1
|
Wang J, Li Z, Zhao Q. Receptor tyrosine kinase-like orphan receptor serves as a potential target in cancer immunotherapy. J Leukoc Biol 2025; 117:qiae141. [PMID: 38973261 DOI: 10.1093/jleuko/qiae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 05/16/2024] [Indexed: 07/09/2024] Open
Abstract
Receptor tyrosine kinase-like orphan receptor (ROR), consisting of ROR1 and ROR2, is a conserved family of receptor tyrosine kinase superfamily that plays crucial roles during embryonic development with limited expression in adult normal tissues. However, it is overexpressed in a range of hematological malignancies and solid tumors and functions in cellular processes including cell survival, polarity, and migration, serving as a potential target in cancer immunotherapy. This review summarizes the expression and structure of ROR in developmental morphogenesis and its function in cancers associated with Wnt5a signaling and highlights the cancer immunotherapy strategies targeting ROR.
Collapse
Affiliation(s)
- Jiaqi Wang
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
| | - Zhoufang Li
- Core Research Facilities, Southern University of Science and Technology, Shenzhen 518055, China
| | - Qi Zhao
- Cancer Centre, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR 999078, China
| |
Collapse
|
2
|
Leng F, Huang J, Wu L, Zhang J, Lin X, Deng R, Zhu J, Li Z, Li Z, Wang Y, Zhang H, Lu D, Kipps TJ, Zhang S. Targeting ROR2 homooligomerization disrupts ROR2-dependent signaling and suppresses stem-like cell properties of human breast adenocarcinoma. iScience 2025; 28:111589. [PMID: 39829682 PMCID: PMC11742321 DOI: 10.1016/j.isci.2024.111589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 01/22/2025] Open
Abstract
Breast cancer stem-like cells (CSCs) are enriched following treatment with chemotherapy, and posited as having a high level of plasticity and enhanced tumor-initiation capacity, which can enable cancer relapse. Here, we show that such features are shared by breast cancer (BCA) cells that express receptor tyrosine kinase-like orphan receptor (ROR2), which is expressed primarily during embryogenesis and by various cancers. We find that Wnt5a can induce ROR2 homooligomerization to activate noncanonical Wnt signaling and enhance tumor-initiation capacity of BCA cells. Molecular analysis reveals that the cysteine-rich domain and transmembrane domain are required for ROR2 homooligomerization to activate ROR2. Treatment with a newly generated monoclonal antibody (mAb) specific for ROR2 can block Wnt5a-induced ROR2 homooligomerization, ROR2-dependent noncanonical Wnt signaling, and impair the capacity of BCA patient-derived xenografts to initiate tumor in immune-deficient mice. Collectively, these results indicate that targeting ROR2 (e.g., using mAb) suppresses BCA stemness and, thereby, may prevent BCA relapse.
Collapse
Affiliation(s)
- Feng Leng
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jiajia Huang
- State Key Laboratory of Oncology in South China, Department of Medical Oncology, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Liufeng Wu
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jianchao Zhang
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Xinxin Lin
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Ruhuan Deng
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Jinhang Zhu
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Zhen Li
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Zhenghao Li
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Yimeng Wang
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Han Zhang
- Xenta Biomedical Science Co., Ltd, Guangzhou 510060, China
| | - Desheng Lu
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Thomas J. Kipps
- Moores Cancer Center, University of California, San Diego, San Diego, CA 92037, USA
| | - Suping Zhang
- Shenzhen Key Laboratory of Precision Medicine for Hematological Malignancies, Guangdong Key Laboratory for Genome Stability and Human Disease Prevention, Department of Pharmacology, School of Basic Medical Sciences, Base for International Science and Technology Cooperation: Carson Cancer Stem Cell Vaccines R&D Center, International Cancer Center, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
- Moores Cancer Center, University of California, San Diego, San Diego, CA 92037, USA
| |
Collapse
|
3
|
Zhou H, Zhang S, Jin X, A C, Gong P, Zhao S. The Electric Field Guided HaCaT Cell Migration Through the EGFR/p38 MAPK/Akt Pathway. Curr Issues Mol Biol 2024; 47:16. [PMID: 39852131 PMCID: PMC11763975 DOI: 10.3390/cimb47010016] [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: 11/21/2024] [Revised: 12/16/2024] [Accepted: 12/28/2024] [Indexed: 01/26/2025] Open
Abstract
Previous studies have shown that the endogenous electric field (EF) is an overriding cure in guiding cell migration toward the wound center to promote wound healing, but the mechanism underlying is unclear. In this study, we investigated the molecular mechanism of electric field-guided cell migration in human keratinocyte HaCaT cells. Our results showed that HaCaT cells migrate toward the anode under EFs. The phosphorylation levels of p38 MAPK and Akt were obviously elevated in the EF. Knocking down p38 MAPK obviously abolished directed migration of HaCaT cells under the EFs. Inhibiting p38 MAPK by SB203580 impaired the EF-guided cell migration. The electric field may guide HaCaT cell migration through the EGFR/p38 MAPK/Akt pathway.
Collapse
Affiliation(s)
| | | | | | | | - Peng Gong
- School of Life Sciences, Yunnan Normal University, Kunming 650500, China; (H.Z.); (S.Z.); (X.J.); (C.A.)
| | - Sanjun Zhao
- School of Life Sciences, Yunnan Normal University, Kunming 650500, China; (H.Z.); (S.Z.); (X.J.); (C.A.)
| |
Collapse
|
4
|
Kamizaki K, Katsukawa M, Yamamoto A, Fukada SI, Uezumi A, Endo M, Minami Y. Ror2 signaling regulated by differential Wnt proteins determines pathological fate of muscle mesenchymal progenitors. Cell Death Dis 2024; 15:784. [PMID: 39468010 PMCID: PMC11519583 DOI: 10.1038/s41419-024-07173-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 10/30/2024]
Abstract
Skeletal muscle mesenchymal progenitors (MPs) play a critical role in supporting muscle regeneration. However, under pathological conditions, they contribute to intramuscular adipose tissue accumulation, involved in muscle diseases, including muscular dystrophy and sarcopenia, age-related muscular atrophy. How MP fate is determined in these different contexts remains unelucidated. Here, we report that Ror2, a non-canonical Wnt signaling receptor, is selectively expressed in MPs and regulates their pathological features in a differential ligand-dependent manner. We identified Wnt11 and Wnt5b as ligands of Ror2. In vitro, Wnt11 inhibited MP senescence, which is required for normal muscle regeneration, and Wnt5b promoted MP proliferation. We further found that both Wnts are abundant in degenerating muscle and synergistically stimulate Ror2, leading to unwanted MP proliferation and eventually intramuscular adipose tissue accumulation. These findings provide evidence that Ror2-mediated signaling elicited by differential Wnts plays a critical role in determining the pathological fate of MPs.
