1
|
Wang H, Li J, Yu K, Lu Y, Ma M, Li Y. The cellular localization and oncogenic or tumor suppressive effects of angiomiotin-like protein 2 in tumor and normal cells. IUBMB Life 2024; 76:764-779. [PMID: 38717123 DOI: 10.1002/iub.2830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/10/2024] [Indexed: 10/19/2024]
Abstract
Angiomiotin (AMOT) family comprises three members: AMOT, AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). AMOTL2 is widely expressed in endothelial cells, epithelial cells, and various cancer cells. Specifically, AMOTL2 predominantly localizes in the cytoplasm and nucleus in human normal cells, whereas associates with cell-cell junctions and actin cytoskeleton in non-human cells, and locates at cell junctions or within the recycling endosomes in cancer cells. AMOTL2 is implicated in regulation of tube formation, cell polarity, and shape, although the specific impact on tumorigenesis remains to be conclusively determined. It has been shown that AMOTL2 enhances tumor growth and metastasis in pancreatic, breast, and colon cancer, however inhibits cell proliferation and migration in lung, hepatocellular cancer, and glioblastoma. In addition to its role in cell shape and cytoskeletal dynamics through co-localization with F-actin, AMOTL2 modulates the transcription of Yes-associated protein (YAP) by binding to it, thereby affecting its phosphorylation and cellular sequestration. Furthermore, the stability and cellular localization of AMOTL2, influenced by its phosphorylation and ubiquitination mediated by specific proteins, affects its cellular function. Additionally, we observe that AMOTL2 is predominantly downregulated in some tumors, but significantly elevated in colorectal adenocarcinoma (COAD). Moreover, overall analysis, GSEA and ROC curve analysis indicate that AMOTL2 exerts as an oncogenic protein in COAD by modulating Wnt pathway, participating in synthesis of collagen formation, and interacting with extracellular matrix receptor. In addition, AMOTL2 potentially regulates the distribution of immune cells infiltration in COAD. In summary, AMOTL2 probably functions as an oncogene in COAD. Consequently, further in-depth mechanistic research is required to elucidate the precise roles of AMOTL2 in various cancers.
Collapse
Affiliation(s)
- Huizhen Wang
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jing Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Kexun Yu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yida Lu
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mengdi Ma
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yongxiang Li
- Department of General Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Wang Y, Yu FX. Angiomotin family proteins in the Hippo signaling pathway. Bioessays 2024; 46:e2400076. [PMID: 38760875 DOI: 10.1002/bies.202400076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 05/03/2024] [Accepted: 05/08/2024] [Indexed: 05/19/2024]
Abstract
The Motin family proteins (Motins) are a class of scaffolding proteins consisting of Angiomotin (AMOT), AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). Motins play a pivotal role in angiogenesis, tumorigenesis, and neurogenesis by modulating multiple cellular signaling pathways. Recent findings indicate that Motins are components of the Hippo pathway, a signaling cascade involved in development and cancer. This review discusses how Motins are integrated into the Hippo signaling network, as either upstream regulators or downstream effectors, to modulate cell proliferation and migration. The repression of YAP/TAZ by Motins contributes to growth inhibition, whereas subcellular localization of Motins and their interactions with actin fibers are critical in regulating cell migration. The net effect of Motins on cell proliferation and migration may contribute to their diverse biological functions.
Collapse
Affiliation(s)
- Yu Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fa-Xing Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, International Co-laboratory of Medical Epigenetics and Metabolism, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Wang H, Ye M, Jin X. Role of angiomotin family members in human diseases (Review). Exp Ther Med 2024; 27:258. [PMID: 38766307 PMCID: PMC11099588 DOI: 10.3892/etm.2024.12546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 10/23/2023] [Indexed: 05/22/2024] Open
Abstract
Angiomotin (Amot) family members, including Amot, Amot-like protein 1 (Amotl1) and Amot-like protein 2 (Amotl2), have been found to interact with angiostatins. In addition, Amot family members are involved in various physiological and pathological functions such as embryonic development, angiogenesis and tumorigenesis. Some studies have also demonstrated its regulation in signaling pathways such as the Hippo signaling pathway, AMPK signaling pathway and mTOR signaling pathways. Amot family members play an important role in neural stem cell differentiation, dendritic formation and synaptic maturation. In addition, an increasing number of studies have focused on their function in promoting and/or suppressing cancer, but the underlying mechanisms remain to be elucidated. The present review integrated relevant studies on upstream regulation and downstream signals of Amot family members, as well as the latest progress in physiological and pathological functions and clinical applications, hoping to offer important ideas for further research.
Collapse
Affiliation(s)
- Haoyun Wang
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, P.R. China
- Department of Radiotherapy, The First Hospital of Ningbo University, Ningbo, Zhejiang 315010, P.R. China
| |
Collapse
|
4
|
Flamier A, Bisht P, Richards A, Tomasello DL, Jaenisch R. Human iPS cell-derived sensory neurons can be infected by SARS-CoV-2. iScience 2023; 26:107690. [PMID: 37680484 PMCID: PMC10480666 DOI: 10.1016/j.isci.2023.107690] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/10/2023] [Accepted: 08/17/2023] [Indexed: 09/09/2023] Open
Abstract
COVID-19 has impacted billions of people since 2019 and unfolded a major healthcare crisis. With an increasing number of deaths and the emergence of more transmissible variants, it is crucial to better understand the biology of the disease-causing virus, the SARS-CoV-2. Peripheral neuropathies appeared as a specific COVID-19 symptom occurring at later stages of the disease. In order to understand the impact of SARS-CoV-2 on the peripheral nervous system, we generated human sensory neurons from induced pluripotent stem cells that we infected with the SARS-CoV-2 strain WA1/2020 and the variants delta and omicron. Using single-cell RNA sequencing, we found that human sensory neurons can be infected by SARS-CoV-2 but are unable to produce infectious viruses. Our data indicate that sensory neurons can be infected by the original WA1/2020 strain of SARS-CoV-2 as well as the delta and omicron variants, yet infectability differs between the original strain and the variants.
Collapse
Affiliation(s)
- Anthony Flamier
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Punam Bisht
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | | | | | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
5
|
Sanchez MP, Escouflaire C, Baur A, Bottin F, Hozé C, Boussaha M, Fritz S, Capitan A, Boichard D. X-linked genes influence various complex traits in dairy cattle. BMC Genomics 2023; 24:338. [PMID: 37337145 DOI: 10.1186/s12864-023-09438-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND The search for quantitative trait loci (QTL) affecting traits of interest in mammals is frequently limited to autosomes, with the X chromosome excluded because of its hemizygosity in males. This study aimed to assess the importance of the X chromosome in the genetic determinism of 11 complex traits related to milk production, milk composition, mastitis resistance, fertility, and stature in 236,496 cows from three major French dairy breeds (Holstein, Montbéliarde, and Normande) and three breeds of regional importance (Abondance, Tarentaise, and Vosgienne). RESULTS Estimates of the proportions of heritability due to autosomes and X chromosome (h²X) were consistent among breeds. On average over the 11 traits, h²X=0.008 and the X chromosome explained ~ 3.5% of total genetic variance. GWAS was performed within-breed at the sequence level (~ 200,000 genetic variants) and then combined in a meta-analysis. QTL were identified for most breeds and traits analyzed, with the exception of Tarentaise and Vosgienne and two fertility traits. Overall, 3, 74, 59, and 71 QTL were identified in Abondance, Montbéliarde, Normande, and Holstein, respectively, and most were associated with the most-heritable traits (milk traits and stature). The meta-analyses, which assessed a total of 157 QTL for the different traits, highlighted new QTL and refined the positions of some QTL found in the within-breed analyses. Altogether, our analyses identified a number of functional candidate genes, with the most notable being GPC3, MBNL3, HS6ST2, and DMD for dairy traits; TMEM164, ACSL4, ENOX2, HTR2C, AMOT, and IRAK1 for udder health; MAMLD1 and COL4A6 for fertility; and NRK, ESX1, GPR50, GPC3, and GPC4 for stature. CONCLUSIONS This study demonstrates the importance of the X chromosome in the genetic determinism of complex traits in dairy cattle and highlights new functional candidate genes and variants for these traits. These results could potentially be extended to other species as many X-linked genes are shared among mammals.
Collapse
Affiliation(s)
- Marie-Pierre Sanchez
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, 78350, France.
| | | | | | - Fiona Bottin
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, 78350, France
| | | | - Mekki Boussaha
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, 78350, France
| | | | - Aurélien Capitan
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, 78350, France
| | - Didier Boichard
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, 78350, France
| |
Collapse
|
6
|
Amirifar P, Kissil J. The role of Motin family proteins in tumorigenesis-an update. Oncogene 2023; 42:1265-1271. [PMID: 36973516 DOI: 10.1038/s41388-023-02677-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
The Motin protein family consists of three members: AMOT (p80 and p130 isoforms), AMOT-like protein 1 (AMOTL1), and AMOT-like protein 2 (AMOTL2). The family members play an important role in processes such as cell proliferation, migration, angiogenesis, tight junction formation, and cell polarity. These functions are mediated through the involvement of the Motins in the regulation of different signal transduction pathways, including those regulated by small G-proteins and the Hippo-YAP pathway. One of the more characterized aspects of Motin family function is their role in regulating signaling through the Hippo-YAP pathway, and while some studies suggest a YAP-inhibitory function other studies indicate the Motins are required for YAP activity. This duality is also reflected in previous reports, often contradictory, that suggest the Motin proteins can function as oncogenes or tumor suppressors in tumorigenesis. In this review we summarize recent findings and integrate that with the existing work describing the multifunctional role of the Motins in different cancers. The emerging picture suggests that the Motin protein function is cell-type and context dependent and that further investigation in relevant cell types and whole organism models is required for the elucidation of the function of this protein family.
