1
|
Aliya S, Farani MR, Kim E, Kim S, Gupta VK, Kumar K, Huh YS. Therapeutic targeting of the tumor microenvironments with cannabinoids and their analogs: Update on clinical trials. ENVIRONMENTAL RESEARCH 2023; 231:115862. [PMID: 37146933 DOI: 10.1016/j.envres.2023.115862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/07/2023]
Abstract
Cancer is a major global public health concern that affects both industrialized and developing nations. Current cancer chemotherapeutic options are limited by side effects, but plant-derived alternatives and their derivatives offer the possibilities of enhanced treatment response and reduced side effects. A plethora of recently published articles have focused on treatments based on cannabinoids and cannabinoid analogs and reported that they positively affect healthy cell growth and reverse cancer-related abnormalities by targeting aberrant tumor microenvironments (TMEs), lowering tumorigenesis, preventing metastasis, and/or boosting the effectiveness of chemotherapy and radiotherapy. Furthermore, TME modulating systems are receiving much interest in the cancer immunotherapy field because it has been shown that TMEs have significant impacts on tumor progression, angiogenesis, invasion, migration, epithelial to mesenchymal transition, metastasis and development of drug resistance. Here, we have reviewed the effective role of cannabinoids, their analogs and cannabinoid nano formulations on the cellular components of TME (endothelial cells, pericytes, fibroblast and immune cells) and how efficiently it retards the progression of carcinogenesis is discussed. The article summarizes the existing research on the molecular mechanisms of cannabinoids regulation of the TME and finally highlights the human studies on cannabinoids' active interventional clinical trials. The conclusion outlines the need for future research involving clinical trials of cannabinoids to demonstrate their efficacy and activity as a treatment/prevention for various types of human malignancies.
Collapse
Affiliation(s)
- Sheik Aliya
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea
| | | | - Eunsu Kim
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Suheon Kim
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Vivek Kumar Gupta
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Krishan Kumar
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea
| | - Yun Suk Huh
- Department of Biological Engineering, Inha University, Incheon, 22212, Republic of Korea.
| |
Collapse
|
2
|
Gaspar TB, Lopes JM, Soares P, Vinagre J. An update on genetically engineered mouse models of pancreatic neuroendocrine neoplasms. Endocr Relat Cancer 2022; 29:R191-R208. [PMID: 36197786 DOI: 10.1530/erc-22-0166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/29/2022] [Indexed: 11/09/2022]
Abstract
Pancreatic neuroendocrine neoplasms (PanNENs) are rare and clinically challenging entities. At the molecular level, PanNENs' genetic profile is well characterized, but there is limited knowledge regarding the contribution of the newly identified genes to tumor initiation and progression. Genetically engineered mouse models (GEMMs) are the most versatile tool for studying the plethora of genetic variations influencing PanNENs' etiopathogenesis and behavior over time. In this review, we present the state of the art of the most relevant PanNEN GEMMs available and correlate their findings with the human neoplasms' counterparts. We discuss the historic GEMMs as the most used and with higher translational utility models. GEMMs with Men1 and glucagon receptor gene germline alterations stand out as the most faithful models in recapitulating human disease; RIP-Tag models are unique models of early-onset, highly vascularized, invasive carcinomas. We also include a section of the most recent GEMMs that evaluate pathways related to cell cycle and apoptosis, Pi3k/Akt/mTOR, and Atrx/Daxx. For the latter, their tumorigenic effect is heterogeneous. In particular, for Atrx/Daxx, we will require more in-depth studies to evaluate their contribution; even though they are prevalent genetic events in PanNENs, they have low/inexistent tumorigenic capacity per se in GEMMs. Researchers planning to use GEMMs can find a road map of the main clinical features in this review, presented as a guide that summarizes the chief milestones achieved. We identify pitfalls to overcome, concerning the novel designs and standardization of results, so that future models can replicate human disease more closely.
Collapse
Affiliation(s)
- Tiago Bordeira Gaspar
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar da Universidade do Porto, Porto, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - José Manuel Lopes
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
- Department of Pathology, Centro Hospitalar e Universitário de São João, Porto, Portugal
| | - Paula Soares
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| | - João Vinagre
- i3S - Instituto de Investigação e Inovação em Saúde, Porto, Portugal
- Ipatimup - Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
- FMUP - Faculdade de Medicina da Universidade do Porto, Porto, Portugal
| |
Collapse
|
3
|
Bakshi HA, Faruck HL, Ravesh Z, Ansari P, Hannan JMA, Hashimoto R, Takayama K, Farzand R, Nasef MM, Mensah A, Aljabali AAA, Mishra V, Charbe NB, Goyal R, Negi P, Serrano-Aroca Á, Bahar B, El-Tanani M, Courtenay AJ, McCarron P, Jack IG, Tambuwala MM. Therapeutic Potential of Cannabinoids on Tumor Microenvironment: A Molecular Switch in Neoplasia Transformation. Integr Cancer Ther 2022; 21:15347354221096766. [PMID: 35796303 PMCID: PMC9425895 DOI: 10.1177/15347354221096766] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The efficacy of chemotherapy depends on the tumor microenvironment. This microenvironment consists of a complex cellular network that can exert both stimulatory and inhibitory effects on tumor genesis. Given the increasing interest in the effectiveness of cannabis, cannabinoids have gained much attention as a potential chemotherapy drug. Cannabinoids are a group of marker compounds found in Cannabis sativa L., more commonly known as marijuana, a psychoactive drug used since ancient times for pain management. Although the anticancer potential of C. sativa, has been recognized previously, increased attention was generated after discovering the endocannabinoid system and the successful production of cannabinoid receptors. In vitro and in vivo studies on various tumor models have shown therapeutic efficiency by modifying the tumor microenvironment. However, despite extensive attention regarding potential therapeutic implications of cannabinoids, considerable clinical and preclinical analysis is needed to adequately define the physiological, pharmacological, and medicinal aspects of this range of compounds in various disorders covered in this review. This review summarizes the key literature surrounding the role of cannabinoids in the tumor microenvironment and their future promise in cancer treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rabia Farzand
