1
|
Kamei CN, Sampson WGB, Albertz C, Aries O, Wolf A, Upadhyay RM, Hughes SM, Schenk H, Bonnet F, Draper BW, McCracken KW, Marciano DK, Oxburgh L, Drummond IA. Multiple Wnt signaling pathways direct epithelial tubule interconnection in the regenerating zebrafish kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645545. [PMID: 40196581 PMCID: PMC11974930 DOI: 10.1101/2025.03.26.645545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Epithelial tubule fusion is fundamental for kidney morphogenesis. Differentiating nephron tubules interconnect with collecting system epithelia to generate a lumenal pathway for fluid excretion. In the adult zebrafish kidney, nephrogenesis occurs as a regenerative response to injury and provides a model to explore cell signaling pathways required for tubule interconnection. We show that canonical Wnt signaling at the junction between two tubules induces a mesenchymal, invasive cell phenotype and is required, along with Src kinase and rac1, to generate basal cell protrusions. The Wnt ligands wnt9b and wnt4 are both required for new nephron formation after injury. Mutation in wnt4 or treatment with the canonical Wnt inhibitor IWR1 blocks formation of basal protrusions in forming nephrons. Mutation in the Wnt receptor frizzled9b reveals a fusion-associated non-canonical Wnt pathway that acts to 1) restrict canonical Wnt gene expression, 2) drive Rho kinase-dependent apical constriction of epithelial cells, and 3) position basal protrusions and generate orthogonal tubule lumenal connections. As a result, frizzled9b mutant nephrons fail to fully interconnect with target distal tubules. Our results indicate that canonical and non-canonical Wnt signaling interact in the same cells to orient and drive tubule interconnection in the regenerating zebrafish kidney.
Collapse
|
2
|
Qian Z, Lin W, Cai X, Wu J, Ke K, Ye Z, Wu F. WYC-209 inhibited GC malignant progression by down-regulating WNT4 through RARα. Cancer Biol Ther 2024; 25:2299288. [PMID: 38178596 PMCID: PMC10773637 DOI: 10.1080/15384047.2023.2299288] [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: 08/08/2023] [Accepted: 12/21/2023] [Indexed: 01/06/2024] Open
Abstract
Gastric cancer (GC) has been a major health burden all over the world but there are fewer promising chemotherapeutic drugs due to its multidrug resistance. It has been reported that WYC-209 suppresses the growth and metastasis of tumor-repopulating cells but the effect on GC was not explored. MTT, colony formation, and transwell assays were performed to examine the effects of WYC-209 on the proliferation, colony growth, and mobility of GC cells. Western blotting and qRT-PCR were used to detect the expression of proteins and mRNA. RNA-seq and enrichment analyses were conducted for the differentially expressed genes and enriched biological processes and pathways. The rescue experiments were carried out for further validation. Besides, we constructed xenograft model to confirm the effect of WYC-209 in vivo. The dual-luciferase reporter and Chromatin immunoprecipitation were implemented to confirm the underlying mechanism. WYC-209 exerted excellent anti-cancer effects both in vitro and in vivo. Based on RNA-seq and enrichment analyses, we found that Wnt family member 4 (WNT4) was significantly down-regulated. More importantly, WNT4 overexpression breached the inhibitory effect of WYC-209 on GC progression. Mechanically, WYC-209 significantly promoted the binding between retinoic acid receptor α (RARα) and WNT4 promoter. WYC-209 exerts anti-tumor effects in GC by down-regulating the expression of WNT4 via RARα.
Collapse
Affiliation(s)
- Zhenyuan Qian
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Wenfa Lin
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xufan Cai
- School of Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianzhang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Kun Ke
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zaiyuan Ye
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Fang Wu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| |
Collapse
|
3
|
Li J, Zhang H, Wang J, Deng M, Li Z, Jiang W, Xu K, Wu L, Dong Z, Liu J, Ding Q, Yu H. Development and Validation of an AI-Driven System for Automatic Literature Analysis and Molecular Regulatory Network Construction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405395. [PMID: 39373342 PMCID: PMC11600262 DOI: 10.1002/advs.202405395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/06/2024] [Indexed: 10/08/2024]
Abstract
Decoding gene regulatory networks is essential for understanding the mechanisms underlying many complex diseases. GENET is developed, an automated system designed to extract and visualize extensive molecular relationships from published biomedical literature. Using natural language processing, entities and relations are identified from a randomly selected set of 1788 scientific articles, and visualized in a filterable knowledge graph. The performance of GENET is evaluated and compared with existing methods. The named entity recognition model has achieved an overall precision of 94.23% (4835/5131; 93.56-94.84%), recall of 97.72% (4835/4948; 97.27-98.10%), and an F1 score of 95.94%. The relation extraction model has demonstrated an overall precision of 91.63% (2593/2830; 90.55-92.59%), recall of 89.17% (2593/2908; 87.99-90.25%), and an F1 score of 90.38%. GENET significantly outperforms existing methods in extracting molecular relationships (P < 0.001). Additionally, GENET has successfully predicted WNT family member 4 regulates insulin-like growth factor 2 via signal transducer and activator of transcription 3 in colon cancer. With RNA sequencing data and multiple immunofluorescence, the authenticity of this prediction is validated, supporting the promising feasibility of GENET.
