1
|
Choi SH, Kim DY. Regulation of Tumor Microenvironment through YAP/TAZ under Tumor Hypoxia. Cancers (Basel) 2024; 16:3030. [PMID: 39272887 PMCID: PMC11394240 DOI: 10.3390/cancers16173030] [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: 07/16/2024] [Revised: 08/22/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
In solid tumors such as hepatocellular carcinoma (HCC), hypoxia is one of the important mechanisms of cancer development that closely influences cancer development, survival, and metastasis. The development of treatments for cancer was temporarily revolutionized by immunotherapy but continues to be constrained by limited response rates and the resistance and high costs required for the development of new and innovative strategies. In particular, solid tumors, including HCC, a multi-vascular tumor type, are sensitive to hypoxia and generate many blood vessels for metastasis and development, making it difficult to treat HCC, not only with immunotherapy but also with drugs targeting blood vessels. Therefore, in order to develop a treatment strategy for hypoxic tumors, various mechanisms must be explored and analyzed to treat these impregnable solid tumors. To date, tumor growth mechanisms linked to hypoxia are known to be complex and coexist with various signal pathways, but recently, mechanisms related to the Hippo signal pathway are emerging. Interestingly, Hippo YAP/TAZ, which appear during early tumor and normal tumor growth, and YAP/TAZ, which appear during hypoxia, help tumor growth and proliferation in different directions. Peculiarly, YAP/TAZ, which have different phosphorylation directions in the hypoxic environment of tumors, are involved in cancer proliferation and metastasis in various carcinomas, including HCC. Analyzing the mechanisms that regulate the function and expression of YAP in addition to HIF in the complex hypoxic environment of tumors may lead to a variety of anti-cancer strategies and combining HIF and YAP/TAZ may develop the potential to change the landscape of cancer treatment.
Collapse
Affiliation(s)
- Sung Hoon Choi
- Institute of Health & Environment, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs Inc., Seoul 08826, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
- Yonsei Liver Cancer Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea
| |
Collapse
|
2
|
Hantusch B, Kenner L, Stanulović VS, Hoogenkamp M, Brown G. Targeting Androgen, Thyroid Hormone, and Vitamin A and D Receptors to Treat Prostate Cancer. Int J Mol Sci 2024; 25:9245. [PMID: 39273194 PMCID: PMC11394715 DOI: 10.3390/ijms25179245] [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: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 09/15/2024] Open
Abstract
The nuclear hormone family of receptors regulates gene expression. The androgen receptor (AR), upon ligand binding and homodimerization, shuttles from the cytosol into the nucleus to activate gene expression. Thyroid hormone receptors (TRs), retinoic acid receptors (RARs), and the vitamin D receptor (VDR) are present in the nucleus bound to chromatin as a heterodimer with the retinoid X receptors (RXRs) and repress gene expression. Ligand binding leads to transcription activation. The hormonal ligands for these receptors play crucial roles to ensure the proper conduct of very many tissues and exert effects on prostate cancer (PCa) cells. Androgens support PCa proliferation and androgen deprivation alone or with chemotherapy is the standard therapy for PCa. RARγ activation and 3,5,3'-triiodo-L-thyronine (T3) stimulation of TRβ support the growth of PCa cells. Ligand stimulation of VDR drives growth arrest, differentiation, and apoptosis of PCa cells. Often these receptors are explored as separate avenues to find treatments for PCa and other cancers. However, there is accumulating evidence to support receptor interactions and crosstalk of regulatory events whereby a better understanding might lead to new combinatorial treatments.
Collapse
Affiliation(s)
- Brigitte Hantusch
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
| | - Lukas Kenner
- Department of Pathology, Department for Experimental and Laboratory Animal Pathology, Medical University of Vienna, 1010 Vienna, Austria;
- Comprehensive Cancer Center, Medical University Vienna, 1090 Vienna, Austria
- Unit of Laboratory Animal Pathology, University of Veterinary Medicine Vienna, 1210 Vienna, Austria
- Department of Molecular Biology, Umeå University, 901 87 Umeå, Sweden
- Christian Doppler Laboratory for Applied Metabolomics, Medical University Vienna, 1090 Vienna, Austria
- Center for Biomarker Research in Medicine (CBmed), 8010 Graz, Austria
| | - Vesna S. Stanulović
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Maarten Hoogenkamp
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK; (V.S.S.); (M.H.)
| | - Geoffrey Brown
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
3
|
Powała K, Żołek T, Brown G, Kutner A. Molecular Interactions of Selective Agonists and Antagonists with the Retinoic Acid Receptor γ. Int J Mol Sci 2024; 25:6568. [PMID: 38928275 PMCID: PMC11203493 DOI: 10.3390/ijms25126568] [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: 05/21/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
All-trans retinoic acid (ATRA), the major active metabolite of all-trans retinol (vitamin A), is a key hormonal signaling molecule. In the adult organism, ATRA has a widespread influence on processes that are crucial to the growth and differentiation of cells and, in turn, the acquisition of mature cell functions. Therefore, there is considerable potential in the use of retinoids to treat diseases. ATRA binds to the retinoic acid receptors (RAR) which, as activated by ATRA, selectively regulate gene expression. There are three main RAR isoforms, RARα, RARβ, and RARγ. They each have a distinct role, for example, RARα and RARγ regulate myeloid progenitor cell differentiation and hematopoietic stem cell maintenance, respectively. Hence, targeting an isoform is crucial to developing retinoid-based therapeutics. In principle, this is exemplified when ATRA is used to treat acute promyelocytic leukemia (PML) and target RARα within PML-RARα oncogenic fusion protein. ATRA with arsenic trioxide has provided a cure for the once highly fatal leukemia. Recent in vitro and in vivo studies of RARγ have revealed the potential use of agonists and antagonists to treat diseases as diverse as cancer, heterotopic ossification, psoriasis, and acne. During the final drug development there may be a need to design newer compounds with added modifications to improve solubility, pharmacokinetics, or potency. At the same time, it is important to retain isotype specificity and activity. Examination of the molecular interactions between RARγ agonists and the ligand binding domain of RARγ has revealed aspects to ligand binding that are crucial to RARγ selectivity and compound activity and key to designing newer compounds.
Collapse
Affiliation(s)
- Katarzyna Powała
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland
| | - Teresa Żołek
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland
| | - Geoffrey Brown
- School of Biomedical Sciences, Institute of Clinical Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK;
| | - Andrzej Kutner
- Department of Drug Chemistry Pharmaceutical and Biomedical Analysis, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha, 02-097 Warsaw, Poland;
| |
Collapse
|
4
|
Hussein NA, Ebid SA, Ahmad MA, Khedr GE, Saad DM. The possible correlation between miR-762, Hippo signaling pathway, TWIST1, and SMAD3 in lung cancer and chronic inflammatory diseases. Sci Rep 2024; 14:8246. [PMID: 38589525 PMCID: PMC11001855 DOI: 10.1038/s41598-024-58704-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/02/2024] [Indexed: 04/10/2024] Open
Abstract
MicroRNAs are small RNA molecules that have a significant role in translational repression and gene silencing through binding to downstream target mRNAs. MiR-762 can stimulate the proliferation and metastasis of various types of cancer. Hippo pathway is one of the pathways that regulate tissue development and carcinogenesis. Dysregulation of this pathway plays a vital role in the progression of cancer. This study aimed to evaluate the possible correlation between miR-762, the Hippo signaling pathway, TWIST1, and SMAD3 in patients with lung cancer, as well as patients with chronic inflammatory diseases. The relative expression of miR-762, MST1, LATS2, YAP, TWIST1, and SMAD3 was determined in 50 lung cancer patients, 30 patients with chronic inflammatory diseases, and 20 healthy volunteers by real-time PCR. The levels of YAP protein and neuron-specific enolase were estimated by ELISA and electrochemiluminescence immunoassay, respectively. Compared to the control group, miR-762, YAP, TWIST1, and SMAD3 expression were significantly upregulated in lung cancer patients and chronic inflammatory patients, except SMAD3 was significantly downregulated in chronic inflammatory patients. MST1, LATS2, and YAP protein were significantly downregulated in all patients. MiR-762 has a significant negative correlation with MST1, LATS2, and YAP protein in lung cancer patients and with MST1 and LATS2 in chronic inflammatory patients. MiR-762 may be involved in the induction of malignant behaviors in lung cancer through suppression of the Hippo pathway. MiR-762, MST1, LATS2, YAP mRNA and protein, TWIST1, and SMAD3 may be effective diagnostic biomarkers in both lung cancer patients and chronic inflammatory patients. High YAP, TWIST1, SMA3 expression, and NSE level are associated with a favorable prognosis for lung cancer.
Collapse
Affiliation(s)
- Neveen A Hussein
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt.
| | - Samia A Ebid
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Mohammad A Ahmad
- Clinical Pathology Department, Military Medical Academy, Cairo, Egypt
| | - Gamal E Khedr
- Clinical Pathology Department, Tanta Cancer Center, Tanta, Egypt
| | - Dina M Saad
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
5
|
Venghateri JB, Dassa B, Morgenstern D, Shreberk-Shaked M, Oren M, Geiger B. Deciphering the involvement of the Hippo pathway co-regulators, YAP/TAZ in invadopodia formation and matrix degradation. Cell Death Dis 2023; 14:290. [PMID: 37185904 PMCID: PMC10130049 DOI: 10.1038/s41419-023-05769-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 03/21/2023] [Accepted: 03/23/2023] [Indexed: 05/17/2023]
Abstract
Invadopodia are adhesive, actin-rich protrusions formed by metastatic cancer cells that degrade the extracellular matrix and facilitate invasion. They support the metastatic cascade by a spatially and temporally coordinated process whereby invading cells bind to the matrix, degrade it by specific metalloproteinases, and mechanically penetrate diverse tissue barriers by forming actin-rich extensions. However, despite the apparent involvement of invadopodia in the metastatic process, the molecular mechanisms that regulate invadopodia formation and function are still largely unclear. In this study, we have explored the involvement of the key Hippo pathway co-regulators, namely YAP, and TAZ, in invadopodia formation and matrix degradation. Toward that goal, we tested the effect of depletion of YAP, TAZ, or both on invadopodia formation and activity in multiple human cancer cell lines. We report that the knockdown of YAP and TAZ or their inhibition by verteporfin induces a significant elevation in matrix degradation and invadopodia formation in several cancer cell lines. Conversely, overexpression of these proteins strongly suppresses invadopodia formation and matrix degradation. Proteomic and transcriptomic profiling of MDA-MB-231 cells, following co-knockdown of YAP and TAZ, revealed a significant change in the levels of key invadopodia-associated proteins, including the crucial proteins Tks5 and MT1-MMP (MMP14). Collectively, our findings show that YAP and TAZ act as negative regulators of invadopodia formation in diverse cancer lines, most likely by reducing the levels of essential invadopodia components. Dissecting the molecular mechanisms of invadopodia formation in cancer invasion may eventually reveal novel targets for therapeutic applications against invasive cancer.