- University of Huddersfield, Queensgate, Huddersfield, UK
| | | | | | | | - Vijay Mishra
- Lovely Professional University, Phagwara, Punjab, India
| | | | - Rohit Goyal
- Shoolini University, Solan, Himachal Pradesh, India
| | - Poonam Negi
- Shoolini University, Solan, Himachal Pradesh, India
| | | | | | | | | | - Paul McCarron
- Ulster University, Coleraine, County Londonderry, UK
| | - Iain G Jack
- Ulster University, Coleraine, County Londonderry, UK
| | | |
Collapse
|
4
|
Detjen K, Hammerich L, Özdirik B, Demir M, Wiedenmann B, Tacke F, Jann H, Roderburg C. Models of Gastroenteropancreatic Neuroendocrine Neoplasms: Current Status and Future Directions. Neuroendocrinology 2021; 111:217-236. [PMID: 32615560 DOI: 10.1159/000509864] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022]
Abstract
Gastroenteropancreatic neuroendocrine neoplasms (GEP-NENs) are a rare, heterogeneous group of tumors that originate from the endocrine system of the gastrointestinal tract and pancreas. GEP-NENs are subdivided according to their differentiation into well-differentiated neuroendocrine tumors (NETs) and poorly differentiated neuroendocrine carcinomas (NECs). Since GEP-NENs represent rare diseases, only limited data from large prospective, randomized clinical trials are available, and recommendations for treatment of GEP-NEN are in part based on data from retrospective analyses or case series. In this context, tractable disease models that reflect the situation in humans and that allow to recapitulate the different clinical aspects and disease stages of GEP-NET or GEP-NEC are urgently needed. In this review, we highlight available data on mouse models for GEP-NEN. We discuss how these models reflect tumor biology of human disease and whether these models could serve as a tool for understanding the pathogenesis of GEP-NEN and for disease modeling and pharmacosensitivity assays, facilitating prediction of treatment response in patients. In addition, open issues applicable for future developments will be discussed.
Collapse
Affiliation(s)
- Katharina Detjen
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Burcin Özdirik
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Bertram Wiedenmann
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Henning Jann
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany
| | - Christoph Roderburg
- Department of Hepatology and Gastroenterology, Charité - University Medicine Berlin, Campus Virchow Klinikum and Charité Campus Mitte, Berlin, Germany,
| |
Collapse
|
5
|
Cancer immune control needs senescence induction by interferon-dependent cell cycle regulator pathways in tumours. Nat Commun 2020; 11:1335. [PMID: 32165639 PMCID: PMC7067802 DOI: 10.1038/s41467-020-14987-6] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 02/07/2020] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint blockade (ICB)-based or natural cancer immune responses largely eliminate tumours. Yet, they require additional mechanisms to arrest those cancer cells that are not rejected. Cytokine-induced senescence (CIS) can stably arrest cancer cells, suggesting that interferon-dependent induction of senescence-inducing cell cycle regulators is needed to control those cancer cells that escape from killing. Here we report in two different cancers sensitive to T cell-mediated rejection, that deletion of the senescence-inducing cell cycle regulators p16Ink4a/p19Arf (Cdkn2a) or p21Cip1 (Cdkn1a) in the tumour cells abrogates both the natural and the ICB-induced cancer immune control. Also in humans, melanoma metastases that progressed rapidly during ICB have losses of senescence-inducing genes and amplifications of senescence inhibitors. Metastatic cells also resist CIS. Such genetic and functional alterations are infrequent in metastatic melanomas regressing during ICB. Thus, activation of tumour-intrinsic, senescence-inducing cell cycle regulators is required to stably arrest cancer cells that escape from eradication. The growth of cancer cells can be stably arrested by cytokine-induced senescence. Here, the authors show that cancers with defects in senescence-inducing cell cycle regulator pathways are resistant to immune checkpoint blockade.
Collapse
|
6
|
Maugein A, Diedisheim M, Bailly K, Scharfmann R, Albagli O. The RB gene family controls the maturation state of the EndoC-βH2 human pancreatic β-cells. Differentiation 2020; 113:1-9. [PMID: 32120156 DOI: 10.1016/j.diff.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 02/17/2020] [Accepted: 02/17/2020] [Indexed: 01/18/2023]
Abstract
The functional maturation of human pancreatic β-cells remains poorly understood. EndoC-βH2 is a human β-cell line with a reversible immortalized phenotype. Removal of the two oncogenes, SV40LT and hTERT introduced for its propagation, stops proliferation, triggers cell size increase and senescence, promotes mitochondrial activity and amplifies several β-cell traits and functions. Overall, these events recapitulate several aspects of functional β-cell maturation. We report here that selective depletion of SV40LT, but not of hTERT, is sufficient to revert EndoC-βH2 immortalization. SV40LT inhibits the activity of the RB family members and of P53. In EndoC-βH2 cells, the knock-down of RB itself, and, to a lesser extent, of its relative P130, precludes most events triggered by SV40LT depletion. In contrast, the knock-down of P53 does not prevent reversion of immortalization. Thus, an increase in RB and P130 activity, but not in P53 activity, is required for functional maturation of EndoC-βH2 cells upon SV40LT-depletion. In addition, RB and/or P130 depletion in SV40LT-expressing EndoC-βH2 cells decreases cell size, stimulates proliferation, and decreases the expression of key β-cell genes. Thus, despite SV40LT expression, EndoC-βH2 cells have a residual RB activity, which when suppressed reverts them to a more immature phenotype. These results show that the expression and activity levels of RB family members, especially RB itself, regulate the maturation state of EndoC-βH2 cells.
