1
|
Yang H, Ji Y, Liu D, Chai O, Qi Z, Guo R, Sun B, Wang F. LLGL2 targets the Hedgehog signaling pathway to influence malignant progression of endometrial cancer. Cell Signal 2025; 127:111553. [PMID: 39647764 DOI: 10.1016/j.cellsig.2024.111553] [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: 09/14/2024] [Revised: 11/24/2024] [Accepted: 12/03/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND Endometrial cancer (EC) is a malignant tumor of the endometrial epithelium, with endometrial adenocarcinoma being the most common type. It has a good overall prognosis, but recurrence and metastasis are often associated with poorer outcomes. LLGL2 is closely linked to tumorigenesis; however, its in endometrial carcinoma is unknown. This study investigated the biological function of LLGL2 in endometrial cancer and its intrinsic molecular mechanisms in promoting the malignant progression of the disease. METHODS The expression of LLGL2 in endometrial cancer was analyzed using the TCGA database via UALCAN and validated through western blot analysis, RT-qPCR, and immunohistochemistry. Additionally, the correlation between LLGL2 and the clinicopathological features of patients was examined. The effects of LLGL2 on the proliferation and migration of endometrial cancer cells were assessed using EdU proliferation assay, clone formation assay, Transwell assay, scratch assay, and the detection of proliferation and metastasis markers. Furthermore, the impact of LLGL2 on key genes of the Hedgehog signaling pathway, including SHH, PTCH1, SMO, and GLI1, was explored using western blot analysis and RT-qPCR. The expression of SHH, PTCH1 and GLI1 was also detected in cells treated with the Hedgehog signaling pathway inhibitor (JK184) in endometrial cancer cells overexpressing LLGL2. Finally, the role of LLGL2 in tumor growth was investigated in vivo using sh-LLGL2 and OE-LLGL2 nude mouse tumorigenic model. RESULTS LLGL2 expression was upregulated in endometrial cancer tissues compared to normal endometrium. LLGL2 expression was significantly correlated with tumor histological grading (p < 0.01). LLGL2 promoted proliferation, migration, and invasive abilities of endometrial cancer cells. In the nude mouse tumorigenic model, LLGL2 deficiency inhibited the growth of subcutaneously transplanted tumors, and overexpression of LLGL2 promoted the growth of tumors. At the molecular level, LLGL2 may promote the transduction of the Hedgehog signaling pathway in endometrial cancer cells. CONCLUSION LLGL2 may be involved in the development of endometrial cancer as an oncogene, leading to aberrant activation of the Hedgehog signaling pathway. Therefore, LLGL2 is a potential new target for the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Hua Yang
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Yuqing Ji
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Dong Liu
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Ou Chai
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, 300134 Tianjin, China
| | - Zhiying Qi
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Ruimeng Guo
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China
| | - Bei Sun
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, 300134 Tianjin, China.
| | - Fang Wang
- Department of Gynaecology, the Second Hospital of Tianjin Medical University, 300211 Tianjin, China..
| |
Collapse
|
2
|
Hedayat S, Cascione L, Cunningham D, Schirripa M, Lampis A, Hahne JC, Tunariu N, Hong SP, Marchetti S, Khan K, Fontana E, Angerilli V, Delrieux M, Nava Rodrigues D, Procaccio L, Rao S, Watkins D, Starling N, Chau I, Braconi C, Fotiadis N, Begum R, Guppy N, Howell L, Valenti M, Cribbes S, Kolozsvari B, Kirkin V, Lonardi S, Ghidini M, Passalacqua R, Elghadi R, Magnani L, Pinato DJ, Di Maggio F, Ghelardi F, Sottotetti E, Vetere G, Ciracì P, Vlachogiannis G, Pietrantonio F, Cremolini C, Cortellini A, Loupakis F, Fassan M, Valeri N. Circulating microRNA Analysis in a Prospective Co-clinical Trial Identifies MIR652-3p as a Response Biomarker and Driver of Regorafenib Resistance Mechanisms in Colorectal Cancer. Clin Cancer Res 2024; 30:2140-2159. [PMID: 38376926 DOI: 10.1158/1078-0432.ccr-23-2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE The multi-kinase inhibitor (mKi) regorafenib has demonstrated efficacy in chemorefractory patients with metastatic colorectal cancer (mCRC). However, lack of predictive biomarkers and concerns over significant toxicities hamper the use of regorafenib in clinical practice. EXPERIMENTAL DESIGN Serial liquid biopsies were obtained at baseline and monthly until disease progression in chemorefractory patients with mCRC treated with regorafenib in a phase II clinical trial (PROSPECT-R n = 40; NCT03010722) and in a multicentric validation cohort (n = 241). Tissue biopsies collected at baseline, after 2 months and at progression in the PROSPECT-R trial were used to establish patient-derived organoids (PDO) and for molecular analyses. MicroRNA profiling was performed on baseline bloods using the NanoString nCounter platform and results were validated by digital-droplet PCR and/or ISH in paired liquid and tissue biopsies. PDOs co-cultures and PDO-xenotransplants were generated for functional analyses. RESULTS Large-scale microRNA expression analysis in longitudinal matched liquid and tissue biopsies from the PROSPECT-R trial identified MIR652-3p as a biomarker of clinical benefit to regorafenib. These findings were confirmed in an independent validation cohort and in a "control" group of 100 patients treated with lonsurf. Using ex vivo co-culture assays paired with single-cell RNA-sequencing of PDO established pre- and post-treatment, we modeled regorafenib response observed in vivo and in patients, and showed that MIR652-3p controls resistance to regorafenib by impairing regorafenib-induced lethal autophagy and by orchestrating the switch from neo-angiogenesis to vessel co-option. CONCLUSIONS Our results identify MIR652-3p as a potential biomarker and as a driver of cell and non-cell-autonomous mechanisms of resistance to regorafenib.
Collapse
Affiliation(s)
- Somaieh Hedayat
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Luciano Cascione
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Bellinzona, Switzerland
| | - David Cunningham
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Marta Schirripa
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Andrea Lampis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Jens C Hahne
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Sung Pil Hong
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Silvia Marchetti
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Khurum Khan
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Elisa Fontana
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Valentina Angerilli
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Mia Delrieux
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Daniel Nava Rodrigues
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Letizia Procaccio
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Sheela Rao
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicos Fotiadis
- Department of Interventional Radiology, The Royal Marsden Hospital, London, United Kingdom
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naomy Guppy
- Breast Cancer Now Nina Barough Pathology Core Facility, The Institute of Cancer Research, London, United Kingdom
| | - Louise Howell
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Melanie Valenti
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | | | - Vladimir Kirkin
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sara Lonardi
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Raghad Elghadi
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Luca Magnani
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - David J Pinato
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Federica Di Maggio
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate Francesco Salvatore, Via Gaetano Salvatore, Naples, Italy
| | - Filippo Ghelardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Guglielmo Vetere
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Paolo Ciracì
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Georgios Vlachogiannis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessio Cortellini
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Fotios Loupakis
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Nicola Valeri
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
3
|
Portela M, Mukherjee S, Paul S, La Marca JE, Parsons LM, Veraksa A, Richardson HE. The Drosophila tumour suppressor Lgl and Vap33 activate the Hippo pathway through a dual mechanism. J Cell Sci 2024; 137:jcs261917. [PMID: 38240353 PMCID: PMC10911279 DOI: 10.1242/jcs.261917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/10/2024] [Indexed: 02/12/2024] Open
Abstract
The tumour suppressor, Lethal (2) giant larvae [Lgl; also known as L(2)gl], is an evolutionarily conserved protein that was discovered in the vinegar fly Drosophila, where its depletion results in tissue overgrowth and loss of cell polarity. Lgl links cell polarity and tissue growth through regulation of the Notch and the Hippo signalling pathways. Lgl regulates the Notch pathway by inhibiting V-ATPase activity via Vap33. How Lgl regulates the Hippo pathway was unclear. In this current study, we show that V-ATPase activity inhibits the Hippo pathway, whereas Vap33 acts to activate Hippo signalling. Vap33 physically and genetically interacts with the actin cytoskeletal regulators RtGEF (Pix) and Git, which also bind to the Hippo protein (Hpo) and are involved in the activation of the Hippo pathway. Additionally, we show that the ADP ribosylation factor Arf79F (Arf1), which is a Hpo interactor, is involved in the inhibition of the Hippo pathway. Altogether, our data suggest that Lgl acts via Vap33 to activate the Hippo pathway by a dual mechanism: (1) through interaction with RtGEF, Git and Arf79F, and (2) through interaction and inhibition of the V-ATPase, thereby controlling epithelial tissue growth.
Collapse
Affiliation(s)
- Marta Portela
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3002, Australia
| | - Swastik Mukherjee
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Sayantanee Paul
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - John E. La Marca
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Blood Cells and Blood Cancer Division, Water and Eliza Hall Institute, Melbourne, Victoria, 3052, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Victoria, 3010, Australia
- Genome Engineering and Cancer Modelling Program, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, 3084, Australia
| | - Linda M. Parsons
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3002, Australia
| | - Alexey Veraksa
- Department of Biology, University of Massachusetts Boston, Boston, MA 02125, USA
| | - Helena E. Richardson
- Department of Biochemistry & Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, 3086, Australia
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, 3002, Australia
- Sir Peter MacCallum Department of Oncology, Department of Anatomy and Neuroscience, Department of Biochemistry and Molecular Biology, University of Melbourne, Melbourne, Victoria, 3010, Australia
| |
Collapse
|
4
|
Bii VM, Rudoy D, Klezovitch O, Vasioukhin V. Lethal giant larvae gene family ( Llgl1 and Llgl2 ) functions as a tumor suppressor in mouse skin epidermis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531408. [PMID: 36945368 PMCID: PMC10028895 DOI: 10.1101/2023.03.06.531408] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/11/2023]
Abstract
Loss of cell polarity and tissue disorganization occurs in majority of epithelial cancers. Studies in simple model organisms identified molecular mechanisms responsible for the establishment and maintenance of cellular polarity, which play a pivotal role in establishing proper tissue architecture. The exact role of these cell polarity pathways in mammalian cancer is not completely understood. Here we analyzed the mammalian orthologs of drosophila apical-basal polarity gene lethal giant larvae ( lgl ), which regulates asymmetric stem cell division and functions as a tumor suppressor in flies. There are two mammalian orthologs of lgl ( Llgl1 and Llgl2 ). To determine the role of the entire lgl signaling pathway in mammals we generated mice with ablation of both Llgl1 and Llgl2 in skin epidermis using K14-Cre ( Llgl1/2 -/- cKO mice). Surprisingly, we found that ablation of Llgl1/2 genes does not impact epidermal polarity in adult mice. However, old Llgl1/2 cKO mice present with focal skin lesions which are missing epidermal layer and ripe with inflammation. To determine the role of lgl signaling pathway in cancer we generated Trp53 -/- /Llgl1/2 -/- cKO and Trp53 -/+ /Llgl1/2 -/- cKO mice. Loss of Llgl1/2 promoted squamous cell carcinoma (SCC) development in Trp53 -/- cKO and caused SCC in Trp53 -/+ cKO mice, while no cancer was observed in Trp53 -/+ cKO controls. Mechanistically, we show that ablation of Llgl1/2 causes activation of aPKC and upregulation of NF-kB signaling pathway, which may be necessary for SCC in Trp53 -/+ /Llgl1/2 -/- cKO mice. We conclude that Lgl signaling pathway functions as a tumor suppressor in mammalian skin epidermis.
