51
|
Carter P, Schnell U, Chaney C, Tong B, Pan X, Ye J, Mernaugh G, Cotton JL, Margulis V, Mao J, Zent R, Evers BM, Kapur P, Carroll TJ. Deletion of Lats1/2 in adult kidney epithelia leads to renal cell carcinoma. J Clin Invest 2021; 131:e144108. [PMID: 34060480 DOI: 10.1172/jci144108] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 04/22/2021] [Indexed: 11/17/2022] Open
Affiliation(s)
- Phoebe Carter
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ulrike Schnell
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Christopher Chaney
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Betty Tong
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Xinchao Pan
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jianhua Ye
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Glenda Mernaugh
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Jennifer L Cotton
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | | | - Junhao Mao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Roy Zent
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Bret M Evers
- Department of Pathology, and.,Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, Texas, USA.Renal cell carcinoma (RCC) is the most common kidney cancer in humans. Misregulation of the Hippo/Warts pathway is frequently reported in RCC, suggesting a role in disease formation/progression. Paradoxically, misregulation of this pathway is also observed in non-tumorigenic kidney diseases, raising questions as to its specific role in RCC. Here, we show that ablation of the Warts kinases Lats1 and Lats2 in mature renal epithelia was sufficient to cause metastatic RCC in mice. Distinct tumors with sarcomatoid histology were present in mutant kidneys 3 months after genetic ablation. Tumor formation required the downstream effectors Yap and Taz, and treatment with verteporfin, a drug that inhibits Yap activity, could slow progression of the disease. Examination of human tissues showed that among histological subtypes of RCC, nuclear YAP was most commonly observed in sarcomatoid RCC. However, analysis of transcriptomic data from human RCC revealed a unique subset with a molecular signature that closely resembled the transcriptome of Lats, mutants. Together, these findings show that misregulation of the Warts pathway is sufficient to drive renal tumor formation in mice and suggest that human tumors with active YAP may represent a unique subset of RCCs that can be therapeutically targeted
| | - Payal Kapur
- Department of Urology.,Department of Pathology, and
| | - Thomas J Carroll
- Department of Molecular Biology and Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
52
|
Zilli F, Marques Ramos P, Auf der Maur P, Jehanno C, Sethi A, Coissieux M, Eichlisberger T, Sauteur L, Rouchon A, Bonapace L, Pinto Couto J, Rad R, Jensen MR, Banfi A, Stadler MB, Bentires‐Alj M. The NFIB-ERO1A axis promotes breast cancer metastatic colonization of disseminated tumour cells. EMBO Mol Med 2021; 13:e13162. [PMID: 33751828 PMCID: PMC8033524 DOI: 10.15252/emmm.202013162] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 01/29/2021] [Accepted: 02/01/2021] [Indexed: 12/11/2022] Open
Abstract
Metastasis is the main cause of deaths related to solid cancers. Active transcriptional programmes are known to regulate the metastatic cascade but the molecular determinants of metastatic colonization remain elusive. Using an inducible piggyBac (PB) transposon mutagenesis screen, we have shown that overexpression of the transcription factor nuclear factor IB (NFIB) alone is sufficient to enhance primary mammary tumour growth and lung metastatic colonization. Mechanistically and functionally, NFIB directly increases expression of the oxidoreductase ERO1A, which enhances HIF1α-VEGFA-mediated angiogenesis and colonization, the last and fatal step of the metastatic cascade. NFIB is thus clinically relevant: it is preferentially expressed in the poor-prognostic group of basal-like breast cancers, and high expression of the NFIB/ERO1A/VEGFA pathway correlates with reduced breast cancer patient survival.
Collapse
Affiliation(s)
- Federica Zilli
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | - Pedro Marques Ramos
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Priska Auf der Maur
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Charly Jehanno
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Atul Sethi
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Marie‐May Coissieux
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| | | | - Loïc Sauteur
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Adelin Rouchon
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Laura Bonapace
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Joana Pinto Couto
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Novartis Institutes for Biomedical ResearchBaselSwitzerland
| | - Roland Rad
- Department of Medicine IITUM School of MedicineInstitute of Molecular Oncology and Functional GenomicsCenter for Translational Cancer Research (TranslaTUM)Technische Universität MünchenMünchenGermany
- German Cancer Consortium (DKTK)German Cancer Research Center (DKFZ)HeidelbergGermany
| | | | - Andrea Banfi
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
| | - Michael B Stadler
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
- Swiss Institute of BioinformaticsBaselSwitzerland
| | - Mohamed Bentires‐Alj
- Department of BiomedicineDepartment of SurgeryUniversity Hospital BaselUniversity of BaselBaselSwitzerland
- Friedrich Miescher Institute for Biomedical ResearchBaselSwitzerland
| |
Collapse
|
53
|
|
54
|
Ma S, Wu Z, Yang F, Zhang J, Johnson RL, Rosenfeld MG, Guan KL. Hippo signalling maintains ER expression and ER + breast cancer growth. Nature 2021; 591:E1-E10. [PMID: 33658690 DOI: 10.1038/s41586-020-03131-5] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022]
Affiliation(s)
- Shenghong Ma
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Zhengming Wu
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Feng Yang
- Howard Hughes Medical Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jianmin Zhang
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, USA
| | - Randy L Johnson
- Department of Cancer Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael G Rosenfeld
- Howard Hughes Medical Institute, Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Kun-Liang Guan
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA. .,Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
55
|
Quinn HM, Vogel R, Popp O, Mertins P, Lan L, Messerschmidt C, Landshammer A, Lisek K, Château-Joubert S, Marangoni E, Koren E, Fuchs Y, Birchmeier W. YAP and β-Catenin Cooperate to Drive Oncogenesis in Basal Breast Cancer. Cancer Res 2021; 81:2116-2127. [PMID: 33574090 DOI: 10.1158/0008-5472.can-20-2801] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 12/15/2020] [Accepted: 02/08/2021] [Indexed: 11/16/2022]
Abstract
Targeting cancer stem cells (CSC) can serve as an effective approach toward limiting resistance to therapies. While basal-like (triple-negative) breast cancers encompass cells with CSC features, rational therapies remain poorly established. We show here that the receptor tyrosine kinase Met promotes YAP activity in basal-like breast cancer and find enhanced YAP activity within the CSC population. Interfering with YAP activity delayed basal-like cancer formation, prevented luminal to basal transdifferentiation, and reduced CSC. YAP knockout mammary glands revealed a decrease in β-catenin target genes, suggesting that YAP is required for nuclear β-catenin activity. Mechanistically, nuclear YAP interacted with β-catenin and TEAD4 at gene regulatory elements. Proteomic patient data revealed an upregulation of the YAP signature in basal-like breast cancers. Our findings demonstrate that in basal-like breast cancers, β-catenin activity is dependent on YAP signaling and controls the CSC program. These findings suggest that targeting the YAP/TEAD4/β-catenin complex offers a potential therapeutic strategy for eradicating CSCs in basal-like breast cancers. SIGNIFICANCE: These findings show that YAP cooperates with β-catenin in basal-like breast cancer to regulate CSCs and that targeting this interaction may be a novel CSC therapy for patients with basal-like breast cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/8/2116/F1.large.jpg.
Collapse
Affiliation(s)
- Hazel M Quinn
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Regina Vogel
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Philipp Mertins
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | - Linxiang Lan
- Adult Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom.,The Institute of Cancer Research, London, United Kingdom
| | - Clemens Messerschmidt
- Computer Science Department, Humboldt-Universität, Berlin, Germany.,Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Alexandro Landshammer
- Dept. of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Kamil Lisek
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany
| | | | | | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Technion Israel Institute of Technology, Haifa, Israel
| | - Walter Birchmeier
- Cancer Research Program, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Association, Berlin, Germany.
| |
Collapse
|
56
|
Su D, Li Y, Guan L, Li Q, Shi C, Ma X, Song Y. Elevated MST1 leads to apoptosis via depletion of YAP1 in cardiomyocytes exposed to high glucose. Mol Med 2021; 27:13. [PMID: 33568044 PMCID: PMC7874454 DOI: 10.1186/s10020-021-00267-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 01/01/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gestational diabetes mellitus is a risk factor for congenital heart defects. The article aimed to investigate the expression and roles of MST1, YAP1, Last1/2 and Survivin in modulating HG-induced cardiomyocyte apoptosis and maternal diabetes-induced heart abnormality. METHODS Diabetes mellitus was induced in rats using streptozotocin. The protein expression and phosphorylation analysis in fetal heart tissue was assessed by western blot and immunohistochemical staining. Hoechst 33342 staining assay was performed to explore H9C2 apoptosis. The gene and protein expression in H9C2 cells was assessed by quantitative PCR and western blot. Knockdown of gene expression was assessed by RNA interference. RESULTS Our results revealed that increased MST1 protein levels in the heart tissues of the offspring of diabetic rats in vivo and in H9C2 cardiomyocytes under HG treatment in vitro, respectively. Knockdown and overexpression experiments showed that MST1 played a key role in mediating HG-induced apoptosis in cardiomyocytes. Downregulation of YAP1 was associated with HG-induced, MST1-mediated cardiomyocyte apoptosis. Further study showed that MST1 downregulated the protein level of YAP1 through mediation of YAP1 phosphorylation on Ser127 and Ser397; this process also required LATS1/2 participation. MST1 overexpression increased the phosphorylation levels of LATS1/2, which were also shown to be increased in the heart tissues of diabetic offspring. We also found that YAP1 mediated the expression of Survivin during HG-induced apoptosis, and the Survivin-inhibitor YM155 partially inhibited the role of YAP1 in suppressing apoptosis induced by HG in cardiomyocytes. CONCLUSION These findings reveal a regulatory mechanism of MST1/YAP1/Survivin signaling in modulating cardiomyocyte apoptosis in vitro and maternal diabetes-induced congenital heart defects in vivo.
Collapse
Affiliation(s)
- Dongmei Su
- Department of Genetics, Center for Genetics, National Research Institute for Family Planning, Health Department, 12, Dahuisi Road, Haidian, Beijing, 100081, China.,Graduate School, Peking Union Medical College, Beijing, China
| | - Yanhua Li
- Department of Teaching and Research of Obstetrics and Gynecology, Shandong Medical College, Linyi, Shandong, China.,Department of Obstetrics, Linyi People's Hospital, 27, Jiefang Road, Linyi, 276003, Shandong, China
| | - Lina Guan
- Department of Genetics, Center for Genetics, National Research Institute for Family Planning, Health Department, 12, Dahuisi Road, Haidian, Beijing, 100081, China
| | - Qian Li
- Department of Genetics, Center for Genetics, National Research Institute for Family Planning, Health Department, 12, Dahuisi Road, Haidian, Beijing, 100081, China
| | - Cuige Shi
- Department of Genetics, Center for Genetics, National Research Institute for Family Planning, Health Department, 12, Dahuisi Road, Haidian, Beijing, 100081, China
| | - Xu Ma
- Department of Genetics, Center for Genetics, National Research Institute for Family Planning, Health Department, 12, Dahuisi Road, Haidian, Beijing, 100081, China. .,Graduate School, Peking Union Medical College, Beijing, China.
| | - Yonghui Song
- Department of Obstetrics, Linyi People's Hospital, 27, Jiefang Road, Linyi, 276003, Shandong, China
| |
Collapse
|
57
|
Zhou X, Li W, Liu Y, Amon A. Cross-compartment signal propagation in the mitotic exit network. eLife 2021; 10:e63645. [PMID: 33481703 PMCID: PMC7822594 DOI: 10.7554/elife.63645] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 01/06/2021] [Indexed: 12/26/2022] Open
Abstract
In budding yeast, the mitotic exit network (MEN), a GTPase signaling cascade, integrates spatial and temporal cues to promote exit from mitosis. This signal integration requires transmission of a signal generated on the cytoplasmic face of spindle pole bodies (SPBs; yeast equivalent of centrosomes) to the nucleolus, where the MEN effector protein Cdc14 resides. Here, we show that the MEN activating signal at SPBs is relayed to Cdc14 in the nucleolus through the dynamic localization of its terminal kinase complex Dbf2-Mob1. Cdc15, the protein kinase that activates Dbf2-Mob1 at SPBs, also regulates its nuclear access. Once in the nucleus, priming phosphorylation of Cfi1/Net1, the nucleolar anchor of Cdc14, by the Polo-like kinase Cdc5 targets Dbf2-Mob1 to the nucleolus. Nucleolar Dbf2-Mob1 then phosphorylates Cfi1/Net1 and Cdc14, activating Cdc14. The kinase-primed transmission of the MEN signal from the cytoplasm to the nucleolus exemplifies how signaling cascades can bridge distant inputs and responses.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of TechnologyCambridgeUnited States
| | - Wenxue Li
- Yale Cancer Biology Institute, Department of Pharmacology, Yale UniversityWest HavenUnited States
| | - Yansheng Liu
- Yale Cancer Biology Institute, Department of Pharmacology, Yale UniversityWest HavenUnited States
| | - Angelika Amon
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of TechnologyCambridgeUnited States
| |
Collapse
|
58
|
Tan S, Zhao Z, Qiao Y, Zhang B, Zhang T, Zhang M, Qi J, Wang X, Meng M, Zhou Q. Activation of the tumor suppressive Hippo pathway by triptonide as a new strategy to potently inhibit aggressive melanoma cell metastasis. Biochem Pharmacol 2021; 185:114423. [PMID: 33476574 DOI: 10.1016/j.bcp.2021.114423] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 01/02/2021] [Accepted: 01/13/2021] [Indexed: 02/08/2023]
Abstract
Metastatic melanoma has a very high mortality rate despite the availability of chemotherapy, radiotherapy, and immunotherapy; therefore, more effective therapeutics are needed. The Hippo pathway plays an inhibitory role in melanoma progression, but the tumor suppressors Salvador homolog-1 (SAV1) and large tumor suppressor 1 (LATS1) in this pathway are down-regulated in melanoma. As a result, the downstream oncogenic Yes-associated protein (YAP) is active, resulting in uncontrolled melanoma growth and metastasis. Therapeutics for remedying SAV1 and LATS1 deficiency in melanoma have not yet been reported in the literature. Here, we show that the small molecule triptonide (MW 358 Da) robustly suppressed melanoma cell tumorigenicity, migration, and invasion. Furthermore, triptonide markedly reduced tumor growth and melanoma lung metastasis in tumor-bearing mice with low toxicity. Molecular mechanistic studies revealed that triptonide promoted SAV1 and LATS1 expression, strongly activated the tumor-suppressive Hippo pathway, degraded oncogenic YAP via the lysosomal pathway, and reduced levels of tumorigenic microphthalmia-associated transcription factor (MITF) in melanoma cells. Triptonide also strongly inhibited activation of AKT, a SAV1-binding signaling protein. Collectively, our results conceptually demonstrate that induction of SAV1 and LATS1 expression and activation of the tumor-suppressive Hippo pathway by triptonide potently inhibits aggressive melanoma cell growth and metastasis. These findings suggest a new strategy for developing therapeutics to treat metastatic melanoma and highlight a novel drug candidate against aggressive melanoma.
