1
|
Trelford CB, Shepherd TG. LKB1 biology: assessing the therapeutic relevancy of LKB1 inhibitors. Cell Commun Signal 2024; 22:310. [PMID: 38844908 PMCID: PMC11155146 DOI: 10.1186/s12964-024-01689-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024] Open
Abstract
Liver Kinase B1 (LKB1), encoded by Serine-Threonine Kinase 11 (STK11), is a master kinase that regulates cell migration, polarity, proliferation, and metabolism through downstream adenosine monophosphate-activated protein kinase (AMPK) and AMPK-related kinase signalling. Since genetic screens identified STK11 mutations in Peutz-Jeghers Syndrome, STK11 mutants have been implicated in tumourigenesis labelling it as a tumour suppressor. In support of this, several compounds reduce tumour burden through upregulating LKB1 signalling, and LKB1-AMPK agonists are cytotoxic to tumour cells. However, in certain contexts, its role in cancer is paradoxical as LKB1 promotes tumour cell survival by mediating resistance against metabolic and oxidative stressors. LKB1 deficiency has also enhanced the selectivity and cytotoxicity of several cancer therapies. Taken together, there is a need to develop LKB1-specific pharmacological compounds, but prior to developing LKB1 inhibitors, further work is needed to understand LKB1 activity and regulation. However, investigating LKB1 activity is strenuous as cell/tissue type, mutations to the LKB1 signalling pathway, STE-20-related kinase adaptor protein (STRAD) binding, Mouse protein 25-STRAD binding, splicing variants, nucleocytoplasmic shuttling, post-translational modifications, and kinase conformation impact the functional status of LKB1. For these reasons, guidelines to standardize experimental strategies to study LKB1 activity, associate proteins, spliced isoforms, post-translational modifications, and regulation are of upmost importance to the development of LKB1-specific therapies. Therefore, to assess the therapeutic relevancy of LKB1 inhibitors, this review summarizes the importance of LKB1 in cell physiology, highlights contributors to LKB1 activation, and outlines the benefits and risks associated with targeting LKB1.
Collapse
Affiliation(s)
- Charles B Trelford
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada.
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada.
| | - Trevor G Shepherd
- The Mary &, John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, 790 Commissioners Road East, Room A4‑921, London, ON, N6A 4L6, Canada
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics and Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
2
|
Lagoudaki ED, Koutsopoulos AV, Sfakianaki M, Papadaki C, Manikis GC, Voutsina A, Trypaki M, Tsakalaki E, Fiolitaki G, Hatzidaki D, Yiachnakis E, Koumaki D, Mavroudis D, Tzardi M, Stathopoulos EN, Marias K, Georgoulias V, Souglakos J. LKB1 Loss Correlates with STING Loss and, in Cooperation with β-Catenin Membranous Loss, Indicates Poor Prognosis in Patients with Operable Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:1818. [PMID: 38791897 PMCID: PMC11120022 DOI: 10.3390/cancers16101818] [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: 04/07/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
To investigate the incidence and prognostically significant correlations and cooperations of LKB1 loss of expression in non-small cell lung cancer (NSCLC), surgical specimens from 188 metastatic and 60 non-metastatic operable stage I-IIIA NSCLC patients were analyzed to evaluate their expression of LKB1 and pAMPK proteins in relation to various processes. The investigated factors included antitumor immunity response regulators STING and PD-L1; pro-angiogenic, EMT and cell cycle targets, as well as metastasis-related (VEGFC, PDGFRα, PDGFRβ, p53, p16, Cyclin D1, ZEB1, CD24) targets; and cell adhesion (β-catenin) molecules. The protein expression levels were evaluated via immunohistochemistry; the RNA levels of LKB1 and NEDD9 were evaluated via PCR, while KRAS exon 2 and BRAFV600E mutations were evaluated by Sanger sequencing. Overall, loss of LKB1 protein expression was observed in 21% (51/248) patients and correlated significantly with histotype (p < 0.001), KRAS mutations (p < 0.001), KC status (concomitant KRAS mutation and p16 downregulation) (p < 0.001), STING loss (p < 0.001), and high CD24 expression (p < 0.001). STING loss also correlated significantly with loss of LKB1 expression in the metastatic setting both overall (p = 0.014) and in lung adenocarcinomas (LUACs) (p = 0.005). Additionally, LKB1 loss correlated significantly with a lack of or low β-catenin membranous expression exclusively in LUACs, both independently of the metastatic status (p = 0.019) and in the metastatic setting (p = 0.007). Patients with tumors yielding LKB1 loss and concomitant nonexistent or low β-catenin membrane expression experienced significantly inferior median overall survival of 20.50 vs. 52.99 months; p < 0.001 as well as significantly greater risk of death (HR: 3.32, 95% c.i.: 1.71-6.43; p <0.001). Our findings underscore the impact of the synergy of LKB1 with STING and β-catenin in NSCLC, in prognosis.
Collapse
Affiliation(s)
- Eleni D. Lagoudaki
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Anastasios V. Koutsopoulos
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Maria Sfakianaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Chara Papadaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Georgios C. Manikis
- Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece; (G.C.M.); (K.M.)
| | - Alexandra Voutsina
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Maria Trypaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Eleftheria Tsakalaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Georgia Fiolitaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Dora Hatzidaki
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
| | - Emmanuel Yiachnakis
- Laboratory of Bio-Medical Data Analysis Digital Applications and Interdisciplinary Approaches, University of Crete, 71003 Heraklion, Greece;
| | - Dimitra Koumaki
- Department of Dermatology, University General Hospital of Heraklion, Voutes, 71500 Heraklion, Greece;
| | - Dimitrios Mavroudis
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - Maria Tzardi
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Efstathios N. Stathopoulos
- Department of Pathology, University General Hospital of Heraklion, 71500 Heraklion, Greece; (A.V.K.); (M.T.); (E.N.S.)
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
| | - Kostas Marias
- Foundation for Research and Technology Hellas (FORTH), 70013 Heraklion, Greece; (G.C.M.); (K.M.)
| | - Vassilis Georgoulias
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| | - John Souglakos
- School of Medicine, University of Crete, 70013 Heraklion, Greece; (D.M.); (V.G.); (J.S.)
- Laboratory of Translational Oncology, School of Medicine, University of Crete, 70013 Heraklion, Greece; (M.S.); (C.P.); (A.V.); (M.T.); (E.T.); (G.F.); (D.H.)
