1
|
Huang J, Luo S, Shen J, Lee M, Chen R, Ma S, Sun LQ, Li JJ. Cellular polarity pilots breast cancer progression and immunosuppression. Oncogene 2025; 44:783-793. [PMID: 40057606 PMCID: PMC11913746 DOI: 10.1038/s41388-025-03324-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 02/03/2025] [Accepted: 02/19/2025] [Indexed: 03/19/2025]
Abstract
Disrupted cellular polarity (DCP) is a hallmark of solid cancer, the malignant disease of epithelial tissues, which occupies ~90% of all human cancers. DCP has been identified to affect not only the cancer cell's aggressive behavior but also the migration and infiltration of immune cells, although the precise mechanism of DCP-affected tumor-immune cell interaction remains unclear. This review discusses immunosuppressive tumor microenvironments (TME) caused by DCP-driven tumor cell proliferation with DCP-impaired immune cell functions. We will revisit the fundamental roles of cell polarity (CP) proteins in sustaining mammary luminal homeostasis, epithelial transformation, and breast cancer progression. Then, the current data on CP involvement in immune cell activation, maturation, migration, and tumor infiltration are evaluated. The CP status on the immune effector cells and their targeted tumor cells are highlighted in tumor immune regulation, including the antigen presentation and the formation of immune synapses (IS). CP-regulated antigen presentation and delivery and the formation of IS between the immune cells, especially between the immune effectors and tumor cells, will be addressed. Alterations of CP on the tumor cells, infiltrated immune effector cells, or both are discussed with these aspects. We conclude that CP-mediated tumor aggressiveness coupled with DCP-impaired immune cell disability may decide the degree of immunosuppressive status and responsiveness to immune checkpoint blockade (ICB). Further elucidating the dynamics of CP- or DCP-mediated immune regulation in TME will provide more critical insights into tumor-immune cell dynamics, which is required to invent more effective approaches for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Huang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shufeng Luo
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha
| | - Juan Shen
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Maya Lee
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Rachel Chen
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Lun-Quan Sun
- Hunan Key Laboratory of Molecular Radiation Oncology, Xiangya Cancer Center, Central South University, China, Hunan, Changsha.
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, California, USA.
- NCI-designated Comprehensive Cancer Center, University of California Davis School of Medicine, Sacramento, California, USA.
| |
Collapse
|
2
|
Hong GL, Kim KH, Kim YJ, Lee HJ, Cho SP, Han SY, Yang SW, Lee JS, Kang SK, Lim JS, Jung JY. Novel role of LLGL2 silencing in autophagy: reversing epithelial-mesenchymal transition in prostate cancer. Biol Res 2024; 57:25. [PMID: 38720397 PMCID: PMC11077766 DOI: 10.1186/s40659-024-00499-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
PURPOSE Prostate cancer (PCa) is a major urological disease that is associated with significant morbidity and mortality in men. LLGL2 is the mammalian homolog of Lgl. It acts as a tumor suppressor in breast and hepatic cancer. However, the role of LLGL2 and the underlying mechanisms in PCa have not yet been elucidated. Here, we investigate the role of LLGL2 in the regulation of epithelial-mesenchymal transition (EMT) in PCa through autophagy in vitro and in vivo. METHODS PC3 cells were transfected with siLLGL2 or plasmid LLGL2 and autophagy was examined. Invasion, migration, and wound healing were assessed in PC3 cells under autophagy regulation. Tumor growth was evaluated using a shLLGL2 xenograft mouse model. RESULTS In patients with PCa, LLGL2 levels were higher with defective autophagy and increased EMT. Our results showed that the knockdown of LLGL2 induced autophagy flux by upregulating Vps34 and ATG14L. LLGL2 knockdown inhibits EMT by upregulating E-cadherin and downregulating fibronectin and α-SMA. The pharmacological activation of autophagy by rapamycin suppressed EMT, and these effects were reversed by 3-methyladenine treatment. Interestingly, in a shLLGL2 xenograft mouse model, tumor size and EMT were decreased, which were improved by autophagy induction and worsened by autophagy inhibition. CONCLUSION Defective expression of LLGL2 leads to attenuation of EMT due to the upregulation of autophagy flux in PCa. Our results suggest that LLGL2 is a novel target for alleviating PCa via the regulation of autophagy.
Collapse
Affiliation(s)
- Geum-Lan Hong
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Kyung-Hyun Kim
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Yae-Ji Kim
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Hui-Ju Lee
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Sung-Pil Cho
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Seung-Yun Han
- Department of Anatomy, College of Medicine, Konyang University, Daejeon, 35365, Republic of Korea
| | - Seung Woo Yang
- Department of Urology, College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Jong-Soo Lee
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea
| | - Shin-Kwang Kang
- Department of Thoracic and Cardiovascular Surgery, College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Jae-Sung Lim
- Department of Urology, College of Medicine, Chungnam National University Hospital, Daejeon, 35015, Republic of Korea
| | - Ju-Young Jung
- Department of Veterinary Medicine, Institute of Veterinary Science, College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yusung-gu, Daejeon, 34134, Republic of Korea.