Collapse
Affiliation(s)
- Koki Kamizaki
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Mitsuko Katsukawa
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Ayano Yamamoto
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - So-Ichiro Fukada
- Laboratory of Stem Cell Regeneration and Adaptation, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
| | - Akiyoshi Uezumi
- Division of Cell Heterogeneity, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mitsuharu Endo
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Yasuhiro Minami
- Division of Cell Physiology, Department of Physiology and Cell Biology, Graduate School of Medicine, Kobe University, Kobe, Japan.
| |
Collapse
|
5
|
Piastra V, Ganci F, Sacconi A, Pranteda A, Allegretti M, Bernardini R, Serra M, Lupo B, Dell'Aquila E, Ferretti G, Pescarmona E, Bartolazzi A, Blandino G, Trusolino L, Bossi G. Repurposed AT9283 triggers anti-tumoral effects by targeting MKK3 oncogenic functions in Colorectal Cancer. J Exp Clin Cancer Res 2024; 43:234. [PMID: 39164711 PMCID: PMC11334304 DOI: 10.1186/s13046-024-03150-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 08/04/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer-related deaths worldwide, with a survival rate near to 10% when diagnosed at an advanced stage. Hence, the identification of new molecular targets to design more selective and efficient therapies is urgently required. The Mitogen activated protein kinase kinase 3 (MKK3) is a dual-specificity threonine/tyrosine protein kinase that, activated in response to cellular stress and inflammatory stimuli, regulates a plethora of biological processes. Previous studies revealed novel MKK3 roles in supporting tumor malignancy, as its depletion induces autophagy and cell death in cancer lines of different tumor types, including CRC. Therefore, MKK3 may represent an interesting new therapeutic target in advanced CRC, however selective MKK3 inhibitors are currently not available. METHODS The study involved transcriptomic based drug repurposing approach and confirmatory assays with CRC lines, primary colonocytes and a subset of CRC patient-derived organoids (PDO). Investigations in vitro and in vivo were addressed. RESULTS The repurposing approach identified the multitargeted kinase inhibitor AT9283 as a putative compound with MKK3 depletion-mimicking activities. Indeed, AT9283 drops phospho- and total-MKK3 protein levels in tested CRC models. Likely the MKK3 silencing, AT9283 treatment: i) inhibited cell proliferation promoting autophagy and cell death in tested CRC lines and PDOs; ii) resulted well-tolerated by CCD-18Co colonocytes; iii) reduced cancer cell motility inhibiting CRC cell migration and invasion; iv) inhibited COLO205 xenograft tumor growth. Mechanistically, AT9283 abrogated MKK3 protein levels mainly through the inhibition of aurora kinase A (AURKA), impacting on MKK3/AURKA protein-protein interaction and protein stability therefore uncovering the relevance of MKK3/AURKA crosstalk in sustaining CRC malignancy in vitro and in vivo. CONCLUSION Overall, we demonstrated that the anti-tumoral effects triggered by AT9283 treatment recapitulated the MKK3 depletion effects in all tested CRC models in vitro and in vivo, suggesting that AT9283 is a repurposed drug. According to its good tolerance when tested with primary colonocytes (CCD-18CO), AT9283 is a promising drug for the development of novel therapeutic strategies to target MKK3 oncogenic functions in late-stage and metastatic CRC patients.
Collapse
Affiliation(s)
- Valentina Piastra
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Department of Science, University Roma Tre, Rome, Italy
| | - Federica Ganci
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Andrea Sacconi
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Angelina Pranteda
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Department of Science, University Roma Tre, Rome, Italy
| | - Matteo Allegretti
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Roberta Bernardini
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture (CIMETA), University of Rome Tor Vergata, Rome, Italy
| | - Martina Serra
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
- Interdepartmental Center for Comparative Medicine, Alternative Techniques and Aquaculture (CIMETA), University of Rome Tor Vergata, Rome, Italy
| | - Barbara Lupo
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Emanuela Dell'Aquila
- Second Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Gianluigi Ferretti
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Armando Bartolazzi
- Pathology Research Laboratory, St Andrea University Hospital, Rome, Italy
| | - Giovanni Blandino
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy
| | - Livio Trusolino
- Candiolo Cancer Institute - FPO IRCCS, Candiolo, Turin, Italy
- Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Gianluca Bossi
- Translational Oncology Research Unit, Department of Diagnostic Research and Technological Innovation, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, Rome, 53 - 00144, Italy.
| |
Collapse
|
6
|
Hong JW, Yu Y, Wang LS, Li Z, Zhang R, Wang Q, Ding Z, Zhang JP, Zhang MR, Xu LC. BMP4 Regulates EMT to be Involved in non-Syndromic Cleft lip With or Without Palate. Cleft Palate Craniofac J 2023; 60:1462-1473. [PMID: 35702016 DOI: 10.1177/10556656221105762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE In the previous study, we identified bone morphogenetic protein 4 (BMP4) responsible for non-syndromic cleft lip with or without cleft palate (NSCL/P). We aimed to elucidate the effects and mechanisms of BMP4 on epithelial-mesenchymal transition (EMT) through Smad1 signaling pathway to be involved in NSCL/P. METHODS The human oral epidermoid carcinoma cells (KBs) were transfected with plasmids or small interfering RNA (siRNA) to build the models. The migration of the cells was evaluated by transwell assay. Western blotting and quantitative real-time reverse transcription-polymerase chain reaction (qRT-PCR) were used to detect the expressions of BMP4, E-cadherin, N-cadherin, EMT-related transcription factors snal1 and snal2, matrix metalloproteinase 2 (MMP2), MMP9, Smad1, and phosphorylated Smad1. RESULTS In the overexpression group, the migration number of cells was increased significantly. The protein expression of E-cadherin was decreased significantly, while the protein expression level of the N-cadherin was increased significantly. The protein and mRNA expressions of MMP2, MMP9, snal1, and snal2 were significantly higher. The expression level of Smad1 was not significantly changed, while the phosphorylation of Smad1 was significantly increased. In the BMP4-siRNA group, the migrating number cells was significantly decreased. The protein expression of E-cadherin was increased significantly, while the expression of N-cadherin was significantly decreased. The protein and mRNA expressions of MMP2, MMP9, snal1, and snal2 were significantly lower than that of the control group. The expressions of Smad1 and phosphorylation of Smad1 were not significantly changed. CONCLUSIONS BMP4 enhances cell migration and promotes cell EMT through Smad1 signaling pathway. Abnormal BMP4 mediates migration and EMT through other relevant signaling pathways resulting in NSCL/P. The study provides new insight into the mechanisms of NSCL/P associated with BMP4.n.
