1
|
Jiang X, Xu Z, Jiang S, Wang H, Xiao M, Shi Y, Wang K. PDZ and LIM Domain-Encoding Genes: Their Role in Cancer Development. Cancers (Basel) 2023; 15:5042. [PMID: 37894409 PMCID: PMC10605254 DOI: 10.3390/cancers15205042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
PDZ-LIM family proteins (PDLIMs) are a kind of scaffolding proteins that contain PDZ and LIM interaction domains. As protein-protein interacting molecules, PDZ and LIM domains function as scaffolds to bind to a variety of proteins. The PDLIMs are composed of evolutionarily conserved proteins found throughout different species. They can participate in cell signal transduction by mediating the interaction of signal molecules. They are involved in many important physiological processes, such as cell differentiation, proliferation, migration, and the maintenance of cellular structural integrity. Studies have shown that dysregulation of the PDLIMs leads to tumor formation and development. In this paper, we review and integrate the current knowledge on PDLIMs. The structure and function of the PDZ and LIM structural domains and the role of the PDLIMs in tumor development are described.
Collapse
Affiliation(s)
| | | | | | | | | | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; (X.J.); (Z.X.); (S.J.); (H.W.); (M.X.)
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu 322000, China; (X.J.); (Z.X.); (S.J.); (H.W.); (M.X.)
| |
Collapse
|
2
|
Cox OT, O’Sullivan N, Tresse E, Ward S, Buckley N, O’Connor R. PDLIM2 is highly expressed in Breast Cancer tumour-associated macrophages and is required for M2 macrophage polarization. Front Oncol 2022; 12:1028959. [PMID: 36531051 PMCID: PMC9749823 DOI: 10.3389/fonc.2022.1028959] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/04/2022] [Indexed: 03/04/2025] Open
Abstract
The PDZ-LIM domain-containing protein 2 (PDLIM2) regulates cell polarity and the protein stability of key transcription factors in epithelial and hemopoietic cells. We previously reported that PDLIM2 is more highly expressed in Triple Negative Breast Cancer (TNBC) than in other breast cancer types or normal breast tissue. In the course of the TNBC study, it was noted that PDLIM2 was highly expressed in the stroma of PDLIM2-expressing tumours. Here, we investigated the phenotype of these stromal cells and whether any infiltrating immune population was linked to PDLIM2 expression. We found that high PDLIM2 expression in breast tumours was associated with higher levels of infiltrating M2 macrophages, but was not associated with infiltrating T cell sub-populations. We then tested whether PDLIM2 contributes to macrophage differentiation or function by using cultures of bone marrow-derived macrophages from wildtype and Pdlim2 knockout mice. This demonstrated that PDLIM2 is required for naïve macrophage migration and for the full adoption of IL-4-induced M2 polarization, including expression of M2 phenotypic markers, cell adhesion and cell migration. TLR4-, TLR3- or IFNγ-induced M1 macrophage activity was less dependent on PDLIM2. Finally, analysis of publicly available breast cancer datasets showed that high PDLIM2 expression is associated with increased M2 macrophage infiltration. We conclude that PDLIM2 expression influences the tumour associated stroma and, in particular, M2 macrophage infiltration that may contribute to the progression of TNBC or other subsets of breast cancer.
Collapse
Affiliation(s)
- Orla T. Cox
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Neil O’Sullivan
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Emilie Tresse
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Stephanie Ward
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, Cork, Ireland
| | - Niamh Buckley
- School of Pharmacy and Patrick G. Johnson Centre for Cancer Research, Queens University, Belfast, United Kingdom
| | - Rosemary O’Connor
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, BioSciences Institute, University College Cork, Cork, Ireland
| |
Collapse
|
3
|
High Expression of PDLIM2 Predicts a Poor Prognosis in Prostate Cancer and Is Correlated with Epithelial-Mesenchymal Transition and Immune Cell Infiltration. J Immunol Res 2022; 2022:2922832. [PMID: 35707002 PMCID: PMC9192325 DOI: 10.1155/2022/2922832] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Purpose To elucidate the clinical and prognostic role of PDZ and LIM domain protein (PDLIM) genes and the association to epithelial-mesenchymal transition (EMT) and immune cell infiltration in patients with prostate cancer (PRAD). Methods The data of RNA-seq, DNA methylation, and clinical features of PRAD patients were collected from The Cancer Genome Atlas (TCGA) database to define the prognostic value of PDLIM gene expression and the association with EMT and immune cell infiltration. A tissue microarray including 134 radical prostatectomy specimens was served as validation by immunohistochemistry (IHC) staining analysis. Results The mRNA levels of PDLIM1/2/3/4/6/7 were significantly downregulated, while PDLIM5 was upregulated in PRAD (P < 0.05). High expression of PDLIM2 mRNA suggests poor progression free interval in PRAD patients. DNA methylation of PDLIM2 was correlated with its mRNA expression level, and that the cg22973076 methylation site in PDLIM2 was associated with shorter PFI (P < 0.05) in PRAD. Single-sample gene-set enrichment and gene functional enrichment results showed that PDLIM2 was correlated with EMT and immune processes. Spearman's test showed a significant correlation with six reported EMT signatures and several EMT signature-related genes. Tumor microenvironment analysis revealed that the PDLIM2 mRNA expression was positively correlated with the immune score, stromal score, and various tumor infiltrating immune cells. Additionally, the results showed that patients in the high-PDLIM2 mRNA expression group may be more sensitive to immune checkpoint blockade therapy. Finally, IHC analysis further implicated the protein level of PDLIM2 was upregulated in PRAD and acts as a novel potential biomarker in predicting tumor progression. Conclusion Our study suggests that PDLIM family genes might be significantly correlated with oncogenesis and the progression of PRAD. PDLIM2 correlated with EMT and immune cell infiltration by acting as an oncogene in PRAD, which may serve as a potential prognostic biomarker for PRAD patients.
