance immune cell infiltration and activation. Recent clinical trials, particularly those involving novel immunotherapeutic agents and tumor vaccines, are examined to understand their efficacy and limitations. Our analysis reveals that combining immunotherapy with chemotherapy, radiation therapy, or drugs targeting epigenetic processes shows promise, improving overall survival rates and response to treatment. For instance, trials utilizing checkpoint inhibitors in combination with standard chemotherapies have extended disease-free survival by up to 6 months compared to chemotherapy alone. Importantly, vaccines targeting specific tumor neoantigens have shown the potential to increase patient survival. However, these approaches also face significant challenges, including overcoming the immunosuppressive tumor microenvironment and enhancing the delivery and efficacy of therapeutic agents. By providing an overview of both the promising results and the obstacles encountered, this review aims to highlight ongoing efforts to refine immunotherapy approaches for better patient outcomes.
Collapse
Affiliation(s)
- Chris T P Do
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jack Y Prochnau
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Angel Dominguez
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Manjeet K Rao
- Greehey Children's Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| |
Collapse
|
17
|
Zhao Y, Feng Y, Sun F, Li L, Chen J, Song Y, Zhu W, Hu X, Li Z, Kong F, Du Y, Kong X. Optimized rAAV8 targeting acinar KLF4 ameliorates fibrosis in chronic pancreatitis via exosomes-enriched let-7s suppressing pancreatic stellate cells activation. Mol Ther 2024; 32:2624-2640. [PMID: 38956871 PMCID: PMC11405174 DOI: 10.1016/j.ymthe.2024.06.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 05/14/2024] [Accepted: 06/20/2024] [Indexed: 07/04/2024] Open
Abstract
Chronic pancreatitis (CP) is marked by progressive fibrosis and the activation of pancreatic stellate cells (PSCs), accompanied by the destruction of pancreatic parenchyma, leading to the loss of acinar cells (ACs). Few research studies have explored the mechanism by which damaged ACs (DACs) contribute to PSCs activation and pancreatic fibrosis. Currently, there are no effective drugs for curing CP or limiting the progression of pancreatic fibrosis. In this research, co-culture with intact acinar cells (IACs) suppressed PSC activation, while co-culture with DACs did the opposite. Krüppel-like factor 4 (KLF4) was significantly upregulated in DACs and was established as the key molecule that switches ACs from PSCs-suppressor to PSCs-activator. We revealed the exosomes of IACs contributed to the anti-activated function of IACs-CS on PSCs. MiRNome profiling showed that let-7 family is significantly enriched in IAC-derived exosomes (>30% miRNome), which partially mediates IACs' suppressive impacts on PSCs. Furthermore, it has been observed that the enrichment of let-7 in exosomes was influenced by the expression level of KLF4. Mechanistic studies demonstrated that KLF4 in ACs upregulated Lin28A, thereby decreasing let-7 levels in AC-derived exosomes, and thus promoting PSCs activation. We utilized an adeno-associated virus specifically targeting KLF4 in ACs (shKLF4-pAAV) to suppress PSCs activation in CP, resulting in reduced pancreatic fibrosis. IAC-derived exosomes hold potential as potent weapons against PSCs activation via let-7s, while activated KLF4/Lin28A signaling in DACs diminished such functions. ShKLF4-pAAV holds promise as a novel therapeutic approach for CP.