Collapse
Affiliation(s)
- Jia-Wei Hong
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Yue Yu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Lu-Shan Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Zheng Li
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Rui Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Qi Wang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Zhen Ding
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Jin-Peng Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Mei-Rong Zhang
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| | - Li-Chun Xu
- Key Lab of Environment and Health, School of Public Health, Xuzhou Medical University, Jiangsu, China
| |
Collapse
|
7
|
Maleki F, Razmi H, Rashidi MR, Yousefi M, Ramazani S, Ghorbani M, Hojjat-Farsangi M, Shahpasand K. Detection of receptor tyrosine kinase-orphan receptor-2 using an electrochemical immunosensor modified with electrospun nanofibers comprising polyvinylpyrrolidone, soy, and gold nanoparticles. Mikrochim Acta 2023; 190:418. [PMID: 37770707 DOI: 10.1007/s00604-023-06002-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/16/2023] [Indexed: 09/30/2023]
Abstract
An electrochemical immunosensing platform was developed for the detection of receptor tyrosine kinase-orphan receptor-2 (ROR2) at a glassy carbon electrode (GCE) modified with the electrospun nanofiber containing polyvinylpyrrolidone (PVP), soy, and Au nanoparticles (AuNPs). The PVP/soy/AuNP nanofiber exhibited good electrochemical behavior due to synergistic effects between PVP, soy, and AuNPs. The PVP/soy in the modified film provided good mechanical strength, high porosity, flexible structures, and high specific surface area. On the other hand, the presence of AuNPs effectively improved conductivity, as well as the immobilization of anti-ROR2 on the modified GCE, leading to enhanced sensitivity. Various characterization approaches such as FE-SEM, FTIR, and EDS were used for investigating the morphological and structural features, and the elemental composition. The designed immunosensor performance was investigated using electrochemical impedance spectroscopy (EIS), cyclic voltammetry (CV), and differential pulse voltammetry (DPV). Under optimum conditions with a working potential range from -0.2 to 0.6 V (vs. SCE), sensitivity, linear range (LR), limit of detection (LOD), and correlation coefficient (R2) were acquired at 122.26 μA/cm2 dec, 0.01-1000 pg/mL, 3.39 fg/mL, and 0.9974, respectively. Furthermore, the determination of ROR2 in human plasma samples using the designed immunosensing platform was examined and exhibited satisfactory results including good selectivity against other proteins, reproducibility, and cyclic stability.
Collapse
Affiliation(s)
- Fatemeh Maleki
- Department of Chemistry, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 53714-161, Iran
| | - Habib Razmi
- Department of Chemistry, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, 53714-161, Iran.
| | | | - Mehdi Yousefi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soghra Ramazani
- Faculty of Textile Engineering, Urmia University of Technology, Urmia, 5716693188, Iran
| | - Marjan Ghorbani
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, BioClinicum, Karolinska University Hospital Solna and Karolinska Institute, 17164, Stockholm, Sweden
| | - Kourosh Shahpasand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology (RI-SCBT), Tehran, Iran
| |
Collapse
|
8
|
Corti C, Boscolo Bielo L, Schianca AC, Salimbeni BT, Criscitiello C, Curigliano G. Future potential targets of antibody-drug conjugates in breast cancer. Breast 2023; 69:312-322. [PMID: 36996620 PMCID: PMC10063401 DOI: 10.1016/j.breast.2023.03.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 02/06/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Metastatic breast cancer (BC) remains an incurable disease. Besides endocrine and targeted agents, chemotherapy is still a relevant therapeutic option for this disease. Recently, antibody-drug conjugates (ADCs) have shown to overcome the lack of tumor specificity and systemic toxicity typically associated with traditional chemotherapies, thus improving the therapeutic index. To effectively exploit this technological breakthrough, identification of optimal target antigens (Ags) is of utmost importance. To make the ideal target, differential expression of target Ags between healthy and cancer tissues, as well as specific mechanisms of ADC internalization after Ag-antibody interaction are required. Therefore, several in silico strategies to identify and characterize new promising candidate Ags have been developed. If initial in vitro and in vivo positive data are documented, thus providing a biological rationale for further Ag investigation, early phase clinical trials are designed. In BC, these strategies have already led to the development of effective ADCs, namely trastuzumab emtansine (T-DM1), trastuzumab deruxtecan (T-DXd) and sacituzumab govitecan (SG), primarily targeting HER2 and TROP-2. However, promising new Ags are currently under investigation, with encouraging results especially coming from targeting HER3, FRα, Tissue Factor, LIV-1, ROR1-2, and B7-H4. In this review, we describe the landscape of emergent and future potential targets (i.e., other than HER2 and TROP-2) investigated in BC for ADC development. Predominant target expression, function, preclinical rationale, potential clinical implication, as well as preliminary clinical trial results are provided.