Collapse
Affiliation(s)
- Jubina Balan Venghateri
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Bareket Dassa
- Bioinformatics Unit, Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | | | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Benjamin Geiger
- Department of Immunology and Regenerative Biology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
6
|
Wang Y, Chen H, Yu J, Kang W, To KF. Recent insight into the role and therapeutic potential of YAP/TAZ in gastrointestinal cancers. Biochim Biophys Acta Rev Cancer 2022; 1877:188787. [PMID: 36041574 DOI: 10.1016/j.bbcan.2022.188787] [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: 06/06/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 11/18/2022]
Abstract
With the rapid development of cancer treatment, gastrointestinal (GI) cancers are still the most prevalent malignancies with high morbidity and mortality worldwide. Dysregulation of the Hippo signaling pathway has been recognized to play a critical role during cancer development and adopted for monitoring disease progression and therapy response. Despite the well-documented tumor proliferation and metastasis, recent efforts in two core Hippo components, Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ), have identified as the driving forces behind cancer metabolism, stemness, tumor immunity, and therapy resistance. Understanding the molecular mechanisms by which YAP/TAZ facilitates the tumorigenesis and progression of GI cancer, and identifying novel therapeutic strategies for targeting YAP/TAZ are crucial to GI cancer treatment and prevention. In this study, we summarize the latest findings on the function and regulatory mechanisms of YAP/TAZ in GI cancers, and highlight the translational significance of targeting YAP/TAZ for cancer therapies.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Huarong Chen
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jun Yu
- Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Wei Kang
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Ka Fai To
- Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; Institute of Digestive Disease, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China; State Key Laboratory of Translational Oncology, Sir Y.K. Pao Cancer Centre, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China.
| |
Collapse
|
7
|
Zhang Y, Wang X, Zhou X. Functions of Yes-association protein (YAP) in cancer progression and anticancer therapy resistance. BRAIN SCIENCE ADVANCES 2022. [DOI: 10.26599/bsa.2022.9050008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
The Hippo pathway, a highly conserved kinase cascade, regulates cell proliferation, apoptosis, organ size, and tissue homeostasis. Dysregulation of this pathway reportedly plays an important role in the progression of various human cancers. Yes-association protein (YAP), the Hippo pathway’s core effector, is considered a marker for cancer therapy and patient prognosis. In addition, studies have indicated that YAP is involved in promoting anticancer drug resistance. This review summarizes current knowledge on YAP’s role in cancer progression, anticancer drug resistance, and advances in the development of YAP-targeting drugs. A thorough understanding of the complex interactions among molecular, cellular, and environmental factors concerning YAP function in cancer progression may provide new insight into the underlying mechanism of anticancer drug resistance. It might lead to improved prognosis through novel combined therapies.
Collapse
Affiliation(s)
- Yu Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiang Wang
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- The Graduate School, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- These authors contributed equally to this work
| | - Xiuping Zhou
- Institute of Nervous System Diseases, Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
- Department of Neurosurgery, The Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, Jiangsu, China
| |
Collapse
|
8
|
Brás MM, Sousa SR, Carneiro F, Radmacher M, Granja PL. Mechanobiology of Colorectal Cancer. Cancers (Basel) 2022; 14:1945. [PMID: 35454852 PMCID: PMC9028036 DOI: 10.3390/cancers14081945] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022] Open
Abstract
In this review, the mechanobiology of colorectal cancer (CRC) are discussed. Mechanotransduction of CRC is addressed considering the relationship of several biophysical cues and biochemical pathways. Mechanobiology is focused on considering how it may influence epithelial cells in terms of motility, morphometric changes, intravasation, circulation, extravasation, and metastization in CRC development. The roles of the tumor microenvironment, ECM, and stroma are also discussed, taking into account the influence of alterations and surface modifications on mechanical properties and their impact on epithelial cells and CRC progression. The role of cancer-associated fibroblasts and the impact of flow shear stress is addressed in terms of how it affects CRC metastization. Finally, some insights concerning how the knowledge of biophysical mechanisms may contribute to the development of new therapeutic strategies and targeting molecules and how mechanical changes of the microenvironment play a role in CRC disease are presented.
Collapse
Affiliation(s)
- Maria Manuela Brás
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
- Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Susana R. Sousa
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
- Instituto Superior de Engenharia do Porto (ISEP), Instituto Politécnico do Porto (IPP), 4200-072 Porto, Portugal
| | - Fátima Carneiro
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), 4200-465 Porto, Portugal
- Serviço de Patologia, Centro Hospitalar Universitário de São João (CHUSJ), 4200-319 Porto, Portugal
- Faculdade de Medicina da Universidade do Porto (FMUP), 4200-319 Porto, Portugal
| | - Manfred Radmacher
- Institute for Biophysics, University of Bremen, 28334 Bremen, Germany
| | - Pedro L. Granja
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; (M.M.B.); (S.R.S.); (F.C.); (P.L.G.)
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
9
|
Jiang Y, Xie WJ, Chen RW, You WW, Ye WL, Chen H, Chen WX, Xu JP. The Hippo Signaling Core Components YAP and TAZ as New Prognostic Factors in Lung Cancer. Front Surg 2022; 9:813123. [PMID: 35388363 PMCID: PMC8977465 DOI: 10.3389/fsurg.2022.813123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 01/31/2022] [Indexed: 01/15/2023] Open
Abstract
BackgroundThe Hippo pathway is an essential signaling cascade that regulates cell and organ growth. However, there is no consensus about (i) the expression levels of the Hippo signaling core components yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) in lung cancer, especially in small cell lung cancer (SCLC), or (ii) their association with the prognosis of patients with SCLC.MethodsWe screened relevant articles and identified eligible studies in the PubMed, EMBASE, COCHRANE, and WanFang databases. A combined analysis was performed to investigate (i) the expression levels of the major effectors, YAP and TAZ, in lung cancer and its subsets and (ii) their prognostic role in lung cancer, especially in SCLC.ResultsIn total, 6 studies related to TAZ and 13 studies concerning YAP were enrolled in this meta-analysis. We found that high TAZ expression was significantly associated with poor overall survival (OS) of patients with non-small cell lung cancer (NSCLC) in the overall population [Ph < 0.001, crude hazard ratio (HR) = 1.629, 95% CI = 1.199–2.214 for TAZ expression; Ph = 0.029, adjusted HR = 2.127, 95% CI = 1.307–3.460 for TAZ], the Caucasian population (Ph = 0.043, crude HR = 1.233, 95% CI = 1.030–1.477 for TAZ expression), and the Asian population (Ph = 0.551, adjusted HR = 2.676, 95% CI = 1.798–3.982 for TAZ). Moreover, there was a significant negative association between YAP expression and an unsatisfactory survival of patients with lung cancer (Ph = 0.327, crude HR = 1.652, 95% CI = 1.211–2.253 for YAP expression) and patients with NSCLC [disease-free survival (DFS): Ph = 0.693, crude HR = 2.562, 95% CI = 1.876–3.499 for YAP expression; Ph = 0.920, crude HR = 2.617, 95% CI = 1.690–4.052 for YAP-mRNA; OS: Ph = 0.878, crude HR = 1.777, 95% CI = 1.233–2.562 for YAP expression], especially in the Asian population (DFS: Ph = 0.414, crude HR = 2.515, 95% CI = 1.755–3.063; OS: Ph = 0.712, crude HR = 1.772, 95% CI = 1.214–2.587). However, no association was observed in the multivariate combined analysis. High YAP expression was significantly associated with short OS of patients with SCLC in our combined multivariate analysis in the Asian population (Ph = 0.289, crude HR = 4.482, 95% CI = 2.182–9.209), but not with crude data (Ph = 0.033, crude HR = 1.654, 95% CI = 0.434–6.300).ConclusionThe Hippo pathway is involved in carcinogenesis and progression of NSCLC and SCLC, and high expression levels of YAP and TAZ are independent and novel prognostic factors for lung cancer.
Collapse
Affiliation(s)
- Yu Jiang
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
- Yu Jiang
| | - Wen-Jing Xie
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Rong-Wei Chen
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Wei-Wei You
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Wei-Lin Ye
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Hong Chen
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
| | - Wen-Xu Chen
- Department of Clinical Laboratory, Fuzhou Second Hospital Affiliated to Xiamen University, Fuzhou, China
- *Correspondence: Wen-Xu Chen
| | - Jian-Ping Xu
- Department of Clinical Laboratory Medicine, Fujian Medical University, Fuzhou, China
- Jian-Ping Xu
| |
Collapse
|
10
|
Kudryashova TV, Dabral S, Nayakanti S, Ray A, Goncharov DA, Avolio T, Shen Y, Rode A, Pena A, Jiang L, Lin D, Baust J, Bachman TN, Graumann J, Ruppert C, Guenther A, Schmoranzer M, Grobs Y, Lemay SE, Tremblay E, Breuils-Bonnet S, Boucherat O, Mora AL, DeLisser H, Zhao J, Zhao Y, Bonnet S, Seeger W, Pullamsetti SS, Goncharova EA. Noncanonical HIPPO/MST Signaling via BUB3 and FOXO Drives Pulmonary Vascular Cell Growth and Survival. Circ Res 2022; 130:760-778. [PMID: 35124974 PMCID: PMC8897250 DOI: 10.1161/circresaha.121.319100] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 01/19/2022] [Indexed: 11/16/2022]
Abstract
RATIONALE The MSTs (mammalian Ste20-like kinases) 1/2 are members of the HIPPO pathway that act as growth suppressors in adult proliferative diseases. Pulmonary arterial hypertension (PAH) manifests by increased proliferation and survival of pulmonary vascular cells in small PAs, pulmonary vascular remodeling, and the rise of pulmonary arterial pressure. The role of MST1/2 in PAH is currently unknown. OBJECTIVE To investigate the roles and mechanisms of the action of MST1 and MST2 in PAH. METHODS AND RESULTS Using early-passage pulmonary vascular cells from PAH and nondiseased lungs and mice with smooth muscle-specific tamoxifen-inducible Mst1/2 knockdown, we found that, in contrast to canonical antiproliferative/proapoptotic roles, MST1/2 act as proproliferative/prosurvival molecules in human PAH pulmonary arterial vascular smooth muscle cells and pulmonary arterial adventitial fibroblasts and support established pulmonary vascular remodeling and pulmonary hypertension in mice with SU5416/hypoxia-induced pulmonary hypertension. By using unbiased proteomic analysis, gain- and loss-of function approaches, and pharmacological inhibition of MST1/2 kinase activity by XMU-MP-1, we next evaluated mechanisms of regulation and function of MST1/2 in PAH pulmonary vascular cells. We found that, in PAH pulmonary arterial adventitial fibroblasts, the proproliferative function of MST1/2 is caused by IL-6-dependent MST1/2 overexpression, which induces PSMC6-dependent downregulation of forkhead homeobox type O 3 and hyperproliferation. In PAH pulmonary arterial vascular smooth muscle cells, MST1/2 acted via forming a disease-specific interaction with BUB3 and supported ECM (extracellular matrix)- and USP10-dependent BUB3 accumulation, upregulation of Akt-mTORC1, cell proliferation, and survival. Supporting our in vitro observations, smooth muscle-specific Mst1/2 knockdown halted upregulation of Akt-mTORC1 in small muscular PAs of mice with SU5416/hypoxia-induced pulmonary hypertension. CONCLUSIONS Together, this study describes a novel proproliferative/prosurvival role of MST1/2 in PAH pulmonary vasculature, provides a novel mechanistic link from MST1/2 via BUB3 and forkhead homeobox type O to the abnormal proliferation and survival of pulmonary arterial vascular smooth muscle cells and pulmonary arterial adventitial fibroblasts, remodeling and pulmonary hypertension, and suggests new target pathways for therapeutic intervention.