- Department of Medical Oncology, University General Hospital of Heraklion, 71500 Heraklion, Greece
| |
Collapse
|
3
|
Shao X, Dang Y, Zhang T, Bai N, Huang J, Guo M, Sun L, Li M, Sun X, Zhang X, Han F, Zhang N, Zhuang H, Li Y. LINC00869 Promotes Hepatocellular Carcinoma Metastasis via Protrusion Formation. Mol Cancer Res 2024; 22:282-294. [PMID: 37934195 DOI: 10.1158/1541-7786.mcr-23-0414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/05/2023] [Accepted: 11/03/2023] [Indexed: 11/08/2023]
Abstract
Coordination of filament assembly and membrane remodeling is required for the directional migration of cancer cells. The Wiskott-Aldrich syndrome protein (WASP) recruits the actin-related protein (ARP) 2/3 complex to assemble branched actin networks. The goal of our study was to assess the potential regulatory role exerted by the novel long noncoding RNA (lncRNA) LINC00869 on hepatocellular carcinoma (HCC) cells. We used HCC cells to overexpress or knockdown LINC00869, analyzed patient data from publicly available databases and Cancer Hospital Affiliated with Zhengzhou University, and used a xenograft mouse model of HCC to study the molecular mechanism associated with LINC00869 expression. We found that high levels of LINC00869 expression were associated with poor prognosis in patients with HCC. Next, we detected an interaction between LINC00869 and both WASP and ARP2 in HCC cells, and observed a modulatory effect of LINC00869 on the phosphorylation of WASP at Y291 and the activity of cell division control protein 42 (CDC42). These modulatory roles were required for WASP/CDC42 activity on F-actin polymerization to enhance membrane protrusion formation and maintain persistent cell polarization. This, in turn, promoted the migration and invasion abilities of HCC cells. Finally, we confirmed the role of LINC00869in vivo, using the tumor xenograft mouse model; and identified a positive correlation between LINC00869 expression levels and the phosphorylation levels of WASP in HCC samples. Overall, our findings suggest a unique mechanism by which LINC00869 orchestrates membrane protrusion during migration and invasion of HCC cells. IMPLICATIONS LncRNA LINC00869 regulates the activity of CDC42-WASP pathway and positively affects protrusion formation in HCC cells, which expands the current understanding of lncRNA functions as well as gives a better understanding of carcinogenesis.
Collapse
Affiliation(s)
- Xiaowen Shao
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yamei Dang
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Tingting Zhang
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Nan Bai
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jianing Huang
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Mengya Guo
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Li Sun
- Department of Gynaecology and Obstetrics, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Minghe Li
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xiao Sun
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xinran Zhang
- Department of Pharmacy, Tianjin First Central Hospital, Tianjin, China
| | - Feng Han
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Ning Zhang
- Translational Cancer Research Center, Peking University First Hospital, Beijing, China
- Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Medical University, Tianjin, China
| | - Hao Zhuang
- Department of Hepatic Biliary Pancreatic Surgery, Cancer Hospital Affiliated to Zhengzhou University and Henan Cancer Hospital, Zhengzhou, Henan, China
| | - Yongmei Li
- Department of Pathogen Biology and Department of Genetics, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| |
Collapse
|
4
|
Tilston-Lunel AM, Varelas X. Polarity in respiratory development, homeostasis and disease. Curr Top Dev Biol 2023; 154:285-315. [PMID: 37100521 DOI: 10.1016/bs.ctdb.2023.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
The respiratory system is composed of a multitude of cells that organize to form complex branched airways that end in alveoli, which respectively function to guide air flow and mediate gas exchange with the bloodstream. The organization of the respiratory sytem relies on distinct forms of cell polarity, which guide lung morphogenesis and patterning in development and provide homeostatic barrier protection from microbes and toxins. The stability of lung alveoli, the luminal secretion of surfactants and mucus in the airways, and the coordinated motion of multiciliated cells that generate proximal fluid flow, are all critical functions regulated by cell polarity, with defects in polarity contributing to respiratory disease etiology. Here, we summarize the current knowledge of cell polarity in lung development and homeostasis, highlighting key roles for polarity in alveolar and airway epithelial function and outlining relationships with microbial infections and diseases, such as cancer.
Collapse
|
5
|
Bajaj R, Warner AN, Fradette JF, Gibbons DL. Dance of The Golgi: Understanding Golgi Dynamics in Cancer Metastasis. Cells 2022; 11:1484. [PMID: 35563790 PMCID: PMC9102947 DOI: 10.3390/cells11091484] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/24/2022] [Indexed: 12/17/2022] Open
Abstract
The Golgi apparatus is at the center of protein processing and trafficking in normal cells. Under pathological conditions, such as in cancer, aberrant Golgi dynamics alter the tumor microenvironment and the immune landscape, which enhances the invasive and metastatic potential of cancer cells. Among these changes in the Golgi in cancer include altered Golgi orientation and morphology that contribute to atypical Golgi function in protein trafficking, post-translational modification, and exocytosis. Golgi-associated gene mutations are ubiquitous across most cancers and are responsible for modifying Golgi function to become pro-metastatic. The pharmacological targeting of the Golgi or its associated genes has been difficult in the clinic; thus, studying the Golgi and its role in cancer is critical to developing novel therapeutic agents that limit cancer progression and metastasis. In this review, we aim to discuss how disrupted Golgi function in cancer cells promotes invasion and metastasis.
Collapse
Affiliation(s)
- Rakhee Bajaj
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Amanda N. Warner
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| | - Jared F. Fradette
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
| | - Don L. Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA; (R.B.); (A.N.W.); (J.F.F.)
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030, USA
| |
Collapse
|
6
|
Sumbly V, Landry I. Unraveling the Role of STK11/LKB1 in Non-small Cell Lung Cancer. Cureus 2022; 14:e21078. [PMID: 35165542 PMCID: PMC8826623 DOI: 10.7759/cureus.21078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2022] [Indexed: 12/25/2022] Open
Abstract
There are two major groups of lung cancer: non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). NSCLCs can be further separated into three different categories: lung adenocarcinoma, squamous cell carcinoma, and large cell carcinoma. Pulmonary adenocarcinomas represent nearly half of all lung cancer cases and are known to be caused by smoking, certain occupational exposures, and specific genetic mutations. Scientists have noticed that most NSCLCs are driven by defects in the following genes: EGFR, BRAF, ALK, MET, and HER. Abnormalities in the STK11/LKB1 gene have also been shown to induce lung adenocarcinoma. LKB1-deficient cancer cells contain an overactive AMPK “energy sensor,” which inhibits cellular death and promotes glucose, lipid, and protein synthesis via the mTOR protein complex. Studies have also discovered that the loss of STK11/LKB1 favors oncogenesis by creating an immunosuppressive environment for tumors to grow and accelerate events such as angiogenesis, epithelial-mesenchymal transition (EMT), and cell polarity destabilization. STK11/LKB1-mutant lung cancers are currently treated with radiotherapy with or without chemotherapy. Recent clinical trials studying the effects of glutaminase inhibitors, mTOR inhibitors, and anti-PD-L1 therapy in lung cancer patients have yielded promising results. This narrative review provides an overview of the STK11/LKB1 gene and its role in cancer development. Additionally, a summary of the LKB1/APMK/mTOR is provided.