Collapse
Affiliation(s)
- Jia Li
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Hailin Zhang
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Jiamin Wang
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Mei Deng
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Zhiyong Li
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Wei Jiang
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Kejin Xu
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Lianlian Wu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Zehua Dong
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Jun Liu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Nursing Department of Renmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
| | - Qianshan Ding
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| | - Honggang Yu
- Department of GastroenterologyRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Key Laboratory of Digestive SystemRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Hubei Provincial Clinical Research Center for Digestive Disease Minimally Invasive IncisionRenmin Hospital of Wuhan UniversityWuhanHubei430060P. R. China
- Engineering Research Center for Artificial Intelligence Endoscopy Interventional Treatment of Hubei ProvinceWuhanHubei430060P. R. China
| |
Collapse
|
4
|
Kulkarni P, Basu R, Bonn T, Low B, Mazurek N, Kopchick JJ. Growth Hormone Upregulates Melanoma Drug Resistance and Migration via Melanoma-Derived Exosomes. Cancers (Basel) 2024; 16:2636. [PMID: 39123364 PMCID: PMC11311539 DOI: 10.3390/cancers16152636] [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: 06/25/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
Drug resistance in melanoma is a major hindrance in cancer therapy. Growth hormone (GH) plays a pivotal role in contributing to the resistance to chemotherapy. Knocking down or blocking the GH receptor has been shown to sensitize the tumor cells to chemotherapy. Extensive studies have demonstrated that exosomes, a subset of extracellular vesicles, play an important role in drug resistance by transferring key factors to sensitize cancer cells to chemotherapy. In this study, we explore how GH modulates exosomal cargoes from melanoma cells and their role in drug resistance. We treated the melanoma cells with GH, doxorubicin, and the GHR antagonist, pegvisomant, and analyzed the exosomes released. Additionally, we administered these exosomes to the recipient cells. The GH-treated melanoma cells released exosomes with elevated levels of ABC transporters (ABCC1 and ABCB1), N-cadherin, and MMP2, enhancing drug resistance and migration in the recipient cells. GHR antagonism reduced these exosomal levels, restoring drug sensitivity and attenuating migration. Overall, our findings highlight a novel role of GH in modulating exosomal cargoes that drive chemoresistance and metastasis in melanoma. This understanding provides insights into the mechanisms of GH in melanoma chemoresistance and suggests GHR antagonism as a potential therapy to overcome chemoresistance in melanoma treatment.
Collapse
Affiliation(s)
- Prateek Kulkarni
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Reetobrata Basu
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
| | - Taylor Bonn
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Department of Nutrition, Ohio University, Athens, OH 45701, USA
| | - Beckham Low
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Department of Biological Sciences, Ohio University, Athens, OH 45701, USA
| | - Nathaniel Mazurek
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Environmental and Plant Biology, Ohio University, Athens, OH 45701, USA
| | - John J. Kopchick
- Edison Biotechnology Institute, Ohio University, Athens, OH 45701, USA; (P.K.); (R.B.); (T.B.); (B.L.); (N.M.)
- Molecular and Cellular Biology Program, Ohio University, Athens, OH 45701, USA
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA
| |
Collapse
|
5
|
Kurzava Kendall L, Ma Y, Yang T, Lubecka K, Stefanska B. Epigenetic Effects of Resveratrol on Oncogenic Signaling in Breast Cancer. Nutrients 2024; 16:699. [PMID: 38474826 DOI: 10.3390/nu16050699] [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: 01/30/2024] [Revised: 02/23/2024] [Accepted: 02/26/2024] [Indexed: 03/14/2024] Open
Abstract
The crosstalk between oncogenic signaling pathways plays a crucial role in driving cancer development. We previously demonstrated that dietary polyphenols, specifically resveratrol (RSV) and other stilbenoids, epigenetically target oncogenes for silencing via DNA hypermethylation in breast cancer. In the present study, we identify signal transduction regulators among RSV-hypermethylated targets and investigate the functional role of RSV-mediated DNA hypermethylation in the regulation of Hedgehog and Wnt signaling. Non-invasive ER-positive MCF-7 and highly invasive triple-negative MCF10CA1a human breast cancer cell lines were used as experimental models. Upon 9-day exposure to 15 µM RSV, pyrosequencing and qRT-PCR were performed to assess DNA methylation and expression of GLI2 and WNT4, which are upstream regulators of the Hedgehog and Wnt pathways, respectively. Our results showed that RSV led to a DNA methylation increase within GLI2 and WNT4 enhancers, which was accompanied by decreases in gene expression. Consistently, we observed the downregulation of genes downstream of the Hedgehog and Wnt signaling, including common targets shared by both pathways, CCND1 and CYR61. Further analysis using chromatin immunoprecipitation identified increased H3K27 trimethylation and decreased H3K9 and H3K27 acetylation, along with abolishing OCT1 transcription factor binding. Those changes indicate a transcriptionally silent chromatin state at GLI2 and WNT4 enhancers. The inhibition of the Wnt signal transduction was confirmed using a phospho-antibody array that demonstrated suppression of positive and stimulation of negative Wnt regulators. In conclusion, our results provide scientific evidence for dietary polyphenols as epigenetics-modulating agents that act to re-methylate and silence oncogenes, reducing the oncogenic signal transduction. Targeting such an action could be an effective strategy in breast cancer prevention and/or adjuvant therapy.