Collapse
|
4
|
Guo ZS, Qu Z. PDLIM2: Signaling pathways and functions in cancer suppression and host immunity. Biochim Biophys Acta Rev Cancer 2021; 1876:188630. [PMID: 34571051 DOI: 10.1016/j.bbcan.2021.188630] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/30/2021] [Accepted: 09/22/2021] [Indexed: 12/20/2022]
Abstract
PDZ and LIM domains-containing proteins play pivotal functions in cell cytoskeleton organization, cell polarization and differentiation. As a key member of the family, PDLIM2 regulates stability and activity of transcription factors such as NF-κB, STATs and β-catenin, and thus exert it functions in inflammation, immunity, and cancer. PDLIM2 functions as a tumor suppressor in multiple tissues and it is often genetically mutated or epigenetically silenced in human cancers derived from lung, breast, ovarian and other histologies. However, in certain types of cancers, PDLIM2 may promote cancer cell proliferation and metastases. Therefore, PDLIM2 is added to a long list of genes that can function as tumor suppressor or oncogenic protein. During tumorigenesis induced by oncogenic viruses, PDLIM2 is a key target. Through promotion of NF-κB/RelA and STAT3 degradation, PDLIM2 enhances expression of proteins involved in antigen presentation and promotes T-cell activation while repressing multidrug resistance genes, thereby rendering mutated cells susceptible to immune surveillance and cytotoxicity mediated by immune cells and chemotherapeutic drugs. Intriguingly, PDLIM2 in alveolar macrophages (AMs) plays key roles in monitoring lung tumorigenesis, as its selective genetic deletion leads to constitutive activation of STAT3, driving monocyte differentiation to AMs with pro-tumorigenic polarization and activation. PDLIM2 has also been explored as a therapeutic target for cancer therapy. At the end of this review, we provide perspectives on this important molecule and discuss the future directions of both basic and translational studies.
Collapse
Affiliation(s)
- Zong Sheng Guo
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Zhaoxia Qu
- UPMC Hillman Cancer Center, Pittsburgh, PA, USA; Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
5
|
Frank CN, Hou X, Petrosyan A, Villani V, Zhao R, Hansen JR, Clair G, Salem F, De Filippo RE, Cravedi P, Lemley KV, Perin L. Effect of disease progression on the podocyte cell cycle in Alport Syndrome. Kidney Int 2021; 101:106-118. [PMID: 34562503 DOI: 10.1016/j.kint.2021.08.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/18/2021] [Accepted: 08/19/2021] [Indexed: 01/16/2023]
Abstract
Progression of glomerulosclerosis is associated with loss of podocytes with subsequent glomerular tuft instability. It is thought that a diminished number of podocytes may be able to preserve tuft stability through cell hypertrophy associated with cell cycle reentry. At the same time, reentry into the cell cycle risks podocyte detachment if podocytes cross the G1/S checkpoint and undergo abortive cytokinesis. In order to study cell cycle dynamics during chronic kidney disease (CKD) development, we used a FUCCI model (fluorescence ubiquitination-based cell cycle indicator) of mice with X-linked Alport Syndrome. This model exhibits progressive CKD and expresses fluorescent reporters of cell cycle stage exclusively in podocytes. With the development of CKD, an increasing fraction of podocytes in vivo were found to be in G1 or later cell cycle stages. Podocytes in G1 and G2 were hypertrophic. Heterozygous female mice, with milder manifestations of CKD, showed G1 fraction numbers intermediate between wild-type and male Alport mice. Proteomic analysis of podocytes in different cell cycle phases showed differences in cytoskeleton reorganization and metabolic processes between G0 and G1 in disease. Additionally, in vitro experiments confirmed that damaged podocytes reentered the cell cycle comparable to podocytes in vivo. Importantly, we confirmed the upregulation of PDlim2, a highly expressed protein in podocytes in G1, in a patient with Alport Syndrome, confirming our proteomics data in the human setting. Thus, our data showed that in the Alport model of progressive CKD, podocyte cell cycle distribution is altered, suggesting that cell cycle manipulation approaches may have a role in the treatment of various progressive glomerular diseases characterized by podocytopenia.
Collapse
Affiliation(s)
- Camille Nicolas Frank
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA; Division of Nephrology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Xiaogang Hou
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Astgik Petrosyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Valentina Villani
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA
| | - Rui Zhao
- Biological Science Division, Integrative Omics, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Joshua R Hansen
- Biological Science Division, Integrative Omics, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Geremy Clair
- Biological Science Division, Integrative Omics, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Fadi Salem
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Roger E De Filippo
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Paolo Cravedi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kevin V Lemley
- Division of Nephrology, Department of Pediatrics, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, California, USA; Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA.