Collapse
Affiliation(s)
- Parisa Amirifar
- Department of Molecular Oncology, Cancer Biology Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Joseph Kissil
- Department of Molecular Oncology, Cancer Biology Evolution Program, H. Lee Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
7
|
Flamier A, Bisht P, Richards A, Tomasello D, Jaenisch R. Human iPS cell-derived sensory neurons can be infected by SARS-CoV-2 strain WA1/2020 as well as variants delta and omicron. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.10.523422. [PMID: 36711852 PMCID: PMC9882040 DOI: 10.1101/2023.01.10.523422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
COVID-19 has impacted billions of people in the world since 2019 and unfolded a major healthcare crisis. With an increasing number of deaths and the emergence of more transmissible variants, it is crucial to better understand the biology of the disease-causing virus, the SARS-CoV-2. Peripheral neuropathies appeared as a specific COVID-19 symptom occurring at later stages of the disease. In order to understand the impact of SARS-CoV-2 on the peripheral nervous system, we generated human sensory neurons from induced pluripotent stem cells that we infected with the SARS-CoV-2 strain WA1/2020 and the variants delta and omicron. Using single cell RNA sequencing, we found that human sensory neurons can be infected by SARS-CoV-2 but are unable to produce new viruses. Our data suggests that sensory neurons can be infected by the original WA1/2020 strain of SARS-CoV-2 as well as the delta and omicron variants.
Collapse
|
8
|
Hwang D, Kim M, Kim S, Kwon MR, Kang YS, Kim D, Kang HC, Lim DS. AMOTL2 mono-ubiquitination by WWP1 promotes contact inhibition by facilitating LATS activation. Life Sci Alliance 2021; 4:4/10/e202000953. [PMID: 34404733 PMCID: PMC8372784 DOI: 10.26508/lsa.202000953] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 08/06/2021] [Accepted: 08/06/2021] [Indexed: 11/24/2022] Open
Abstract
This work reveals a novel function of WWP1 E3 ligase in the mono-ubiquitination of AMOTL2, which enables the binding and activation of LATS kinases upon contact inhibition. Contact inhibition is a key cellular phenomenon that prevents cells from hyper-proliferating upon reaching confluence. Although not fully characterized, a critical driver of this process is the Hippo signaling pathway, whose downstream effector yes-associated protein plays pivotal roles in cell growth and differentiation. Here, we provide evidence that the E3 ligase WWP1 (WW-domain containing protein 1) mono-ubiquitinates AMOTL2 (angiomotin-like 2) at K347 and K408. Mono-ubiquitinated AMOTL2, in turn, interacts with the kinase LATS2, which facilitates recruitment of the upstream Hippo pathway component SAV1 and ultimately promotes yes-associated protein phosphorylation and subsequent cytoplasmic sequestration and/or degradation. Furthermore, contact inhibition induced by high cell density promoted the localization and stabilization of WWP1 at cell junctions, where it interacted with Crumbs polarity proteins. Notably, the Crumbs complex was functionally important for AMOTL2 mono-ubiquitination and LATS activation under high cell density conditions. These findings delineate a functionally important molecular mechanism in which AMOTL2 mono-ubiquitination by WWP1 at cell junctions and LATS activation are tightly coupled to upstream cell density cues.
Collapse
Affiliation(s)
- Daehee Hwang
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Miju Kim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Soyeon Kim
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Mi Ra Kwon
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Ye-Seul Kang
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Dahyun Kim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| | - Ho-Chul Kang
- Genomic Instability Research Center, Ajou University School of Medicine, Suwon, Korea
| | - Dae-Sik Lim
- National Creative Research Center for Cell Plasticity, Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, Korea
| |
Collapse
|
9
|
Li D, Shen Y, Ren H, Wang L, Yang J, Wang Y. Angiomotin-p130 inhibits vasculogenic mimicry formation of small cell lung cancer independently of Smad2/3 signal pathway. J Bioenerg Biomembr 2021; 53:295-305. [PMID: 33712992 DOI: 10.1007/s10863-021-09891-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 03/05/2021] [Indexed: 01/20/2023]
Abstract
Lung cancer, the most concerning malignancy worldwide and one of the leading causes of cancer-related deaths. Growing evidence indicates that Angiomotin (Amot)-p130 plays an important role in types of cancer, including breast cancer and gastric cancer. Moreover, evidence suggested that the low Amot-p130 expression correlates with the poor prognosis of lung cancer patients, however, the role and mechanism of Amot-p130 in lung cancer is still unclear. In this study, we showed that Amot-p130 expression was reduced in lung cancer tissues, compared with the adjacent para-carcinoma tissues. In addition, we observed that the reduced expression of Amot-p130 was associated with vasculogenic mimicry (VM) channels formation in lung cancer tissues. Amot-p130 expression was differently expression in lung cancer cell line H446, H1688 and H2227 compared with the normal human lung cells HFL1. To clarify the role of Amot-p130 in lung cancer, we constructed the Amot-p130 expressing H446 cells and Amot-p130 silencing H1299 cells. We confirmed that Amot-p130 overexpression inhibited the migration and invasion of lung cancer cells, whereas its silence promoted cell migration and invasion. Interestingly, we also found that Amot-p130 overexpression suppressed VM tube formation in H446 cells, while its knockdown promoted VM tube formation in H2227 cells. Further studies suggested that Amot-p130 plays roles in M tube formation of lung cancer cell V are independent on smad2/3 signaling pathway. Finally, inoculation of Amot-p130 expressing H446 cells and Amot-p130 silencing H1299 cells into nude mice suppressed tumor growth, when compared with the control group. Based on these results, Amot-p130 serves as a possible diagnostic and therapeutic target in lung cancer patients, and may be an effective mediator of VM formation in lung cancer.
Collapse
Affiliation(s)
- Dan Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yanwei Shen
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Hui Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Li Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jin Yang
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Yuan Wang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
| |
Collapse
|
10
|
Sang T, Yang J, Liu J, Han Y, Li Y, Zhou X, Wang X. AMOT suppresses tumor progression via regulating DNA damage response signaling in diffuse large B-cell lymphoma. Cancer Gene Ther 2021; 28:1125-1135. [PMID: 33414519 DOI: 10.1038/s41417-020-00258-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 10/22/2020] [Accepted: 11/04/2020] [Indexed: 12/29/2022]
Abstract
Angiomotin (AMOT) is a membrane protein that is aberrantly expressed in a variety of solid tumors. Accumulating evidence support that AMOT is involved in the pathological processes of tumor proliferation, apoptosis, and invasion. However, the potential role of AMOT in the pathogenesis of diffuse large B-cell lymphoma (DLBCL) remains elusive. In the present study, we investigated the expression level and biological function of AMOT in DLBCL. AMOT expression was significantly reduced in DLBCL biopsy section, and low AMOT expression was associated with poor clinical prognosis. Overexpression of AMOT by lentivirus in human DLBCL cells induced cell viability inhibition concomitant with an increased percentage of cells in G1 phase and decreased percentage in S phase. Moreover, AMOT upregulation increased the sensitivity of DLBCL cells to doxorubicin. Furthermore, overexpression of AMOT led to reduced activation of key kinases for the DNA damage response (DDR). The above results indicated that AMOT acts as a tumor suppressor via inhibition of the DDR, thus reducing the viability while increasing the chemosensitivity in DLBCL. In summary, AMOT may be a novel potential target for DLBCL therapeutic intervention.
Collapse
Affiliation(s)
- Tan Sang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,School of Medicine, Shandong University, Jinan, Shandong, 250012, China.,Department of Hematology, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Juan Yang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Jiarui Liu
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Yang Han
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China.,School of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China. .,School of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China. .,National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250021, China. .,School of Medicine, Shandong University, Jinan, Shandong, 250012, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, Shandong, 250021, China. .,National Clinical Research Center for Hematologic Diseases, Jinan, Shandong, 250021, China.
| |
Collapse
|
11
|
miR-205 in Breast Cancer: State of the Art. Int J Mol Sci 2020; 22:ijms22010027. [PMID: 33375067 PMCID: PMC7792793 DOI: 10.3390/ijms22010027] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/11/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
Despite its controversial roles in different cancer types, miR-205 has been mainly described as an oncosuppressive microRNA (miRNA), with some contrasting results, in breast cancer. The role of miR-205 in the occurrence or progression of breast cancer has been extensively studied since the first evidence of its aberrant expression in tumor tissues versus normal counterparts. To date, it is known that the expression of miR-205 in the different subtypes of breast cancer is decreasing from the less aggressive subtype, estrogen receptor/progesterone receptor positive breast cancer, to the more aggressive, triple negative breast cancer, influencing metastasis capability, response to therapy and patient survival. In this review, we summarize the most important discoveries that have highlighted the functional role of this miRNA in breast cancer initiation and progression, in stemness maintenance, in the tumor microenvironment, its potential role as a biomarker and its relevance in normal breast physiology—the still open questions. Finally, emerging evidence reveals the role of some lncRNAs in breast cancer progression as sponges of miR-205. Here, we also reviewed the studies in this field.
Collapse
|
12
|
Using Phosphatidylinositol Phosphorylation as Markers for Hyperglycemic Related Breast Cancer. Int J Mol Sci 2020; 21:ijms21072320. [PMID: 32230859 PMCID: PMC7177416 DOI: 10.3390/ijms21072320] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 02/06/2023] Open
Abstract
Studies have suggested that type 2 diabetes (T2D) is associated with a higher incidence of breast cancer and related mortality rates. T2D postmenopausal women have an ~20% increased chance of developing breast cancer, and women with T2D and breast cancer have a 50% increase in mortality compared to breast cancer patients without diabetes. This correlation has been attributed to the general activation of insulin receptor signaling, glucose metabolism, phosphatidylinositol (PI) kinases, and growth pathways. Furthermore, the presence of breast cancer specific PI kinase and/or phosphatase mutations enhance metastatic breast cancer phenotypes. We hypothesized that each of the breast cancer subtypes may have characteristic PI phosphorylation profiles that are changed in T2D conditions. Therefore, we sought to characterize the PI phosphorylation when equilibrated in normal glycemic versus hyperglycemic serum conditions. Our results suggest that hyperglycemia leads to: 1) A reduction in PI3P and PIP3, with increased PI4P that is later converted to PI(3,4)P2 at the cell surface in hormone receptor positive breast cancer; 2) a reduction in PI3P and PI4P with increased PIP3 surface expression in human epidermal growth factor receptor 2-positive (HER2+) breast cancer; and 3) an increase in di- and tri-phosphorylated PIs due to turnover of PI3P in triple negative breast cancer. This study begins to describe some of the crucial changes in PIs that play a role in T2D related breast cancer incidence and metastasis.