Collapse
Affiliation(s)
| | - Yuexi Ma
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Tony Yang
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Katarzyna Lubecka
- Department of Biomedical Chemistry, Medical University of Lodz, 92-215 Lodz, Poland
| | - Barbara Stefanska
- Food, Nutrition and Health Program, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
6
|
Sen P, Roy Acharyya S, Arora A, Ghosh SS. An in-silico approach to understand the potential role of Wnt inhibitory factor-1 (WIF-1) in the inhibition of the Wnt signalling pathway. J Biomol Struct Dyn 2024; 42:326-345. [PMID: 36995086 DOI: 10.1080/07391102.2023.2192810] [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: 06/28/2022] [Accepted: 03/12/2023] [Indexed: 03/31/2023]
Abstract
WIF1 (Wnt inhibitory factor 1) is a potent tumour suppressor gene which is epigenetically silenced in numerous malignancies. The associations of WIF1 protein with the Wnt pathway molecules have not been fully explored, despite their involvement in the downregulation of several malignancies. In the present study, a computational approach encompassing the expression, gene ontology analysis and pathway analysis is employed to obtain an insight into the role of the WIF1 protein. Moreover, the interaction of the WIF1 domain with the Wnt pathway molecules was carried out to ascertain the tumour-suppressive role of the domain, along with the determination of their plausible interactions. Initially, the protein-protein interaction network analysis endowed us with the Wnt ligands (such as Wnt1, Wnt3a, Wnt4, Wnt5a, Wnt8a and Wnt9a), along with the Frizzled receptors (Fzd1 and Fzd2) and the low-density lipoprotein complex (Lrp5/6) as the foremost interactors of the protein. Further, the expression analysis of the aforementioned genes and proteins was determined using The Cancer Genome Atlas to comprehend the significance of the signalling molecules in the major cancer subtypes. Moreover, the associations of the aforementioned macromolecular entities with the WIF1 domain were explored using the molecular docking studies, whereas the dynamics and stability of the assemblage were investigated using 100 ns molecular dynamics simulations. Therefore, providing us insights into the plausible roles of WIF1 in inhibiting the Wnt pathways in various malignancies.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Plaboni Sen
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Suchandra Roy Acharyya
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Arisha Arora
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Siddhartha Sankar Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| |
Collapse
|
7
|
Boeschen M, Kuhn CK, Wirtz H, Seyfarth HJ, Frille A, Lordick F, Hacker UT, Obeck U, Stiller M, Bläker H, von Laffert M. Comparative bioinformatic analysis of KRAS, STK11 and KEAP1 (co-)mutations in non-small cell lung cancer with a special focus on KRAS G12C. Lung Cancer 2023; 184:107361. [PMID: 37699269 DOI: 10.1016/j.lungcan.2023.107361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 09/14/2023]
Abstract
OBJECTIVES Mutations in STK11 (STK11MUT) and KEAP1 (KEAP1MUT) occur frequently in non-small cell lung cancer (NSCLC) and are often co-mutated with KRAS. Several studies linked the co-occurrence of KRASMUT + STK11MUT, as well as KRASMUT + KEAP1MUT to reduced response to immune checkpoint inhibitors (ICI) and even a negative impact on survival. Data focusing STK11 + KEAP1 co-mutations or the triple mutation (KRAS + STK11 + KEAP1) are scarce. The recent availability of KRAS-G12C inhibitors increases the clinical relevance of those co-mutations in KRAS-mutated NSCLC. MATERIALS AND METHODS We present a comprehensive bioinformatic analysis encompassing six datasets retrieved from cBioPortal. RESULTS Independent of the treatment, triple mutations and STK11MUT + KEAP1MUT were significantly associated with a reduced overall survival (OS). Across treatments, OS of patients with a KRAS G12C triple mutation was significantly reduced compared to patients with KRAS G12C-only. Under ICI-therapy, there was no significant difference in OS between patients harboring the KRAS G12C-only and patients with the KRAS G12C triple mutation, but a significant difference between patients harboring KRAS non-G12C and KRAS non-G12C triple mutations. Triple mutated primary tumors showed a significantly increased frequency of distant metastases to bone and adrenal glands compared to KRAS-only mutated tumors. Additionally, our drug response analysis in cancer cell lines harboring the triple mutations revealed the WNT pathway inhibitor XAV-939 as a potential future drug candidate for this mutational situation. CONCLUSION The triple mutation status may serve as a negative prognostic and predictive factor across treatments compared to KRASMUT-only. KRAS G12C generally seems to be a negative predictive marker for ICI-therapy.
Collapse
Affiliation(s)
- Myriam Boeschen
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| | - Christina Katharina Kuhn
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, University of Leipzig, Johannisallee 30, 04103 Leipzig, Germany
| | - Hubert Wirtz
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Hans-Jürgen Seyfarth
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Armin Frille
- Department of Respiratory Medicine, Leipzig University Medical Center, Liebigstrasse 20, 04103 Leipzig
| | - Florian Lordick
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrich T Hacker
- Department of Medicine II, University Cancer Center Leipzig (UCCL), Leipzig University Medical Center, Leipzig, Germany
| | - Ulrike Obeck
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Mathias Stiller
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Hendrik Bläker
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany
| | - Maximilian von Laffert
- Institute of Pathology, Leipzig University Medical Center, Liebigstraße 26, 04103 Leipzig, Germany.
| |
Collapse
|
8
|
Kiewisz J, Waśniewski T, Kieżun J, Skowrońska A, Kaczmarek MM, Szóstak B, Kowalczyk AE, Kmieć Z. WNT4 Gene and Protein Expression in Endometrial Cancer and Its Significance. Cancers (Basel) 2023; 15:4780. [PMID: 37835474 PMCID: PMC10571897 DOI: 10.3390/cancers15194780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND The inappropriate action of WNT4 and estrogens affects uterine homeostasis and function, and may lead to endometrial cancer (EC). OBJECTIVE The aim was to evaluate the alterations of WNT4 gene expression and WNT4 protein immunoreactivity (Ir) in EC, considering tumor characteristics, the clinicopathological association and estrogen dependence. METHODS WNT4 mRNA levels were compared between benign (control) endometrium (n = 8) and endometroid EC (EEC) and non-endometroid EC (non-EEC) samples (n = 28) using the real-time PCR technique. The WNT4-Ir and ERα-Ir were evaluated by immunohistochemistry (IHC). WNT4 mRNA gene and WNT4-Ir were correlated with clinicopathological and blood morphological parameters. Overall survival (OS) was assessed. The bioanalysis was utilized to study WNT4 expression in large patient cohort (n = 549). RESULTS WNT4 gene expression was decreased in EC samples (specifically in EEC but not in non-EEC) compared to the control. The WNT4 gene expression was also decreased in EC samples categorized by the tumor characteristics. There was no statistical difference in WNT4-Ir or ERα-Ir between the control and EC. There was no correlation between OS and WNT4 gene expression and WNT4-Ir. Bioanalysis showed that WNT4 and ESR1 gene expression alterations tended to be mutually exclusive. An alteration in WNT4 expression was found in different histological tumor types in a large group of EC patients. CONCLUSIONS There is a great need to evaluate the molecular background of EC. Our study suggests that the WNT4 gene has the potential to be a marker of functional estrogen signaling in EEC.
