1
|
Fernández-Nogueira P, Linzoain-Agos P, Cueto-Remacha M, De la Guia-Lopez I, Recalde-Percaz L, Parcerisas A, Gascon P, Carbó N, Gutierrez-Uzquiza A, Fuster G, Bragado P. Role of semaphorins, neuropilins and plexins in cancer progression. Cancer Lett 2024; 606:217308. [PMID: 39490515 DOI: 10.1016/j.canlet.2024.217308] [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: 08/02/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Progress in understanding nervous system-cancer interconnections has emphasized the functional role of semaphorins (SEMAs) and their receptors, neuropilins (NRPs) and plexins (PLXNs), in cancer progression. SEMAs are a conserved and extensive family of broadly expressed soluble and membrane-associated proteins that were first described as regulators of axon guidance and neural and vascular development. However, recent advances have shown that they can have a dual role in cancer progression, acting either as tumor promoters or suppressors. SEMAs effects result from their interaction with specific co-receptors/receptors NRPs/PLXNs, that have also been described to play a role in cancer progression. They can influence both cancer cells and tumor microenvironment components modulating various aspects of tumorigenesis such as oncogenesis, tumor growth, invasion and metastatic spread or treatment resistance. In this review we focus on the role of these axon guidance signals and their receptors and co-receptors in various aspects of cancer. Furthermore, we also highlight their potential application as novel approaches for cancer treatment in the future.
Collapse
Affiliation(s)
- P Fernández-Nogueira
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - P Linzoain-Agos
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - M Cueto-Remacha
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - I De la Guia-Lopez
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - L Recalde-Percaz
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Parcerisas
- Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain
| | - P Gascon
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - N Carbó
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain
| | - A Gutierrez-Uzquiza
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain
| | - G Fuster
- Department of Biomedicine, School of Medicine, Universitat de Barcelona, 08028, Barcelona, Spain; Biosciences Department, Faculty of Sciences, Technology and Engineering, University of Vic. Central University of Catalonia (UVic-UCC), 08500, Vic, Catalonia, Spain; Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Institute of Biomedicine of the Universitat de Barcelona (IBUB), 08028, Barcelona, Spain; Tissue Repair and Regeneration Laboratory (TR2Lab), Institute of Research and Innovation of Life Sciences and Health, Catalunya Central (IRIS-CC), 08500, Vic, Catalonia, Spain.
| | - P Bragado
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Complutense University of Madrid, 28040, Madrid, Spain; Health Research Institute of the Hospital Clínico San Carlos, 28040, Madrid, Spain.
| |
Collapse
|
2
|
Thielman NRJ, Funes V, Davuluri S, Ibanez HE, Sun WC, Fu J, Li K, Muth S, Pan X, Fujiwara K, Dwayne L Thomas Ii, Henderson M, Teh SS, Zhu Q, Thompson E, Jaffee EM, Kolodkin A, Meng F, Zheng L. Semaphorin 3D promotes pancreatic ductal adenocarcinoma progression and metastasis through macrophage reprogramming. SCIENCE ADVANCES 2024; 10:eadp0684. [PMID: 39413197 DOI: 10.1126/sciadv.adp0684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Axon guidance molecules are frequently altered in pancreatic ductal adenocarcinoma (PDA) and influence PDA progression. However, the molecular mechanism remained unclear. Using genetically engineered mouse models to examine semaphorin 3D (SEMA3D), we identified a dual role for tumor- and nerve-derived SEMA3D in the malignant transformation of pancreatic epithelial cells and invasive PDA development. Pancreatic-specific knockout of the SEMA3D gene from the KRASG12D and TP53R172H mutation knock-in, PDX1-Cre(KPC) mouse model demonstrated delayed tumor initiation, prolonged survival, absence of metastasis, and reduced M2 macrophage expression. Mechanistically, tumor- and nerve-derived SEMA3D indirectly reprograms macrophages through KRASMUT-dependent ARF6 signaling in PDA cells, resulting in increased lactate production, which is sensed by GPCR132 on macrophages to stimulate protumorigenic M2 polarization. Multiplex immunohistochemistry demonstrated increased M2-polarized macrophages proximal to nerves in SEMA3D-expressing human PDA tissue. This study suggests that altered SEMA3D expression leads to an acquisition of cancer-promoting functions, and nerve-derived SEMA3D is "hijacked" by PDA cells to support growth and metastasis in a KRASMUT-dependent manner.
Collapse
Affiliation(s)
- Noelle R J Thielman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Vanessa Funes
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Sanjana Davuluri
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Molecular Microbiology and Immunology, Johns Hopkins University School of Public Health, Baltimore, MD 21287, USA
| | - Hector E Ibanez
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wei-Chih Sun
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Juan Fu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Keyu Li
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Division of Pancreatic Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Stephen Muth
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Xingyi Pan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Kenji Fujiwara
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Surgery, Kimura Hospital and Department of Surgery; Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Dwayne L Thomas Ii
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - MacKenzie Henderson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Selina Shiqing Teh
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Qingfeng Zhu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth Thompson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Skip Viragh Center for Pancreatic Cancer, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Cancer Convergence Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | - Alex Kolodkin
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neuroscience, Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Fengxi Meng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Shanghai Eye and ENT Hospital, Fudan University, Shanghai 200031, China
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Skip Viragh Center for Pancreatic Cancer, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
3
|
Brichkina A, Ems M, Suezov R, Singh R, Lutz V, Picard FSR, Nist A, Stiewe T, Graumann J, Daude M, Diederich WE, Finkernagel F, Chung HR, Bartsch DK, Roth K, Keber C, Denkert C, Huber M, Gress TM, Lauth M. DYRK1B blockade promotes tumoricidal macrophage activity in pancreatic cancer. Gut 2024; 73:1684-1701. [PMID: 38834297 PMCID: PMC11420735 DOI: 10.1136/gutjnl-2023-331854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 05/15/2024] [Indexed: 06/06/2024]
Abstract
OBJECTIVE Highly malignant pancreatic ductal adenocarcinoma (PDAC) is characterised by an abundant immunosuppressive and fibrotic tumour microenvironment (TME). Future therapeutic attempts will therefore demand the targeting of tumours and stromal compartments in order to be effective. Here we investigate whether dual specificity and tyrosine phosphorylation-regulated kinase 1B (DYRK1B) fulfil these criteria and represent a promising anticancer target in PDAC. DESIGN We used transplantation and autochthonous mouse models of PDAC with either genetic Dyrk1b loss or pharmacological DYRK1B inhibition, respectively. Mechanistic interactions between tumour cells and macrophages were studied in direct or indirect co-culture experiments. Histological analyses used tissue microarrays from patients with PDAC. Additional methodological approaches included bulk mRNA sequencing (transcriptomics) and proteomics (secretomics). RESULTS We found that DYRK1B is mainly expressed by pancreatic epithelial cancer cells and modulates the influx and activity of TME-associated macrophages through effects on the cancer cells themselves as well as through the tumour secretome. Mechanistically, genetic ablation or pharmacological inhibition of DYRK1B strongly attracts tumoricidal macrophages and, in addition, downregulates the phagocytosis checkpoint and 'don't eat me' signal CD24 on cancer cells, resulting in enhanced tumour cell phagocytosis. Consequently, tumour cells lacking DYRK1B hardly expand in transplantation experiments, despite their rapid growth in culture. Furthermore, combining a small-molecule DYRK1B-directed therapy with mammalian target of rapamycin inhibition and conventional chemotherapy stalls the growth of established tumours and results in a significant extension of life span in a highly aggressive autochthonous model of PDAC. CONCLUSION In light of DYRK inhibitors currently entering clinical phase testing, our data thus provide a novel and clinically translatable approach targeting both the cancer cell compartment and its microenvironment.