Collapse
|
13
|
MiR-205 Dysregulations in Breast Cancer: The Complexity and Opportunities. Noncoding RNA 2019; 5:ncrna5040053. [PMID: 31752366 PMCID: PMC6958506 DOI: 10.3390/ncrna5040053] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 11/12/2019] [Accepted: 11/13/2019] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are endogenous non-coding small RNAs that downregulate target gene expression by imperfect base-pairing with the 3' untranslated regions (3'UTRs) of target gene mRNAs. MiRNAs play important roles in regulating cancer cell proliferation, stemness maintenance, tumorigenesis, cancer metastasis, and cancer therapeutic resistance. While studies have shown that dysregulation of miRNA-205-5p (miR-205) expression is controversial in different types of human cancers, it is generally observed that miR-205-5p expression level is downregulated in breast cancer and that miR-205-5p exhibits a tumor suppressive function in breast cancer. This review focuses on the role of miR-205-5p dysregulation in different subtypes of breast cancer, with discussions on the effects of miR-205-5p on breast cancer cell proliferation, epithelial-mesenchymal transition (EMT), metastasis, stemness and therapy-resistance, as well as genetic and epigenetic mechanisms that regulate miR-205-5p expression in breast cancer. In addition, the potential diagnostic and therapeutic value of miR-205-5p in breast cancer is also discussed. A comprehensive list of validated miR-205-5p direct targets is presented. It is concluded that miR-205-5p is an important tumor suppressive miRNA capable of inhibiting the growth and metastasis of human breast cancer, especially triple negative breast cancer. MiR-205-5p might be both a potential diagnostic biomarker and a therapeutic target for metastatic breast cancer.
Collapse
|
14
|
Zhang H, Sun J, Ju W, Li B, Lou Y, Zhang G, Liang G, Luo X. Apatinib suppresses breast cancer cells proliferation and invasion via angiomotin inhibition. Am J Transl Res 2019; 11:4460-4469. [PMID: 31396349 PMCID: PMC6684907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 06/04/2019] [Indexed: 06/10/2023]
Abstract
Breast cancer is a leading cause of cancer-related death in the women worldwide. Apatinib is a novel tyrosine kinase inhibitor that selectively binds and inhibits vascular endothelial growth factor receptor 2 (VEGFR-2). The clinical trials have demonstrated the objective efficacy of Apatinib against metastatic breast cancer. However, the underlying mechanism is not well established. In the present study, the breast cell lines, BT-474 and MCF-7, were investigated. The effect of Apatinib on the cell viability was determined using CCK-8 assay. The migration, invasion, cell cycle distribution and the downstream signaling of VEGFR-2 in the cells were determined after 48 h treatment with this drug. Subsequently, Vector of angiomotin (AMOT) cDNA was transfected into MCF-7 cells. The cells were either exposed to Apatinib or vehicle and then examined for cell viabilities, migration, invasion, cell cycle distribution and the downstream signaling of VEGFR-2. Apatinib demonstrated a dose-dependent, significant inhibition of cell viabilities, migration and invasion of BT-474 and MCF-7 cells, with an increase in the percentage of cells in G1 phase and a decrease in S phase. In addition, in MCF-7 cells, Apatinib decreased AMOT expression, accompanied with the decreased expression of LATS1/2, YAP, ERK1/2 phosphorylation and cyclin D1. The inhibitory effect of Apatinib on the cell activities and protein expressions were significantly suppressed by AMOT overexpression. The results of this study indicated that Apatinib inhibited MCF-7 cell proliferation and invasion through AMOT/VEGFR-2 pathway.
Collapse
Affiliation(s)
- Haige Zhang
- Department of Radiation Oncology, Zhongshan Hospital, Fudan UniversityShanghai 200032, China
- Department of Radiation Oncology, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| | - Jing Sun
- Department of Radiation Oncology, Zhongshan Hospital, Fudan UniversityShanghai 200032, China
| | - Wencui Ju
- Department of Radiation Oncology, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| | - Bin Li
- Department of Radiation Oncology, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| | - Yunfeng Lou
- Department of Radiation Oncology, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| | - Guoqiang Zhang
- Department of Infectious Disease, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| | - Gaofeng Liang
- Medical College, Henan University of Science and TechnologyLuoyang 471023, Henan, China
| | - Xiaoyong Luo
- Department of Radiation Oncology, Luoyang Central Hospital Affiliated to Zhengzhou UniversityLuoyang 471000, Henan, China
| |
Collapse
|
15
|
Hall L, Donovan E, Araya M, Idowa E, Jiminez-Segovia I, Folck A, Wells CD, Kimble-Hill AC. Identification of Specific Lysines and Arginines That Mediate Angiomotin Membrane Association. ACS OMEGA 2019; 4:6726-6736. [PMID: 31179409 PMCID: PMC6547806 DOI: 10.1021/acsomega.9b00165] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Accepted: 03/28/2019] [Indexed: 05/17/2023]
Abstract
The family of Angiomotin (Amot) proteins regulate several biological pathways associated with cellular differentiation, proliferation, and migration. These adaptor proteins target proteins to the apical membrane, actin fibers, or the nucleus. A major function of the Amot coiled-coil homology (ACCH) domain is to initiate protein interactions with the cellular membrane, particularly those containing phosphatidylinositol lipids. The work presented in this article uses several ACCH domain lysine/arginine mutants to probe the relative importance of individual residues for lipid binding. This identified four lysine and three arginine residues that mediate full lipid binding. Based on these findings, three of these residues were mutated to glutamates in the Angiomotin 80 kDa splice form and were incorporated into human mammary cell lines. Results show that mutating three of these residues in the context of full-length Angiomotin reduced the residence of the protein at the apical membrane. These findings provide new insight into how the ACCH domain mediates lipid binding to enable Amot proteins to control epithelial cell growth.
Collapse
Affiliation(s)
- Le’Celia Hall
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Emily Donovan
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Michael Araya
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Eniola Idowa
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Ilse Jiminez-Segovia
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Anthony Folck
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Clark D. Wells
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| | - Ann C. Kimble-Hill
- Department of Biochemistry
and Molecular Biology, Indiana University
School of Medicine, Room MS 4053, 635 Barnhill Drive, Indianapolis, Indiana 46202, United
States
| |
Collapse
|
16
|
Angiomotin-p130 inhibits β-catenin stability by competing with Axin for binding to tankyrase in breast cancer. Cell Death Dis 2019; 10:179. [PMID: 30792381 PMCID: PMC6385204 DOI: 10.1038/s41419-019-1427-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 01/02/2023]
Abstract
Growing evidence indicates that Angiomotin (Amot)-p130 and Amot-p80 have different physiological functions. We hypothesized that Amot-p130 is a tumor suppressor gene in breast cancer, in contrast with the canonical oncogenicity of Amot-p80 or total Amot. To clarify the role of Amot-p130 in breast cancer, we performed real-time quantitative PCR, western blotting, flow cytometry, microarray, immunofluorescence, immunoprecipitation, and tumor sphere-formation assays in vitro, as well as tumorigenesis and limited-dilution analysis in vivo. In this study, we showed that Amot-p130 inhibited the proliferation, migration, and invasion of breast cancer cells. Interestingly, transcriptional profiles indicated that genes differentially expressed in response to Amot-p130 knockdown were mostly related to β-catenin signaling in MCF7 cells. More importantly, most of the downstream partners of β-catenin were associated with stemness. In a further validation, Amot-p130 inhibited the cancer stem cell potential of breast cancer cells both in vitro and in vivo. Mechanistically, Amot-p130 decreased β-catenin stability by competing with Axin for binding to tankyrase, leading to a further inhibition of the WNT pathway. In conclusions, Amot-p130 functions as a tumor suppressor gene in breast cancer, disrupting β-catenin stability by competing with Axin for binding to tankyrase. Amot-p130 was identified as a potential target for WNT pathway-targeted therapies in breast cancer.
Collapse
|
17
|
Peck C, Virtanen P, Johnson D, Kimble-Hill AC. Using the Predicted Structure of the Amot Coiled Coil Homology Domain to Understand Lipid Binding. INDIANA UNIVERSITY JOURNAL OF UNDERGRADUATE RESEARCH 2018; 4:27-46. [PMID: 30957019 PMCID: PMC6448796 DOI: 10.14434/iujur.v4i1.24528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiomotins (Amots) are a family of adapter proteins that modulate cellular polarity, differentiation, proliferation, and migration. Amot family members have a characteristic lipid-binding domain, the coiled coil homology (ACCH) domain that selectively targets the protein to membranes, which has been directly linked to its regulatory role in the cell. Several spot blot assays were used to validate the regions of the domain that participate in its membrane association, deformation, and vesicle fusion activity, which indicated the need for a structure to define the mechanism. Therefore, we sought to understand the structure-function relationship of this domain in order to find ways to modulate these signaling pathways. After many failed attempts to crystallize the ACCH domain of each Amot family member for structural analysis, we decided to pursue homologous models that could be refined using small angle x-ray scattering data. Theoretical models were produced using the homology software SWISS-MODEL and threading software I-TASSER and LOMETS, followed by comparison to SAXS data for model selection and refinement. We present a theoretical model of the domain that is driven by alpha helices and short random coil regions. These alpha helical regions form a classic dimer interface followed by two wide spread legs that we predict to be the lipid binding interface.