Collapse
Affiliation(s)
- Jolanta Kiewisz
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (J.K.); (A.E.K.); (Z.K.)
| | - Tomasz Waśniewski
- Department of Gynecology and Obstetrics, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland;
| | - Jacek Kieżun
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (J.K.); (A.E.K.); (Z.K.)
| | - Agnieszka Skowrońska
- Department of Human Physiology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland
| | - Monika M. Kaczmarek
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Science, 10-748 Olsztyn, Poland
| | - Błażej Szóstak
- Department of Pathomorphology, The Regional Specialist Hospital, 10-561 Olsztyn, Poland
| | - Anna E. Kowalczyk
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (J.K.); (A.E.K.); (Z.K.)
| | - Zbigniew Kmieć
- Department of Human Histology and Embryology, School of Medicine, Collegium Medicum, University of Warmia and Mazury, 10-082 Olsztyn, Poland; (J.K.); (A.E.K.); (Z.K.)
- Department of Histology, Medical University of Gdansk, 80-211 Gdansk, Poland
| |
Collapse
|
9
|
Zhang X, Schalke B, Kvell K, Kriegsmann K, Kriegsmann M, Graeter T, Preissler G, Ott G, Kurz K, Bulut E, Ströbel P, Marx A, Belharazem D. WNT4 overexpression and secretion in thymic epithelial tumors drive an autocrine loop in tumor cells in vitro. Front Oncol 2022; 12:920871. [PMID: 35965500 PMCID: PMC9372913 DOI: 10.3389/fonc.2022.920871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundWNT4-driven non-canonical signaling is crucial for homeostasis and age-related involution of the thymus. Abnormal WNT signaling is important in many cancers, but the role of WNT signaling in thymic tumors is largely unknown.Materials & MethodsExpression and function of WNT4 and FZD6 were analyzed using qRT–PCR, Western blot, ELISA, in biopsies of non-neoplastic thymi (NT), thymoma and thymic carcinomas. ShRNA techniques and functional assays were used in primary thymic epithelial cells (pTECs) and TC cell line 1889c. Cells were conventionally (2D) grown and in three-dimensional (3D) spheroids.ResultsIn biopsy, WHO classified B3 thymomas and TCs showed increased WNT4 expression compared with NTs. During short-term 2D culture, WNT4 expression and secretion declined in neoplastic pTECs but not in 3D spheroids or medium supplemented with recombinant WNT4 cultures. Under the latter condition, the growth of pTECs was accompanied by increased expression of non-canonical targets RAC1 and JNK. Down-regulation of WNT4 by shRNA induced cell death in pTECs derived from B3 thymomas and led to decreased RAC1, but not JNK protein phosphorylation. Pharmacological inhibition of NF-κB decreased both RAC1 and JNK phosphorylation in neoplastic pTECs.ConclusionsLack of the age-related decline of non-canonical WNT4 expression in TETs and restoration of declining WNT4 expression through exogeneous WNT4 or 3D culture of pTECs hints at an oncogenic role of WNT4 in TETs and is compatible with the WNT4 autocrine loop model. Crosstalk between WNT4 and NF-κB signaling may present a promising target for combined interventions in TETs.
Collapse
Affiliation(s)
- Xiaonan Zhang
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Berthold Schalke
- Department of Neurology, University of Regensburg, Regensburg, Germany
| | - Krisztian Kvell
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, University of Pecs, Pecs, Hungary
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Mark Kriegsmann
- Translational Lung Research Centre Heidelberg, German Centre for Lung Research, Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Thomas Graeter
- Department of Thoracic Surgery, University Medical Centre Erlangen, Erlangen, Germany
| | - Gerhard Preissler
- Department of Thoraxic Surgery, Clinic Schillerhöhe, Robert-Bosch-Hospital, Gerlingen, Löwenstein, Germany
| | - German Ott
- Department of Clinical Pathology, Robert-Bosch-Hospital, Stuttgart, Germany
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Katrin Kurz
- Department of Clinical Pathology, Robert-Bosch-Hospital, Stuttgart, Germany
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, Robert-Bosch-Hospital, Stuttgart, Germany
| | - Elena Bulut
- Department of Thoraxic Surgery, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Djeda Belharazem
- Institute of Pathology and Medical Research Center, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
- *Correspondence: Djeda Belharazem,
| |
Collapse
|
10
|
Assidi M, Buhmeida A, Al-Zahrani MH, Al-Maghrabi J, Rasool M, Naseer MI, Alkhatabi H, Alrefaei AF, Zari A, Elkhatib R, Abuzenadah A, Pushparaj PN, Abu-Elmagd M. The Prognostic Value of the Developmental Gene FZD6 in Young Saudi Breast Cancer Patients: A Biomarkers Discovery and Cancer Inducers OncoScreen Approach. Front Mol Biosci 2022; 9:783735. [PMID: 35237656 PMCID: PMC8883113 DOI: 10.3389/fmolb.2022.783735] [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: 09/26/2021] [Accepted: 01/05/2022] [Indexed: 12/21/2022] Open
Abstract
Wnt signalling receptors, Frizzleds (FZDs), play a pivotal role in many cellular events during embryonic development and cancer. Female breast cancer (BC) is currently the worldwide leading incident cancer type that cause 1 in 6 cancer-related death. FZD receptors expression in cancer was shown to be associated with tumour development and patient outcomes including recurrence and survival. FZD6 received little attention for its role in BC and hence we analysed its expression pattern in a Saudi BC cohort to assess its prognostic potential and unravel the impacted signalling pathway. Paraffin blocks from approximately 405 randomly selected BC patients aged between 25 and 70 years old were processed for tissue microarray using an automated tissue arrayer and then subjected to FZD6 immunohistochemistry staining using the Ventana platform. Besides, Ingenuity Pathway Analysis (IPA) knowledgebase was used to decipher the upstream and downstream regulators of FZD6 in BC. TargetScan and miRabel target-prediction databases were used to identify the potential microRNA to regulate FZD6 expression in BC. Results showed that 60% of the BC samples had a low expression pattern while 40% showed a higher expression level. FZD6 expression analysis showed a significant correlation with tumour invasion (p < 0.05), and borderline significance with tumour grade (p = 0.07). FZD6 expression showed a highly significant association with the BC patients’ survival outcomes. This was mainly due to the overall patients’ cohort where tumours with FZD6 elevated expression showed higher recurrence rates (DFS, p < 0.0001, log-rank) and shorter survival times (DSS, p < 0.02, log-rank). Interestingly, the FZD6 prognostic value was more potent in younger BC patients as compared to those with late onset of the disease. TargetScan microRNA target-prediction analysis and validated by miRabel showed that FZD6 is a potential target for a considerable number of microRNAs expressed in BC. The current study demonstrates a potential prognostic role of FZD6 expression in young BC female patients and provides a better understanding of the involved molecular silencing machinery of the Wnt/FZD6 signalling. Our results should provide a better understanding of FZD6 role in BC by adding more knowledge that should help in BC prevention and theranostics.