| |
Collapse
|
6
|
OXER1 and RACK1-associated pathway: a promising drug target for breast cancer progression. Oncogenesis 2020; 9:105. [PMID: 33311444 PMCID: PMC7732991 DOI: 10.1038/s41389-020-00291-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 11/06/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Recent data indicate that receptor for activated C kinase 1 (RACK1) is a putative prognostic marker and drug target in breast cancer (BC). High RACK1 expression is negatively associated with overall survival, as it seems to promote BC progression. In tumors, RACK1 expression is controlled by a complex balance between glucocorticoids and androgens. Given the fact that androgens and androgenic derivatives can inhibit BC cell proliferation and migration, the role of androgen signaling in regulating RACK1 transcription in mammary tumors is of pivotal interest. Here, we provide evidence that nandrolone (19-nortosterone) inhibits BC cell proliferation and migration by antagonizing the PI3K/Akt/NF-κB signaling pathway, which eventually results in RACK1 downregulation. We also show that nandrolone impairs the PI3K/Akt/NF-κB signaling pathway and decreases RACK1 expression via binding to the membrane-bound receptor, oxoeicosanoid receptor 1 (OXER1). High levels of OXER1 are observed in several BC cell lines and correlate with RACK1 expression and poor prognosis. Our data provide evidence on the role played by the OXER1-dependent intracellular pathway in BC progression and shed light on the mechanisms underlying membrane-dependent androgen effects on RACK1 regulation. Besides the mechanistic relevance, the results of the study are of interest from a translational prospective. In fact, they identify a new and actionable pathway to be used for the design of innovative and rational therapeutic strategies in the context of the personalized treatment of BC. In addition, they draw attention on nandrolone-based compounds that lack hormonal activity as potential anti-tumor agents.
Collapse
|
7
|
Kiely M, Tse LA, Koka H, Wang D, Lee P, Wang F, Wu C, Tsang KH, Chan WC, Law SH, Zhang H, Karlins E, Zhu B, Hutchinson A, Hicks B, Zhu B, Yang XR. Age-related DNA methylation in paired normal and tumour breast tissue in Chinese breast cancer patients. Epigenetics 2020; 16:677-691. [PMID: 32970968 PMCID: PMC8143246 DOI: 10.1080/15592294.2020.1819661] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Age-related DNA methylation is a potential mechanism contributing to breast cancer development. Studies of primarily Caucasian women have identified many CpG sites of age-related methylation in non-diseased breast tissue possibly driving cancer development over time. There is a paucity of studies involving Asian women whose ages at breast cancer onset are usually younger than Caucasians. We identified the 181 most consistent age-related methylation events in non-diseased breast tissue across published studies. Age-related methylation events were measured in adjacent normal and breast tumour tissue in an exclusively Asian population at the previously identified age-related methylation sites. Age-related methylation was found in 118 probes in adjacent normal breast tissue. Methylation of 99% of these sites was increased with age and predominantly located on CpG islands in promoter regions. To ascertain biological relevance to breast cancer, we focused on the 37 sites with overall higher methylation in tumour compared to adjacent normal samples. Some sites positively related to age, including AQP5 and CORO6, inversely correlated with gene expression. Several others have known involvement in suppression of carcinogenesis including GPC5 and SST, suggesting that perturbation of epigenetic regulation at these sites due to ageing may contribute to the progression of carcinogenesis. This study highlights an age-related methylation landscape in non-tumour tissue, consistent not just across studies, but also across different populations. We present candidate age-related methylation sites warranting further investigation as potential epigenetic drivers of breast cancer. They may serve as potential targets of site-specific demethylation intervention strategies for the prevention of age-related breast cancer.
Collapse
Affiliation(s)
- Maeve Kiely
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Lap Ah Tse
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Hela Koka
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Difei Wang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA.,Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Priscilla Lee
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Feng Wang
- Jockey Club School of Public Health and Primary Care, The Chinese University of Hong Kong, Hong Kong, China
| | - Cherry Wu
- North District Hospital, Hong Kong, China
| | | | | | | | - Han Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Eric Karlins
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA.,Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA.,Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA.,Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA.,Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| |
Collapse
|
8
|
Buoso E, Masi M, Long A, Chiappini C, Travelli C, Govoni S, Racchi M. Ribosomes as a nexus between translation and cancer progression: Focus on ribosomal Receptor for Activated C Kinase 1 (RACK1) in breast cancer. Br J Pharmacol 2020; 179:2813-2828. [PMID: 32726469 DOI: 10.1111/bph.15218] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/30/2020] [Accepted: 07/16/2020] [Indexed: 12/19/2022] Open
Abstract
Ribosomes coordinate spatiotemporal control of gene expression, contributing to the acquisition and maintenance of cancer phenotype. The link between ribosomes and cancer is found in the roles of individual ribosomal proteins in tumorigenesis and cancer progression, including the ribosomal protein, receptor for activated C kinase 1 (RACK1). RACK1 regulates cancer cell invasion and is localized in spreading initiation centres, structural adhesion complexes containing RNA binding proteins and poly-adenylated mRNAs that suggest a local translation process. As RACK1 is a ribosomal protein directly involved in translation and in breast cancer progression, we propose a new molecular mechanism for breast cancer cell migration and invasion, which considers the molecular differences between epithelial and mesenchymal cell profiles in order to characterize and provide novel targets for therapeutic strategies. Hence, we provide an analysis on how ribosomes translate cancer progression with a final focus on the ribosomal protein RACK1 in breast cancer.
Collapse
Affiliation(s)
- Erica Buoso
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Mirco Masi
- Department of Drug Sciences, University of Pavia, Pavia, Italy.,Scuola Universitaria Superiore IUSS Pavia, Pavia, Italy
| | - Aideen Long
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College, Dublin, Ireland
| | | | | | - Stefano Govoni
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| | - Marco Racchi
- Department of Drug Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
9
|
Soetje B, Fuellekrug J, Haffner D, Ziegler WH. Application and Comparison of Supervised Learning Strategies to Classify Polarity of Epithelial Cell Spheroids in 3D Culture. Front Genet 2020; 11:248. [PMID: 32292417 PMCID: PMC7119422 DOI: 10.3389/fgene.2020.00248] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/02/2020] [Indexed: 12/16/2022] Open
Abstract
Three-dimensional culture systems that allow generation of monolayered epithelial cell spheroids are widely used to study epithelial function in vitro. Epithelial spheroid formation is applied to address cellular consequences of (mono)-genetic disorders, that is, ciliopathies, in toxicity testing, or to develop treatment options aimed to restore proper epithelial cell characteristics and function. With the potential of a high-throughput method, the main obstacle to efficient application of the spheroid formation assay so far is the laborious, time-consuming, and bias-prone analysis of spheroid images by individuals. Hundredths of multidimensional fluorescence images are blinded, rated by three persons, and subsequently, differences in ratings are compared and discussed. Here, we apply supervised learning and compare strategies based on machine learning versus deep learning. While deep learning approaches can directly process raw image data, machine learning requires transformed data of features extracted from fluorescence images. We verify the accuracy of both strategies on a validation data set, analyse an experimental data set, and observe that different strategies can be very accurate. Deep learning, however, is less sensitive to overfitting and experimental batch-to-batch variations, thus providing a rather powerful and easily adjustable classification tool.