Collapse
Affiliation(s)
- Alicia Maugein
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Marc Diedisheim
- Assistance Publique - Hôpitaux de Paris, Diabetology Department, Paris University, Cochin Hospital, and INSERM, UMRS 1138, Centre de Recherche des Cordeliers, Paris University, 75006, Paris, France
| | - Karine Bailly
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Raphaël Scharfmann
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France
| | - Olivier Albagli
- Paris University, Institut Cochin, INSERM, U1016, CNRS, UMR8104, 75014, Paris, France.
| |
Collapse
|
7
|
Scharfmann R, Staels W, Albagli O. The supply chain of human pancreatic β cell lines. J Clin Invest 2019; 129:3511-3520. [PMID: 31478912 PMCID: PMC6715382 DOI: 10.1172/jci129484] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with type 1 or type 2 diabetes have an insufficiency in their functional β cell mass. To advance diabetes treatment and to work toward a cure, a better understanding of how to protect the pancreatic β cells against autoimmune or metabolic assaults (e.g., obesity, gestation) will be required. Over the past decades, β cell protection has been extensively investigated in rodents both in vivo and in vitro using isolated islets or rodent β cell lines. Transferring these rodent data to humans has long been challenging, at least partly for technical reasons: primary human islet preparations were scarce and functional human β cell lines were lacking. In 2011, we described a robust protocol of targeted oncogenesis in human fetal pancreas and produced the first functional human β cell line, and in subsequent years additional lines with specific traits. These cell lines are currently used by more than 150 academic and industrial laboratories worldwide. In this Review, we first explain how we developed the human β cell lines and why we think we succeeded where others, despite major efforts, did not. Next, we discuss the use of such functional human β cell lines and share some perspectives on their use to advance diabetes research.
Collapse
Affiliation(s)
- Raphael Scharfmann
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| | - Willem Staels
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
- Beta Cell Neogenesis (BENE), Vrije Universiteit Brussel, Brussels, Belgium
| | - Olivier Albagli
- INSERM U1016, Institut Cochin, Université Paris Descartes, Paris, France
| |
Collapse
|
8
|
Buicko JL, Finnerty BM, Zhang T, Kim BJ, Fahey TJ, Nancy Du YC. Insights into the biology and treatment strategies of pancreatic neuroendocrine tumors. ACTA ACUST UNITED AC 2019; 2. [PMID: 31535089 DOI: 10.21037/apc.2019.06.02] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Pancreatic neuroendocrine tumors (PNETs) are the second most common primary pancreatic neoplasms after pancreatic ductal adenocarcinoma. PNETs present with widely various clinical manifestation and unfavorable survival rate. The recent advances in next generation sequencing have significantly increased our understanding of the molecular landscape of PNETs and help guide the development of targeted therapies. This review intends to outline a holistic picture of the tumors by discussing current understanding of clinical presentations, up-to-date treatment strategies, novel mouse models, and molecular biology of PNETs. Furthermore, we will provide insight into the future development of more effective targeted therapies that are necessary to manage PNETs.
Collapse
Affiliation(s)
- Jessica L Buicko
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Tiantian Zhang
- Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Bu Jung Kim
- Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Thomas J Fahey
- Department of Surgery, Weill Cornell Medicine, New York, NY 10065, USA
| | - Yi-Chieh Nancy Du
- Department of Pathology & Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| |
Collapse
|
9
|
Albagli O, Maugein A, Huijbregts L, Bredel D, Carlier G, Martin P, Scharfmann R. New α- and SIN γ-retrovectors for safe transduction and specific transgene expression in pancreatic β cell lines. BMC Biotechnol 2019; 19:35. [PMID: 31208395 PMCID: PMC6580483 DOI: 10.1186/s12896-019-0531-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 06/06/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Viral vectors are invaluable tools to transfer genes and/or regulatory sequences into differentiated cells such as pancreatic cells. To date, several kinds of viral vectors have been used to transduce different pancreatic cell types, including insulin-producing β cells. However, few studies have used vectors derived from « simple » retroviruses, such as avian α- or mouse γ-retroviruses, despite their high experimental convenience. Moreover, such vectors were never designed to specifically target transgene expression into β cells. RESULTS We here describe two novel α- or SIN (Self-Inactivating) γ-retrovectors containing the RIP (Rat Insulin Promoter) as internal promoter. These two retrovectors are easily produced in standard BSL2 conditions, rapidly concentrated if needed, and harbor a large multiple cloning site. For the SIN γ-retrovector, either the VSV-G (pantropic) or the retroviral ecotropic (rodent specific) envelope was used. For the α-retrovector, we used the A type envelope, as its receptor, termed TVA, is only naturally present in avian cells and can efficiently be provided to mammalian β cells through either exogenous expression upon cDNA transfer or gesicle-mediated delivery of the protein. As expected, the transgenes cloned into the two RIP-containing retrovectors displayed a strong preferential expression in β over non-β cells compared to transgenes cloned in their non-RIP (CMV- or LTR-) regulated counterparts. We further show that RIP activity of both retrovectors mirrored fluctuations affecting endogenous INSULIN gene expression in human β cells. Finally, both α- and SIN γ-retrovectors were extremely poorly mobilized by the BXV1 xenotropic retrovirus, a common invader of human cells grown in immunodeficient mice, and, most notably, of human β cell lines. CONCLUSION Our novel α- and SIN γ-retrovectors are safe and convenient tools to stably and specifically express transgene(s) in mammalian β cells. Moreover, they both reproduce some regulatory patterns affecting INSULIN gene expression. Thus, they provide a helpful tool to both study the genetic control of β cell function and monitor changes in their differentiation status.