Collapse
Affiliation(s)
- Anna Brichkina
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
- Present address: Institute of Systems Immunology, Center for Tumor and Immune Biology, Marburg, Germany
| | - Miriam Ems
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Roman Suezov
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Rajeev Singh
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Veronika Lutz
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Felix S R Picard
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University Marburg, Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University Marburg, Marburg, Germany
- Institute for Molecular Oncology, German Center for Lung Research (DZL), Marburg, Germany
| | - Johannes Graumann
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- Institute of Translational Proteomics, Philipps University, Marburg, Germany
| | - Michael Daude
- Medicinal Chemistry Core Facility, Philipps University Marburg, Marburg, Germany
| | - Wibke E Diederich
- Medicinal Chemistry Core Facility, Philipps University Marburg, Marburg, Germany
- Department of Medicinal chemistry, Center for Tumor and Immune Biology, Marburg, Germany
| | - Florian Finkernagel
- Bioinformatics Core Facility, Center for Tumor and Immune Biology, Marburg, Germany
| | - Ho-Ryun Chung
- Institute for Medical Bioinformatics and Biostatistics, Institute for Molecular Biology and Tumor Research, Marburg, Germany
| | - Detlef K Bartsch
- Department of Visceral, Thoracic and Vascular Surgery, Philipps-University Marburg, Marburg, Germany
| | - Katrin Roth
- Cell Imaging Core Facility, Center for Tumor Biology and Immunology, Philipps-University Marburg, Marburg, Hessen, Germany
| | - Corinna Keber
- Institute of Pathology, University Hospital of Giessen-Marburg, Marburg, Germany
| | - Carsten Denkert
- Institute of Pathology, University Hospital of Giessen-Marburg, Marburg, Germany
| | - Magdalena Huber
- Institute of Systems Immunology, Philipps-Universitat Marburg, Marburg, Hessen, Germany
| | - Thomas M Gress
- Department of Gastroenterology, Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| | - Matthias Lauth
- Department of Gastroenterology Endocrinology and Metabolism, Center for Tumor and Immune Biology, Marburg, Germany
| |
Collapse
|
4
|
Alamri A. Sema-3E/PlexinD1 axis modulates dendritic cell phenotypes and functions: Current status and future implications. Hum Immunol 2024; 85:110815. [PMID: 38772051 DOI: 10.1016/j.humimm.2024.110815] [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: 01/27/2024] [Revised: 05/03/2024] [Accepted: 05/15/2024] [Indexed: 05/23/2024]
Abstract
This comprehensive research review explores the complex interplay between the Sema-3E/PlexinD1 axis and dendritic cells (DCs), highlighting its critical role in immune modulation with implications for clinical application Critical regulators of immune responses Dendritic cells are central to adaptive immunity, and the Sema-3E /PlexinD1 axis emerges as a key modulator affecting their phenotypes and functions Review delineates the impact of this signaling axis on DC maturation, migration, antigen presentation, and cytokine production, unravels its multifaceted role in shaping the immune response. Recognizing the limitations and gaps in current knowledge, the study highlights the need for further studies to condition downstream signaling events and related information experienced by the Sema-3E/PlexinD1 axis emphasizes the clarity of the immune system. The review concludes by identifying opportunities for translation, focusing on therapeutic and diagnostic potential. It highlights the importance of collaborative, interdisciplinary efforts to address the challenges and harness the therapeutic and pathological potential of targeting the Sema-3E/PlexinD1 axis, thus opening the way for transformative advances in immunology and clinical medicine.
Collapse
Affiliation(s)
- Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
5
|
Grimsrud MM, Forster M, Goeppert B, Hemmrich-Stanisak G, Sax I, Grzyb K, Braadland PR, Charbel A, Metzger C, Albrecht T, Steiert TA, Schlesner M, Manns MP, Vogel A, Yaqub S, Karlsen TH, Schirmacher P, Boberg KM, Franke A, Roessler S, Folseraas T. Whole-exome sequencing reveals novel cancer genes and actionable targets in biliary tract cancers in primary sclerosing cholangitis. Hepatol Commun 2024; 8:e0461. [PMID: 38967597 PMCID: PMC11227357 DOI: 10.1097/hc9.0000000000000461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Accepted: 03/13/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND People with primary sclerosing cholangitis (PSC) have a 20% lifetime risk of biliary tract cancer (BTC). Using whole-exome sequencing, we characterized genomic alterations in tissue samples from BTC with underlying PSC. METHODS We extracted DNA from formalin-fixed, paraffin-embedded tumor and paired nontumor tissue from 52 resection or biopsy specimens from patients with PSC and BTC and performed whole-exome sequencing. Following copy number analysis, variant calling, and filtering, putative PSC-BTC-associated genes were assessed by pathway analyses and annotated to targeted cancer therapies. RESULTS We identified 53 candidate cancer genes with a total of 123 nonsynonymous alterations passing filtering thresholds in 2 or more samples. Of the identified genes, 19% had not previously been implicated in BTC, including CNGA3, KRT28, and EFCAB5. Another subset comprised genes previously implicated in hepato-pancreato-biliary cancer, such as ARID2, ELF3, and PTPRD. Finally, we identified a subset of genes implicated in a wide range of cancers such as the tumor suppressor genes TP53, CDKN2A, SMAD4, and RNF43 and the oncogenes KRAS, ERBB2, and BRAF. Focal copy number variations were found in 51.9% of the samples. Alterations in potential actionable genes, including ERBB2, MDM2, and FGFR3 were identified and alterations in the RTK/RAS (p = 0.036), TP53 (p = 0.04), and PI3K (p = 0.043) pathways were significantly associated with reduced overall survival. CONCLUSIONS In this exome-wide characterization of PSC-associated BTC, we delineated both PSC-specific and universal cancer genes. Our findings provide opportunities for a better understanding of the development of BTC in PSC and could be used as a platform to develop personalized treatment approaches.