Collapse
Affiliation(s)
- Birga Soetje
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hanover, Germany
| | - Joachim Fuellekrug
- Molecular Cell Biology Laboratory, Internal Medicine IV, University Hospital Heidelberg, Heidelberg, Germany
| | - Dieter Haffner
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hanover, Germany
| | - Wolfgang H. Ziegler
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hanover, Germany
| |
Collapse
|
10
|
Cui L, Cheng Z, Hu K, Pang Y, Liu Y, Qian T, Quan L, Dai Y, Pang Y, Ye X, Shi J, Fu L. Prognostic value of the PDLIM family in acute myeloid leukemia. Am J Transl Res 2019; 11:6124-6131. [PMID: 31632581 PMCID: PMC6789254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
Acute myeloid leukemia (AML) is a genetically complex, highly aggressive hematological malignancy. Prognosis is usually with grim. PDZ and LIM domain proteins (PDLIM) are involved in the regulation of a variety of biological processes, including cytoskeletal organization, cell differentiation, organ development, neural signaling or tumorigenesis. The clinical and prognostic value of the PDLIM family in AML is unclear. To understand the role of PDLIM expression in AML, The Cancer Genome Atlas (TCGA) database was screened and 155 de novo AML patients with complete clinical information and the expression data of the PDLIM family were included in the study. The clinical and molecular characteristics associated with the expression of different members of the PDLIM family were summarized using various statistical methods. In 84 patients who only received chemotherapy, univariate analysis indicated that high expression of PDLIM2 or PDLIM7 was associated with shorter EFS and OS (both P<0.05 for PDLIM2, and both P<0.01 for PDLIM7). Multivariate analysis suggested that high expression of PDLIM7 was an independent risk factor for EFS and OS (both P<0.05). In the other 71 patients who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT), survival was unaffected by PDLIM expressions. In summary, high expression of PDLIM2 and PDLIM7, especially the latter, could serve as adverse prognostic factors for AML, but their prognostic effects could be reversed by allo-HSCT.
Collapse
Affiliation(s)
- Longzhen Cui
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Translational Medicine Center, Huaihe Hospital of Henan UniversityKaifeng 475000, Henan, China
- Department of Hematology, Huaihe Hospital of Henan UniversityKaifeng 475000, Henan, China
| | - Zhiheng Cheng
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Kai Hu
- Department of Hematology and Lymphoma Research Center, Peking University, Third HospitalBeijing 100191, China
| | - Yifan Pang
- Department of Medicine, William Beaumont HospitalRoyal Oak, MI 48073, USA
| | - Yan Liu
- Translational Medicine Center, Huaihe Hospital of Henan UniversityKaifeng 475000, Henan, China
| | - Tingting Qian
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Liang Quan
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Yifeng Dai
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of GroningenGroningen, Netherlands
| | - Ying Pang
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Xu Ye
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
| | - Jinlong Shi
- Department of Medical Big Data, Chinese PLA General HospitalBeijing 100853, China
| | - Lin Fu
- Department of Hematology, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhou 510260, Guangdong, China
- Department of Hematology, Huaihe Hospital of Henan UniversityKaifeng 475000, Henan, China
| |
Collapse
|
11
|
Cox OT, Edmunds SJ, Simon-Keller K, Li B, Moran B, Buckley NE, Bustamante-Garrido M, Healy N, O'Flanagan CH, Gallagher WM, Kennedy RD, Bernards R, Caldas C, Chin SF, Marx A, O'Connor R. PDLIM2 Is a Marker of Adhesion and β-Catenin Activity in Triple-Negative Breast Cancer. Cancer Res 2019; 79:2619-2633. [PMID: 30885980 DOI: 10.1158/0008-5472.can-18-2787] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 01/09/2019] [Accepted: 03/12/2019] [Indexed: 11/16/2022]
Abstract
The PDLIM2 protein regulates stability of transcription factors including NF-κB and STATs in epithelial and hemopoietic cells. PDLIM2 is strongly expressed in certain cancer cell lines that exhibit an epithelial-to-mesenchymal phenotype, and its suppression is sufficient to reverse this phenotype. PDLIM2 supports the epithelial polarity of nontransformed breast cells, suggesting distinct roles in tumor suppression and oncogenesis. To better understand its overall function, we investigated PDLIM2 expression and activity in breast cancer. PDLIM2 protein was present in 60% of tumors diagnosed as triple-negative breast cancer (TNBC), and only 20% of other breast cancer subtypes. High PDLIM2 expression in TNBC was positively correlated with adhesion signaling and β-catenin activity. Interestingly, PDLIM2 was restricted to the cytoplasm/membrane of TNBC cells and excluded from the nucleus. In breast cell lines, PDLIM2 retention in the cytoplasm was controlled by cell adhesion, and translocation to the nucleus was stimulated by insulin-like growth factor-1 or TGFβ. Cytoplasmic PDLIM2 was associated with active β-catenin and ectopic expression of PDLIM2 was sufficient to increase β-catenin levels and its transcriptional activity in reporter assays. Suppression of PDLIM2 inhibited tumor growth in vivo, whereas overexpression of PDLIM2 disrupted growth in 3D cultures. These results suggest that PDLIM2 may serve as a predictive biomarker for a large subset of TNBC whose phenotype depends on adhesion-regulated β-catenin activity and which may be amenable to therapies that target these pathways. SIGNIFICANCE: This study shows that PDLIM2 expression defines a subset of triple-negative breast cancer that may benefit from targeting the β-catenin and adhesion signaling pathways. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/10/2619/F1.large.jpg.