| |
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: 1] [Impact Index Per Article: 1.0] [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
|
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
|
5
|
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
|
6
|
Pramanick D, Singh R, Roy B, Mazumdar A, Ghose S. “Loss of polarity of basal cells” – The term revisited in the light of genomics. J Oral Biol Craniofac Res 2022; 12:248-252. [DOI: 10.1016/j.jobcr.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 10/18/2022] Open
|
7
|
Ecovoiu AA, Ratiu AC, Micheu MM, Chifiriuc MC. Inter-Species Rescue of Mutant Phenotype-The Standard for Genetic Analysis of Human Genetic Disorders in Drosophila melanogaster Model. Int J Mol Sci 2022; 23:2613. [PMID: 35269756 PMCID: PMC8909942 DOI: 10.3390/ijms23052613] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/16/2022] Open
Abstract
Drosophila melanogaster (the fruit fly) is arguably a superstar of genetics, an astonishing versatile experimental model which fueled no less than six Nobel prizes in medicine. Nowadays, an evolving research endeavor is to simulate and investigate human genetic diseases in the powerful D. melanogaster platform. Such a translational experimental strategy is expected to allow scientists not only to understand the molecular mechanisms of the respective disorders but also to alleviate or even cure them. In this regard, functional gene orthology should be initially confirmed in vivo by transferring human or vertebrate orthologous transgenes in specific mutant backgrounds of D. melanogaster. If such a transgene rescues, at least partially, the mutant phenotype, then it qualifies as a strong candidate for modeling the respective genetic disorder in the fruit fly. Herein, we review various examples of inter-species rescue of relevant mutant phenotypes of the fruit fly and discuss how these results recommend several human genes as candidates to study and validate genetic variants associated with human diseases. We also consider that a wider implementation of this evolutionist exploratory approach as a standard for the medicine of genetic disorders would allow this particular field of human health to advance at a faster pace.
Collapse
Affiliation(s)
- Alexandru Al. Ecovoiu
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania;
| | - Attila Cristian Ratiu
- Department of Genetics, Faculty of Biology, University of Bucharest, 060101 Bucharest, Romania;
| | - Miruna Mihaela Micheu
- Department of Cardiology, Clinical Emergency Hospital of Bucharest, 014461 Bucharest, Romania;
| | - Mariana Carmen Chifiriuc
- The Research Institute of the University of Bucharest and Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania;
| |
Collapse
|
8
|
Ma R, Gong D, You H, Xu C, Lu Y, Bergers G, Werb Z, Klein OD, Petritsch CK, Lu P. LGL1 binds to Integrin β1 and inhibits downstream signaling to promote epithelial branching in the mammary gland. Cell Rep 2022; 38:110375. [PMID: 35172155 PMCID: PMC9113222 DOI: 10.1016/j.celrep.2022.110375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/08/2021] [Accepted: 01/20/2022] [Indexed: 11/29/2022] Open
Abstract
Branching morphogenesis is a fundamental process by which organs in invertebrates and vertebrates form branches to expand their surface areas. The current dogma holds that directional cell migration determines where a new branch forms and thus patterns branching. Here, we asked whether mouse Lgl1, a homolog of the Drosophila tumor suppressor Lgl, regulates epithelial polarity in the mammary gland. Surprisingly, mammary glands lacking Lgl1 have normal epithelial polarity, but they form fewer branches. Moreover, we find that Lgl1 null epithelium is unable to directionally migrate, suggesting that migration is not essential for mammary epithelial branching as expected. We show that LGL1 binds to Integrin β1 and inhibits its downstream signaling, and Integrin β1 overexpression blocks epithelial migration, thus recapitulating the Lgl1 null phenotype. Altogether, we demonstrate that Lgl1 modulation of Integrin β1 signaling is essential for directional migration and that epithelial branching in invertebrates and the mammary gland is fundamentally distinct. Ma et al. show that Lgl1 is essential for mammary gland branching morphogenesis but not epithelial polarity. Lgl1 is required for directional migration by regulating Integrin β1 signaling levels and focal adhesion strengths. Finally, branching mechanisms are distinct between mammary gland and Drosophila systems where directional migration is indispensable.
Collapse
Affiliation(s)
- Rongze Ma
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Difei Gong
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Huanyang You
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Chongshen Xu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Yunzhe Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Gabriele Bergers
- VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, 3000 Leuven, Belgium
| | - Zena Werb
- Department of Anatomy and Program in Developmental and Stem Cell Biology, University of California, San Francisco, San Francisco, CA 94143-0452, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, UCSF Box 0422, 513 Parnassus Avenue, HSE1508, San Francisco, CA 94143-0422, USA
| | - Claudia K Petritsch
- Department of Neurological Surgery, Stanford University, Palo Alto, CA 94305, USA
| | - Pengfei Lu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
9
|
Zhang Y, Yuan P, Ma X, Deng Q, Gao J, Yang J, Zhang T, Zhang C, Zhang W. Deletion of Smooth Muscle Lethal Giant Larvae 1 Promotes Neointimal Hyperplasia in Mice. Front Pharmacol 2022; 13:834296. [PMID: 35140622 PMCID: PMC8819082 DOI: 10.3389/fphar.2022.834296] [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: 12/13/2021] [Accepted: 01/06/2022] [Indexed: 12/01/2022] Open
Abstract
Vascular smooth muscle cell (VSMC) proliferation and migration contribute to neointimal hyperplasia after injury, which causes vascular remodeling related to arteriosclerosis, hypertension, and restenosis. Lethal giant larvae 1 (LGL1) is a highly conserved protein and plays an important role in cell polarity and tumor suppression. However, whether LGL1 affects neointimal hyperplasia is still unknown. In this study, we used smooth muscle-specific LGL1 knockout (LGL1SMKO) mice generated by cross-breeding LGL1flox/flox mice with α-SMA-Cre mice. LGL1 expression was significantly decreased during both carotid artery ligation in vivo and PDGF-BB stimulation in vitro. LGL1 overexpression inhibited the proliferation and migration of VSMCs. Mechanistically, LGL1 could bind with signal transducer and activator of transcription 3 (STAT3) and promote its degradation via the proteasomal pathway. In the carotid artery ligation animal model, smooth muscle-specific deletion of LGL1 accelerated neointimal hyperplasia, which was attenuated by the STAT3 inhibitor SH-4-54. In conclusion, LGL1 may inhibit neointimal hyperplasia by repressing VSMC proliferation and migration via promoting STAT3 proteasomal degradation.