Collapse
Affiliation(s)
- Marit M. Grimsrud
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Michael Forster
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Benjamin Goeppert
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
- Institute of Pathology, Hospital RKH Kliniken Ludwigsburg, Ludwigsburg, Germany
- Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| | | | - Irmi Sax
- Biomedical Informatics, Data Mining and Data Analytics, University of Augsburg, Augsburg, Germany
| | - Krzysztof Grzyb
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Peder R. Braadland
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Alphonse Charbel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Carmen Metzger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thomas Albrecht
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Tim Alexander Steiert
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Matthias Schlesner
- Biomedical Informatics, Data Mining and Data Analytics, University of Augsburg, Augsburg, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Arndt Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | - Sheraz Yaqub
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Hepatobiliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Tom H. Karlsen
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Kirsten M. Boberg
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-University, Kiel, Germany
| | - Stephanie Roessler
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Trine Folseraas
- Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Norwegian PSC Research Center, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Research Institute of Internal Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
- Section for Gastroenterology, Department of Transplantation Medicine, Division of Surgery, Inflammatory Medicine and Transplantation, Oslo University Hospital Rikshospitalet, Oslo, Norway
| |
Collapse
|
6
|
Atkins D, Rosas JM, Månsson LK, Shahverdi N, Dey SS, Pitenis AA. Survival-Associated Cellular Response Maintained in Pancreatic Ductal Adenocarcinoma (PDAC) Switched Between Soft and Stiff 3D Microgel Culture. ACS Biomater Sci Eng 2024; 10:2177-2187. [PMID: 38466617 PMCID: PMC11005012 DOI: 10.1021/acsbiomaterials.3c01079] [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: 08/03/2023] [Revised: 02/26/2024] [Accepted: 02/26/2024] [Indexed: 03/13/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for about 90% of all pancreatic cancer cases. Five-year survival rates have remained below 12% since the 1970s, in part due to the difficulty in detection prior to metastasis (migration and invasion into neighboring organs and glands). Mechanical memory is a concept that has emerged over the past decade that may provide a path toward understanding how invading PDAC cells "remember" the mechanical properties of their diseased ("stiff", elastic modulus, E ≈ 10 kPa) microenvironment even while invading a healthy ("soft", E ≈ 1 kPa) microenvironment. Here, we investigated the role of mechanical priming by culturing a dilute suspension of PDAC (FG) cells within a 3D, rheologically tunable microgel platform from hydrogels with tunable mechanical properties. We conducted a suite of acute (short-term) priming studies where we cultured PDAC cells in either a soft (E ≈ 1 kPa) or stiff (E ≈ 10 kPa) environment for 6 h, then removed and placed them into a new soft or stiff 3D environment for another 18 h. Following these steps, we conducted RNA-seq analyses to quantify gene expression. Initial priming in the 3D culture showed persistent gene expression for the duration of the study, regardless of the subsequent environments (stiff or soft). Stiff 3D culture was associated with the downregulation of tumor suppressors (LATS1, BCAR3, CDKN2C), as well as the upregulation of cancer-associated genes (RAC3). Immunofluorescence staining (BCAR3, RAC3) further supported the persistence of this cellular response, with BCAR3 upregulated in soft culture and RAC3 upregulated in stiff-primed culture. Stiff-primed genes were stratified against patient data found in The Cancer Genome Atlas (TCGA). Upregulated genes in stiff-primed 3D culture were associated with decreased survival in patient data, suggesting a link between patient survival and mechanical priming.
Collapse
Affiliation(s)
- Dixon
J. Atkins
- Department
of Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Jonah M. Rosas
- Department
of Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Lisa K. Månsson
- Materials
Department, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| | - Nima Shahverdi
- Molecular,
Cellular, and Developmental Biology Department, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Siddharth S. Dey
- Department
of Chemical Engineering, University
of California Santa Barbara, Santa
Barbara, California 93106, United States
- Department
of Bioengineering, University of California
Santa Barbara, Santa Barbara, California 93106, United States
| | - Angela A. Pitenis
- Materials
Department, University of California Santa
Barbara, Santa
Barbara, California 93106, United States
| |
Collapse
|
7
|
Dudgeon C, Casabianca A, Harris C, Ogier C, Bellina M, Fiore S, Bernet A, Ducarouge B, Goldschneider D, Su X, Pitarresi J, Hezel A, De S, Narrow W, Soliman F, Shields C, Vendramini-Costa DB, Prela O, Wang L, Astsaturov I, Mehlen P, Carpizo DR. Netrin-1 feedforward mechanism promotes pancreatic cancer liver metastasis via hepatic stellate cell activation, retinoid, and ELF3 signaling. Cell Rep 2023; 42:113369. [PMID: 37922311 DOI: 10.1016/j.celrep.2023.113369] [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: 01/03/2023] [Revised: 09/04/2023] [Accepted: 10/18/2023] [Indexed: 11/05/2023] Open
Abstract
The biology of metastatic pancreatic ductal adenocarcinoma (PDAC) is distinct from that of the primary tumor due to changes in cell plasticity governed by a distinct transcriptome. Therapeutic strategies that target this distinct biology are needed. We detect an upregulation of the neuronal axon guidance molecule Netrin-1 in PDAC liver metastases that signals through its dependence receptor (DR), uncoordinated-5b (Unc5b), to facilitate metastasis in vitro and in vivo. The mechanism of Netrin-1 induction involves a feedforward loop whereby Netrin-1 on the surface of PDAC-secreted extracellular vesicles prepares the metastatic niche by inducing hepatic stellate cell activation and retinoic acid secretion that in turn upregulates Netrin-1 in disseminated tumor cells via RAR/RXR and Elf3 signaling. While this mechanism promotes PDAC liver metastasis, it also identifies a therapeutic vulnerability, as it can be targeted using anti-Netrin-1 therapy to inhibit metastasis using the Unc5b DR cell death mechanism.
Collapse
Affiliation(s)
- Crissy Dudgeon
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Anthony Casabianca
- Department of Surgery, Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Chris Harris
- Department of Surgery, Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Charline Ogier
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Mélanie Bellina
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée "La Ligue," LabEx DEVweCAN, Institut Convergence PLAsCAN, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, 69008 Lyon, France; Netris Pharma, 69008 Lyon, France
| | - Stephany Fiore
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée "La Ligue," LabEx DEVweCAN, Institut Convergence PLAsCAN, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, 69008 Lyon, France
| | - Agnes Bernet
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée "La Ligue," LabEx DEVweCAN, Institut Convergence PLAsCAN, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, 69008 Lyon, France; Netris Pharma, 69008 Lyon, France
| | | | | | - Xiaoyang Su
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Jason Pitarresi
- Department of Medicine, Division of Hematology/Oncology, University of Massachusetts, Worcester, MA, USA
| | - Aram Hezel
- Department of Medicine, Division of Medical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Subhajyoti De
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Wade Narrow
- Department of Surgery, Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | - Fady Soliman