Collapse
Affiliation(s)
- Orla T Cox
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Shelley J Edmunds
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Katja Simon-Keller
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Germany
| | - Bo Li
- School of Biomolecular & Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Bruce Moran
- School of Biomolecular & Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Niamh E Buckley
- School of Pharmacy, Queens University Belfast, Belfast, Northern Ireland
| | - Milan Bustamante-Garrido
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Nollaig Healy
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Ciara H O'Flanagan
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - William M Gallagher
- School of Biomolecular & Biomedical Science, Conway Institute, University College Dublin, Dublin, Ireland
| | - Richard D Kennedy
- Centre for Cancer Research and Cell Biology, Queens University Belfast, Northern Ireland
| | - René Bernards
- Division of Molecular Carcinogenesis and Cancer Genomics Netherlands, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Carlos Caldas
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Suet-Feung Chin
- Cancer Research UK Cambridge Research Institute, Li Ka Shing Centre, Cambridge, UK
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Germany
| | - Rosemary O'Connor
- Cell Biology Laboratory, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland.
| |
Collapse
|
12
|
Zhang B, Xie F, Aziz AUR, Shao S, Li W, Deng S, Liao X, Liu B. Heat Shock Protein 27 Phosphorylation Regulates Tumor Cell Migration under Shear Stress. Biomolecules 2019; 9:biom9020050. [PMID: 30704117 PMCID: PMC6406706 DOI: 10.3390/biom9020050] [Citation(s) in RCA: 8] [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: 10/25/2018] [Revised: 01/25/2019] [Accepted: 01/25/2019] [Indexed: 01/02/2023] Open
Abstract
Heat shock protein 27 (HSP27) is a multifunctional protein that undergoes significant changes in its expression and phosphorylation in response to shear stress stimuli, suggesting that it may be involved in mechanotransduction. However, the mechanism of HSP27 affecting tumor cell migration under shear stress is still not clear. In this study, HSP27-enhanced cyan fluorescent protein (ECFP) and HSP27-Ypet plasmids are constructed to visualize the self-polymerization of HSP27 in living cells based on fluorescence resonance energy transfer technology. The results show that shear stress induces polar distribution of HSP27 to regulate the dynamic structure at the cell leading edge. Shear stress also promotes HSP27 depolymerization to small molecules and then regulates polar actin accumulation and focal adhesion kinase (FAK) polar activation, which further promotes tumor cell migration. This study suggests that HSP27 plays an important role in the regulation of shear stress-induced HeLa cell migration, and it also provides a theoretical basis for HSP27 as a potential drug target for metastasis.
Collapse
Affiliation(s)
- Baohong Zhang
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Fei Xie
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Aziz Ur Rehman Aziz
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Shuai Shao
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Wang Li
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Sha Deng
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| | - Xiaoling Liao
- Institute of Biomedical Engineering, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Bo Liu
- School of Biomedical Engineering, Dalian University of Technology, Liaoning IC Technology Key Lab, Dalian 116024, China.
| |
Collapse
|
13
|
Wu WM, Liao YC. Downregulation of C-Terminal Tensin-Like Protein (CTEN) Suppresses Prostate Cell Proliferation and Contributes to Acinar Morphogenesis. Int J Mol Sci 2018; 19:ijms19103190. [PMID: 30332774 PMCID: PMC6214133 DOI: 10.3390/ijms19103190] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 10/09/2018] [Accepted: 10/12/2018] [Indexed: 12/20/2022] Open
Abstract
C-terminal tensin-like protein (CTEN) is a member of tensin family, which is crucial for the assembly of cell-matrix adhesome. Unlike other tensins, CTEN is selectively expressed only in a few tissues such as the prostate. However, the biological relevance of CTEN in normal prostate is poorly understood. In this study, we revealed that CTEN is selectively expressed in the prostate epithelial cells and enriched in the basal compartment. Knockdown of CTEN in RWPE-1 cells suppresses cell proliferation and results in G1/S cell cycle arrest as well as the accumulation of cyclin-dependent kinase (CDK) inhibitors, p21 and p27. Moreover, the expression of CTEN is decreased during acinar morphogenesis using Matrigel-based three-dimensional (3D) culture. In the course of acinar formation, induction of CTEN reactivates focal adhesion kinase (FAK) Y397 phosphorylation and disrupts the acini structure. This study, to our knowledge, is the first report demonstrating that downregulation of CTEN is required for luminal differentiation and acinar formation.