Collapse
Affiliation(s)
- Vikram Sumbly
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals/Queens, Jamaica, USA
| | - Ian Landry
- Internal Medicine, Icahn School of Medicine at Mount Sinai, New York City Health and Hospitals/Queens, Jamaica, USA
| |
Collapse
|
7
|
Modulating Tumor Microenvironment: A Review on STK11 Immune Properties and Predictive vs Prognostic Role for Non-small-cell Lung Cancer Immunotherapy. Curr Treat Options Oncol 2021; 22:96. [PMID: 34524570 DOI: 10.1007/s11864-021-00891-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2021] [Indexed: 01/07/2023]
Abstract
OPINION STATEMENT The quest for immunotherapy (IT) biomarkers is an element of highest clinical interest in both solid and hematologic tumors. In non-small-cell lung cancer (NSCLC) patients, besides PD-L1 expression evaluation with its intrinsic limitations, tissue and circulating parameters, likely portraying the tumor and its stromal/immune counterparts, have been proposed as potential predictors of IT responsiveness. STK11 mutations have been globally labeled as markers of IT resistance. After a thorough literature review, STK11 mutations condition the prognosis of NSCLC patients receiving ICI-containing regimens, implying a relevant biological and clinical significance. On the other hand, waiting for prospective and solid data, the putative negative predictive value of STK11 inactivation towards IT is sustained by less evidence. The physiologic regulation of multiple cellular pathways performed by STK11 likely explains the multifaceted modifications in tumor cells, stroma, and tumor immune microenvironment (TIME) observed in STK11 mutant lung cancer, particularly explored in the molecular subgroup of KRAS co-mutation. IT approaches available thus far in NSCLC, mainly represented by anti-PD-1/PD-L1 inhibitors, are not promising in the case of STK11 inactivation. Perceptive strategies aimed at modulating the TIME, regardless of STK11 status or specifically addressed to STK11-mutated cases, will hopefully provide valid therapeutic options to be adopted in the clinical practice.
Collapse
|
8
|
Nerve growth factor regulates liver cancer cell polarity and motility. Mol Med Rep 2021; 23:288. [PMID: 33649819 PMCID: PMC7905331 DOI: 10.3892/mmr.2021.11927] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023] Open
Abstract
Nerve growth factor (NGF), a prototypical neurotrophic factor essential for neuronal cell proliferation and survival, has been implicated as a marker of tumor progression, as well as a potential target for novel therapeutic approaches in cancer. To investigate the functional potential of NGF in liver cancer in the present study, a stable NGF-overexpressing HepG2 cell line was generated. The scratch-wound assay was used to investigate cell motility and polarity. Western blotting was performed to evaluate the expression levels of epithelial-mesenchymal transition (EMT)-related proteins, including E-cadherin, N-cadherin and vimentin. Moreover, immunofluorescence was performed to investigate the arrangement of the actin cytoskeleton. Cell anoikis resistance was examined using a suspension culture model and cell apoptosis was examined via flow cytometry. The present results indicated that NGF overexpression in HepG2 cells disrupted HepG2 cell polarity and promoted cell motility. Furthermore, NGF overexpression induced EMT and actin cytoskeleton rearrangement in HepG2 cells, as well as enhanced anoikis resistance and prevented cellular apoptosis. Notably, a tropomyosin receptor kinase A receptor inhibitor blocked NGF-induced cell motility and apoptosis. Therefore, it was suggested that NGF serves a critical role in the invasion and metastasis of liver cancer. The use of NGF as a biomarker or potential new target could lead to the development of novel factors for diagnosis or for improving therapeutic strategies in liver cancer.
Collapse
|
9
|
2-methoxyestradiol sensitizes breast cancer cells to taxanes by targeting centrosomes. Oncotarget 2020; 11:4479-4489. [PMID: 33400733 PMCID: PMC7721614 DOI: 10.18632/oncotarget.27810] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Centrosomes amplification is a hallmark of cancer. We hypothesize that 2-methoxyestradiol (2-ME) sensitizes breast cancer (BC) cells to taxanes by targeting amplified centrosomes. We assessed the extent by which 2-ME together with paclitaxel (PTX) induces centrosome alterations with subsequent mitotic catastrophe in different BC subtypes. 2-ME induced a significant reduction in PTX IC50 values in all cells tested ranging from 28–44% (P < 0.05). Treatment with both PTX and 2-ME significantly increased the number of misaligned metaphases compared to PTX alone (34%, 100% and 52% for MCF7, MDA-MB231 and SUM149, respectively; P < 0.05). The number of cells with multipolar spindle formation was significantly increased (81%, 220% and 285% for MCF7, MDA-MB231 and SUM 149, respectively; P < 0.05). PTX and 2-ME treatment significantly increased interphase declustering in cancer cells (56% for MCF7, 208% for MDA-MB231 and 218% for SUM149, respectively; P < 0.05) and metaphase declustering (1.4-fold, 1.56-fold and 2.48-fold increase for MCF7, MDA-MB231 and SUM149, respectively; P < 0.05). This report is the first to document centrosome declustering as a mechanism of action of 2-ME and provides a potential approach for reducing taxane toxicity in cancer treated patients.
Collapse
|
10
|
Beyond LKB1 Mutations in Non-Small Cell Lung Cancer: Defining LKB1less Phenotype to Optimize Patient Selection and Treatment. Pharmaceuticals (Basel) 2020; 13:ph13110385. [PMID: 33202760 PMCID: PMC7697441 DOI: 10.3390/ph13110385] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 01/10/2023] Open
Abstract
LKB1 is frequently mutated in non-small cell lung cancer (NSCLC). LKB1-mutated NSCLCs often have a dismal prognosis and receive lower benefit from the currently available therapies. LKB1 acts as a cell emergency brake in low-energy conditions, by modulating the activity of crucial anabolic enzymes. Thus, loss of LKB1 activity leads to the enhancement of tumor cell proliferation also under conditions of energy shortage. This unrestrained growth may be exploited as an Achilles heel in NSCLC, i.e., by inhibiting mitochondrial respiration. Recently, clinical trials have started to investigate the efficacy of metabolism-based treatments in NSCLCs. To date, enrollment of patients within these trials is based on LKB1 loss of function status, defined by mutation in the gene or by complete absence of immunohistochemical staining. However, LKB1 impairment could be the consequence of epigenetic regulations that partially or completely abrogate protein expression. These epigenetic regulations result in LKB1 wild-type tumors with aggressiveness and vulnerabilities similar to those of LKB1-mutated ones. In this review, we introduced the definition of the “LKB1less phenotype”, and we summarized all currently known features linked to this status, in order to optimize selection and treatment of NSCLC patients with impaired LKB1 function.