Collapse
Affiliation(s)
- Tatiana V. Kudryashova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Swati Dabral
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Sreenath Nayakanti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Arnab Ray
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dmitry A. Goncharov
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Theodore Avolio
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yuanjun Shen
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Analise Rode
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Andressa Pena
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lifeng Jiang
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
| | - Derek Lin
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
| | - Jeffrey Baust
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Timothy N. Bachman
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Johannes Graumann
- Biomolecular Mass Spectrometry, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Clemens Ruppert
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, 35392, Germany
| | - Andreas Guenther
- Department of Internal Medicine, Member of the DZL, Member of CPI, Justus Liebig University, Giessen, 35392, Germany
| | - Mario Schmoranzer
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
| | - Yann Grobs
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Sarah Eve Lemay
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Eve Tremblay
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | | | - Olivier Boucherat
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Ana L. Mora
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Horace DeLisser
- Department of Pathology and Laboratory Medicine, Pulmonary Vascular Disease Program, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jing Zhao
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Yutong Zhao
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Sébastien Bonnet
- Department of Medicine, Faculty of Medicine, Université Laval, Quebec City, Canada
| | - Werner Seeger
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the DZL, Justus Liebig University, Giessen, Germany
| | - Soni S. Pullamsetti
- Max Planck Institute for Heart and Lung Research, Department of Lung Development and Remodeling, Member of the German Center for Lung Research (DZL), Bad Nauheim, Germany
- Department of Internal Medicine, Universities of Giessen and Marburg Lung Center (UGMLC), Institute for Lung Health (ILH), Cardio-Pulmonary Institute (CPI), Member of the DZL, Justus Liebig University, Giessen, Germany
| | - Elena A. Goncharova
- Lung Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of California, Davis School of Medicine, Davis, CA, USA
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
11
|
Kim M, Ly SH, Xie Y, Duronio GN, Ford-Roshon D, Hwang JH, Sulahian R, Rennhack JP, So J, Gjoerup O, Talamas JA, Grandclaudon M, Long HW, Doench JG, Sethi NS, Giannakis M, Hahn WC. YAP1 and PRDM14 converge to promote cell survival and tumorigenesis. Dev Cell 2022; 57:212-227.e8. [PMID: 34990589 PMCID: PMC8827663 DOI: 10.1016/j.devcel.2021.12.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 08/15/2021] [Accepted: 12/03/2021] [Indexed: 01/26/2023]
Abstract
The transcriptional co-activator YAP1 oncogene is the downstream effector of the Hippo pathway, which regulates tissue homeostasis, organ size, regeneration, and tumorigenesis. Multiple cancers are dependent on sustained expression of YAP1 for cell proliferation, survival, and tumorigenesis, but the molecular basis of this oncogene dependency is not well understood. To identify genes that can functionally substitute for YAP1, we performed a genome-scale genetic rescue screen in YAP1-dependent colon cancer cells expressing an inducible YAP1-specific shRNA. We found that the transcription factor PRDM14 rescued cell proliferation and tumorigenesis upon YAP1 suppression in YAP1-dependent cells, xenografts, and colon cancer organoids. YAP1 and PRDM14 individually activated the transcription of calmodulin 2 (CALM2) and a glucose transporter SLC2A1 upon YAP1 suppression, and CALM2 or SLC2A1 expression was required for the rescue of YAP1 suppression. Together, these findings implicate PRDM14-mediated transcriptional upregulation of CALM2 and SLC2A1 as key components of oncogenic YAP1 signaling and dependency.
Collapse
Affiliation(s)
- Miju Kim
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Seav Huong Ly
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Yingtian Xie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Gina N Duronio
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Dane Ford-Roshon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Justin H Hwang
- Masonic Cancer Center and Department of Medicine, University of Minnesota-Twin Cities, Minneapolis, MN 55455, USA
| | - Rita Sulahian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan P Rennhack
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jonathan So
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Ole Gjoerup
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Jessica A Talamas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | - Henry W Long
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - John G Doench
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nilay S Sethi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - William C Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
12
|
Cianci P, Restini E. Artificial intelligence in colorectal cancer management. Artif Intell Cancer 2021; 2:79-89. [DOI: 10.35713/aic.v2.i6.79] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/22/2021] [Accepted: 12/29/2021] [Indexed: 02/06/2023] Open
Abstract
Artificial intelligence (AI) is a new branch of computer science involving many disciplines and technologies. Since its application in the medical field, it has been constantly studied and developed. AI includes machine learning and neural networks to create new technologies or to improve existing ones. Various AI supporting systems are available for a personalized and novel strategy for the management of colorectal cancer (CRC). This mini-review aims to summarize the progress of research and possible clinical applications of AI in the investigation, early diagnosis, treatment, and management of CRC, to offer elements of knowledge as a starting point for new studies and future applications.
Collapse
Affiliation(s)
- Pasquale Cianci
- Department of Surgery and Traumatology, ASL BAT, Lorenzo Bonomo Hospital, Andria 76123, Puglia, Italy
| | - Enrico Restini
- Department of Surgery and Traumatology, ASL BAT, Lorenzo Bonomo Hospital, Andria 76123, Puglia, Italy
| |
Collapse
|
13
|
Lei T, Gao Y, Duan Y, Cui C, Zhang L, Si M. Inhibition of zinc finger protein 367 exerts a tumor suppressive role in colorectal cancer by affecting the activation of oncogenic YAP1 signaling. ENVIRONMENTAL TOXICOLOGY 2021; 36:2278-2290. [PMID: 34351699 DOI: 10.1002/tox.23341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/12/2021] [Accepted: 07/24/2021] [Indexed: 06/13/2023]
Abstract
Zinc finger protein 367 (ZNF367) has been documented as a new cancer-related protein that exerts a pivotal role in the carcinogenesis of multiple cancers. However, whether ZNF367 plays a role in colorectal cancer has not been fully understood. Our data showed that ZNF367 expression was higher in colorectal cancer. Depletion of ZNF367 weakened the proliferative and invasive capabilities of colorectal cancer cells. Up-regulation of ZNF367 enhanced the in vitro malignant features of colorectal cancer cells. Knockdown of ZNF367 impeded the activation of Yes-associated protein (YAP1). Reactivation of YAP1 reversed the ZNF367-knockdown-mediated anticancer effects. Suppression of YAP1 significantly abolished ZNF367-overexpression-induced tumor-promotion effects. Depletion of ZNF367 repressed the tumorigenicity of colorectal cancer cells in vivo. These findings demonstrate that ZNF367 is overexpressed in colorectal cancer and acts as a potential tumor-promoter that contributes to the proliferation and invasion of colorectal cancer by enhancing the activation of YAP1 signaling.
Collapse
Affiliation(s)
- Ting Lei
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of General Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Ya Gao
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yuhong Duan
- Endocrinology Department, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Chunli Cui
- Department of Pharmacy, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Li Zhang
- Institutional Pharmacy, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| | - Mingming Si
- Department of General Surgery, Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, China
| |
Collapse
|
14
|
lncRNA cytoskeleton regulator RNA (CYTOR): Diverse functions in metabolism, inflammation and tumorigenesis, and potential applications in precision oncology. Genes Dis 2021; 10:415-429. [DOI: 10.1016/j.gendis.2021.08.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/20/2021] [Indexed: 12/19/2022] Open
|
15
|
Zhu M, Peng R, Liang X, Lan Z, Tang M, Hou P, Song JH, Mak CSL, Park J, Zheng SE, Huang A, Ma X, Chen R, Chang Q, Logothetis CJ, Jain AK, Lin SH, Katayama H, Hanash S, Wang G. P4HA2-induced prolyl hydroxylation suppresses YAP1-mediated prostate cancer cell migration, invasion, and metastasis. Oncogene 2021; 40:6049-6056. [PMID: 34471235 PMCID: PMC8526415 DOI: 10.1038/s41388-021-02000-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 08/09/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
Yes-associated protein 1 (YAP1), a key player in the Hippo pathway, has been shown to play a critical role in tumor progression. However, the role of YAP1 in prostate cancer cell invasion, migration, and metastasis is not well defined. Through functional, transcriptomic, epigenomic, and proteomic analyses, we showed that prolyl hydroxylation of YAP1 plays a critical role in the suppression of cell migration, invasion, and metastasis in prostate cancer. Knockdown (KD) or knockout (KO) of YAP1 led to an increase in cell migration, invasion, and metastasis in prostate cancer cells. Microarray analysis showed that the EMT pathway was activated in Yap1-KD cells. ChIP-seq analysis showed that YAP1 target genes are enriched in pathways regulating cell migration. Mass spectrometry analysis identified P4H prolyl hydroxylase in the YAP1 complex and YAP1 was hydroxylated at multiple proline residues. Proline-to-alanine mutations of YAP1 isoform 3 identified proline 174 as a critical residue, and its hydroxylation suppressed cell migration, invasion, and metastasis. KO of P4ha2 led to an increase in cell migration and invasion, which was reversed upon Yap1 KD. Our study identified a novel regulatory mechanism of YAP1 by which P4HA2-dependent prolyl hydroxylation of YAP1 determines its transcriptional activities and its function in prostate cancer metastasis.
Collapse
Affiliation(s)
- Ming Zhu
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ruiqing Peng
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Sun Yat-sen University Cancer Center; State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xin Liang
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zhengdao Lan
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ming Tang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Pingping Hou
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jian H. Song
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Celia Sze Ling Mak
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jiwon Park
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shui-er Zheng
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ailing Huang
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Xingdi Ma
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ruidong Chen
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Qing Chang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher J. Logothetis
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhinav K. Jain
- Department of Epigenetics and Molecular Carcinogenesis & Epigenomics Profiling Core Facility, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sue-Hwa Lin
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hiroyuki Katayama
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Samir Hanash
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Guocan Wang
- Department of Genitourinary Medical Oncology and the David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA.,Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
16
|
Sun R, He XY, Mei C, Ou CL. Role of exosomal long non-coding RNAs in colorectal cancer. World J Gastrointest Oncol 2021; 13:867-878. [PMID: 34457192 PMCID: PMC8371516 DOI: 10.4251/wjgo.v13.i8.867] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/11/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
Exosomes are a class of small extracellular vesicles, 30-150 nm in diameter, that transfer biological information (e.g., DNA, RNA, and protein) via cell-to-cell communication. Exosomes play critical roles in the occurrence and development of human cancers, including colorectal cancer (CRC). Recent studies have shown that long non-coding RNAs (lncRNAs) can be encapsulated in exosomes, which transfer lncRNAs from secretory cells into recipient cells. This process affects the progression of CRC, since exosomal lncRNAs display special and extensive functions in CRC tumorigenesis, including malignant proliferation, metastasis, chemoresistance, and inflammatory response. Moreover, due to their specificity and sensitivity, exosomal lncRNAs are released into body fluids (e.g., urine, sputum, and plasma), which have the potential to be biomarkers of CRC tumorigenesis within screening efforts and medical and epidemiologic research. In this review, we aim to clarify the function and mechanism of exosomal lncRNAs in CRC tumorigenesis and provide a strategy for early diagnosis and medical treatment of this malignancy.