Collapse
|
18
|
Kimble-Hill AC, Petrache HI, Seifert S, Firestone MA. Reorganization of Ternary Lipid Mixtures of Nonphosphorylated Phosphatidylinositol Interacting with Angiomotin. J Phys Chem B 2018; 122:8404-8415. [PMID: 29877706 PMCID: PMC6351316 DOI: 10.1021/acs.jpcb.7b12641] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Phosphatidylinositol (PI) lipids are necessary for many cellular signaling pathways of membrane associated proteins, such as angiomotin (Amot). The Amot family regulates cellular polarity, growth, and migration. Given the low concentration of PI lipids in these membranes, it is likely that such protein-membrane interactions are stabilized by lipid domains or small lipid clusters. By small-angle X-ray scattering, we show that nonphosphorylated PI lipids induce lipid demixing in ternary mixtures of phosphatidylcholine (PC) and phosphatidylethanolamine (PE), likely because of preferential interactions between the head groups of PE and PI. These results were obtained in the presence of buffer containing tris(hydroxymethyl)aminomethane, 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid, NaCl, ethylenediaminetetraacetic acid, dithiothreitol, and benzamidine at pH 8.0 that in previous work showed an ability to cause PC to phase separate but are necessary to stabilize Amot for in vitro experimentation. Collectively, this provided a framework for determining the effect of Amot on lipid organization. Using fluorescence spectroscopy, we were able to show that the association of Amot with this lipid platform causes significant reorganization of the lipid into a more homogenous structure. This reorganization mechanism could be the basis for Amot membrane association and fusogenic activity previously described in the literature and should be taken into consideration in future protein-membrane interaction studies.
Collapse
Affiliation(s)
- Ann C. Kimble-Hill
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, MS 4053, 635 Barnhill Dr., Indianapolis, Indiana 46202, United States
| | - Horia I. Petrache
- Department of Physics, Indiana University Purdue University Indianapolis, LD 154, 402 N. Blackford Street, Indianapolis, Indiana 46202, United States
| | - Soenke Seifert
- X-Ray Science Division, Advanced Photon Source, Argonne National Laboratory, Argonne, Illinois 60439, United States
| | - Millicent A. Firestone
- Center for Integrated Nanotechnologies, Los Alamos National Laboratory, MPA-CINT, MS K771, Los Alamos, New Mexico 87545, United States
| |
Collapse
|
19
|
Angiomotin regulates prostate cancer cell proliferation by signaling through the Hippo-YAP pathway. Oncotarget 2018; 8:10145-10160. [PMID: 28052036 PMCID: PMC5354648 DOI: 10.18632/oncotarget.14358] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 12/13/2016] [Indexed: 02/05/2023] Open
Abstract
Angiomotin (AMOT) is a family of proteins found to be a component of the apical junctional complex of vertebrate epithelial cells and is recently found to play important roles in neurofibromatosis type 2 (NF-2). Whether AMOT plays a role in prostate cancer (PCa) is unknown. AMOT is expressed as two isoforms, AMOTp80 and AMOTp130, which has a 409 aa N-terminal domain that is absent in AMOTp80. Both AMOTp80 and AMOTp130 are expressed in LNCaP and C4-2B4, but at a low to undetectable level in PC3, DU145, and BPH1 cells. Further study showed that AMOTp130 and AMOTp80 have distinct functions in PCa cells. We found that AMOTp80, but not AMOT p130, functioned as a tumor promoter by enhancing PCa cell proliferation. Mechanistic studies showed that AMOTp80 signaled through the Hippo pathway by promoting nuclear translocation of YAP, resulting in an increased expression of YAP target protein BMP4. Moreover, inhibition of BMP receptor activity by LDN-193189 abrogates AMOTp80-mediated cell proliferation. Together, this study reveals a novel mechanism whereby the AMOTp80-Merlin-MST1-LATS-YAP-BMP4 pathway leads to AMOTp80-induced tumor cell proliferation.
Collapse
|
20
|
Xu G, Zhu H, Zhang M, Xu J. Histone deacetylase 3 is associated with gastric cancer cell growth via the miR-454-mediated targeting of CHD5. Int J Mol Med 2018; 41:155-163. [PMID: 29115379 PMCID: PMC5746286 DOI: 10.3892/ijmm.2017.3225] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Accepted: 09/07/2017] [Indexed: 12/20/2022] Open
Abstract
Gastric cancer (GC) is the third leading cause of cancer-related mortality in China and worlwide; hence, the identification of GC-related genes is necessary for the development of effective treatment strategies. In this study, histone deacetylase 3 (HDAC3) was identified as the most significantly upregulated cancer-related gene in GC tissues by microarray. In accordance with this, HDAC3 expression was found to be upregulated in GC cell lines/tissues. Further experiments indicated that the knockdown of HDAC3 decreased GC cell viability, reduced the colony formation number and decreased tumor weight. To explore the underlying mechanisms, the overexpression of HDAC3 was induced by transfection with an overexpression plasmid, followed by miRNA microarray, and we identified miR-454 as the most markedly upregulated miRNA. Accordingly, miR-454 expression was upregulated in GC cell lines/tissues and a high level of miR-454 indicated a high HDAC3 expression in GC tissues, and miR-454 knockdown reduced cell viability. In addition, a high level of miR-454 was significantly associated with an advanced clinical stage, lymph node metastases and a poor prognosis of patients with GC. Furthermore, CHD5 was identified as a direct target of miR-454. CHD5 was downregulated in GC tissues/cell lines and the expresssion of CHD5 inversely correlated with the level of miR-454 in GC tissues. Taken together, these observations indicate that HDAC3 is associated with GC cell growth via the miR-454-mediated targeting of CHD5.
Collapse
Affiliation(s)
| | | | - Minghui Zhang
- Department of Oncology, People's Hospital of Pudong, Shanghai 201299, P.R. China
| | - Jinhua Xu
- Department of Oncology, People's Hospital of Pudong, Shanghai 201299, P.R. China
| |
Collapse
|
21
|
Hong SA, Son MW, Cho J, Jang SH, Lee HJ, Lee JH, Cho HD, Oh MH, Lee MS. Low angiomotin-p130 with concomitant high Yes-associated protein 1 expression is associated with adverse prognosis of advanced gastric cancer. APMIS 2017; 125:996-1006. [PMID: 28885730 DOI: 10.1111/apm.12750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 07/14/2017] [Indexed: 11/26/2022]
Abstract
Angiomotin (AMOT) promotes angiogenesis and plays a role in neovascularization during tumorigenesis. Recently, the AMOT isoform, AMOT-p130, was shown to exert a regulatory effect on Yes-associated protein 1 (YAP1), a major downstream effector of the Hippo pathway. The specific roles of AMOT-p130 and YAP1 in advanced gastric cancer (AGC) are yet to be established. In this study, a total of 166 patients with AGC were enrolled, and AMOT-p130 and YAP1 levels were analyzed by immunohistochemistry using tissue microarrays. Low AMOT-p130 together with high YAP1 expression (n = 30, 18.1%) was associated with high T stage (p = 0.042), high TNM stage (p = 0.025), and venous invasion (p = 0.048). A Kaplan-Meier survival analysis with log-rank test revealed a significant correlation with decreased AMOT-p130 coupled with high nuclear YAP1 expression with shorter overall survival (p = 0.0045) and disease-free survival (p = 0.0028). Furthermore, multivariate analyses showed that the low AMOT-p130/high YAP1 expression profile was an independent prognostic factor for disease-free survival (p = 0.008, HR = 1.874, CI, 1.177-2.986) and overall survival (p = 0.012, HR = 1.903, CI, 1.152-3.143). Our findings collectively demonstrate that low AMOT-p130 combined with high YAP1 expression is correlated with an unfavorable AGC prognosis.
Collapse
Affiliation(s)
- Soon Auck Hong
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Myoung Won Son
- Department of General Surgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Junhun Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Si-Hyong Jang
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Hyun Ju Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Ji-Hye Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Hyun Deuk Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Mee-Hye Oh
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Moon Soo Lee
- Department of General Surgery, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
22
|
Hildebrand S, Hultin S, Subramani A, Petropoulos S, Zhang Y, Cao X, Mpindi J, Kalloniemi O, Johansson S, Majumdar A, Lanner F, Holmgren L. The E-cadherin/AmotL2 complex organizes actin filaments required for epithelial hexagonal packing and blastocyst hatching. Sci Rep 2017; 7:9540. [PMID: 28842668 PMCID: PMC5572699 DOI: 10.1038/s41598-017-10102-w] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022] Open
Abstract
Epithelial cells connect via cell-cell junctions to form sheets of cells with separate cellular compartments. These cellular connections are essential for the generation of cellular forms and shapes consistent with organ function. Tissue modulation is dependent on the fine-tuning of mechanical forces that are transmitted in part through the actin connection to E-cadherin as well as other components in the adherens junctions. In this report we show that p100 amotL2 forms a complex with E-cadherin that associates with radial actin filaments connecting cells over multiple layers. Genetic inactivation or depletion of amotL2 in epithelial cells in vitro or zebrafish and mouse in vivo, resulted in the loss of contractile actin filaments and perturbed epithelial packing geometry. We further showed that AMOTL2 mRNA and protein was expressed in the trophectoderm of human and mouse blastocysts. Genetic inactivation of amotL2 did not affect cellular differentiation but blocked hatching of the blastocysts from the zona pellucida. These results were mimicked by treatment with the myosin II inhibitor blebbistatin. We propose that the tension generated by the E-cadherin/AmotL2/actin filaments plays a crucial role in developmental processes such as epithelial geometrical packing as well as generation of forces required for blastocyst hatching.