Collapse
|
5
|
Sollazzo M, Paglia S, Di Giacomo S, Grifoni D. Apoptosis inhibition restrains primary malignant traits in different Drosophila cancer models. Front Cell Dev Biol 2023; 10:1043630. [PMID: 36704198 PMCID: PMC9871239 DOI: 10.3389/fcell.2022.1043630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023] Open
Abstract
Tumor cells exploit multiple mechanisms to evade apoptosis, hence the strategies aimed at reactivating cell death in cancer. However, recent studies are revealing that dying cells play remarkable pro-oncogenic roles. Among the mechanisms promoting cell death, cell competition, elicited by disparities in MYC activity in confronting cells, plays the primary role of assuring tissue robustness during development from Drosophila to mammals: cells with high MYC levels (winners) overproliferate while killing suboptimal neighbors (losers), whose death is essential to process completion. This mechanism is coopted by tumor cells in cancer initiation, where host cells succumb to high-MYC-expressing precancerous neighbors. Also in this case, inhibition of cell death restrains aberrant cell competition and rescues tissue structure. Inhibition of apoptosis may thus emerge as a good strategy to counteract cancer progression in competitive contexts; of note, we recently found a positive correlation between cell death amount at the tumor/stroma interface and MYC levels in human cancers. Here we used Drosophila to investigate the functional role of competition-dependent apoptosis in advanced cancers, observing dramatic changes in mass dimensions and composition following a boost in cell competition, rescued by apoptosis inhibition. This suggests the role of competition-dependent apoptosis be not confined to the early stages of tumorigenesis. We also show that apoptosis inhibition, beside restricting cancer mass, is sufficient to rescue tissue architecture and counteract cell migration in various cancer contexts, suggesting that a strong activation of the apoptotic pathways intensifies cancer burden by affecting distinct phenotypic traits at different stages of the disease.
Collapse
Affiliation(s)
- Manuela Sollazzo
- CanceЯEvolutionLab, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Simona Paglia
- CanceЯEvolutionLab, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Simone Di Giacomo
- CanceЯEvolutionLab, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Daniela Grifoni
- CanceЯEvolutionLab, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy,CanceЯEvolutionLab, Department of Life, Health and Environmental Sciences, University of L’Aquila, L’Aquila, Italy,*Correspondence: Daniela Grifoni,
| |
Collapse
|
6
|
Cell polarity and extrusion: How to polarize extrusion and extrude misspolarized cells? Curr Top Dev Biol 2023; 154:131-167. [PMID: 37100516 DOI: 10.1016/bs.ctdb.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
The barrier function of epithelia is one of the cornerstones of the body plan organization of metazoans. It relies on the polarity of epithelial cells which organizes along the apico-basal axis the mechanical properties, signaling as well as transport. This barrier function is however constantly challenged by the fast turnover of epithelia occurring during morphogenesis or adult tissue homeostasis. Yet, the sealing property of the tissue can be maintained thanks to cell extrusion: a series of remodeling steps involving the dying cell and its neighbors leading to seamless cell expulsion. Alternatively, the tissue architecture can also be challenged by local damages or the emergence of mutant cells that may alter its organization. This includes mutants of the polarity complexes which can generate neoplastic overgrowths or be eliminated by cell competition when surrounded by wild type cells. In this review, we will provide an overview of the regulation of cell extrusion in various tissues focusing on the relationship between cell polarity, cell organization and the direction of cell expulsion. We will then describe how local perturbations of polarity can also trigger cell elimination either by apoptosis or by cell exclusion, focusing specifically on how polarity defects can be directly causal to cell elimination. Overall, we propose a general framework connecting the influence of polarity on cell extrusion and its contribution to aberrant cell elimination.
Collapse
|
7
|
Chatterjee D, Costa CAM, Wang XF, Jevitt A, Huang YC, Deng WM. Single-cell transcriptomics identifies Keap1-Nrf2 regulated collective invasion in a Drosophila tumor model. eLife 2022; 11:80956. [PMID: 36321803 PMCID: PMC9708074 DOI: 10.7554/elife.80956] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/01/2022] [Indexed: 11/30/2022] Open
Abstract
Apicobasal cell polarity loss is a founding event in epithelial-mesenchymal transition and epithelial tumorigenesis, yet how pathological polarity loss links to plasticity remains largely unknown. To understand the mechanisms and mediators regulating plasticity upon polarity loss, we performed single-cell RNA sequencing of Drosophila ovaries, where inducing polarity-gene l(2)gl-knockdown (Lgl-KD) causes invasive multilayering of the follicular epithelia. Analyzing the integrated Lgl-KD and wildtype transcriptomes, we discovered the cells specific to the various discernible phenotypes and characterized the underlying gene expression. A genetic requirement of Keap1-Nrf2 signaling in promoting multilayer formation of Lgl-KD cells was further identified. Ectopic expression of Keap1 increased the volume of delaminated follicle cells that showed enhanced invasive behavior with significant changes to the cytoskeleton. Overall, our findings describe the comprehensive transcriptome of cells within the follicle cell tumor model at the single-cell resolution and identify a previously unappreciated link between Keap1-Nrf2 signaling and cell plasticity at early tumorigenesis.
Collapse
Affiliation(s)
- Deeptiman Chatterjee
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, United States
| | - Caique Almeida Machado Costa
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, United States
| | - Xian-Feng Wang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, United States
| | - Allison Jevitt
- Department of Biological Science, Florida State University, Tallahassee, United States
| | - Yi-Chun Huang
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, United States
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, Tulane Cancer Center, New Orleans, United States.,Department of Biological Science, Florida State University, Tallahassee, United States
| |
Collapse
|
8
|
Cartón-García F, Brotons B, Anguita E, Dopeso H, Tarragona J, Nieto R, García-Vidal E, Macaya I, Zagyva Z, Dalmau M, Sánchez-Martín M, van Ijzendoorn SCD, Landolfi S, Hernandez-Losa J, Schwartz Jr S, Matias-Guiu X, Ramón y Cajal S, Martínez-Barriocanal Á, Arango D. Myosin Vb as a tumor suppressor gene in intestinal cancer. Oncogene 2022; 41:5279-5288. [DOI: 10.1038/s41388-022-02508-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 10/08/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
|
9
|
Kim KH, Hong GL, Kim YJ, Lee HJ, Jung JY. Silencing of LLGL2 Suppresses the Estradiol-Induced BPH-1 Cell Proliferation through the Regulation of Autophagy. Biomedicines 2022; 10:biomedicines10081981. [PMID: 36009528 PMCID: PMC9406103 DOI: 10.3390/biomedicines10081981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/05/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Lethal giant larvae (Lgl) is an apical-basal polarity gene first identified in Drosophila. LLGL2 is one of the mammalian homologs of Lgl. However, little is known about its function in the prostate. In this study, to explore the new role of LLGL2 in the prostate, we examined the proliferative activity of a BPH-1 cell line, a well-established model for the human prostate biology of benign prostatic hyperplasia (BPH). The expression of LLGL2 was dose-dependently increased in BPH-1 cells after treatment with 17β-estradiol (E2). Additionally, E2 treatment increased the proliferation of the BPH-1 cells. However, the knockdown of LLGL2 with siRNA significantly suppressed the proliferation of the E2-treated BPH-1 cells. Moreover, si-llgl2 treatment up-regulated the expression of LC-3B, ATG7, and p-beclin, which are known to play a pivotal role in autophagosome formation in E2-treated BPH-1 cells. Overexpression of LLGL2 was able to further prove these findings by showing the opposite results from the knockdown of LLGL2 in E2-treated BPH-1 cells. Collectively, our results suggest that LLGL2 is closely involved in the proliferation of prostate cells by regulating autophagosome formation. These results provide a better understanding of the mechanism involved in the effect of LLGL2 on prostate cell proliferation. LLGL2 might serve as a potential target in the diagnosis and/or treatment of human BPH.
Collapse
Affiliation(s)
| | | | | | | | - Ju-Young Jung
- Correspondence: ; Tel.: +82-42-821-8899; Fax: +82-42-821-7926
| |
Collapse
|
10
|
Munnik C, Xaba MP, Malindisa ST, Russell BL, Sooklal SA. Drosophila melanogaster: A platform for anticancer drug discovery and personalized therapies. Front Genet 2022; 13:949241. [PMID: 36003330 PMCID: PMC9393232 DOI: 10.3389/fgene.2022.949241] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer is a complex disease whereby multiple genetic aberrations, epigenetic modifications, metabolic reprogramming, and the microenvironment contribute to the development of a tumor. In the traditional anticancer drug discovery pipeline, drug candidates are usually screened in vitro using two-dimensional or three-dimensional cell culture. However, these methods fail to accurately mimic the human disease state. This has led to the poor success rate of anticancer drugs in the preclinical stages since many drugs are abandoned due to inefficacy or toxicity when transitioned to whole-organism models. The common fruit fly, Drosophila melanogaster, has emerged as a beneficial system for modeling human cancers. Decades of fundamental research have shown the evolutionary conservation of key genes and signaling pathways between flies and humans. Moreover, Drosophila has a lower genetic redundancy in comparison to mammals. These factors, in addition to the advancement of genetic toolkits for manipulating gene expression, allow for the generation of complex Drosophila genotypes and phenotypes. Numerous studies have successfully created Drosophila models for colorectal, lung, thyroid, and brain cancers. These models were utilized in the high-throughput screening of FDA-approved drugs which led to the identification of several compounds capable of reducing proliferation and rescuing phenotypes. More noteworthy, Drosophila has also unlocked the potential for personalized therapies. Drosophila ‘avatars’ presenting the same mutations as a patient are used to screen multiple therapeutic agents targeting multiple pathways to find the most appropriate combination of drugs. The outcomes of these studies have translated to significant responses in patients with adenoid cystic carcinoma and metastatic colorectal cancers. Despite not being widely utilized, the concept of in vivo screening of drugs in Drosophila is making significant contributions to the current drug discovery pipeline. In this review, we discuss the application of Drosophila as a platform in anticancer drug discovery; with special focus on the cancer models that have been generated, drug libraries that have been screened and the status of personalized therapies. In addition, we elaborate on the biological and technical limitations of this system.
Collapse
Affiliation(s)
- Chamoné Munnik
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Malungi P. Xaba
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Sibusiso T. Malindisa
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
| | - Bonnie L. Russell
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- Buboo (Pty) Ltd, The Innovation Hub, Pretoria, South Africa
| | - Selisha A. Sooklal
- Department of Life and Consumer Sciences, University of South Africa, Pretoria, South Africa
- *Correspondence: Selisha A. Sooklal,
| |
Collapse
|
11
|
Weidle UH, Nopora A. MicroRNAs Involved in Small-cell Lung Cancer as Possible Agents for Treatment and Identification of New Targets. Cancer Genomics Proteomics 2021; 18:591-603. [PMID: 34479913 DOI: 10.21873/cgp.20283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/13/2021] [Accepted: 07/15/2021] [Indexed: 11/10/2022] Open
Abstract
Small-cell lung cancer, a neuro-endocrine type of lung cancers, responds very well to chemotherapy-based agents. However, a high frequency of relapse due to adaptive resistance is observed. Immunotherapy-based treatments with checkpoint inhibitors has resulted in improvement of treatment but the responses are not as impressive as in other types of tumor. Therefore, identification of new targets and treatment modalities is an important issue. After searching the literature, we identified eight down-regulated microRNAs involved in radiation- and chemotherapy-induced resistance, as well as three up-regulated and four down-regulated miRNAs with impacts on proliferation, invasion and apoptosis of small-cell lung cancer cells in vitro. Furthermore, one up-regulated and four down-regulated microRNAs with in vivo activity in SCLC cell xenografts were identified. The identified microRNAs are candidates for inhibition or reconstitution therapy. The corresponding targets are candidates for inhibition or functional reconstitution with antibody-based moieties or small molecules.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| | - Adam Nopora
- Roche Pharma Research and Early Development, Roche Innovation Center Munich, Penzberg, Germany
| |
Collapse
|
12
|
Modeling Notch-Induced Tumor Cell Survival in the Drosophila Ovary Identifies Cellular and Transcriptional Response to Nuclear NICD Accumulation. Cells 2021; 10:cells10092222. [PMID: 34571871 PMCID: PMC8465586 DOI: 10.3390/cells10092222] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/02/2021] [Accepted: 08/23/2021] [Indexed: 12/26/2022] Open
Abstract
Notch is a conserved developmental signaling pathway that is dysregulated in many cancer types, most often through constitutive activation. Tumor cells with nuclear accumulation of the active Notch receptor, NICD, generally exhibit enhanced survival while patients experience poorer outcomes. To understand the impact of NICD accumulation during tumorigenesis, we developed a tumor model using the Drosophila ovarian follicular epithelium. Using this system we demonstrated that NICD accumulation contributed to larger tumor growth, reduced apoptosis, increased nuclear size, and fewer incidents of DNA damage without altering ploidy. Using bulk RNA sequencing we identified key genes involved in both a pre- and post- tumor response to NICD accumulation. Among these are genes involved in regulating double-strand break repair, chromosome organization, metabolism, like raptor, which we experimentally validated contributes to early Notch-induced tumor growth. Finally, using single-cell RNA sequencing we identified follicle cell-specific targets in NICD-overexpressing cells which contribute to DNA repair and negative regulation of apoptosis. This valuable tumor model for nuclear NICD accumulation in adult Drosophila follicle cells has allowed us to better understand the specific contribution of nuclear NICD accumulation to cell survival in tumorigenesis and tumor progression.