Collapse
|
11
|
Granado-Martínez P, Garcia-Ortega S, González-Sánchez E, McGrail K, Selgas R, Grueso J, Gil R, Naldaiz-Gastesi N, Rhodes AC, Hernandez-Losa J, Ferrer B, Canals F, Villanueva J, Méndez O, Espinosa-Gil S, Lizcano JM, Muñoz-Couselo E, García-Patos V, Recio JA. STK11 (LKB1) missense somatic mutant isoforms promote tumor growth, motility and inflammation. Commun Biol 2020; 3:366. [PMID: 32647375 PMCID: PMC7347935 DOI: 10.1038/s42003-020-1092-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Elucidating the contribution of somatic mutations to cancer is essential for personalized medicine. STK11 (LKB1) appears to be inactivated in human cancer. However, somatic missense mutations also occur, and the role/s of these alterations to this disease remain unknown. Here, we investigated the contribution of four missense LKB1 somatic mutations in tumor biology. Three out of the four mutants lost their tumor suppressor capabilities and showed deficient kinase activity. The remaining mutant retained the enzymatic activity of wild type LKB1, but induced increased cell motility. Mechanistically, LKB1 mutants resulted in differential gene expression of genes encoding vesicle trafficking regulating molecules, adhesion molecules and cytokines. The differentially regulated genes correlated with protein networks identified through comparative secretome analysis. Notably, three mutant isoforms promoted tumor growth, and one induced inflammation-like features together with dysregulated levels of cytokines. These findings uncover oncogenic roles of LKB1 somatic mutations, and will aid in further understanding their contributions to cancer development and progression. Paula Granado-Martínez, Sara Ortega, Elena González-Sánchez et al. report a functional analysis of four cancer-associated mutant isoforms of the gene STK11 using cell-based and animal models. They find the mutant isoforms no longer show tumor suppressor activity, promote tumor growth, and affect the regulation of cytokines and genes involved in vesicle trafficking.
Collapse
Affiliation(s)
- Paula Granado-Martínez
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Sara Garcia-Ortega
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Elena González-Sánchez
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Kimberley McGrail
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Rafael Selgas
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Judit Grueso
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Experimental Therapeutics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Rosa Gil
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Neia Naldaiz-Gastesi
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Biodonostia, Neurosciences Area, Group of Neuromuscular Diseases, San Sebastian, 20014, Spain
| | - Ana C Rhodes
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Barcelona Clinic Liver Cancer (BCLC) Group, Liver Unit, Hospital Clínic of Barcelona, University of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, 08036, Spain
| | - Javier Hernandez-Losa
- Anatomy Pathology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Berta Ferrer
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Anatomy Pathology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Francesc Canals
- Proteomics Laboratory, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Josep Villanueva
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Olga Méndez
- Preclinical Research Program, Vall d'Hebron Institute of Oncology (VHIO, Barcelona, 08035, Spain
| | - Sergio Espinosa-Gil
- Protein Kinases and Signal Transduction Laboratory, Neuroscience Institute and Molecular Biology and Biochemistry Department, UAB, Bellaterra, Barcelona, 08193, Spain
| | - José M Lizcano
- Protein Kinases and Signal Transduction Laboratory, Neuroscience Institute and Molecular Biology and Biochemistry Department, UAB, Bellaterra, Barcelona, 08193, Spain
| | - Eva Muñoz-Couselo
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Clinical Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital, Barcelona-UAB, Barcelona, 08035, Spain
| | - Vicenç García-Patos
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.,Dermatology Department, Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain
| | - Juan A Recio
- Biomedical Research in Melanoma-Animal Models and Cancer Laboratory- Vall d'Hebron Research Institute VHIR-Vall d'Hebron Hospital Barcelona-UAB, Barcelona, 08035, Spain.
| |
Collapse
|
12
|
Jossin Y. Molecular mechanisms of cell polarity in a range of model systems and in migrating neurons. Mol Cell Neurosci 2020; 106:103503. [PMID: 32485296 DOI: 10.1016/j.mcn.2020.103503] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 04/20/2020] [Accepted: 05/23/2020] [Indexed: 01/09/2023] Open
Abstract
Cell polarity is defined as the asymmetric distribution of cellular components along an axis. Most cells, from the simplest single-cell organisms to highly specialized mammalian cells, are polarized and use similar mechanisms to generate and maintain polarity. Cell polarity is important for cells to migrate, form tissues, and coordinate activities. During development of the mammalian cerebral cortex, cell polarity is essential for neurogenesis and for the migration of newborn but as-yet undifferentiated neurons. These oriented migrations include both the radial migration of excitatory projection neurons and the tangential migration of inhibitory interneurons. In this review, I will first describe the development of the cerebral cortex, as revealed at the cellular level. I will then define the core molecular mechanisms - the Par/Crb/Scrib polarity complexes, small GTPases, the actin and microtubule cytoskeletons, and phosphoinositides/PI3K signaling - that are required for asymmetric cell division, apico-basal and front-rear polarity in model systems, including C elegans zygote, Drosophila embryos and cultured mammalian cells. As I go through each core mechanism I will explain what is known about its importance in radial and tangential migration in the developing mammalian cerebral cortex.
Collapse
Affiliation(s)
- Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium.
| |
Collapse
|
13
|
Krahn MP. Phospholipids of the Plasma Membrane - Regulators or Consequence of Cell Polarity? Front Cell Dev Biol 2020; 8:277. [PMID: 32411703 PMCID: PMC7198698 DOI: 10.3389/fcell.2020.00277] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 03/31/2020] [Indexed: 12/15/2022] Open
Abstract
Cell polarity is a key feature of many eukaryotic cells, including neurons, epithelia, endothelia and asymmetrically dividing stem cells. Apart from the specific localization of proteins to distinct domains of the plasma membrane, most of these cells exhibit an asymmetric distribution of phospholipids within the plasma membrane too. Notably, research over the last years has revealed that many known conserved regulators of apical-basal polarity in epithelial cells are capable of binding to phospholipids, which in turn regulate the localization and to some extent the function of these proteins. Conversely, phospholipid-modifying enzymes are recruited and controlled by polarity regulators, demonstrating an elaborated balance between asymmetrically localized proteins and phospholipids, which are enriched in certain (micro)domains of the plasma membrane. In this review, we will focus on our current understanding of apical-basal polarity and the implication of phospholipids within the plasma membrane during the cell polarization of epithelia and migrating cells.