Collapse
Affiliation(s)
- Wei-Ming Wu
- Department of Biochemical Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan.
| | - Yi-Chun Liao
- Department of Biochemical Science and Technology, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan.
| |
Collapse
|
14
|
Stanicka J, Rieger L, O’Shea S, Cox O, Coleman M, O’Flanagan C, Addario B, McCabe N, Kennedy R, O’Connor R. FES-related tyrosine kinase activates the insulin-like growth factor-1 receptor at sites of cell adhesion. Oncogene 2018. [DOI: 10.1038/s41388-017-0113-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
15
|
Kamal M, Holliday DL, Morrison EE, Speirs V, Toomes C, Bell SM. Loss of CSMD1 expression disrupts mammary duct formation while enhancing proliferation, migration and invasion. Oncol Rep 2017; 38:283-292. [PMID: 28534981 DOI: 10.3892/or.2017.5656] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
Abstract
The CUB and sushi multiple domains 1 (CSMD1) gene maps to chromosome 8p23, a region deleted in many cancers. Loss of CSMD1 expression is associated with poor prognosis in breast cancer suggesting that it acts as a tumour suppressor in this cancer. However, the function of CSMD1 is largely unknown. Herein, we investigated CSMD1 functions in cell line models. CSMD1 expression was suppressed in MCF10A and LNCaP cells using short hairpin RNA. Functional assays were performed focusing on the 'normal' MCF10A cell line. Suppression of CSMD1 significantly increased the proliferation, cell migration and invasiveness of MCF10A cells compared to shcontrols. shCSMD1 cells also showed significantly reduced adhesion to Matrigel and fibronectin. In a three-dimensional Matrigel model of MCF10A cells, reduced CSMD1 expression resulted in the development of larger and more poorly differentiated breast acini-like structures that displayed impaired lumen formation. Loss of CSMD1 expression disrupts a model of mammary duct formation while enhancing proliferation, migration and invasion. Our data suggest that CSMD1 is involved in the suppression of a transformed phenotype.
Collapse
Affiliation(s)
- Mohamed Kamal
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Deborah L Holliday
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Ewan E Morrison
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Valerie Speirs
- Leeds Institute of Cancer and Pathology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Carmel Toomes
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| | - Sandra M Bell
- Leeds Institute of Biomedical and Clinical Sciences, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds LS9 7TF, UK
| |
Collapse
|
16
|
Caromile LA, Dortche K, Rahman MM, Grant CL, Stoddard C, Ferrer FA, Shapiro LH. PSMA redirects cell survival signaling from the MAPK to the PI3K-AKT pathways to promote the progression of prostate cancer. Sci Signal 2017; 10:10/470/eaag3326. [PMID: 28292957 DOI: 10.1126/scisignal.aag3326] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Increased abundance of the prostate-specific membrane antigen (PSMA) on prostate epithelium is a hallmark of advanced metastatic prostate cancer (PCa) and correlates negatively with prognosis. However, direct evidence that PSMA functionally contributes to PCa progression remains elusive. We generated mice bearing PSMA-positive or PSMA-negative PCa by crossing PSMA-deficient mice with transgenic PCa (TRAMP) models, enabling direct assessment of PCa incidence and progression in the presence or absence of PSMA. Compared with PSMA-positive tumors, PSMA-negative tumors were smaller, lower-grade, and more apoptotic with fewer blood vessels, consistent with the recognized proangiogenic function of PSMA. Relative to PSMA-positive tumors, tumors lacking PSMA had less than half the abundance of type 1 insulin-like growth factor receptor (IGF-1R), less activity in the survival pathway mediated by PI3K-AKT signaling, and more activity in the proliferative pathway mediated by MAPK-ERK1/2 signaling. Biochemically, PSMA interacted with the scaffolding protein RACK1, disrupting signaling between the β1 integrin and IGF-1R complex to the MAPK pathway, enabling activation of the AKT pathway instead. Manipulation of PSMA abundance in PCa cell lines recapitulated this signaling pathway switch. Analysis of published databases indicated that IGF-1R abundance, cell proliferation, and expression of transcripts for antiapoptotic markers positively correlated with PSMA abundance in patients, suggesting that this switch may be relevant to human PCa. Our findings suggest that increase in PSMA in prostate tumors contributes to progression by altering normal signal transduction pathways to drive PCa progression and that enhanced signaling through the IGF-1R/β1 integrin axis may occur in other tumors.
Collapse
Affiliation(s)
- Leslie Ann Caromile
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Kristina Dortche
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - M Mamunur Rahman
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Christina L Grant
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Christopher Stoddard
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Fernando A Ferrer
- Department of Urology, New York Medical College, Valhalla, NY 10595, USA
| | - Linda H Shapiro
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
17
|
Bassiri K, Ferluga S, Sharma V, Syed N, Adams CL, Lasonder E, Hanemann CO. Global Proteome and Phospho-proteome Analysis of Merlin-deficient Meningioma and Schwannoma Identifies PDLIM2 as a Novel Therapeutic Target. EBioMedicine 2017; 16:76-86. [PMID: 28126595 PMCID: PMC5474504 DOI: 10.1016/j.ebiom.2017.01.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Revised: 01/13/2017] [Accepted: 01/13/2017] [Indexed: 12/20/2022] Open
Abstract
Loss or mutation of the tumour suppressor Merlin predisposes individuals to develop multiple nervous system tumours, including schwannomas and meningiomas, sporadically or as part of the autosomal dominant inherited condition Neurofibromatosis 2 (NF2). These tumours display largely low grade features but their presence can lead to significant morbidity. Surgery and radiotherapy remain the only treatment options despite years of research, therefore an effective therapeutic is required. Unbiased omics studies have become pivotal in the identification of differentially expressed genes and proteins that may act as drug targets or biomarkers. Here we analysed the proteome and phospho-proteome of these genetically defined tumours using primary human tumour cells to identify upregulated/activated proteins and/or pathways. We identified over 2000 proteins in comparative experiments between Merlin-deficient schwannoma and meningioma compared to human Schwann and meningeal cells respectively. Using functional enrichment analysis we highlighted several dysregulated pathways and Gene Ontology terms. We identified several proteins and phospho-proteins that are more highly expressed in tumours compared to controls. Among proteins jointly dysregulated in both tumours we focused in particular on PDZ and LIM domain protein 2 (PDLIM2) and validated its overexpression in several tumour samples, while not detecting it in normal cells. We showed that shRNA mediated knockdown of PDLIM2 in both primary meningioma and schwannoma leads to significant reductions in cellular proliferation. To our knowledge, this is the first comprehensive assessment of the NF2-related meningioma and schwannoma proteome and phospho-proteome. Taken together, our data highlight several commonly deregulated factors, and indicate that PDLIM2 may represent a novel, common target for meningioma and schwannoma. Proteome and phosphoproteome of Merlin-deficient schwannomas and meningiomas were analysed. Comparative studies highlighted several pathways relevant for therapeutic intervention. PDLIM2 was identified as a novel, commonly upregulated protein in both tumours. PDLIM2 knockdown led to a significant reduction in proliferation in both cell types.