Collapse
Affiliation(s)
- Sebastian Hildebrand
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet and Division of Obstetrics and Gynecology, Karolinska University Hospital, Huddinge, Sweden.,Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Sara Hultin
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Aravindh Subramani
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Sophie Petropoulos
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet and Division of Obstetrics and Gynecology, Karolinska University Hospital, Huddinge, Sweden
| | - Yuanyuan Zhang
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
| | - Xiaofang Cao
- Department of Medical Biochemistry and Microbiology, Uppsala Biomedical Center (BMC), Uppsala University, Uppsala, Sweden
| | - John Mpindi
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Olli Kalloniemi
- Medical Biotechnology, VTT Technical Research Centre of Finland, Turku, Finland.,Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Staffan Johansson
- Department of Medical Biochemistry and Microbiology, Uppsala Biomedical Center (BMC), Uppsala University, Uppsala, Sweden
| | - Arindam Majumdar
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden.,Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, 46285, USA
| | - Fredrik Lanner
- Department of Clinical Sciences, Intervention and Technology (CLINTEC), Karolinska Institutet and Division of Obstetrics and Gynecology, Karolinska University Hospital, Huddinge, Sweden.
| | - Lars Holmgren
- Department of Oncology-Pathology, Cancer Centrum Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
23
|
Lv M, Shen Y, Yang J, Li S, Wang B, Chen Z, Li P, Liu P, Yang J. Angiomotin Family Members: Oncogenes or Tumor Suppressors? Int J Biol Sci 2017; 13:772-781. [PMID: 28656002 PMCID: PMC5485632 DOI: 10.7150/ijbs.19603] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 04/09/2017] [Indexed: 12/17/2022] Open
Abstract
Angiomotin (Amot) family contains three members: Amot (p80 and p130 isoforms), Amot-like protein 1 (Amotl1), and Amot-like protein 2 (Amotl2). Amot proteins play an important role in tube formation and migration of endothelial cells and the regulation of tight junctions, polarity, and epithelial-mesenchymal transition in epithelial cells. Moreover, these proteins regulate the proliferation and migration of cancer cells. In most cancers, Amot family members promote the proliferation and invasion of cancer cells, including breast cancer, osteosarcoma, colon cancer, prostate cancer, head and neck squamous cell carcinoma, cervical cancer, liver cancer, and renal cell cancer. However, in glioblastoma, ovarian cancer, and lung cancer, Amot inhibits the growth of cancer cells. In addition, there are controversies on the regulation of Yes-associated protein (YAP) by Amot. Amot promotes either the internalization of YAP into the nucleus or the retention of YAP in the cytoplasm of different cell types. Moreover, Amot regulates the AMPK, mTOR, Wnt, and MAPK signaling pathways. However, it is unclear whether Amot is an oncogene or a tumor suppressor gene in different cellular processes. This review focuses on the multifunctional roles of Amot in cancers.
Collapse
Affiliation(s)
- Meng Lv
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Yanwei Shen
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Jiao Yang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Shuting Li
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Biyuan Wang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Zheling Chen
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Pan Li
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| | - Peijun Liu
- Center for Translational Medicine, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Yang
- Department of Oncology, the First Affiliated Hospital of Xian Jiaotong University, Xi'an, Shaanxi Province ,710061, P.R. China
| |
Collapse
|
24
|
Jang SH, Cho HD, Lee JH, Lee HJ, Hong SA, Cho J, Kim HJ, Oh MH. Reduced angiomotin p130 expression correlates with poor prognosis in lung adenocarcinoma. J Clin Pathol 2016; 70:625-630. [PMID: 27980054 DOI: 10.1136/jclinpath-2016-204071] [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: 08/13/2016] [Revised: 11/21/2016] [Accepted: 11/27/2016] [Indexed: 12/31/2022]
Abstract
AIMS Lung cancer is the leading cause of cancer-related deaths worldwide, and it still results in a poor prognosis despite research and development of a treatment modality. Angiomotin (AMOT) was first described as a protein involved in angiogenesis, and although the oncogenic and tumour-suppressive roles of AMOT were recently reported, the biological function of AMOT has not yet been clarified. The aim of this study was thus to evaluate the relationship between reduced AMOT p130 expression and clinicopathological parameters, including patients' survival. METHODS We enrolled 67 patients with lung adenocarcinoma in this study and measured the immunoreactivity of AMOT p130 in a tissue microarray. The data were analysed using a χ2 test, Cox regression hazards model and log-rank test with Kaplan-Meier curves. RESULTS Reduced AMOT p130 expression is related to lung adenocarcinoma developed at a young age with statistical significance, but there is no statistical significance for the other clinicopathological parameters. Kaplan-Meier curves with log-rank test showed that reduced AMOT p130 expression had significantly better survival rate compared with the retained group (p=0.002). Univariable and multivariable analyses of the disease free survival revealed that the decreased AMOT expression was an independent prognostic factor (p=0.004, p=0.008, respectively). CONCLUSIONS Decreased AMOT p130 could be an independent indicator of poor survival in patients with lung adenocarcinoma.
Collapse
Affiliation(s)
- Si-Hyong Jang
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Hyun Deuk Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Ji-Hye Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Hyun Ju Lee
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Soon Auck Hong
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Junhun Cho
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Han Jo Kim
- Department of Internal Medicine, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| | - Mee-Hye Oh
- Department of Pathology, Soonchunhyang University Cheonan Hospital, College of Medicine, Soonchunhyang University, Cheonan, Korea
| |
Collapse
|
25
|
Xiao J, Jin K, Wang J, Ma J, Zhang J, Jiang N, Wang H, Luo X, Fei J, Wang Z, Yang X, Ma D. Conditional knockout of TFPI-1 in VSMCs of mice accelerates atherosclerosis by enhancing AMOT/YAP pathway. Int J Cardiol 2016; 228:605-614. [PMID: 27875740 DOI: 10.1016/j.ijcard.2016.11.195] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/06/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Tissue factor pathway inhibitor-1 (TFPI-1) has multiple functions and its precise role and molecular mechanism during the development of atherosclerosis are not clear. OBJECTIVES To determine the effect and molecular mechanism of TFPI-1 deficiency in vascular smooth muscle cells (VSMCs) in atherosclerosis in the apolipoprotein E knockout (ApoE-/-) mouse. METHODS AND RESULTS A mouse model with a conditional knockout of TFPI-1 in VSMCs in an atherosclerosis-prone background (ApoE-/-) was generated. Mice were fed a high fat diet for 18weeks and were then euthanized. Arterial trees and aortas were stained with Sudan IV and were labeled via immunohistochemistry. Cell proliferation and migration of VSMCs in atherosclerotic plaques were assessed. More atherosclerotic lesions and higher levels of proliferation and migration of VSMCs were observed in TFPI-1fl/fl/Sma-Cre+ApoE-/-mice. An interaction between TFPI-1 and angiomotin (AMOT) was identified in human VSMCs by mass spectrometry, immunoprecipitation and co-localization analyses. Signal pathway changes were detected by Western blot analysis, and the expression levels of target genes were determined by real-time PCR. Decreased phosphorylation of AMOT and Yes-associated protein 1 (YAP) in TFPI-1fl/fl/Sma-Cre+ApoE-/- mice resulted in increased expression levels of snail family zinc finger 2 (SLUG) and connective tissue growth factor (CTGF), which are target genes of the Hippo signaling pathway that have been verified as atherosclerosis candidate genes. CONCLUSION Deficiency in TFPI-1 in the VSMCs of ApoE-/- mice accelerated the development of atherosclerosis by promoting the proliferation and migration of VSMCs which may be caused by the decreased phosphorylation of AMOT and YAP. SIGNIFICANCE TFPI-1 has been found to has an anticoagulant activity, induce cell apoptosis and prevent cell proliferation. For the first time, we constructed a line of conditional knockout mice in which the TPFI-1 gene is deleted in VSMCs. We found that TFPI-1 deficiency clearly promoted the development of atherosclerosis when these mice were crossed into an ApoE-/-background. One notable feature of atherosclerosis is the proliferation and migration of smooth muscle cells. Previous reports involved TFPI-1 do not completely explain the proliferation and migration of VSMCs because heterozygous TF deficient (TF±) mice bred in an ApoE-/- background did not show diminished atherosclerosis compared to TF+/+ mice bred in the same background. Our results first confirmed that TFPI-1 interacts with AMOT, which led to a decrease in the phosphorylation of YAP and further increased the genes expression of the proliferation and migration involved. Our results further confirmed that atherosclerosis was a localized disease.
Collapse
Affiliation(s)
- Jiajun Xiao
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Kaiyue Jin
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Jiping Wang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Jing Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Jin Zhang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Nan Jiang
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China
| | - Huijun Wang
- Cardiovascular Center, Children's Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Xinping Luo
- Department of Cardiovascular Medicine, Huashan Hospital Affiliated to Fudan University, Shanghai 200032, China
| | - Jian Fei
- Shanghai Research Centre for Model Organisms, Shanghai 201203,China
| | - Zhugang Wang
- Shanghai Research Centre for Model Organisms, Shanghai 201203,China
| | - Xiao Yang
- Institute of Geriatrics, PLA Postgraduate School of Medicine, PLA General Hospital, Beijing 100853, China
| | - Duan Ma
- Key Laboratory of Metabolism and Molecular Medicine, Ministry of Education, Collaborative Innovation Center of Genetics and Development, Department of Biochemistry and Molecular Biology, Institute of Biomedical Sciences, School of Basic Medical Sciences, Fudan University, Shanghai 20032, China; Cardiovascular Center, Children's Hospital Affiliated to Fudan University, Shanghai 200032, China.
| |
Collapse
|
26
|
Zhang Y, Yuan J, Zhang X, Yan F, Huang M, Wang T, Zheng X, Zhang M. Angiomotin promotes the malignant potential of colon cancer cells by activating the YAP-ERK/PI3K-AKT signaling pathway. Oncol Rep 2016; 36:3619-3626. [PMID: 27779692 DOI: 10.3892/or.2016.5194] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 06/07/2016] [Indexed: 11/06/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related deaths with an increasing incidence in China. The aberrant expression of angiomotin (AMOT) has been confirmed in a variety of tumors and can interact with Yes-associated protein (YAP) to either promote or suppress the progression of cancer. Unfortunately, its role in CRC remains poorly elucidated. Herein, higher levels of AMOT were observed in CRC cell lines. Upregulation of AMOT in LoVo cells markedly increased cell proliferation and apoptotic resistance to 5-fluorouracil. Moreover, its increase also promoted cell invasion and migration. Simultaneously, AMOT silencing markedly attenuated the growth and metastatic potential of HCT116 cells. Notably, AMOT upregulation promoted the activity of YAP by decreasing the expression of phosphorylated YAP and YAP in the cytoplasm and increasing YAP levels in the nucleus. Further mechanistic analysis corroborated that transfection with YAP siRNA notably diminished cell growth, invasion and migration in the AMOT‑overexpressing LoVo cells. Additionally, upregulation of AMOT induced the activation of the ERK and AKT pathways by YAP expression, both associated with the development of CRC. Collectively, these results suggest that AMOT may function as an oncogene in the progression of CRC by activating the YAP-ERK/PI3K-AKT signaling pathway. Therefore, this study presents a promising therapeutic target for CRC.