Collapse
Affiliation(s)
- Ru Sun
- Department of Blood Transfusion, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Xiao-Yun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Cheng Mei
- Department of Blood Transfusion, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| | - Chun-Lin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan Province, China
| |
Collapse
|
17
|
Noe O, Filipiak L, Royfman R, Campbell A, Lin L, Hamouda D, Stanbery L, Nemunaitis J. Adenomatous polyposis coli in cancer and therapeutic implications. Oncol Rev 2021; 15:534. [PMID: 34267890 PMCID: PMC8256374 DOI: 10.4081/oncol.2021.534] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/22/2021] [Indexed: 02/06/2023] Open
Abstract
Inactivating mutations of the adenomatous polyposis coli (APC) gene and consequential upregulation of the Wnt signaling pathway are critical initiators in the development of colorectal cancer (CRC), the third most common cancer in the United States for both men and women. Emerging evidence suggests APCmutations are also found in gastric, breast and other cancers. The APC gene, located on chromosome 5q, is responsible for negatively regulating the b-catenin/Wnt pathway by creating a destruction complex with Axin/Axin2, GSK-3b, and CK1. In the event of an APC mutation, b-catenin accumulates, translocates to the cell nucleus and increases the transcription of Wnt target genes that have carcinogenic consequences in gastrointestinal epithelial stem cells. A literature review was conducted to highlight carcinogenesis related to APC mutations, as well as preclinical and clinical studies for potential therapies that target steps in inflammatory pathways, including IL-6 transduction, and Wnt pathway signaling regulation. Although a range of molecular targets have been explored in murine models, relatively few pharmacological agents have led to substantial increases in survival for patients with colorectal cancer clinically. This article reviews a range of molecular targets that may be efficacious targets for tumors with APC mutations.
Collapse
Affiliation(s)
- Olivia Noe
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Louis Filipiak
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Rachel Royfman
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Austin Campbell
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Leslie Lin
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Danae Hamouda
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | - Laura Stanbery
- Department of Medicine, University of Toledo College of Medicine and Life Sciences, Toledo, OH
| | | |
Collapse
|
18
|
Yin F, Dong J, Kang LI, Liu X. Hippo-YAP signaling in digestive system tumors. Am J Cancer Res 2021; 11:2495-2507. [PMID: 34249412 PMCID: PMC8263672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/23/2021] [Indexed: 06/13/2023] Open
Abstract
The Hippo pathway is an evolutionally conserved pathway and plays an important role in regulating tissue hemostasis and organ size control. Deregulation of the Hippo pathway is implicated in various human digestive system tumors. The past two decades have witnessed the discovery and elucidation of key signaling components and molecular mechanisms of the Hippo pathway. Among these, the signaling transducers YAP/TAZ are in the center of this complex network to sense and respond to extracellular cues such as cell contact, matrix stiffness and growth factors. In this review, we summarize the biological and clinical significance of Hippo-YAP signaling in the digestive system tumors, and explore the novel therapeutic strategies for targeting Hippo-YAP signaling.
Collapse
Affiliation(s)
- Feng Yin
- Department of Pathology and Anatomical Sciences, University of MissouriColumbia, Missouri, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred and Pamela Buffett Cancer Center, University of Nebraska Medical CenterOmaha, Nebraska, USA
| | - Liang-I Kang
- Department of Pathology and Immunology, Washington University School of MedicineSt. Louis, Missouri, USA
| | - Xiuli Liu
- Department of Pathology, Immunology and Lab Medicine, University of FloridaGainesville, Florida, USA
| |
Collapse
|
19
|
Wu G, Hao C, Qi X, Nie J. Effect of Yes Associated Protein 1 Silence on Proliferation and Apoptosis of Bladder Cancer Cells. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Yes Associated Protein 1 (YAP) can act as either an oncoprotein or a tumor suppressor in different cellular contexts. However, the reports about the direct role of YAP silence in bladder cancer cells are rare. We designed loss-off-function experiments to investigate the effect of YAP
knockdown on bladder cancer cell proliferation, cell cycle and cell apoptosis. We examined YAP expression in human bladder cancer and paracancerous tissues using RT-qPCR, western blot and immunohisto-chemistry. YAP short hairpin RNA (shRNA) was successfully constructed and transfected into
T24 cells to knockdown YAP. Cell proliferation, cell cycle and cell apoptosis were analyzed by CCK-8 and flow cytometry. We found the expression levels of YAP mRNA and protein were significantly increased in the bladder cancer tissues when compared with that in the paracancerous tissues. shRNA
YAP inhibited cell proliferation, induced cell cycle arrest at G1 phase, and induced cell apoptosis. In conclusion, our findings provided the first evidence that YAP knockdown could inhibit cell proliferation and induce cell apoptosis of bladder cancer cells. YAP inhibition may be beneficial
in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Gaoliang Wu
- Department of Urology, Jiangxi Cancer, Hospital, Nanchang 330029, Jiangxi, P. R. China
| | - Chao Hao
- Department of Urology, Jiangxi Cancer, Hospital, Nanchang 330029, Jiangxi, P. R. China
| | - Xueliang Qi
- Department of Urology, Jiangxi Cancer, Hospital, Nanchang 330029, Jiangxi, P. R. China
| | - Jianqiang Nie
- Department of Urology, Jiangxi Cancer, Hospital, Nanchang 330029, Jiangxi, P. R. China
| |
Collapse
|
20
|
He X, Yu B, Kuang G, Wu Y, Zhang M, Cao P, Ou C. Long noncoding RNA DLEU2 affects the proliferative and invasive ability of colorectal cancer cells. J Cancer 2021; 12:428-437. [PMID: 33391439 PMCID: PMC7738996 DOI: 10.7150/jca.48423] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/24/2020] [Indexed: 12/17/2022] Open
Abstract
Emerging evidence indicates that long noncoding RNAs (lncRNAs) are closely associated with colorectal cancer (CRC) tumorigenesis. One example is lncRNA Deleted in Lymphocytic Leukemia 2 (DLEU2). However, how DLEU2 contributes to CRC is still poorly understood. This study sought to investigate the effects of DLEU2 on CRC pathogenesis, and the underlying mechanism involved. Using a quantitative real-time polymerase chain reaction (qRT-PCR) assay, we demonstrated that the expression levels of DLEU2 in 45 pairs of CRC tissues were higher than those in the corresponding normal colon mucosal tissues. In addition, CRC patients with high DLEU2 expression levels exhibited poor overall survival (OS) and recurrence-free survival (RFS), as determined by analyses and measurements from the GEO and GEPIA databases. When DLEU2 was silenced using short interfering RNA (siRNA) in CRC cell line, the results demonstrated that DLEU2 silencing suppressed CRC cell tumorigenesis in vitro, which was associated with decreased expression of cyclin dependent kinase 6(CDK6), ZEB1, and ZEB2 as well as enhancing the expression of Cyclin-dependent kinase inhibitor 1A (CDKN1A). Taken together, the results of this study suggested that DLEU2 may play critical roles in the progression of CRC and may serve as a prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Bingbing Yu
- Department of Pathology, Dezhou People's Hospital, Dezhou 253056, Shandong, China
| | - Gaoyan Kuang
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Yongrong Wu
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, Hunan, China
| | - Meili Zhang
- Department of Pathology, Dezhou People's Hospital, Dezhou 253056, Shandong, China
| | - Pengfei Cao
- Department of Hematology, Xiangya hospital, Central South University, Changsha 410008, Hunan, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
21
|
Increased Cytoplasmic Yes-associated Protein (YAP) Expression in Mismatch Repair Protein-Proficient Colorectal Cancer With High-grade Tumor Budding and Reduced Autophagy Activity. Appl Immunohistochem Mol Morphol 2020; 29:305-312. [PMID: 33264106 DOI: 10.1097/pai.0000000000000888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 10/26/2020] [Indexed: 01/07/2023]
Abstract
Yes-associated protein (YAP) is a transcriptional coactivator regulated by autophagy that stimulates colorectal cancer (CRC) progression through activation of epithelial-mesenchymal transition (EMT), represented by tumor budding. The associations between these components in CRC are unknown. Archived surgically resected CRCs with known mismatch repair protein (MMR) status were retrieved (n=81; 2010 to 2016). Electronic medical records were reviewed for clinicopathologic variables including pathologic TNM stage and clinical stage. Tumor budding was graded according to consensus guidelines. Cytoplasmic and nuclear YAP and p62 (autophagy substrate) immunoreactivity were semiquantitatively scored within tumor samples. The Student t test, Fisher exact test, χ2 test, and Spearman correlation coefficient were performed with P<0.05 as a significance level. MMR proficiency (MMR-P) status correlated with high-grade tumor budding. The extent of cytoplasmic YAP staining and pathologic N stage was associated with tumor budding in multivariate analysis. Cytoplasmic YAP expression correlated with higher cytoplasmic p62 expression, suggesting an inverse correlation between autophagy activation and cytoplasmic YAP expression. Nuclear YAP expression correlated with pathologic N stage and clinical stage. A correlation between MMR-P status and tumor budding, combined with correlations between cytoplasmic YAP, tumor budding and p62 raise the possibility of 2 distinct neoplastic pathways concerning autophagy and YAP; one displaying relative activation of YAP and EMT, being commonly observed in MMR-P, and another with less active YAP and EMT, but active autophagy, being commonly seen in MMR-deficient CRC. Nuclear YAP staining could be useful in prognostication.
Collapse
|
22
|
Gulfo J, Rotondo F, Ávalos de León CG, Cornide-Petronio ME, Fuster C, Gracia-Sancho J, Jiménez-Castro MB, Peralta C. FGF15 improves outcomes after brain dead donor liver transplantation with steatotic and non-steatotic grafts in rats. J Hepatol 2020; 73:1131-1143. [PMID: 32422221 DOI: 10.1016/j.jhep.2020.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 04/29/2020] [Accepted: 05/06/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Donation after brain death (DBD) grafts are associated with reduced graft quality and function post liver transplantation (LT). We aimed to elucidate i) the impact of FGF15 levels on DBD grafts; ii) whether this impact resulted from altered intestinal FXR-FGF15; iii) whether administration of FGF15 to donors after brain death could confer a benefit on graft function post LT; and iv) whether FGF15 affects bile acid (BA) accumulation. METHODS Steatotic and non-steatotic grafts from DBD donors and donors without brain death were transplanted in rats. FGF15 was administered alone or combined with either a BA (cholic acid) or a YAP inhibitor. RESULTS Brain death induced intestinal damage and downregulation of FXR. The resulting reduced intestinal FGF15 was associated with low hepatic FGF15 levels, liver damage and regenerative failure. Hepatic FGFR4-Klb - the receptor for FGF15 - was downregulated whereas CYP7A1 was overexpressed, resulting in BA accumulation. FGF15 administration to DBD donors increased hepatic FGFR4-Klb, reduced CYP7A1 and normalized BA levels. The benefit of FGF15 on liver damage was reversed by cholic acid, whereas its positive effect on regeneration was maintained. YAP signaling in DBD donors was activated after FGF15 treatment. When a YAP inhibitor was administered, the benefits of FGF15 on regeneration were abolished, whereas its positive effect on hepatic damage remained. Neither the Hippo-YAP-BA nor the BA-IQGAP1-YAP axis was involved in the benefits of FGF15. CONCLUSION Alterations in the gut-liver axis contribute to the reduced quality of DBD grafts and the associated pathophysiology of LT. FGF15 pre-treatment in DBD donors protected against damage and promoted cell proliferation. LAY SUMMARY After brain death, potential liver donors have reduced intestinal FXR, which is associated with reduced intestinal, circulatory and hepatic levels of FGF15. A similar reduction in the cell-surface receptor complex Fgfr4/Klb is observed, whereas CYP7A1 is overexpressed; together, these molecular events result in the dangerous accumulation of bile acids, leading to damage and regenerative failure in brain dead donor grafts. Herein, we demonstrate that when such donors receive appropriate doses of FGF15, CYP7A1 levels and hepatic bile acid toxicity are reduced, and liver regeneration is promoted.