Loss or mutation of the protein Merlin causes a genetic condition known as Neurofibromatosis 2 (NF2) characterised by the growth of schwannomas and meningiomas. We analysed several of these tumour samples and identified over 2000 proteins in comparative experiments between Merlin-deficient schwannoma and meningioma compared to normal controls. We identified PDZ and LIM domain protein 2 (PDLIM2) as overexpressed in both tumour types and further showed that knockdown of PDLIM2 leads to significant reductions in cellular proliferation. Taken together, our data highlight several deregulated signalling pathways, and indicate that PDLIM2 may represent a novel, common target for meningioma and schwannoma.
Collapse
Affiliation(s)
- Kayleigh Bassiri
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Research Way, Derriford, Plymouth PL6 8BU, UK
| | - Sara Ferluga
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Research Way, Derriford, Plymouth PL6 8BU, UK
| | - Vikram Sharma
- School of Biomedical and Healthcare Sciences, Plymouth University, Drakes Circus, Plymouth PL4 8AA, UK
| | - Nelofer Syed
- John Fulcher Neuro-oncology Laboratory, Division of Brain Sciences, Faculty of Medicine, Imperial College London, London W6 8RP, UK
| | - Claire L Adams
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Research Way, Derriford, Plymouth PL6 8BU, UK
| | - Edwin Lasonder
- School of Biomedical and Healthcare Sciences, Plymouth University, Drakes Circus, Plymouth PL4 8AA, UK
| | - C Oliver Hanemann
- Institute of Translational and Stratified Medicine, Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Research Way, Derriford, Plymouth PL6 8BU, UK.
| |
Collapse
|
18
|
Dowling CM, Walsh D, Coffey JC, Kiely PA. The importance of selecting the appropriate reference genes for quantitative real time PCR as illustrated using colon cancer cells and tissue. F1000Res 2016. [PMID: 26962435 DOI: 10.12688/f1000research.7656.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Quantitative real-time reverse-transcription polymerase chain reaction (RT-qPCR) remains the most sensitive technique for nucleic acid quantification. Its popularity is reflected in the remarkable number of publications reporting RT-qPCR data. Careful normalisation within RT-qPCR studies is imperative to ensure accurate quantification of mRNA levels. This is commonly achieved through the use of reference genes as an internal control to normalise the mRNA levels between different samples. The selection of appropriate reference genes can be a challenge as transcript levels vary with physiology, pathology and development, making the information within the transcriptome flexible and variable. In this study, we examined the variation in expression of a panel of nine candidate reference genes in HCT116 and HT29 2-dimensional and 3-dimensional cultures, as well as in normal and cancerous colon tissue. Using normfinder we identified the top three most stable genes for all conditions. Further to this we compared the change in expression of a selection of PKC coding genes when the data was normalised to one reference gene and three reference genes. Here we demonstrated that there is a variation in the fold changes obtained dependent on the number of reference genes used. As well as this, we highlight important considerations namely; assay efficiency tests, inhibition tests and RNA assessment which should also be implemented into all RT-qPCR studies. All this data combined demonstrates the need for careful experimental design in RT-qPCR studies to help eliminate false interpretation and reporting of results.
Collapse
Affiliation(s)
- Catríona M Dowling
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland; Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Dara Walsh
- 4i Centre for Interventions in Infection, Inflammation and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - John C Coffey
- 4i Centre for Interventions in Infection, Inflammation and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Patrick A Kiely
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland; Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| |
Collapse
|
19
|
Kang M, Lee KH, Lee HS, Park YH, Jeong CW, Ku JH, Kim HH, Kwak C. PDLIM2 suppression efficiently reduces tumor growth and invasiveness of human castration-resistant prostate cancer-like cells. Prostate 2016; 76:273-285. [PMID: 26499308 DOI: 10.1002/pros.23118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2015] [Accepted: 10/14/2015] [Indexed: 11/07/2022]
Abstract
BACKGROUND Although PDLIM2 gene may have a context-dependent role in various human malignancies and can be a potential therapeutic target, only a limited number of in vitro studies addressed the molecular functions of PDLIM2 in prostate cancer. Here, we aimed to explore the role of PDLIM2 and the effect of the PDLIM2 gene suppression on oncogenic phenotypes of human castration-resistant prostate cancer (CRPC)-like cells. METHODS We used human CRPC-like cell lines (PC3, DU145, and C4-2B) for our experiments. Transcription levels of PDLIM2 and relevant genes were measured by real time-PCR and protein expression was analyzed by western blot. Cell viability, proliferation, clonogenic growth, and tumor sphere formation were examined after a specific inhibition of PDLIM2 using RNA interference. Flow cytometry was used to examine apoptotic cell death and cell cycle disturbances. Wound healing and transwell migration assays were performed to investigate the invasion capabilities of CRPC-like cells. Additionally, key oncogenic signaling pathways were examined using western blot. Lastly, we evaluated the in vivo efficacy of PDLIM2 suppression on tumor growth of human CRPC xenografts in mice. RESULTS We observed a significant enhancement of PDLIM2 expression in human CRPC-like cell lines, while a specific inhibition of PDLIM2 reduced cell viability and proliferation due to apoptotic cell death. Conversely, PDLIM2 overexpression significantly reduced cell proliferation compared to the negative control in androgen-sensitive LNCaP cells. Moreover, PDLIM2 suppression led to a decrease of clonogenic growth and tumor sphere formation in three-dimensional cultures with the G2/M cell cycle arrest in human CRPC-like cells. PDLIM2 inhibition also attenuated cellular migration and invasion capabilities of human CRPC-like cells, and reduced the expression of mesenchymal marker. Among several oncogenic signaling pathways, only the MAPK/ERK signaling cascade was decreased by PDLIM2 inhibition and reciprocally, ERK inhibition down-regulated PDLIM2 expression. Importantly, PDLIM2 inhibition remarkably compromised tumor growth in a human CRPC xenograft model. CONCLUSION In summary, the suppression of PDLIM2 significantly reduced such oncogenic phenotypes as proliferation, clonogenicity, invasiveness, and tumor cell growth in human CRPC-like cells both in vitro and in vivo, indicating that PDLIM2 may be considered a novel therapeutic target gene for treating human CRPC.