Collapse
Affiliation(s)
- Ya Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peidong Yuan
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoping Ma
- Department of Obstetrics and Gynecology, Liaocheng People’s Hospital, Liaocheng, China
| | - Qiming Deng
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Jianmin Yang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianran Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Tianran Zhang, ; Cheng Zhang, ; Wencheng Zhang,
| | - Cheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Tianran Zhang, ; Cheng Zhang, ; Wencheng Zhang,
| | - Wencheng Zhang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Cardiovascular Disease Research Center of Shandong First Medical University, Central Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Tianran Zhang, ; Cheng Zhang, ; Wencheng Zhang,
| |
Collapse
|
10
|
Drosophila Accessory Gland: A Complementary In Vivo Model to Bring New Insight to Prostate Cancer. Cells 2021; 10:cells10092387. [PMID: 34572036 PMCID: PMC8468328 DOI: 10.3390/cells10092387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/03/2021] [Accepted: 09/08/2021] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer is the most common cancer in aging men. Despite recent progress, there are still few effective treatments to cure its aggressive and metastatic stages. A better understanding of the molecular mechanisms driving disease initiation and progression appears essential to support the development of more efficient therapies and improve patient care. To do so, multiple research models, such as cell culture and mouse models, have been developed over the years and have improved our comprehension of the biology of the disease. Recently, a new model has been added with the use of the Drosophila accessory gland. With a high level of conservation of major signaling pathways implicated in human disease, this functional equivalent of the prostate represents a powerful, inexpensive, and rapid in vivo model to study epithelial carcinogenesis. The purpose of this review is to quickly overview the existing prostate cancer models, including their strengths and limitations. In particular, we discuss how the Drosophila accessory gland can be integrated as a convenient complementary model by bringing new understanding in the mechanisms driving prostate epithelial tumorigenesis, from initiation to metastatic formation.
Collapse
|
11
|
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
|
12
|
Dillard C, Reis JGT, Rusten TE. RasV12; scrib-/- Tumors: A Cooperative Oncogenesis Model Fueled by Tumor/Host Interactions. Int J Mol Sci 2021; 22:ijms22168873. [PMID: 34445578 PMCID: PMC8396170 DOI: 10.3390/ijms22168873] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
The phenomenon of how oncogenes and tumor-suppressor mutations can synergize to promote tumor fitness and cancer progression can be studied in relatively simple animal model systems such as Drosophila melanogaster. Almost two decades after the landmark discovery of cooperative oncogenesis between oncogenic RasV12 and the loss of the tumor suppressor scribble in flies, this and other tumor models have provided new concepts and findings in cancer biology that has remarkable parallels and relevance to human cancer. Here we review findings using the RasV12; scrib-/- tumor model and how it has contributed to our understanding of how these initial simple genetic insults cooperate within the tumor cell to set in motion the malignant transformation program leading to tumor growth through cell growth, cell survival and proliferation, dismantling of cell-cell interactions, degradation of basement membrane and spreading to other organs. Recent findings have demonstrated that cooperativity goes beyond cell intrinsic mechanisms as the tumor interacts with the immediate cells of the microenvironment, the immune system and systemic organs to eventually facilitate malignant progression.
Collapse
Affiliation(s)
- Caroline Dillard
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Correspondence: (C.D.); (T.E.R.)
| | - José Gerardo Teles Reis
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway;
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, 0379 Oslo, Norway
- Correspondence: (C.D.); (T.E.R.)
| |
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
|
Millet-Boureima C, He S, Le TBU, Gamberi C. Modeling Neoplastic Growth in Renal Cell Carcinoma and Polycystic Kidney Disease. Int J Mol Sci 2021; 22:3918. [PMID: 33920158 PMCID: PMC8070407 DOI: 10.3390/ijms22083918] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/06/2021] [Accepted: 04/08/2021] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) and autosomal dominant polycystic kidney disease (ADPKD) share several characteristics, including neoplastic cell growth, kidney cysts, and limited therapeutics. As well, both exhibit impaired vasculature and compensatory VEGF activation of angiogenesis. The PI3K/AKT/mTOR and Ras/Raf/ERK pathways play important roles in regulating cystic and tumor cell proliferation and growth. Both RCC and ADPKD result in hypoxia, where HIF-α signaling is activated in response to oxygen deprivation. Primary cilia and altered cell metabolism may play a role in disease progression. Non-coding RNAs may regulate RCC carcinogenesis and ADPKD through their varied effects. Drosophila exhibits remarkable conservation of the pathways involved in RCC and ADPKD. Here, we review the progress towards understanding disease mechanisms, partially overlapping cellular and molecular dysfunctions in RCC and ADPKD and reflect on the potential for the agile Drosophila genetic model to accelerate discovery science, address unresolved mechanistic aspects of these diseases, and perform rapid pharmacological screens.
Collapse
Affiliation(s)
- Cassandra Millet-Boureima
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
| | - Thi Bich Uyen Le
- Department of Biology, Concordia University, Montreal, QC H4B 1R6, Canada; (C.M.-B.); (S.H.); (T.B.U.L.)