Collapse
Affiliation(s)
- Yating Zhao
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yongpu Feng
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Fengyuan Sun
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Lei Li
- Digestive Endoscopy Center, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Jiayu Chen
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Yingxiao Song
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Wenbo Zhu
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Xiulin Hu
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China
| | - Zhaoshen Li
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Fanyang Kong
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Yiqi Du
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| | - Xiangyu Kong
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital, National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
18
|
Mustafa M, Abbas K, Alam M, Habib S, Zulfareen, Hasan GM, Islam S, Shamsi A, Hassan I. Investigating underlying molecular mechanisms, signaling pathways, emerging therapeutic approaches in pancreatic cancer. Front Oncol 2024; 14:1427802. [PMID: 39087024 PMCID: PMC11288929 DOI: 10.3389/fonc.2024.1427802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Pancreatic adenocarcinoma, a clinically challenging malignancy constitutes a significant contributor to cancer-related mortality, characterized by an inherently poor prognosis. This review aims to provide a comprehensive understanding of pancreatic adenocarcinoma by examining its multifaceted etiologies, including genetic mutations and environmental factors. The review explains the complex molecular mechanisms underlying its pathogenesis and summarizes current therapeutic strategies, including surgery, chemotherapy, and emerging modalities such as immunotherapy. Critical molecular pathways driving pancreatic cancer development, including KRAS, Notch, and Hedgehog, are discussed. Current therapeutic strategies, including surgery, chemotherapy, and radiation, are discussed, with an emphasis on their limitations, particularly in terms of postoperative relapse. Promising research areas, including liquid biopsies, personalized medicine, and gene editing, are explored, demonstrating the significant potential for enhancing diagnosis and treatment. While immunotherapy presents promising prospects, it faces challenges related to immune evasion mechanisms. Emerging research directions, encompassing liquid biopsies, personalized medicine, CRISPR/Cas9 genome editing, and computational intelligence applications, hold promise for refining diagnostic approaches and therapeutic interventions. By integrating insights from genetic, molecular, and clinical research, innovative strategies that improve patient outcomes can be developed. Ongoing research in these emerging fields holds significant promise for advancing the diagnosis and treatment of this formidable malignancy.
Collapse
Affiliation(s)
- Mohd Mustafa
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Kashif Abbas
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mudassir Alam
- Department of Zoology, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Safia Habib
- Department of Biochemistry, J.N. Medical College, Faculty of Medicine, Aligarh Muslim University, Aligarh, India
| | - Zulfareen
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Sidra Islam
- Department of Inflammation & Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Anas Shamsi
- Center of Medical and Bio-Allied Health Sciences Research (CMBHSR), Ajman University, Ajman, United Arab Emirates
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
19
|
Yang K, Xie R, Xiao G, Zhao Z, Ding M, Lin T, Tsang YS, Chen Y, Xu D, Fei J. The integration of single-cell and bulk RNA-seq atlas reveals ERS-mediated acinar cell damage in acute pancreatitis. J Transl Med 2024; 22:346. [PMID: 38605381 PMCID: PMC11010368 DOI: 10.1186/s12967-024-05156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 04/02/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Acute pancreatitis (AP) is a clinically common acute abdominal disease, whose pathogenesis remains unclear. The severe patients usually have multiple complications and lack specific drugs, leading to a high mortality and poor outcome. Acinar cells are recognized as the initial site of AP. However, there are no precise single-cell transcriptomic profiles to decipher the landscape of acinar cells during AP, which are the missing pieces of jigsaw we aimed to complete in this study. METHODS A single-cell sequencing dataset was used to identify the cell types in pancreas of AP mice and to depict the transcriptomic maps in acinar cells. The pathways' activities were evaluated by gene sets enrichment analysis (GSEA) and single-cell gene sets variation analysis (GSVA). Pseudotime analysis was performed to describe the development trajectories of acinar cells. We also constructed the protein-protein interaction (PPI) network and identified the hub genes. Another independent single-cell sequencing dataset of pancreas samples from AP mice and a bulk RNA sequencing dataset of peripheral blood samples from AP patients were also analyzed. RESULTS In this study, we identified genetic markers of each cell type in the pancreas of AP mice based on single-cell sequencing datasets and analyzed the transcription changes in acinar cells. We found that acinar cells featured acinar-ductal metaplasia (ADM), as well as increased endocytosis and vesicle transport activity during AP. Notably, the endoplasmic reticulum stress (ERS) and ER-associated degradation (ERAD) pathways activated by accumulation of unfolded/misfolded proteins in acinar cells could be pivotal for the development of AP. CONCLUSION We deciphered the distinct roadmap of acinar cells in the early stage of AP at single-cell level. ERS and ERAD pathways are crucially important for acinar homeostasis and the pathogenesis of AP.