Collapse
Affiliation(s)
- Shijie Tan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Zhe Zhao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Jiangsu 215123, PR China
| | - Yingnan Qiao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Bin Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; Center of Systems Medicine, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, PR China; Suzhou Institute of Systems Medicine, Suzhou 215123, PR China
| | - Tong Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Mengli Zhang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China
| | - Jindan Qi
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Xiaohua Wang
- School of Nursing, Soochow University, Suzhou, Jiangsu 215006, PR China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China.
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, Key Laboratory of Thrombosis and Hemostasis, Ministry of Health, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, PR China; State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Soochow University, Suzhou, Jiangsu 215123, PR China; National Clinical Research Center for Hematology Diseases, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, PR China.
| |
Collapse
|
59
|
Muranen TA, Khan S, Fagerholm R, Aittomäki K, Cunningham JM, Dennis J, Leslie G, McGuffog L, Parsons MT, Simard J, Slager S, Soucy P, Easton DF, Tischkowitz M, Spurdle AB, Schmutzler RK, Wappenschmidt B, Hahnen E, Hooning MJ, Singer CF, Wagner G, Thomassen M, Pedersen IS, Domchek SM, Nathanson KL, Lazaro C, Rossing CM, Andrulis IL, Teixeira MR, James P, Garber J, Weitzel JN, Jakubowska A, Yannoukakos D, John EM, Southey MC, Schmidt MK, Antoniou AC, Chenevix-Trench G, Blomqvist C, Nevanlinna H. Association of germline variation with the survival of women with BRCA1/2 pathogenic variants and breast cancer. NPJ Breast Cancer 2020; 6:44. [PMID: 32964118 PMCID: PMC7483417 DOI: 10.1038/s41523-020-00185-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 08/11/2020] [Indexed: 02/02/2023] Open
Abstract
Germline genetic variation has been suggested to influence the survival of breast cancer patients independently of tumor pathology. We have studied survival associations of genetic variants in two etiologically unique groups of breast cancer patients, the carriers of germline pathogenic variants in BRCA1 or BRCA2 genes. We found that rs57025206 was significantly associated with the overall survival, predicting higher mortality of BRCA1 carrier patients with estrogen receptor-negative breast cancer, with a hazard ratio 4.37 (95% confidence interval 3.03-6.30, P = 3.1 × 10-9). Multivariable analysis adjusted for tumor characteristics suggested that rs57025206 was an independent survival marker. In addition, our exploratory analyses suggest that the associations between genetic variants and breast cancer patient survival may depend on tumor biological subgroup and clinical patient characteristics.
Collapse
Affiliation(s)
- Taru A. Muranen
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Sofia Khan
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
- University of Turku and Åbo Akademi University, Turku Bioscience Centre, Turku, Finland
| | - Rainer Fagerholm
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| | - Kristiina Aittomäki
- University of Helsinki, Department of Clinical Genetics, Helsinki University Hospital, Helsinki, Finland
| | - Julie M. Cunningham
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN USA
| | - Joe Dennis
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Goska Leslie
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Lesley McGuffog
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Michael T. Parsons
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
| | - Jacques Simard
- CHU de Quebec Research Center, Genomics Center, Québec City, QC Canada
| | - Susan Slager
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN USA
| | - Penny Soucy
- CHU de Quebec Research Center, Genomics Center, Québec City, QC Canada
| | - Douglas F. Easton
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
| | - Marc Tischkowitz
- McGill University, Program in Cancer Genetics, Departments of Human Genetics and Oncology, Montréal, QC Canada
- University of Cambridge, Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Amanda B. Spurdle
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
| | - kConFab Investigators
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
- University of Turku and Åbo Akademi University, Turku Bioscience Centre, Turku, Finland
- University of Helsinki, Department of Clinical Genetics, Helsinki University Hospital, Helsinki, Finland
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
- CHU de Quebec Research Center, Genomics Center, Québec City, QC Canada
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
- McGill University, Program in Cancer Genetics, Departments of Human Genetics and Oncology, Montréal, QC Canada
- University of Cambridge, Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
- Medical University of Vienna, Dept of OB/GYN and Comprehensive Cancer Center, Vienna, Austria
- Odense University Hospital, Department of Clinical Genetics, Odence C, Denmark
- Aalborg University Hospital, Molecular Diagnostics, Aalborg, Denmark
- Aalborg University, Dept of Clinical Medicine, Aalborg, Denmark
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Abramson Cancer Center, Philadelphia, PA USA
- ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Molecular Diagnostic Unit, Hereditary Cancer Program, Barcelona, Spain
- Rigshospitalet, Copenhagen University Hospital, Center for Genomic Medicine, Copenhagen, Denmark
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON Canada
- Portuguese Oncology Institute, Department of Genetics, Porto, Portugal
- University of Porto, Biomedical Sciences Institute (ICBAS), Porto, Portugal
- Peter MacCallum Cancer Center, Parkville Familial Cancer Centre, Melbourne, VIC Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC Australia
- Dana-Farber Cancer Institute, Cancer Risk and Prevention Clinic, Boston, MA USA
- City of Hope, Clinical Cancer Genomics, Duarte, CA USA
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
- Pomeranian Medical University, Independent Laboratory of Molecular Biology and Genetic Diagnostics, Szczecin, Poland
- National Centre for Scientific Research ‘Demokritos’, Molecular Diagnostics Laboratory, INRASTES, Athens, Greece
- Stanford Cancer Institute, Stanford University School of Medicine, Department of Medicine, Division of Oncology, Stanford, CA USA
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, VIC Australia
- The University of Melbourne, Department of Clinical Pathology, Melbourne, VIC Australia
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
- University of Helsinki, Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Örebro University Hospital, Department of Oncology, Örebro, Sweden
| | - Rita K. Schmutzler
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Barbara Wappenschmidt
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Eric Hahnen
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
| | - Maartje J. Hooning
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
| | - HEBON Investigators
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
- University of Turku and Åbo Akademi University, Turku Bioscience Centre, Turku, Finland
- University of Helsinki, Department of Clinical Genetics, Helsinki University Hospital, Helsinki, Finland
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
- CHU de Quebec Research Center, Genomics Center, Québec City, QC Canada
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
- McGill University, Program in Cancer Genetics, Departments of Human Genetics and Oncology, Montréal, QC Canada
- University of Cambridge, Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
- Medical University of Vienna, Dept of OB/GYN and Comprehensive Cancer Center, Vienna, Austria
- Odense University Hospital, Department of Clinical Genetics, Odence C, Denmark
- Aalborg University Hospital, Molecular Diagnostics, Aalborg, Denmark
- Aalborg University, Dept of Clinical Medicine, Aalborg, Denmark
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Abramson Cancer Center, Philadelphia, PA USA
- ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Molecular Diagnostic Unit, Hereditary Cancer Program, Barcelona, Spain
- Rigshospitalet, Copenhagen University Hospital, Center for Genomic Medicine, Copenhagen, Denmark
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON Canada
- Portuguese Oncology Institute, Department of Genetics, Porto, Portugal
- University of Porto, Biomedical Sciences Institute (ICBAS), Porto, Portugal
- Peter MacCallum Cancer Center, Parkville Familial Cancer Centre, Melbourne, VIC Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC Australia
- Dana-Farber Cancer Institute, Cancer Risk and Prevention Clinic, Boston, MA USA
- City of Hope, Clinical Cancer Genomics, Duarte, CA USA
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
- Pomeranian Medical University, Independent Laboratory of Molecular Biology and Genetic Diagnostics, Szczecin, Poland
- National Centre for Scientific Research ‘Demokritos’, Molecular Diagnostics Laboratory, INRASTES, Athens, Greece
- Stanford Cancer Institute, Stanford University School of Medicine, Department of Medicine, Division of Oncology, Stanford, CA USA
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, VIC Australia
- The University of Melbourne, Department of Clinical Pathology, Melbourne, VIC Australia
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
- University of Helsinki, Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Örebro University Hospital, Department of Oncology, Örebro, Sweden
| | - Christian F. Singer
- Medical University of Vienna, Dept of OB/GYN and Comprehensive Cancer Center, Vienna, Austria
| | - Gabriel Wagner
- Medical University of Vienna, Dept of OB/GYN and Comprehensive Cancer Center, Vienna, Austria
| | - Mads Thomassen
- Odense University Hospital, Department of Clinical Genetics, Odence C, Denmark
| | - Inge Sokilde Pedersen
- Aalborg University Hospital, Molecular Diagnostics, Aalborg, Denmark
- Aalborg University, Dept of Clinical Medicine, Aalborg, Denmark
| | - Susan M. Domchek
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Abramson Cancer Center, Philadelphia, PA USA
| | - Katherine L. Nathanson
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Abramson Cancer Center, Philadelphia, PA USA
| | - Conxi Lazaro
- ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Molecular Diagnostic Unit, Hereditary Cancer Program, Barcelona, Spain
| | - Caroline Maria Rossing
- Rigshospitalet, Copenhagen University Hospital, Center for Genomic Medicine, Copenhagen, Denmark
| | - Irene L. Andrulis
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON Canada
| | - Manuel R. Teixeira
- Portuguese Oncology Institute, Department of Genetics, Porto, Portugal
- University of Porto, Biomedical Sciences Institute (ICBAS), Porto, Portugal
| | - Paul James
- Peter MacCallum Cancer Center, Parkville Familial Cancer Centre, Melbourne, VIC Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC Australia
| | - Judy Garber
- Dana-Farber Cancer Institute, Cancer Risk and Prevention Clinic, Boston, MA USA
| | | | - SWE-BRCA Investigators
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
- University of Turku and Åbo Akademi University, Turku Bioscience Centre, Turku, Finland
- University of Helsinki, Department of Clinical Genetics, Helsinki University Hospital, Helsinki, Finland
- Mayo Clinic, Department of Laboratory Medicine and Pathology, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
- CHU de Quebec Research Center, Genomics Center, Québec City, QC Canada
- Mayo Clinic, Department of Health Sciences Research, Rochester, MN USA
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Oncology, Cambridge, UK
- McGill University, Program in Cancer Genetics, Departments of Human Genetics and Oncology, Montréal, QC Canada
- University of Cambridge, Department of Medical Genetics, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Hereditary Breast and Ovarian Cancer, Cologne, Germany
- Faculty of Medicine and University Hospital Cologne, University of Cologne, Center for Molecular Medicine Cologne (CMMC), Cologne, Germany
- Erasmus MC Cancer Institute, Department of Medical Oncology, Family Cancer Clinic, Rotterdam, The Netherlands
- Medical University of Vienna, Dept of OB/GYN and Comprehensive Cancer Center, Vienna, Austria
- Odense University Hospital, Department of Clinical Genetics, Odence C, Denmark
- Aalborg University Hospital, Molecular Diagnostics, Aalborg, Denmark
- Aalborg University, Dept of Clinical Medicine, Aalborg, Denmark
- Perelman School of Medicine at the University of Pennsylvania, Department of Medicine, Abramson Cancer Center, Philadelphia, PA USA
- ICO-IDIBELL (Bellvitge Biomedical Research Institute, Catalan Institute of Oncology), CIBERONC, Molecular Diagnostic Unit, Hereditary Cancer Program, Barcelona, Spain
- Rigshospitalet, Copenhagen University Hospital, Center for Genomic Medicine, Copenhagen, Denmark
- Lunenfeld-Tanenbaum Research Institute of Mount Sinai Hospital, Fred A. Litwin Center for Cancer Genetics, Toronto, ON Canada
- University of Toronto, Department of Molecular Genetics, Toronto, ON Canada
- Portuguese Oncology Institute, Department of Genetics, Porto, Portugal
- University of Porto, Biomedical Sciences Institute (ICBAS), Porto, Portugal
- Peter MacCallum Cancer Center, Parkville Familial Cancer Centre, Melbourne, VIC Australia
- The University of Melbourne, Sir Peter MacCallum Department of Oncology, Melbourne, VIC Australia
- Dana-Farber Cancer Institute, Cancer Risk and Prevention Clinic, Boston, MA USA
- City of Hope, Clinical Cancer Genomics, Duarte, CA USA
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
- Pomeranian Medical University, Independent Laboratory of Molecular Biology and Genetic Diagnostics, Szczecin, Poland
- National Centre for Scientific Research ‘Demokritos’, Molecular Diagnostics Laboratory, INRASTES, Athens, Greece
- Stanford Cancer Institute, Stanford University School of Medicine, Department of Medicine, Division of Oncology, Stanford, CA USA
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, VIC Australia
- The University of Melbourne, Department of Clinical Pathology, Melbourne, VIC Australia
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
- University of Helsinki, Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Örebro University Hospital, Department of Oncology, Örebro, Sweden
| | - Anna Jakubowska
- Pomeranian Medical University, Department of Genetics and Pathology, Szczecin, Poland
- Pomeranian Medical University, Independent Laboratory of Molecular Biology and Genetic Diagnostics, Szczecin, Poland
| | - Drakoulis Yannoukakos
- National Centre for Scientific Research ‘Demokritos’, Molecular Diagnostics Laboratory, INRASTES, Athens, Greece
| | - Esther M. John
- Stanford Cancer Institute, Stanford University School of Medicine, Department of Medicine, Division of Oncology, Stanford, CA USA
| | - Melissa C. Southey
- Monash University, Precision Medicine, School of Clinical Sciences at Monash Health, Clayton, VIC Australia
- The University of Melbourne, Department of Clinical Pathology, Melbourne, VIC Australia
| | - Marjanka K. Schmidt
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Division of Molecular Pathology, Amsterdam, The Netherlands
- The Netherlands Cancer Institute-Antoni van Leeuwenhoek hospital, Division of Psychosocial Research and Epidemiology, Amsterdam, The Netherlands
| | - Antonis C. Antoniou
- University of Cambridge, Centre for Cancer Genetic Epidemiology, Department of Public Health and Primary Care, Cambridge, UK
| | - Georgia Chenevix-Trench
- QIMR Berghofer Medical Research Institute, Department of Genetics and Computational Biology, Brisbane, QLD Australia
| | - Carl Blomqvist
- University of Helsinki, Department of Oncology, Helsinki University Hospital, Helsinki, Finland
- Örebro University Hospital, Department of Oncology, Örebro, Sweden
| | - Heli Nevanlinna
- University of Helsinki, Department of Obstetrics and Gynecology, Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
60
|
Mohammadi S, Arefnezhad R, Danaii S, Yousefi M. New insights into the core Hippo signaling and biological macromolecules interactions in the biology of solid tumors. Biofactors 2020; 46:514-530. [PMID: 32445262 DOI: 10.1002/biof.1634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/16/2020] [Accepted: 04/17/2020] [Indexed: 12/26/2022]
Abstract
As an evolutionarily conserved pathway, Hippo signaling pathway impacts different pathology and physiology processes such as wound healing, tissue repair/size and regeneration. When some components of Hippo signaling dysregulated, it affects cancer cells proliferation. Moreover, the relation Hippo pathway with other signaling including Wnt, TGFβ, Notch, and EGFR signaling leaves effect on the proliferation of cancer cells. Utilizing a number of therapeutic approaches, such as siRNAs and long noncoding RNA (lncRNA) to prevent cancer cells through the targeting of Hippo pathways, can provide new insights into cancer target therapy. The purpose of present review, first of all, is to demonstrate the importance of Hippo signaling and its relation with other signaling pathways in cancer. It also tries to demonstrate targeting Hippo signaling progress in cancer therapy.