Collapse
Affiliation(s)
- Olivier Albagli
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France.
| | - Alicia Maugein
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France
| | - Lukas Huijbregts
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France
| | - Delphine Bredel
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France.,Present Address: Laboratoire de Recherche Translationnelle en Immunothérapie, Institut Gustave Roussy, 114 Rue Edouard Vaillant, 94800, Villejuif, France
| | - Géraldine Carlier
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France
| | - Patrick Martin
- Université Côte d'Azur, CNRS UMR7277 INSERM U1099, iBV (Institut de Biologie Valrose), Université Nice Sophia Antipolis, Bâtiment Sciences Naturelles; UFR Sciences, Parc Valrose, 28, avenue Valrose, 06108, Nice Cedex 2, France
| | - Raphaël Scharfmann
- INSERM U1016, CNRS UMR8104, Institut Cochin, Université Paris Descartes, 123 Boulevard de Port-Royal, 75014, Paris, France
| |
Collapse
|
10
|
Conditional deletion of RB1 in the Tie2 lineage leads to aortic valve regurgitation. PLoS One 2018; 13:e0190623. [PMID: 29304157 PMCID: PMC5755794 DOI: 10.1371/journal.pone.0190623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023] Open
Abstract
Objective Aortic valve disease is a complex process characterized by valve interstitial cell activation, disruption of the extracellular matrix culminating in valve mineralization occurring over many years. We explored the function of the retinoblastoma protein (pRb) in aortic valve disease, given its critical role in mesenchymal cell differentiation including bone development and mineralization. Approach and results We generated a mouse model of conditional pRb knockout (cKO) in the aortic valve regulated by Tie2-Cre-mediated excision of floxed RB1 alleles. Aged pRb cKO animals showed significantly more aortic valve regurgitation by echocardiography compared to pRb het control animals. The pRb cKO aortic valves had increased leaflet thickness without increased cellular proliferation. Histologic studies demonstrated intense α-SMA expression in pRb cKO leaflets associated with disorganized extracellular matrix and increased leaflet stiffness. The pRb cKO mice also showed increased circulating cytokine levels. Conclusions Our studies demonstrate that pRb loss in the Tie2-lineage that includes aortic valve interstitial cells is sufficient to cause age-dependent aortic valve dysfunction.
Collapse
|
11
|
Aristizabal Prada ET, Auernhammer CJ. Targeted therapy of gastroenteropancreatic neuroendocrine tumours: preclinical strategies and future targets. Endocr Connect 2018; 7:R1-R25. [PMID: 29146887 PMCID: PMC5754510 DOI: 10.1530/ec-17-0286] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 11/16/2017] [Indexed: 12/12/2022]
Abstract
Molecular targeted therapy of advanced neuroendocrine tumours (NETs) of the gastroenteropancreatic (GEP) system currently encompasses approved therapy with the mammalian target of rapamycin (mTOR) inhibitor everolimus and the multi-tyrosinkinase inhibitor sunitinib. However, clinical efficacy of these treatment strategies is limited by low objective response rates and limited progression-free survival due to tumour resistance. Further novel strategies for molecular targeted therapy of NETs of the GEP system are needed. This paper reviews preclinical research models and signalling pathways in NETs of the GEP system. Preclinical and early clinical data on putative novel targets for molecular targeted therapy of NETs of the GEP system are discussed, including PI3K, Akt, mTORC1/mTORC2, GSK3, c-Met, Ras-Raf-MEK-ERK, embryogenic pathways (Hedgehog, Notch, Wnt/beta-catenin, TGF-beta signalling and SMAD proteins), tumour suppressors and cell cycle regulators (p53, cyclin-dependent kinases (CDKs) CDK4/6, CDK inhibitor p27, retinoblastoma protein (Rb)), heat shock protein HSP90, Aurora kinase, Src kinase family, focal adhesion kinase and epigenetic modulation by histone deacetylase inhibitors.
Collapse
Affiliation(s)
- E T Aristizabal Prada
- Department of Internal Medicine IVCampus Grosshadern, University-Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - C J Auernhammer
- Department of Internal Medicine IVCampus Grosshadern, University-Hospital, Ludwig-Maximilians-University of Munich, Munich, Germany
| |
Collapse
|
12
|
Abstract
The core aspect of the senescent phenotype is a stable state of cell cycle arrest. However, this is a disguise that conceals a highly active metabolic cell state with diverse functionality. Both the cell-autonomous and the non-cell-autonomous activities of senescent cells create spatiotemporally dynamic and context-dependent tissue reactions. For example, the senescence-associated secretory phenotype (SASP) provokes not only tumour-suppressive but also tumour-promoting responses. Senescence is now increasingly considered to be an integrated and widespread component that is potentially important for tumour development, tumour suppression and the response to therapy.
Collapse
Affiliation(s)
- Pedro A Pérez-Mancera
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Andrew R J Young
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| | - Masashi Narita
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
13
|
T-helper-1-cell cytokines drive cancer into senescence. Nature 2013; 494:361-5. [PMID: 23376950 DOI: 10.1038/nature11824] [Citation(s) in RCA: 545] [Impact Index Per Article: 45.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2012] [Accepted: 12/06/2012] [Indexed: 02/07/2023]
Abstract
Cancer control by adaptive immunity involves a number of defined death and clearance mechanisms. However, efficient inhibition of exponential cancer growth by T cells and interferon-γ (IFN-γ) requires additional undefined mechanisms that arrest cancer cell proliferation. Here we show that the combined action of the T-helper-1-cell cytokines IFN-γ and tumour necrosis factor (TNF) directly induces permanent growth arrest in cancers. To safely separate senescence induced by tumour immunity from oncogene-induced senescence, we used a mouse model in which the Simian virus 40 large T antigen (Tag) expressed under the control of the rat insulin promoter creates tumours by attenuating p53- and Rb-mediated cell cycle control. When combined, IFN-γ and TNF drive Tag-expressing cancers into senescence by inducing permanent growth arrest in G1/G0, activation of p16INK4a (also known as CDKN2A), and downstream Rb hypophosphorylation at serine 795. This cytokine-induced senescence strictly requires STAT1 and TNFR1 (also known as TNFRSF1A) signalling in addition to p16INK4a. In vivo, Tag-specific T-helper 1 cells permanently arrest Tag-expressing cancers by inducing IFN-γ- and TNFR1-dependent senescence. Conversely, Tnfr1(-/-)Tag-expressing cancers resist cytokine-induced senescence and grow aggressively, even in TNFR1-expressing hosts. Finally, as IFN-γ and TNF induce senescence in numerous murine and human cancers, this may be a general mechanism for arresting cancer progression.