Collapse
|
13
|
Leng S, Xie F, Liu J, Shen J, Quan G, Wen T. LLGL2 Increases Ca 2+ Influx and Exerts Oncogenic Activities via PI3K/AKT Signaling Pathway in Hepatocellular Carcinoma. Front Oncol 2021; 11:683629. [PMID: 34178676 PMCID: PMC8223678 DOI: 10.3389/fonc.2021.683629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/20/2021] [Indexed: 02/05/2023] Open
Abstract
Background Lethal giant larvae (Lgl), scaffolding proteins, regulate the epithelial cell apicobasal polarity in Drosophila. They play important roles in asymmetric cell division, cell migration, and progenitor cells self-renewal as tumor suppressors. One of Lgl mammalian homologues proteins, LLGL2 overexpression has been reported in ER+ breast cancer and promotes tumor proliferation through regulating leucine uptake. Nonetheless, the role of LLGL2 in hepatocellular carcinoma (HCC) is still unknown. Methods TCGA dataset mining, qRT-PCR, Western blot along with immunohistochemistry assays were employed to explore LLGL2 expression in human HCC samples and cell lines. Moreover, the clinical value of LLGL2 was investigated in 156 HCC patients. Furthermore, the role as well as the molecular mechanism of LLGL2 in the progression of HCC was explored through a series of in vitro and in vivo experiments. Results LLGL2 was up-regulated in HCC tissues, which was related with certain clinicopathological features including tumor number, vascular invasion as well as advanced stage. High expression of LLGL2 predicted poor prognosis after hepatectomy. LLGL2 promoted HCC cells proliferation, migration and invasion through PI3K/ATK signaling by promoting calcium ion influx. Conclusion Our study identified that LLGL2 is a tumor promoter in HCC for the first time, which could potentially be utilized as a new biomarker and a therapeutic target for HCC.
Collapse
Affiliation(s)
- Shusheng Leng
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China.,General Surgery Department, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Fei Xie
- Department of Hepatobiliary, Pancreatic and Splenic Surgery, The First People's Hospital of Neijiang City, Neijiang, China
| | - Junyi Liu
- Central Laboratory, Affiliated Hospital/Clinical Medical College of Chengdu University, Chengdu, China
| | - Junyi Shen
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| | - Guangqian Quan
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| | - Tianfu Wen
- Department of Liver Surgery and Liver Transplantation Center, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Wang Y, Zhang Y, Sang B, Zhu X, Yu R, Zhou X. Human giant larvae-1 promotes migration and invasion of malignant glioma cells by regulating N-cadherin. Oncol Lett 2021; 21:167. [PMID: 33552285 PMCID: PMC7798033 DOI: 10.3892/ol.2021.12428] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022] Open
Abstract
Human giant larvae-1 (Hugl-1) is a human homologue of Drosophila tumor suppressor lethal (2)-giant larvae and has been reported to be involved in the development of human malignancies. Previous studies performed by our group demonstrated that Hugl-1 inhibits glioma cell proliferation in an intracranial model of nude mice. However, the exact molecular mechanisms underlying the participation of Hugl-1 in glioma invasion and migration, and in the depolarizing process remain largely unknown. Utilizing the U251-MG cells with stable expression of Hugl-1, the present study used wound healing, Transwell invasion and western blot assays to explore the role and specific mechanism of Hugl-1 in glioma invasion and migration. The results of the present study demonstrated that overexpression of Hugl-1 decreased cell-cell adhesion and increased cell-cell extracellular matrix adhesion. In addition, overexpression of Hugl-1 promoted pseudopodia formation, glioma cell migration and invasion. The molecular mechanism of action involved the negative regulation of N-cadherin protein levels by Hugl-1. Overexpression or knockdown of N-cadherin partially suppressed or enhanced the effects of Hugl-1 on glioma cell migration and invasion, respectively. Furthermore, Hugl-1 inhibited cell proliferation, while promoting cell migration, which suggests that it may serve a two-sided biological role in cellular processes. Taken together, these results suggest that Hugl-1 promotes the migration and invasion of malignant glioma cells by decreasing N-cadherin expression. Thus, Hugl-1 may be applied in the development of targeted and personalized treatment.
Collapse
Affiliation(s)
- Yan Wang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Yu Zhang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Ben Sang
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xianlong Zhu
- The Graduate School, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China.,Department of Neurosurgery, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, P.R. China
| |
Collapse
|
15
|
Miao Y, Cao F, Li P, Liu P. DNA methylation of Hugl-2 is a prognostic biomarker in kidney renal clear cell carcinoma. Clin Exp Pharmacol Physiol 2021; 48:44-53. [PMID: 32754907 PMCID: PMC7821335 DOI: 10.1111/1440-1681.13390] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 07/17/2020] [Accepted: 07/30/2020] [Indexed: 01/13/2023]
Abstract
It has been reported that loss of Hugl-2 contributes to tumour formation and progression in vitro and in vivo. However, whether Hugl-2 levels decrease during kidney renal clear cell carcinoma (KIRC) and the mechanism involved remain unknown. This study aimed to investigate whether DNA methylation of Hugl-2 reduces its expression, leading to the progression and poor prognosis of KIRC. Hugl-2 methylation and mRNA expression and KIRC clinicopathological data were extracted from The Cancer Genome Atlas (TCGA), and relationships among these factors were analyzed using UALCAN, MethHC, Wanderer and LinkedOmics web tools. We found that Hugl-2 mRNA and protein levels were reduced in KIRC tissues. Moreover, Hugl-2 mRNA levels were related to tumour grade and overall survival, and Hugl-2 methylation was increased in KIRC. According to the results of methylation-specific PCR, KIRC cells had higher Hugl-2 DNA methylation levels than HKC cells. Moreover, Hugl-2 DNA methylation correlated negatively with Hugl-2 mRNA and was also related to the pathology and T stage of KIRC patients. KIRC patients with high Hugl-2 DNA methylation also had shorter overall survival. Additionally, methylation of cg08827674, a Hugl-2 probe, was related to pathologic stage, T stage, neoplasm histologic grade, serum calcium level without laterality, M stage, N stage, and ethnicity. Furthermore, treatment with the DNA methylation inhibitor decitabine resulted in upregulation of Hugl-2 mRNA and protein levels in KIRC cell lines. These results indicate that Hugl-2 DNA methylation may be both a prognostic marker and a therapeutic target in KIRC.
Collapse
Affiliation(s)
- Yi Miao
- Center for Translational MedicineThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Fang Cao
- Center for Translational MedicineThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Pingping Li
- Center for Translational MedicineThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| | - Peijun Liu
- Center for Translational MedicineThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
- Key Laboratory for Tumor Precision Medicine of Shaanxi ProvinceThe First Affiliated Hospital of Xi’an Jiaotong UniversityXi’anChina
| |
Collapse
|
16
|
Nandy N, Roy JK. Rab11 is essential for lgl mediated JNK-Dpp signaling in dorsal closure and epithelial morphogenesis in Drosophila. Dev Biol 2020; 464:188-201. [PMID: 32562757 DOI: 10.1016/j.ydbio.2020.06.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 06/11/2020] [Accepted: 06/11/2020] [Indexed: 11/30/2022]
Abstract
Dorsal closure during Drosophila embryogenesis provides a robust genetic platform to study the basic cellular mechanisms that govern epithelial wound healing and morphogenesis. As dorsal closure proceeds, the lateral epithelial tissue (LE) adjacent to the dorsal opening advance contra-laterally, with a simultaneous retraction of the amnioserosa. The process involves a fair degree of coordinated cell shape changes in the dorsal most epithelial (DME) cells as well as a few penultimate rows of lateral epithelial (LE) cells (collectively referred here as Dorsolateral Epithelial (DLE) cells), lining the periphery of the amnioserosa, which in due course of time extend contra-laterally and ultimately fuse over the dorsal hole, giving rise to a dorsal epithelial continuum. The JNK-Dpp signaling in the dorsolateral epidermis, plays an instrumental role in guiding their fate during this process. A large array of genes have been reported to be involved in the regulation of this core signaling pathway, yet the mechanisms by which they do so is hitherto unclear, which forms the objective of our present study. Here we show a probable mechanism via which lgl, a conserved tumour suppressor gene, regulates the JNK-Dpp pathway during dorsal closure and epithelial morphogenesis. A conditional/targeted knock-down of lgl in the dorsolateral epithelium of embryos results in failure of dorsal closure. Interestingly, we also observed a similar phenotype in a Rab11 knockdown condition. Our experiment suggests Rab11 to be interacting with lgl as they seem to synergize in order to regulate the core JNK-Dpp signaling pathway during dorsal closure and also during adult thorax closure process.
Collapse
Affiliation(s)
- Nabarun Nandy
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, India
| | - Jagat Kumar Roy
- Cytogenetics Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
17
|
Abedrabbo M, Ravid S. Scribble, Lgl1, and myosin II form a complex in vivo to promote directed cell migration. Mol Biol Cell 2020; 31:2234-2248. [PMID: 32697665 PMCID: PMC7550706 DOI: 10.1091/mbc.e19-11-0657] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Scribble (Scrib) and Lethal giant larvae 1 (Lgl1) are conserved polarity proteins that play important roles in different forms of cell polarity. The roles of Scrib and Lgl1 in apical-basal cell polarity have been studied extensively, but little is known about their roles in the cell polarity of migrating cells. Furthermore, the effect of Scrib and Lgl1 interaction on cell polarity is largely unknown. In this study, we show that Scrib, through its leucine-rich repeat domain, forms a complex in vivo with Lgl1. Scrib also forms a complex with myosin II, and Scrib, Lgl1, and myosin II colocalize at the leading edge of migrating cells. The cellular localization and the cytoskeletal association of Scrib and Lgl1 are interdependent, as depletion of either protein affects its counterpart. In addition, depletion of either Scrib or Lgl1 disrupts the cellular localization of myosin II. We show that depletion of either Scrib or Lgl1 affects cell adhesion through the inhibition of focal adhesion disassembly. Finally, we show that Scrib and Lgl1 are required for proper cell polarity of migrating cells. These results provide new insights into the mechanism regulating the cell polarity of migrating cells by Scrib, Lgl1, and myosin II.