Collapse
Affiliation(s)
- Kaige Yang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Rongli Xie
- Department of General Surgery, Ruijin Hospital LuWan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guohui Xiao
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhifeng Zhao
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Min Ding
- Department of General Surgery, Ruijin Hospital LuWan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Tingyu Lin
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiu Sing Tsang
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Chen
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dan Xu
- Department of Emergency, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jian Fei
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Department of General Surgery, Ruijin Hospital LuWan Branch, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
20
|
Zheng C, Wang J, Wang J, Zhang Q, Liang T. Cell of Origin of Pancreatic cancer: Novel Findings and Current Understanding. Pancreas 2024; 53:e288-e297. [PMID: 38277420 PMCID: PMC11882172 DOI: 10.1097/mpa.0000000000002301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/08/2023] [Indexed: 01/28/2024]
Abstract
ABSTRACT Pancreatic ductal adenocarcinoma (PDAC) stands as one of the most lethal diseases globally, boasting a grim 5-year survival prognosis. The origin cell and the molecular signaling pathways that drive PDAC progression are not entirely understood. This review comprehensively outlines the categorization of PDAC and its precursor lesions, expounds on the creation and utility of genetically engineered mouse models used in PDAC research, compiles a roster of commonly used markers for pancreatic progenitors, duct cells, and acinar cells, and briefly addresses the mechanisms involved in the progression of PDAC. We acknowledge the value of precise markers and suitable tracing tools to discern the cell of origin, as it can facilitate the creation of more effective models for PDAC exploration. These conclusions shed light on our existing understanding of foundational genetically engineered mouse models and focus on the origin and development of PDAC.
Collapse
Affiliation(s)
- Chenlei Zheng
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Jianing Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Junli Wang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
| | - Qi Zhang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| | - Tingbo Liang
- From the Department of Hepatobiliary and Pancreatic Surgery
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases
- The Innovation Center for the Study of Pancreatic Diseases of Zhejiang Province
- Zhejiang University Cancer Center, Hangzhou, China
| |
Collapse
|
21
|
Nipper M, Xu Y, Liu J, Yin X, Liu Z, Ye Z, Zhang J, Chen Y, Wang P. TGFβ and Hippo Signaling Pathways Coordinate to Promote Acinar to Ductal Metaplasia in Human Pancreas. Cells 2024; 13:186. [PMID: 38247878 PMCID: PMC10813953 DOI: 10.3390/cells13020186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 12/04/2023] [Accepted: 12/09/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND & AIMS Acinar-to-ductal metaplasia (ADM) serves as a precursor event in the development of pancreatic ductal adenocarcinoma (PDAC) upon constitutive environmental and genetical stress. While the role of ADM in PDAC progression has been established, the molecular mechanisms underlying human ADM remain elusive. We previously demonstrated the induction of ADM in human acinar cells through the transforming growth factor beta (TGFβ) signaling pathway. We aim to investigate the interaction between TGFβ and Hippo pathways in mediating ADM. METHODS RNA-sequencing was conducted on sorted normal primary human acinar, ductal, and AD (acinar cells that have undergone ADM) cells. ATAC-seq analysis was utilized to reveal the chromatin accessibility in these three cell types. ChIP-Seq of YAP1, SMAD4, and H3K27ac was performed to identify the gene targets of YAP1 and SMAD4. The role of YAP1/TAZ in ADM-driven cell proliferation, as well as in oncogenic KRAS driven proliferation, was assessed using sphere formation assay. RESULTS AD cells have a unique transcription profile, with upregulated genes in open chromatin states in acinar cells. YAP1 and SMAD4 co-occupy the loci of ADM-related genes, including PROM1, HES1, and MMP7, co-regulating biological functions such as cell adhesion, cell migration, and inflammation. Overexpression of YAP1/TAZ promoted acinar cell proliferation but still required the TGFβ pathway. YAP1/TAZ were also crucial for TGFβ-induced sphere formation and were necessary for KRAS-induced proliferation. CONCLUSIONS Our study reveals the intricate transition between acinar and AD states in human pancreatic tissues. It unveils the complex interaction between the Hippo and TGF-β pathways during ADM, highlighting the pivotal role of YAP1/TAZ and SMAD4 in PDAC initiation.
Collapse
Affiliation(s)
- Michael Nipper
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (M.N.); (Y.X.); (J.L.); (X.Y.)
| | - Yi Xu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (M.N.); (Y.X.); (J.L.); (X.Y.)
| | - Jun Liu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (M.N.); (Y.X.); (J.L.); (X.Y.)
| | - Xue Yin
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (M.N.); (Y.X.); (J.L.); (X.Y.)
| | - Zhijie Liu
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA;
| | - Zhengqing Ye
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (Z.Y.); (Y.C.)
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Jianmin Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (Z.Y.); (Y.C.)
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pei Wang
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; (M.N.); (Y.X.); (J.L.); (X.Y.)
| |
Collapse
|