Collapse
|
14
|
Nolan-Stevaux O, Truitt MC, Pahler JC, Olson P, Guinto C, Lee DC, Hanahan D. Differential contribution to neuroendocrine tumorigenesis of parallel egfr signaling in cancer cells and pericytes. Genes Cancer 2011; 1:125-41. [PMID: 20975924 PMCID: PMC2958675 DOI: 10.1177/1947601909358722] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Factors associated with tumor sensitivity to epidermal growth factor receptor (EGFR) inhibitors in the context of wild-type EGFR remain elusive. This study investigates the mechanistic basis of responsiveness to EGFR inhibitors in the RIP1-Tag2 (RT2) mouse model of pancreatic neuroendocrine tumorigenesis (PNET). Upon treatment of RT2 mice with EGFR inhibitors, PNET tumors harboring wild-type, nonamplified alleles of Egfr grow at a markedly reduced rate and display a significant increase in tumor cell apoptosis, as well as reduced neovascularization. The authors identify Tgf-α and Hb-egf as key limiting mediators of separable pathological functions of Egfr in neuroendocrine tumor progression: Tgf-α mutant tumors present with an elevated apoptotic index, whereas Hb-egf mutant lesions exhibit decreased angiogenic switching and neovascularization. This study not only associates Tgf-α and Hb-egf expression with wild-type Egfr oncogenicity but also ascribes the proangiogenic activity of Egfr in this tumor model to a novel mesenchymal Hb-egf/Egfr signaling axis, whereby endothelial and pericyte-derived Hb-egf activates Egfr specifically in tumor-associated perivascular cells, leading to increased pericyte coverage of the tumor endothelium and enhanced angiogenesis.
Collapse
Affiliation(s)
- Olivier Nolan-Stevaux
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Wirt SE, Adler AS, Gebala V, Weimann JM, Schaffer BE, Saddic LA, Viatour P, Vogel H, Chang HY, Meissner A, Sage J. G1 arrest and differentiation can occur independently of Rb family function. ACTA ACUST UNITED AC 2010; 191:809-25. [PMID: 21059851 PMCID: PMC2983066 DOI: 10.1083/jcb.201003048] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Repression of E2F target genes is required for cell cycle arrest in Rb family (Rb, p107, and p130)-deficient cells. The ability of progenitor cells to exit the cell cycle is essential for proper embryonic development and homeostasis, but the mechanisms governing cell cycle exit are still not fully understood. Here, we tested the requirement for the retinoblastoma (Rb) protein and its family members p107 and p130 in G0/G1 arrest and differentiation in mammalian cells. We found that Rb family triple knockout (TKO) mouse embryos survive until days 9–11 of gestation. Strikingly, some TKO cells, including in epithelial and neural lineages, are able to exit the cell cycle in G0/G1 and differentiate in teratomas and in culture. This ability of TKO cells to arrest in G0/G1 is associated with the repression of key E2F target genes. Thus, G1 arrest is not always dependent on Rb family members, which illustrates the robustness of cell cycle regulatory networks during differentiation and allows for the identification of candidate pathways to inhibit the expansion of cancer cells with mutations in the Rb pathway.
Collapse
Affiliation(s)
- Stacey E Wirt
- Department of Pediatrics, Stanford Medical School, Stanford, CA 94305, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Olson P, Lu J, Zhang H, Shai A, Chun MG, Wang Y, Libutti SK, Nakakura EK, Golub TR, Hanahan D. MicroRNA dynamics in the stages of tumorigenesis correlate with hallmark capabilities of cancer. Genes Dev 2009; 23:2152-65. [PMID: 19759263 DOI: 10.1101/gad.1820109] [Citation(s) in RCA: 216] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
While altered expression of microRNAs (miRs) in tumors has been well documented, it remains unclear how the miR transcriptome intersects neoplastic progression. By profiling the miR transcriptome we identified miR expression signatures associated with steps in tumorigenesis and the acquisition of hallmark capabilities in a prototypical mouse model of cancer. Metastases and a rare subset of primary tumors shared a distinct miR signature, implicating a discrete lineage for metastatic tumors. The miR-200 family is strongly down-regulated in metastases and met-like primary tumors, thereby relieving repression of the mesenchymal transcription factor Zeb1, which in turn suppresses E-cadherin. Treatment with a clinically approved angiogenesis inhibitor normalized angiogenic signature miRs in primary tumors, while altering expression of metastatic signature miRs similarly to liver metastases, suggesting their involvement in adaptive resistance to anti-angiogenic therapy via enhanced metastasis. Many of the miR changes associated with specific stages and hallmark capabilities in the mouse model are similarly altered in human tumors, including cognate pancreatic neuroendocrine tumors, implying a generality.