- Haematology-Oncology Research Group, National University Cancer Institute, Singapore 119228, Singapore
| | - Chiara Gamberi
- Department of Biology, Coastal Carolina University, Conway, SC 29528-6054, USA
| |
Collapse
|
15
|
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
|
16
|
Finding relationships among biological entities. LOGIC AND CRITICAL THINKING IN THE BIOMEDICAL SCIENCES 2020. [PMCID: PMC7499094 DOI: 10.1016/b978-0-12-821364-3.00005-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Confusion over the concepts of “relationships” and “similarities” lies at the heart of many battles over the direction and intent of research projects. Here is a short story that demonstrates the difference between the two concepts: You look up at the clouds, and you begin to see the shape of a lion. The cloud has a tail, like a lion’s tale, and a fluffy head, like a lion’s mane. With a little imagination the mouth of the lion seems to roar down from the sky. You have succeeded in finding similarities between the cloud and a lion. If you look at a cloud and you imagine a tea kettle producing a head of steam and you recognize that the physical forces that create a cloud and the physical forces that produced steam from a heated kettle are the same, then you have found a relationship. Most popular classification algorithms operate by grouping together data objects that have similar properties or values. In so doing, they may miss finding the true relationships among objects. Traditionally, relationships among data objects are discovered by an intellectual process. In this chapter, we will discuss the scientific gains that come when we classify biological entities by relationships, not by their similarities.
Collapse
|
17
|
Liu N, Cui W, Chen M, Zhang X, Song X, Pan C. A 21-bp indel within the LLGL1 gene is significantly associated with litter size in goat. Anim Biotechnol 2019; 32:213-218. [PMID: 31646948 DOI: 10.1080/10495398.2019.1677682] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The scribble cell polarity complex component (LLGL1) is part of the cytoskeletal network and is involved in maintaining cell polarity and epithelial integrity. Based on the whole-genome sequencing analysis in goat, LLGL1 gene is suggested as a putative important candidate gene affecting litter size in Shaanbei White Cashmere Goats (SBWC). Therefore, the objective of this study was to uncover the possible novel insertion/deletion (Indel) variant in goat LLGL1 gene and to evaluate its association with litter size of SBWC (n = 827). Using the PCR detection and DNA sequencing, the 21-bp indel in the upstream of LLGL1 was firstly founded and two genotypes were identified: II (insertion/insertion) and ID (insertion/deletion), respectively. Association analyses revealed that the 21-bp indel was significantly correlated with litter size (p = 0.017). Notably, the individuals with II genotype were significantly greater than that of the genotype ID, and the 'I' allele was dominant. Additionally, the remarkable influence of the indel on traits might be related to the change of DEAF-1-related (NUDR) binding site through bioinformatics analysis. Briefly, the 21-bp indel within the goat LLGL1 gene could be an effective DNA molecular marker and provide valuable theoretical basis for marker-assisted selection (MAS) in goat industry.
Collapse
Affiliation(s)
- Nuan Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Wenbo Cui
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Mingyue Chen
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xuelian Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| | - Xiaoyue Song
- Shaanxi Provincial Engineering and Technology Research Center of Cashmere Goats, Yulin University, Yulin, Shaanxi, China.,Life Science Research Center, Yulin University, Yulin, Shaanxi, China
| | - Chuanying Pan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
18
|
Villegas SN. One hundred years of Drosophila cancer research: no longer in solitude. Dis Model Mech 2019; 12:12/4/dmm039032. [PMID: 30952627 PMCID: PMC6505481 DOI: 10.1242/dmm.039032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022] Open
Abstract
When Mary Stark first described the presence of tumours in the fruit fly Drosophila melanogaster in 1918, would she ever have imagined that flies would become an invaluable organism for modelling and understanding oncogenesis? And if so, would she have expected it to take 100 years for this model to be fully accredited? This Special Article summarises the efforts and achievements of Drosophilists to establish the fly as a valid model in cancer research through different scientific periods.
Collapse
Affiliation(s)
- Santiago Nahuel Villegas
- Instituto de Neurociencias, Consejo Superior de Investigaciones Científicas (CSIC) and Universidad Miguel Hernandez (UMH), Campus de Sant Joan, Apartado 18, 03550 Sant Joan, Alicante, Spain
| |
Collapse
|
19
|
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
|
20
|
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: 147] [Impact Index Per Article: 24.5] [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
|
21
|
Kurelac I, Iommarini L, Vatrinet R, Amato LB, De Luise M, Leone G, Girolimetti G, Umesh Ganesh N, Bridgeman VL, Ombrato L, Columbaro M, Ragazzi M, Gibellini L, Sollazzo M, Feichtinger RG, Vidali S, Baldassarre M, Foriel S, Vidone M, Cossarizza A, Grifoni D, Kofler B, Malanchi I, Porcelli AM, Gasparre G. Inducing cancer indolence by targeting mitochondrial Complex I is potentiated by blocking macrophage-mediated adaptive responses. Nat Commun 2019; 10:903. [PMID: 30796225 PMCID: PMC6385215 DOI: 10.1038/s41467-019-08839-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 01/30/2019] [Indexed: 02/08/2023] Open
Abstract
Converting carcinomas in benign oncocytomas has been suggested as a potential anti-cancer strategy. One of the oncocytoma hallmarks is the lack of respiratory complex I (CI). Here we use genetic ablation of this enzyme to induce indolence in two cancer types, and show this is reversed by allowing the stabilization of Hypoxia Inducible Factor-1 alpha (HIF-1α). We further show that on the long run CI-deficient tumors re-adapt to their inability to respond to hypoxia, concordantly with the persistence of human oncocytomas. We demonstrate that CI-deficient tumors survive and carry out angiogenesis, despite their inability to stabilize HIF-1α. Such adaptive response is mediated by tumor associated macrophages, whose blockage improves the effect of CI ablation. Additionally, the simultaneous pharmacological inhibition of CI function through metformin and macrophage infiltration through PLX-3397 impairs tumor growth in vivo in a synergistic manner, setting the basis for an efficient combinatorial adjuvant therapy in clinical trials.