Collapse
Affiliation(s)
- Solmaz Mohammadi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Shahla Danaii
- Gynecology Department, Eastern Azerbaijan ACECR ART Center, Eastern Azerbaijan Branch of ACECR, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Depatment of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
61
|
Jin L, Wu Z, Wang Y, Zhao X. Cryptotanshinone attenuates the stemness of non-small cell lung cancer cells via promoting TAZ translocation from nuclear to cytoplasm. Chin Med 2020; 15:66. [PMID: 32612672 PMCID: PMC7325009 DOI: 10.1186/s13020-020-00348-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 06/23/2020] [Indexed: 12/26/2022] Open
Abstract
Background Cancer stem cells (CSCs) are regarded as the root of tumor progression, thus representing an anti-cancer therapy through targeting this cell sub-population. Methods Non-small cell lung cancer (NSCLC) CSCs were enriched by non-adherent spheroid formation analysis. Lentivirus infection was used to stably change gene expression. Cell cycle, EdU incorporation, cell apoptosis, cell viability, ALDH1 activity, spheroid formation and in vivo tumor initiation assays were performed to detect the effects of Cryptotanshinone (CT), a traditional Chinese herb medicine, on the stemness of NSCLC cells. RNA-sequencing combined qRT-PCR and western blot analysis were constructed to explore the underlying mechanism contributing to CT-mediated effects. Results CT could attenuate the stemness of NSCLC CSCs, as evident by the reduced spheroid formation ability, stemness marker expression and ALDH1 activity. Additionally, CT provoked NSCLC CSCs entry into the cell cycle. RNA-sequencing analysis showed that Hippo signaling pathway was highly enriched in NSCLC CSCs with CT treatment. Further experiments disclosed that CT decreased TAZ (a regulatory master of Hippo pathway) expression via promoting its nuclear-cytoplasm translocation in NSCLC CSCs. Also, overexpression of TAZ partially saved the attenuation of CT on the stemness of NSCLC CSCs. Notably, CT enhanced the sensitivity of tyrosine kinase inhibitor (TKI) and chemotherapy in NSCLC CSCs. Conclusions This work reveals that CT attenuates NSCLC CSC stemness, implying the possibility of CT as an adjuvant therapy for NSCLC.
Collapse
Affiliation(s)
- Linling Jin
- Department of Respiratory & Critical Care Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029 People's Republic of China
| | - Zhenzhen Wu
- Department of Respiratory & Critical Care Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029 People's Republic of China
| | - Yanli Wang
- Department of Respiratory & Critical Care Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029 People's Republic of China
| | - Xin Zhao
- Department of Respiratory & Critical Care Medicine, Jiangsu Province Hospital, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Rd, Nanjing, 210029 People's Republic of China
| |
Collapse
|
62
|
Cao X, Wang C, Liu J, Zhao B. Regulation and functions of the Hippo pathway in stemness and differentiation. Acta Biochim Biophys Sin (Shanghai) 2020; 52:736-748. [DOI: 10.1093/abbs/gmaa048] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 12/20/2019] [Accepted: 02/24/2020] [Indexed: 12/15/2022] Open
Abstract
Abstract
The Hippo pathway plays important roles in organ development, tissue regeneration, and human diseases, such as cancer. In the canonical Hippo pathway, the MST1/2-LATS1/2 kinase cascade phosphorylates and inhibits transcription coactivators Yes-associated protein and transcription coactivator with PDZ-binding motif and thus regulates transcription of genes important for cell proliferation and apoptosis. However, recent studies have depicted a much more complicate picture of the Hippo pathway with many new components and regulatory stimuli involving both chemical and mechanical signals. Furthermore, accumulating evidence indicates that the Hippo pathway also plays important roles in the determination of cell fates, such as self-renewal and differentiation. Here, we review regulations of the Hippo pathway and its functions in stemness and differentiation emphasizing recent discoveries.
Collapse
Affiliation(s)
- Xiaolei Cao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Chenliang Wang
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Jiyang Liu
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
| | - Bin Zhao
- MOE key Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China, and
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
63
|
Kyriazoglou A, Liontos M, Zakopoulou R, Kaparelou M, Tsiara A, Papatheodoridi AM, Georgakopoulou R, Zagouri F. The Role of the Hippo Pathway in Breast Cancer Carcinogenesis, Prognosis, and Treatment: A Systematic Review. Breast Care (Basel) 2020; 16:6-15. [PMID: 33716627 DOI: 10.1159/000507538] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
Background The Hippo pathway is a developmental pathway recently discovered in Drosophila melanogaster; in mammals it normally controls organ development and wound healing. Hippo signaling is deregulated in breast cancer (BC). MST1/2 and LATS1/2 kinases are the upstream molecular elements of Hippo signaling which phosphorylate and regulate the two effectors of Hippo signaling, YAP1 and TAZ cotranscriptional activators. The two molecular effectors of the Hippo pathway facilitate their activity through TEAD transcription factors. Several molecular pathways with known oncogenic functions cross-talk with the Hippo pathway. Methods A systematic review studying the correlation of the Hippo pathway with BC tumorigenesis, prognosis, and treatment was performed. Results Recent literature highlights the critical role of Hippo signaling in a wide spectrum of biological mechanisms in BC. Discussion The Hippo pathway has a crucial position in BC molecular biology, cellular behavior, and response to treatment. Targeting the Hippo pathway could potentially improve the prognosis and outcome of BC patients.
Collapse
Affiliation(s)
| | - Michalis Liontos
- Department of Clinical Therapeutics, General Hospital Alexandra, Athens, Greece
| | - Roubini Zakopoulou
- Department of Clinical Therapeutics, General Hospital Alexandra, Athens, Greece
| | - Maria Kaparelou
- Department of Clinical Therapeutics, General Hospital Alexandra, Athens, Greece
| | - Anna Tsiara
- Department of Clinical Therapeutics, General Hospital Alexandra, Athens, Greece
| | | | | | - Flora Zagouri
- Department of Clinical Therapeutics, General Hospital Alexandra, Athens, Greece
| |
Collapse
|
64
|
Ménard A, Abou Nader N, Levasseur A, St-Jean G, Le Gad-Le Roy M, Boerboom D, Benoit-Biancamano MO, Boyer A. Targeted Disruption of Lats1 and Lats2 in Mice Impairs Adrenal Cortex Development and Alters Adrenocortical Cell Fate. Endocrinology 2020; 161:5815549. [PMID: 32243503 PMCID: PMC7211035 DOI: 10.1210/endocr/bqaa052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Accepted: 04/02/2020] [Indexed: 02/08/2023]
Abstract
It has recently been shown that the loss of the Hippo signaling effectors Yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) in adrenocortical steroidogenic cells impairs the postnatal maintenance of the adrenal gland. To further explore the role of Hippo signaling in mouse adrenocortical cells, we conditionally deleted the key Hippo kinases large tumor suppressor homolog kinases 1 and -2 (Lats1 and Lats2, two kinases that antagonize YAP and TAZ transcriptional co-regulatory activity) in steroidogenic cells using an Nr5a1-cre strain (Lats1flox/flox;Lats2flox/flox;Nr5a1-cre). We report here that developing adrenocortical cells adopt characteristics of myofibroblasts in both male and female Lats1flox/flox;Lats2flox/flox;Nr5a1-cre mice, resulting in a loss of steroidogenic gene expression, adrenal failure and death by 2 to 3 weeks of age. A marked accumulation of YAP and TAZ in the nuclei of the myofibroblast-like cell population with an accompanying increase in the expression of their transcriptional target genes in the adrenal glands of Lats1flox/flox;Lats2flox/flox;Nr5a1-cre animals suggested that the myofibroblastic differentiation could be attributed in part to YAP and TAZ. Taken together, our results suggest that Hippo signaling is required to maintain proper adrenocortical cell differentiation and suppresses their differentiation into myofibroblast-like cells.
Collapse
Affiliation(s)
- Amélie Ménard
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Nour Abou Nader
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Adrien Levasseur
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Guillaume St-Jean
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Marie Le Gad-Le Roy
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Derek Boerboom
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Marie-Odile Benoit-Biancamano
- Département de Pathologie et Microbiologie Vétérinaire, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
| | - Alexandre Boyer
- Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Canada
- Correspondence: Alexandre Boyer, Centre de Recherche en Reproduction et Fertilité, Faculté de Médecine Vétérinaire, Université de Montréal, 3200 rue Sicotte, St-Hyacinthe, QC, J2S 7C6, Canada. E-mail:
| |
Collapse
|
65
|
ARID1A determines luminal identity and therapeutic response in estrogen-receptor-positive breast cancer. Nat Genet 2020; 52:198-207. [PMID: 31932695 DOI: 10.1038/s41588-019-0554-0] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 11/21/2019] [Indexed: 12/14/2022]
Abstract
Mutations in ARID1A, a subunit of the SWI/SNF chromatin remodeling complex, are the most common alterations of the SWI/SNF complex in estrogen-receptor-positive (ER+) breast cancer. We identify that ARID1A inactivating mutations are present at a high frequency in advanced endocrine-resistant ER+ breast cancer. An epigenome CRISPR-CAS9 knockout (KO) screen identifies ARID1A as the top candidate whose loss determines resistance to the ER degrader fulvestrant. ARID1A inactivation in cells and in patients leads to resistance to ER degraders by facilitating a switch from ER-dependent luminal cells to ER-independent basal-like cells. Cellular plasticity is mediated by loss of ARID1A-dependent SWI/SNF complex targeting to genomic sites of the luminal lineage-determining transcription factors including ER, forkhead box protein A1 (FOXA1) and GATA-binding factor 3 (GATA3). ARID1A also regulates genome-wide ER-FOXA1 chromatin interactions and ER-dependent transcription. Altogether, we uncover a critical role for ARID1A in maintaining luminal cell identity and endocrine therapeutic response in ER+ breast cancer.