Collapse
Affiliation(s)
- Peter Olson
- Diabetes Center, University of California at San Francisco, San Francisco, California 94143, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Staton CA, Brown NJ, Reed MWR. Current status and future prospects for anti-angiogenic therapies in cancer. Expert Opin Drug Discov 2009; 4:961-79. [DOI: 10.1517/17460440903196737] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
18
|
A mouse model for chronic lymphocytic leukemia based on expression of the SV40 large T antigen. Blood 2009; 114:119-27. [DOI: 10.1182/blood-2009-01-198937] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Abstract
The simian virus 40 (SV40) T antigen is a potent oncogene able to transform many cell types and has been implicated in leukemia and lymphoma. In this report, we have achieved sporadic SV40 T-antigen expression in mature B cells in mice, by insertion of a SV40 T antigen gene in opposite transcriptional orientation in the immunoglobulin (Ig) heavy (H) chain locus between the D and JH segments. SV40 T-antigen expression appeared to result from retention of the targeted germline allele and concomitant antisense transcription of SV40 large T in mature B cells, leading to chronic lymphocytic leukemia (CLL). Although B-cell development was unperturbed in young mice, aging mice showed accumulation of a monoclonal B-cell population in which the targeted IgH allele was in germline configuration and the wild-type IgH allele had a productive V(D)J recombination. These leukemic B cells were IgDlowCD5+ and manifested nonrandom usage of V, D, and J segments. VH regions were either unmutated, with preferential usage of the VH11 family, or manifested extensive somatic hypermutation. Our findings provide an animal model for B-CLL and show that pathways activated by SV40 T antigen play important roles in the pathogenesis of B-CLL.
Collapse
|
19
|
Zheng HC, Nakamura T, Zheng Y, Nakanishi Y, Tabuchi Y, Uchiyama A, Takahashi H, Takano Y. SV40 T antigen disrupted the cell metabolism and the balance between proliferation and apoptosis in lens tumors of transgenic mice. J Cancer Res Clin Oncol 2009; 135:1521-32. [PMID: 19466455 DOI: 10.1007/s00432-009-0599-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2009] [Accepted: 04/27/2009] [Indexed: 11/25/2022]
Abstract
PURPOSE Simian Vacuolating Virus 40 (SV40) T antigen perturbed p53 and RB to cause cell malignant transformation. The purpose of this study was to identify the molecular changes during lens carcinogenesis and cancer progression induced by SV40 T antigen. METHODS The different lens lesions of alpha A-crystallin/SV40 T antigen transgenic mice were examined using cDNA microarray, immunohistochemistry and TUNEL to scan the influenced molecules and signal pathways. RESULTS There appeared dysplasia, carcinoma in situ, followed by invasion inside or outside eyeball, and final metastasis into lymph node or lung. Cell functions largely changed from such many aspects as cell cycle, cell morphology, cell development, cell-to-cell signaling and so forth since lens carcinogenesis. The significant differences were observed in such signaling pathways as metabolism about carbohydrate, amino acid, nucleotides, Xenobiotics and nitrogen (P < 0.05).The remarkable distinction of cell proliferation and cell death was found after carcinoma began to invade. There was significant alteration in cell growth, cell cycle, cell-to-cell signaling and metabolism since carcinoma invasion outside the eyeball happened. Parafibromin, Stat 1alpha, Mek kinase-1, CK2alpha, GRP78, Arp2 and Apr3 were not expressed in wild-type mice lens, but in others. The proliferative levels of dysplasia, carcinoma in situ and invasive carcinoma inside eyeballs were statistically higher than other groups (P < 0.05). The apoptotic levels of dysplasia were significantly higher than wild-type control (P < 0.05), but lower than the others (P < 0.05). CONCLUSION SV40 T antigen remarkably targeted the cell metabolism and disrupted the balance between proliferation and apoptosis during the lens carcinogenesis and following progression.
Collapse
Affiliation(s)
- Hua-chuan Zheng
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, China Medical University, Shenyang, China.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Sáenz Robles MT, Pipas JM. T antigen transgenic mouse models. Semin Cancer Biol 2009; 19:229-35. [PMID: 19505650 DOI: 10.1016/j.semcancer.2009.02.002] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 02/03/2009] [Accepted: 02/06/2009] [Indexed: 01/12/2023]
Abstract
The study of polyomavirus has benefited immensely from two scientific methodologies, cell culture and in vitro studies on one side and the use of transgenic mice as experimental models on the other. Both approaches allowed us to identify cellular products targeted by the viruses, the consequences of these interactions at the phenotypic and molecular level, and thus the potential roles of the targets within their normal cellular context. In particular, cell culture and in vitro reports suggest a model explaining partially how SV40 large T antigen contributes to oncogenic transformation. In most cases, T antigen induces cell cycle entry by inactivation of the Rb proteins (pRb, p130, and p107), thus activating E2F-dependent transcription and subsequent S-phase entry. Simultaneously, T antigen blocks p53 activity and therefore prevents the ensuing cell-cycle arrest and apoptosis. For the most part, studies of T antigen expression in transgenic mice support this model, but the use of T antigen mutants and their expression in different tissue and cell type settings have expanded our knowledge of the model system and raised important questions regarding tumorigenic mechanisms functioning in vivo.
Collapse
|
21
|
Willimsky G, Czéh M, Loddenkemper C, Gellermann J, Schmidt K, Wust P, Stein H, Blankenstein T. Immunogenicity of premalignant lesions is the primary cause of general cytotoxic T lymphocyte unresponsiveness. ACTA ACUST UNITED AC 2008; 205:1687-700. [PMID: 18573907 PMCID: PMC2442645 DOI: 10.1084/jem.20072016] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Cancer is sporadic in nature, characterized by an initial clonal oncogenic event and usually a long latency. When and how it subverts the immune system is unknown. We show, in a model of sporadic immunogenic cancer, that tumor-specific tolerance closely coincides with the first tumor antigen recognition by B cells. During the subsequent latency period until tumors progress, the mice acquire general cytotoxic T lymphocyte (CTL) unresponsiveness, which is associated with high transforming growth factor (TGF) beta1 levels and expansion of immature myeloid cells (iMCs). In mice with large nonimmunogenic tumors, iMCs expand but TGF-beta1 serum levels are normal, and unrelated CTL responses are undiminished. We conclude that (a) tolerance to the tumor antigen occurs at the premalignant stage, (b) tumor latency is unlikely caused by CTL control, and (c) a persistent immunogenic tumor antigen causes general CTL unresponsiveness but tumor burden and iMCs per se do not.