Collapse
Affiliation(s)
- Ivana Kurelac
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Luisa Iommarini
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Renaud Vatrinet
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Laura Benedetta Amato
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Monica De Luise
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Giulia Leone
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Giulia Girolimetti
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Nikkitha Umesh Ganesh
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | | | - Luigi Ombrato
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK
| | - Marta Columbaro
- Laboratory of Musculoskeletal Cell Biology, IRCCS Istituto Ortopedico Rizzoli, Via Giulio Cesare Pupilli 1, 40136, Bologna, Italy
| | - Moira Ragazzi
- Anatomia Patologica, Azienda Ospedaliera S. Maria Nuova di Reggio Emilia, Viale Risorgimento 80, 42123, Reggio Emilia, Italy
| | - Lara Gibellini
- Dipartimento di Scienze Mediche e Chirurgiche materno infantili e dell'adulto, Università degli Studi di Modena e Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Manuela Sollazzo
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Rene Gunther Feichtinger
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Silvia Vidali
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Maurizio Baldassarre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Sarah Foriel
- Khondrion BV, Philips van Leydenlaan 15, 6525 EX, Nijmegen, The Netherlands
- Radboud Center for Mitochondrial Medicine (RCMM) at the Department of Pediatrics, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6500 HB, Nijmegen, The Netherlands
| | - Michele Vidone
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy
| | - Andrea Cossarizza
- Dipartimento di Scienze Mediche e Chirurgiche materno infantili e dell'adulto, Università degli Studi di Modena e Reggio Emilia, Via del Pozzo 71, 41124, Modena, Italy
| | - Daniela Grifoni
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy
| | - Barbara Kofler
- Research Program for Receptor Biochemistry and Tumor Metabolism, Department of Pediatrics, University Hospital of the Paracelsus Medical University, Muellner Hauptstraße 48, 5020, Salzburg, Austria
| | - Ilaria Malanchi
- Tumor-Host Interaction Lab, The Francis Crick Institute, 1 Midland Rd, NW1 1AT, London, UK.
| | - Anna Maria Porcelli
- Dipartimento di Farmacia e Biotecnologie, Università di Bologna, Via Selmi 3, 40126, Bologna, Italy.
- Centro Interdipartimentale di Ricerca Industriale Scienze della Vita e Tecnologie per la Salute, Università di Bologna, Via Tolara di Sopra 41/E, 40064, Ozzano dell'Emilia, Italy.
| | - Giuseppe Gasparre
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
- Centro di Ricerca Biomedica Applicata (CRBA), Università di Bologna, Via Massarenti 9, 40138, Bologna, Italy.
| |
Collapse
|
22
|
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
Affiliation(s)
| | | | | | | | | | | | - Daniela Grifoni
- Cancer Evolution Laboratory, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Portela M, Yang L, Paul S, Li X, Veraksa A, Parsons LM, Richardson HE. Lgl reduces endosomal vesicle acidification and Notch signaling by promoting the interaction between Vap33 and the V-ATPase complex. Sci Signal 2018; 11:11/533/eaar1976. [PMID: 29871910 DOI: 10.1126/scisignal.aar1976] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Epithelial cell polarity is linked to the control of tissue growth and tumorigenesis. The tumor suppressor and cell polarity protein lethal-2-giant larvae (Lgl) promotes Hippo signaling and inhibits Notch signaling to restrict tissue growth in Drosophila melanogaster Notch signaling is greater in lgl mutant tissue than in wild-type tissue because of increased acidification of endosomal vesicles, which promotes the proteolytic processing and activation of Notch by γ-secretase. We showed that the increased Notch signaling and tissue growth defects of lgl mutant tissue depended on endosomal vesicle acidification mediated by the vacuolar adenosine triphosphatase (V-ATPase). Lgl promoted the activity of the V-ATPase by interacting with Vap33 (VAMP-associated protein of 33 kDa). Vap33 physically and genetically interacted with Lgl and V-ATPase subunits and repressed V-ATPase-mediated endosomal vesicle acidification and Notch signaling. Vap33 overexpression reduced the abundance of the V-ATPase component Vha44, whereas Lgl knockdown reduced the binding of Vap33 to the V-ATPase component Vha68-3. Our data indicate that Lgl promotes the binding of Vap33 to the V-ATPase, thus inhibiting V-ATPase-mediated endosomal vesicle acidification and thereby reducing γ-secretase activity, Notch signaling, and tissue growth. Our findings implicate the deregulation of Vap33 and V-ATPase activity in polarity-impaired epithelial cancers.
Collapse
Affiliation(s)
- Marta Portela
- Department of Biochemistry and Genetics, 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.,Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute, Avenida Doctor Arce, 37, Madrid 28002, Spain
| | - Liu Yang
- Department of Biology, University of Massachusetts, Boston, MA 02125, USA
| | - Sayantanee Paul
- Department of Biology, University of Massachusetts, Boston, MA 02125, USA
| | - Xia Li
- Department of Mathematics and Statistics, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Alexey Veraksa
- Department of Biology, University of Massachusetts, Boston, MA 02125, USA
| | - Linda M Parsons
- Cell Cycle and Development Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria 3002, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Helena E Richardson
- Department of Biochemistry and Genetics, 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
|
24
|
Saito Y, Desai RR, Muthuswamy SK. Reinterpreting polarity and cancer: The changing landscape from tumor suppression to tumor promotion. Biochim Biophys Acta Rev Cancer 2018; 1869:103-116. [DOI: 10.1016/j.bbcan.2017.12.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 12/08/2017] [Indexed: 12/21/2022]
|
25
|
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
|
26
|
Stephens R, Lim K, Portela M, Kvansakul M, Humbert PO, Richardson HE. The Scribble Cell Polarity Module in the Regulation of Cell Signaling in Tissue Development and Tumorigenesis. J Mol Biol 2018; 430:3585-3612. [PMID: 29409995 DOI: 10.1016/j.jmb.2018.01.011] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 01/22/2023]
Abstract
The Scribble cell polarity module, comprising Scribbled (Scrib), Discs-large (Dlg) and Lethal-2-giant larvae (Lgl), has a tumor suppressive role in mammalian epithelial cancers. The Scribble module proteins play key functions in the establishment and maintenance of different modes of cell polarity, as well as in the control of tissue growth, differentiation and directed cell migration, and therefore are major regulators of tissue development and homeostasis. Whilst molecular details are known regarding the roles of Scribble module proteins in cell polarity regulation, their precise mode of action in the regulation of other key cellular processes remains enigmatic. An accumulating body of evidence indicates that Scribble module proteins play scaffolding roles in the control of various signaling pathways, which are linked to the control of tissue growth, differentiation and cell migration. Multiple Scrib, Dlg and Lgl interacting proteins have been discovered, which are involved in diverse processes, however many function in the regulation of cellular signaling. Herein, we review the components of the Scrib, Dlg and Lgl protein interactomes, and focus on the mechanism by which they regulate cellular signaling pathways in metazoans, and how their disruption leads to cancer.