- Rutgers Robert Wood-Johnson Medical School, New Brunswick, NJ, USA
| | - Cory Shields
- Wilmot Cancer Center, University of Rochester, Rochester, NY, USA
| | | | - Orjola Prela
- Department of Surgery, Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Lan Wang
- Department of Pathology and Laboratory Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Igor Astsaturov
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory - Equipe labellisée "La Ligue," LabEx DEVweCAN, Institut Convergence PLAsCAN, Centre de Recherche en Cancérologie de Lyon (CRCL), INSERM U1052-CNRS UMR5286, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, 69008 Lyon, France; Netris Pharma, 69008 Lyon, France
| | - Darren R Carpizo
- Department of Surgery, Division of Surgical Oncology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA; Wilmot Cancer Center, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
8
|
Thielman NRJ, Funes V, Davuluri S, Ibanez HE, Sun WC, Fu J, Li K, Muth S, Pan X, Fujiwara K, Thomas D, Henderson M, Teh SS, Zhu Q, Thompson E, Jaffee EM, Kolodkin A, Meng F, Zheng L. Tumor- and Nerve-Derived Axon Guidance Molecule Promotes Pancreatic Ductal Adenocarcinoma Progression and Metastasis through Macrophage Reprogramming. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563862. [PMID: 37961340 PMCID: PMC10634802 DOI: 10.1101/2023.10.24.563862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Axon guidance molecules were found to be the gene family most frequently altered in pancreatic ductal adenocarcinoma (PDA) through mutations and copy number changes. However, the exact molecular mechanism regarding PDA development remained unclear. Using genetically engineered mouse models to examine one of the axon guidance molecules, semaphorin 3D (SEMA3D), we found a dual role for tumor-derived SEMA3D in malignant transformation of pancreatic epithelial cells and a role for nerve-derived SEMA3D in PDA development. This was demonstrated by the pancreatic-specific knockout of the SEMA3D gene from the KRAS G12D and TP53 R 172 H mutation knock-in, PDX1-Cre (KPC) mouse model which demonstrated a delayed tumor initiation and growth comparing to the original KPC mouse model. Our results showed that SEMA3D knockout skews the macrophages in the pancreas away from M2 polarization, providing a potential mechanistic role of tumor-derived SEMA3D in PDA development. The KPC mice with the SEMA3D knockout remained metastasis-free, however, died from primary tumor growth. We then tested the hypothesis that a potential compensation mechanism could result from SEMA3D which is naturally expressed by the intratumoral nerves. Our study further revealed that nerve-derived SEMA3D does not reprogram macrophages directly, but reprograms macrophages indirectly through ARF6 signaling and lactate production in PDA tumor cells. SEMA3D increases tumor-secreted lactate which is sensed by GPCR132 on macrophages and subsequently stimulates pro-tumorigenic M2 polarization in vivo. Tumor intrinsic- and extrinsic-SEMA3D induced ARF6 signaling through its receptor Plexin D1 in a mutant KRAS-dependent manner. Consistently, RNA sequencing database analysis revealed an association of higher KRAS MUT expression with an increase in SEMA3D and ARF6 expression in human PDAs. Moreover, multiplex immunohistochemistry analysis showed an increased number of M2-polarized macrophages proximal to nerves in human PDA tissue expressing SEMA3D. Thus, this study suggests altered expression of SEMA3D in tumor cells lead to acquisition of cancer-promoting functions and the axon guidance signaling originating from nerves is "hijacked" by tumor cells to support their growth. Other axon guidance and neuronal development molecules may play a similar dual role which is worth further investigation. One sentence summary Tumor- and nerve-derived SEMA3D promotes tumor progression and metastasis through macrophage reprogramming in the tumor microenvironment. STATEMENT OF SIGNIFICANCE This study established the dual role of axon guidance molecule, SEMA3D, in the malignant transformation of pancreatic epithelial cells and of nerve-derived SEMA3D in PDA progression and metastasis. It revealed macrophage reprogramming as the mechanism underlying bothroles. Together, this research elucidated how inflammatory responses promote invasive PDA progression and metastasis through an oncogenic process.
Collapse
|
9
|
Fard D, Giraudo E, Tamagnone L. Mind the (guidance) signals! Translational relevance of semaphorins, plexins, and neuropilins in pancreatic cancer. Trends Mol Med 2023; 29:817-829. [PMID: 37598000 DOI: 10.1016/j.molmed.2023.07.009] [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: 06/24/2023] [Revised: 07/19/2023] [Accepted: 07/19/2023] [Indexed: 08/21/2023]
Abstract
Pancreatic cancer is a major cause of demise worldwide. Although key associated genetic changes have been discovered, disease progression is sustained by pathogenic mechanisms that are poorly understood at the molecular level. In particular, the tissue microenvironment of pancreatic adenocarcinoma (PDAC) is usually characterized by high stromal content, scarce recruitment of immune cells, and the presence of neuronal fibers. Semaphorins and their receptors, plexins and neuropilins, comprise a wide family of regulatory signals that control neurons, endothelial and immune cells, embryo development, and normal tissue homeostasis, as well as the microenvironment of human tumors. We focus on the role of these molecular signals in pancreatic cancer progression, as revealed by experimental research and clinical studies, including novel approaches for cancer treatment.
Collapse
Affiliation(s)
- Damon Fard
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy
| | - Enrico Giraudo
- Department of Science and Drug Technology, University of Turin, Turin, Italy; Candiolo Cancer Institute, FPO IRCCS, Candiolo, Turin, Italy
| | - Luca Tamagnone
- Università Cattolica del Sacro Cuore, Department of Life Sciences and Public Health, Rome, Italy; Fondazione Policlinico Gemelli, IRCCS, Rome, Italy.
| |
Collapse
|
10
|
Martínez-Ramos S, Rafael-Vidal C, Malvar-Fernández B, Rodriguez-Trillo A, Veale D, Fearon U, Conde C, Conde-Aranda J, Radstake TRDJ, Pego-Reigosa JM, Reedquist KA, García S. HOXA5 is a key regulator of class 3 semaphorins expression in the synovium of rheumatoid arthritis patients. Rheumatology (Oxford) 2023; 62:2621-2630. [PMID: 36398888 PMCID: PMC10321103 DOI: 10.1093/rheumatology/keac654] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 11/08/2022] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE Class 3 semaphorins are reduced in the synovial tissue of RA patients and these proteins are involved in the pathogenesis of the disease. The aim of this study was to identify the transcription factors involved in the expression of class 3 semaphorins in the synovium of RA patients. METHODS Protein and mRNA expression in synovial tissue from RA and individuals at risk (IAR) patients, human umbilical vein endothelial cells (HUVEC) and RA fibroblast-like synoviocytes (FLS) was determined by ELISA, immunoblotting and quantitative PCR. TCF-3, EBF-1 and HOXA5 expression was knocked down using siRNA. Cell viability, migration and invasion were determined using MTT, calcein, wound closure and invasion assays, respectively. RESULTS mRNA expression of all class 3 semaphorins was significantly lower in the synovium of RA compared with IAR patients. In silico analysis suggested TCF-3, EBF-1 and HOXA5 as transcription factors involved in the expression of these semaphorins. TCF-3, EBF-1 and HOXA5 silencing significantly reduced the expression of several class 3 semaphorin members in FLS and HUVEC. Importantly, HOXA5 expression was significantly reduced in the synovium of RA compared with IAR patients and was negatively correlated with clinical disease parameters. Additionally, TNF-α down-regulated the HOXA5 expression in FLS and HUVEC. Finally, HOXA5 silencing enhanced the migratory and invasive capacities of FLS and the viability of HUVEC. CONCLUSION HOXA5 expression is reduced during the progression of RA and could be a novel therapeutic strategy for modulating the hyperplasia of the synovium, through the regulation of class 3 semaphorins expression.