Collapse
Affiliation(s)
- Gerald Willimsky
- Institute of Immunology, Charité Campus Benjamin Franklin, 12200 Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Müller-Hermelink N, Braumüller H, Pichler B, Wieder T, Mailhammer R, Schaak K, Ghoreschi K, Yazdi A, Haubner R, Sander CA, Mocikat R, Schwaiger M, Förster I, Huss R, Weber WA, Kneilling M, Röcken M. TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 2008; 13:507-18. [PMID: 18538734 DOI: 10.1016/j.ccr.2008.04.001] [Citation(s) in RCA: 237] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 05/23/2007] [Accepted: 04/08/2008] [Indexed: 12/12/2022]
Abstract
Immune responses may arrest tumor growth by inducing tumor dormancy. The mechanisms leading to either tumor dormancy or promotion of multistage carcinogenesis by adaptive immunity are poorly characterized. Analyzing T antigen (Tag)-induced multistage carcinogenesis in pancreatic islets, we show that Tag-specific CD4+ T cells home selectively into the tumor microenvironment around the islets, where they either arrest or promote transition of dysplastic islets into islet carcinomas. Through combined TNFR1 signaling and IFN-gamma signaling, Tag-specific CD4+ T cells induce antiangiogenic chemokines and prevent alpha(v)beta(3) integrin expression, tumor angiogenesis, tumor cell proliferation, and multistage carcinogenesis, without destroying Tag-expressing islet cells. In the absence of either TNFR1 signaling or IFN-gamma signaling, the same T cells paradoxically promote angiogenesis and multistage carcinogenesis. Thus, tumor-specific T cells can directly survey multistage carcinogenesis through cytokine signaling.
Collapse
MESH Headings
- Animals
- Antigens, Viral, Tumor/genetics
- Antigens, Viral, Tumor/metabolism
- Blood Glucose/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/transplantation
- Cell Movement
- Cell Proliferation
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Immunotherapy/methods
- Insulinoma/blood supply
- Insulinoma/genetics
- Insulinoma/immunology
- Insulinoma/metabolism
- Insulinoma/pathology
- Insulinoma/therapy
- Integrin alphaVbeta3/metabolism
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Knockout
- Mice, Transgenic
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Pancreatic Neoplasms/blood supply
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Th1 Cells/immunology
- Th1 Cells/pathology
- Time Factors
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Nele Müller-Hermelink
- Department of Dermatology, Eberhard Karls University, Liebermeisterstrasse 25, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Shen Q, Sun Q, Wei X, Dong J, Zhang R, Wu P, Jin Y, Feng J, Li H, Hu Y. Generation and characterization of islet cell tumor in pTet-on/pTRE-SV40Tag double-transgenic mice model. J Biosci Bioeng 2007; 104:14-21. [PMID: 17697978 DOI: 10.1263/jbb.104.14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2007] [Accepted: 04/05/2007] [Indexed: 01/04/2023]
Abstract
A line of double-transgenic mice that develop neoplasms arising primarily in the pancreas was established. In these mice, the oncogene SV40 T antigen (Tag) was detected in the pancreas with and without the control of Tet-on system. The transgenic mice that developed pancreatic tumors as early as 20 weeks of age showed hypoglycemia on a blood glucose test. Pathological and immunohistochemical characterizations demonstrated that the tumors belonged to neuroendocrine neoplasms arising from pancreatic islets. A change in IGFs/IGF-1R signaling pathway was detected using real-time PCR analysis. A potential association between the IGFs/IGF-1R system and SV40Tag was studied to further explain the cancerogenesis of the double-transgenic mice by Western blot analysis and immunoprecipitation experiments. The results suggest that a Tag transgenic mice model could be used to study the molecular mechanism of the tumorigenesis of islets.
Collapse
Affiliation(s)
- Qian Shen
- Key Lab of Brain Functional Genomics, MOE & STCSM, Shanghai Institute of Brain Functional Genomics, East China Normal University, Shanghai, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Durán I, Salazar R, Casanovas O, Arrazubi V, Vilar E, Siu LL, Yao J, Tabernero J. New drug development in digestive neuroendocrine tumors. Ann Oncol 2007; 18:1307-13. [PMID: 17301070 DOI: 10.1093/annonc/mdm009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The traditional cytotoxic agents are of limited efficacy in the treatment of neuroendocrine tumors of the gastrointestinal tract (NETs). Recent investigations have brought up a number of biological features in this family of neoplasms that could represent targets for anticancer treatment. NETs seem to have an extraordinary tumor vascularization with high expression of proangiogenic molecules such as the vascular endothelial growth factor along with overexpression of certain tyrosine kinase receptors such as the epidermal growth factor receptor (EGFR), the insulin growth factor receptor (IGFR) and their downstream signaling pathway components (PI3K-AKT-mTOR). The rationale of an antiangiogenic approach in the treatment of NETs and the use of other pharmacological strategies such as EGFR, IGFR and mammalian target of rapamycin inhibitors are discussed. Additionally, the emerging results of recent clinical trials with these targeted drugs are presented.