Collapse
Affiliation(s)
- José Gulfo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Floriana Rotondo
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | | | | | - Carla Fuster
- Pathology Department, Hospital Clinic, Barcelona, Spain
| | - Jordi Gracia-Sancho
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas (CIBEREHD), Barcelona, Spain; Liver Vascular Biology Research Group, IDIBAPS, Barcelona, Spain
| | | | - Carmen Peralta
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas (CIBEREHD), Barcelona, Spain.
| |
Collapse
|
23
|
Wang Y, Nie H, He X, Liao Z, Zhou Y, Zhou J, Ou C. The emerging role of super enhancer-derived noncoding RNAs in human cancer. Theranostics 2020; 10:11049-11062. [PMID: 33042269 PMCID: PMC7532672 DOI: 10.7150/thno.49168] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 08/23/2020] [Indexed: 02/06/2023] Open
Abstract
Super enhancers (SEs) are large clusters of adjacent enhancers that drive the expression of genes which regulate cellular identity; SE regions can be enriched with a high density of transcription factors, co-factors, and enhancer-associated epigenetic modifications. Through enhanced activation of their target genes, SEs play an important role in various diseases and conditions, including cancer. Recent studies have shown that SEs not only activate the transcriptional expression of coding genes to directly regulate biological functions, but also drive the transcriptional expression of non-coding RNAs (ncRNAs) to indirectly regulate biological functions. SE-derived ncRNAs play critical roles in tumorigenesis, including malignant proliferation, metastasis, drug resistance, and inflammatory response. Moreover, the abnormal expression of SE-derived ncRNAs is closely related to the clinical and pathological characterization of tumors. In this review, we summarize the functions and roles of SE-derived ncRNAs in tumorigenesis and discuss their prospective applications in tumor therapy. A deeper understanding of the potential mechanism underlying the action of SE-derived ncRNAs in tumorigenesis may provide new strategies for the early diagnosis of tumors and targeted therapy.
Collapse
|
24
|
Wang Y, He X, Nie H, Zhou J, Cao P, Ou C. Application of artificial intelligence to the diagnosis and therapy of colorectal cancer. Am J Cancer Res 2020; 10:3575-3598. [PMID: 33294256 PMCID: PMC7716173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/14/2020] [Indexed: 06/12/2023] Open
Abstract
Artificial intelligence (AI) is a relatively new branch of computer science involving many disciplines and technologies, including robotics, speech recognition, natural language and image recognition or processing, and machine learning. Recently, AI has been widely applied in the medical field. The effective combination of AI and big data can provide convenient and efficient medical services for patients. Colorectal cancer (CRC) is a common type of gastrointestinal cancer. The early diagnosis and treatment of CRC are key factors affecting its prognosis. This review summarizes the research progress and clinical application value of AI in the investigation, early diagnosis, treatment, and prognosis of CRC, to provide a comprehensive theoretical basis for AI as a promising diagnostic and treatment tool for CRC.
Collapse
Affiliation(s)
- Yutong Wang
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Xiaoyun He
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
- Department of Endocrinology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Hui Nie
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South UniversityChangsha 410008, Hunan, China
| |
Collapse
|
25
|
Li D, Bao J, Yao J, Li J. lncRNA USP2-AS1 promotes colon cancer progression by modulating Hippo/YAP1 signaling. Am J Transl Res 2020; 12:5670-5682. [PMID: 33042447 PMCID: PMC7540120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 08/22/2020] [Indexed: 06/11/2023]
Abstract
Dysregulation of Hippo signaling by long non-coding RNA (lncRNA) contributes to colon adenocarcinoma (COAD) progression, while the underlying mechanisms remain elusive. Our study shows that lncRNA USP2-AS1 is a Yes-associated protein 1 (YAP1) binding lncRNA, and inactivates Hippo signaling in COAD cells. Moreover, our data indicated that USP2-AS1 lowered the phosph-YAP (S127), elevated the total level of YAP1, and triggered the expression of downstream target genes in COAD cells. The loss- and gain-of function assays demonstrated that USP2-AS1 promotes cellular proliferation and metastasis of COAD cells. Clinically, the USP2-AS1 levels were significantly elevated in COAD tissues and were positively correlated with tumor grade, size, and TNM stage. Collectively, these findings demonstrated that USP2-AS1 modulates and regulates Hippo signaling in COAD and could be a valuable therapeutic target.
Collapse
Affiliation(s)
- Dongying Li
- Department of Digestive, The First Affiliated Hospital of Zhengzhou University No. 1 East Jianshe Road, Zhengzhou 450052, Henan, China
| | - Jie Bao
- Department of Digestive, The First Affiliated Hospital of Zhengzhou University No. 1 East Jianshe Road, Zhengzhou 450052, Henan, China
| | - Jiayuan Yao
- Department of Digestive, The First Affiliated Hospital of Zhengzhou University No. 1 East Jianshe Road, Zhengzhou 450052, Henan, China
| | - Jiansheng Li
- Department of Digestive, The First Affiliated Hospital of Zhengzhou University No. 1 East Jianshe Road, Zhengzhou 450052, Henan, China
| |
Collapse
|
26
|
Xie P, Mo JL, Liu JH, Li X, Tan LM, Zhang W, Zhou HH, Liu ZQ. Pharmacogenomics of 5-fluorouracil in colorectal cancer: review and update. Cell Oncol (Dordr) 2020; 43:989-1001. [PMID: 32474853 DOI: 10.1007/s13402-020-00529-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is a disease with high morbidity and mortality rates. 5-fluorouracil (5-FU) is the first-line recommended drug for chemotherapy in patients with CRC, and it has a good effect on a variety of other solid tumors as well. Unfortunately, however, due to the emergence of drug resistance the effectiveness of treatment may be greatly reduced. In the past decade, major progress has been made in the field of 5-FU drug resistance in terms of molecular mechanisms, pre-clinical (animal) models and clinical trials. CONCLUSIONS In this article we systematically review and update current knowledge on 5-FU pharmacogenomics related to drug uptake and activation, the expression and activity of target enzymes (DPD, TS and MTHFR) and key signaling pathways in CRC. Furthermore, a summary of drug combination strategies aimed at targeting specific genes and/or pathways to reverse 5-FU resistance is provided. Based on this, we suggest that causal relationships between genes, pathways and drug sensitivity should be systematically considered from a multidimensional perspective. In the design of research methods, emerging technologies such as CRISPR-Cas, TALENS and patient-derived xenograft models should be applied as far as possible to improve the accuracy of clinically relevant results.
Collapse
Affiliation(s)
- Pan Xie
- Department of Clinical Pharmacology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, People's Republic of China
| | - Jun-Luan Mo
- Shenzhen Center for Chronic Disease Control, 518020, Shenzhen, People's Republic of China
| | - Jin-Hong Liu
- Shenzhen Center for Chronic Disease Control, 518020, Shenzhen, People's Republic of China
| | - Xi Li
- Department of Clinical Pharmacology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, People's Republic of China
| | - Li-Ming Tan
- Department of Pharmacy, The Second People's Hospital of Huaihua City, 418000, Huaihua, People's Republic of China
| | - Wei Zhang
- Department of Clinical Pharmacology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, People's Republic of China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China.,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, People's Republic of China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology and National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, People's Republic of China. .,Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, 410078, Changsha, People's Republic of China.
| |
Collapse
|
27
|
Wang B, Shao W, Shi Y, Liao J, Chen X, Wang C. Verteporfin induced SUMOylation of YAP1 in endometrial cancer. Am J Cancer Res 2020; 10:1207-1217. [PMID: 32368396 PMCID: PMC7191103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 03/01/2020] [Indexed: 06/11/2023] Open
Abstract
Yes-associated protein (YAP or YAP1) has been proposed to function as an oncogene in most cancers, with nuclear localization of YAP1 correlating with poor prognosis. Photosensitizer Verteporfin has been proven as an inhibitor of YAP1 through preventing the combination of YAP1 with TEA domain transcription factor (TEAD). We showed previously that the total and phospho-levels of YAP1 were related to the clinical characteristics and outcomes in endometrial cancer (EC) patients, and that YAP1 promoted the proliferation and metastasis of EC cells in vitro cell line studies and in animal models. We also reported that Verteporfin inhibited cell growth and induced cell death through inhibiting YAP1 in EC in our previous study. However, the mechanism of how Verteporfin inhibits the function of YAP1 remains unclear. In this study, we analyzed the global effects of Verteporfin on cell function by using Reverse Phase Protein Arrays (RPPA) and Ingenuity Pathway Analysis (IPA). Furthermore, we demonstrated that Verteporfin induced the SUMOylation of YAP1 for the first time. Interestingly, we found that the SUMOylation of YAP1 was regulated by YAP1 phosphorylation. Together, our study revealed a novel mechanism by which Verteporfin inhibits the function of YAP1 through regulating YAP1 SUMOylation. Our study may provide a rationale for the clinical use of Verteporfin in endometrial cancer by targeting YAP1.
Collapse
Affiliation(s)
- Bo Wang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Wenyu Shao
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Yue Shi
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Jiongbo Liao
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Xiaojun Chen
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| | - Chao Wang
- Department of Obstetrics and Gynecology, Obstetrics and Gynecology Hospital, Fudan UniversityShanghai, People’s Republic of China
- Shanghai Key Laboratory of Female Reproductive Endocrine Related DiseasesShanghai 200011, People’s Republic of China
| |
Collapse
|
28
|
Duan LM, Liu JY, Yu CW, Fan JX, Li T, Yang JX, Zheng YB, Liu FC, He ZT, Yuan HL, Wu XH, Luo CL. PLCε knockdown prevents serine/glycine metabolism and proliferation of prostate cancer by suppressing YAP. Am J Cancer Res 2020; 10:196-210. [PMID: 32064161 PMCID: PMC7017741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 12/29/2019] [Indexed: 06/10/2023] Open
Abstract
The metabolic reprogramming is an important basis for the development of many tumors, including prostate cancer (PCa). Metabolic changes in many amino acids consist of serine and glycine affect the biological behavior of them. Phospholipase C epsilon (PLCε) plays an important role as an oncogene. However, its role in regulating amino acid metabolism remains unclear. In this study, results found significantly positive correlation between PLCε and Yes-associated protein (YAP) in PCa tissues. LC-MS/MS and GC-MS results further displayed abnormally elevated levels of serine, glycine and its some downstream metabolites in the blood of PCa patients. Secondly, PLCε knockdown can inhibit serine/glycine producing and proliferation of PCa both in vivo and in vitro. Mechanistically, PLCε may affect the serine/glycine metabolism by regulating dephosphorylation and nuclear translocation of YAP. More interestingly, verteporfin (VP, a specific inhibitor of YAP) could effectively enhance the PLCε-depletion induced inhibition of serine/glycine secretion and growth. Overall, this research revealed the possibility of anomalous serine/glycine levels in the blood for the diagnosis of PCa, identified the important role of the PLCε/YAP axis in regulating serine/glycine metabolism, cell proliferation and tumor growth, and suggested the combination of VP with PLCε-depletion may provide a new idea for the treatment of PCa.