Collapse
Affiliation(s)
- Sara Martínez-Ramos
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Carlos Rafael-Vidal
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Beatriz Malvar-Fernández
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Angela Rodriguez-Trillo
- Laboratorio de Reumatología Experimental y Observacional, Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico, Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Douglas Veale
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Rheumatology EULAR Centre of Excellence, St Vincent's University Hospital and University College Dublin, Dublin, Ireland
- Department of Molecular Rheumatology, Trinity Biomedical Science Institute, Trinity College Dublin, Dublin, Ireland
| | - Carmen Conde
- Laboratorio de Reumatología Experimental y Observacional, Servicio de Reumatología, Instituto de Investigación Sanitaria de Santiago (IDIS), Hospital Clínico, Universitario de Santiago de Compostela (CHUS), Servizo Galego de Saude (SERGAS), Santiago de Compostela, Spain
| | - Javier Conde-Aranda
- Molecular and Cellular Gastroenterology, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Timothy R D J Radstake
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Jose María Pego-Reigosa
- Rheumatology & Immuno-mediated Diseases Research Group (IRIDIS), Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO, Vigo, Spain
- Rheumatology Department, University Hospital Complex of Vigo, Vigo, Spain
| | - Kris A Reedquist
- Department of Rheumatology and Clinical Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, University of Utrecht, Utrecht, The Netherlands
| | - Samuel García
- Correspondence to: Samuel García, Rheumatology & Immune-mediated Diseases (IRIDIS) Group, Galicia Sur Health Research Institute (IIS Galicia Sur), Hospital Álvaro Cunqueiro, Estrada Clara Campoamor No. 341, Beade, 36312 Vigo (Pontevedra), Spain. E-mail:
| |
Collapse
|
11
|
Bica C, Tirpe A, Nutu A, Ciocan C, Chira S, Gurzau ES, Braicu C, Berindan-Neagoe I. Emerging roles and mechanisms of semaphorins activity in cancer. Life Sci 2023; 318:121499. [PMID: 36775114 DOI: 10.1016/j.lfs.2023.121499] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/08/2023] [Accepted: 02/08/2023] [Indexed: 02/12/2023]
Abstract
Semaphorins are regulatory molecules that are linked to the modulation of several cancer processes, such as angiogenesis, cancer cell invasiveness and metastasis, tumor growth, as well as cancer cell survival. Semaphorin (SEMA) activity depends on the cancer histotypes and their particularities. In broad terms, the effects of SEMAs result from their interaction with specific receptors/co-receptors - Plexins, Neuropilins and Integrins - and the subsequent effects upon the downstream effectors (e.g. PI3K/AKT, MAPK/ERK). The present article serves as an integrative review work, discussing the broad implications of semaphorins in cancer, focusing on cell proliferation/survival, angiogenesis, invasion, metastasis, stemness, and chemo-resistance/response whilst highlighting their heterogeneity as a family. Herein, we emphasized that semaphorins are largely implicated in cancer progression, interacting with the tumor microenvironment components. Whilst some SEMAs (e.g. SEMA3A, SEMA3B) function widely as tumor suppressors, others (e.g. SEMA3C) act as pro-tumor semaphorins. The differences observed in terms of the biological structure of SEMAs and the particularities of each cancer histotypes require that each semaphorin be viewed as a unique entity, and its roles must be researched accordingly. A more in-depth and comprehensive view of the molecular mechanisms that promote and sustain the malignant behavior of cancer cells is of utmost importance.
Collapse
Affiliation(s)
- Cecilia Bica
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Alexandru Tirpe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania; Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 8 Victor Babes Street, 400012 Cluj-Napoca, Romania.
| | - Andreea Nutu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Cristina Ciocan
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Sergiu Chira
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Eugen S Gurzau
- Cluj School of Public Health, College of Political, Administrative and Communication Sciences, Babes-Bolyai University, 7 Pandurilor Street, Cluj-Napoca, Romania; Environmental Health Center, 58 Busuiocului Street, 400240 Cluj-Napoca, Romania.
| | - Cornelia Braicu
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca 400337, Romania.
| |
Collapse
|
12
|
Liu D, Li J, Qi F, You H. Semaphorins and their receptors in pancreatic cancer: Mechanisms and therapeutic opportunities. Front Oncol 2023; 12:1106762. [PMID: 36713527 PMCID: PMC9880980 DOI: 10.3389/fonc.2022.1106762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 12/23/2022] [Indexed: 01/15/2023] Open
Abstract
Pancreatic cancer (PC) is a malignant tumor with high malignancy that is difficult to diagnose and treat. PC is a major medical problem because of its low early diagnosis rate, high surgical mortality rate, low cure rate, and expensive related testing cost. Therefore, the significance of finding new markers for PC is self-evident. Semaphorins (Semas) have been shown to affect angiogenesis and lymphangiogenesis and can also directly affect the behavior of tumor cells. The expression and related action targets of its family members on PC are summarized in this review.
Collapse
Affiliation(s)
- Dahai Liu
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Jie Li
- School of Medicine, Foshan University, Foshan, Guangdong, China
| | - Fei Qi
- Department of Pulmonary and Critical Care Medicine, Peking University Shenzhen Hospital, Shenzhen, China
| | - Hua You
- Department of Pediatric Hematology and Oncology, Children’s Hospital of Chongqing Medical University, Chongqing, China,Laboratory for Excellence in Systems Biomedicine of Pediatric Oncology, Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China,*Correspondence: Hua You,
| |
Collapse
|
13
|
The Role of Neural Signaling in the Pancreatic Cancer Microenvironment. Cancers (Basel) 2022; 14:cancers14174269. [PMID: 36077804 PMCID: PMC9454556 DOI: 10.3390/cancers14174269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Pancreatic cancer is a highly lethal malignant disease with a dense stroma, called the tumor microenvironment. Accumulating evidence indicates the important role of sympathetic, parasympathetic, and sensory nerves in the tumor microenvironment of various cancers, including pancreatic cancer. Cancer cells and neural cells interact with each other to form a complex network and cooperatively promote cancer growth and invasion. In this review article, we describe the current understanding of the role of nerves in the tumor microenvironment. Abstract Pancreatic cancer is one of the most lethal malignant diseases. Various cells in the tumor microenvironment interact with tumor cells and orchestrate to support tumor progression. Several kinds of nerves are found in the tumor microenvironment, and each plays an essential role in tumor biology. Recent studies have shown that sympathetic, parasympathetic, and sensory neurons are found in the pancreatic cancer microenvironment. Neural signaling not only targets neural cells, but tumor cells and immune cells via neural receptors expressed on these cells, through which tumor growth, inflammation, and anti-tumor immunity are affected. Thus, these broad-range effects of neural signaling in the pancreatic cancer microenvironment may represent novel therapeutic targets. The modulation of neural signaling may be a therapeutic strategy targeting the whole tumor microenvironment. In this review, we describe the current understanding of the role of nerves in the tumor microenvironment of various cancers, with an emphasis on pancreatic cancer. We also discuss the underlying mechanisms and the possibility of therapeutic applications.
Collapse
|
14
|
Brisset M, Grandin M, Bernet A, Mehlen P, Hollande F. Dependence receptors: new targets for cancer therapy. EMBO Mol Med 2021; 13:e14495. [PMID: 34542930 PMCID: PMC8573599 DOI: 10.15252/emmm.202114495] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/11/2021] [Accepted: 08/11/2021] [Indexed: 12/22/2022] Open
Abstract
Dependence receptors are known to promote survival and positive signaling such as proliferation, migration, and differentiation when activated, but to actively trigger apoptosis when unbound to their ligand. Their abnormal regulation was shown to be an important feature of tumorigenesis, allowing cancer cells to escape apoptosis triggered by these receptors while promoting in parallel major aspects of tumorigenesis such as proliferation, angiogenesis, invasiveness, and chemoresistance. This involvement in multiple cancer hallmarks has raised interest in dependence receptors as targets for cancer therapy. Although additional studies remain necessary to fully understand the complexity of signaling pathways activated by these receptors and to target them efficiently, it is now clear that dependence receptors represent very exciting targets for future cancer treatment. This manuscript reviews current knowledge on the contribution of dependence receptors to cancer and highlights the potential for therapies that activate pro-apoptotic functions of these proteins.