Collapse
Affiliation(s)
- Maha Abedrabbo
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| | - Shoshana Ravid
- Department of Biochemistry and Molecular Biology, The Institute of Medical Research Israel-Canada, The Hebrew University-Hadassah Medical School, Jerusalem 91120, Israel
| |
Collapse
|
18
|
Loss of LLGL1 Expression Correlates with Diffuse Gastric Cancer and Distant Peritoneal Metastases. Can J Gastroenterol Hepatol 2019; 2019:2920493. [PMID: 31058107 PMCID: PMC6463686 DOI: 10.1155/2019/2920493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 02/21/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Loss of LLGL1 has been associated with loss of cellular adhesion and dissemination of cells from colorectal cancer and malignant melanoma. Regulation and relevance of LLGL1 were analyzed in gastric cancer patients with lymphatic and distant dissemination. Furthermore, LLGL1 expression was analyzed in relation to the cellular adhesion protein E-cadherin. METHODS LLGL1 and E-cadherin transcription levels were evaluated in 56 gastric cancer patients and five gastric cancer cell lines. IHC staining for LLGL1 was performed on 39 gastric cancer specimens. LLGL1 was stably transfected into LLGL1 negative gastric cancer cell line SNU16 (del(17) (p11.2)) for functional in vitro assays and a xenograft bioassay. RESULTS Gastric cancer specimens and cell lines displayed LLGL1 and E-cadherin expression levels with variable intensity. In gastric mucosa, LLGL1 exhibited weak cytoplasmic and strong cortical staining. Loss of LLGL1 expression occurred in 65% of gastric cancers and significantly correlated with loss of E-cadherin expression (P=0.00009). Loss of LLGL1 expression was associated with the diffuse type of gastric cancer (P=0.029) with peritoneal carcinomatosis (M1; P=0.006) and with female gender (P=0.017). Stable reexpression of LLGL1 in SNU16 cells significantly increased both plastic surface adhesion and extracellular matrix proteins laminin and fibronectin, but had no impact on in vitro proliferation, apoptosis, or invasion or on in vivo proliferation or differentiation in our xenograft bioassay. CONCLUSION LLGL1 is coexpressed with E-cadherin. Loss of expression of either protein is associated with diffuse gastric cancer and peritoneal metastases. LLGL1 does not impact on proliferation or epithelial-mesenchymal transition (EMT) rather increasing cellular adhesion.
Collapse
|
19
|
Mirzoyan Z, Sollazzo M, Allocca M, Valenza AM, Grifoni D, Bellosta P. Drosophila melanogaster: A Model Organism to Study Cancer. Front Genet 2019; 10:51. [PMID: 30881374 PMCID: PMC6405444 DOI: 10.3389/fgene.2019.00051] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/21/2019] [Indexed: 12/26/2022] Open
Abstract
Cancer is a multistep disease driven by the activation of specific oncogenic pathways concomitantly with the loss of function of tumor suppressor genes that act as sentinels to control physiological growth. The conservation of most of these signaling pathways in Drosophila, and the ability to easily manipulate them genetically, has made the fruit fly a useful model organism to study cancer biology. In this review we outline the basic mechanisms and signaling pathways conserved between humans and flies responsible of inducing uncontrolled growth and cancer development. Second, we describe classic and novel Drosophila models used to study different cancers, with the objective to discuss their strengths and limitations on their use to identify signals driving growth cell autonomously and within organs, drug discovery and for therapeutic approaches.
Collapse
Affiliation(s)
- Zhasmine Mirzoyan
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Mariateresa Allocca
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | | | - Daniela Grifoni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Paola Bellosta
- Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy.,Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy.,Department of Biosciences, University of Milan, Milan, Italy.,Department of Medicine, NYU Langone Medical Center, New York, NY, United States
| |
Collapse
|
20
|
Zhang T, Zhang S, Song X, Zhao X, Hou C, Li Z, Gao J. Loss of Lgl1 Disrupts the Radial Glial Fiber-guided Cortical Neuronal Migration and Causes Subcortical Band Heterotopia in Mice. Neuroscience 2018; 400:132-145. [PMID: 30597194 DOI: 10.1016/j.neuroscience.2018.12.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/16/2022]
Abstract
Radial glial cells (RGCs) are neuronal progenitors and function as scaffolds for neuronal radial migration in the developing cerebral cortex. These functions depend on a polarized radial glial scaffold, which is of fundamental importance for brain development. Lethal giant larvae 1 (Lgl1), a key regulator for cell polarity from Drosophila to mammals, plays a key role in tumorigenesis and brain development. To overcome neonatal lethality in Lgl1-null mice and clarify the role of Lgl1 in mouse cerebral cortex development and function, we created Lgl1 dorsal telencephalon-specific knockout mice mediated by Emx1-Cre. Lgl1Emx1 conditional knockout (CKO) mice had normal life spans and could be used for function research. Histology results revealed that the mutant mice displayed an ectopic cortical mass in the dorsolateral hemispheric region between the normotopic cortex and the subcortical white matter, resembling human subcortical band heterotopia (SBH). The Lgl1Emx1 CKO cortex showed disrupted adherens junctions (AJs), which were accompanied by ectopic RGCs and intermediate progenitors, and disorganization of the radial glial fiber system. The early- and late-born neurons failed to reach the destined position along the disrupted radial glial fiber scaffold and instead accumulated in ectopic positions and formed SBH. Additionally, the absence of Lgl1 led to severe abnormalities in RGCs, including hyperproliferation, impaired differentiation, and increased apoptosis. Lgl1Emx1 CKO mice also displayed deficiencies in anxiety-related behaviors. We concluded that Lgl1 is essential for RGC development and neural migration during cerebral cortex development.
Collapse
Affiliation(s)
- Tingting Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Sen Zhang
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xinli Song
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Xiaohan Zhao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Congzhe Hou
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China
| | - Zhenzu Li
- Department of Bioengineering, Shandong Polytechnic, Jinan 250104, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan 250100, China.
| |
Collapse
|
21
|
Shen H, Chai H, Li M, Zhou Z, Liang Y, Yang Z, Huang H, Liu X, Zhang B. Robust sparse accelerated failure time model for survival analysis. Technol Health Care 2018; 26:55-63. [PMID: 29689755 PMCID: PMC6004954 DOI: 10.3233/thc-174141] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
To identify the bio-mark genes related to disease with high dimension and low sample size gene expression data, various regression approaches with different regularization methods have been proposed to solve this problem. Nevertheless, high-noises in biological data significantly reduce the performances of methods. The accelerated failure time (AFT) modelwas designed for gene selection and survival time estimation in cancer survival analysis. In this article, we proposed a novel robust sparse accelerated failure time model (RS-AFT) through combining the least absolute deviation (LAD) and Lq regularization. An iterative weighted linear programming algorithm without regularization parameter tuning was proposed to solve this RS-AFT model. The results of the experiments show our method has better performancebothin gene selection and survival time estimationthan some widely used regularization methods such as lasso, elastic net and SCAD. Hence we thought the RS-AFT model may be a competitive regularization method in cancer survival analysis.
Collapse
Affiliation(s)
| | | | | | | | - Yong Liang
- Corresponding author: Yong Liang, Faculty of Information Technology and State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau 999078, China. Tel.: +853 63869506; Fax: +853 88972034; E-mail: .
| | | | | | | | | |
Collapse
|
22
|
Greenwood E, Maisel S, Ebertz D, Russ A, Pandey R, Schroeder J. Llgl1 prevents metaplastic survival driven by epidermal growth factor dependent migration. Oncotarget 2018; 7:60776-60792. [PMID: 27542214 PMCID: PMC5308616 DOI: 10.18632/oncotarget.11320] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/02/2016] [Indexed: 12/18/2022] Open
Abstract
We have previously demonstrated that Llgl1 loss results in a gain of mesenchymal phenotypes and a loss of apicobasal and planar polarity. We now demonstrate that these changes represent a fundamental shift in cellular phenotype. Llgl1 regulates the expression of multiple cell identity markers, including CD44, CD49f, and CD24, and the nuclear translocation of TAZ and Slug. Cells lacking Llgl1 form mammospheres, where survival and transplantability is dependent upon the Epidermal Growth Factor Receptor (EGFR). Additionally, Llgl1 loss allows cells to grow in soft-agar and maintain prolonged survival as orthotopic transplants in NOD-SCIDmice. Lineage tracing and wound healing experiments demonstrate that mammosphere survival is due to enhanced EGF-dependent migration. The loss of Llgl1 drives EGFR mislocalization and an EGFR mislocalization point mutation (P667A) drives these same phenotypes, including activation of AKT and TAZ nuclear translocation. Together, these data indicate that the loss of Llgl1 results in EGFR mislocalization, promoting pre-neoplastic changes.
Collapse
Affiliation(s)
- Erin Greenwood
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Sabrina Maisel
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| | - David Ebertz
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona
| | - Atlantis Russ
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona
| | - Ritu Pandey
- Arizona Cancer Center, University of Arizona, Tucson, Arizona.,Department of Cell and Molecular Medicine, University of Arizona, Tucson, Arizona
| | - Joyce Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona.,Arizona Cancer Center, University of Arizona, Tucson, Arizona.,BIO5 Institute, University of Arizona, Tucson, Arizona.,Genetics Program, University of Arizona, Tucson, Arizona.,Cancer Biology Program, University of Arizona, Tucson, Arizona
| |
Collapse
|
23
|
Parvy JP, Hodgson JA, Cordero JB. Drosophila as a Model System to Study Nonautonomous Mechanisms Affecting Tumour Growth and Cell Death. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7152962. [PMID: 29725601 PMCID: PMC5872677 DOI: 10.1155/2018/7152962] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/04/2018] [Indexed: 12/26/2022]
Abstract
The study of cancer has represented a central focus in medical research for over a century. The great complexity and constant evolution of the pathology require the use of multiple research model systems and interdisciplinary approaches. This is necessary in order to achieve a comprehensive understanding into the mechanisms driving disease initiation and progression, to aid the development of appropriate therapies. In recent decades, the fruit fly Drosophila melanogaster and its associated powerful genetic tools have become a very attractive model system to study tumour-intrinsic and non-tumour-derived processes that mediate tumour development in vivo. In this review, we will summarize recent work on Drosophila as a model system to study cancer biology. We will focus on the interactions between tumours and their microenvironment, including extrinsic mechanisms affecting tumour growth and how tumours impact systemic host physiology.
Collapse
Affiliation(s)
- Jean-Philippe Parvy
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Joseph A. Hodgson
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Julia B. Cordero
- CRUK Beatson Institute, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
- Wolfson Wohl Cancer Research Centre, Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1QH, UK
| |
Collapse
|
24
|
Daniel SG, Russ AD, Guthridge KM, Raina AI, Estes PS, Parsons LM, Richardson HE, Schroeder JA, Zarnescu DC. miR-9a mediates the role of Lethal giant larvae as an epithelial growth inhibitor in Drosophila. Biol Open 2018; 7:bio.027391. [PMID: 29361610 PMCID: PMC5829493 DOI: 10.1242/bio.027391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Drosophila lethal giant larvae (lgl) encodes a conserved tumor suppressor with established roles in cell polarity, asymmetric division, and proliferation control. Lgl's human orthologs, HUGL1 and HUGL2, are altered in human cancers, however, its mechanistic role as a tumor suppressor remains poorly understood. Based on a previously established connection between Lgl and Fragile X protein (FMRP), a miRNA-associated translational regulator, we hypothesized that Lgl may exert its role as a tumor suppressor by interacting with the miRNA pathway. Consistent with this model, we found that lgl is a dominant modifier of Argonaute1 overexpression in the eye neuroepithelium. Using microarray profiling we identified a core set of ten miRNAs that are altered throughout tumorigenesis in Drosophila lgl mutants. Among these are several miRNAs previously linked to human cancers including miR-9a, which we found to be downregulated in lgl neuroepithelial tissues. To determine whether miR-9a can act as an effector of Lgl in vivo, we overexpressed it in the context of lgl knock-down by RNAi and found it able to reduce the overgrowth phenotype caused by Lgl loss in epithelia. Furthermore, cross-comparisons between miRNA and mRNA profiling in lgl mutant tissues and human breast cancer cells identified thrombospondin (tsp) as a common factor altered in both fly and human breast cancer tumorigenesis models. Our work provides the first evidence of a functional connection between Lgl and the miRNA pathway, demonstrates that miR-9a mediates Lgl's role in restricting epithelial proliferation, and provides novel insights into pathways controlled by Lgl during tumor progression. Summary: Mir-9a overexpression can suppress the overgrowth phenotype caused by Lgl knock-down in epithelia. Gene profiling identifies pathways dysregulated in lgl mutants and shared features between flies and human cancer cells.