Collapse
|
66
|
Yin L, Li W, Xu A, Shi H, Wang K, Yang H, Wang R, Peng B. SH3BGRL2 inhibits growth and metastasis in clear cell renal cell carcinoma via activating hippo/TEAD1-Twist1 pathway. EBioMedicine 2020; 51:102596. [PMID: 31911271 PMCID: PMC7000347 DOI: 10.1016/j.ebiom.2019.12.005] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 12/06/2019] [Accepted: 12/06/2019] [Indexed: 12/12/2022] Open
Abstract
Background Clear cell renal cell carcinoma (ccRCC) is one of the most prevalent malignancies in the world, and tumor metastasis is still the main reason for disease progression. Accumulating evidence shows that SH3BGRL2 may play a key role in tumor progression and metastasis. However, the role of SH3BGRL2 in ccRCC has not been systematically investigated and remains elusive. Methods The clinical significance of SH3BGRL2 was evaluated by bioinformatic analysis and tissue microarray (TMA) samples. SH3BGRL2 expression was determined by RT-PCR, western blot and immunohistochemistry staining. Tumor suppressive effect of SH3BGRL2 was determined by both in vitro and in vivo studies. Western blot, chromatin immunoprecipitation assay and luciferase report assay were applied for mechanism dissection. Findings SH3BGRL2 was crucial for epithelial-mesenchymal transition (EMT) progression and metastasis in ccRCC. Clinically, SH3BGRL2 was identified as an independent prognostic factor for ccRCC patients. Gain- and loss-of-function results suggested that SH3BGRL2 played a critical role in cell proliferation, migration and invasion. Mechanistically, we found that SH3BGRL2 acted as a tumor suppressor through Hippo/TEAD1 signaling, then TEAD1 altered Twist1 expression at the transcriptional level via directly binding to its promoter region. Interpretation Our findings established that SH3BGRL2 performed as a tumor suppressor and modulator via Hippo/TEAD1-Twist1 signaling in ccRCC, and the alteration of SH3BGRL2 could serve as a functional response biomarker of tumor progression and metastasis in ccRCC.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Wenjia Li
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Aiming Xu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Heng Shi
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Huan Yang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Ronghao Wang
- School of basic medical sciences, Southwest Medical University, Luzhou, China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.
| |
Collapse
|
67
|
Gao Q, López-Knowles E, Cheang MCU, Morden J, Ribas R, Sidhu K, Evans D, Martins V, Dodson A, Skene A, Holcombe C, Mallon E, Evans A, Bliss JM, Robertson J, Smith I, Martin LA, Dowsett M. Impact of aromatase inhibitor treatment on global gene expression and its association with antiproliferative response in ER+ breast cancer in postmenopausal patients. Breast Cancer Res 2019; 22:2. [PMID: 31892336 PMCID: PMC6938628 DOI: 10.1186/s13058-019-1223-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 11/04/2019] [Indexed: 12/31/2022] Open
Abstract
Background Endocrine therapy reduces breast cancer mortality by 40%, but resistance remains a major clinical problem. In this study, we sought to investigate the impact of aromatase inhibitor (AI) therapy on gene expression and identify gene modules representing key biological pathways that relate to early AI therapy resistance. Methods Global gene expression was measured on pairs of core-cut biopsies taken at baseline and at surgery from 254 patients with ER-positive primary breast cancer randomised to receive 2-week presurgical AI (n = 198) or no presurgical treatment (control n = 56) from the POETIC trial. Data from the AI group was adjusted to eliminate artefactual process-related changes identified in the control group. The response was assessed by changes in the proliferation marker, Ki67. Results High baseline ESR1 expression associated with better AI response in HER2+ tumours but not HER2− tumours. In HER2− tumours, baseline expression of 48 genes associated with poor antiproliferative response (p < 0.005) including PERP and YWHAQ, the two most significant, and the transcription co-regulators (SAP130, HDAC4, and NCOA7) which were among the top 16 most significant. Baseline gene signature scores measuring cell proliferation, growth factor signalling (ERBB2-GS, RET/GDNF-GS, and IGF-1-GS), and immune activity (STAT1-GS) were significantly higher in poor AI responders. Two weeks of AI caused downregulation of genes involved in cell proliferation and ER signalling, as expected. Signature scores of E2F activation and TP53 dysfunction after 2-week AI were associated with poor AI response in both HER2− and HER2+ patients. Conclusions There is a high degree of heterogeneity in adaptive mechanisms after as little as 2-week AI therapy; however, all appear to converge on cell cycle regulation. Our data support the evaluation of whether an E2F signatures after short-term exposure to AI may identify those patients most likely to benefit from the early addition of CDK4/6 inhibitors. Trial registration ISRCTN, ISRCTN63882543, registered on 18 December 2007.
Collapse
Affiliation(s)
- Qiong Gao
- Breast Cancer Now Research Centre, ICR, London, UK
| | - Elena López-Knowles
- Breast Cancer Now Research Centre, ICR, London, UK.,Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Maggie Chon U Cheang
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | - James Morden
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | | | - Kally Sidhu
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - David Evans
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Vera Martins
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Andrew Dodson
- Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK
| | - Anthony Skene
- Royal Bournemouth Hospital, Castle Lane East, Bournemouth, UK
| | - Chris Holcombe
- Royal Liverpool University Hospital, 200 London Road, Liverpool, UK
| | | | | | - Judith M Bliss
- Clinical Trials and Statistics Unit, The Institute of Cancer Research, London, UK
| | | | - Ian Smith
- Breast Unit, Royal Marsden Hospital, London, UK
| | | | - Mitch Dowsett
- Breast Cancer Now Research Centre, ICR, London, UK. .,Ralph Lauren Centre for Breast Cancer Research, Royal Marsden Hospital, London, UK.
| | | |
Collapse
|
68
|
Tang F, Gao R, Jeevan-Raj B, Wyss CB, Kalathur RKR, Piscuoglio S, Ng CKY, Hindupur SK, Nuciforo S, Dazert E, Bock T, Song S, Buechel D, Morini MF, Hergovich A, Matthias P, Lim DS, Terracciano LM, Heim MH, Hall MN, Christofori G. LATS1 but not LATS2 represses autophagy by a kinase-independent scaffold function. Nat Commun 2019; 10:5755. [PMID: 31848340 PMCID: PMC6917744 DOI: 10.1038/s41467-019-13591-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/14/2019] [Indexed: 12/27/2022] Open
Abstract
Autophagy perturbation represents an emerging therapeutic strategy in cancer. Although LATS1 and LATS2 kinases, core components of the mammalian Hippo pathway, have been shown to exert tumor suppressive activities, here we report a pro-survival role of LATS1 but not LATS2 in hepatocellular carcinoma (HCC) cells. Specifically, LATS1 restricts lethal autophagy in HCC cells induced by sorafenib, the standard of care for advanced HCC patients. Notably, autophagy regulation by LATS1 is independent of its kinase activity. Instead, LATS1 stabilizes the autophagy core-machinery component Beclin-1 by promoting K27-linked ubiquitination at lysine residues K32 and K263 on Beclin-1. Consequently, ubiquitination of Beclin-1 negatively regulates autophagy by promoting inactive dimer formation of Beclin-1. Our study highlights a functional diversity between LATS1 and LATS2, and uncovers a scaffolding role of LATS1 in mediating a cross-talk between the Hippo signaling pathway and autophagy.
Collapse
Affiliation(s)
- Fengyuan Tang
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Ruize Gao
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Beena Jeevan-Raj
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Christof B Wyss
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | | - Charlotte K Y Ng
- Institute of Pathology, University Hospital Basel, Basel, Switzerland
| | | | - Sandro Nuciforo
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Eva Dazert
- Biozentrum, University of Basel, Basel, Switzerland
| | - Thomas Bock
- Biozentrum, University of Basel, Basel, Switzerland
| | - Shuang Song
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - David Buechel
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marco F Morini
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | - Patrick Matthias
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Dae-Sik Lim
- Korea Advanced Institute of Science and Technology, Daejeon, 34141, Republic of Korea
| | | | - Markus H Heim
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | | | | |
Collapse
|
69
|
Moon S, Lee OH, Lee S, Lee J, Park H, Park M, Chang EM, Park KH, Choi Y. STK3/4 Expression Is Regulated in Uterine Endometrial Cells during the Estrous Cycle. Cells 2019; 8:cells8121643. [PMID: 31847471 PMCID: PMC6952811 DOI: 10.3390/cells8121643] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/11/2022] Open
Abstract
The uterus is dynamically regulated in response to various signaling triggered by hormones during the estrous cycle. The Hippo signaling pathway is known as an important signaling for regulating cellular processes during development by balancing between cell growth and apoptosis. Serine/threonine protein kinase 3/4 (STK3/4) is a key component of the Hippo signaling network. However, the regulation of STK3/4-Hippo signaling in the uterus is little known. In this study, we investigated the regulation and expression of STK3/4 in the uterine endometrium during the estrous cycle. STK3/4 expression was dynamically regulated in the uterus during the estrous cycle. STK3/4 protein expression was gradually increased from the diestrus stage and reached the highest in the estrus stage. STK3/4 was exclusively localized in the luminal and glandular epithelial cells of the uterus, and phosphorylated STK3/4 was also increased at the estrus stage. Moreover, the increase of STK3/4 expression in uteri was induced by administration of estradiol, but not by progesterone injection in ovariectomized mice. Pretreatment with an estrogen receptor antagonist ICI 182,780 reduced estrogen-induced STK3/4 expression and its phosphorylation. The estrogen-induced STK3/4 expression was related to the increase in phosphorylation of downstream targets including LATS1/2 and YAP. These findings suggest that STK3/4-Hippo signaling acts a novel signaling pathway in the uterine epithelium and STK3/4-Hippo is one of key molecules for connecting between the estrogen downstream signaling pathway and the Hippo signaling pathway leading to regulate dynamic uterine epithelium during the estrous cycle.
Collapse
Affiliation(s)
- Sohyeon Moon
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center, Konkuk University, Seoul 05029, Korea; (S.M.); (J.L.); (H.P.)
| | - Ok-Hee Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Korea; (O.-H.L.); (S.L.); (K.-H.P.)
| | - Sujin Lee
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Korea; (O.-H.L.); (S.L.); (K.-H.P.)
| | - Jihyun Lee
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center, Konkuk University, Seoul 05029, Korea; (S.M.); (J.L.); (H.P.)
| | - Haeun Park
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center, Konkuk University, Seoul 05029, Korea; (S.M.); (J.L.); (H.P.)
| | - Miseon Park
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul 06135, Korea; (M.P.); (E.M.C.)
| | - Eun Mi Chang
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul 06135, Korea; (M.P.); (E.M.C.)
| | - Keun-Hong Park
- Department of Biomedical Science, CHA University, Gyeonggi-do 13488, Korea; (O.-H.L.); (S.L.); (K.-H.P.)
| | - Youngsok Choi
- Department of Stem Cell and Regenerative Biotechnology, Humanized Pig Research Center, Konkuk University, Seoul 05029, Korea; (S.M.); (J.L.); (H.P.)
- Correspondence: ; Tel.: +82-2-450-3969
| |
Collapse
|
70
|
Initiation of human mammary cell tumorigenesis by mutant KRAS requires YAP inactivation. Oncogene 2019; 39:1957-1968. [PMID: 31772328 PMCID: PMC7044112 DOI: 10.1038/s41388-019-1111-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 05/14/2019] [Accepted: 05/27/2019] [Indexed: 01/13/2023]
Abstract
High YAP activity is associated with poor prognosis human breast cancers, but its role during the initial stage of mammary cell transformation is unknown. To address this question, we designed experiments that exploit the ability of KRASG12D-transduced subsets of freshly isolated normal human mammary cells to form invasive tumors rapidly and efficiently when transplanted into immunodeficient mice. Initial examination of the newly developing tumors thus generated revealed a consistent marked loss of nuclear YAP, independent of the initial primary human mammary cell type transduced. Conversely, co-transduction of the same subsets of primary human mammary cells with KRASG12D plus the constitutively active YAPS127A prevented tumor formation. These findings contrast with the enhanced display of transformed properties obtained when the immortalized, but non-tumorigenic MCF10A cells are transduced just with YAPS127A. In addition, we show that YAPS127A-transduction of the human MDA-MB-231 breast cancer cell line (that carry a similar KRAS mutation) enhances their metastatic activity in vivo. We also discover that the KRASG12D-induced early loss of YAP in primary human mammary cells is associated with their induced secretion of amphiregulin. Collectively, these findings suggest that YAP can differentially affect the acquisition of malignant properties by human mammary cells at different stages of their transformation.
Collapse
|
71
|
A CD146 FACS Protocol Enriches for Luminal Keratin 14/19 Double Positive Human Breast Progenitors. Sci Rep 2019; 9:14843. [PMID: 31619692 PMCID: PMC6795797 DOI: 10.1038/s41598-019-50903-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/20/2019] [Indexed: 12/13/2022] Open
Abstract
Human breast cancer is believed to arise in luminal progenitors within the normal breast. A subset of these are double positive (DP) for basal and luminal keratins and localizes to a putative stem cell zone within ducts. We here present a new protocol based on a combination of CD146 with CD117 and CD326 which provides an up to thirty fold enrichment of the DP cells. We show by expression profiling, colony formation, and morphogenesis that CD146high/CD117high/CD326high DP cells belong to a luminal progenitor compartment. While these DP cells are located quite uniformly in ducts, with age a variant type of DP (vDP) cells, which is mainly CD146-negative, accumulates in lobules. Intriguingly, in specimens with BRCA1 mutations known to predispose for cancer, higher frequencies of lobular vDP cells are observed. We propose that vDP cells are strong candidates for tracing the cellular origin of breast cancer.
Collapse
|
72
|
Breast Cancer Stem Cells as Drivers of Tumor Chemoresistance, Dormancy and Relapse: New Challenges and Therapeutic Opportunities. Cancers (Basel) 2019; 11:cancers11101569. [PMID: 31619007 PMCID: PMC6826533 DOI: 10.3390/cancers11101569] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is the most frequent cancer among women worldwide. Therapeutic strategies to prevent or treat metastatic disease are still inadequate although great progress has been made in treating early-stage breast cancer. Cancer stem-like cells (CSCs) that are endowed with high plasticity and self-renewal properties have been shown to play a key role in breast cancer development, progression, and metastasis. A subpopulation of CSCs that combines tumor-initiating capacity and a dormant/quiescent/slow cycling status is present throughout the clinical history of breast cancer patients. Dormant/quiescent/slow cycling CSCs are a key component of tumor heterogeneity and they are responsible for chemoresistance, tumor migration, and metastatic dormancy, defined as the ability of CSCs to survive in target organs and generate metastasis up to two decades after diagnosis. Understanding the strategies that are used by CSCs to resist conventional and targeted therapies, to interact with their niche, to escape immune surveillance, and finally to awaken from dormancy is of key importance to prevent and treat metastatic cancer. This review summarizes the current understanding of mechanisms involved in CSCs chemoresistance, dissemination, and metastasis in breast cancer, with a particular focus on dormant cells. Finally, we discuss how advancements in the detection, molecular understanding, and targeting of dormant CSCs will likely open new therapeutic avenues for breast cancer treatment.