Collapse
Affiliation(s)
- Michael P. Krahn
- Department of Medical Cell Biology, Medical Clinic D, University Hospital of Münster, Münster, Germany
| |
Collapse
|
14
|
Chen D, Yang L, Chen X, Zhang X, Liu Y, Guo Z, Zhang LW. Automated contour analysis of multi-cellular spheroids spreading through high content imaging. Phys Biol 2018; 15:026006. [PMID: 29251623 DOI: 10.1088/1478-3975/aaa27b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The morphology of 2D cell colonies has been studied to understand tumor metastasis in the past decades. However, 2D cell cultures are lacking many features of 3D tissues, and their physiological behaviors are quite different from solid tumors in vivo. In this work, we studied the multi-cellular tumor spheroid (MCTS) spreading on the substrate, which keeps parts of 3D tissue characteristics and facilitates cell tracking through 2D imaging. By using a high content imaging system (HCS), we tracked multiple spheroids in one single 96-well plate for 36 h. An automated algorithm based on Otsu's method was developed to investigate the morphological details of spheroids through the quantification of radius length and its coefficients of variation. Spheroid spreading is altered by the PIP-platin, which was a novel platinum based drug previously reported by us with an inhibitory effect on cell migration. All parameters showed dose dependent decreases when PIP-platin concentration increased, indicating the inhibition of spheroid expansion by this compound. To investigate the surface roughness of spheroids affected by the drug, we applied the Fourier parameter β and the normalized standard deviation of the radius STD r / [Formula: see text], which were found inversely proportional to the concentrations of PIP-platin. Particularly at the low drug concentrations, the indices of contour roughness appeared to be more sensitive than spheroid sizes, which could be the potential morphological markers for high content screening of drugs.
Collapse
Affiliation(s)
- Dandan Chen
- School of Radiation Medicine and Protection, Medical College, Soochow University, Suzhou, Jiangsu 215006, People's Republic of China. Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Suzhou, Jiangsu 215006, People's Republic of China. Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Suzhou, Jiangsu 215006, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
15
|
Zhang C, Xiao X, Chen M, Aldharee H, Chen Y, Long W. Liver kinase B1 restoration promotes exosome secretion and motility of lung cancer cells. Oncol Rep 2017; 39:376-382. [PMID: 29138862 PMCID: PMC5783601 DOI: 10.3892/or.2017.6085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022] Open
Abstract
Liver kinase B1 (LKB1) regulates a variety of cellular functions, including cell polarity, energy metabolism and cell growth, by targeting multiple signaling pathways such as AMPK/mTOR and p53. LKB1 functions as a tumor suppressor in sporadic cancers including lung cancer. Extracellular vesicles such as exosomes secreted by cancer cells modulate the tumor microenvironment and progression by targeting both tumor cells (autocrine actions) and other types of cells associated with tumors (paracrine actions). While the roles of LKB1 in cellular signaling in general is well-studied, its specific role in exosome-mediated signaling remains to be explored. To this purpose, we reintroduced LKB1 into H460 and A549 lung cancer cells that are endogenously deficient in LKB1 expression. Notably, we found that while restoration of LKB1 significantly reduced lung cancer cell growth as expected, it greatly promoted cell motility and enhanced the release of exosomes. In addition, exosomes isolated from H460 cells with stable restoration of LKB1 had much higher ability in stimulating lung cancer cell migration than did those from H460 cells lacking LKB1. Mechanistically, restoration of LKB1 in H460 cells inhibited cellular expression and exosomal secretion of migration-suppressing microRNAs (miRNAs), including miR-125a, miR-126 and let7b. Taken together, the present study revealed a new role for LKB1 in promoting cell motility by downregulating migration-suppressing miRNA expression and exosome secretion.
Collapse
Affiliation(s)
- Cheng Zhang
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| | - Xiang Xiao
- Department of Pharmacology and Toxicology, Wright State University, Dayton, OH 45435, USA
| | - Minyi Chen
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| | - Hitham Aldharee
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| | - Yanfang Chen
- Department of Pharmacology and Toxicology, Wright State University, Dayton, OH 45435, USA
| | - Weiwen Long
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH 45435, USA
| |
Collapse
|
16
|
Thaiparambil J, Mansour O, El-Zein R. Effect of Benzo[a]Pyrene on Spindle Misorientation and Fidelity of Chromosome Segregation in Lung Epithelial BEAS-2B Cells. Toxicol Sci 2017; 162:167-176. [DOI: 10.1093/toxsci/kfx229] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Jose Thaiparambil
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas
| | - Oula Mansour
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas
| | - Randa El-Zein
- Department of Radiology, Houston Methodist Research Institute, Houston, Texas
| |
Collapse
|
17
|
Cell Division Cycle 42 plays a Cell type-Specific role in Lung Tumorigenesis. Sci Rep 2017; 7:10407. [PMID: 28871124 PMCID: PMC5583260 DOI: 10.1038/s41598-017-10891-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/15/2017] [Indexed: 12/17/2022] Open
Abstract
Cell division cycle 42 (CDC42) plays important roles in polarity establishment and maintenance as well as cell cycle progression and cell division. Although disruption of cell polarity is a prerequisite in epithelial tumor initiation, the roles of CDC42 in tumorigenesis are still poorly understood. Here we find that Cdc42 deficiency inhibits the KrasG12D-induced lung alveoli tumor formation, while conversely promotes bronchiole tumor formation in mice. Bronchial Cdc42 loss destroys contact inhibition potentially through cell polarity disruption, and results in increased tumor formation. In contrast, deletion of Cdc42 in alveoli cells prevents KrasG12D-induced cell proliferation, which leads to reduced tumor formation. Further analyses of clinical specimens uncover a significant positive correlation between CDC42 and type II alveolar epithelial cells marker SP-A, indicating the potential importance of CDC42 in this specific subset of lung cancer. Collectively, we identify the lineage-specific function of CDC42 in lung tumorigenesis potentially through the regulation of cell polarity integrity.