Collapse
Affiliation(s)
- Scott G Daniel
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Atlantis D Russ
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA.,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Kathryn M Guthridge
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia
| | - Ammad I Raina
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Patricia S Estes
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Linda M Parsons
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia.,Department of Genetics, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Helena E Richardson
- Cell Cycle and Development Laboratory, Research Division, Peter MacCallum Cancer Center, Melbourne, Victoria 3000, Australia.,Sir Peter MacCallum Department of Oncology, Department of Anatomy & Neuroscience, Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3000, Australia.,Department of Biochemistry & Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Joyce A Schroeder
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA.,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| | - Daniela C Zarnescu
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA .,Genetics Graduate Interdisciplinary Program, University of Arizona, Tucson, AZ 85721, USA.,Arizona Cancer Center, University of Arizona, Tucson, AZ 85721, USA
| |
Collapse
|
25
|
Lim KH, Suresh B, Park JH, Kim YS, Ramakrishna S, Baek KH. Ubiquitin-specific protease 11 functions as a tumor suppressor by modulating Mgl-1 protein to regulate cancer cell growth. Oncotarget 2018; 7:14441-57. [PMID: 26919101 PMCID: PMC4924727 DOI: 10.18632/oncotarget.7581] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Accepted: 01/31/2016] [Indexed: 12/17/2022] Open
Abstract
The Lethal giant larvae (Lgl) gene encodes a cortical cytoskeleton protein, Lgl, and is involved in maintaining cell polarity and epithelial integrity. Previously, we observed that Mgl-1, a mammalian homologue of the Drosophila tumor suppressor protein Lgl, is subjected to degradation via ubiquitin-proteasome pathway, and scaffolding protein RanBPM prevents the turnover of the Mgl-1 protein. Consequently, overexpression of RanBPM enhances Mgl-1-mediated cell proliferation and migration. Here, we analyzed the ability of ubiquitin-specific protease 11 (USP11) as a novel regulator of Mgl-1 and it requires RanBPM to regulate proteasomal degradation of Mgl-1. USP11 showed deubiquitinating activity and stabilized Mgl-1 protein. However, USP11-mediated Mgl-1 stabilization was inhibited in RanBPM-knockdown cells. Furthermore, in the cancer cell migration, the regulation of Mgl-1 by USP11 required RanBPM expression. In addition, an in vivo study revealed that depletion of USP11 leads to tumor formation. Taken together, the results indicated that USP11 functions as a tumor suppressor through the regulation of Mgl-1 protein degradation via RanBPM.
Collapse
Affiliation(s)
- Key-Hwan Lim
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| | - Bharathi Suresh
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| | - Jung-Hyun Park
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| | - Young-Soo Kim
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| | - Suresh Ramakrishna
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| | - Kwang-Hyun Baek
- Department of Biomedical Science, CHA University, Gyeonggi-Do 463-400, Republic of Korea
| |
Collapse
|
26
|
Calleja M, Morata G, Casanova J. Tumorigenic Properties of Drosophila Epithelial Cells Mutant for lethal giant larvae. Dev Dyn 2018; 245:834-43. [PMID: 27239786 DOI: 10.1002/dvdy.24420] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/23/2016] [Accepted: 05/23/2016] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Mutations in Drosophila tumor suppressor genes (TSGs) lead to the formation of invasive tumors in the brain and imaginal discs. RESULTS Here we studied the tumorigenic properties of imaginal discs mutant for the TSG gene lethal giant larvae (lgl). lgl mutant cells display the characteristic features of mammalian tumor cells: they can proliferate indefinitely, induce additional tracheogenesis (an insect counterpart of vasculogenesis) and invade neighboring tissues. Lgl mutant tissues exhibit high apoptotic levels, which lead to the activation of the Jun-N-Terminal Kinase (JNK) pathway. We propose that JNK is a key factor in the acquisition of these tumorigenic properties; it promotes cell proliferation and induces high levels of Mmp1 and confers tumor cells capacity to invade wild-type tissue. Noteworthy, lgl RNAi-mediated down-regulation does not produce similar transformations in the central nervous system (CNS), thereby indicating a fundamental difference between the cells of developing imaginal discs and those of differentiated organs. We discuss these results in the light of the "single big-hit origin" of some human pediatric or developmental cancers. CONCLUSIONS Down-regulation of lgl in imaginal discs is sufficient to enhance tracheogenesis and to promote invasion and colonization of other larval structures including the CNS. Developmental Dynamics 245:834-843, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Ginés Morata
- Centro de Biología Molecular, CSIC-UAM, Madrid, Spain
| | - Jordi Casanova
- Institut de Biologia Molecular de Barcelona (CSIC) Barcelona, Catalonia, Spain.,Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology, Barcelona, Catalonia, Spain
| |
Collapse
|
27
|
Sollazzo M, Genchi C, Paglia S, Di Giacomo S, Pession A, de Biase D, Grifoni D. High MYC Levels Favour Multifocal Carcinogenesis. Front Genet 2018; 9:612. [PMID: 30619451 PMCID: PMC6297171 DOI: 10.3389/fgene.2018.00612] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 11/20/2018] [Indexed: 02/05/2023] Open
Abstract
The term "field cancerisation" describes the formation of tissue sub-areas highly susceptible to multifocal tumourigenesis. In the earlier stages of cancer, cells may indeed display a series of molecular alterations that allow them to proliferate faster, eventually occupying discrete tissue regions with irrelevant morphological anomalies. This behaviour recalls cell competition, a process based on a reciprocal fitness comparison: when cells with a growth advantage arise in a tissue, they are able to commit wild-type neighbours to death and to proliferate at their expense. It is known that cells expressing high MYC levels behave as super-competitors, able to kill and replace less performant adjacent cells; given MYC upregulation in most human cancers, MYC-mediated cell competition is likely to pioneer field cancerisation. Here we show that MYC overexpression in a sub-territory of the larval wing epithelium of Drosophila is sufficient to trigger a number of cellular responses specific to mammalian pre-malignant tissues. Moreover, following induction of different second mutations, high MYC-expressing epithelia were found to be susceptible to multifocal growth, a hallmark of mammalian pre-cancerous fields. In summary, our study identified an early molecular alteration implicated in field cancerisation and established a genetically amenable model which may help study the molecular basis of early carcinogenesis.
Collapse
|
28
|
Yang W, Zhou C, Luo M, Shi X, Li Y, Sun Z, Zhou F, Chen Z, He J. MiR-652-3p is upregulated in non-small cell lung cancer and promotes proliferation and metastasis by directly targeting Lgl1. Oncotarget 2017; 7:16703-15. [PMID: 26934648 PMCID: PMC4941345 DOI: 10.18632/oncotarget.7697] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Accepted: 02/06/2016] [Indexed: 12/17/2022] Open
Abstract
Our previous study found that miR-652-3p is markedly upregulated in the serum of patients with NSCLC and suggesting that miR-652-3p is a potential biomarker for the early diagnosis of NSCLC. In this study, we detected the expression of miR-652-3p in NSCLC tumor tissues and cell lines and investigated the effect of miR-652-3p on the proliferation and metastasis of NSCLC cells. Our results showed that the expression of miR-652-3p was significantly upregulated in tumor tissues of 50 patients with NSCLC, and it was significantly higher in patients with positive lymph node metastasis, advanced TNM stage and poor prognosis. Using functional analyses by overexpressing or suppressing miR-652-3p in NSCLC cells, we demonstrated that miR-652-3p promoted cell proliferation, migration, invasion and inhibited cell apoptosis. Moreover, the lethal(2) giant larvae 1 (Lgl1) was identified as a direct and functional target of miR-652-3p. Overexpression or knockdown of miR-652-3p led to decreased or increased expression of Lgl1 protein, and the binding site mutation of LLGL1 3'UTR abrogated the responsiveness of the luciferase reporters to miR-652-3p. Overexpression of Lgl1 partially attenuated the function of miR-652-3p. Collectively, these results revealed that miR-652-3p execute a tumor-promoter function in NSCLC through direct binding and regulating the expression of Lgl1.
Collapse
Affiliation(s)
- Wenhui Yang
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Chengcheng Zhou
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Mei Luo
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Xuejiao Shi
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Yuan Li
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zengmiao Sun
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Fang Zhou
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Zhaoli Chen
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| | - Jie He
- Department of Thoracic Surgery, Cancer Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100021, China
| |
Collapse
|
29
|
Di Giacomo S, Sollazzo M, de Biase D, Ragazzi M, Bellosta P, Pession A, Grifoni D. Human Cancer Cells Signal Their Competitive Fitness Through MYC Activity. Sci Rep 2017; 7:12568. [PMID: 28974715 PMCID: PMC5626713 DOI: 10.1038/s41598-017-13002-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 09/13/2017] [Indexed: 02/08/2023] Open
Abstract
MYC-mediated cell competition is a cell-cell interaction mechanism known to play an evolutionary role during development from Drosophila to mammals. Cells expressing low levels of MYC, called losers, are committed to die by nearby cells with high MYC activity, called winners, that overproliferate to compensate for cell loss, so that the fittest cells be selected for organ formation. Given MYC's consolidated role in oncogenesis, cell competition is supposed to be relevant to cancer, but its significance in human malignant contexts is largely uncharacterised. Here we show stereotypical patterns of MYC-mediated cell competition in human cancers: MYC-upregulating cells and apoptotic cells were indeed repeatedly found at the tumour-stroma interface and within the tumour parenchyma. Cell death amount in the stromal compartment and MYC protein level in the tumour were highly correlated regardless of tumour type and stage. Moreover, we show that MYC modulation in heterotypic co-cultures of human cancer cells is sufficient as to subvert their competitive state, regardless of genetic heterogeneity. Altogether, our findings suggest that the innate role of MYC-mediated cell competition in development is conserved in human cancer, with malignant cells using MYC activity to colonise the organ at the expense of less performant neighbours.
Collapse
Affiliation(s)
- Simone Di Giacomo
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, Bologna, 40126, Italy.
| | - Manuela Sollazzo
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, Bologna, 40126, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, Bologna, 40126, Italy
| | - Moira Ragazzi
- Pathology Unit, IRCCS Arcispedale Santa Maria Nuova, Via Amendola 2, 42122, Reggio Emilia, Italy
| | - Paola Bellosta
- Center for Integrate Biology (CIBIO), University of Trento, Via Sommarive 9, Povo, (TN), 38123, Italy
| | - Annalisa Pession
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, Bologna, 40126, Italy
| | - Daniela Grifoni
- Department of Pharmacy and Biotechnology, University of Bologna, Via Selmi 3, Bologna, 40126, Italy.
| |
Collapse
|
30
|
Klunder LJ, Faber KN, Dijkstra G, van IJzendoorn SCD. Mechanisms of Cell Polarity-Controlled Epithelial Homeostasis and Immunity in the Intestine. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027888. [PMID: 28213466 DOI: 10.1101/cshperspect.a027888] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Intestinal epithelial cell polarity is instrumental to maintain epithelial homeostasis and balance communications between the gut lumen and bodily tissue, thereby controlling the defense against gastrointestinal pathogens and maintenance of immune tolerance to commensal bacteria. In this review, we highlight recent advances with regard to the molecular mechanisms of cell polarity-controlled epithelial homeostasis and immunity in the human intestine.