Collapse
Affiliation(s)
- Chiara Corti
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy.
| | - Luca Boscolo Bielo
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Ambra Carnevale Schianca
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Beatrice Taurelli Salimbeni
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Carmen Criscitiello
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology (DIPO), University of Milan, Milan, Italy
| |
Collapse
|
9
|
Xiao X, Miao X, Duan S, Liu S, Cao Q, Wu R, Tao C, Zhao J, Qu Q, Markiewicz A, Peng R, Chen Y, Żaczek A, Liu J. Single-Cell Enzymatic Screening for Epithelial Mesenchymal Transition with an Ultrasensitive Superwetting Droplet-Array Microchip. SMALL METHODS 2023:e2300096. [PMID: 37086121 DOI: 10.1002/smtd.202300096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/16/2023] [Indexed: 05/03/2023]
Abstract
The phenotypic changes of circulating tumor cells (CTCs) during the epithelial-mesenchymal transition (EMT) have been a hot topic in tumor biology and cancer therapeutic development. Here, an integrated platform of single-cell fluorescent enzymatic assays with superwetting droplet-array microchips (SDAM) for ultrasensitive functional screening of epithelial-mesenchymal sub-phenotypes of CTCs is reported. The SDAM can generate high-density, volume well-defined droplet (0.66 nL per droplet) arrays isolating single tumor cells via a discontinuous dewetting effect. It enables sensitive detection of MMP9 enzyme activities secreted by single tumor cells, correlating to their epithelial-mesenchymal sub-phenotypes. In the pilot clinical double-blind tests, the authors have demonstrated that SDAM assays allow for rapid identification and functional screening of CTCs with different epithelial-mesenchymal properties. The consistency with the clinical outcomes validates the usefulness of single-cell secreted MMP9 as a biomarker for selective CTC screening and tumor metastasis monitoring. Convenient addressing and recovery of individual CTCs from SDAM have been demonstrated for gene mutation sequencing, immunostaining, and transcriptome analysis, revealing new understandings of the signaling pathways between MMP9 secretion and the EMT regulation of CTCs. The SDAM approach combined with sequencing technologies promises to explore the dynamic EMT plasticity of tumors at the single-cell level.
Collapse
Affiliation(s)
- Xiang Xiao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Xinxing Miao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Shanzhou Duan
- Department of thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215123, P. R. China
| | - Sidi Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Qinghua Cao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Renfei Wu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Chengcheng Tao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Jian Zhao
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Qing Qu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, Gdańsk, 80-211, Poland
| | - Rui Peng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| | - Yongbing Chen
- Department of thoracic Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, 215123, P. R. China
| | - Anna Żaczek
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, Medical University of Gdansk, Gdańsk, 80-211, Poland
| | - Jian Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Joint International Research Laboratory of Carbon-Based Functional Materials and Devices, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
10
|
Castro MV, Barbero GA, Máscolo P, Villanueva MB, Nsengimana J, Newton-Bishop J, Illescas E, Quezada MJ, Lopez-Bergami P. ROR2 promotes epithelial-mesenchymal transition by hyperactivating ERK in melanoma. J Cell Commun Signal 2023; 17:75-88. [PMID: 35723796 PMCID: PMC10030744 DOI: 10.1007/s12079-022-00683-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 05/23/2022] [Indexed: 12/19/2022] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a protein with important functions during embryogenesis that is dysregulated in human cancer. An intriguing feature of this receptor is that it plays opposite roles in different tumor types either promoting or inhibiting tumor progression. Understanding the complex role of this receptor requires a more profound exploration of both the altered biological and molecular mechanisms. Here, we describe that ROR2 promotes Epithelial-Mesenchymal Transition (EMT) by inducing cadherin switch and the upregulation of the transcription factors ZEB1, Twist, Slug, Snail, and HIF1A, together with a mesenchymal phenotype and increased migration. We show that ROR2 activates both p38 and ERK mitogen-activated protein kinase pathways independently of Wnt5a. Further, we demonstrated that the upregulation of EMT-related proteins depends on the hyperactivation of the ERK pathway far above the typical high constitutive activity observed in melanoma. In addition, ROR2 also promoted ERK phosphorylation, EMT, invasion, and necrosis in xenotransplanted mice. ROR2 also associates with EMT in tumor samples from melanoma patients where analysis of large cohorts revealed that increased ROR2 levels are linked to EMT signatures. This important role of ROR2 translates into melanoma patient' s prognosis since elevated ROR2 levels reduced overall survival and distant metastasis-free survival of patients with lymph node metastasis. In sum, these results demonstrate that ROR2 contributes to melanoma progression by inducing EMT and necrosis and can be an attractive therapeutic target for melanoma.
Collapse
Affiliation(s)
- María Victoria Castro
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Gastón Alexis Barbero
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Paula Máscolo
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
| | - María Belén Villanueva
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Jérémie Nsengimana
- Biostatistics Research Group, Population Health Sciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | | | - Edith Illescas
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
| | - María Josefina Quezada
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602., 1405, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina.
| |
Collapse
|
11
|
Lopez-Bergami P. ROR2, a driver of "phenotype switching" in melanoma? Cancer Cell Int 2022; 22:288. [PMID: 36127680 PMCID: PMC9487041 DOI: 10.1186/s12935-022-02711-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 09/08/2022] [Indexed: 12/03/2022] Open
Abstract
Receptor tyrosine kinase-like orphan receptor 2 (ROR2) is a receptor for the Wnt5a ligand that was shown to play a dual role in cancer. ROR2 was shown to either suppress or promote tumor progression in different tumor types by regulating the same biological processes (i.e. proliferation, invasion) in opposite ways. We have recently observed that ROR2 plays multiple and somewhat contradictory roles in melanoma where it impairs cell proliferation but promotes migration, EMT and chemoresistance. In the present article, ROR2 is proposed to be a major driver of “phenotype switching” in melanoma that can tilt the cellular behavior toward proliferative or invasive phenotypes. This function of ROR2 has therapeutic implications since it would provide an opportunity for targeting specific phenotypes such as invasive and drug-resistant ones by inhibiting ROR2.
Collapse
Affiliation(s)
- Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Hidalgo 775, 6th Floor, Lab 602, 1405, Buenos Aires, Argentina. .,Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), 1425, Buenos Aires, Argentina.
| |
Collapse
|
12
|
Dissection of the MKK3 Functions in Human Cancer: A Double-Edged Sword? Cancers (Basel) 2022; 14:cancers14030483. [PMID: 35158751 PMCID: PMC8833818 DOI: 10.3390/cancers14030483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/02/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023] Open
Abstract
The role played by MKK3 in human cancer is controversial. MKK3 is an evolutionarily conserved protein kinase that activates in response to a variety of stimuli. Phosphorylates, specifically the p38MAPK family proteins, contribute to the regulation of a plethora of cellular processes such as proliferation, differentiation, apoptosis, invasion, and cell migration. Genes in carcinogenesis are classified as oncogenes and tumor suppressors; however, a clear distinction is not always easily made as it depends on the cell context and tissue specificity. The aim of this study is the examination of the potential contribution of MKK3 in cancer through a systematic analysis of the recent literature. The overall results reveal a complex scenario of MKK3’s involvement in cancer. The oncogenic functions of MKK3 were univocally documented in several solid tumors, such as colorectal, prostate cancer, and melanoma, while its tumor-suppressing functions were described in glioblastoma and gastric cancer. Furthermore, a dual role of MKK3 as an oncogene as well as tumor a suppressor has been described in breast, cervical, ovarian, liver, esophageal, and lung cancer. However, overall, more evidence points to its role as an oncogene in these diseases. This review indicates that the oncogenic and tumor-suppressing roles of MKK3 are strictly dependent on the tumor type and further suggests that MKK3 could represent an efficient putative molecular target that requires contextualization within a specific tumor type in order to adequately evaluate its potential effectiveness in designing novel anticancer therapies.