Collapse
Affiliation(s)
- Mourad Assidi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdelbaset Buhmeida
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maryam H. Al-Zahrani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Jaudah Al-Maghrabi
- Department of Pathology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
| | - Mahmood Rasool
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Muhammad I. Naseer
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heba Alkhatabi
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulmajeed F. Alrefaei
- Department of Biology, Jamoum University College, Umm Al-Qura University, Mecca, Saudi Arabia
| | - Ali Zari
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Razan Elkhatib
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Adel Abuzenadah
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Peter N. Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Transdisciplinary Research, Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Muhammad Abu-Elmagd
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- *Correspondence: Muhammad Abu-Elmagd,
| |
Collapse
|
11
|
Roles and action mechanisms of WNT4 in cell differentiation and human diseases: a review. Cell Death Discov 2021; 7:287. [PMID: 34642299 PMCID: PMC8511224 DOI: 10.1038/s41420-021-00668-w] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/24/2021] [Accepted: 09/20/2021] [Indexed: 02/07/2023] Open
Abstract
WNT family member 4 (WNT4), which belongs to the conserved WNT protein family, plays an important role in the development and differentiation of many cell types during the embryonic development and adult homeostasis. Increasing evidence has shown that WNT4 is a special ligand that not only activates the β-catenin independent pathway but also acts on β-catenin signaling based on different cellular processes. This article is a summary of the current knowledge about the expression, regulation, and function of WNT4 ligands and their signal pathways in cell differentiation and human disease processes. WNT4 is a promoter in osteogenic differentiation in bone marrow stromal cells (BMSCs) by participating in bone homeostasis regulation in osteoporotic diseases. Non-canonical WNT4 signaling is necessary for metabolic maturation of pancreatic β-cell. WNT4 is also necessary for decidual cell differentiation and decidualization, which plays an important role in preeclampsia. WNT4 promotes neuronal differentiation of neural stem cell and dendritic cell (DC) into conventional type 1 DC (cDC1). Besides, WNT4 mediates myofibroblast differentiation in the skin, kidney, lung, and liver during scarring or fibrosis. On the negative side, WNT4 is highly expressed in cancer tissues, playing a pro-carcinogenic role in many cancer types. This review provides an overview of the progress in elucidating the role of WNT4 signaling pathway components in cell differentiation in adults, which may provide useful clues for the diagnosis, prevention, and therapy of human diseases.
Collapse
|
12
|
Shafaroudi AM, Sharifi-Zarchi A, Rahmani S, Nafissi N, Mowla SJ, Lauria A, Oliviero S, Matin MM. Expression and Function of C1orf132 Long-Noncoding RNA in Breast Cancer Cell Lines and Tissues. Int J Mol Sci 2021; 22:6768. [PMID: 34201896 PMCID: PMC8268529 DOI: 10.3390/ijms22136768] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/25/2021] [Accepted: 06/02/2021] [Indexed: 11/24/2022] Open
Abstract
miR-29b2 and miR-29c play a suppressive role in breast cancer progression. C1orf132 (also named MIR29B2CHG) is the host gene for generating both microRNAs. However, the region also expresses longer transcripts with unknown functions. We employed bioinformatics and experimental approaches to decipher C1orf132 expression and function in breast cancer tissues. We also used the CRISPR/Cas9 technique to excise a predicted C1orf132 distal promoter and followed the behavior of the edited cells by real-time PCR, flow cytometry, migration assay, and RNA-seq techniques. We observed that C1orf132 long transcript is significantly downregulated in triple-negative breast cancer. We also identified a promoter for the longer transcripts of C1orf132 whose functionality was demonstrated by transfecting MCF7 cells with a C1orf132 promoter-GFP construct. Knocking-out the promoter by means of CRISPR/Cas9 revealed no alterations in the expression of the neighboring genes CD46 and CD34, while the expression of miR-29c was reduced by half. Furthermore, the promoter knockout elevated the migration ability of the edited cells. RNA sequencing revealed many up- and downregulated genes involved in various cellular pathways, including epithelial to mesenchymal transition and mammary gland development pathways. Altogether, we are reporting here the existence of an additional/distal promoter with an enhancer effect on miR-29 generation and an inhibitory effect on cell migration.
Collapse
Affiliation(s)
| | - Ali Sharifi-Zarchi
- Department of Computer Engineering, Sharif University of Technology, Tehran 11155-11365, Iran; (A.S.-Z.); (S.R.)
| | - Saeid Rahmani
- Department of Computer Engineering, Sharif University of Technology, Tehran 11155-11365, Iran; (A.S.-Z.); (S.R.)