Collapse
Affiliation(s)
- Leon J Klunder
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Klaas Nico Faber
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Gerard Dijkstra
- Department of Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| | - Sven C D van IJzendoorn
- Department of Cell Biology, University Medical Center Groningen, University of Groningen, 9713 GZ Groningen, the Netherlands
| |
Collapse
|
31
|
Paglia S, Sollazzo M, Di Giacomo S, de Biase D, Pession A, Grifoni D. Failure of the PTEN/aPKC/Lgl Axis Primes Formation of Adult Brain Tumours in Drosophila. BIOMED RESEARCH INTERNATIONAL 2017; 2017:2690187. [PMID: 29445734 PMCID: PMC5763105 DOI: 10.1155/2017/2690187] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/02/2017] [Accepted: 11/08/2017] [Indexed: 02/05/2023]
Abstract
Different regions in the mammalian adult brain contain immature precursors, reinforcing the concept that brain cancers, such as glioblastoma multiforme (GBM), may originate from cells endowed with stem-like properties. Alterations of the tumour suppressor gene PTEN are very common in primary GBMs. Very recently, PTEN loss was shown to undermine a specific molecular axis, whose failure is associated with the maintenance of the GBM stem cells in mammals. This axis is composed of PTEN, aPKC, and the polarity determinant Lethal giant larvae (Lgl): PTEN loss promotes aPKC activation through the PI3K pathway, which in turn leads to Lgl inhibition, ultimately preventing stem cell differentiation. To find the neural precursors responding to perturbations of this molecular axis, we targeted different neurogenic regions of the Drosophila brain. Here we show that PTEN mutation impacts aPKC and Lgl protein levels also in Drosophila. Moreover, we demonstrate that PI3K activation is not sufficient to trigger tumourigenesis, while aPKC promotes hyperplastic growth of the neuroepithelium and a noticeable expansion of the type II neuroblasts. Finally, we show that these neuroblasts form invasive tumours that persist and keep growing in the adult, leading the affected animals to untimely death, thus displaying frankly malignant behaviours.
Collapse
Affiliation(s)
- Simona Paglia
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Manuela Sollazzo
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Simone Di Giacomo
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Dario de Biase
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Annalisa Pession
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| | - Daniela Grifoni
- Department of “Pharmacy and Biotechnology”, University of Bologna, Via Selmi 3, 40126 Bologna, Italy
| |
Collapse
|
32
|
Li Z, Zhang T, Lin Z, Hou C, Zhang J, Men Y, Li H, Gao J. Lgl1 Is Required for Olfaction and Development of Olfactory Bulb in Mice. PLoS One 2016; 11:e0162126. [PMID: 27603780 PMCID: PMC5014313 DOI: 10.1371/journal.pone.0162126] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/17/2016] [Indexed: 11/27/2022] Open
Abstract
Lethal giant larvae 1 (Lgl1) was initially identified as a tumor suppressor in Drosophila and functioned as a key regulator of epithelial polarity and asymmetric cell division. In this study, we generated Lgl1 conditional knockout mice mediated by Pax2-Cre, which is expressed in olfactory bulb (OB). Next, we examined the effects of Lgl1 loss in the OB. First, we determined the expression patterns of Lgl1 in the neurogenic regions of the embryonic dorsal region of the LGE (dLGE) and postnatal OB. Furthermore, the Lgl1 conditional mutants exhibited abnormal morphological characteristics of the OB. Our behavioral analysis exhibited greatly impaired olfaction in Lgl1 mutant mice. To elucidate the possible mechanisms of impaired olfaction in Lgl1 mutant mice, we investigated the development of the OB. Interestingly, reduced thickness of the MCL and decreased density of mitral cells (MCs) were observed in Lgl1 mutant mice. Additionally, we observed a dramatic loss in SP8+ interneurons (e.g. calretinin and GABAergic/non-dopaminergic interneurons) in the GL of the OB. Our results demonstrate that Lgl1 is required for the development of the OB and the deletion of Lgl1 results in impaired olfaction in mice.
Collapse
Affiliation(s)
- Zhenzu Li
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Tingting Zhang
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Zhuchun Lin
- Jinan First People's Hospital, Jinan, 250011, Shandong, China
| | - Congzhe Hou
- The Second Hospital of Shandong University, Jinan, 250000, Shandong, China
| | - Jian Zhang
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Yuqin Men
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| | - Huashun Li
- SARITEX Center for Stem Cell, Engineering Translational Medicine, Shanghai East Hospital, Advanced Institute of Translational Medicine, Tongji University School of Medicine, Shanghai, 200123, China.,Center for Stem Cell&Nano-Medicine, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 200123, China.,Shenzhen Key Laboratory for Molecular Biology of Neural Development, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518000, Guangdong, China
| | - Jiangang Gao
- Institute of Developmental Biology, School of Life Science, Shandong University, Jinan, 250100, Shandong, China
| |
Collapse
|
33
|
Abstract
Cancer is a complex disease that affects multiple organs. Whole-body animal models provide important insights into oncology that can lead to clinical impact. Here, we review novel concepts that Drosophila studies have established for cancer biology, drug discovery, and patient therapy. Genetic studies using Drosophila have explored the roles of oncogenes and tumor-suppressor genes that when dysregulated promote cancer formation, making Drosophila a useful model to study multiple aspects of transformation. Not limited to mechanism analyses, Drosophila has recently been showing its value in facilitating drug development. Flies offer rapid, efficient platforms by which novel classes of drugs can be identified as candidate anticancer leads. Further, we discuss the use of Drosophila as a platform to develop therapies for individual patients by modeling the tumor's genetic complexity. Drosophila provides both a classical and a novel tool to identify new therapeutics, complementing other more traditional cancer tools.
Collapse
Affiliation(s)
- M Sonoshita
- Icahn School of Medicine at Mount Sinai, New York, NY, United States; Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - R L Cagan
- Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| |
Collapse
|
34
|
Yadav AK, Srikrishna S, Gupta SC. Cancer Drug Development Using Drosophila as an in vivo Tool: From Bedside to Bench and Back. Trends Pharmacol Sci 2016; 37:789-806. [PMID: 27298020 DOI: 10.1016/j.tips.2016.05.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 12/14/2022]
Abstract
The fruit fly Drosophila melanogaster has been used for modeling cancer and as an in vivo tool for the validation and/or development of cancer therapeutics. The impetus for the use of Drosophila in cancer research stems from the high conservation of its signaling pathways, lower genetic redundancy, short life cycle, genetic amenability, and ease of maintenance. Several cell signaling pathways in Drosophila have been used for cancer drug development. The efficacy of combination therapy and uptake/bioavailability of drugs have also been studied. Drosophila has been validated using several FDA-approved drugs, suggesting a potential application of this model in drug repurposing. The model is emerging as a powerful tool for high-throughput screening and should significantly reduce the cost and time associated with drug development. In this review we discuss the applications of Drosophila in cancer drug development. The advantages and limitations of the model are discussed.
Collapse
Affiliation(s)
- Amarish Kumar Yadav
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India
| | - Saripella Srikrishna
- Cancer and Neurobiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India.
| | - Subash Chandra Gupta
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi 221 005, India.
| |
Collapse
|
35
|
Gandalovičová A, Vomastek T, Rosel D, Brábek J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget 2016; 7:25022-49. [PMID: 26872368 PMCID: PMC5041887 DOI: 10.18632/oncotarget.7214] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Apico-basal polarity is typical of cells present in differentiated epithelium while front-rear polarity develops in motile cells. In cancer development, the transition from epithelial to migratory polarity may be seen as the hallmark of cancer progression to an invasive and metastatic disease. Despite the morphological and functional dissimilarity, both epithelial and migratory polarity are controlled by a common set of polarity complexes Par, Scribble and Crumbs, phosphoinositides, and small Rho GTPases Rac, Rho and Cdc42. In epithelial tissues, their mutual interplay ensures apico-basal and planar cell polarity. Accordingly, altered functions of these polarity determinants lead to disrupted cell-cell adhesions, cytoskeleton rearrangements and overall loss of epithelial homeostasis. Polarity proteins are further engaged in diverse interactions that promote the establishment of front-rear polarity, and they help cancer cells to adopt different invasion modes. Invading cancer cells can employ either the collective, mesenchymal or amoeboid invasion modes or actively switch between them and gain intermediate phenotypes. Elucidation of the role of polarity proteins during these invasion modes and the associated transitions is a necessary step towards understanding the complex problem of metastasis. In this review we summarize the current knowledge of the role of cell polarity signaling in the plasticity of cancer cell invasiveness.
Collapse
Affiliation(s)
- Aneta Gandalovičová
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Tomáš Vomastek
- Institute of Microbiology, Academy of Sciences of The Czech Republic, Videňská, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| |
Collapse
|
36
|
Dong W, Zhang X, Liu W, Chen YJ, Huang J, Austin E, Celotto AM, Jiang WZ, Palladino MJ, Jiang Y, Hammond GRV, Hong Y. A conserved polybasic domain mediates plasma membrane targeting of Lgl and its regulation by hypoxia. J Cell Biol 2015; 211:273-86. [PMID: 26483556 PMCID: PMC4621827 DOI: 10.1083/jcb.201503067] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/02/2015] [Indexed: 01/01/2023] Open
Abstract
The plasma membrane targeting of Lgl, a key polarity and tumor suppressor protein, is mediated by electrostatic interactions between a polybasic motif in Lgl and phospholipids on the plasma membrane, and this mechanism is regulated by hypoxia and aPKC-phosphorylation. Lethal giant larvae (Lgl) plays essential and conserved functions in regulating both cell polarity and tumorigenesis in Drosophila melanogaster and vertebrates. It is well recognized that plasma membrane (PM) or cell cortex localization is crucial for Lgl function in vivo, but its membrane-targeting mechanisms remain poorly understood. Here, we discovered that hypoxia acutely and reversibly inhibits Lgl PM targeting through a posttranslational mechanism that is independent of the well-characterized atypical protein kinase C (aPKC) or Aurora kinase–mediated phosphorylations. Instead, we identified an evolutionarily conserved polybasic (PB) domain that targets Lgl to the PM via electrostatic binding to membrane phosphatidylinositol phosphates. Such PB domain–mediated PM targeting is inhibited by hypoxia, which reduces inositol phospholipid levels on the PM through adenosine triphosphate depletion. Moreover, Lgl PB domain contains all the identified phosphorylation sites of aPKC and Aurora kinases, providing a molecular mechanism by which phosphorylations neutralize the positive charges on the PB domain to inhibit Lgl PM targeting.
Collapse
Affiliation(s)
- Wei Dong
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Xuejing Zhang
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Weijie Liu
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Yi-jiun Chen
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Juan Huang
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261 Nanjing Medical University, Nanjing 210029, China
| | - Erin Austin
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Alicia M Celotto
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Wendy Z Jiang
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Michael J Palladino
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Yu Jiang
- Department of Pharmacology & Chemical Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Gerald R V Hammond
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| | - Yang Hong
- Department of Cell Biology, University of Pittsburgh Medical School, Pittsburgh, PA 15261
| |
Collapse
|
37
|
Liu X, Lu D, Ma P, Liu H, Cao Y, Sang B, Zhu X, Shi Q, Hu J, Yu R, Zhou X. Hugl-1 inhibits glioma cell growth in intracranial model. J Neurooncol 2015; 125:113-21. [PMID: 26341367 DOI: 10.1007/s11060-015-1901-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 08/29/2015] [Indexed: 12/28/2022]
Abstract
Drosophila lethal (2) giant larvae (lgl) has been reported as a tumor suppressor and could regulate the Drosophila hippo signaling. Human giant larvae-1(Hugl-1), one human homologue of Drosophila lgl, also has been reported to be involved in the development of some human cancers. However, whether Hugl-1 is associated with the pathogenesis of malignant gliomas remains poorly understood. In the present work, we examined the effect of Hugl-1 on glioma cell growth both in vitro and in vivo. Firstly, we found that Hugl-1 protein levels decreased in the human glioma tissues, suggesting that Hugl-1 is involved in glioma progression. Unfortunately, either stably or transiently over-expressing Hugl-1 did not affect glioma cell proliferation in vitro. In addition, Hugl-1 over-expression did not regulate hippo signaling pathway. Interestingly, over-expression of Hugl-1 not only inhibited gliomagenesis but also markedly inhibited cell proliferation and promoted the apoptosis of U251 cells in an orthotopic model of nude mice. Taken together, this study provides the evidence that Hugl-1 inhibits glioma cell growth in intracranial model of nude mice, suggesting that Hugl-1 might be a potential tumor target for glioma therapy.