Collapse
|
73
|
Yang K, Zeng L, Ge A, Chen Z, Bao T, Long Z, Ge J, Huang L. Investigating the regulation mechanism of baicalin on triple negative breast cancer's biological network by a systematic biological strategy. Biomed Pharmacother 2019; 118:109253. [PMID: 31545288 DOI: 10.1016/j.biopha.2019.109253] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2019] [Revised: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To investigate the regulation mechanism of baicalin on triple negative breast cancer (TNBC)'s biological network by a systematic biological strategy and cytology experiment. METHODS A systematic biological methodology is utilized to predict the potential targets of baicalin, collect the genes of TNBC, and analyze the TNBC and baicalin's network. After the systematic biological analysis is performed, the cytology experiment, real-time quantitative PCR (qPCR) is used to validate the key biological processes and signaling pathways. RESULTS After systematic biological analysis, two networks were constructed and analyzed: (1) TNBC network; (2) Baicalin-TNBC protein-protein interaction (PPI) network. Several TNBC-related, treatment-related targets, clusters, signaling pathways and biological processes were found. Cytology experiment shows that baicalin can inhibit the proliferation, migration and invasion of breast cancer MDA-MB-231 cells in vitro (P < 0.05). The results of qPCR showed that baicalin increase the expression of E-cadherin mRNA, and decrease the expression of vimentin, β-catenin, c-Myc and MMP-7 mRNA in LPS-induced breast cancer MDA-MB-231 cells (P < 0.05). CONCLUSION Baicalin may achieve anti-tumor effects through regulating the targets, biological processes and pathways found in this research.
Collapse
Affiliation(s)
- Kailin Yang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China; Capital Medical University, Beijing, China
| | - Liuting Zeng
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Anqi Ge
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan Province, China
| | - Zhouhua Chen
- The Second People's Hospital of Xiangtan City, Xiangtan, Hunan Province, China
| | - Tingting Bao
- Beijing University of Chinese Medicine, Beijing, China
| | - Zhiyong Long
- Shantou University Medical College, Shantou, Guangdong Province, China
| | - Jinwen Ge
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| | - Lizhong Huang
- Hunan University of Chinese Medicine, Changsha, Hunan Province, China.
| |
Collapse
|
74
|
Fu NY, Nolan E, Lindeman GJ, Visvader JE. Stem Cells and the Differentiation Hierarchy in Mammary Gland Development. Physiol Rev 2019; 100:489-523. [PMID: 31539305 DOI: 10.1152/physrev.00040.2018] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The mammary gland is a highly dynamic organ that undergoes profound changes within its epithelium during puberty and the reproductive cycle. These changes are fueled by dedicated stem and progenitor cells. Both short- and long-lived lineage-restricted progenitors have been identified in adult tissue as well as a small pool of multipotent mammary stem cells (MaSCs), reflecting intrinsic complexity within the epithelial hierarchy. While unipotent progenitor cells predominantly execute day-to-day homeostasis and postnatal morphogenesis during puberty and pregnancy, multipotent MaSCs have been implicated in coordinating alveologenesis and long-term ductal maintenance. Nonetheless, the multipotency of stem cells in the adult remains controversial. The advent of large-scale single-cell molecular profiling has revealed striking changes in the gene expression landscape through ontogeny and the presence of transient intermediate populations. An increasing number of lineage cell-fate determination factors and potential niche regulators have now been mapped along the hierarchy, with many implicated in breast carcinogenesis. The emerging diversity among stem and progenitor populations of the mammary epithelium is likely to underpin the heterogeneity that characterizes breast cancer.
Collapse
Affiliation(s)
- Nai Yang Fu
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Emma Nolan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Geoffrey J Lindeman
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| | - Jane E Visvader
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore, Singapore; Tumour-Host Interaction Laboratory, Francis Crick Institute, London, United Kingdom; Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia; Royal Melbourne Hospital and Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia; and Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
75
|
Pan Z, Tian Y, Cao C, Niu G. The Emerging Role of YAP/TAZ in Tumor Immunity. Mol Cancer Res 2019; 17:1777-1786. [PMID: 31308148 DOI: 10.1158/1541-7786.mcr-19-0375] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/03/2019] [Accepted: 07/10/2019] [Indexed: 11/16/2022]
Abstract
Yes-associated protein (YAP)/WW domain-containing transcription regulator 1 (TAZ) is an important transcriptional regulator and effector of the Hippo signaling pathway that has emerged as a critical determinant of malignancy in many human tumors. YAP/TAZ expression regulates the cross-talk between immune cells and tumor cells in the tumor microenvironment through its influence on T cells, myeloid-derived suppressor cells, and macrophages. However, the mechanisms underlying these effects are poorly understood. An improved understanding of the role of YAP/TAZ in tumor immunity is essential for exploring innovative tumor treatments and making further breakthroughs in antitumor immunotherapy. This review primarily focuses on the role of YAP/TAZ in immune cells, their interactions with tumor cells, and how this impacts on tumorigenesis, progression, and therapy resistance.
Collapse
Affiliation(s)
- Zhaoji Pan
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| | - Yiqing Tian
- Xinyi People's Hospital, Xinyi, Xuzhou, Jiangsu, P.R. China.
| | - Chengsong Cao
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| | - Guoping Niu
- Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Medical College of Southeast University, Xuzhou, Jiangsu, P.R. China
| |
Collapse
|
76
|
Feng X, Zhang M, Wang B, Zhou C, Mu Y, Li J, Liu X, Wang Y, Song Z, Liu P. CRABP2 regulates invasion and metastasis of breast cancer through hippo pathway dependent on ER status. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:361. [PMID: 31419991 PMCID: PMC6697986 DOI: 10.1186/s13046-019-1345-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
Background Triple Negative Breast cancer (TNBC) is incurable cancer with higher rates of relapse and shorter overall survival compared with other subtypes of breast cancer. Cellular retinoic acid binding protein 2 (CRABP2) belongs to fatty acid binding protein (FABP) family which binds with all-trans retinoic acid (RA). Previous studies from the database have reported the patients with high expression of CRABP2 showed different prognosis in ER+ and ER− breast cancer. However, its biological role and exact mechanism in breast cancer remain unknown. This aim of this study was to explore how CRABP2 regulated invasion and metastasis based on the estrogen receptor-α (herein called ER) status in breast cancer. Methods Immunohistochemical staining method was used to analyze the expression of CRABP2 in human breast cancer tissues. Lentivirus vector-based shRNA technique was used to test the functional relevance of CRABP2 knockdown in breast tumors. Tail vein injection model was used to examine the lung metastasis. Co-immunoprecipitation, Western blotting, immunofluorescence, and quantitative reverse transcription polymerase chain reaction (RT-qPCR) were conducted to investigate the underlying mechanism that influenced the ER to the regulation of CRABP2 to Lats1. Results We observed that knockdown of CRABP2 promotes EMT, invasion and metastasis of ER+ breast cancer cells in vitro and in vivo, whereas overexpression of CRABP2 yields the reverse results. In ER+ mammary cancer cells, the interaction of CRABP2 and Lats1 suppress the ubiquitination of Lats1 to activate Hippo pathway to inhibit the invasion and metastasis of ER+ mammary cancer. However, in ER− mammary cancer cells, the interaction of CRABP2 and Lats1 promote the ubiquitination of Lats1 to inactivate Hippo pathway to promote the invasion and metastasis of ER− mammary cancer. Conclusions Our findings indicate that CRABP2 can suppress invasion and metastasis of ER+ breast cancer and promote invasion and metastasis of ER− breast cancer by regulating the stability of Lats1 in vitro and in vivo, and it provides new ideas for breast cancer therapy. Electronic supplementary material The online version of this article (10.1186/s13046-019-1345-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xuefei Feng
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Miao Zhang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Bo Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Can Zhou
- Department of Breast Surgery, the first Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Yudong Mu
- Department of Clinical LaboratoryTumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Juan Li
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Xiaoxu Liu
- Department of Breast Surgery, the first Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Yaochun Wang
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China
| | - Zhangjun Song
- Department of Breast Disease Center, Tumor Hospital of Shaanxi Province, Affiliated to the Medical College of Xi'an Jiaotong University, 309 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.
| | - Peijun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China. .,Key Laboratory for Tumor Precision Medicine of Shaanxi Province, the First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta Western Rd, Xi'an, 710061, Shaanxi Province, China.
| |
Collapse
|
77
|
Yin L, Li W, Wang G, Shi H, Wang K, Yang H, Peng B. NR1B2 suppress kidney renal clear cell carcinoma (KIRC) progression by regulation of LATS 1/2-YAP signaling. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:343. [PMID: 31391070 PMCID: PMC6686564 DOI: 10.1186/s13046-019-1344-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 07/24/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Kidney Renal Clear Cell Carcinoma (KIRC) accounts for 75% of all renal cancers. Previous study had conflict evidences regarding NR1B2 role in cancer, and its expression and biological role in KIRC remained unclear. Our aims were to characterize the role of NR1B2 in KIRC. METHODS NR1B2 expression in TCGA database were analyzed. Clinical KIRC samples were examined by RT-PCR, western blot and tissue microarray (TMA). The relationship between NR1B2 expression and the clinical characteristics were evaluated. KIRC cell line were stably overexpressed NR1B2 or with an NR1B2 knocked down using lentivirus system. The cells were analyzed by migration and invasion assay, then injected into nude mice to assess tumor growth and metastasis. EMT marker expression and LATS 1/2-YAP pathway demonstration were detected by the TCGA database and western blot. RESULTS The expression of NR1B2 in KIRC was significantly down-regulated in the TCGA database and our clinical samples. Moreover, NR1B2 expression negatively correlated with tumor stage and positively correlated with overall and disease-free survival rate. Univariate and multivariate analyses indicated the expression level of NR1B2 could be used as an independent factor for predicting the prognosis of KIRC. Overexpression NR1B2 significantly inhibited and knockdown NR1B2 markedly promoted KIRC cell invasion and metastasis both in vitro and in vivo. Mechanistic investigations revealed that NR1B2 might be a tumor suppressor to inhibit EMT through the LATS1/2-YAP pathway. CONCLUSIONS our results defined NR1B2 as a tumor suppressor in KIRC that restricted EMT by the LATS1/2-YAP pathway.
Collapse
Affiliation(s)
- Lei Yin
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Wenjia Li
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guangchun Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Heng Shi
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.,Department of Urology, Shanghai Tenth People's Hospital, Nanjing Medical University, Nanjing, China
| | - Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China
| | - Huan Yang
- Department of Urology, Tongji Hospital,Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine in Tongji University, Shanghai, China.
| |
Collapse
|
78
|
Tharmapalan P, Mahendralingam M, Berman HK, Khokha R. Mammary stem cells and progenitors: targeting the roots of breast cancer for prevention. EMBO J 2019; 38:e100852. [PMID: 31267556 PMCID: PMC6627238 DOI: 10.15252/embj.2018100852] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/11/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022] Open
Abstract
Breast cancer prevention is daunting, yet not an unsurmountable goal. Mammary stem and progenitors have been proposed as the cells-of-origin in breast cancer. Here, we present the concept of limiting these breast cancer precursors as a risk reduction approach in high-risk women. A wealth of information now exists for phenotypic and functional characterization of mammary stem and progenitor cells in mouse and human. Recent work has also revealed the hormonal regulation of stem/progenitor dynamics as well as intrinsic lineage distinctions between mammary epithelial populations. Leveraging these insights, molecular marker-guided chemoprevention is an achievable reality.
Collapse
Affiliation(s)
| | - Mathepan Mahendralingam
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| | - Hal K Berman
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| | - Rama Khokha
- Princess Margaret Cancer CentreUniversity Health NetworkUniversity of TorontoTorontoONCanada
| |
Collapse
|
79
|
Pellacani D, Tan S, Lefort S, Eaves CJ. Transcriptional regulation of normal human mammary cell heterogeneity and its perturbation in breast cancer. EMBO J 2019; 38:e100330. [PMID: 31304632 PMCID: PMC6627240 DOI: 10.15252/embj.2018100330] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 10/22/2018] [Accepted: 11/08/2018] [Indexed: 12/18/2022] Open
Abstract
The mammary gland in adult women consists of biologically distinct cell types that differ in their surface phenotypes. Isolation and molecular characterization of these subpopulations of mammary cells have provided extensive insights into their different transcriptional programs and regulation. This information is now serving as a baseline for interpreting the heterogeneous features of human breast cancers. Examination of breast cancer mutational profiles further indicates that most have undergone a complex evolutionary process even before being detected. The consequent intra-tumoral as well as inter-tumoral heterogeneity of these cancers thus poses major challenges to deriving information from early and hence likely pervasive changes in potential therapeutic interest. Recently described reproducible and efficient methods for generating human breast cancers de novo in immunodeficient mice transplanted with genetically altered primary cells now offer a promising alternative to investigate initial stages of human breast cancer development. In this review, we summarize current knowledge about key transcriptional regulatory processes operative in these partially characterized subpopulations of normal human mammary cells and effects of disrupting these processes in experimentally produced human breast cancers.