Collapse
Affiliation(s)
- Li-Mei Duan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Jia-Yu Liu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Chao-Wen Yu
- Center for Clinical Molecular Medicine, Children’s Hospital of Chongqing Medical UniversityChongqing 400014, China
| | - Jia-Xin Fan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Ting Li
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Jin-Xiao Yang
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Yong-Bo Zheng
- Department of Urology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Feng-Chun Liu
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Zhen-Ting He
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Hong-Ling Yuan
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| | - Xiao-Hou Wu
- Department of Urology, The First Affiliated Hospital of Chongqing Medical UniversityChongqing 400016, China
| | - Chun-Li Luo
- Key Laboratory of Clinical Laboratory Diagnostics (Ministry of Education), School of Laboratory Medicine, Chongqing Medical UniversityChongqing 400016, China
| |
Collapse
|
29
|
Jiménez-Martínez M, Ostalé CM, van der Burg LR, Galán-Martínez J, Hardwick JCH, López-Pérez R, Hawinkels LJAC, Stamatakis K, Fresno M. DUSP10 Is a Regulator of YAP1 Activity Promoting Cell Proliferation and Colorectal Cancer Progression. Cancers (Basel) 2019; 11:cancers11111767. [PMID: 31717606 PMCID: PMC6896144 DOI: 10.3390/cancers11111767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/31/2019] [Accepted: 11/05/2019] [Indexed: 12/24/2022] Open
Abstract
Cell contact inhibition (CCI) is deregulated in cancer. Colorectal cancer (CRC) is the third most commonly diagnosed cancer worldwide. We found that dual-specificity phosphatase 10 (DUSP10) is involved in CRC. DUSP10 overexpression increased the growth of CRC cell lines and mouse xenografts, while the opposite phenotype was observed by DUSP10 silencing. High cell density (HD) induced DUSP10 expression in CRC cell lines, particularly within the nucleus. Yes-associated protein 1 (YAP1) is activated by dephosphorylation, controlling organ growth and CCI, both processes being deregulated in CRC. Expression levels and localization of DUSP10 matched with YAP1 levels in CRC cell lines. DUSP10 and YAP1 co-immunoprecipitated and their interaction was dependent on YAP1 Ser397. The existence of DUSP10 and YAP1 pathway in vivo was confirmed by using a transgenic Drosophila model. Finally, in CRC patients’ samples, high levels of nuclear DUSP10 correlated with nuclear YAP1 in epithelial tumor tissue. Strong nuclear DUSP10 staining also correlated with high tumor stage and poor survival. Overall, these findings describe a DUSP10–YAP1 molecular link in CRC cell lines promoting cell growth in HD. We present evidence suggesting a pro-tumorigenic role of nuclear DUSP10 expression in CRC patients.
Collapse
Affiliation(s)
- Marta Jiménez-Martínez
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (M.J.-M.); (J.G.-M.); (R.L.-P.); (K.S.)
- Department of Molecular Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria de La Princesa (IIS-P), 28006 Madrid, Spain
| | - Cristina M. Ostalé
- Department of Development and Regeneration, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain;
| | - Lennart R. van der Burg
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (L.R.v.d.B.); (J.C.H.H.); (L.J.A.C.H.)
| | - Javier Galán-Martínez
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (M.J.-M.); (J.G.-M.); (R.L.-P.); (K.S.)
- Department of Molecular Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria de La Princesa (IIS-P), 28006 Madrid, Spain
| | - James C. H. Hardwick
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (L.R.v.d.B.); (J.C.H.H.); (L.J.A.C.H.)
| | - Ricardo López-Pérez
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (M.J.-M.); (J.G.-M.); (R.L.-P.); (K.S.)
| | - Lukas J. A. C. Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; (L.R.v.d.B.); (J.C.H.H.); (L.J.A.C.H.)
| | - Konstantinos Stamatakis
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (M.J.-M.); (J.G.-M.); (R.L.-P.); (K.S.)
- Department of Molecular Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria de La Princesa (IIS-P), 28006 Madrid, Spain
| | - Manuel Fresno
- Department of Cell Biology and Immunology, Centro de Biología Molecular “Severo Ochoa” (CSIC-UAM), 28049 Madrid, Spain; (M.J.-M.); (J.G.-M.); (R.L.-P.); (K.S.)
- Department of Molecular Biology, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Instituto de Investigación Sanitaria de La Princesa (IIS-P), 28006 Madrid, Spain
- Correspondence: ; Tel.: +34-911-964-565
| |
Collapse
|
30
|
He X, Li S, Yu B, Kuang G, Wu Y, Zhang M, He Y, Ou C, Cao P. Up-regulation of LINC00467 promotes the tumourigenesis in colorectal cancer. J Cancer 2019; 10:6405-6413. [PMID: 31772673 PMCID: PMC6856745 DOI: 10.7150/jca.32216] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
Recent studies have reported that long non-coding RNAs (lncRNAs) are associated with the tumourigenesis of colorectal cancer (CRC); however, several of these are yet to be identified and characterised. In this study, we report a novel lncRNA, LINC00467, which was significantly up-regulated in CRC; we investigated its function and mechanism in CRC. Our study demonstrated that LINC00467 levels in 45 pairs of CRC tissues were higher than those in the corresponding normal colon mucosal tissues. We used the Gene Expression Omnibus (GEO) and Gene Expression Profiling Interactive Analysis (GEPIA) databases for the analysis and measurement of clinical samples, and observed that the CRC patients with high LINC00467 expression levels showed poor overall survival (OS) and recurrent-free survival (RFS) rates. Furthermore, following the short interfering RNA (siRNA) knockdown of LINC00467 in the CRC cell line, the results demonstrated that LINC00467 suppresses the proliferation, invasion and metastasis of CRC cells in vitro. Moreover, its molecular mechanism of LINC00467 decreased the expression of Cyclin D1, Cyclin A1, CDK2, CDK4 and Twist1 as well as enhanced the expression of E‑cadherin. Collectively, these findings suggest that LINC00467 may be crucial in the progression and development of CRC, and may serve as a potential therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Shen Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Bingbing Yu
- Department of Pathology, Dezhou People's Hospital, Dezhou, Shandong 253056, China
| | - Gaoyan Kuang
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Yongrong Wu
- Department of Orthopedics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan 410007, China
| | - Meili Zhang
- Department of Pathology, Dezhou People's Hospital, Dezhou, Shandong 253056, China
| | - Yuxiang He
- Department of Oncology, Xiangya Hospital, Central South University,Changsha, Hunan 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Pengfei Cao
- Department of Hematology, Xiangya hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| |
Collapse
|
31
|
Wang S, Kim J, Lee C, Oh D, Han J, Kim TJ, Kim SW, Seo YS, Oh SH, Jung Y. Tumor necrosis factor-inducible gene 6 reprograms hepatic stellate cells into stem-like cells, which ameliorates liver damage in mouse. Biomaterials 2019; 219:119375. [DOI: 10.1016/j.biomaterials.2019.119375] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 07/19/2019] [Accepted: 07/20/2019] [Indexed: 12/12/2022]
|
32
|
Role of Muscarinic Acetylcholine Signaling in Gastrointestinal Cancers. Biomedicines 2019; 7:biomedicines7030058. [PMID: 31405140 PMCID: PMC6783861 DOI: 10.3390/biomedicines7030058] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 07/30/2019] [Accepted: 08/07/2019] [Indexed: 12/31/2022] Open
Abstract
In the tumor microenvironment, various stromal and immune cells accumulate and interact with cancer cells to contribute to tumor progression. Among stromal players, nerves have recently been recognized as key regulators of tumor growth. More neurotransmitters, such as catecholamines and acetylcholine (ACh), are present in tumors, as the cells that secrete neurotransmitters accumulate by the release of neurotrophic factors from cancer cells. In this short review, we focus on the role of nerve signaling in gastrointestinal (GI) cancers. Given that muscarinic acetylcholine receptor signaling seems to be a dominant regulator of GI stem cells and cancers, we review the function and mechanism of the muscarinic ACh pathway as a regulator of GI cancer progression. Accumulating evidence suggests that ACh, which is secreted from nerves and tuft cells, stimulates GI epithelial stem cells and contributes to cancer progression via muscarinic receptors.
Collapse
|
33
|
Riddiough GE, Fifis T, Muralidharan V, Perini MV, Christophi C. Searching for the link; mechanisms underlying liver regeneration and recurrence of colorectal liver metastasis post partial hepatectomy. J Gastroenterol Hepatol 2019; 34:1276-1286. [PMID: 30828863 DOI: 10.1111/jgh.14644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 12/13/2022]
Abstract
Despite excellent treatment of primary colorectal cancer, the majority of deaths occur as a result of metastasis to the liver. Recent population studies have estimated that one quarter of patients with colorectal cancer will incur synchronous or metachronous colorectal liver metastasis. However, only one quarter of these patients will be eligible for potentially curative resection. Tumor recurrence occurs in reportedly 60% of patients undergoing hepatic resection, and the majority of intrahepatic recurrence occurs within the first 6 months of surgery. The livers innate ability to restore its homeostatic size, and volume facilitates major hepatic resection that currently offers the only chance of cure to patients with extensive hepatic metastases. Experimental and clinical evidence supports the notion that following partial hepatectomy, liver regeneration (LR) paradoxically drives tumor progression and increases the risk of recurrence. It is becoming increasingly clear that the processes that drive liver organogenesis, regeneration, and tumor progression are inextricably linked. This presents a major hurdle in the management of colorectal liver metastasis and other hepatic malignancies because therapies that reduce the risk of recurrence without hampering LR are sought. The processes and pathways underlying these phenomena are multiple, complex, and cross-communicate. In this review, we will summarize the common mechanisms contributing to both LR and tumor recurrence.