Collapse
Affiliation(s)
- Xuejiao Liu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Dong Lu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Peng Ma
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Huaqiang Liu
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Yuewen Cao
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Ben Sang
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Xianlong Zhu
- The Graduate School, Xuzhou Medical College, Xuzhou, Jiangsu, People's Republic of China
| | - Qiong Shi
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jinxia Hu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical College, 84 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
- Brain Hospital, Affiliated Hospital of Xuzhou Medical College, 99 West Huai-hai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
38
|
Ravid S. The tumor suppressor Lgl1 regulates front-rear polarity of migrating cells. Cell Adh Migr 2015; 8:378-83. [PMID: 25482644 DOI: 10.4161/cam.29387] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Cell migration is a highly integrated, multistep process that plays an important role in physiological and pathological processes. The migrating cell is highly polarized, with complex regulatory pathways that integrate its component processes spatially and temporally. The Drosophila tumor suppressor, Lethal (2) giant larvae (Lgl), regulates apical-basal polarity in epithelia and asymmetric cell division. But little is known about the role of Lgl in establishing cell polarity in migrating cells. Recently, we showed that the mammalian Lgl1 interacts directly with non-muscle myosin IIA (NMIIA), inhibiting its ability to assemble into filaments in vitro. Lgl1 also regulates the cellular localization of NMIIA, the maturation of focal adhesions, and cell migration. We further showed that phosphorylation of Lgl1 by aPKCζ prevents its interaction with NMIIA and is important for Lgl1 and acto-NMII cytoskeleton cellular organization. Lgl is a critical downstream target of the Par6-aPKC cell polarity complex; we showed that Lgl1 forms two distinct complexes in vivo, Lgl1-NMIIA and Lgl1-Par6-aPKCζ in different cellular compartments. We further showed that aPKCζ and NMIIA compete to bind directly to Lgl1 through the same domain. These data provide new insights into the role of Lgl1, NMIIA, and Par6-aPKCζ in establishing front-rear polarity in migrating cells. In this commentary, I discuss the role of Lgl1 in the regulation of the acto-NMII cytoskeleton and its regulation by the Par6-aPKCζ polarity complex, and how Lgl1 activity may contribute to the establishment of front-rear polarity in migrating cells.
Collapse
Affiliation(s)
- Shoshana Ravid
- a Department of Biochemistry and Molecular Biology; The Institute of Medical Research Israel-Canada ; The Hebrew University-Hadassah Medical School ; Jerusalem , Israel
| |
Collapse
|
39
|
Yamashita K, Ide M, Furukawa KT, Suzuki A, Hirano H, Ohno S. Tumor suppressor protein Lgl mediates G1 cell cycle arrest at high cell density by forming an Lgl-VprBP-DDB1 complex. Mol Biol Cell 2015; 26:2426-38. [PMID: 25947136 PMCID: PMC4571298 DOI: 10.1091/mbc.e14-10-1462] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Accepted: 04/28/2015] [Indexed: 11/25/2022] Open
Abstract
Lgl is a conserved tumor suppressor suggested to be involved in cell polarity regulation and suppression of cell proliferation. Lgl inhibits formation of the VprBP-DDB1-Cul4A-Roc1 ubiquitin E3 ligase complex, which is implicated in cell cycle progression, by promoting formation of the Lgl-VprBP-DDB1 complex to prevent overproliferation. Lethal giant larvae (Lgl) is an evolutionarily conserved tumor suppressor whose loss of function causes disrupted epithelial architecture with enhanced cell proliferation and defects in cell polarity. A role for Lgl in the establishment and maintenance of cell polarity via suppression of the PAR-aPKC polarity complex is established; however, the mechanism by which Lgl regulates cell proliferation is not fully understood. Here we show that depletion of Lgl1 and Lgl2 in MDCK epithelial cells results in overproliferation and overproduction of Lgl2 causes G1 arrest. We also show that Lgl associates with the VprBP-DDB1 complex independently of the PAR-aPKC complex and prevents the VprBP-DDB1 subunits from binding to Cul4A, a central component of the CRL4 [VprBP] ubiquitin E3 ligase complex implicated in G1- to S-phase progression. Consistently, depletion of VprBP or Cul4 rescues the overproliferation of Lgl-depleted cells. In addition, the affinity between Lgl2 and the VprBP-DDB1 complex increases at high cell density. Further, aPKC-mediated phosphorylation of Lgl2 negatively regulates the interaction between Lgl2 and VprBP-DDB1 complex. These results suggest a mechanism protecting overproliferation of epithelial cells in which Lgl plays a critical role by inhibiting formation of the CRL4 [VprBP] complex, resulting in G1 arrest.
Collapse
Affiliation(s)
- Kazunari Yamashita
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama 236-0004, Japan
| | - Mariko Ide
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama 236-0004, Japan
| | - Kana T Furukawa
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama 236-0004, Japan
| | - Atsushi Suzuki
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama 236-0004, Japan Molecular Cellular Biology Laboratory, Graduate School of Medical Life Science, Yokohama City University, Yokohama 230-0045, Japan
| | - Hisashi Hirano
- Supramolecular Biology, International Graduate School of Arts and Sciences, Yokohama City University, Yokohama 230-0045, Japan
| | - Shigeo Ohno
- Department of Molecular Biology, Graduate School of Medical Science, Yokohama City University, Yokohama 236-0004, Japan
| |
Collapse
|
40
|
Winkler IS, Blaschke JD, Davis DJ, Stireman JO, O'Hara JE, Cerretti P, Moulton JK. Explosive radiation or uninformative genes? Origin and early diversification of tachinid flies (Diptera: Tachinidae). Mol Phylogenet Evol 2015; 88:38-54. [PMID: 25841383 DOI: 10.1016/j.ympev.2015.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 03/20/2015] [Accepted: 03/25/2015] [Indexed: 12/01/2022]
Abstract
Molecular phylogenetic studies at all taxonomic levels often infer rapid radiation events based on short, poorly resolved internodes. While such rapid episodes of diversification are an important and widespread evolutionary phenomenon, much of this poor phylogenetic resolution may be attributed to the continuing widespread use of "traditional" markers (mitochondrial, ribosomal, and some nuclear protein-coding genes) that are often poorly suited to resolve difficult, higher-level phylogenetic problems. Here we reconstruct phylogenetic relationships among a representative set of taxa of the parasitoid fly family Tachinidae and related outgroups of the superfamily Oestroidea. The Tachinidae are one of the most species rich, yet evolutionarily recent families of Diptera, providing an ideal case study for examining the differential performance of loci in resolving phylogenetic relationships and the benefits of adding more loci to phylogenetic analyses. We assess the phylogenetic utility of nine genes including both traditional genes (e.g., CO1 mtDNA, 28S rDNA) and nuclear protein-coding genes newly developed for phylogenetic analysis. Our phylogenetic findings, based on a limited set of taxa, include: a close relationship between Tachinidae and the calliphorid subfamily Polleninae, monophyly of Tachinidae and the subfamilies Exoristinae and Dexiinae, subfamily groupings of Dexiinae+Phasiinae and Tachininae+Exoristinae, and robust phylogenetic placement of the somewhat enigmatic genera Strongygaster, Euthera, and Ceracia. In contrast to poor resolution and phylogenetic incongruence of "traditional genes," we find that a more selective set of highly informative genes is able to more precisely identify regions of the phylogeny that experienced rapid radiation of lineages, while more accurately depicting their phylogenetic context. Although much expanded taxon sampling is necessary to effectively assess the monophyly of and relationships among major tachinid lineages and their relatives, we show that a small number of well-chosen nuclear protein-coding genes can successfully resolve even difficult phylogenetic problems.
Collapse
Affiliation(s)
- Isaac S Winkler
- Department of Biological Sciences, Wright State University, Dayton, OH 45435, USA; Department of Biology, Linfield College, McMinnville, OR 97128, USA
| | - Jeremy D Blaschke
- Department of Entomology and Plant Pathology, University of Tennessee, Knoxville, TN 37996, USA
| | - Daniel J Davis
- Department of Biological Sciences, Wright State University, Dayton, OH 45435, USA
| | - John O Stireman
- Department of Biological Sciences, Wright State University, Dayton, OH 45435, USA.
| | - James E O'Hara
- Canadian National Collection of Insects, Agriculture and Agri-Food Canada, 960 Carling Avenue, Ottawa, Ontario K1A 0C6, Canada
| | - Pierfilippo Cerretti
- DAFNAE-Entomology, Università degli Studi di Padova, Viale dell'Università 16, 35020 Legnaro (Padova), Italy; Dipartimento di Biologia e Biotecnologie 'Charles Darwin', 'Sapienza' Università di Roma, Piazzale A. Moro 5, 00185 Rome, Italy
| | - John K Moulton
- Department of Entomology and Plant Pathology, University of Tennessee, Knoxville, TN 37996, USA
| |
Collapse
|
41
|
Lin WH, Asmann YW, Anastasiadis PZ. Expression of polarity genes in human cancer. Cancer Inform 2015; 14:15-28. [PMID: 25991909 PMCID: PMC4390136 DOI: 10.4137/cin.s18964] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/10/2015] [Accepted: 02/12/2015] [Indexed: 01/01/2023] Open
Abstract
Polarity protein complexes are crucial for epithelial apical–basal polarity and directed cell migration. Since alterations of these processes are common in cancer, polarity proteins have been proposed to function as tumor suppressors or oncogenic promoters. Here, we review the current understanding of polarity protein functions in epithelial homeostasis, as well as tumor formation and progression. As most previous studies focused on the function of single polarity proteins in simplified model systems, we used a genomics approach to systematically examine and identify the expression profiles of polarity genes in human cancer. The expression profiles of polarity genes were distinct in different human tissues and classified cancer types. Additionally, polarity expression profiles correlated with disease progression and aggressiveness, as well as with identified cancer types, where specific polarity genes were commonly altered. In the case of Scribble, gene expression analysis indicated its common amplification and upregulation in human cancer, suggesting a tumor promoting function.
Collapse
Affiliation(s)
- Wan-Hsin Lin
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Yan W Asmann
- Department of Health Sciences Research, Mayo Clinic, Jacksonville, FL, USA
| | | |
Collapse
|
42
|
Grifoni D, Sollazzo M, Fontana E, Froldi F, Pession A. Multiple strategies of oxygen supply in Drosophila malignancies identify tracheogenesis as a novel cancer hallmark. Sci Rep 2015; 5:9061. [PMID: 25762498 PMCID: PMC4357021 DOI: 10.1038/srep09061] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 02/16/2015] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis is the term used to describe all the alterations in blood vessel growth induced by a tumour mass following hypoxic stress. The occurrence of multiple strategies of vessel recruitment favours drug resistance, greatly complicating the treatment of certain tumours. In Drosophila, oxygen is conveyed to the internal organs by the tracheal system, a closed tubular network whose role in cancer growth is so far unexplored. We found that, as observed in human cancers, Drosophila malignant cells suffer from oxygen shortage, release pro-tracheogenic factors, co-opt nearby vessels and get incorporated into the tracheal walls. We also found that the parallelisms observed in cellular behaviours are supported by genetic and molecular conservation. Finally, we identified a molecular circuitry associated with the differentiation of cancer cells into tracheal cells. In summary, our findings identify tracheogenesis as a novel cancer hallmark in Drosophila, further expanding the power of the fly model in cancer research.