Collapse
Affiliation(s)
- Davide Pellacani
- Terry Fox LaboratoryBritish Columbia Cancer AgencyVancouverBCCanada
| | - Susanna Tan
- Terry Fox LaboratoryBritish Columbia Cancer AgencyVancouverBCCanada
| | - Sylvain Lefort
- Terry Fox LaboratoryBritish Columbia Cancer AgencyVancouverBCCanada
| | - Connie J Eaves
- Terry Fox LaboratoryBritish Columbia Cancer AgencyVancouverBCCanada
| |
Collapse
|
80
|
Zhu C, Li L, Zhang Z, Bi M, Wang H, Su W, Hernandez K, Liu P, Chen J, Chen M, Huang THM, Chen L, Liu Z. A Non-canonical Role of YAP/TEAD Is Required for Activation of Estrogen-Regulated Enhancers in Breast Cancer. Mol Cell 2019; 75:791-806.e8. [PMID: 31303470 DOI: 10.1016/j.molcel.2019.06.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 05/06/2019] [Accepted: 06/05/2019] [Indexed: 12/26/2022]
Abstract
YAP/TEAD are nuclear effectors of the Hippo pathway, regulating organ size and tumorigenesis largely through promoter-associated function. However, their function as enhancer regulators remains poorly understood. Through an in vivo proximity-dependent labeling (BioID) technique, we identified YAP1 and TEAD4 protein as co-regulators of ERα on enhancers. The binding of YAP1/TEAD4 to ERα-bound enhancers is augmented upon E2 stimulation and is required for the induction of E2/ERα target genes and E2-induced oncogenic cell growth. Furthermore, their enhancer binding is a prerequisite for enhancer activation marked by eRNA transcription and for the recruitment of the enhancer activation machinery component MED1. The binding of TEAD4 on active ERE-containing enhancers is independent of its DNA-binding behavior, and instead, occurs through protein-tethering trans-binding. Our data reveal a non-canonical function of YAP1 and TEAD4 as ERα cofactors in regulating cancer growth, highlighting the potential of YAP/TEAD as possible actionable drug targets for ERα+ breast cancer.
Collapse
Affiliation(s)
- Chi Zhu
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Li Li
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Department of Oncology, The Second People's Hospital of Jiaozuo, Jiaozuo City, Henan 454001, China
| | - Zhao Zhang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Mingjun Bi
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Hu Wang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Wenyue Su
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Karen Hernandez
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Pingping Liu
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Junqiang Chen
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Mingqiu Chen
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA; Fujian Cancer Hospital & Fujian Medical University Cancer Hospital, Fuzhou, Fujian 350014, China
| | - Tim Hui-Ming Huang
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Lizhen Chen
- Barshop Institute for Longevity and Aging Studies, Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| | - Zhijie Liu
- Department of Molecular Medicine, Mays Cancer Center, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229, USA.
| |
Collapse
|
81
|
Xu S, Pang L, Liu Y, Lian X, Mo K, Lv R, Zhu H, Lv C, Lin J, Sun J, Xu L, Wang S. Akt plays indispensable roles during the first cell lineage differentiation of mouse. J Mol Histol 2019; 50:369-374. [PMID: 31190160 DOI: 10.1007/s10735-019-09833-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 06/10/2019] [Indexed: 01/06/2023]
Abstract
The first cell lineage differentiation occurs during the development of mouse 8-cell embryo to blastocyst. Akt is a potent kinase whose role during blastocyst formation has not been elucidated. In the present study, immunofluorescence results showed that the Akt protein was specifically localized to the outer cells of the morula. Akt-specific inhibitor MK2206 significantly inhibited mouse blastocyst formation and resulted in decreased expression of the trophectoderm marker Cdx2 and led to granular distribution of ERα in the cytoplasm. Furthermore, knockdown of ERα by siRNA microinjection can also lead to a decrease in the development rate of mouse blastocysts, accompanied by a decrease in the expression level of Yap protein. We conclude that Akt may be indispensable for the first cell lineage differentiation of mouse.
Collapse
Affiliation(s)
- Songhua Xu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fuzhou, 350122, People's Republic of China.,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Lili Pang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fuzhou, 350122, People's Republic of China
| | - Yue Liu
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fuzhou, 350122, People's Republic of China
| | - Xiuli Lian
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Kaien Mo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Ruimin Lv
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Huimin Zhu
- Fujian Key Laboratory of Medical Bioinformatics, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China.,Key Laboratory of Ministry of Education for Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Chengyu Lv
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Jianmin Lin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Jiandong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Lixuan Xu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Shie Wang
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine of Fujian Province University, Fuzhou, 350122, People's Republic of China. .,Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, 350122, People's Republic of China.
| |
Collapse
|
82
|
Tecalco-Cruz AC, Ramírez-Jarquín JO, Cruz-Ramos E. Estrogen Receptor Alpha and its Ubiquitination in Breast Cancer Cells. Curr Drug Targets 2019; 20:690-704. [DOI: 10.2174/1389450119666181015114041] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/09/2018] [Accepted: 10/09/2018] [Indexed: 12/23/2022]
Abstract
More than 70% of all breast cancer cases are estrogen receptor alpha-positive (ERα). ERα is a member of the nuclear receptor family, and its activity is implicated in the gene transcription linked to the proliferation of breast cancer cells, as well as in extranuclear signaling pathways related to the development of resistance to endocrine therapy. Protein-protein interactions and posttranslational modifications of ERα underlie critical mechanisms that modulate its activity. In this review, the relationship between ERα and ubiquitin protein (Ub), was investigated in the context of breast cancer cells. Interestingly, Ub can bind covalently or non-covalently to ERα resulting in either a proteolytic or non-proteolytic fate for this receptor. Thereby, Ub-dependent molecular pathways that modulate ERα signaling may play a central role in breast cancer progression, and consequently, present critical targets for treatment of this disease.
Collapse
Affiliation(s)
- Angeles C. Tecalco-Cruz
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Josué O. Ramírez-Jarquín
- Instituto de Fisiologia Celular. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| | - Eduardo Cruz-Ramos
- Instituto de Investigaciones Biomedicas. Universidad Nacional Autonoma de Mexico. Mexico City, 04510, Mexico
| |
Collapse
|
83
|
Obradović MMS, Hamelin B, Manevski N, Couto JP, Sethi A, Coissieux MM, Münst S, Okamoto R, Kohler H, Schmidt A, Bentires-Alj M. Glucocorticoids promote breast cancer metastasis. Nature 2019; 567:540-544. [PMID: 30867597 DOI: 10.1038/s41586-019-1019-4] [Citation(s) in RCA: 279] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 02/09/2019] [Indexed: 02/07/2023]
Abstract
Diversity within or between tumours and metastases (known as intra-patient tumour heterogeneity) that develops during disease progression is a serious hurdle for therapy1-3. Metastasis is the fatal hallmark of cancer and the mechanisms of colonization, the most complex step in the metastatic cascade4, remain poorly defined. A clearer understanding of the cellular and molecular processes that underlie both intra-patient tumour heterogeneity and metastasis is crucial for the success of personalized cancer therapy. Here, using transcriptional profiling of tumours and matched metastases in patient-derived xenograft models in mice, we show cancer-site-specific phenotypes and increased glucocorticoid receptor activity in distant metastases. The glucocorticoid receptor mediates the effects of stress hormones, and of synthetic derivatives of these hormones that are used widely in the clinic as anti-inflammatory and immunosuppressive agents. We show that the increase in stress hormones during breast cancer progression results in the activation of the glucocorticoid receptor at distant metastatic sites, increased colonization and reduced survival. Our transcriptomics, proteomics and phospho-proteomics studies implicate the glucocorticoid receptor in the activation of multiple processes in metastasis and in the increased expression of kinase ROR1, both of which correlate with reduced survival. The ablation of ROR1 reduced metastatic outgrowth and prolonged survival in preclinical models. Our results indicate that the activation of the glucocorticoid receptor increases heterogeneity and metastasis, which suggests that caution is needed when using glucocorticoids to treat patients with breast cancer who have developed cancer-related complications.
Collapse
Affiliation(s)
- Milan M S Obradović
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Wellmera AG, Basel, Switzerland
| | - Baptiste Hamelin
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nenad Manevski
- Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.,UCB Celltech, Development Sciences, Slough, UK
| | - Joana Pinto Couto
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Novartis Institutes for BioMedical Research, Basel, Switzerland
| | - Atul Sethi
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.,Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Marie-May Coissieux
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Simone Münst
- Institute of Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Ryoko Okamoto
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland.,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Hubertus Kohler
- Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland
| | - Alexander Schmidt
- Proteomics Core Facility, Biozentrum, University of Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Department of Biomedicine, Department of Surgery, University Hospital Basel, University of Basel, Basel, Switzerland. .,Friedrich Miescher Institute for Biomedical Research, Basel, Switzerland.
| |
Collapse
|
84
|
Lescoat A, Jégo P, Lecureur V. M-CSF and GM-CSF monocyte-derived macrophages in systemic sclerosis: the two sides of the same coin? Ann Rheum Dis 2019; 78:e19. [PMID: 29439999 DOI: 10.1136/annrheumdis-2018-213112] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 01/25/2018] [Indexed: 01/02/2023]
Affiliation(s)
- Alain Lescoat
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, F-35000 Rennes, France
- Médecine Interne, CHU Rennes, Rennes, France
| | - Patrick Jégo
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, F-35000 Rennes, France
- Médecine Interne, CHU Rennes, Rennes, France
| | - Valérie Lecureur
- Univ Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S 1085, F-35000 Rennes, France
| |
Collapse
|
85
|
Rahmat MB, Zhang S, Koh CG. POPX2 is a novel LATS phosphatase that regulates the Hippo pathway. Oncotarget 2019; 10:1525-1538. [PMID: 30863499 PMCID: PMC6407677 DOI: 10.18632/oncotarget.26689] [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: 10/25/2018] [Accepted: 02/09/2019] [Indexed: 12/23/2022] Open
Abstract
The Hippo pathway regulates cell proliferation, survival, apoptosis and differentiation. During carcinogenesis, members of the Hippo pathway are mutated to avoid anoikis and promote anchorage independent growth. Although many regulators of the Hippo pathway have been reported, negative regulators of the hippo kinases are not well studied. Through an interactome screen, we found that POPX2 phosphatase interacts with several of the Hippo pathway core kinases, including LATS1 which is the direct kinase regulating the transcription co-activators, YAP and TAZ. Phosphorylated YAP/TAZ are retained in the cytoplasm and prevented from translocation into the nucleus to activate transcription of target genes. We found that POPX2 could dephosphorylate LATS1 on Threonine-1079, leading to inactivation of LATS1 kinase. As a result, YAP/TAZ are not phosphorylated and are able to translocate into the nucleus to activate target genes involved in cell proliferation. Furthermore, POPX2 knock-out using CRISPR in the highly metastatic MDA-MB-231 breast cancer cells results in decreased cell proliferation and impairment of anchorage independent growth. We propose that POPX2 act as a suppressor of the Hippo pathway through LATS1 dephosphorylation and inactivation.
Collapse
Affiliation(s)
| | - Songjing Zhang
- School of Biological Sciences, Nanyang Technological University, Singapore
| | - Cheng-Gee Koh
- School of Biological Sciences, Nanyang Technological University, Singapore
| |
Collapse
|
86
|
Xu Y, Ji K, Wu M, Hao B, Yao KT, Xu Y. A miRNA-HERC4 pathway promotes breast tumorigenesis by inactivating tumor suppressor LATS1. Protein Cell 2019; 10:595-605. [PMID: 30710319 PMCID: PMC6626598 DOI: 10.1007/s13238-019-0607-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
The E3 ligase HERC4 is overexpressed in human breast cancer and its expression levels correlated with the prognosis of breast cancer patients. However, the roles of HERC4 in mammary tumorigenesis remain unclear. Here we demonstrate that the knockdown of HERC4 in human breast cancer cells dramatically suppressed their proliferation, survival, migration, and tumor growth in vivo, while the overexpression of HERC4 promoted their aggressive tumorigenic activities. HERC4 is a new E3 ligase for the tumor suppressor LATS1 and destabilizes LATS1 by promoting the ubiquitination of LATS1. miRNA-136-5p and miRNA-1285-5p, expression of which is decreased in human breast cancers and is inversely correlated with the prognosis of breast cancer patients, are directly involved in suppressing the expression of HERC4. In summary, we discover a miRNA-HERC4-LATS1 pathway that plays important roles in the pathogenesis of breast cancer and represents new therapeutic targets for human breast cancer.
Collapse
Affiliation(s)
- Youqin Xu
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China
| | - Kaiyuan Ji
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China
| | - Meng Wu
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China
| | - Bingtao Hao
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China.
| | - Kai-Tai Yao
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China.
| | - Yang Xu
- Guangdong Provincial Key laboratory of Tumor Immunotherapy, School of Basic Medical Sciences, Cancer Research Institute, Southern Medical University, Guangzhou, 510632, China. .,Division of Biological Sciences, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
87
|
Hypermethylated LATS2 gene with decreased expression in female breast cancer: A case control study from North India. Gene 2018; 676:156-163. [PMID: 30010037 DOI: 10.1016/j.gene.2018.07.033] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/25/2018] [Accepted: 07/11/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND LATS2, a presumed tumor suppressor gene located on chromosome 13q11-12 is involved in cell growth related activity like regulation of cell cycle at G1/S. The reduced expression of LATS2 has been reported in many tumors; including tumors of Breast, which is to the best of our knowledge has not been studied in north Indian female breast cancer population. OBJECTIVE Here, we looked upon the expression pattern and methylation status of the LATS2 gene in north Indian female breast cancer cases to further strengthen its role as a tumor suppressor gene and more importantly as a cancer biomarker. METHODS mRNA expression level was determined by real time PCR in 140 Breast cancer patients, Protein expression was studied by Immunohistochemistry and Promoter methylation was studied by Methylation specific PCR. All findings were correlated with clinicopathological features. RESULTS LATS2 mRNA expression was remarkably downregulated in 67.85% (95/140) cases. The expression of Large Associated Tumor Suppressor 2 at protein level was also absent in 67.85% (95/140) cases. The absence of LATS2 protein strongly correlated with promoter hypermethylation where 91 out of a total of 107 hyper methylated cases showed absence of protein (91/107, 85%). The absence of LATS2 protein was strongly significant with HER2 neu status (0.01), TNM staging (0.009) and Molecular subtype (0.024). CONCLUSION The decreased expression in breast cancer seems to be associated with hypermethylation of LATS2 promoter regions. Further LATS2 as a tumor suppressor can be recognized as a promising Biomarker in Breast cancer pathogenesis. Though, further studies, targeting larger sets of breast cancer population are required to establish LATS2 as a promising biomarker.