Collapse
Affiliation(s)
- Morgan Brisset
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| | - Mélodie Grandin
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| | - Agnès Bernet
- Apoptosis, Cancer and Development LaboratoryCentre de Recherche en Cancérologie de Lyon, INSERM U1052‐CNRS UMR5286Centre Léon BérardUniversité de LyonLyonFrance
| | - Patrick Mehlen
- Apoptosis, Cancer and Development LaboratoryCentre de Recherche en Cancérologie de Lyon, INSERM U1052‐CNRS UMR5286Centre Léon BérardUniversité de LyonLyonFrance
| | - Frédéric Hollande
- Department of Clinical Pathology, Victorian Comprehensive Cancer CentreThe University of MelbourneMelbourneVic.Australia
- University of Melbourne Centre for Cancer ResearchVictorian Comprehensive Cancer CentreMelbourneVic.Australia
| |
Collapse
|
15
|
Ahammad I. A comprehensive review of tumor proliferative and suppressive role of semaphorins and therapeutic approaches. Biophys Rev 2020; 12:1233-1247. [PMID: 32577918 PMCID: PMC7575654 DOI: 10.1007/s12551-020-00709-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/15/2020] [Indexed: 01/05/2023] Open
Abstract
Semaphorins have been traditionally known as axon guidance proteins that negatively regulate axonal growth. However, in the past couple of decades, their versatile role in so many other biological processes has come to prominence as well. One such example is their role in cancer. In this review article, the focus was on the tumor proliferative and tumor suppressive role of all 20 semaphorin family members under the 7 semaphorin classes found in vertebrates and invertebrates as well as the ongoing and emerging therapeutic approaches to combat semaphorin-mediated cancers. Except sema6C, 19 of the 20 non-viral semaphorin family members have been discovered to be associated with cancer in one way or another. Eleven semaphorin family members have been discovered to be tumor proliferative and 8 to be tumor suppressive. Six therapeutic avenues and their safety profiles have been discussed which are currently at use or at the various stages of development. Finally, perspectives on which approach is the best for treating cancers associated with semaphorins have been given.
Collapse
Affiliation(s)
- Ishtiaque Ahammad
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh.
| |
Collapse
|
16
|
Song W, Ren J, Wang C, Ge Y, Fu T. Analysis of Circular RNA-Related Competing Endogenous RNA Identifies the Immune-Related Risk Signature for Colorectal Cancer. Front Genet 2020; 11:505. [PMID: 32582276 PMCID: PMC7283524 DOI: 10.3389/fgene.2020.00505] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
Background Recent papers have described circular RNAs (circRNAs) playing important roles in the development and progression of colorectal cancer (CRC). However, the expression profiles of circRNAs and their functions in CRC have rarely been studied. The objective was to identify circRNAs involved in the carcinogenesis and progression of CRC and to explore potential molecular mechanisms as a competitive endogenous RNA (ceRNA). Moreover, we aimed to establish an immune-related gene signature for predicting the overall survival (OS) of CRC. Methods The expression patterns of circRNA, miRNA, mRNA, and clinicopathological data were collected from the GEO and TCGA databases. A ceRNA network would be established, and the functional enrichment analyses were performed. The protein-protein interaction network (PPI) was constructed, and hub genes were identified using a cytohub plugin. Subsequently, an immune-related signature was developed based on mRNAs in the ceRNA network. In addition, OS-nomogram was constructed by combining an immune-related signature and clinicopathological characterization to predict the OS. Results We established a circRNA-miRNA-mRNA ceRNA network. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that the mRNAs were mainly enriched in neuroactive ligand-receptor interaction, Wnt signaling pathway, cell adhesion molecules (CAMs), and renin secretion. PPI network and module analysis identified 10 hub genes, and the circRNA-miRNA hub gene regulatory modules was established. After univariate and multivariate analysis, seven immune-related genes in the ceRNA network were used to construct the immune-related signature. Patients were divided into low-risk and high-risk groups, and there were significant differences in the OS. The ROC of the nomogram indicated the satisfactory accuracy and predictive power. Furthermore, we established a prognostic nomogram based on immune-related risk score and clinical characterization. The ROC and calibration curves revealed the accuracy of the nomogram. In addition, the high-risk score was positively correlated with six immune infiltrating cells (P < 0.05). Conclusion We screened the key genes and established a circRNA-related ceRNA network involved in CRC, which will assist in understanding the molecular mechanisms underlying the carcinogenesis and progression. Moreover, our proposed immune-based signature may predict survival and reflect the immune status of CRC patients.
Collapse
Affiliation(s)
- Wei Song
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Ren
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, China
| | - Chuntao Wang
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuhang Ge
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tao Fu
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
17
|
Haider MT, Saito H, Zarrer J, Uzhunnumpuram K, Nagarajan S, Kari V, Horn-Glander M, Werner S, Hesse E, Taipaleenmäki H. Breast cancer bone metastases are attenuated in a Tgif1-deficient bone microenvironment. Breast Cancer Res 2020; 22:34. [PMID: 32272947 PMCID: PMC7146874 DOI: 10.1186/s13058-020-01269-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/20/2020] [Indexed: 12/21/2022] Open
Abstract
Background Osteoclast activation is a hallmark of breast cancer-induced bone disease while little is known about the role of osteoblasts in this process. Recently, we identified the homeodomain protein TG-interacting factor-1 (Tgif1) as a crucial regulator of osteoblast function. In this study, we demonstrate that lack of Tgif1 also restricts the progression of breast cancer bone metastases. Methods Transwell migration assays were used to investigate the osteoblast-breast cancer cell interaction in vitro. Molecular analyses included RNA sequencing, immunoblotting, and qRT-PCR. To determine the role of Tgif1 in metastatic bone disease, 4T1 breast cancer cells were injected intracardially into mice with a germ line deletion of Tgif1 (Tgif1−/−) or control littermates (Tgif1+/+). Progression of bone metastases and alterations in the bone microenvironment were assessed using bioluminescence imaging, immunofluorescence staining, confocal microscopy, and histomorphometry. Results Medium conditioned by osteoblasts stimulated breast cancer cell migration, indicating a potential role of osteoblasts during bone metastasis progression. Tgif1 expression was strongly increased in osteoblasts upon stimulation by breast cancer cells, demonstrating the implication of Tgif1 in the osteoblast-breast cancer cell interaction. Indeed, conditioned medium from osteoblasts of Tgif1−/− mice failed to induce breast cancer cell migration compared to control, suggesting that Tgif1 in osteoblasts augments cancer cell motility. Semaphorin 3E (Sema3E), which is abundantly secreted by Tgif1−/− osteoblasts, dose-dependently reduced breast cancer cell migration while silencing of Sema3E expression in Tgif1−/− osteoblasts partially restored the impaired migration. In vivo, we observed a decreased number of breast cancer bone metastases in Tgif1−/− mice compared to control littermates. Consistently, the presence of single breast cancer cells or micro-metastases in the tibiae was reduced in Tgif1−/− mice. Breast cancer cells localized in close proximity to Endomucin-positive vascular cells as well as to osteoblasts. Although Tgif1 deficiency did not affect the bone marrow vasculature, the number and activity of osteoblasts were reduced compared to control. This suggests that the protective effect on bone metastases might be mediated by osteoblasts rather than by the bone marrow vasculature. Conclusion We propose that the lack of Tgif1 in osteoblasts increases Sema3E expression and attenuates breast cancer cell migration as well as metastases formation.