Collapse
Affiliation(s)
- Daniela Grifoni
- Department of "Farmacia e Biotecnologie", University of Bologna, Bologna, Italy
| | - Manuela Sollazzo
- Department of "Farmacia e Biotecnologie", University of Bologna, Bologna, Italy
| | - Elisabetta Fontana
- Department of "Farmacia e Biotecnologie", University of Bologna, Bologna, Italy
| | - Francesca Froldi
- Department of "Farmacia e Biotecnologie", University of Bologna, Bologna, Italy
| | - Annalisa Pession
- Department of "Farmacia e Biotecnologie", University of Bologna, Bologna, Italy
| |
Collapse
|
43
|
Cao F, Miao Y, Xu K, Liu P. Lethal (2) giant larvae: an indispensable regulator of cell polarity and cancer development. Int J Biol Sci 2015; 11:380-9. [PMID: 25798058 PMCID: PMC4366637 DOI: 10.7150/ijbs.11243] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 01/21/2015] [Indexed: 01/04/2023] Open
Abstract
Cell polarity is one of the most basic properties of all normal cells and is essential for regulating numerous biological processes. Loss of polarity is considered a hallmark for cancer. Multiple polarity proteins are implicated in maintenance of cell polarity. Lethal (2) giant larvae (Lgl) is one of polarity proteins that plays an important role in regulating cell polarity, asymmetric division as well as tumorigenesis. Lgl proteins in different species have similar structures and conserved functions. Lgl acts as an indispensable regulator of cell biological function, including cell polarity and asymmetric division, through interplaying with other polarity proteins, regulating exocytosis, mediating cytoskeleton and being involved in signaling pathways. Furthermore, Lgl plays a role of a tumor suppressor, and the aberrant expression of Hugl, a human homologue of Lgl, contributes to multiple cancers. However, the exact functions of Lgl and the underlying mechanisms remain enigmatic. In this review, we will give an overview of the Lgl functions in cell polarity and cancer development, discuss the potential mechanisms underlying these functions, and raise our conclusion of previous studies and points of view about the future studies.
Collapse
Affiliation(s)
- Fang Cao
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Yi Miao
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Kedong Xu
- 2. Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| | - Peijun Liu
- 1. Center for Translational Medicine, The First Affiliated Hospital of Xian Jiaotong University, College of Medicine, Xi'an, China
| |
Collapse
|
44
|
Pelissier-Rota MA, Chartier NT, Jacquier-Sarlin MR. Dynamic Regulation of Adherens Junctions: Implication in Cell Differentiation and Tumor Development. INTERCELLULAR COMMUNICATION IN CANCER 2015:53-149. [DOI: 10.1007/978-94-017-7380-5_4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
45
|
Nam KH, Kim MA, Choe G, Kim WH, Lee HS. Deregulation of the cell polarity protein Lethal giant larvae 2 (Lgl2) correlates with gastric cancer progression. Gastric Cancer 2014; 17:610-20. [PMID: 24337435 DOI: 10.1007/s10120-013-0324-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2013] [Accepted: 11/24/2013] [Indexed: 02/07/2023]
Abstract
BACKGROUND We investigated the roles of Lethal giant larvae 2 (Lgl2), an epithelial cell polarity protein, during gastric carcinogenesis and gastric cancer (GC) progression and evaluated the correlation of Lgl2 with epithelial-mesenchymal transition (EMT) markers. METHODS Lgl2 protein and mRNA expression were determined by immunohistochemistry and mRNA in situ hybridization in a large series of GC and preneoplastic lesions. Additionally, expression of 7 EMT markers was examined by immunohistochemistry. RESULTS Loss of membrane Lgl2 staining in GC was observed in 347 of 409 GCs. Lgl2 loss was associated with diffuse histological type (P < 0.001), advanced stage (P = 0.021), and worse prognosis (P = 0.047). Furthermore, Lgl2 loss correlated with reduced E-cadherin expression (P < 0.01) and increased expression of vimentin (P < 0.01). Combined analysis of Lgl2 and the EMT markers, S100A4 and MMP2, improved predictions of patient outcomes. During gastric carcinogenesis, membrane expression of Lgl2 was progressively lost in 4 % of normal mucosa, 75 % of intestinal metaplasia, 58 % of gastric dysplasia, 69 % of intestinal type GC, and 96 % of diffuse type GC. CONCLUSIONS Our results suggest that Lgl2 loss occurs at an early stage of gastric carcinogenesis and contributes to GC progression.
Collapse
Affiliation(s)
- Kyung Han Nam
- Department of Pathology, Seoul National University Bundang Hospital, 173-82 Gumi-ro, Bundang-gu, Seongnam-si, Gyeonggi-do, 463-707, Korea
| | | | | | | | | |
Collapse
|
46
|
Abstract
Apico-basal polarity is a cardinal molecular feature of adult eukaryotic epithelial cells and appears to be involved in several key cellular processes including polarized cell migration and maintenance of tissue architecture. Epithelial cell polarity is maintained by three well-conserved polarity complexes, namely, PAR, Crumbs and SCRIB. The location and interaction between the components of these complexes defines distinct structural domains of epithelial cells. Establishment and maintenance of apico-basal polarity is regulated through various conserved cell signalling pathways including TGF beta, Integrin and WNT signalling. Loss of cell polarity is a hallmark for carcinoma, and its underlying molecular mechanism is beginning to emerge from studies on model organisms and cancer cell lines. Moreover, deregulated expression of apico-basal polarity complex components has been reported in human tumours. In this review, we provide an overview of the apico-basal polarity complexes and their regulation, their role in cell migration, and finally their involvement in carcinogenesis.
Collapse
Affiliation(s)
- Mohammed Khursheed
- Centre for DNA Fingerprinting and Diagnostics (CDFD), Nampally, Hyderabad 500 001, India
| | | |
Collapse
|
47
|
Das SS, Nanda GG, Alone DP. Artemisinin and curcumin inhibit Drosophila brain tumor, prolong life span, and restore locomotor activity. IUBMB Life 2014; 66:496-506. [PMID: 24975030 DOI: 10.1002/iub.1284] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 06/11/2014] [Accepted: 06/11/2014] [Indexed: 11/10/2022]
Abstract
Deletion of tumor suppressor gene, lethal(2)giant larvae [l(2)gl], leads to brain tumor in Drosophila melanogaster at larval stage of development and severe brain dysplasia in mice. We have studied the effect of two potential antitumor drugs artemisinin and curcumin in the perspective of inhibiting l(2)gl brain tumor. Efficacies of these drugs are characterized morphologically by measuring brain sizes of untreated and treated larvae on the basis of tumor inhibition and anatomically by looking at the cellular patterning via antibody staining of the third instar Drosophila larval brains. Behavioral experiments were done in form of locomotion to correlate tumor inhibition with the revival of brain function and longevity assays to assess general health span. It was observed that both drugs show antitumor properties individually and in combination when larvae were treated with these drugs. We also found evidence for reactive oxygen species-mediated action of these drugs. Both the drugs when treated individually or together show better median life span and locomotory response. Although the efficacies of various treatments varied, overall, the positive effects of artemisinin and curcumin demonstrate a potential applicability of these drugs against brain tumor in higher organisms. It also paves a way for a simpler model system for screening such natural products for antitumor property.
Collapse
Affiliation(s)
- Sourajit Soumyaranjan Das
- School of Biological Sciences, National Institute of Science Education and Research (NISER), Bhubaneswar, Odisha, India
| | | | | |
Collapse
|
48
|
Gont A, Hanson JEL, Lavictoire SJ, Parolin DA, Daneshmand M, Restall IJ, Soucie M, Nicholas G, Woulfe J, Kassam A, Da Silva VF, Lorimer IAJ. PTEN loss represses glioblastoma tumor initiating cell differentiation via inactivation of Lgl1. Oncotarget 2014; 4:1266-79. [PMID: 23907540 PMCID: PMC3787156 DOI: 10.18632/oncotarget.1164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme is an aggressive and incurable type of brain tumor. A subset of undifferentiated glioblastoma cells, known as glioblastoma tumor initiating cells (GTICs), has an essential role in the malignancy of this disease and also appears to mediate resistance to radiation therapy and chemotherapy. GTICs retain the ability to differentiate into cells with reduced malignant potential, but the signaling pathways controlling differentiation are not fully understood at this time. PTEN loss is a very common in glioblastoma multiforme and leads to aberrant activation of the phosphoinositide 3-kinase pathway. Increased signalling through this pathway leads to activation of multiple protein kinases, including atypical protein kinase C. In Drosophila, active atypical protein kinase C has been shown to promote the self-renewal of neuroblasts, inhibiting their differentiation along a neuronal lineage. This effect is mediated by atypical protein kinase c-mediated phosphorylation and inactivation of Lgl, a protein that was first characterized as a tumour suppressor in Drosophila. The effects of the atypical protein kinase C/Lgl pathway on the differentiation status of GTICs, and its potential link to PTEN loss, have not been assessed previously. Here we show that PTEN loss leads to the phosphorylation and inactivation of Lgl by atypical protein kinase C in glioblastoma cells. Re-expression of PTEN in GTICs promoted their differentiation along a neuronal lineage. This effect was also seen when atypical protein kinase C was knocked down using RNA interference, and when a non-phosphorylatable, constitutively active form of Lgl was expressed in GTICs. Thus PTEN loss, acting via atypical protein kinase C activation and Lgl inactivation, helps to maintain GTICs in an undifferentiated state.
Collapse
Affiliation(s)
- Alexander Gont
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rewiring cell polarity signaling in cancer. Oncogene 2014; 34:939-50. [PMID: 24632617 DOI: 10.1038/onc.2014.59] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Revised: 02/07/2014] [Accepted: 02/11/2014] [Indexed: 02/08/2023]
Abstract
Disrupted cell polarity is a feature of epithelial cancers. The Crumbs, Par and Scribble polarity complexes function to specify and maintain apical and basolateral membrane domains, which are essential to organize intracellular signaling pathways that maintain epithelial homeostasis. Disruption of apical-basal polarity proteins facilitates rewiring of oncogene and tumor suppressor signaling pathways to deregulate proliferation, apoptosis, invasion and metastasis. Moreover, apical-basal polarity integrates intracellular signaling with the microenvironment by regulating metabolic signaling, extracellular matrix remodeling and tissue level organization. In this review, we discuss recent advances in our understanding of how polarity proteins regulate diverse signaling pathways throughout cancer progression from initiation to metastasis.
Collapse
|
50
|
Hawkins ED, Oliaro J, Ramsbottom KM, Ting SB, Sacirbegovic F, Harvey M, Kinwell T, Ghysdael J, Johnstone RW, Humbert PO, Russell SM. Lethal giant larvae 1 tumour suppressor activity is not conserved in models of mammalian T and B cell leukaemia. PLoS One 2014; 9:e87376. [PMID: 24475281 PMCID: PMC3903681 DOI: 10.1371/journal.pone.0087376] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2013] [Accepted: 12/24/2013] [Indexed: 01/04/2023] Open
Abstract
In epithelial and stem cells, lethal giant larvae (Lgl) is a potent tumour suppressor, a regulator of Notch signalling, and a mediator of cell fate via asymmetric cell division. Recent evidence suggests that the function of Lgl is conserved in mammalian haematopoietic stem cells and implies a contribution to haematological malignancies. To date, direct measurement of the effect of Lgl expression on malignancies of the haematopoietic lineage has not been tested. In Lgl1−/− mice, we analysed the development of haematopoietic malignancies either alone, or in the presence of common oncogenic lesions. We show that in the absence of Lgl1, production of mature white blood cell lineages and long-term survival of mice are not affected. Additionally, loss of Lgl1 does not alter leukaemia driven by constitutive Notch, c-Myc or Jak2 signalling. These results suggest that the role of Lgl1 in the haematopoietic lineage might be restricted to specific co-operating mutations and a limited number of cellular contexts.
Collapse
Affiliation(s)
- Edwin D. Hawkins
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- * E-mail: (EDH); (SMR)
| | - Jane Oliaro
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Kelly M. Ramsbottom
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Stephen B. Ting
- Stem Cell Research Group, Australian Centre for Blood Diseases, Monash University and Alfred Health, Melbourne, Victoria, Australia
| | - Faruk Sacirbegovic
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Michael Harvey
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Tanja Kinwell
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jacques Ghysdael
- Institut Curie, Centre Universitaire, Bat 110 91405, Orsay, France
- Centre National de la Recherche Scientifique UMR 3306, Orsay, France
- INSERM (Institut National de la Santé et de la Recherche Médicale) U1005, Orsay, France
| | - Ricky W. Johnstone
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Patrick O. Humbert
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Cell Cycle and Cancer Genetics Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sarah M. Russell
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
- Centre for Micro-Photonics, Swinburne University of Technology, Hawthorn, Victoria, Australia
- * E-mail: (EDH); (SMR)
| |
Collapse
|