| | - Nahid Nafissi
- Surgical Department, School of Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran;
| | - Seyed Javad Mowla
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran 14115-154, Iran;
| | - Andrea Lauria
- Department of Life Sciences and Systems Biology, University of Turin, 10126 Turin, Italy;
- Italian Institute for Genomic Medicine, 10060 Candiolo, Italy
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, University of Turin, 10126 Turin, Italy;
- Italian Institute for Genomic Medicine, 10060 Candiolo, Italy
| | - Maryam M. Matin
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran;
- Novel Diagnostics and Therapeutics Research Group, Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| |
Collapse
|
13
|
Zhang C, Cui T, Cai R, Wangpaichitr M, Mirsaeidi M, Schally AV, Jackson RM. Growth Hormone-Releasing Hormone in Lung Physiology and Pulmonary Disease. Cells 2020; 9:E2331. [PMID: 33096674 PMCID: PMC7589146 DOI: 10.3390/cells9102331] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/10/2020] [Accepted: 10/17/2020] [Indexed: 01/11/2023] Open
Abstract
Growth hormone-releasing hormone (GHRH) is secreted primarily from the hypothalamus, but other tissues, including the lungs, produce it locally. GHRH stimulates the release and secretion of growth hormone (GH) by the pituitary and regulates the production of GH and hepatic insulin-like growth factor-1 (IGF-1). Pituitary-type GHRH-receptors (GHRH-R) are expressed in human lungs, indicating that GHRH or GH could participate in lung development, growth, and repair. GHRH-R antagonists (i.e., synthetic peptides), which we have tested in various models, exert growth-inhibitory effects in lung cancer cells in vitro and in vivo in addition to having anti-inflammatory, anti-oxidative, and pro-apoptotic effects. One antagonist of the GHRH-R used in recent studies reviewed here, MIA-602, lessens both inflammation and fibrosis in a mouse model of bleomycin lung injury. GHRH and its peptide agonists regulate the proliferation of fibroblasts through the modulation of extracellular signal-regulated kinase (ERK) and Akt pathways. In addition to downregulating GH and IGF-1, GHRH-R antagonist MIA-602 inhibits signaling pathways relevant to inflammation, including p21-activated kinase 1-signal transducer and activator of transcription 3/nuclear factor-kappa B (PAK1-STAT3/NF-κB and ERK). MIA-602 induces fibroblast apoptosis in a dose-dependent manner, which is an effect that is likely important in antifibrotic actions. Taken together, the novel data reviewed here show that GHRH is an important peptide that participates in lung homeostasis, inflammation, wound healing, and cancer; and GHRH-R antagonists may have therapeutic potential in lung diseases.
Collapse
Affiliation(s)
- Chongxu Zhang
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
| | - Tengjiao Cui
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
| | - Renzhi Cai
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
| | - Medhi Wangpaichitr
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
| | - Mehdi Mirsaeidi
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Andrew V. Schally
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33101, USA
- Department of Pathology and Sylvester Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | - Robert M. Jackson
- Research Service, Miami VAHS, Miami, FL 33125, USA; (C.Z.); (T.C.); (R.C.); (M.W.); (M.M.); (A.V.S.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| |
Collapse
|
14
|
Marques P, Grossman AB, Korbonits M. The tumour microenvironment of pituitary neuroendocrine tumours. Front Neuroendocrinol 2020; 58:100852. [PMID: 32553750 DOI: 10.1016/j.yfrne.2020.100852] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 05/26/2020] [Accepted: 06/02/2020] [Indexed: 02/06/2023]
Abstract
The tumour microenvironment (TME) includes a variety of non-neoplastic cells and non-cellular elements such as cytokines, growth factors and enzymes surrounding tumour cells. The TME emerged as a key modulator of tumour initiation, progression and invasion, with extensive data available in many cancers, but little is known in pituitary tumours. However, the understanding of the TME of pituitary tumours has advanced thanks to active research in this field over the last decade. Different immune and stromal cell subpopulations, and several cytokines, growth factors and matrix remodelling enzymes, have been characterised in pituitary tumours. Studying the TME in pituitary tumours may lead to a better understanding of tumourigenic mechanisms, identification of biomarkers useful to predict aggressive disease, and development of novel therapies. This review summarises the current knowledge on the different TME cellular/non-cellular elements in pituitary tumours and provides an overview of their role in tumourigenesis, biological behaviour and clinical outcomes.
Collapse
Affiliation(s)
- Pedro Marques
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Ashley B Grossman
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| | - Márta Korbonits
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK.
| |
Collapse
|
15
|
Hu M, Xie J, Liu Z, Wang X, Liu M, Wang J. Comprehensive Analysis Identifying Wnt Ligands Gene Family for Biochemical Recurrence in Prostate Adenocarcinoma and Construction of a Nomogram. J Comput Biol 2020; 27:1656-1667. [PMID: 32298604 DOI: 10.1089/cmb.2019.0397] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
There is little research to explore the relationship between Wnt ligands gene family and biochemical recurrence of prostate adenocarcinoma. The purpose of this study was to systematically evaluate the role of Wnt ligands gene family in biochemical recurrence in prostate adenocarcinoma. RNA-seq transcriptome data and clinicopathological data of 489 prostate adenocarcinoma tissues and 51 nontumor tissues were obtained from The Cancer Genome Atlas. We developed a risk score model with the least absolute shrinkage and selection operator Cox regression algorithm. We used the X-tile program to derive the best threshold for risk scores, dividing patients into high-, intermediate-, and low-risk groups. Gene set enrichment analysis (GSEA) was performed. Nomogram was constructed based on the risk score and clinical features. The risk score = (0.192 × expression level of Wnt9A) + (0.732 × expression level of Wnt8B) + (0.051 × expression level of Wnt7B) + (-0.320 × expression level of Wnt3A). The risk score was an independent prognostic factor, with a hazard ratio of 1.298 (95% confidence interval: 1.046-1.612; p = 0.018). GSEA revealed that the Kyoto Encyclopedia of Genes and Genomes pathway of the four selected genes was closely related to malignancy-related biological processes. Nomogram was constructed based on the risk score and clinical features. The C index was 0.719, and the calibration curve showed that the nomogram performed well. In general, we comprehensively evaluated the association between Wnt ligands gene family and biochemical recurrence of prostate cancer. We developed a risk score model based on messenger RNA expression levels of several selected Wnt ligand family genes (Wnt3A, Wnt7B, Wnt8B, and Wnt9A), which was significantly associated with biochemical recurrence of prostate cancer. Our results might be helpful for future molecular studies focusing on the biochemical recurrence of prostate cancer.