Collapse
Affiliation(s)
- Marie-Therese Haider
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hiroaki Saito
- Institute of Molecular Musculoskeletal Research, University Hospital, LMU Munich, Munich, Germany
| | - Jennifer Zarrer
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kevin Uzhunnumpuram
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sankari Nagarajan
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen Center for Molecular Biosciences, Göttingen, Germany.,Present address: Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, UK
| | - Vijayalakshmi Kari
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Göttingen Center for Molecular Biosciences, Göttingen, Germany
| | - Michael Horn-Glander
- University Cancer Center Hamburg, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Werner
- Institute of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Eric Hesse
- Institute of Molecular Musculoskeletal Research, University Hospital, LMU Munich, Munich, Germany
| | - Hanna Taipaleenmäki
- Molecular Skeletal Biology Laboratory, Department of Trauma, Hand and Reconstructive Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
18
|
Zhang X, Klamer B, Li J, Fernandez S, Li L. A pan-cancer study of class-3 semaphorins as therapeutic targets in cancer. BMC Med Genomics 2020; 13:45. [PMID: 32241267 PMCID: PMC7118829 DOI: 10.1186/s12920-020-0682-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Initially characterized as axon guidance factors, semaphorins also have been implicated to have critical roles in multiple physiological and developmental functions, including the regulation of immune responses, angiogenesis, organ formation, and the etiology of multiple forms of cancer. Moreover, their contribution in immunity and the regulation of tumour microenvironment is becoming increasingly recognized. Here, we provide a comprehensive analysis of class-3 semaphorins, the only secreted family of genes among veterbrate semaphorins, in terms of their expression profiles and their association with patient survival. We also relate their role with immune subtypes, tumour microenvironment, and drug sensitivity using a pan-cancer study. RESULTS Expression profiles of class-3 semaphorins (SEMA3s) and their association with patient survival and tumour microenvironment were studied in 31 cancer types using the TCGA pan-cancer data. The expression of SEMA3 family varies in different cancer types with striking inter- and intra- cancer heterogeneity. In general, our results show that SEMA3A, SEMA3C, and SEMA3F are primarily upregulated in cancer cells, while the rest of SEMA3s are mainly down-regulated in the tested tumours. The expression of SEMA3 family members was frequently associated with patient overall survival. However, the direction of the association varied with regards to the particular SEMA3 isoform queried and the specific cancer type tested. More specifically, SEMA3A and SEMA3E primarily associate with a poor prognosis of survival, while SEMA3G typically associates with survival advantage. The rest of SEMA3s show either survival advantage or disadvantage dependent on cancer type. In addition, all SEMA3 genes show significant association with immune infiltrate subtypes, and they also correlate with level of stromal cell infiltration and tumour cell stemness with various degrees. Finally, our study revealed that SEMA3 genes, especially SEMA3C and SEMA3F may contribute to drug induced cancer cell resistance. CONCLUSIONS Our systematic analysis of class-3 semaphorin gene expression and their association with immune infiltrates, tumour microenvironment and cancer patient outcomes highlights the need to study each SEMA3 member as a separate entity within each specific cancer type. Also our study validated the identification of class-3 semaphorin signals as promising therapeutic targets in cancer although further laboratory validation still needed.
Collapse
Affiliation(s)
- Xiaoli Zhang
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA.
| | - Brett Klamer
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| | - Jin Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| | - Soledad Fernandez
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| | - Lang Li
- Department of Biomedical Informatics, College of Medicine, The Ohio State University, 320B Lincoln Tower, 1800 Cannon Dr., Columbus, OH, 43210, USA
| |
Collapse
|
19
|
Jurcak NR, Rucki AA, Muth S, Thompson E, Sharma R, Ding D, Zhu Q, Eshleman JR, Anders RA, Jaffee EM, Fujiwara K, Zheng L. Axon Guidance Molecules Promote Perineural Invasion and Metastasis of Orthotopic Pancreatic Tumors in Mice. Gastroenterology 2019; 157:838-850.e6. [PMID: 31163177 PMCID: PMC6707836 DOI: 10.1053/j.gastro.2019.05.065] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/25/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Little is known about mechanisms of perineural invasion (PNI) by pancreatic ductal adenocarcinomas (PDAs) or other tumors. Annexin A2 (ANXA2) regulates secretion of SEMA3D, an axon guidance molecule, which binds and activates the receptor PLXND1 to promote PDA invasion and metastasis. We investigated whether axon guidance molecules promote PNI and metastasis by PDA cells in mice. METHODS We performed studies in a dorsal root ganglion (DRG) invasion system, wild-type C57BL/6 mice (controls), mice with peripheral sensory neuron-specific disruption of PlxnD1 (PLAC mice), LSL-KRASG12D/+;LSL-TP53R172H/+;PDX-1-CRE+/+ (KPC) mice, and KPC mice crossed with ANXA2-knockout mice (KPCA mice). PDA cells were isolated from KPC mice and DRG cells were isolated from control mice. Levels of SEMA3D or ANXA2 were knocked down in PDA cells with small hairpin and interfering RNAs and cells were analyzed by immunoblots in migration assays, with DRGs and with or without antibodies against PLXND1. PDA cells were injected into the pancreas of control and PLAC mice, growth of tumors was assessed, and tumor samples were analyzed by histology. DRG cells were incubated with SEMA3D and analyzed by live imaging. We measured levels of SEMA3D and PLXND1 in PDA specimens from patients with PNI and calculated distances between tumor cells and nerves. RESULTS DRG cells increase the migration of PDC cells in invasion assays; knockdown of SEMA3D in PDA cells or antibody blockade of PLXND1 on DRG cells reduced this invasive activity. In mice, orthotopic tumors grown from PDA cells with knockdown of SEMA3D, and in PLAC mice, orthotopic tumors grown from PDA cells, had reduced innervation and formed fewer metastases than orthotopic tumors grown from PDA cells in control mice. Increased levels of SEMA3D and PLXND1 in human PDA specimens associated with PNI. CONCLUSIONS DRG cells increase the migratory and invasive activities of pancreatic cancer cells, via secretion of SEMA3D by pancreatic cells and activation of PLXND1 on DRGs. Knockdown of SEMA3D and loss of neural PLXND1 reduces innervation of orthotopic PDAs and metastasis in mice. Increased levels of SEMA3D and PLXND1 in human PDA specimens associated with PNI. Strategies to disrupt the axon guidance pathway mediated by SEMA3D and PLXND1 might be developed to slow progression of PDA.
Collapse
MESH Headings
- Animals
- Annexin A2/deficiency
- Annexin A2/genetics
- Annexin A2/metabolism
- Axon Guidance/genetics
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/metabolism
- Carcinoma, Pancreatic Ductal/secondary
- Cell Communication
- Cell Movement
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Gene Expression Regulation, Neoplastic
- Genes, p53
- Genes, ras
- Genetic Predisposition to Disease
- Homeodomain Proteins/genetics
- Humans
- Intracellular Signaling Peptides and Proteins
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Invasiveness
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Neuronal Outgrowth
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Phenotype
- Semaphorins/genetics
- Semaphorins/metabolism
- Signal Transduction
- Trans-Activators/genetics
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Noelle R Jurcak
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Agnieszka A Rucki
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Graduate Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Stephen Muth
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth Thompson
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Rajni Sharma
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ding Ding
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qingfeng Zhu
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - James R Eshleman
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert A Anders
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Elizabeth M Jaffee
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland; Skip Viragh Center for Pancreatic Cancer, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kenji Fujiwara
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Skip Viragh Center for Pancreatic Cancer, Johns Hopkins University School of Medicine, Baltimore, Maryland; JSPS Overseas Research Fellow, Japan Society for the Promotion of Science, Tokyo, Japan
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland; Skip Viragh Center for Pancreatic Cancer, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
20
|
Jurcak N, Zheng L. Signaling in the microenvironment of pancreatic cancer: Transmitting along the nerve. Pharmacol Ther 2019; 200:126-134. [PMID: 31047906 PMCID: PMC6626552 DOI: 10.1016/j.pharmthera.2019.04.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/25/2019] [Indexed: 12/17/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDA) is a dismal malignant disease with the lowest stage-combined overall survival rate compared to any other cancer type. PDA has a unique tumor microenvironment (TME) comprised of a dense desmoplastic reaction comprising over two-thirds of the total tumor volume. The TME is comprised of cellular and acellular components that all orchestrate different signaling mechanisms together to promote tumorigenesis and disease progression. Particularly, the neural portion of the TME has recently been appreciated in PDA progression. Neural remodeling and perineural invasion (PNI), the neoplastic invasion of tumor cells into nerves, are common adverse histological characteristics of PDA associated with a worsened prognosis and increased cancer aggressiveness. The TME undergoes dramatic neural hypertrophy and increased neural density that is associated with many signaling pathways to promote cell invasion. PNI is also considered one of the main routes for cancer recurrence and metastasis after surgical resection, which remains the only current cure for PDA. Recent studies have shown multiple cell types in the TME signal through autocrine and paracrine mechanisms to enhance perineural invasion, pancreatic neural remodeling and disease progression in PDA. This review summarizes the current findings of the signaling mechanisms and cellular and molecular players involved in neural signaling in the TME of PDA.