Collapse
Affiliation(s)
- Georgina E Riddiough
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Theodora Fifis
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | | | - Marcos V Perini
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| | - Christopher Christophi
- Department of Surgery, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
| |
Collapse
|
34
|
Chen L, Tang RZ, Ruan J, Zhu XB, Yang Y. Up-regulation of THY1 attenuates interstitial pulmonary fibrosis and promotes lung fibroblast apoptosis during acute interstitial pneumonia by blockade of the WNT signaling pathway. Cell Cycle 2019; 18:670-681. [PMID: 30829553 DOI: 10.1080/15384101.2019.1578144] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Acute interstitial pneumonia (AIP) is an idiopathic pulmonary disease featuring rapid progressive dyspnea and respiratory failure. These symptoms typically develop within several days or weeks in patients without any pre-existing lung disease or external chest disease. Thymocyte differentiation antigen-1 (THY1) has been reported to have an effect on lung fibroblast proliferation and fibrogenic signaling. In this study, the mechanism of THY1 in AIP in influencing pulmonary fibrosis in terms of lung fibroblast proliferation and apoptosis was examined. An AIP mouse model with the pathological changes of lung tissues observed was established to identify the role of THY1 in the pathogenesis of AIP. The expression of THY1, a key regulator of the WNT pathway β-catenin and fibroblasts markers MMP-2, Occludin, α-SMA and Vimentin were determined. Lung fibroblasts of mice were isolated, in which THY1 expression was altered to identify roles THY1 plays in cell viability and apoptosis. A TOP/TOPflash assay was utilized to determine the activation of WNT pathway. Decrement of pulmonary fibrosis was achieved through THY1 up-regulation. The expression of MMP-2, Occludin, α-SMA, Vimentin and β-catenin, and the extent of β-catenin phosphorylation, significantly decreased, thereby indicating that THY1 overexpression inactivated WNT. Cell proliferation was inhibited and apoptosis was accelerated in lung fibroblasts transfected with vector carrying overexpressed THY1. Altogether, this study defines the potential role of THY1 in remission of AIP, via the upregulation of THY1, which renders the WNT pathway inactive. This inactivation of the WNT signaling pathway could alleviate pulmonary fibrosis by reducing lung fibroblast proliferation in AIP. Abbreviations: AIP: Acute interstitial pneumonia; ILDs: interstitial lung diseases; DAD: diffuse alveolar damage; SPF: specific-pathogen-free; NC: negative control; HCMV: human cytomegalovirus; HE: Hematoxylin-eosin; RIPA: radio-immunoprecipitation assay; SDS-PAGE: sodium dodecyl sulfate-polyacrylamide gel electrophoresis; BSA: bovine serum albumin; HRP: horseradish peroxidase; ECL: electrochemiluminescence; FBS: fetal bovine serum; DMSO: dimethyl sulfoxide; OD: optical density.
Collapse
Affiliation(s)
- Lin Chen
- a Department of Respiratory and Critical Care Medicine , Sichuan Academy of Medical Sciences & Sichuan Province People's Hospital , Chengdu , P.R. China
| | - Rong-Zhen Tang
- b Department of Aged Infectious Diseases , Sichuan Academy of Medical Sciences & Sichuan Province People's Hospital , Chengdu , P.R. China
| | - Jia Ruan
- c Department of Respiratory Diseases , Sichuan West China Hospital Geriatric Center-Fifth People's Hospital of Sichuan Province , Chengdu , P.R. China
| | - Xiao-Bo Zhu
- d Department of Respiratory Diseases , Ziyang City People's Hospital , Ziyang , P.R. China
| | - Yang Yang
- a Department of Respiratory and Critical Care Medicine , Sichuan Academy of Medical Sciences & Sichuan Province People's Hospital , Chengdu , P.R. China
| |
Collapse
|
35
|
Blanchard TG, Czinn SJ, Banerjee V, Sharda N, Bafford AC, Mubariz F, Morozov D, Passaniti A, Ahmed H, Banerjee A. Identification of Cross Talk between FoxM1 and RASSF1A as a Therapeutic Target of Colon Cancer. Cancers (Basel) 2019; 11:cancers11020199. [PMID: 30744076 PMCID: PMC6406751 DOI: 10.3390/cancers11020199] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 12/20/2022] Open
Abstract
Metastatic colorectal cancer (mCRC) is characterized by the expression of cellular oncogenes, the loss of tumor suppressor gene function. Therefore, identifying integrated signaling between onco-suppressor genes may facilitate the development of effective therapy for mCRC. To investigate these pathways we utilized cell lines and patient derived organoid models for analysis of gene/protein expression, gene silencing, overexpression, and immunohistochemical analyses. An inverse relationship in expression of oncogenic FoxM1 and tumor suppressor RASSF1A was observed in various stages of CRC. This inverse correlation was also observed in mCRC cells lines (T84, Colo 205) treated with Akt inhibitor. Inhibition of FoxM1 expression in mCRC cells as well as in our ex vivo model resulted in increased RASSF1A expression. Reduced levels of RASSF1A expression were found in normal cells (RWPE-1, HBEpc, MCF10A, EC) stimulated with exogenous VEGF165. Downregulation of FoxM1 also coincided with increased YAP phosphorylation, indicative of tumor suppression. Conversely, downregulation of RASSF1A coincided with FoxM1 overexpression. These studies have identified for the first time an integrated signaling pathway between FoxM1 and RASSF1A in mCRC progression, which may facilitate the development of novel therapeutic options for advanced colon cancer therapy.
Collapse
Affiliation(s)
- Thomas G Blanchard
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Steven J Czinn
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Vivekjyoti Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Neha Sharda
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Andrea C Bafford
- Department of Surgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Fahad Mubariz
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Dennis Morozov
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | - Antonino Passaniti
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- The Marlene & Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
- Department of Biochemistry & Molecular Biology and Program in Molecular Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | - Aditi Banerjee
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| |
Collapse
|
36
|
Andl T, Andl CD, Zhang Y. Two-edged sword: how activation of the "proto-oncogene" yes-associated protein 1 in lung squamous cell carcinoma can surprisingly inhibit tumor growth. J Thorac Dis 2019; 10:S3870-S3874. [PMID: 30631502 DOI: 10.21037/jtd.2018.10.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Thomas Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL, USA
| | - Claudia D Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL, USA
| | - Yuhang Zhang
- Division of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
37
|
Yu HX, Yao Y, Bu FT, Chen Y, Wu YT, Yang Y, Chen X, Zhu Y, Wang Q, Pan XY, Meng XM, Huang C, Li J. Blockade of YAP alleviates hepatic fibrosis through accelerating apoptosis and reversion of activated hepatic stellate cells. Mol Immunol 2019; 107:29-40. [PMID: 30639476 DOI: 10.1016/j.molimm.2019.01.004] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 12/20/2018] [Accepted: 01/04/2019] [Indexed: 02/07/2023]
Abstract
Yes-associated protein (YAP) is a significant downstream protein in the Hippo signaling pathway with important functions in cell proliferation, apoptosis, invasion and migration. YAP also plays a role in the progression and development of various liver diseases. In hepatic fibrosis development and reversion, the proliferation and apoptosis of activated hepatic stellate cells (HSCs) play a critical role. However, the contribution of YAP to hepatic fibrosis progression and reversion and the underlying mechanism have not been investigated. Here we investigated the expression and function of YAP in the proliferation and apoptosis of activated HSCs. We found that YAP expression was increased in liver fibrosis tissues from CCl4-induced model mice and restored to normal level after stopping CCl4 injection and 6 weeks of spontaneously recovery. YAP expression was elevated in HSC-T6 cells treated with TGF-β1 and recovered after MDI treatment. Silencing of YAP inhibited the activation and proliferation of HSC-T6 cells stimulated by TGF-β1. In addition, the apoptosis of activated HSC-T6 cells silenced for YAP was slightly enhanced. Furthermore, over-expression of YAP repressed the reversion of activated HSC-T6 cells mediated by MDI reversal. We found that HSC-T6 cells activated by TGF-β1 showed higher levels of nuclear YAP compared with MDI-treated cells, indicating that YAP was activated in HSC-T6 cells treated by TGF-β1. We also found that loss of YAP attenuated Wnt/β-catenin pathway activity in activated HSC-T6 cells. Treatment of VP, an inhibitor of the YAP-TEAD complex, reduced both activation and proliferation of HSC-T6 cells and increased apoptosis. Together these results indicated that reduced expression of YAP contributes to acquisition of the quiescent phenotype in HSCs. Our results suggest that YAP may be a useful target in HSCs activation and reversion.
Collapse
Affiliation(s)
- Hai-Xia Yu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yao Yao
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Fang-Tian Bu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yu Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yu-Ting Wu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yang Yang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xin Chen
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Yan Zhu
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Qin Wang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xue-Yin Pan
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Xiao-Ming Meng
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China
| | - Cheng Huang
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| | - Jun Li
- The Key Laboratory of Major Autoimmune Diseases, Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China; The Key Laboratory of Anti-inflammatory of Immune Medicines, Ministry of Education, Hefei, China; Institute for Liver Diseases of Anhui Medical University, Hefei, China.
| |
Collapse
|
38
|
Zhang S, Wei Q, Yang Y, Qin H, Li X, Cai S, Ma Y. Loss of Yes-associated Protein Represents an Aggressive Subtype of Colorectal Cancer. J Cancer 2019; 10:689-696. [PMID: 30719167 PMCID: PMC6360423 DOI: 10.7150/jca.28333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 10/27/2018] [Indexed: 01/15/2023] Open
Abstract
Background: Yes-associated protein (YAP) is a downstream effecter of Hippo signaling pathway, and has been linked to the initiation and development of colorectal cancer (CRC). However, the clinical significance of YAP in CRC remains controversial. This study was designed to investigate the clinical significance of YAP in CRC. Methods: We selected 206 eligible patients diagnosed with CRC from 2003 to 2007. Tissue microarray (TMA) blocks were made from 206 formalin-fixed paraffin-embedded CRC tissues and 158 corresponding normal colonic tissues. Using the TMA blocks, we performed immunohistochemical staining of YAP and assessed its expression status in different subcellular locations. The patients were divided into four groups according to the expression status of YAP in the cytoplasm and nucleus. Statistical analysis was performed to explore the correlation between YAP expression and clinicopathological features and overall survival (OS) in CRC patients. Results: Our results showed that both cytoplasmic YAP and nuclear YAP were overexpressed in CRC tissues compared to normal colonic tissues. Complete loss of YAP expression in CRC was significantly correlated with larger tumor size (p=0.023), proximal tumor location (p=0.038), higher tumor grade (p=0.022) and worse OS (p<0.001). Univariate and multivariate Cox regression analyses revealed that complete loss of YAP expression was an independent indicator of poor prognosis in CRC (p<0.001). Conclusions: Loss of YAP expression correlates with poor prognosis and may represent a subgroup with more aggressive biological features in CRC.
Collapse
Affiliation(s)
- Sheng Zhang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qing Wei
- Department of Pathology, Shanghai Tenth People's Hospital, Tongji University, Shanghai, 200072, China
| | - Yongzhi Yang
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Huanlong Qin
- Department of GI Surgery, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Xinxiang Li
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Sanjun Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yanlei Ma
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| |
Collapse
|
39
|
He X, Su W, Zhou Y, Ge X, Zhou J, Ou C. CircPVT1: a bridge linking Hippo pathway and human cancers. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:S91. [PMID: 30740412 DOI: 10.21037/atm.2018.11.06] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Xiaoyun He
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weiping Su
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiaolu Ge
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jianhua Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
40
|
Affiliation(s)
- Eekhoon Jho
- Department of Life Science, University of Seoul, Seoul, Republic of Korea
| |
Collapse
|
41
|
Gan Y, Li Y, Li T, Shu G, Yin G. CCNA2 acts as a novel biomarker in regulating the growth and apoptosis of colorectal cancer. Cancer Manag Res 2018; 10:5113-5124. [PMID: 30464611 PMCID: PMC6217169 DOI: 10.2147/cmar.s176833] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Colorectal cancer (CRC) is considered to be the most prevalent malignant tumors that contribute to high cancer-related mortality. However, the signaling pathways involved in CRC and CRC-driven genes are largely unknown. We seek to discover a novel biomarker in CRC. MATERIALS AND METHODS All clinical CRC samples (n=33) were from Xiangya Hospital. We first selected CCNA2 by integrated bioinformatics analysis of four GSE databases. Next, the expression of CCNA2 in tissues and cell lines was verified by quantitative real-time PCR. The effects of CCNA2 on cell growth, proliferation, cell cycle, and apoptosis were examined by in vitro assays. RESULTS We identified 498 shared DEGs (294 upregulated and 204 downregulated), and the top ten hub genes were selected by integrated analysis. These hub genes were significantly overexpressed in CRC samples and were positively correlated. Our data revealed that the expression of CCNA2 in CRC tissues is higher than that in normal tissues. The CCNA2 knockdown could significantly suppress CRC cell growth by impairing cell cycle progression and inducing cell apoptosis. CONCLUSION CCNA2, as a novel oncogenic gene, plays a role in regulating cancer cell growth and apoptosis. It could be used as a new biomarker for diagnosis and therapy in CRC.