Collapse
Affiliation(s)
- I Durán
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Maser RS, Choudhury B, Campbell PJ, Feng B, Wong KK, Protopopov A, O'Neil J, Gutierrez A, Ivanova E, Perna I, Lin E, Mani V, Jiang S, McNamara K, Zaghlul S, Edkins S, Stevens C, Brennan C, Martin ES, Wiedemeyer R, Kabbarah O, Nogueira C, Histen G, Aster J, Mansour M, Duke V, Foroni L, Fielding AK, Goldstone AH, Rowe JM, Wang YA, Look AT, Stratton MR, Chin L, Futreal PA, DePinho RA. Chromosomally unstable mouse tumours have genomic alterations similar to diverse human cancers. Nature 2007; 447:966-71. [PMID: 17515920 PMCID: PMC2714968 DOI: 10.1038/nature05886] [Citation(s) in RCA: 304] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2007] [Accepted: 04/30/2007] [Indexed: 01/07/2023]
Abstract
Highly rearranged and mutated cancer genomes present major challenges in the identification of pathogenetic events driving the neoplastic transformation process. Here we engineered lymphoma-prone mice with chromosomal instability to assess the usefulness of mouse models in cancer gene discovery and the extent of cross-species overlap in cancer-associated copy number aberrations. Along with targeted re-sequencing, our comparative oncogenomic studies identified FBXW7 and PTEN to be commonly deleted both in murine lymphomas and in human T-cell acute lymphoblastic leukaemia/lymphoma (T-ALL). The murine cancers acquire widespread recurrent amplifications and deletions targeting loci syntenic to those not only in human T-ALL but also in diverse human haematopoietic, mesenchymal and epithelial tumours. These results indicate that murine and human tumours experience common biological processes driven by orthologous genetic events in their malignant evolution. The highly concordant nature of genomic events encourages the use of genomically unstable murine cancer models in the discovery of biological driver events in the human oncogenome.
Collapse
Affiliation(s)
- Richard S Maser
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chen M, Rahman L, Voeller D, Kastanos E, Yang SX, Feigenbaum L, Allegra C, Kaye FJ, Steeg P, Zajac-Kaye M. Transgenic expression of human thymidylate synthase accelerates the development of hyperplasia and tumors in the endocrine pancreas. Oncogene 2007; 26:4817-24. [PMID: 17297449 DOI: 10.1038/sj.onc.1210273] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Thymidylate synthase (TS) is an essential enzyme for DNA synthesis and repair and elevated levels of TS have been identified as an important prognostic biomarker for colorectal cancer and several other common human malignancies. In addition, TS gene expression has been linked with cell-cycle regulation and cell proliferation through the ability of retinoblastoma protein to repress the transcriptional activation of E2F target genes such as TS. Therefore, overproduction of TS could participate in the progression to a neoplastic phenotype. Consistent with this model, a recent study has suggested that ectopic TS expression can induce a transformed phenotype in mammalian cells. To investigate the role of deregulated TS activity in tumor development, we generated transgenic mice that express high levels of catalytically active human TS (hTS) exclusively in the pancreas and low levels of hTS in multiple other tissues. Analyses of pancreatic tissue in TS transgenic mice revealed abnormalities within the endocrine pancreas, ranging from pancreatic islet hyperplasia to the detection of islet cell tumors. Overexpression of hTS in murine islets provides a new model to study genetic alterations associated with the progression from normal cells to hyperplasia to islet cell tumors, and suggests that this mouse model may be useful for regulating TS activity in vivo for development of cancer prevention and new therapies.
Collapse
Affiliation(s)
- M Chen
- Molecular Therapeutics Program, Center for Cancer Research, Bethesda, MD, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Once thought incapable of significant proliferation, the pancreatic beta-cell has recently been shown to harbor immense powers of self-renewal. Pancreatic beta-cells, the sole source of insulin in vertebrate animals, can grow facultatively to a degree unmatched by other organs in experimental animals. beta-cell growth matches changes in systemic insulin demand, which increase during common physiologic states such as aging, obesity, and pregnancy. Compensatory changes in beta-cell mass are controlled by beta-cell proliferation. Here we review recent advances in our understanding of the intrinsic factors and mechanisms that control beta-cell cycle progression. Dysregulation of beta-cell proliferation is emerging as a fundamental feature in the pathogenesis of human disease states such as cancer and diabetes mellitus. New experimental observations and studies of these diseases suggest that beta-cell fate and expansion are coordinately regulated. We speculate on how these advances may accelerate the discovery of new strategies for the treatment of diseases characterized by a deficiency or excess of beta-cells.
Collapse
Affiliation(s)
- Jeremy J Heit
- Departments of Developmental Biology and Medicine (Oncology Division), Stanford University School of Medicine, Stanford, California 94305, USA.
| | | | | |
Collapse
|
28
|
Loffler KA, Biondi CA, Gartside MG, Serewko-Auret MM, Duncan R, Tonks ID, Mould AW, Waring P, Muller HK, Kay GF, Hayward NK. Lack of augmentation of tumor spectrum or severity in dual heterozygous Men1 and Rb1 knockout mice. Oncogene 2006; 26:4009-17. [PMID: 17173065 DOI: 10.1038/sj.onc.1210163] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
To identify possible genetic interactions between the mechanisms of tumor suppression of menin and pRb, we intercrossed mice with targeted deletions of Men1 and Rb1, and compared tumor development in cohorts of animals carrying single or dual mutations of these tumor-suppressor genes. In mice lacking one copy of Men1, pancreatic islet and anterior pituitary adenomas are common. In animals lacking one copy of Rb1, intermediate pituitary and thyroid tumors occur at high frequency, with less frequent development of pancreatic islet hyperplasia and parathyroid lesions. In mice heterozygous for both Men1 and Rb1, pancreatic hyperplasia and tumors of the intermediate pituitary and thyroid occurred at high frequency. Serum measurements of calcium and glucose did not vary significantly between genotypic groups. Loss of heterozygosity at the Rb1 locus was common in pituitary and thyroid tumors, whereas loss of menin was observed in pancreatic and parathyroid lesions. The tumor spectrum in the double heterozygotes was a combination of pathologies seen in each of the individual heterozygotes, without decrease in age of onset, indicating independent, non-additive effects of the two mutations. Together with the lack of increased tumor spectrum, this suggests that menin and pRb function in a common pathway of tumor suppression.
Collapse
Affiliation(s)
- K A Loffler
- Cancer and Cell Biology Divison, Queensland Institute of Medical Research, Herston, Queensland, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|