Collapse
Affiliation(s)
- Yan Zhang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Jun Yuan
- Department of Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xinli Zhang
- Radio Immunity Center, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Futang Yan
- Department of Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Minggang Huang
- CT Department, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Tao Wang
- Department of Radiology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiao Zheng
- Department of Clinical Laboratory, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Ming Zhang
- Department of Medical Imaging, The First Affiliated Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
27
|
Couderc C, Boin A, Fuhrmann L, Vincent-Salomon A, Mandati V, Kieffer Y, Mechta-Grigoriou F, Del Maestro L, Chavrier P, Vallerand D, Brito I, Dubois T, De Koning L, Bouvard D, Louvard D, Gautreau A, Lallemand D. AMOTL1 Promotes Breast Cancer Progression and Is Antagonized by Merlin. Neoplasia 2016; 18:10-24. [PMID: 26806348 PMCID: PMC4735628 DOI: 10.1016/j.neo.2015.11.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 11/18/2015] [Accepted: 11/23/2015] [Indexed: 11/29/2022] Open
Abstract
The Hippo signaling network is a key regulator of cell fate. In the recent years, it was shown that its implication in cancer goes well beyond the sole role of YAP transcriptional activity and its regulation by the canonical MST/LATS kinase cascade. Here we show that the motin family member AMOTL1 is an important effector of Hippo signaling in breast cancer. AMOTL1 connects Hippo signaling to tumor cell aggressiveness. We show that both canonical and noncanonical Hippo signaling modulates AMOTL1 levels. The tumor suppressor Merlin triggers AMOTL1 proteasomal degradation mediated by the NEDD family of ubiquitin ligases through direct interaction. In parallel, YAP stimulates AMOTL1 expression. The loss of Merlin expression and the induction of Yap activity that are frequently observed in breast cancers thus result in elevated AMOTL1 levels. AMOTL1 expression is sufficient to trigger tumor cell migration and stimulates proliferation by activating c-Src. In a large cohort of human breast tumors, we show that AMOTL1 protein levels are upregulated during cancer progression and that, importantly, the expression of AMOTL1 in lymph node metastasis appears predictive of the risk of relapse. Hence we uncover an important mechanism by which Hippo signaling promotes breast cancer progression by modulating the expression of AMOTL1.
Collapse
Affiliation(s)
| | - Alizée Boin
- Institut Curie, Paris, France; CNRS UMR144, Paris, France
| | - Laetitia Fuhrmann
- Institut Curie, Paris, France; CNRS UMR144, Paris, France; Department of Biopathology, Paris, France
| | - Anne Vincent-Salomon
- Institut Curie, Paris, France; Department of Biopathology, Paris, France; INSERM U934, Paris, France
| | - Vinay Mandati
- Institut Curie, Paris, France; CNRS UMR144, Paris, France
| | - Yann Kieffer
- Institut Curie, Paris, France; Stress and Cancer Laboratory, INSERM U830, France
| | | | | | | | - David Vallerand
- Institut Curie, Paris, France; Département de Recherche Translationnelle, Laboratoire d'Investigation Préclinique, Paris, France
| | - Isabelle Brito
- Institut Curie, Paris, France; INSERM U900, Paris, France; Mines ParisTech, Fontainebleau, France
| | - Thierry Dubois
- Institut Curie, Paris, France; Département de Recherche Translationnelle, Breast Cancer Biology Group, France
| | | | - Daniel Bouvard
- INSERM U823, Institut Albert Bonniot, Grenoble, France; Université Joseph Fourier, Grenoble, France
| | - Daniel Louvard
- Institut Curie, Paris, France; CNRS UMR144, Paris, France
| | | | | |
Collapse
|
28
|
Campbell CI, Samavarchi-Tehrani P, Barrios-Rodiles M, Datti A, Gingras AC, Wrana JL. The RNF146 and tankyrase pathway maintains the junctional Crumbs complex through regulation of angiomotin. J Cell Sci 2016; 129:3396-411. [PMID: 27521426 DOI: 10.1242/jcs.188417] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/22/2016] [Indexed: 12/11/2022] Open
Abstract
The Crumbs complex is an important determinant of epithelial apical-basal polarity that functions in regulation of tight junctions, resistance to epithelial-to-mesenchymal transitions and as a tumour suppressor. Although the functional role of the Crumbs complex is being elucidated, its regulation is poorly understood. Here, we show that suppression of RNF146, an E3 ubiquitin ligase that recognizes ADP-ribosylated substrates, and tankyrase, a poly(ADP-ribose) polymerase, disrupts the junctional Crumbs complex and disturbs the function of tight junctions. We show that RNF146 binds a number of polarity-associated proteins, in particular members of the angiomotin (AMOT) family. Accordingly, AMOT proteins are ADP-ribosylated by TNKS2, which drives ubiquitylation by RNF146 and subsequent degradation. Ablation of RNF146 or tankyrase, as well as overexpression of AMOT, led to the relocation of PALS1 (a Crumbs complex component) from the apical membrane to internal puncta, a phenotype that is rescued by AMOTL2 knockdown. We thus reveal a new function of RNF146 and tankyrase in stabilizing the Crumbs complex through downregulation of AMOT proteins at the apical membrane.
Collapse
Affiliation(s)
- Craig I Campbell
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Payman Samavarchi-Tehrani
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Miriam Barrios-Rodiles
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Alessandro Datti
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5
| | - Anne-Claude Gingras
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5 Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | - Jeffrey L Wrana
- Center for Systems Biology, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, Canada M5G 1X5 Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
29
|
Zhang H, Fan Q. MicroRNA-205 inhibits the proliferation and invasion of breast cancer by regulating AMOT expression. Oncol Rep 2015; 34:2163-70. [PMID: 26239614 DOI: 10.3892/or.2015.4148] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/29/2015] [Indexed: 11/06/2022] Open
Abstract
It has been reported that the expression of angiomotin (AMOT) is upregulated in breast cancer. However, the regulatory mechanism remains unknown. In the present study, we aimed to ascertain whether the expression of AMOT is regulated by microRNAs (miRNAs) in breast cancer. In the present study, miR-205 was significantly downregulated in breast cancer samples and it was identified to directly target the 3'-untranslated region (3'-UTR) of AMOT in breast cancer MCF-7 cells by luciferase assay. miR-205 and small interfering RNA (siRNA)-mediated AMOT-knockdown experiments revealed that miR-205 significantly inhibited the proliferation and the invasion of MCF-7 cells through a decrease in the expression of AMOT, yet had no effect on apoptosis. Furthermore, we observed that the overexpression of AMOT partially reversed the inhibitory effect of miR-205 on the growth and the invasion of MCF-7 cells. The data indicated that miR-205 regulated the proliferation and the invasion of breast cancer cells through suppression of AMOT expression, at least partly. Therefore, the disordered decreased expression of miR-205 and the resulting AMOT upregulation contributes to breast carcinogenesis, and miR-205-AMOT represents a new potential therapeutic target for the treatment of breast carcinoma.
Collapse
Affiliation(s)
- Haige Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
30
|
Sun Z, Prodduturi N, Sun SY, Thompson EA, Kocher JPA. Chromosome X genomic and epigenomic aberrations and clinical implications in breast cancer by base resolution profiling. Epigenomics 2015; 7:1099-110. [PMID: 26039248 DOI: 10.2217/epi.15.43] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
AIM Abnormal inactivation or loss of inactivated X chromosome (Xi) is implicated in women's cancer. However, the underlying mechanisms and clinical relevance are little known. MATERIALS & METHODS High-throughput sequencing was conducted on breast cancer cell lines for copy number, RNA expression and 5'-methylcytosine in ChrX. The results were examined in primary breast tumors. RESULTS & CONCLUSION Breast cancer cells demonstrated reduced or total loss of hemimethylation. Most cell lines lost part or one of X chromosomes. Cell lines without ChrX loss were more active in gene expression. DNA methylation was corroborated with Xi control lincRNA XIST. Similar transcriptome and DNA methylation changes were observed in primary breast cancer datasets with clinical phenotype associations. Dramatic genomic and epigenomic changes in ChrX may be used for potential diagnostic or prognostic markers in breast cancer.
Collapse
Affiliation(s)
- Zhifu Sun
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Naresh Prodduturi
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic, Rochester, MN 55905, USA
| | - Susan Y Sun
- Medical School, University of Minnesota, Minneapolis, MN 55455, USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jean-Pierre A Kocher
- Department of Health Sciences Research, Division of Biomedical Statistics & Informatics, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
31
|
LV MENG, LV MEILING, CHEN LING, QIN TIANJIE, ZHANG XIAO, LIU PEIJUN, YANG JIN. Angiomotin promotes breast cancer cell proliferation and invasion. Oncol Rep 2015; 33:1938-46. [DOI: 10.3892/or.2015.3780] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 01/21/2015] [Indexed: 11/06/2022] Open
|
32
|
Shi P, Feng J, Chen C. Hippo pathway in mammary gland development and breast cancer. Acta Biochim Biophys Sin (Shanghai) 2015; 47:53-9. [PMID: 25467757 DOI: 10.1093/abbs/gmu114] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Accumulated evidence suggests that the Hippo signaling pathway plays crucial roles in mammary gland development and breast cancer. Key components of the Hippo pathway regulate breast epithelial cell proliferation, migration, invasion, and stemness. Additionally, the Hippo pathway regulates breast tumor growth, metastasis, and drug resistance. It is expected that the Hippo pathway will provide novel therapeutic targets for breast cancer. This review will discuss and summarize the roles of several core components of the Hippo pathway in mammary gland development and breast cancer.