Collapse
Affiliation(s)
- Minyong Kang
- Department of Urology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyoung-Hwa Lee
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Hye Sun Lee
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Yong Hyun Park
- Department of Urology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chang Wook Jeong
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Ja Hyeon Ku
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Hyeon Hoe Kim
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| | - Cheol Kwak
- Department of Urology, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
20
|
Dowling CM, Walsh D, Coffey JC, Kiely PA. The importance of selecting the appropriate reference genes for quantitative real time PCR as illustrated using colon cancer cells and tissue. F1000Res 2016; 5:99. [PMID: 26962435 PMCID: PMC4768652 DOI: 10.12688/f1000research.7656.2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/04/2016] [Indexed: 12/16/2022] Open
Abstract
Quantitative real-time reverse-transcription polymerase chain reaction (RT-qPCR) remains the most sensitive technique for nucleic acid quantification. Its popularity is reflected in the remarkable number of publications reporting RT-qPCR data. Careful normalisation within RT-qPCR studies is imperative to ensure accurate quantification of mRNA levels. This is commonly achieved through the use of reference genes as an internal control to normalise the mRNA levels between different samples. The selection of appropriate reference genes can be a challenge as transcript levels vary with physiology, pathology and development, making the information within the transcriptome flexible and variable. In this study, we examined the variation in expression of a panel of nine candidate reference genes in HCT116 and HT29 2-dimensional and 3-dimensional cultures, as well as in normal and cancerous colon tissue. Using normfinder we identified the top three most stable genes for all conditions. Further to this we compared the change in expression of a selection of PKC coding genes when the data was normalised to one reference gene and three reference genes. Here we demonstrated that there is a variation in the fold changes obtained dependent on the number of reference genes used. As well as this, we highlight important considerations namely; assay efficiency tests, inhibition tests and RNA assessment which should also be implemented into all RT-qPCR studies. All this data combined demonstrates the need for careful experimental design in RT-qPCR studies to help eliminate false interpretation and reporting of results.
Collapse
Affiliation(s)
- Catríona M Dowling
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland; Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Dara Walsh
- 4i Centre for Interventions in Infection, Inflammation and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - John C Coffey
- 4i Centre for Interventions in Infection, Inflammation and Immunity, Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| | - Patrick A Kiely
- Department of Life Sciences, and Materials and Surface Science Institute, University of Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland; Graduate Entry Medical School, University of Limerick, Limerick, Ireland
| |
Collapse
|
21
|
Cox OT, O’Shea S, Tresse E, Bustamante-Garrido M, Kiran-Deevi R, O’Connor R. IGF-1 Receptor and Adhesion Signaling: An Important Axis in Determining Cancer Cell Phenotype and Therapy Resistance. Front Endocrinol (Lausanne) 2015; 6:106. [PMID: 26191041 PMCID: PMC4490239 DOI: 10.3389/fendo.2015.00106] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 06/19/2015] [Indexed: 11/13/2022] Open
Abstract
IGF-1R expression and activation levels generally cannot be correlated in cancer cells, suggesting that cellular proteins may modulate IGF-1R activity. Strong candidates for such modulation are found in cell-matrix and cell-cell adhesion signaling complexes. Activated IGF-1R is present at focal adhesions, where it can stabilize β1 integrin and participate in signaling complexes that promote invasiveness associated with epithelial mesenchymal transition (EMT) and resistance to therapy. Whether IGF-1R contributes to EMT or to non-invasive tumor growth may be strongly influenced by the degree of extracellular matrix engagement and the presence or absence of key proteins in IGF-1R-cell adhesion complexes. One such protein is PDLIM2, which promotes both cell polarization and EMT by regulating the stability of transcription factors including NFκB, STATs, and beta catenin. PDLIM2 exhibits tumor suppressor activity, but is also highly expressed in certain invasive cancers. It is likely that distinct adhesion complex proteins modulate IGF-1R signaling during cancer progression or adaptive responses to therapy. Thus, identifying the key modulators will be important for developing effective therapeutic strategies and predictive biomarkers.
Collapse
Affiliation(s)
- Orla T. Cox
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Sandra O’Shea
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Emilie Tresse
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Milan Bustamante-Garrido
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Ravi Kiran-Deevi
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
| | - Rosemary O’Connor
- Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland
- *Correspondence: Rosemary O’Connor, Cell Biology Laboratory, BioSciences Institute, School of Biochemistry and Cell Biology, University College Cork, Cork, Ireland,
| |
Collapse
|