Collapse
|
13
|
Castro MV, Lopez-Bergami P. Cellular and molecular mechanisms implicated in the dual role of ROR2 in cancer. Crit Rev Oncol Hematol 2022; 170:103595. [PMID: 35032666 DOI: 10.1016/j.critrevonc.2022.103595] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/03/2022] [Accepted: 01/10/2022] [Indexed: 02/07/2023] Open
Abstract
ROR1 and ROR2 are Wnt receptors that are critical for β-catenin-independent Wnt pathways and have been linked to processes driving tumor progression, such as cell proliferation, survival, invasion, and therapy resistance. Both receptors have garnered interest as potential therapeutic targets since they are largely absent in adult tissue, are overexpressed in several cancers, and, as members of the receptor tyrosine kinase family, are easier to target than all other components of the pathway. Unlike ROR1 which always promotes tumorigenesis, ROR2 has a very complex role in cancer acting either to promote or inhibit tumor progression in different tumor types. In the present article, we summarize the findings on ROR2 expression in cancer patients and its impact on clinical outcome. Further, we review the biological processes and signaling pathways regulated by ROR2 that explain its dual role in cancer. Finally, we describe the ongoing strategies to target ROR2 in cancer.
Collapse
Affiliation(s)
- María Victoria Castro
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Buenos Aires, 1405, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, 1425, Argentina
| | - Pablo Lopez-Bergami
- Centro de Estudios Biomédicos, Básicos, Aplicados y Desarrollo (CEBBAD), Universidad Maimónides, Buenos Aires, 1405, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, 1425, Argentina.
| |
Collapse
|
14
|
Xie S, Jin L, Yin T, Ren J, Liu W. Microarray analysis and functional prediction of differentially expressed circular RNAs in acquired middle ear cholesteatoma. Biomed Eng Online 2021; 20:129. [PMID: 34922560 PMCID: PMC8684697 DOI: 10.1186/s12938-021-00960-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/16/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Middle ear cholesteatoma is characterized by hyper-proliferation of keratinocytes. Circular RNA (circRNA) plays an essential role in the pathogenesis of many proliferative diseases. However, the role of circRNA in the etiopathogenesis of middle ear cholesteatoma is rarely investigated so far. We aimed to investigate the differential expression profiling of circRNAs between acquired middle ear cholesteatoma and normal skin, and to identify potential circRNAs contributing to the etiopathogenesis of middle ear cholesteatoma. Microarray analysis and functional prediction were performed to investigate the circRNA expression profiling between middle ear cholesteatoma and normal skin. Validation of differentially expressed circRNAs was conducted by qRT-PCR. Prediction of m6A modification was also carried out. RESULTS Microarray analysis displayed that totally 93 up-regulated and 85 down-regulated circRNAs were identified in middle ear cholesteatoma. Through validation, expressions of hsa_circRNA_104327 and hsa_circRNA_404655 were significantly higher, while hsa_circRNA_000319 was significantly down-regulated in cholesteatoma. GO classification, KEGG pathway, and ceRNA network analyses suggested that these differentially expressed circRNAs might play important roles in the etiopathogenesis of middle ear cholesteatoma. Prediction of m6A modification exhibited that hsa_circRNA_000319 possessed 4 m6A sites with very high confidence, and hsa_circRNA_404655 had 3 m6A sites with high confidence. CONCLUSIONS Our study revealed that these differentially expressed circRNAs might contribute to the etiopathogenesis of middle ear cholesteatoma. Further researches should be conducted to investigate the exact mechanism of these differentially expressed circRNAs in the etiopathogenesis of middle ear cholesteatoma. Targeting on these circRNAs may provide a new strategy for middle ear cholesteatoma therapy in the future.
Collapse
Affiliation(s)
- Shumin Xie
- Department of Otolaryngology-Head and Neck Surgery, The Xiangya Hospital, Central South University, Hunan Provincial Key Lab, Otolaryngology Institute of Major Diseases, Changsha, 410008, Hunan, China
| | - Li Jin
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tuanfang Yin
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Jihao Ren
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Wei Liu
- Department of Otolaryngology-Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
15
|
Menck K, Heinrichs S, Wlochowitz D, Sitte M, Noeding H, Janshoff A, Treiber H, Ruhwedel T, Schatlo B, von der Brelie C, Wiemann S, Pukrop T, Beißbarth T, Binder C, Bleckmann A. WNT11/ROR2 signaling is associated with tumor invasion and poor survival in breast cancer. J Exp Clin Cancer Res 2021; 40:395. [PMID: 34911552 PMCID: PMC8672621 DOI: 10.1186/s13046-021-02187-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 11/16/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast cancer has been associated with activation of the WNT signaling pathway, although no driver mutations in WNT genes have been found yet. Instead, a high expression of the alternative WNT receptor ROR2 was observed, in particular in breast cancer brain metastases. However, its respective ligand and downstream signaling in this context remained unknown. METHODS We modulated the expression of ROR2 in human breast cancer cells and characterized their gene and protein expression by RNA-Seq, qRT-PCR, immunoblots and reverse phase protein array (RPPA) combined with network analyses to understand the molecular basis of ROR2 signaling in breast cancer. Using co-immunoprecipitations, we verified the interaction of ROR2 with the identified ligand, WNT11. The functional consequences of WNT11/ROR2 signaling for tumor cell aggressiveness were assessed by microscopy, impedance sensing as well as viability and invasion assays. To evaluate the translational significance of our findings, we performed gene set enrichment, expression and survival analyses on human breast cancer brain metastases. RESULTS We found ROR2 to be highly expressed in aggressive breast tumors and associated with worse metastasis-free survival. ROR2 overexpression induced a BRCAness-like phenotype in a cell-context specific manner and rendered cells resistant to PARP inhibition. High levels of ROR2 were furthermore associated with defects in cell morphology and cell-cell-contacts leading to increased tumor invasiveness. On a molecular level, ROR2 overexpression upregulated several non-canonical WNT ligands, in particular WNT11. Co-immunoprecipitation confirmed that WNT11 indeed interacts with the cysteine-rich domain of ROR2 and triggers its invasion-promoting signaling via RHO/ROCK. Knockdown of WNT11 reversed the pro-invasive phenotype and the cellular changes in ROR2-overexpressing cells. CONCLUSIONS Taken together, our study revealed a novel auto-stimulatory loop in which ROR2 triggers the expression of its own ligand, WNT11, resulting in enhanced tumor invasion associated with breast cancer metastasis.