Collapse
Affiliation(s)
- Peiguo Shi
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming 650223, China University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jing Feng
- Department of laboratory medicine & Central Laboratory, South Medical University Affiliated Fengxian Hospital, Shanghai 201499, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Chinese Academy of Sciences, Kunming Institute of Zoology, Kunming 650223, China
| |
Collapse
|
33
|
Ortiz A, Lee YC, Yu G, Liu HC, Lin SC, Bilen MA, Cho H, Yu-Lee LY, Lin SH. Angiomotin is a novel component of cadherin-11/β-catenin/p120 complex and is critical for cadherin-11-mediated cell migration. FASEB J 2014; 29:1080-91. [PMID: 25466890 DOI: 10.1096/fj.14-261594] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Loss of E-cadherin and up-regulation of mesenchymal cadherins, a hallmark of the epithelial-mesenchymal transition, contributes to migration and dissemination of cancer cells. Expression of human cadherin-11 (Cad11), also known as osteoblast cadherin, in prostate cancer increases the migration of prostate cancer cells. How Cad11 mediates cell migration is unknown. Using the human Cad11 cytoplasmic domain in pulldown assays, we identified human angiomotin (Amot), known to be involved in cell polarity, migration, and Hippo pathway, as a component of the Cad11 protein complex. Deletion analysis showed that the last C-terminal 10 amino acids in Cad11 cytoplasmic domain are required for Amot binding. Further, Cad11 preferentially interacts with Amot-p80 than Amot-p130 isoform and binds directly to the middle domain of Amot-p80. Cad11-Amot interaction affects Cad11-mediated cell migration, but not homophilic adhesion, as deletion of Amot binding motif of Cad11 (Cad11-ΔAmot) did not abolish Cad11-mediated cell-cell adhesion of mouse L cells, but significantly reduced Cad11-mediated cell migration of human C4-2B4 and PC3-mm2 prostate cancer cells and human HEK293T cells. Together, our studies identified Amot-p80 as a novel component of the Cad11 complex and demonstrated that Amot-p80 is critical for Cad11-mediated cell migration.
Collapse
Affiliation(s)
- Angelica Ortiz
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Yu-Chen Lee
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Guoyu Yu
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hsuan-Chen Liu
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Song-Chang Lin
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Melmet Asim Bilen
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Hyojin Cho
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Li-Yuan Yu-Lee
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Sue-Hwa Lin
- Departments of *Translational Molecular Pathology and Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA; University of Texas Graduate School of Biomedical Sciences, Houston, Texas, USA; and Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
34
|
Angiomotin decreases lung cancer progression by sequestering oncogenic YAP/TAZ and decreasing Cyr61 expression. Oncogene 2014; 34:4056-68. [PMID: 25381822 DOI: 10.1038/onc.2014.333] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 08/18/2014] [Accepted: 08/22/2014] [Indexed: 12/15/2022]
Abstract
Lung cancer is the leading cause of cancer death worldwide, with metastasis underlying majority of related deaths. Angiomotin (AMOT), a scaffold protein, has been shown to interact with oncogenic Yes-associated protein/transcriptional co-activator with a PDZ-binding motif (YAP/TAZ) proteins, suggesting a potential role in tumor progression. However, the functional role of AMOT in lung cancer remains unknown. This study aimed to identify the patho-physiological characteristics of AMOT in lung cancer progression. Results revealed that AMOT expression was significantly decreased in clinical lung cancer specimens. Knockdown of AMOT in a low metastatic CL1-0 lung cancer cell line initiated cancer proliferation, migration, invasion and epithelial-mesenchymal transition. The trigger of cancer progression caused by AMOT loss was transduced by decreased cytoplasmic sequestration and increased nuclear translocation of oncogenic co-activators YAP/TAZ, leading to increased expression of the growth factor, Cyr61. Tumor promotion by AMOT knockdown was reversed when YAP/TAZ or Cyr61 was absent. Further, AMOT knockdown increased the growth and spread of Lewis lung carcinoma in vivo. These findings suggest that AMOT is a crucial suppressor of lung cancer metastasis and highlight its critical role as a tumor suppressor and its potential as a prognostic biomarker and therapeutic target for lung cancer.
Collapse
|
35
|
Hakami F, Darda L, Stafford P, Woll P, Lambert DW, Hunter KD. The roles of HOXD10 in the development and progression of head and neck squamous cell carcinoma (HNSCC). Br J Cancer 2014; 111:807-16. [PMID: 25010866 PMCID: PMC4134504 DOI: 10.1038/bjc.2014.372] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Revised: 06/04/2014] [Accepted: 06/09/2014] [Indexed: 01/02/2023] Open
Abstract
Background: HOX gene expression is altered in many cancers; previous microarray revealed changes in HOX gene expression in head and neck squamous cell carcinoma (HNSCC), particularly HOXD10. Methods: HOXD10 expression was assessed by qPCR and immunoblotting in vitro and by immunohistochemistry (IHC) in tissues. Low-expressing cells were stably transfected with HOXD10 and the phenotype assessed with MTS, migration and adhesion assays and compared with the effects of siRNA knockdown in high-HOXD10-expressing cells. Novel HOXD10 targets were identified using expression microarrays, confirmed by reporter assay, and validated in tissues using IHC. Results: HOXD10 expression was low in NOKs, high in most primary tumour cells, and low in lymph node metastasis cells, a pattern confirmed using IHC in tissues. Overexpression of HOXD10 decreased cell invasion but increased proliferation, adhesion and migration, with knockdown causing reciprocal effects. There was no consistent effect on apoptosis. Microarray analysis identified several putative HOXD10-responsive genes, including angiomotin (AMOT-p80) and miR-146a. These were confirmed as HOXD10 targets by reporter assay. Manipulation of AMOT-p80 expression resulted in phenotypic changes similar to those on manipulation of HOXD10 expression. Conclusions: HOXD10 expression varies by stage of disease and produces differential effects: high expression giving cancer cells a proliferative and migratory advantage, and low expression may support invasion/metastasis, in part, by modulating AMOT-p80 levels.
Collapse
Affiliation(s)
- F Hakami
- 1] Unit of Oral & Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK [2] Department of Pathology and Laboratory Medicine, King Abdulaziz Medical City-WR, Jeddah, Saudi Arabia
| | - L Darda
- Unit of Oral & Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - P Stafford
- Unit of Oral & Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - P Woll
- Academic Unit of Clinical Oncology, University of Sheffield, Sheffield S10 2SJ, UK
| | - D W Lambert
- Unit of Oral & Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK
| | - K D Hunter
- 1] Unit of Oral & Maxillofacial Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK [2] Department of Oral Pathology and Biology, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
36
|
The Angiomotins--from discovery to function. FEBS Lett 2014; 588:2693-703. [PMID: 24548561 DOI: 10.1016/j.febslet.2014.02.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 02/05/2014] [Accepted: 02/06/2014] [Indexed: 12/13/2022]
Abstract
Angiomotins were originally identified as angiostatin binding proteins and implicated in the regulation of endothelial cell migration. Recent studies have shed light on the role of Angiomotins and other members of the Motin protein family in epithelial cells and in pathways directly linked to the pathogenesis of cancer. In particular, Motins have been shown to play a role in signaling pathways regulated by small G-proteins and the Hippo-YAP pathway. In this review the role of the Motin protein family in these signaling pathways will be described and open questions will be discussed.
Collapse
|
37
|
Dai X, She P, Chi F, Feng Y, Liu H, Jin D, Zhao Y, Guo X, Jiang D, Guan KL, Zhong TP, Zhao B. Phosphorylation of angiomotin by Lats1/2 kinases inhibits F-actin binding, cell migration, and angiogenesis. J Biol Chem 2013; 288:34041-34051. [PMID: 24106267 DOI: 10.1074/jbc.m113.518019] [Citation(s) in RCA: 122] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Hippo tumor suppressor pathway plays important roles in organ size control through Lats1/2 mediated phosphorylation of the YAP/TAZ transcription co-activators. However, YAP/TAZ independent functions of the Hippo pathway are largely unknown. Here we report a novel role of the Hippo pathway in angiogenesis. Angiomotin p130 isoform (AMOTp130) is phosphorylated on a conserved HXRXXS motif by Lats1/2 downstream of GPCR signaling. Phosphorylation disrupts AMOT interaction with F-actin and correlates with reduced F-actin stress fibers and focal adhesions. Furthermore, phosphorylation of AMOT by Lats1/2 inhibits endothelial cell migration in vitro and angiogenesis in zebrafish embryos in vivo. Thus AMOT is a direct substrate of Lats1/2 mediating functions of the Hippo pathway in endothelial cell migration and angiogenesis.