Collapse
Affiliation(s)
- Yaqi Gan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China,
| | - Yimin Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China,
| | - Tong Li
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China,
| | - Guang Shu
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China,
| | - Gang Yin
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan Province, China,
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, Hunan Province, China,
| |
Collapse
|
42
|
Zhou Z, Zhang HS, Zhang ZG, Sun HL, Liu HY, Gou XM, Yu XY, Huang YH. Loss of HACE1 promotes colorectal cancer cell migration via upregulation of YAP1. J Cell Physiol 2018; 234:9663-9672. [PMID: 30362561 DOI: 10.1002/jcp.27653] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 10/02/2018] [Indexed: 12/12/2022]
Abstract
Colorectal cancer (CRC) is the third-leading cause of cancer mortality worldwide. HACE1 function as a tumor-suppressor gene and is downregulated in several kinds of cancers. However, the distribution and clinical significance of HACE1 in CRC is still not clarified. In this study, we found that the HACE1 expression is greatly downregulated in CRC tissues and cell lines. Moreover, the HACE1 expression was significantly associated with inhibition of CRC cell proliferation, metastasis, and invasion. HACE1 inhibited epithelial-mesenchymal transition in CRC cells. Furthermore, we found that HACE1 altered the protein expression of the Hippo pathway by downregulation of YAP1. HACE1 suppresses the invasive ability of CRC cells by negatively regulating the YAP1 pathway. Our data indicates that HACE1 directly targets YAP1 and induces downregulation of YAP1, thereby increasing the activity of the Hippo pathway. In summary, these findings demonstrated that HACE1-YAP1 axis had an important part in the CRC development and progression.
Collapse
Affiliation(s)
- Zhen Zhou
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Hong-Sheng Zhang
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Zhong-Guo Zhang
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Hong-Liang Sun
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Hui-Yun Liu
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Xiao-Meng Gou
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Xiao-Ying Yu
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| | - Ying-Hui Huang
- Department of Biotechnology, College of Life Science & Bioengineering, Beijing University of Technology, Chaoyang, Beijing, China
| |
Collapse
|
43
|
Real SAS, Parveen F, Rehman AU, Khan MA, Deo SVS, Shukla NK, Husain SA. Aberrant Promoter Methylation of YAP Gene and its Subsequent Downregulation in Indian Breast Cancer Patients. BMC Cancer 2018; 18:711. [PMID: 29970036 PMCID: PMC6031145 DOI: 10.1186/s12885-018-4627-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 06/21/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND YAP, a potent oncogene and major downstream effector of the mammalian Hippo tumor suppressor pathway can act as either oncogene or tumor suppressor gene based on the type of tissue involved. Despite various studies, the role and mechanism through which YAP mediates its tumor suppressor or oncogenic effects are not yet fully understood. Therefore in the present study we aimed to investigate YAP at DNA, mRNA and protein level and also attempted to correlate our molecular findings with various clinicopathological variables of the patients. METHODS The study comprised of a total 137 genetically unrelated women with sporadic breast cancer cases and normal adjacent tissues not infiltrated with tumor. Mutation of YAP gene was analyzed by automated DNA sequencing. YAP promoter methylation was studied using MS-PCR. Expression at mRNA and protein level was studied using qPCR and IHC respectively. RESULTS In our study YAP mRNA expression was found to be 8.65 ± 6.17 fold downregulated in 67.15% cases. The expression of YAP when analyzed at the protein level by IHC was found to be absent in 78.83% cases. Results from MS-PCR analysis showed that YAP promoter methylation plays an important role in declining the expression of YAP protein. The absence of YAP protein coincided with 86.60% methylated cases thereby showing a very strong correlation (p = 0.001). We also investigated YAP mutation at the major check point sites in the Hippo pathway and observed no mutation. A significant association was observed on correlating mRNA expression with clinical stages (p = 0.038) and protein expression with ER status (p = 0.018) among Indian breast cancer patients. CONCLUSION The expression of YAP was found to be downregulated in response to aberrant promoter methylation. The downregulation of YAP are consistent with previous studies suggesting it to have a tumor suppressive role in breast cancer. We did not observe any mutation at the major check point sites in the Hippo pathway.
Collapse
Affiliation(s)
| | - Farah Parveen
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Asad Ur Rehman
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India
| | | | | | - Nootan Kumar Shukla
- Department of Surgical Oncology, All India Institute of Medical Science, New Delhi, 110608, India
| | - Syed Akhtar Husain
- Department of Biosciences, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
44
|
Dehghanian F, Hojati Z, Hosseinkhan N, Mousavian Z, Masoudi-Nejad A. Reconstruction of the genome-scale co-expression network for the Hippo signaling pathway in colorectal cancer. Comput Biol Med 2018; 99:76-84. [PMID: 29890510 DOI: 10.1016/j.compbiomed.2018.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 05/24/2018] [Accepted: 05/24/2018] [Indexed: 01/22/2023]
Abstract
The Hippo signaling pathway (HSP) has been identified as an essential and complex signaling pathway for tumor suppression that coordinates proliferation, differentiation, cell death, cell growth and stemness. In the present study, we conducted a genome-scale co-expression analysis to reconstruct the HSP in colorectal cancer (CRC). Five key modules were detected through network clustering, and a detailed discussion of two modules containing respectively 18 and 13 over and down-regulated members of HSP was provided. Our results suggest new potential regulatory factors in the HSP. The detected modules also suggest novel genes contributing to CRC. Moreover, differential expression analysis confirmed the differential expression pattern of HSP members and new suggested regulatory factors between tumor and normal samples. These findings can further reveal the importance of HSP in CRC.
Collapse
Affiliation(s)
- Fariba Dehghanian
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, P.O. Box 81746-73441, Isfahan, Iran
| | - Zohreh Hojati
- Division of Genetics, Department of Biology, Faculty of Sciences, University of Isfahan, P.O. Box 81746-73441, Isfahan, Iran.
| | - Nazanin Hosseinkhan
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Zaynab Mousavian
- Department of Computer Science, School of Mathematics, Statistics, and Computer Science, University of Tehran, Tehran, Iran; Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
45
|
He X, Yan Q, Kuang G, Wang Y, Cao P, Ou C. Metastasis-associated lung adenocarcinoma transcript 1 regulates tumor progression: old wine in a new bottle. J Thorac Dis 2018; 10:S1088-S1091. [PMID: 29850189 DOI: 10.21037/jtd.2018.04.13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Xiaoyun He
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China.,Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qijia Yan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Gaoyan Kuang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha 410007, China
| | - Yixuan Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chunlin Ou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
46
|
Wang C, Zhu X, Pu C, Song X. Upregulated plasmacytoma variant translocation 1 promotes cell proliferation, invasion and metastasis in colorectal cancer. Mol Med Rep 2018; 17:6598-6604. [PMID: 29512788 PMCID: PMC5928643 DOI: 10.3892/mmr.2018.8669] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 02/27/2018] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence indicates that the long non-coding RNA (lncRNA) plasmacytoma variant translocation 1 (PVT1) is associated with tumourigenesis in various types of cancer. However, its specific effects on the proliferation, invasion and metastasis of colorectal cancer (CRC) are still poorly understood. The present study aimed to investigate PVT1 expression in CRC and explore its role in CRC pathogenesis. The reverse transcriptase‑quantitative polymerase chain reaction (RT‑qPCR) technique was used to assess PVT1 expression in CRC cell lines. Gene Expression Omnibus (GEO) database analysis and measurement of clinical samples was used to analyse the correlation between PVT1 expression, CRC metastasis and overall survival (OS). In addition, knockdown of PVT1 expression was performed using short interfering RNA (siRNA) and RT‑qPCR, western blotting, CCK‑8 assays, tumour cell clone‑formation and Matrigel invasion assays were used to observe its biological functions in HCT116 cells. The present study demonstrated that the expression of PVT1 in CRC cell lines was higher than that in normal colon mucosal cell lines. Using GEO database analysis and the measurement of clinical samples, it was revealed that CRC patients with high PVT1 expression demonstrated poor OS. Multivariate analysis indicated that high PVT1 expression is an independent risk factor for patients with CRC. In addition, PVT1 knockdown suppressed the proliferation, invasion and metastasis of CRC cells in vitro, which were associated with decreasing vimentin, cyclin D1 and cyclin‑dependent kinase 4 expression and enhanced E‑cadherin expression. The results of the present study suggest that PVT1 may serve a critical role in CRC progression and metastasis and may serve as a potential prognostic biomarker for CRC.
Collapse
Affiliation(s)
- Chaoyu Wang
- Department of General Surgery, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Xin Zhu
- Department of General Surgery, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| | - Chibin Pu
- Department of Gastroenterology, Zhongda Hospital, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xuan Song
- Department of General Surgery, People's Hospital of Guizhou Province, Guiyang, Guizhou 550002, P.R. China
| |
Collapse
|
47
|
Senatorov IS, Moniri NH. The role of free-fatty acid receptor-4 (FFA4) in human cancers and cancer cell lines. Biochem Pharmacol 2018; 150:170-180. [PMID: 29452095 DOI: 10.1016/j.bcp.2018.02.011] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 02/12/2018] [Indexed: 12/12/2022]
Abstract
A dietary influence on cancer progression has been evident for many decades, and dietary fatty acids, particularly long chain mono- and polyunsaturated fatty acids, have been shown to play significant roles in influencing growth of a variety of human cancers. The discovery of the family of cell-surface free-fatty acid receptors, which include the long-chain fatty acid receptors FFA1 and FFA4, suggest that many of the effects of dietary fats could be receptor-mediated. FFA4 is ubiquitously expressed and has recently been shown to modulate a variety of important anti-inflammatory and metabolic processes. Since FFA4 is currently an attractive drug target for treatment of metabolic disorders such as diabetes and obesity, understanding its role in cancer progression is critical towards the drug discovery process. In this research update, the current body of knowledge on the role of this receptor in regulating cancer cell proliferation, migration, and invasion, as well as in vivo tumorigenesis is reviewed.
Collapse
Affiliation(s)
- Ilya S Senatorov
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, United States
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA 30341, United States.
| |
Collapse
|