Collapse
Affiliation(s)
- Maolin Hu
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jiangling Xie
- Department of Urology, Qianjiang Central Hospital of Chongqing Municipality, Chongqing, China
| | - Zhifeng Liu
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Xuan Wang
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Ming Liu
- Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| | - Jianye Wang
- Department of Urology, Peking University Fifth School of Clinical Medicine, Beijing, China.,Department of Urology, Beijing Hospital, National Center of Gerontology, Beijing, China
| |
Collapse
|
16
|
Chesnokova V, Melmed S. Growth hormone in the tumor microenvironment. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2019; 63:568-575. [PMID: 31939481 PMCID: PMC7025769 DOI: 10.20945/2359-3997000000186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/27/2019] [Indexed: 12/20/2022]
Abstract
Tumor development is a multistep process whereby local mechanisms enable somatic mutations during preneoplastic stages. Once a tumor develops, it becomes a complex organ composed of multiple cell types. Interactions between malignant and non-transformed cells and tissues create a tumor microenvironment (TME) comprising epithelial cancer cells, cancer stem cells, non-tumorous cells, stromal cells, immune-inflammatory cells, blood and lymphatic vascular network, and extracellular matrix. We review reports and present a hypothesis that postulates the involvement of growth hormone (GH) in field cancerization. We discuss GH contribution to TME, promoting epithelial-to-mesenchymal transition, accumulation of unrepaired DNA damage, tumor vascularity, and resistance to therapy. Arch Endocrinol Metab. 2019;63(6):568-75.
Collapse
Affiliation(s)
- Vera Chesnokova
- Pituitary CenterDepartment of MedicineCedars-Sinai Medical CenterLos AngelesCAUSAPituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shlomo Melmed
- Pituitary CenterDepartment of MedicineCedars-Sinai Medical CenterLos AngelesCAUSAPituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
17
|
Dong JJ, Ying L, Shi KQ. Expression of the Wnt ligands gene family and its relationship to prognosis in hepatocellular carcinoma. Cancer Cell Int 2019; 19:34. [PMID: 30814912 PMCID: PMC6376661 DOI: 10.1186/s12935-019-0743-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 01/31/2019] [Indexed: 12/14/2022] Open
Abstract
Background The Wnt gene family members are known to participate regulating various normal and pathological processes including tumorigenesis. However, the association between Wnt ligands gene family and prognosis in hepatocellular carcinoma has not been systematically studied. Therefore, we evaluated the role of Wnt ligands gene family in hepatocellular carcinoma using publicly available data from The Cancer Genome Atlas (TCGA). Methods Clinical information and RNA-Seq mRNA expression data were derived from TCGA hepatocellular carcinoma cohort. Differences in overall survival (OS) and disease-free survival (DFS) between increased and decreased expression groups (defined by X-tile analyses) of Wnt ligands gene family were compared using Kaplan-Meier method and Cox regression model, with p-values calculated via log-rank test. Gene Set Enrichment Analysis (GSEA) was performed. Results Multivariate analysis adjusted for patient age, sex, BMI, tumor grade, and TMN stage revealed that Wnt1, Wnt3 and Wnt5B expressions were independent prognostic factors for OS and DFS (OS: HR = 0.58, P = 0.006; HR = 0.65, P = 0.03; HR = 0.56, P = 0.023, respectively; DFS: HR = 0.52, P < 0.001; HR = 1.93, P = 0.003; HR = 0.59, P = 0.011, respectively). Furthermore, expression of Wnt1 and Wnt5B was significantly associated with TMN stage (P = 0.02 and P = 0.03 for OS; P = 0.02 and P = 0.02 for DFS). GSEA showed that nucleotide excision repair was differentially enriched in Wnt1 low expression phenotype and aminoacyl trna biosynthesis and basal transcription factors were differentially enriched in Wnt5B low expression phenotype. Conclusions Our results identified associations of several Wnt ligands with prognosis of HCC patients, indicating that these genes could serve as prognostic biomarkers of HCC.
Collapse
Affiliation(s)
- Jia-Jia Dong
- 1Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Li Ying
- 1Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang China
| | - Ke-Qing Shi
- 2Precision Medical Center, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, Wenzhou, China
| |
Collapse
|
18
|
Li Y, Wang M, Huang BW, Ping Y, You J, Gao JQ. Transcriptome-wide elucidation of liposomal formulations for anticancer drug delivery. Int J Nanomedicine 2017; 12:8557-8572. [PMID: 29238192 PMCID: PMC5716676 DOI: 10.2147/ijn.s148975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Although widely used in chemotherapy, free doxorubicin (Dox) might enhance cell malignancy undesirably. Liposomal Dox (Doxlipo) has been clinically approved for the treatment of breast cancer due to reduced systematical toxicity and increased tumor targeting, yet the transcriptome-wide elucidation of the Doxlipo formulations remains elusive. To this end, we explored the impact of two Dox liposomal formulations, Doxlipo mainly containing hydrogenated soy phosphatidylcholine or 1,2-dipalmitoyl-sn-glycero-3-phosphocholine, on the transcriptional pattern of MCF-7 cells. The two types of Dox liposomal formulations with different drug release kinetics were investigated to reveal the relationship between the formulation and tumor malignancy. Interestingly, we found that liposomal formulation significantly altered the transcriptional pattern of a wide range of genes. Under equivalent dosage of Dox, free Dox substantially changed the expression of ANK1, ACTA2, GPR87, GDF15, FZD6, and WNT4 in MCF-7 cells. Notably, free Dox induced much higher expression of ABCB1 and significantly enhanced the cell migration behavior in comparison with HSPC Doxlipo under a similar level of cytotoxicity. Finally, siRNA targeting GPR87 was codelivered with cationic Doxlipo to reduce the expression of malignancy-related genes. Our study, for the first time, provides an overview of the influence of formulation on the malignancy at transcriptional level and reveals the relationship between cytotoxicity and cell malignancy from the formulation aspect, offering valuable reference for the future formulation design for anticancer drug delivery.