Collapse
|
18
|
Coordinated cell motility is regulated by a combination of LKB1 farnesylation and kinase activity. Sci Rep 2017; 7:40929. [PMID: 28102310 PMCID: PMC5244416 DOI: 10.1038/srep40929] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 12/14/2016] [Indexed: 01/07/2023] Open
Abstract
Cell motility requires the precise coordination of cell polarization, lamellipodia formation, adhesion, and force generation. LKB1 is a multi-functional serine/threonine kinase that associates with actin at the cellular leading edge of motile cells and suppresses FAK. We sought to understand how LKB1 coordinates these multiple events by systematically dissecting LKB1 protein domain function in combination with live cell imaging and computational approaches. We show that LKB1-actin colocalization is dependent upon LKB1 farnesylation leading to RhoA-ROCK-mediated stress fiber formation, but membrane dynamics is reliant on LKB1 kinase activity. We propose that LKB1 kinase activity controls membrane dynamics through FAK since loss of LKB1 kinase activity results in morphologically defective nascent adhesion sites. In contrast, defective farnesylation mislocalizes nascent adhesion sites, suggesting that LKB1 farnesylation serves as a targeting mechanism for properly localizing adhesion sites during cell motility. Together, we propose a model where coordination of LKB1 farnesylation and kinase activity serve as a multi-step mechanism to coordinate cell motility during migration.
Collapse
|
19
|
Okon IS, Ding Y, Coughlan KA, Wang Q, Song P, Benbrook DM, Zou MH. Aberrant NRP-1 expression serves as predicator of metastatic endometrial and lung cancers. Oncotarget 2016; 7:7970-8. [PMID: 26701889 PMCID: PMC4884968 DOI: 10.18632/oncotarget.6699] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Accepted: 11/25/2015] [Indexed: 01/13/2023] Open
Abstract
Neuropilin-1 (NRP-1) has emerged as an important driver of tumor-promoting phenotypes of human malignancies. However, incomplete knowledge exists as to how this single-pass transmembrane receptor mediates pleiotropic tumor-promoting functions. The purpose of this study was to evaluate NRP-1 expression and metastatic properties in 94 endometrial cancer and matching serum specimens and in a lung cancer cell line. We found that NRP-1 expression significantly correlated with increased tumoral expression of vascular endothelial growth factor 2 (VEGFR2) and serum levels of hepatocyte growth factor (HGF) and cell growth-stimulating factor (C-GSF). Tumoral NRP-1 also was positively associated with expression of NEDD9, a pro-metastatic protein. In the highly metastatic lung cancer cell line (H1792), stable LKB1 depletion caused increased migration in vitro and accentuated NRP-1 and NEDD9 expression in vivo. Our findings demonstrate that perturbed expression of these targets correlate with metastatic potential of endometrial and lung tumors, providing clinically-relevant biomarker applications for diagnostic and therapeutic targeting.
Collapse
Affiliation(s)
- Imoh S Okon
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | - Ye Ding
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | | | - Qiongxin Wang
- Section of Molecular Medicine, Oklahoma City, OK 73104, USA
| | - Ping Song
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| | - Doris M Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center (OUHSC), Oklahoma City, OK 73104, USA
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA 30302, USA
| |
Collapse
|
20
|
Cunniff B, McKenzie AJ, Heintz NH, Howe AK. AMPK activity regulates trafficking of mitochondria to the leading edge during cell migration and matrix invasion. Mol Biol Cell 2016; 27:2662-74. [PMID: 27385336 PMCID: PMC5007087 DOI: 10.1091/mbc.e16-05-0286] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 06/26/2016] [Indexed: 01/06/2023] Open
Abstract
Mitochondria infiltrate leading edge lamellipodia, increasing local mitochondrial mass and relative ATP concentration. AMPK regulates infiltration of mitochondria into the leading edge of 2D lamellipodia and 3D invadopodia, coupling local metabolic sensing to subcellular targeting of mitochondria during cell movement. Cell migration is a complex behavior involving many energy-expensive biochemical events that iteratively alter cell shape and location. Mitochondria, the principal producers of cellular ATP, are dynamic organelles that fuse, divide, and relocate to respond to cellular metabolic demands. Using ovarian cancer cells as a model, we show that mitochondria actively infiltrate leading edge lamellipodia, thereby increasing local mitochondrial mass and relative ATP concentration and supporting a localized reversal of the Warburg shift toward aerobic glycolysis. This correlates with increased pseudopodial activity of the AMP-activated protein kinase (AMPK), a critically important cellular energy sensor and metabolic regulator. Furthermore, localized pharmacological activation of AMPK increases leading edge mitochondrial flux, ATP content, and cytoskeletal dynamics, whereas optogenetic inhibition of AMPK halts mitochondrial trafficking during both migration and the invasion of three-dimensional extracellular matrix. These observations indicate that AMPK couples local energy demands to subcellular targeting of mitochondria during cell migration and invasion.
Collapse
Affiliation(s)
- Brian Cunniff
- Department of Pathology, University of Vermont, Burlington, VT 05405 University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Andrew J McKenzie
- University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405 Department of Pharmacology, University of Vermont, Burlington, VT 05405
| | - Nicholas H Heintz
- Department of Pathology, University of Vermont, Burlington, VT 05405 University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405
| | - Alan K Howe
- University of Vermont Cancer Center, University of Vermont, Burlington, VT 05405 Department of Pharmacology, University of Vermont, Burlington, VT 05405
| |
Collapse
|
21
|
Gandalovičová A, Vomastek T, Rosel D, Brábek J. Cell polarity signaling in the plasticity of cancer cell invasiveness. Oncotarget 2016; 7:25022-49. [PMID: 26872368 PMCID: PMC5041887 DOI: 10.18632/oncotarget.7214] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Accepted: 01/29/2016] [Indexed: 02/07/2023] Open
Abstract
Apico-basal polarity is typical of cells present in differentiated epithelium while front-rear polarity develops in motile cells. In cancer development, the transition from epithelial to migratory polarity may be seen as the hallmark of cancer progression to an invasive and metastatic disease. Despite the morphological and functional dissimilarity, both epithelial and migratory polarity are controlled by a common set of polarity complexes Par, Scribble and Crumbs, phosphoinositides, and small Rho GTPases Rac, Rho and Cdc42. In epithelial tissues, their mutual interplay ensures apico-basal and planar cell polarity. Accordingly, altered functions of these polarity determinants lead to disrupted cell-cell adhesions, cytoskeleton rearrangements and overall loss of epithelial homeostasis. Polarity proteins are further engaged in diverse interactions that promote the establishment of front-rear polarity, and they help cancer cells to adopt different invasion modes. Invading cancer cells can employ either the collective, mesenchymal or amoeboid invasion modes or actively switch between them and gain intermediate phenotypes. Elucidation of the role of polarity proteins during these invasion modes and the associated transitions is a necessary step towards understanding the complex problem of metastasis. In this review we summarize the current knowledge of the role of cell polarity signaling in the plasticity of cancer cell invasiveness.