Collapse
Affiliation(s)
- Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Saskia Heinrichs
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Darius Wlochowitz
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Maren Sitte
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Helen Noeding
- Institute for Physical Chemistry, Georg August University Göttingen, 37075, Göttingen, Germany
| | - Andreas Janshoff
- Institute for Physical Chemistry, Georg August University Göttingen, 37075, Göttingen, Germany
| | - Hannes Treiber
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Bawarjan Schatlo
- Department of Neurosurgery, University Medical Center Göttingen, 37099, Göttingen, Germany
| | | | - Stefan Wiemann
- Division of Molecular Genome Analysis, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Tobias Pukrop
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
- Department of Internal Medicine III, Hematology and Medical Oncology, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Claudia Binder
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149, Münster, Germany.
- West German Cancer Center, University Hospital Münster, 48149, Münster, Germany.
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099, Göttingen, Germany.
| |
Collapse
|
16
|
Liu D, Sharbeen G, Phillips P, Ford CE. ROR1 and ROR2 expression in pancreatic cancer. BMC Cancer 2021; 21:1199. [PMID: 34763666 PMCID: PMC8582180 DOI: 10.1186/s12885-021-08952-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The Wnt receptors ROR1 and ROR2 are generating increased interest as cancer therapeutic targets but remain understudied in pancreatic ductal adenocarcinoma (PDAC). Compared to canonical Wnt/ β-catenin signalling, the role of noncanonical Wnt signalling in PDAC remains largely unknown. Only one study has investigated the prognostic significance of the noncanonical Wnt signalling receptor, ROR2 in PDAC. No studies have investigated the prognostic role of ROR1 in PDAC. METHODS Here, we performed analysis of ROR1 and ROR2 mRNA expression in three publicly available datasets ICGC-PACA-AU (n = 81), TCGA-PAAD (n = 150) and CPTAC-PDAC (n = 137). ROR1 and ROR2 protein expression from the CPTAC-PDAC discovery cohort were also analysed. Immunohistochemistry (IHC) using the validated anti ROR1 monoclonal antibody (4A5) was performed on the Australian Pancreatic Cancer Genome Initiative (APGI) cohort of PDAC samples (n = 152). Association between ROR1 cytoplasmic staining intensity and clinicopathological parameters including stage, grade and overall survival (OS) was investigated. RESULTS High ROR1 mRNA expression levels correlated with a favourable OS outcome in all of the ICGC-PACA-AU, TCGA-PAAD and CPTAC-PDAC cohorts. ROR1 protein expression was not associated with stage, grade or OS in the APGI cohort. CONCLUSION ROR1 and ROR2 have potential as prognostic markers when measured at the mRNA level in PDAC. Our IHC cohort did not support ROR1 protein expression in predicting OS, and highlighted the discrepancy of prognostic biomarkers when measured by MS, IHC and RNAseq.
Collapse
Affiliation(s)
- Dongli Liu
- Gynaecological Cancer Research Group, Lowy Cancer Research Centre, School of Women's and Children's Health, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - George Sharbeen
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Science, Faculty of Medicine & Health, University of New South Wales, Sydney, Australia
| | - Phoebe Phillips
- Pancreatic Cancer Translational Research Group, Lowy Cancer Research Centre, School of Medical Science, Faculty of Medicine & Health, University of New South Wales, Sydney, Australia
| | - Caroline E Ford
- Gynaecological Cancer Research Group, Lowy Cancer Research Centre, School of Women's and Children's Health, Faculty of Medicine & Health, University of New South Wales, Sydney, New South Wales, 2052, Australia.
| |
Collapse
|
17
|
Bansal S, Limaye AP, Lee J, Fleming T, Poulson C, Omar A, Hachem R, Bharat A, Bremner RM, Smith MA, Mohanakumar T. Circulating exosomes induced by respiratory viral infections in lung transplant recipients activate cellular stress, innate immune pathways and epithelial to mesenchymal transition. Transpl Immunol 2021; 69:101480. [PMID: 34619318 DOI: 10.1016/j.trim.2021.101480] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Chronic lung transplant rejection occurs in over 50% of lung transplant recipients and mechanism of chronic rejection is unknown. Evaluation of potential mechanism of exosomes from lung transplant recipients diagnosed with respiratory viral infection (RVI) in inducing chronic lung allograft dysfunction (CLAD). METHOD Exosomes were isolated from lung transplant recipients followed by DNA and RNA isolation from exosomes. Cell signaling mechanisms were studied by co-culturing exosomes with human epithelial cells. Mice were immunized with exosomes and lung homogenates were studied for immune signaling proteins. RESULTS Exosomes from lung transplant recipients with RVI carry nucleic acids which are capable of inducing innate immune signaling, endoplasmic reticulum stress, and epithelial mesenchymal transition. CONCLUSION Therefore, we propose that RVI can lead to induction of exosomes that initiate the process leading to CLAD in mice models. These novel findings identified the molecular mechanisms by which RVI increases the risk of CLAD.