Collapse
Affiliation(s)
- Rebecca Stephens
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Krystle Lim
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Marta Portela
- Department of Molecular, Cellular and Developmental Neurobiology, Cajal Institute (CSIC), Avenida Doctor Arce, 37, Madrid 28002, Spain
| | - Marc Kvansakul
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Patrick O Humbert
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia; Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Helena E Richardson
- Department of Biochemistry and Genetics, La Trobe Institute of Molecular Science, La Trobe University, Melbourne, Victoria, Australia; Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Anatomy & Neurobiology, University of Melbourne, Melbourne, Victoria 3010, Australia.
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
Milgrom-Hoffman M, Humbert PO. Regulation of cellular and PCP signalling by the Scribble polarity module. Semin Cell Dev Biol 2017; 81:33-45. [PMID: 29154823 DOI: 10.1016/j.semcdb.2017.11.021] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/11/2017] [Accepted: 11/13/2017] [Indexed: 10/18/2022]
Abstract
Since the first identification of the Scribble polarity module proteins as a new class of tumour suppressors that regulate both cell polarity and proliferation, an increasing amount of evidence has uncovered a broader role for Scribble, Dlg and Lgl in the control of fundamental cellular functions and their signalling pathways. Here, we review these findings as well as discuss more specifically the role of the Scribble module in PCP signalling.
Collapse
Affiliation(s)
- Michal Milgrom-Hoffman
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia
| | - Patrick O Humbert
- Department of Biochemistry & Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria 3086, Australia; Department of Biochemistry & Molecular Biology, University of Melbourne, Melbourne, Victoria 3010, Australia; Department of Pathology, University of Melbourne, Melbourne, Victoria 3010, Australia.
| |
Collapse
|
32
|
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
|
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
|
Christofi T, Apidianakis Y. Drosophila and the hallmarks of cancer. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2016; 135:79-110. [PMID: 23615878 DOI: 10.1007/10_2013_190] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
: Cancer was the disease of the twentieth century. Today it is still a leading cause of death worldwide despite being intensively investigated. Abundant knowledge exists regarding the pathological and molecular mechanisms that drive healthy cells to become malignant and form metastatic tumors. The relation of oncogenes and tumor suppressors to the genetic trigger of carcinogenesis is unquestionable. However, the development of the disease requires many characteristics that due to their proven role in cancer are collectively described as the "hallmarks of cancer." We highlight here the historic discoveries made using the model organism Drosophila melanogaster and its contributions to biomedical and cancer research. Flies are utilized as a model organism for the investigation of each and every aspect of cancer hallmarks. Due to the significant conservation between flies and mammals at the signaling and tissue physiology level it is possible to explore the genes and mechanisms responsible for cancer pathogenesis in flies. Recent Drosophila studies suggest novel aspects of therapeutic intervention and are expected to guide cancer research in the twenty-first century.
Collapse
|
35
|
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: 17] [Impact Index Per Article: 1.7] [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
|
36
|
Rho1-Wnd signaling regulates loss-of-cell polarity-induced cell invasion in Drosophila. Oncogene 2015; 35:846-55. [PMID: 25961917 DOI: 10.1038/onc.2015.137] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 03/21/2015] [Accepted: 03/30/2015] [Indexed: 12/11/2022]
Abstract
Both cell polarity and c-Jun N-terminal kinase (JNK) activity are essential to the maintenance of tissue homeostasis, and disruption of either is commonly seen in cancer progression. Despite the established connection between loss-of-cell polarity and JNK activation, much less is known about the molecular mechanism by which aberrant cell polarity induces JNK-mediated cell migration and tumor invasion. Here we show results from a genetic screen using an in vivo invasion model via knocking down cell polarity gene in Drosophila wing discs, and identify Rho1-Wnd signaling as an important molecular link that mediates loss-of-cell polarity-triggered JNK activation and cell invasion. We show that Wallenda (Wnd), a protein kinase of the mitogen-activated protein kinase kinase kinase family, by forming a complex with the GTPase Rho1, is both necessary and sufficient for Rho1-induced JNK-dependent cell invasion, MMP1 activation and epithelial-mesenchymal transition. Furthermore, Wnd promotes cell proliferation and tissue growth through wingless production when apoptosis is inhibited by p35. Finally, Wnd shows oncogenic cooperation with Ras(V12) to trigger tumor growth in eye discs and causes invasion into the ventral nerve cord. Together, our data not only provides a novel mechanistic insight on how cell polarity loss contributes to cell invasion, but also highlights the value of the Drosophila model system to explore human cancer biology.