Collapse
Affiliation(s)
- Aneta Gandalovičová
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Tomáš Vomastek
- Institute of Microbiology, Academy of Sciences of The Czech Republic, Videňská, Prague, Czech Republic
| | - Daniel Rosel
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| | - Jan Brábek
- Department of Cell Biology, Charles University in Prague, Viničná, Prague, Czech Republic
| |
Collapse
|
22
|
Bonastre E, Brambilla E, Sanchez-Cespedes M. Cell adhesion and polarity in squamous cell carcinoma of the lung. J Pathol 2016; 238:606-16. [PMID: 26749265 DOI: 10.1002/path.4686] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 12/18/2015] [Accepted: 12/30/2015] [Indexed: 01/01/2023]
Abstract
Lung cancer is a deadly disease that can roughly be classified into three histopathological groups: lung adenocarcinomas, lung squamous cell carcinomas (LSCCs), and small cell carcinomas. These types of lung cancer are molecularly, phenotypically, and regionally diverse neoplasms, reflecting differences in their cells of origin. LSCCs commonly arise in the airway epithelium of a main or lobar bronchus, which is an important line of defence against the external environment. Furthermore, most LSCCs are characterized histopathologically by the presence of keratinization and/or intercellular bridges, consistent with the molecular features of these tumours, characterized by high levels of transcripts encoding keratins and proteins relevant to intercellular junctions and cell polarity. In this review, the relationships between the molecular features of LSCCs and the types of cell and epithelia of origin are discussed. Recurrent alterations in genes involved in intercellular adhesion and cell polarity in LSCCs are also reviewed, emphasizing the importance of the disruption of PAR3 and the PAR complex. Finally, the possible functional effects of these alterations on epithelial homeostasis, and how they contribute to the development of LSCC, are discussed.
Collapse
Affiliation(s)
- Ester Bonastre
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| | - Elisabeth Brambilla
- Department of Pathology, Institut Albert Bonniot, INSERM U823, University Joseph Fourier, CHU, Grenoble Hopital Michallon, Grenoble, France
| | - Montse Sanchez-Cespedes
- Genes and Cancer Group, Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
23
|
Konen J, Wilkinson S, Lee B, Fu H, Zhou W, Jiang Y, Marcus AI. LKB1 kinase-dependent and -independent defects disrupt polarity and adhesion signaling to drive collagen remodeling during invasion. Mol Biol Cell 2016; 27:1069-84. [PMID: 26864623 PMCID: PMC4814216 DOI: 10.1091/mbc.e15-08-0569] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/05/2016] [Indexed: 12/23/2022] Open
Abstract
LKB1 is a serine/threonine kinase and a commonly mutated gene in lung adenocarcinoma. The majority of LKB1 mutations are truncations that disrupt its kinase activity and remove its C-terminal domain (CTD). Because LKB1 inactivation drives cancer metastasis in mice and leads to aberrant cell invasion in vitro, we sought to determine how compromised LKB1 function affects lung cancer cell polarity and invasion. Using three-dimensional models, we show that LKB1 kinase activity is essential for focal adhesion kinase-mediated cell adhesion and subsequent collagen remodeling but not cell polarity. Instead, cell polarity is overseen by the kinase-independent function of its CTD and more specifically its farnesylation. This occurs through a mesenchymal-amoeboid morphological switch that signals through the Rho-GTPase RhoA. These data suggest that a combination of kinase-dependent and -independent defects by LKB1 inactivation creates a uniquely invasive cell with aberrant polarity and adhesion signaling that drives invasion into the microenvironment.
Collapse
Affiliation(s)
- Jessica Konen
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322 Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322
| | - Scott Wilkinson
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322 Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322
| | - Byoungkoo Lee
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30302
| | - Haian Fu
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322 Department of Pharmacology, Emory University, Atlanta, GA 30322
| | - Wei Zhou
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30302
| | - Adam I Marcus
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322
| |
Collapse
|
24
|
Gomez-Muñoz A, Presa N, Gomez-Larrauri A, Rivera IG, Trueba M, Ordoñez M. Control of inflammatory responses by ceramide, sphingosine 1-phosphate and ceramide 1-phosphate. Prog Lipid Res 2015; 61:51-62. [PMID: 26703189 DOI: 10.1016/j.plipres.2015.09.002] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 09/14/2015] [Accepted: 09/28/2015] [Indexed: 01/04/2023]
Abstract
Inflammation is a network of complex processes involving a variety of metabolic and signaling pathways aiming at healing and repairing damage tissue, or fighting infection. However, inflammation can be detrimental when it becomes out of control. Inflammatory mediators involve cytokines, bioactive lipids and lipid-derived metabolites. In particular, the simple sphingolipids ceramides, sphingosine 1-phosphate, and ceramide 1-phosphate have been widely implicated in inflammation. However, although ceramide 1-phosphate was first described as pro-inflammatory, recent studies show that it has anti-inflammatory properties when produced in specific cell types or tissues. The biological functions of ceramides and sphingosine 1-phosphate have been extensively studied. These sphingolipids have opposing effects with ceramides being potent inducers of cell cycle arrest and apoptosis, and sphingosine 1-phosphate promoting cell growth and survival. However, the biological actions of ceramide 1-phosphate have only been partially described. Ceramide 1-phosphate is mitogenic and anti-apoptotic, and more recently, it has been demonstrated to be key regulator of cell migration. Both sphingosine 1-phosphate and ceramide 1-phosphate are also implicated in tumor growth and dissemination. The present review highlights new aspects on the control of inflammation and cell migration by simple sphingolipids, with special emphasis to the role played by ceramide 1-phosphate in controlling these actions.