Collapse
Affiliation(s)
- Noelle Jurcak
- Graduate Program in Cellular and Molecular Medicine, Baltimore, MD 21287, USA; Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, Baltimore, MD 21287, USA
| | - Lei Zheng
- Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21287, USA; Department of Oncology, Baltimore, MD 21287, USA; Pancreatic Cancer Precision Medicine Program, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
21
|
Vivekanadhan S, Mukhopadhyay D. Divergent roles of Plexin D1 in cancer. Biochim Biophys Acta Rev Cancer 2019; 1872:103-110. [PMID: 31152824 DOI: 10.1016/j.bbcan.2019.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/06/2019] [Accepted: 05/28/2019] [Indexed: 11/18/2022]
Abstract
Plexin D1 belongs to a family of transmembrane proteins called plexins. It was characterized as a receptor for semaphorins and is known to be essential for axonal guidance and vascular patterning. Mutations in Plexin D1 have been implicated in pathologic conditions such as truncus arteriosus and Möbius syndrome. Emerging data show that expression of Plexin D1 is deregulated in several cancers; it can support tumor development by aiding in tumor metastasis and EMT; and conversely, it can act as a dependence receptor and stimulate cell death in the absence of its canonical ligand, semaphorin 3E. The role of Plexin D1 in tumor development and progression is thereby garnering research interest for its potential as a biomarker and as a therapeutic target. In this review, we describe its discovery, structure, mutations, role(s) in cancer, and therapeutic potential.
Collapse
Affiliation(s)
- Sneha Vivekanadhan
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Jacksonville, FL, USA
| | | |
Collapse
|
22
|
Preoperative CT in patients with surgically resectable pancreatic adenocarcinoma: does the time interval between CT and surgery affect survival? Abdom Radiol (NY) 2018; 43:620-628. [PMID: 28695235 DOI: 10.1007/s00261-017-1254-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
PURPOSE The preoperative imaging-to-surgery time interval (ISI) influences the risk of unexpected progression (UP) found at surgery for pancreatic adenocarcinoma. We aimed to assess whether ISI influences disease recurrence and/or survival. METHODS AND MATERIALS A single-institution, ethics board-approved retrospective analysis of all patients who underwent attempted resection of pancreatic (PDAC) or periampullary adenocarcinoma (AmpAC) between 1st January 2010 and 31st December 2015 was performed. All patients underwent preoperative abdominal computed tomography (CT). Exclusion criteria were borderline resectable disease and neoadjuvant chemo/radiotherapy. Patients were followed up until 30th June 2016. The population was divided into ISI ≥/<25 days. Kaplan-Meier and Cox regression survival analyses were performed. RESULTS 239 patients underwent surgical exploration. UP was found in 29 (12.1%) and these patients had longer ISI (median 46 vs. 29 days, p < 0.05). When intention-to-treat analysis was performed, there was no difference in overall survival (OS) between patients with ISI ≥/<25. In those who underwent resection, ISI did not influence disease-free survival (DFS) or OS for PDAC (n = 174). For AmpAC (n = 36), ISI ≥ 25 days was associated with longer OS (p < 0.05) but did not influence DFS. Longer ISI was independently associated with improved OS on regression analysis for AmpAC. CONCLUSION Performing surgery for resectable pancreatic adenocarcinoma within 25 days of abdominal CT reduces the chance of UP but does not confer a survival benefit. For those who undergo resection of AmpAC, a longer ISI was associated with longer OS. This probably represents a more biologically indolent disease in this cohort.
Collapse
|
23
|
Lopez CM, Yu PY, Zhang X, Yilmaz AS, London CA, Fenger JM. MiR-34a regulates the invasive capacity of canine osteosarcoma cell lines. PLoS One 2018; 13:e0190086. [PMID: 29293555 PMCID: PMC5749745 DOI: 10.1371/journal.pone.0190086] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Osteosarcoma (OSA) is the most common bone tumor in children and dogs; however, no substantial improvement in clinical outcome has occurred in either species over the past 30 years. MicroRNAs (miRNAs) are small non-coding RNAs that regulate gene expression and play a fundamental role in cancer. The purpose of this study was to investigate the potential contribution of miR-34a loss to the biology of canine OSA, a well-established spontaneous model of the human disease. METHODOLOGY AND PRINCIPAL FINDINGS RT-qPCR demonstrated that miR-34a expression levels were significantly reduced in primary canine OSA tumors and canine OSA cell lines as compared to normal canine osteoblasts. In canine OSA cell lines stably transduced with empty vector or pre-miR-34a lentiviral constructs, overexpression of miR-34a inhibited cellular invasion and migration but had no effect on cell proliferation or cell cycle distribution. Transcriptional profiling of canine OSA8 cells possessing enforced miR-34a expression demonstrated dysregulation of numerous genes, including significant down-regulation of multiple putative targets of miR-34a. Moreover, gene ontology analysis of down-regulated miR-34a target genes showed enrichment of several biological processes related to cell invasion and motility. Lastly, we validated changes in miR-34a putative target gene expression, including decreased expression of KLF4, SEM3A, and VEGFA transcripts in canine OSA cells overexpressing miR-34a and identified KLF4 and VEGFA as direct target genes of miR-34a. Concordant with these data, primary canine OSA tumor tissues demonstrated increased expression levels of putative miR-34a target genes. CONCLUSIONS These data demonstrate that miR-34a contributes to invasion and migration in canine OSA cells and suggest that loss of miR-34a may promote a pattern of gene expression contributing to the metastatic phenotype in canine OSA.
Collapse
Affiliation(s)
- Cecilia M. Lopez
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
| | - Peter Y. Yu
- Medical Student Research Program, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States of America
| | - Xiaoli Zhang
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, United States of America
| | - Ayse Selen Yilmaz
- Center for Biostatistics, Department of Biomedical Informatics, The Ohio State University, Columbus, Ohio, United States of America
| | - Cheryl A. London
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
- Department of Veterinary Biosciences, College of Veterinary Medicine, Tufts University, New Grafton, Massachusetts, United States of America
| | - Joelle M. Fenger
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|