Collapse
|
37
|
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
|
38
|
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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
Dickerman BK, McDonald JA, Sen GC. The Human dsRNA binding protein PACT is unable to functionally substitute for the Drosophila dsRNA binding protein R2D2. F1000Res 2014; 2:220. [PMID: 24715958 PMCID: PMC3962003 DOI: 10.12688/f1000research.2-220.v2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/19/2014] [Indexed: 11/20/2022] Open
Abstract
The dsRNA binding protein (dsRBP) PACT was first described as an activator of the dsRNA dependent protein kinase PKR in response to stress signals. Additionally, it has been identified as a component of the small RNA processing pathway. A role for PACT in this pathway represents an important interplay between two modes of post-transcriptional gene regulation. The function of PACT in this context is poorly understood. Thus, additional approaches are required to clarify the mechanism by which PACT functions. In this study, the genetic utility of
Drosophila melanogaster was employed to identify dsRNA-binding proteins that are functionally orthologous to PACT. Transgenic
Drosophila expressing human PACT were generated to determine whether PACT is capable of functionally substituting for the
Drosophila dsRBP R2D2, which has a well-defined role in small RNA biogenesis. Results presented here indicate that PACT is unable to substitute for R2D2 at the whole organism level.
Collapse
Affiliation(s)
- Benjamin K Dickerman
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jocelyn A McDonald
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Ganes C Sen
- Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| |
Collapse
|
43
|
Wennmann DO, Schmitz J, Wehr MC, Krahn MP, Koschmal N, Gromnitza S, Schulze U, Weide T, Chekuri A, Skryabin BV, Gerke V, Pavenstädt H, Duning K, Kremerskothen J. Evolutionary and Molecular Facts Link the WWC Protein Family to Hippo Signaling. Mol Biol Evol 2014; 31:1710-23. [DOI: 10.1093/molbev/msu115] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
44
|
Rewiring cell polarity signaling in cancer. Oncogene 2014; 34:939-50. [PMID: 24632617 DOI: 10.1038/onc.2014.59] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [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
|
45
|
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: 17] [Impact Index Per Article: 1.5] [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
|
46
|
Song J, Peng XL, Ji MY, Ai MH, Zhang JX, Dong WG. Hugl-1 induces apoptosis in esophageal carcinoma cells both in vitro and in vivo. World J Gastroenterol 2013; 19:4127-4136. [PMID: 23864775 PMCID: PMC3710414 DOI: 10.3748/wjg.v19.i26.4127] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/15/2013] [Accepted: 05/10/2013] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine whether the human giant larvae homolog 1 gene (Hugl-1/Llg1/Lgl1) exerts tumor suppressor effects in esophageal cancer.
METHODS: We constructed a Hugl-1 expression plasmid, pEZ-M29-Hugl1, for gene transfection. We transfected the pEZ-M29-Hugl1 plasmid into Eca109 esophageal cancer cell lines with Lipofectamine 2000 to overexpress Hugl-1. Real-time reverse transcription-polymerase chain reaction (RT-PCR) and Western blotting were performed to determine the effects of the plasmid on Hugl-1 expression. In vitro cell proliferation and apoptosis were examined separately by cell counting Kit-8 (CCK-8) assay, flow cytometry, and Western blotting before and after the transfection of the plasmid into Eca109 cells. Cell cycle distribution was assessed with flow cytometry. The effect of Hugl-1 overexpressing on tumor growth in vivo was performed with a xenograft tumor model in nude mice. Expression of Hugl-1 in xenograft tumor was analyzed by immunohistochemistry. The transferase-mediated dUTP nick end-labeling (TUNEL) technique was performed to detect and quantitate apoptotic cell.
RESULTS: The transfection efficiency was confirmed with real-time RT-PCR and Western blotting. Our results show that compared with control groups the mRNA levels and protein levels of Hugl-1 in pEZ-M29-Hugl1-treated group were remarkably increased (P < 0.05). The CCK-8 assay demonstrated that the growth of cells overexpressing Hugl-1 was significantly lower than control cells. Cell cycle distribution showed there was a G0/G1 cell cycle arrest in cells overexpressing Hugl-1 (64.09% ± 3.14% vs 50.32% ± 4.60%, 64.09% ± 3.14% vs 49.13% ± 2.24%). Annexin V-fluorescein isothiocyanate revealed that apoptosis was significantly increased in cells overexpressing Hugl-1 compared with control group (17.33% ± 4.76% vs 6.90% ± 1.61%, 17.33% ± 4.76% vs 6.27% ± 0.38%). Moreover, we found that Hugl-1 changes the level of the anti-apoptotic protein Bcl-2 and the pro-apoptotic protein Bax and the activation of both caspase-3 and caspase-9. With a TUNEL assay, we found that Hugl-1 markedly increased the apoptosis rate of Eca109 cells in vivo (60.50% ± 9.11% vs 25.00% ± 12.25%). It was shown that Hugl-1 represents a significantly more effective tumor suppressor gene alone in a xenograft tumor mouse model. This data suggest that Hugl-1 inhibited tumor growth and induced cell apoptosis in vivo.
CONCLUSION: These results suggest that Hugl-1 induces growth suppression and apoptosis in a human esophageal squamous cell carcinoma cell line both in vitro and in vivo.
Collapse
|
47
|
Abstract
The conserved polarity proteins Par6 and aPKC regulate cell polarization processes. However, increasing evidence also suggests that they play a role in oncogenic progression. During tumor progression, epithelial to mesenchymal transition (EMT) delineates an evolutionary conserved process that converts stationary epithelial cells into mesenchymal cells, which have an acquired ability for independent migration and invasion. In addition to signaling pathways that alter genetic programes that trigger the loss of cell-cell adhesion, alternative pathways can alter cell plasticity to regulate cell-cell cohesion and increase invasive potential. One such pathway involves TGFβ-induced phosphorylation of Par6. In epithelial cells, Par6 phosphorylation results in the dissolution of junctional complexes, cytoskeletal remodelling, and increased metastatic potential. Recently, we found that aPKC can also phosphorylate Par6 to drive EMT and increase the migratory potential of non-small cell lung cancer cells. This result has implications with respect to homeostatic and developmental processes involving polarization, and also with respect to cancer progression-particularly since aPKC has been reported to be an oncogenic regulator in various tumor cells.