Collapse
Affiliation(s)
- Sandhya Bansal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Ajit P Limaye
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - John Lee
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Timothy Fleming
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Christin Poulson
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Ashraf Omar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Ramsey Hachem
- Washington University School of Medicine, Department of Medicine, St. Louis, MO, United States of America
| | - Ankit Bharat
- Northwestern University, Chicago, Chicago, IL, United States of America
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America
| | - T Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States of America.
| |
Collapse
|
18
|
Prognostic Significance of ROR2 Expression in Patients with Urothelial Carcinoma. Biomedicines 2021; 9:biomedicines9081054. [PMID: 34440262 PMCID: PMC8392262 DOI: 10.3390/biomedicines9081054] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/16/2021] [Accepted: 08/17/2021] [Indexed: 02/01/2023] Open
Abstract
We investigated the association of receptor tyrosine kinase-like orphan receptor 2 (ROR2) expression with clinicopathological features and oncologic outcomes in large urothelial carcinoma (UC) of the upper tract (UTUC) and urinary bladder (UBUC) cohorts. Through transcriptomic profiling of a published dataset (GSE31684), ROR2 was discovered to be the most upregulated gene during UC progression, focusing on the JNK cascade (GO:0007254). Initially, the evaluation of ROR2 mRNA expression in 50 frozen UBUCs showed significantly upregulated levels in high-stage UC. Moreover, high ROR2 immunoexpression significantly correlated with high tumor stage, high tumor grade, lymph node metastasis, and vascular invasion (all p < 0.05). In multivariate analysis, after adjusting for standard clinicopathological features, ROR2 expression status was an independent prognosticator of cancer-specific survival and metastasis-free survival in UTUC and UBUC (all p < 0.01). In the subgroup analysis, it also significantly predicted bladder tumor recurrence in non-muscle invasive UBUC. Furthermore, the GO enrichment analysis showed that fatty acid, monocarboxylic acid, carboxylic acid metabolic processes, negative regulation of neutrophil migration, and negative regulation of granulocyte and neutrophil chemotaxis were significantly enriched by ROR2 dysregulation. In conclusion, high ROR2 immunoexpression was associated with aggressive pathological characteristics in UC and independently predicted worse prognosis, suggesting it could play roles in clinical risk stratification and therapy decisions.
Collapse
|
19
|
Shi F, Ma M, Zhai R, Ren Y, Li K, Wang H, Xu C, Huang X, Wang N, Zhou F, Yao W. Overexpression of heat shock protein 70 inhibits epithelial-mesenchymal transition and cell migration induced by transforming growth factor-β in A549 cells. Cell Stress Chaperones 2021; 26:505-513. [PMID: 33598875 PMCID: PMC8065086 DOI: 10.1007/s12192-021-01196-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 01/07/2023] Open
Abstract
Heat shock protein 70 (HSP70) is a key member of the HSP family that contributes to a pre-cancerous environment; however, its role in lung cancer remains poorly understood. The present study used geranylgeranylacetone (GGA) to induce HSP70 expression, and transforming growth factor-β (TGF-β) was used to construct an epithelial-mesenchymal transition (EMT) model by stimulating A549 cells in vitro. Western Blot was performed to detect protein levels of NADPH oxidase 4 (NOX4) and the EMT-associated proteins E-cadherin and vimentin both before and after HSP70 expression. Cell morphological changes were observed, and the effect of HSP70 on cell migration ability was detected via the wound healing. The results demonstrated that GGA at 50 and 200 μmol/L could significantly induce HSP70 expression in A549 cells (P < 0.05). Furthermore, HSP70 induced by 200 μmol/L GGA significantly inhibited the changes of E-cadherin, vimentin, and cell morphology induced by TGF-β (P < 0.05), while HSP70 induced by 50 μmol/L GGA did not. The results of the wound healing assay indicated that 200 μmol/L GGA significantly inhibited A549 cell migration induced by TGF-β. Taken together, the results of the present study demonstrated that overexpression of HSP70 inhibited the TGF-β induced EMT process and changed the cell morphology and migratory ability induced by TGF-β in A549 cells.
Collapse
Affiliation(s)
- Fengxian Shi
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Mingze Ma
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ruonan Zhai
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Yanan Ren
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Ke Li
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Hang Wang
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Chunyan Xu
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Xiaowen Huang
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Na Wang
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China
| | - Fang Zhou
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| | - Wu Yao
- Department of Occupational Health and Environmental Health, College of Public Health, Zhengzhou University, Zhengzhou, 450001, Henan, China.
| |
Collapse
|
20
|
Zhang M, Wang Y, Jiang L, Song X, Zheng A, Gao H, Wei M, Zhao L. LncRNA CBR3-AS1 regulates of breast cancer drug sensitivity as a competing endogenous RNA through the JNK1/MEK4-mediated MAPK signal pathway. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:41. [PMID: 33494806 PMCID: PMC7830819 DOI: 10.1186/s13046-021-01844-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
Background Adriamycin (ADR) resistance is one of the main obstacles to improving the clinical prognosis of breast cancer patients. Long noncoding RNAs (lncRNAs) can regulate cell behavior, but the role of these RNAs in the anti-ADR activity of breast cancer remains unclear. Here, we aim to investigate the imbalance of a particular long noncoding RNA, lncRNA CBR3 antisense RNA 1 (CBR3-AS1), and its role in ADR resistance. Methods Microarray analysis of ADR-resistant breast cancer cells was performed to identify CBR3-AS1. CCK-8 and colony formation assays were used to detect the sensitivity of breast cancer cells to ADR. Dual-luciferase reporter, RNA pulldown, IHC and western blot analyses were used to verify the relationship between the expression of CBR3-AS1, miRNA and target genes. For in vivo experiments, the effect of CBR3-AS1 on breast cancer resistance was observed in a xenograft tumor model. The role of CBR3-AS1 in influencing ADR sensitivity was verified by clinical breast cancer specimens from the TCGA, CCLE, and GDSC databases. Results We found that CBR3-AS1 expression was significantly increased in breast cancer tissues and was closely correlated with poor prognosis. CBR3-AS1 overexpression promoted ADR resistance in breast cancer cells in vitro and in vivo. Mechanistically, we identified that CBR3-AS1 functioned as a competitive endogenous RNA by sponging miR-25-3p. MEK4 and JNK1 of the MAPK pathway were determined to be direct downstream proteins of the CBR3-AS1/miR-25-3p axis in breast cancer cells. Conclusions In summary, our findings demonstrate that CBR3-AS1 plays a critical role in the chemotherapy resistance of breast cancer by mediating the miR-25-3p and MEK4/JNK1 regulatory axes. The potential of CBR3-AS1 as a targetable oncogene and therapeutic biomarker of breast cancer was identified. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-021-01844-7.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Yan Wang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Longyang Jiang
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Xinyue Song
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Ang Zheng
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China.,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China
| | - Hua Gao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| | - Lin Zhao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, 110122, Liaoning Province, China. .,Liaoning Key Laboratory of molecular targeted anti-tumor drug development and evaluation; Liaoning Cancer immune peptide drug Engineering Technology Research Center; Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education; China Medical University, Shenyang, Liaoning Province, China.