Collapse
Affiliation(s)
- Xiaoming Dai
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Peilu She
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Fangtao Chi
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Ying Feng
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Huan Liu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Daqing Jin
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Yiqiang Zhao
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Xiaocan Guo
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Dandan Jiang
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Kun-Liang Guan
- Department of Pharmacology and Moores Cancer Center, University of California at San Diego, La Jolla, California 92093-0815
| | - Tao P Zhong
- State Key Laboratory of Genetic Engineering, Department of Genetics, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Bin Zhao
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
38
|
Serum deprivation inhibits the transcriptional co-activator YAP and cell growth via phosphorylation of the 130-kDa isoform of Angiomotin by the LATS1/2 protein kinases. Proc Natl Acad Sci U S A 2013; 110:17368-73. [PMID: 24101513 DOI: 10.1073/pnas.1308236110] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Large tumor suppressor (LATS)1/2 protein kinases transmit Hippo signaling in response to intercellular contacts and serum levels to limit cell growth via the inhibition of Yes-associated protein (YAP). Here low serum and high LATS1 activity are found to enhance the levels of the 130-kDa isoform of angiomotin (Amot130) through phosphorylation by LATS1/2 at serine 175, which then forms a binding site for 14-3-3. Such phosphorylation, in turn, enables the ubiquitin ligase atrophin-1 interacting protein (AIP)4 to bind, ubiquitinate, and stabilize Amot130. Consistently, the Amot130 (S175A) mutant, which lacks LATS phosphorylation, bound AIP4 poorly under all conditions and showed reduced stability. Amot130 and AIP4 also promoted the ubiquitination and degradation of YAP in response to serum starvation, unlike Amot130 (S175A). Moreover, silencing Amot130 expression blocked LATS1 from inhibiting the expression of connective tissue growth factor, a YAP-regulated gene. Concordant with phosphorylated Amot130 specifically mediating these effects, wild-type Amot130 selectively induced YAP phosphorylation and reduced transcription of connective tissue growth factor in an AIP4-dependent manner versus Amot130 (S175A). Further, Amot130 but not Amot130 (S175A) strongly inhibited the growth of MDA-MB-468 breast cancer cells. The dominant-negative effects of Amot130 (S175A) on YAP signaling also support that phosphorylated Amot130 transduces Hippo signaling. Likewise, Amot130 expression provoked premature growth arrest during mammary cell acini formation, whereas Amot130 (S175A)-expressing cells formed enlarged and poorly differentiated acini. Taken together, the phosphorylation of Amot130 by LATS is found to be a key feature that enables it to inhibit YAP-dependent signaling and cell growth.
Collapse
|
39
|
Adler JJ, Heller BL, Bringman LR, Ranahan WP, Cocklin RR, Goebl MG, Oh M, Lim HS, Ingham RJ, Wells CD. Amot130 adapts atrophin-1 interacting protein 4 to inhibit yes-associated protein signaling and cell growth. J Biol Chem 2013; 288:15181-93. [PMID: 23564455 DOI: 10.1074/jbc.m112.446534] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The adaptor protein Amot130 scaffolds components of the Hippo pathway to promote the inhibition of cell growth. This study describes how Amot130 through binding and activating the ubiquitin ligase AIP4/Itch achieves these effects. AIP4 is found to bind and ubiquitinate Amot130 at residue Lys-481. This both stabilizes Amot130 and promotes its residence at the plasma membrane. Furthermore, Amot130 is shown to scaffold a complex containing overexpressed AIP4 and the transcriptional co-activator Yes-associated protein (YAP). Consequently, Amot130 promotes the ubiquitination of YAP by AIP4 and prevents AIP4 from binding to large tumor suppressor 1. Amot130 is found to reduce YAP stability. Importantly, Amot130 inhibition of YAP dependent transcription is reversed by AIP4 silencing, whereas Amot130 and AIP4 expression interdependently suppress cell growth. Thus, Amot130 repurposes AIP4 from its previously described role in degrading large tumor suppressor 1 to the inhibition of YAP and cell growth.
Collapse
Affiliation(s)
- Jacob J Adler
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Nguyen HB, Babcock JT, Wells CD, Quilliam LA. LKB1 tumor suppressor regulates AMP kinase/mTOR-independent cell growth and proliferation via the phosphorylation of Yap. Oncogene 2012; 32:4100-9. [PMID: 23027127 DOI: 10.1038/onc.2012.431] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2012] [Revised: 07/02/2012] [Accepted: 08/07/2012] [Indexed: 12/31/2022]
Abstract
The liver kinase B1 (LKB1) tumor suppressor inhibits cell growth through its regulation of cellular metabolism and apical-basal polarity. The best understood mechanism whereby LKB1 limits cell growth is through activation of the AMP-activated-protein-kinase/mammalian-target-of-rapamycin (AMPK/mTOR) pathway to control metabolism. As LKB1 is also required for polarized epithelial cells to resist hyperplasia, it is anticipated to function through additional mechanisms. Recently, Yes-associated protein (Yap) has emerged as a transcriptional co-activator that modulates tissue homeostasis in response to cell-cell contact. Thus this study examined a possible connection between Yap and LKB1. Restoration of LKB1 expression in HeLa cells, which lack this tumor suppressor, or short-hairpin RNA knockdown of LKB1 in NTERT immortalized keratinocytes, demonstrated that LKB1 promotes Yap phosphorylation, nuclear exclusion and proteasomal degradation. The ability of phosphorylation-defective Yap mutants to rescue LKB1 phenotypes, such as reduced cell proliferation and cell size, suggest that Yap inhibition contributes to LKB1 tumor suppressor function(s). However, failure of Lats1/2 knockdown to suppress LKB1-mediated Yap regulation suggested that LKB1 signals to Yap via a non-canonical pathway. Additionally, LKB1 inhibited Yap independently of either AMPK or mTOR activation. These findings reveal a novel mechanism whereby LKB1 may restrict cancer cell growth via the inhibition of Yap.
Collapse
Affiliation(s)
- H B Nguyen
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | | | | | |
Collapse
|
41
|
The Nedd4-like ubiquitin E3 ligases target angiomotin/p130 to ubiquitin-dependent degradation. Biochem J 2012; 444:279-89. [DOI: 10.1042/bj20111983] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AMOT (angiomotin) is a membrane-associated protein that is expressed in ECs (endothelial cells) and controls migration, TJ (tight junction) formation, cell polarity and angiogenesis. Recent studies have revealed that AMOT and two AMOT-like proteins, AMOTL1 and AMOTL2, play critical roles in the Hippo pathway by regulating the subcellular localization of the co-activators YAP (Yes-associated protein) and TAZ (transcriptional co-activator with PDZ-binding motif). However, it has been unclear how AMOT is regulated. In the present study, we report that AMOT undergoes proteasomal degradation. We identify three members of Nedd4 (neural-precursor-cell-expressed developmentally down-regulated)-like ubiquitin E3 ligases, Nedd4, Nedd4-2 and Itch, as the ubiquitin E3 ligases for the long isoform of AMOT, AMOT/p130. We demonstrate that Nedd4, Nedd4-2 and Itch mediate poly-ubiquitination of AMOT/p130 in vivo. Overexpression of Nedd4, Nedd4-2 or Itch leads to AMOT/p130 proteasomal degradation. Knockdown of Nedd4, Nedd4-2 and Itch causes an accumulation of steady-state level of AMOT/p130. We also show that three L/P-PXY motifs of AMOT/p130 and the WW domains of Nedd4 mediate their interaction. Furthermore, Nedd4-like ubiquitin E3 ligases might compete with YAP for the binding to AMOT/p130, and subsequently targeting AMOT/p130 for ubiquitin-dependent degradation. Together, these observations reveal a novel post-translational regulatory mechanism of AMOT/p130.
Collapse
|
42
|
Li Z, Wang Y, Zhang M, Xu P, Huang H, Wu D, Meng A. The Amotl2 gene inhibits Wnt/β-catenin signaling and regulates embryonic development in zebrafish. J Biol Chem 2012; 287:13005-15. [PMID: 22362771 DOI: 10.1074/jbc.m112.347419] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The Motin family proteins can regulate cell polarity, cell mobility, and proliferation during embryonic development by controlling distinct signaling pathways. In this study, we demonstrate that amotl2 knockdown in zebrafish wild-type embryos results in embryonic dorsalization, and this effect can be antagonized by co-knockdown of the dorsal inducer β-catenin2. Overexpression of amotl2 in masterblind (mbl) homozygous embryos, in which canonical Wnt signaling is up-regulated due to an axin1 mutation, transforms eyeless phenotype into smaller eyes, whereas co-knockdown of amot, amotl1, and amotl2 leads to development of smaller eyes in mbl heterozygotes. In cultured mammalian cells, Motin family members all possess the ability to attenuate Wnt/β-catenin signaling. Focusing on Amotl2, we show that Amotl2 can associate with and trap β-catenin in the Rab11-positive recycling endosomes, and as a result, the amount of β-catenin in the cytosol and nucleus is reduced. Thus, our findings provide novel insights into functions of Motin family members and regulation of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Zhiqiang Li
- State-Key Laboratory of Biomembrane and Membrane Engineering, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | | | | | | | | | | | | |
Collapse
|
43
|
Paramasivam M, Sarkeshik A, Yates JR, Fernandes MJG, McCollum D. Angiomotin family proteins are novel activators of the LATS2 kinase tumor suppressor. Mol Biol Cell 2011; 22:3725-33. [PMID: 21832154 PMCID: PMC3183025 DOI: 10.1091/mbc.e11-04-0300] [Citation(s) in RCA: 161] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The Hippo pathway kinase LATS2 promotes contact inhibition of growth. How LATS2 is activated in response to changes in cell density is unknown. It is found that tight junction protein AMOTL2 is a novel activator of LATS2, raising the possibility that tight junction assembly promotes LATS2-dependent inhibition of cell proliferation. LATS2 kinase functions as part of the Hippo pathway to promote contact inhibition of growth and tumor suppression by phosphorylating and inhibiting the transcriptional coactivator YAP. LATS2 is activated by the MST2 kinase. How LATS2 is activated by MST2 in response to changes in cell density is unknown. Here we identify the angiomotin-family tight junction protein AMOTL2 as a novel activator of LATS2. Like AMOTL2, the other angiomotin-family proteins AMOT and AMOTL1 also activate LATS2 through a novel conserved domain that binds and activates LATS2. AMOTL2 binds MST2, LATS2, and YAP, suggesting that AMOTL2 might serve as a scaffold protein. We show that LATS2, AMOTL2, and YAP all localize to tight junctions, raising the possibility that clustering of Hippo pathway components at tight junctions might function to trigger LATS2 activation and growth inhibition in response to increased cell density.
Collapse
Affiliation(s)
- Murugan Paramasivam
- Department of Microbial and Physiological Systems and Program in Cell Dynamics, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | | | | | | | | |
Collapse
|