Collapse
Affiliation(s)
- Adrian Gunaratne
- Department of Physiology and Pharmacology; Western University; London, ON, Canada
| | | |
Collapse
|
48
|
Asymmetric cell division of stem and progenitor cells during homeostasis and cancer. Cell Mol Life Sci 2013; 71:575-97. [PMID: 23771628 PMCID: PMC3901929 DOI: 10.1007/s00018-013-1386-1] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Revised: 05/21/2013] [Accepted: 05/24/2013] [Indexed: 12/15/2022]
Abstract
Stem and progenitor cells are characterized by their ability to self-renew and produce differentiated progeny. A fine balance between these processes is achieved through controlled asymmetric divisions and is necessary to generate cellular diversity during development and to maintain adult tissue homeostasis. Disruption of this balance may result in premature depletion of the stem/progenitor cell pool, or abnormal growth. In many tissues, including the brain, dysregulated asymmetric divisions are associated with cancer. Whether there is a causal relationship between asymmetric cell division defects and cancer initiation is as yet not known. Here, we review the cellular and molecular mechanisms that regulate asymmetric cell divisions in the neural lineage and discuss the potential connections between this regulatory machinery and cancer.
Collapse
|
49
|
Bazzoun D, Lelièvre S, Talhouk R. Polarity proteins as regulators of cell junction complexes: implications for breast cancer. Pharmacol Ther 2013; 138:418-27. [PMID: 23458609 PMCID: PMC3648792 DOI: 10.1016/j.pharmthera.2013.02.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 02/07/2013] [Indexed: 12/19/2022]
Abstract
The epithelium of multicellular organisms possesses a well-defined architecture, referred to as polarity that coordinates the regulation of essential cell features. Polarity proteins are intimately linked to the protein complexes that make the tight, adherens and gap junctions; they contribute to the proper localization and assembly of these cell-cell junctions within cells and consequently to functional tissue organization. The establishment of cell-cell junctions and polarity are both implicated in the regulation of epithelial modifications in normal and cancer situations. Uncovering the mechanisms through which cell-cell junctions and epithelial polarization are established and how their interaction with the microenvironment directs cell and tissue organization has opened new venues for the development of cancer therapies. In this review, we focus on the breast epithelium to highlight how polarity and cell-cell junction proteins interact together in normal and cancerous contexts to regulate major cellular mechanisms such as migration. The impact of these proteins on epigenetic mechanisms responsible for resetting cells toward oncogenesis is discussed in light of increasing evidence that tissue polarity modulates chromatin function. Finally, we give an overview of recent breast cancer therapies that target proteins involved in cell-cell junctions.
Collapse
Affiliation(s)
- Dana Bazzoun
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), Beirut, Lebanon
| | - Sophie Lelièvre
- Department of Basic Medical Sciences and Center for Cancer Research, Purdue University, IN, U.S.A
| | - Rabih Talhouk
- Department of Biology, Faculty of Arts and Sciences, American University of Beirut (AUB), Beirut, Lebanon
| |
Collapse
|
50
|
Nagasaka K, Seiki T, Yamashita A, Massimi P, Subbaiah VK, Thomas M, Kranjec C, Kawana K, Nakagawa S, Yano T, Taketani Y, Fujii T, Kozuma S, Banks L. A novel interaction between hScrib and PP1γ downregulates ERK signaling and suppresses oncogene-induced cell transformation. PLoS One 2013; 8:e53752. [PMID: 23359326 PMCID: PMC3554735 DOI: 10.1371/journal.pone.0053752] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2012] [Accepted: 12/04/2012] [Indexed: 12/22/2022] Open
Abstract
Previous studies have shown that the cell polarity regulator hScrib interacts with, and consequently controls, the ERK signaling pathway. This interaction occurs through two well-conserved Kinase Interacting Motifs, which allow hScrib to bind ERK1 directly, resulting in a reduction in the levels of phospho-ERK. This suggests that hScrib might recruit a phosphatase to regulate this signaling pathway. Using a proteomic approach we now show that Protein Phosphatase 1γ (PP1γ) is a major interacting partner of hScrib. This interaction is direct and occurs through a conserved PP1γ interaction motif on the hScrib protein, and this interaction appears to be required for hScrib's ability to downregulate ERK phosphorylation. In addition, hScrib also controls the pattern of PP1γ localization, where loss of hScrib enhances the nuclear translocation of PP1γ. Furthermore, we also show that the ability of hScrib to interact with PP1γ is important for the ability of hScrib to suppress oncogene-induced transformation of primary rodent cells. Taken together, these results demonstrate that hScrib acts as a scaffold to integrate the control of the PP1γ and ERK signaling pathways and explains how disruption of hScrib localisation can contribute towards the development of human malignancy.
Collapse
Affiliation(s)
- Kazunori Nagasaka
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takayuki Seiki
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Aki Yamashita
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Paola Massimi
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Trieste, Italy
| | - Vanitha Krishna Subbaiah
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Trieste, Italy
| | - Miranda Thomas
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Trieste, Italy
| | - Christian Kranjec
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Trieste, Italy
| | - Kei Kawana
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shunsuke Nakagawa
- Department of Obstetrics and Gynecology, The Teikyo University School of Medicine, Tokyo, Japan
| | - Tetsu Yano
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yuji Taketani
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tomoyuki Fujii
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shiro Kozuma
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Area Science Park, Trieste, Italy
| |
Collapse
|