| |
Collapse
|
21
|
Gong L, Cai L, Li G, Cai J, Yi X. GADD45B Facilitates Metastasis of Ovarian Cancer Through Epithelial-Mesenchymal Transition. Onco Targets Ther 2021; 14:255-269. [PMID: 33469306 PMCID: PMC7811469 DOI: 10.2147/ott.s281450] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/11/2020] [Indexed: 12/22/2022] Open
Abstract
Background Growth arrest and DNA-damage-inducible 45 beta (GADD45B) is overexpressed and is associated with poor clinical outcomes in many human cancers, but the clinical implication of GADD45B in epithelial ovarian cancer (EOC) remains unclear. Methods Bioinformatics analysis of The Cancer Genome Atlas (TCGA) and gene expression omnibus (GEO) cohorts was used to illustrate the relationship between GADD45B expression and metastasis, as well as the survival time of EOC. GADD45B was downregulated by siRNAs in EOC cells, and migration ability was determined by a transwell assay and wound-healing assay. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and gene set enrichment analysis (GSEA) were conducted to discover the downstream pathway of GADD45B. The regulation of epithelial–mesenchymal transition (EMT) by GADD45B was verified by Western blotting and qRT-PCR. Finally, the correlation of GADD45B expression with EOC metastasis was investigated in EOC tissues by immunohistochemistry. Results Overexpression of GADD45B indicates shorter overall survival time and progression-free survival time, and it is an independent risk factor for poor survival in EOC patients. Elevated GADD45B is related to venous invasion, lymphatic invasion and peritoneal carcinomatosis. Downregulation of GADD45B decreases the migration of ES2 and SKOV3 cells. Further KEGG enrichment analysis and GSEA revealed that EMT may be the downstream pathway of GADD45B. In addition, reduced GADD45B increases the expression of E-cadherin and decreases that of N-cadherin and vimentin. Finally, immunohistochemical analysis of GADD45B expression revealed that the expression of GADD45B in omental metastatic tissues was higher than that in matched primary ovarian cancer tissues. These results suggest that elevated GADD45B promotes the motility of ovarian cancer cells through EMT and is associated with EOC metastasis. Conclusion GADD45B can promote the motility of ovarian cancer cells through EMT, is associated with EOC metastasis, and may be a new biomarker of metastasis and prognosis.
Collapse
Affiliation(s)
- Lanqing Gong
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Liqiong Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Guodong Li
- Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Jing Cai
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| | - Xiaoqing Yi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
22
|
Menck K, Heinrichs S, Baden C, Bleckmann A. The WNT/ROR Pathway in Cancer: From Signaling to Therapeutic Intervention. Cells 2021; 10:cells10010142. [PMID: 33445713 PMCID: PMC7828172 DOI: 10.3390/cells10010142] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/08/2021] [Accepted: 01/11/2021] [Indexed: 12/13/2022] Open
Abstract
The WNT pathway is one of the major signaling cascades frequently deregulated in human cancer. While research had initially focused on signal transduction centered on β-catenin as a key effector activating a pro-tumorigenic transcriptional response, nowadays it is known that WNT ligands can also induce a multitude of β-catenin-independent cellular pathways. Traditionally, these comprise WNT/planar cell polarity (PCP) and WNT/Ca2+ signaling. In addition, signaling via the receptor tyrosine kinase-like orphan receptors (RORs) has gained increasing attention in cancer research due to their overexpression in a multitude of tumor entities. Active WNT/ROR signaling has been linked to processes driving tumor development and progression, such as cell proliferation, survival, invasion, or therapy resistance. In adult tissue, the RORs are largely absent, which has spiked the interest in them for targeted cancer therapy. Promising results in preclinical and initial clinical studies are beginning to unravel the great potential of such treatment approaches. In this review, we summarize seminal findings on the structure and expression of the RORs in cancer, their downstream signaling, and its output in regard to tumor cell function. Furthermore, we present the current clinical anti-ROR treatment strategies and discuss the state-of-the-art, as well as the challenges of the different approaches.
Collapse
Affiliation(s)
- Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Saskia Heinrichs
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Cornelia Baden
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, 48149 Münster, Germany; (K.M.); (S.H.); (C.B.)
- West German Cancer Center, University Hospital Münster, 48149 Münster, Germany
- Department of Hematology/Medical Oncology, University Medical Center Göttingen, 37099 Göttingen, Germany
- Correspondence: ; Tel.: +49-0251-8352712
| |
Collapse
|
23
|
Jiang Y, Xie F, Lv X, Wang S, Liao X, Yu Y, Dai Q, Zhang Y, Meng J, Hu G, Peng Z, Tao L. Mefunidone ameliorates diabetic kidney disease in STZ and db/db mice. FASEB J 2020; 35:e21198. [PMID: 33225469 DOI: 10.1096/fj.202001138rr] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 12/11/2022]
Abstract
Diabetic kidney disease (DKD) is a major cause of end stage renal diseases worldwide. Despite successive interventions for delaying the progression of DKD, current treatments cannot reverse the pathological progression. Mefunidone (MFD) is a new compound with potent antifibrotic properties, but the effect of MFD on DKD remains unknown. Therefore, we investigated the protective effects of MFD in both models of the db/db type 2 diabetes (T2D) and streptozotocin (STZ)-induced type 1 diabetes (T1D) models. Compared with the model group, MFD treatment significantly reduced pathological changes observed by PAS staining, PASM staining, and Masson staining in vivo. To further elucidate the potential mechanisms, we discovered MFD treatment notably restored podocyte function, alleviated inflammation, abated ROS generation, inhibited the TGF-β1/SAMD2/3 pathway, suppressed the phosphorylation levels of MAPKs (ERK1/2, JNK, and P38), and reduced epithelial-to-mesenchymal transition(EMT). In conclusion, these findings demonstrate the effectiveness of MFD in diabetic nephropathy and elucidate its possible mechanism.
Collapse
Affiliation(s)
- Yupeng Jiang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Feifei Xie
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Xin Lv
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Shuting Wang
- Department of Oncology, Xiangya Hospital of Central South University, Changsha, China
| | - Xiaohua Liao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Yue Yu
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Qin Dai
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Yan Zhang
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Gaoyun Hu
- Faculty of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Zhangzhe Peng
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| | - Lijian Tao
- Department of Nephrology, Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|