Collapse
|
88
|
Targeting the Hippo Pathway for Breast Cancer Therapy. Cancers (Basel) 2018; 10:cancers10110422. [PMID: 30400599 PMCID: PMC6266939 DOI: 10.3390/cancers10110422] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 10/31/2018] [Accepted: 11/02/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancer (BC) is one of the most prominent diseases in the world, and the treatments for BC have many limitations, such as resistance and a lack of reliable biomarkers. Currently the Hippo pathway is emerging as a tumor suppressor pathway with its four core components that regulate downstream transcriptional targets. In this review, we introduce the present targeted therapies of BC, and then discuss the roles of the Hippo pathway in BC. Finally, we summarize the evidence of the small molecule inhibitors that target the Hippo pathway, and then discuss the possibilities and future direction of the Hippo-targeted drugs for BC therapy.
Collapse
|
89
|
Furth N, Pateras IS, Rotkopf R, Vlachou V, Rivkin I, Schmitt I, Bakaev D, Gershoni A, Ainbinder E, Leshkowitz D, Johnson RL, Gorgoulis VG, Oren M, Aylon Y. LATS1 and LATS2 suppress breast cancer progression by maintaining cell identity and metabolic state. Life Sci Alliance 2018; 1:e201800171. [PMID: 30456386 PMCID: PMC6238411 DOI: 10.26508/lsa.201800171] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 10/13/2018] [Accepted: 10/15/2018] [Indexed: 02/04/2023] Open
Abstract
In luminal B tumors LATS2 depletion results in metabolic rewiring whereas LATS1 depletion promotes the expression of basal-like features. Deregulated activity of LArge Tumor Suppressor (LATS) tumor suppressors has broad implications on cellular and tissue homeostasis. We examined the consequences of down-regulation of either LATS1 or LATS2 in breast cancer. Consistent with their proposed tumor suppressive roles, expression of both paralogs was significantly down-regulated in human breast cancer, and loss of either paralog accelerated mammary tumorigenesis in mice. However, each paralog had a distinct impact on breast cancer. Thus, LATS2 depletion in luminal B tumors resulted in metabolic rewiring, with increased glycolysis and reduced peroxisome proliferator-activated receptor γ (PPARγ) signaling. Furthermore, pharmacological activation of PPARγ elicited LATS2-dependent death in luminal B-derived cells. In contrast, LATS1 depletion augmented cancer cell plasticity, skewing luminal B tumors towards increased expression of basal-like features, in association with increased resistance to hormone therapy. Hence, these two closely related paralogs play distinct roles in protection against breast cancer; tumors with reduced expression of either LATS1 or LATS2 may rewire signaling networks differently and thus respond differently to anticancer treatments.
Collapse
Affiliation(s)
- Noa Furth
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ioannis S Pateras
- Laboratory of Histology and Embryology Medical School, University of Athens, Athens, Greece
| | - Ron Rotkopf
- Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Vassiliki Vlachou
- Laboratory of Histology and Embryology Medical School, University of Athens, Athens, Greece
| | - Irina Rivkin
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ina Schmitt
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deborah Bakaev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Anat Gershoni
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Elena Ainbinder
- Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Dena Leshkowitz
- Department of Life Sciences Core Facilities, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Randy L Johnson
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vassilis G Gorgoulis
- Laboratory of Histology and Embryology Medical School, University of Athens, Athens, Greece.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Moshe Oren
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Yael Aylon
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
90
|
Ormonde JVS, Li Z, Stegen C, Madrenas J. TAOK3 Regulates Canonical TCR Signaling by Preventing Early SHP-1-Mediated Inactivation of LCK. THE JOURNAL OF IMMUNOLOGY 2018; 201:3431-3442. [PMID: 30373850 DOI: 10.4049/jimmunol.1800284] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 09/25/2018] [Indexed: 01/01/2023]
Abstract
Activation of LCK is required for canonical TCR signaling leading to T cell responses. LCK activation also initiates a negative feedback loop mediated by the phosphatase SHP-1 that turns off TCR signaling. In this article, we report that the thousand-and-one amino acid kinase 3 (TAOK3) is a key regulator of this feedback. TAOK3 is a serine/threonine kinase expressed in many different cell types including T cells. TAOK3-deficient human T cells had impaired LCK-dependent TCR signaling resulting in a defect in IL-2 response to canonical TCR signaling but not to bacterial superantigens, which use an LCK-independent pathway. This impairment was associated with enhanced interaction of LCK with SHP-1 after TCR engagement and rapid termination of TCR signals, a defect corrected by TAOK3 reconstitution. Thus, TAOK3 is a positive regulator of TCR signaling by preventing premature SHP-1-mediated inactivation of LCK. This mechanism may also regulate signaling by other Src family kinase-dependent receptors.
Collapse
Affiliation(s)
- João V S Ormonde
- Microbiome and Disease Tolerance Centre, Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; and
| | - Zhigang Li
- Microbiome and Disease Tolerance Centre, Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; and
| | - Camille Stegen
- Microbiome and Disease Tolerance Centre, Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; and
| | - Joaquín Madrenas
- Microbiome and Disease Tolerance Centre, Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 2B4, Canada; and .,Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA 90277
| |
Collapse
|
91
|
Wang S, Huo D, Ogundiran TO, Ojengbede O, Zheng W, Nathanson KL, Nemesure B, Ambs S, Olopade OI, Zheng Y. Genetic variation in the Hippo pathway and breast cancer risk in women of African ancestry. Mol Carcinog 2018; 57:1311-1318. [PMID: 29873413 PMCID: PMC6662580 DOI: 10.1002/mc.22845] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 05/18/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Gene expression changes within the Hippo pathway were found to be associated with large tumor size and metastasis in breast cancer. The combined effect of genetic variants in genes of this pathway may have a causal role in breast cancer development. We examined 7086 SNPs that were not highly correlated (r2 < 0.8) in 35 Hippo pathway genes using data from the genome-wide association study of breast cancer from the Root Consortium, which includes 3686 participants of African ancestry from Nigeria, United States of America, and Barbados: 1657 cases (403 estrogen receptor-positive [ER+], 374 ER-) and 2029 controls. Gene-level analyses were conducted using improved AdaJoint test for large-scale genetic association studies adjusting for age, study site and the first four eigenvectors from the principal component analysis. SNP-level analyses were conducted with logistic regression. The Hippo pathway was significantly associated with risk of ER+ breast cancer (pathway-level P = 0.019), with WWC1 (Padj = 0.04) being the leading gene. The pathway-level significance was lost without WWC1 (P = 0.12). rs147106204 in the WWC1 gene was the most statistically significant SNP after gene-level adjustment for multiple comparisons (OR = 0.53, 95%CI = 0.41-0.70, Padj = 0.025). We found evidence of an association between genetic variations in the Hippo pathway and ER+ breast cancer. Moreover, WWC1 was identified as the most important genetic susceptibility locus highlighting the importance of genetic epidemiology studies of breast cancer in understudied populations.
Collapse
Affiliation(s)
- Shengfeng Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing, China
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| | - Dezheng Huo
- Department of Public Health Sciences, University of Chicago, Chicago, Illinois, USA
| | | | - Oladosu Ojengbede
- Center for Population and Reproductive Health, College of Medicine, University of Ibadan, Ibadan, Nigeria
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Barbara Nemesure
- Department of Preventive Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Stefan Ambs
- Laboratory of Human Carcinogenesis, National Cancer Institute, Bethesda, Maryland, USA
| | - Olufunmilayo I. Olopade
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| | - Yonglan Zheng
- Center for Clinical Cancer Genetics and Global Health, Department of Medicine, University of Chicago, Chicago, Illinois; USA
| |
Collapse
|
92
|
Gill MK, Christova T, Zhang YY, Gregorieff A, Zhang L, Narimatsu M, Song S, Xiong S, Couzens AL, Tong J, Krieger JR, Moran MF, Zlotta AR, van der Kwast TH, Gingras AC, Sicheri F, Wrana JL, Attisano L. A feed forward loop enforces YAP/TAZ signaling during tumorigenesis. Nat Commun 2018; 9:3510. [PMID: 30158528 PMCID: PMC6115388 DOI: 10.1038/s41467-018-05939-2] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 07/30/2018] [Indexed: 12/13/2022] Open
Abstract
In most solid tumors, the Hippo pathway is inactivated through poorly understood mechanisms that result in the activation of the transcriptional regulators, YAP and TAZ. Here, we identify NUAK2 as a YAP/TAZ activator that directly inhibits LATS-mediated phosphorylation of YAP/TAZ and show that NUAK2 induction by YAP/TAZ and AP-1 is required for robust YAP/TAZ signaling. Pharmacological inhibition or loss of NUAK2 reduces the growth of cultured cancer cells and mammary tumors in mice. Moreover, in human patient samples, we show that NUAK2 expression is elevated in aggressive, high-grade bladder cancer and strongly correlates with a YAP/TAZ gene signature. These findings identify a positive feed forward loop in the Hippo pathway that establishes a key role for NUAK2 in enforcing the tumor-promoting activities of YAP/TAZ. Our results thus introduce a new opportunity for cancer therapeutics by delineating NUAK2 as a potential target for re-engaging the Hippo pathway. The Hippo pathway is frequently dysregulated in cancer. Here, the authors identify NUAK2 as negative regulator of the Hippo pathway from a siRNA kinome screen and show that NUAK2 promotes YAP/TAZ nuclear localisation while NUAK2 is a transcriptional target of YAP/TAZ, thus providing a feed forward loop to promote tumorigenesis.
Collapse
Affiliation(s)
- Mandeep K Gill
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Tania Christova
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Ying Y Zhang
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Alex Gregorieff
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Pathology, McGill University and Research Institute of the McGill University Health Center, Montreal, H4A 3J1, QC, Canada
| | - Liang Zhang
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada.,Department of Biomedical Sciences, College of Veterinary Medicine and Life Sciences, City University of Hong Kong, 999077, Hong Kong, China.,City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, 518057, China
| | - Masahiro Narimatsu
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Siyuan Song
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada
| | - Shawn Xiong
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Amber L Couzens
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Jiefei Tong
- Program in Cell Biology, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Jonathan R Krieger
- SPARC BioCentre, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Michael F Moran
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Program in Cell Biology, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada.,SPARC BioCentre, Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Alexandre R Zlotta
- Department of Surgery, Division of Urology, University of Toronto, Mount Sinai Hospital and University Health Network, Toronto, M5G 1X5, ON, Canada
| | - Theodorus H van der Kwast
- Department of Pathology, Toronto General Hospital, University Health Network, Toronto, ON, M5G 2C4, Canada
| | - Anne-Claude Gingras
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Frank Sicheri
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Jeffrey L Wrana
- Department of Molecular Genetics, University of Toronto, Toronto, ON, M5S 1A8, Canada.,Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Liliana Attisano
- Department of Biochemistry, University of Toronto, Toronto, ON, M5S 1A8, Canada. .,Donnelly Centre, University of Toronto, Toronto, ON, M5S 3E1, Canada.
| |
Collapse
|
93
|
Deng Q, Jiang G, Wu Y, Li J, Liang W, Chen L, Su Q, Li W, Du J, Wong CKC, Chen Z, Wang H. GPER/Hippo-YAP signal is involved in Bisphenol S induced migration of triple negative breast cancer (TNBC) cells. JOURNAL OF HAZARDOUS MATERIALS 2018; 355:1-9. [PMID: 29758456 DOI: 10.1016/j.jhazmat.2018.05.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 04/30/2018] [Accepted: 05/07/2018] [Indexed: 05/20/2023]
Abstract
Nowadays, risk factors of triple-negative breast cancer (TNBC) metastasis are not well identified. Our present study reveals that an industrial chemical, bisphenol S (BPS), can promote the migration, but not the proliferation, of TNBC cells in vitro. BPS activates YAP, a key effector of Hippo pathway, by inhibiting its phosphorylation, which promotes YAP nuclear accumulation and up-regulates its downstream genes such as CTGF and ANKRD1. Inhibition of YAP blocks the BPS-triggered cell migration and up-regulation of fibronectin (FN) and vimentin (Vim). BPS rapidly decreases the phosphorylation levels of LATS1 (Ser909) in TNBC cells, which regulates the activation and functions of YAP. Silencing LATS1/2 by siRNA increases BPS-induced dephosphorylation of YAP and extended the half-life of YAP protein. Inhibition of G protein-coupled estrogen receptor 1 (GPER) and its downstream PLCβ/PKC signals attenuate the effects of BPS-induced YAP dephosphorylation and CTGF up-regulation. Targeted inhibition of GPER/YAP inhibits BPS-induced migration of TNBC cells. Collectively, we reveal that GPER/Hippo-YAP signal is involved in BPS-induced migration of TNBC cells.