Collapse
Affiliation(s)
- Ying Li
- Institute of Pharmaceutics, College of Pharmaceutical Sciences.,Zhejiang Province Key Laboratory of Anticancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Meng Wang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences.,Zhejiang Province Key Laboratory of Anticancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Bu-Wei Huang
- Institute of Pharmaceutics, College of Pharmaceutical Sciences.,Zhejiang Province Key Laboratory of Anticancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| | - Yuan Ping
- School of Materials Science and Engineering, Nanyang Technological University, Singapore
| | - Jian You
- Institute of Pharmaceutics, College of Pharmaceutical Sciences
| | - Jian-Qing Gao
- Institute of Pharmaceutics, College of Pharmaceutical Sciences.,Zhejiang Province Key Laboratory of Anticancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, People's Republic of China
| |
Collapse
|
19
|
Moraleva A, Magoulas C, Polzikov M, Hacot S, Mertani HC, Diaz JJ, Zatsepina O. Involvement of the specific nucleolar protein SURF6 in regulation of proliferation and ribosome biogenesis in mouse NIH/3T3 fibroblasts. Cell Cycle 2017; 16:1979-1991. [PMID: 28873013 DOI: 10.1080/15384101.2017.1371880] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nucleolar proteins which link cell proliferation to ribosome biogenesis are regarded to be potentially oncogenic. Here, in order to examine the involvement of an evolutionary conserved nucleolar protein SURF6/Rrp14 in proliferation and ribosome biogenesis in mammalian cells, we established stably transfected mouse NIH/3T3 fibroblasts capable of conditional overexpression of the protein. Cell proliferation was monitored in real-time, and various cell cycle parameters were quantified based on flow cytometry, Br-dU-labeling and conventional microscopy data. We show that overexpression of SURF6 accelerates cell proliferation and promotes transition through all cell cycle phases. The most prominent SURF6 pro-proliferative effects include a significant reduction of the population doubling time, from 19.8 ± 0.7 to 16.2 ± 0.5 hours (t-test, p < 0.001), and of the length of cell division cycle, from 17.6 ± 0.6 to 14.0 ± 0.4 hours (t-test, p < 0.001). The later was due to the shortening of all cell cycle phases but the length of G1 period was reduced most, from 5.7 ± 0.4 to 3.8 ± 0.3 hours, or by ∼30%, (t-test, p < 0.05). By Northern blots and qRT-PCR, we further showed that the acceleration of cell proliferation was concomitant with an accumulation of rRNA species along both ribosomal subunit maturation pathways. It is evident, therefore, that like the yeast homologue Rrp14, mammalian SURF6 is involved in various steps of rRNA processing during ribosome biogenesis. We concluded that SURF6 is a novel positive regulator of proliferation and G1/S transition in mammals, implicating that SURF6 is a potential oncogenic protein, which can be further studied as a putative target in anti-cancer therapy.
Collapse
Affiliation(s)
- Anastasiia Moraleva
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation
| | - Charalambos Magoulas
- b Centre for Investigative and Diagnostic Oncology, Department of Natural Sciences, School of Science and Technology , Middlesex University , London , United Kingdom
| | - Mikhail Polzikov
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation
| | - Sabine Hacot
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Hichem C Mertani
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Jean-Jacques Diaz
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Olga Zatsepina
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation.,c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| |
Collapse
|
20
|
Abstract
The growth hormone (GH) and insulin-like growth factor-1 (IGF1) axis is the key regulator of longitudinal growth, promoting postnatal bone and muscle growth. The available data suggest that GH expression by tumour cells is associated with the aetiology and progression of various cancers such as endometrial, breast, liver, prostate, and colon cancer. Accordingly there has been increased interest in targeting GH-mediated signal transduction in a therapeutic setting. Because GH has endocrine, autocrine, and paracrine actions, therapeutic strategies will need to take into account systemic and local functions. Activation of related hormone receptors and crosstalk with other signalling pathways are also key considerations.
Collapse
Affiliation(s)
- Jo K Perry
- Liggins Institute, University of Auckland, 1023 Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, University of Auckland, 1023 Auckland, New Zealand
| | - Zheng-Sheng Wu
- Department of Pathology, Anhui Medical University, Hefei, Anhui, PR China
| | - Hichem C Mertani
- Centre de Recherche en Cancérologie de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1052-Centre National de la Recherche Scientifique (CNRS) 5286, Centre Léon Bérard, Université Claude Bernard Lyon I, Université de Lyon, Lyon, France
| | - Tao Zhu
- Hefei National Laboratory for Physical Sciences at Microscale and School of Life Sciences, University of Science and Technology of China, Hefei, Anhui, PR China
| | - Peter E Lobie
- Cancer Science Institute of Singapore and Department of Pharmacology, National University of Singapore, 117456 Singapore; Tsinghua Berkeley Shenzhen Institute, Tsinghua University Graduate School, Shenzhen, PR China.
| |
Collapse
|
21
|
Schmucker HS, Park JP, Coissieux MM, Bentires-Alj M, Feltus FA, Booth BW. RNA Expression Profiling Reveals Differentially Regulated Growth Factor and Receptor Expression in Redirected Cancer Cells. Stem Cells Dev 2017; 26:646-655. [DOI: 10.1089/scd.2016.0340] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Hannah S. Schmucker
- Department of Biological Sciences, Clemson University, Clemson, South Carolina
| | - Jang Pyo Park
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, South Carolina
| | - Marie-May Coissieux
- Friedrich-Miescher Institute for Biomedical Research, Basel, Switzerland
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Friedrich-Miescher Institute for Biomedical Research, Basel, Switzerland
- Department of Biomedicine, University of Basel, University Hospital Basel, Basel, Switzerland
| | - F. Alex Feltus
- Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina
| | - Brian W. Booth
- Institute for Biological Interfaces of Engineering, Clemson University, Clemson, South Carolina
- Department of Bioengineering, Clemson University, Clemson, South Carolina
| |
Collapse
|