Collapse
Affiliation(s)
- Qianqian Deng
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yingmin Wu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Jiexin Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Weiting Liang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Likun Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Qiao Su
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Wuguo Li
- Animal Experiment Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chris K C Wong
- Department of Biology, Hong Kong Baptist University, Hong Kong, China
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China.
| | - Hongsheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
94
|
Jin D, Wu Y, Shao C, Gao Y, Wang D, Guo J. Norcantharidin reverses cisplatin resistance and inhibits the epithelial mesenchymal transition of human non‑small lung cancer cells by regulating the YAP pathway. Oncol Rep 2018; 40:609-620. [PMID: 29901163 PMCID: PMC6072289 DOI: 10.3892/or.2018.6486] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 05/25/2018] [Indexed: 02/06/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for >80% of all lung cancer cases, which are the leading cause of cancer-related mortality worldwide. The clinical efficacy of available therapies for NSCLC is often limited due to the development of resistance to anticancer drugs, particularly to cisplatin (DDP). Norcantharidin (NCTD) is a traditional Chinese medicine used in the treatment of many types of cancer, to which patients do not develop resistance. The aim of the present study was to examine the potential synergistic effects of NCTD and DPP on the viability of the the DDP-resistant NSCLC cell line, A549/DDP. We further explored the potential underlying mechanisms by examining the expression of the oncogene, Yes-associated protein 1 (YAP), whose activation was recently found to be associated with drug resistance. We further examined a series of human lung cancer cell lines and tissues from patients with lung cancer, which revealed that YAP activation contributed to lung cancer initiation, progression and metastasis, and was associated with a poor prognosis, and confering resistance against targeted therapies. Moreover, YAP expression was evaluated in the A549/DDP cells treated with NCTD, DDP, or both drugs. The combined treatment significantly sensitized the A549/DDP cells to DDP-induced growth inhibition by reducing YAP promoter activity (based on transcriptional expression) and the expression of its target genes, connective tissue growth factor (CTGF) and cysteine rich angiogenic inducer 61 (CYR61). Furthermore, compared to the individual treatments, combined treatment increased cell apoptosis and senescence, and decreased epithelial-to-mesenchymal transition and the cell migratory and invasive ability. On the whole, our data indicate that the application of NCTD with reverses DDP resistance and thus, this combined treatment may have promising prospects for use in improving the outcome of patients with NSCLC.
Collapse
Affiliation(s)
- Dan Jin
- Department of Pain Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yan Wu
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Cuijie Shao
- Department of Pain Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Yong Gao
- Department of Pain Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Deqiang Wang
- Department of Pain Medicine, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| | - Jiwei Guo
- Cancer Research Institute, Binzhou Medical University Hospital, Binzhou, Shandong 256603, P.R. China
| |
Collapse
|
95
|
Philippe C, Pinson B, Dompierre J, Pantesco V, Viollet B, Daignan-Fornier B, Moenner M. AICAR Antiproliferative Properties Involve the AMPK-Independent Activation of the Tumor Suppressors LATS 1 and 2. Neoplasia 2018; 20:555-562. [PMID: 29730476 PMCID: PMC5994775 DOI: 10.1016/j.neo.2018.03.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/14/2018] [Accepted: 03/19/2018] [Indexed: 12/11/2022] Open
Abstract
AICAR (Acadesine) is a pharmacological precursor of purine nucleotide biosynthesis with anti-tumoral properties. Although recognized as an AMP mimetic activator of the protein kinase AMPK, the AICAR monophosphate derivative ZMP was also shown to mediate AMPK-independent effects. In order to unveil these AMPK-independent functions, we performed a transcriptomic analysis in AMPKα1/α2 double knockout murine embryonic cells. Kinetic analysis of the cellular response to AICAR revealed the up-regulation of the large tumor suppressor kinases (Lats) 1 and 2 transcripts, followed by the repression of numerous genes downstream of the transcriptional regulators Yap1 and Taz. This transcriptional signature, together with the observation of increased levels in phosphorylation of Lats1 and Yap1 proteins, suggested that the Hippo signaling pathway was activated by AICAR. This effect was observed in both fibroblasts and epithelial cells. Knockdown of Lats1/2 prevented the cytoplasmic delocalization of Yap1/Taz proteins in response to AICAR and conferred a higher resistance to the drug. These results indicate that activation of the most downstream steps of the Hippo cascade participates to the antiproliferative effects of AICAR.
Collapse
Affiliation(s)
- Chloé Philippe
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | - Benoît Pinson
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | - Jim Dompierre
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France
| | | | - Benoît Viollet
- INSERM U1016, Institut Cochin, Paris, France; CNRS (UMR 8104), Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Bertrand Daignan-Fornier
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France.
| | - Michel Moenner
- Université de Bordeaux, IBGC UMR 5095, Bordeaux, France; Centre National de la Recherche Scientifique, IBGC UMR 5095, Bordeaux, France.
| |
Collapse
|
96
|
Abstract
The Hippo pathway is a novel and highly conserved mammalian signaling pathway. Mutations and altered expression of core Hippo pathway components promote the migration, invasion, malignancy, and chemotherapy resistance of breast cancer cells. In cancer metastasis, tumor cells must detach from the primary tumor, invade surrounding tissue, and enter and survive in a foreign microenvironment. The metastatic potential of breast cancer is closely related to individual patient genetic profile. Nevertheless, the exact molecular mechanism that regulates the Hippo pathway in breast cancer metastasis is yet to be fully elucidated. This article discusses the function and regulation of the Hippo pathway, with focus given to its role in the context of breast cancer metastasis.
Collapse
Affiliation(s)
- Changran Wei
- Department of Breast Surgery, Affiliated Hospital of Taishan Medical University, Tai’an, Shandong Province, China
| | - Ying Wang
- Rehabilitation Medicine, Affiliated Hospital of Taishan Medical University, Tai’an, Shandong Province, China
| | - Xiangqi Li
- Department of Breast Surgery, Affiliated Hospital of Taishan Medical University, Tai’an, Shandong Province, China
| |
Collapse
|
97
|
Zheng H, Ke X, Li D, Wang Q, Wang J, Liu X, Deng M, Deng X, Xue Y, Zhu Y, Wang Q. NEDD4 promotes cell growth and motility in hepatocellular carcinoma. Cell Cycle 2018; 17:728-738. [PMID: 29480061 DOI: 10.1080/15384101.2018.1440879] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common causes of cancer-related death worldwide. In China, the situation is even worse as cancer incidence and mortality continue to increase rapidly. Although tremendous progress has been made toward HCC treatments, the benefits for liver cancer patients are still limited. Therefore, it is necessary to identify and develop novel therapeutic methods. Neuronally expressed developmentally downregulated 4 (NEDD4), an E3 ubiquitin ligase, plays a critical role in the development and progression of various types of human cancers. In our study, NEDD4 acts as an oncoprotein in both QGY7703 and SMMC7721 liver cancer cell lines. We found that depletion of NEDD4 by siRNA transfection led to inhibition of cell growth, invasion and migration, and promotion of apoptosis. In contrast, overexpression of NEDD4 via plasmid transfection resulted in facilitated cell proliferation, invasion and migration, and decreased apoptosis. Importantly, we observed that tumor suppressor LATS1, also a core component of Hippo pathway, was negatively regulated by NEDD4 in liver cancer cells. Our findings suggested that NEDD4 may be involved in the HCC progression via regulating LATS1 associated signaling pathway. Therefore, targeting NEDD4-LATS1 signaling could be a potential therapeutic option for HCC treatment.
Collapse
Affiliation(s)
- Hailun Zheng
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Xiquan Ke
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Dapeng Li
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Qiangwu Wang
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Jianchao Wang
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Xiaoyang Liu
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Min Deng
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Xiaojing Deng
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Yongju Xue
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Yu Zhu
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| | - Qizhi Wang
- a Department of Gastroenterology , The First Affiliated Hospital of Bengbu Medical College , Bengbu , Anhui 233004 , P.R. China
| |
Collapse
|
98
|
Wu J, Zhao Z, Zhang H, Kong F, Jiang H, Huang K, Zheng H. LATS1 inhibits metastasis and epithelial-mesenchymal transition in head and neck squamous cell carcinoma. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:2053-2063. [PMID: 31938312 PMCID: PMC6958224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 02/22/2018] [Indexed: 06/10/2023]
Abstract
LATS1 is a serine/threonine kinase of the Hippo signaling pathway that phosphorylates and inactivates transcriptional co-activators YAP1 and WWTR1. To investigate roles of LATS1 expression in head and neck squamous cell carcinomas (HNSCCs), we transfected LATS1-expressing plasmid into B88 cells and examined the phenotypes and their relevant molecules. LATS1 expression was analyzed using immunohistochemistry on tissue microarray, Oncomine, and TCGA databases. LATS1 overexpression was found to suppress growth, migration and invasion, and induce apoptosis, G2 arrest, and mesenchymal to epithelial transition (MET) (P < 0.05). Both increased expression of P21, Bax, and E-cadherin and decreased expression of Cyclin B1, D1, Bcl-2, and MMPs. Twist and N-cadherin were detected in B88 transfectants, in comparison to mock and control by Western blot. Nuclear LATS1 expression was weaker in primary cancers than in normal squamous tissue and dysplasia (P < 0.05) but versa for cytoplasmic counterpart (P < 0.05). Cytoplasmic LATS1 expression was positively correlated with lymph node metastasis (P < 0.05). Survival analysis showed that differentiation degree was an independent factor of long overall and relapse-free survival of HNSCC patients (P < 0.05). According to bioinformatics analysis, we found upregulated LATS1 mRNA expression in HNSCCs (P < 0.05). Cox proportional hazards model indicated that perineural invasion and distant metastasis were independent prognostic factors for overall survival of HNSCC (P < 0.05). These findings suggest nucleocytoplasmic translocation of LATS1 protein and upregulated expression of LATS1 mRNA during tumorigenesis of HNSCC. LATS1 mRNA overexpression may reverse aggressive phenotypes of HNSCC cells, as a gene therapy target.
Collapse
Affiliation(s)
- Jicheng Wu
- Department of Experimental Oncology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Zhijuan Zhao
- Department of Pathology, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou, China
| | - Huiling Zhang
- Department of Experimental Oncology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Fanshuang Kong
- Department of Experimental Oncology, Shengjing Hospital of China Medical UniversityShenyang, China
| | - Huamao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical UniversityJinzhou, China
| | - Keqiang Huang
- Department of Stomatology, The Second Affiliated Hospital of Jinzhou Medical UniversityJinzhou, China
| | - Huachuan Zheng
- Department of Experimental Oncology, Shengjing Hospital of China Medical UniversityShenyang, China
| |
Collapse
|
99
|
Wang T, Qin ZY, Wen LZ, Guo Y, Liu Q, Lei ZJ, Pan W, Liu KJ, Wang XW, Lai SJ, Sun WJ, Wei YL, Liu L, Guo L, Chen YQ, Wang J, Xiao HL, Bian XW, Chen DF, Wang B. Epigenetic restriction of Hippo signaling by MORC2 underlies stemness of hepatocellular carcinoma cells. Cell Death Differ 2018; 25:2086-2100. [PMID: 29555977 DOI: 10.1038/s41418-018-0095-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 02/14/2018] [Accepted: 02/22/2018] [Indexed: 12/12/2022] Open
Abstract
The evolutionarily conserved Hippo signaling pathway is a key regulator of stem cell self-renewal, differentiation, and organ size. While alterations in Hippo signaling are causally linked to uncontrolled cell growth and a broad range of malignancies, genetic mutations in the Hippo pathway are uncommon and it is unclear how the tumor suppressor function of the Hippo pathway is disrupted in human cancers. Here, we report a novel epigenetic mechanism of Hippo inactivation in the context of hepatocellular carcinoma (HCC). We identify a member of the microrchidia (MORC) protein family, MORC2, as an inhibitor of the Hippo pathway by controlling upstream Hippo regulators, neurofibromatosis 2 (NF2) and kidney and brain protein (KIBRA). Mechanistically, MORC2 forms a complex with DNA methyltransferase 3A (DNMT3A) at the promoters of NF2 and KIBRA, leading to their DNA hyper-methylation and transcriptional repression. As a result, NF2 and KIBRA are crucial targets of MORC2 to regulate confluence-induced activation of Hippo signaling and contact inhibition of cell growth under both physiological and pathological conditions. The MORC2-NF2/KIBRA axis is critical for maintaining self-renewal, sorafenib resistance, and oncogenicity of HCC cells in vitro and in nude mice. Furthermore, MORC2 expression is elevated in HCC tissues, associated with stem-like properties of cancer cells, and disease progression in patients. Collectively, MORC2 promotes cancer stemness and tumorigenesis by facilitating DNA methylation-dependent silencing of Hippo signaling and could be a potential molecular target for cancer therapeutics.
Collapse
Affiliation(s)
- Tao Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Zhong-Yi Qin
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Liang-Zhi Wen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yan Guo
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Qin Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Zeng-Jie Lei
- Department of Medical Oncology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, 210002, Jiangsu Province, China
| | - Wei Pan
- Department of Medical Genetics, Second Military Medical University (Navy Medical University), 200433, Shanghai, China
| | - Kai-Jun Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Xing-Wei Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Shu-Jie Lai
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Wen-Jing Sun
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yan-Ling Wei
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Lei Liu
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Ling Guo
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Yu-Qin Chen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Jun Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Hua-Liang Xiao
- Department of Pathology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China
| | - Xiu-Wu Bian
- Institute of Pathology and Southwest Cancer Center, Key Laboratory of Tumor Immunopathology of Ministry of Education of China, Southwest Hospital, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Dong-Feng Chen
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China.
| | - Bin Wang
- Department of Gastroenterology, Institute of Surgery Research, Daping Hospital, Third Military Medical University (Army Medical University), 400042, Chongqing, China.
| |
Collapse
|
100
|
Lin KC, Park HW, Guan KL. Deregulation and Therapeutic Potential of the Hippo Pathway in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050202] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Kimberly C. Lin
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| | - Hyun Woo Park
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California, San Diego, La Jolla, California 92093, USA
| |
Collapse
|