Collapse
Affiliation(s)
- Antonio Gomez-Muñoz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| | - Natalia Presa
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| | - Ana Gomez-Larrauri
- Department of Pneumology, University Hospital of Alava (Osakidetza), Vitoria-Gasteiz, Spain.
| | - Io-Guané Rivera
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| | - Miguel Trueba
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| | - Marta Ordoñez
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, University of the Basque Country (UPV/EHU), P.O. Box 644, 48080 Bilbao, Spain.
| |
Collapse
|
25
|
JIANG SHANSHAN, CHEN RENSHENG, YU JIANAN, LI NIANSHUANG, KE RONG, LUO LINGYU, ZOU JUNRONG, ZHANG JIANGNAN, ZHANG KUNHE, LU NONGHUA, HUANG DEQIANG. Clinical significance and role of LKB1 in gastric cancer. Mol Med Rep 2015; 13:249-56. [DOI: 10.3892/mmr.2015.4508] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 08/07/2015] [Indexed: 11/06/2022] Open
|
26
|
PAK2 is an effector of TSC1/2 signaling independent of mTOR and a potential therapeutic target for Tuberous Sclerosis Complex. Sci Rep 2015; 5:14534. [PMID: 26412398 PMCID: PMC4585940 DOI: 10.1038/srep14534] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 07/22/2015] [Indexed: 11/22/2022] Open
Abstract
Tuberous sclerosis complex (TSC) is caused by inactivating mutations in either TSC1 or TSC2 and is characterized by uncontrolled mTORC1 activation. Drugs that reduce mTOR activity are only partially successful in the treatment of TSC, suggesting that mTOR-independent pathways play a role in disease development. Here, kinome profiles of wild-type and Tsc2−/− mouse embryonic fibroblasts (MEFs) were generated, revealing a prominent role for PAK2 in signal transduction downstream of TSC1/2. Further investigation showed that the effect of the TSC1/2 complex on PAK2 is mediated through RHEB, but is independent of mTOR and p21RAC. We also demonstrated that PAK2 over-activation is likely responsible for the migratory and cell cycle abnormalities observed in Tsc2−/− MEFs. Finally, we detected high levels of PAK2 activation in giant cells in the brains of TSC patients. These results show that PAK2 is a direct effector of TSC1-TSC2-RHEB signaling and a new target for rational drug therapy in TSC.
Collapse
|
27
|
Guo ZM, Li W, Zhao XH. Purine-rich element binding protein alpha promotes invasion and migration of esophageal squamous cell carcinoma KYSE 510 cells by inducing epithelial-mesenchymal transition. Shijie Huaren Xiaohua Zazhi 2014; 22:5579-5586. [DOI: 10.11569/wcjd.v22.i36.5579] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of purine-rich element binding protein alpha (PURα) in the invasion and migration of esophageal squamous cell carcinoma (ESCC) KYSE 510 cells and the underlying mechanisms.
METHODS: An ESCC cell line overexpressing PURα (KYSE 510-PURα) was established, and the expression levels of epithelial-mesenchymal transition associated proteins were determined by comparing with control cells transfected with an empty vector of pCMV6 (KYSE 510-pCMV6). The expression of E-cadherin and vimentin was analyzed by Western blot and immunofluorescent staining. The capabilities of invasion and migration of cancer cells were assessed via transwell and wound healing assays.
RESULTS: PURα was overexpressed in KYSE 510 cells transfected with the pCMV6- PURα vector. The expression of E-cadherin was reduced, and that of Vimentin, N-cadherin and Snail was increased in KYSE 510-PURα cells. Most strikingly, the cell morphology was changed as fibroblasts and the abilities of migration and invasion were altered.
CONCLUSION: Our data suggest that regulation of PURα expression in ESCC cells may induce esophageal epithelial-mesenchymal transition.
Collapse
|
28
|
Li X, Truty MA, Kang Y, Chopin-Laly X, Zhang R, Roife D, Chatterjee D, Lin E, Thomas RM, Wang H, Katz MH, Fleming JB. Extracellular lumican inhibits pancreatic cancer cell growth and is associated with prolonged survival after surgery. Clin Cancer Res 2014; 20:6529-40. [PMID: 25336691 PMCID: PMC4268437 DOI: 10.1158/1078-0432.ccr-14-0970] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE To evaluate the relevance between lumican expression patterns and the clinical course of patients with pancreatic ductal adenocarcinoma (PDAC), and to investigate the role of lumican in PDAC progression. EXPERIMENTAL DESIGN One hundred thirty-one patient tumors were chosen for tissue microarray staining, and Cox regression analysis was used to test the associations between lumican expression and clinical, pathologic, and oncologic outcomes in all patients. Primary PDAC cells and recombinant human lumican protein were used to establish a working model to mimic the in vivo interactions between stromal lumican and PDAC cells. Using this model, we tested the effects of lumican on EGFR signaling via Akt and hypoxia-inducible factor-1α (HIF1α) and its subsequent influence on glucose consumption, lactate production, intracellular ATP, and apoptotic cell death. RESULTS Lumican was present in the stroma surrounding PDAC cells in roughly one-half of primary tumors and the direct xenografts. Patients with stromal lumican were associated with a profound reduction in metastatic recurrence after surgery and 3-fold longer survival than patients without stromal lumican. In PDAC cells, extracellular lumican reduced EGFR expression and phosphorylation through enhanced dimerization and internalization of EGFR and the resultant inhibition of Akt kinase activity. Lumican also reduced HIF1α expression and activity via Akt. PDAC cells with enhanced HIF1α activity were resistant to lumican-induced inhibition of glucose consumption, lactate production, intracellular ATP, and apoptosis. CONCLUSIONS There is a positive association between stromal lumican in primary PDAC tumors and prolonged survival after tumor resection. Lumican plays a restrictive role in EGFR-expressing pancreatic cancer progression.
Collapse
Affiliation(s)
- Xinqun Li
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mark A Truty
- Department of Surgery, Mayo Clinic, Rochester, Minnesota
| | - Ya'an Kang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xavier Chopin-Laly
- Department of Hepato-Biliary and Pancreatic Surgery, Edouard Herriot Hospital, HCL, Lyon, France
| | - Ran Zhang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David Roife
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deyali Chatterjee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - E Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ryan M Thomas
- Department of Surgery, University of Florida, Gainesville, Florida
| | - Huamin Wang
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Matthew H Katz
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jason B Fleming
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
29
|
Ye H, Zhang Y, Geng L, Li Z. Cdc42 expression in cervical cancer and its effects on cervical tumor invasion and migration. Int J Oncol 2014; 46:757-63. [PMID: 25394485 DOI: 10.3892/ijo.2014.2748] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/23/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to examine Cdc42 expression in cervical cancer and explore its effects on invasion and migration capability of cervical cancer cells. Immunohistochemistry was used to detect Cdc42 expression in normal cervical tissues as well as CIN I or below, CIN II or above, and cervical cancer tissues. Western blot analysis was used to explore Cdc42 expression in normal cervical cell line Crl-2614 and cervical cancer cell line HeLa. Plasmids of constitutively active Cdc42 (Cdc42 CA), wild-type Cdc42 (Cdc42 WT) and dominant negative Cdc42 (Cdc42 DN) were transfected, respectively, into HeLa cells to investigate the impacts of Cdc42 on migration and invasion of cervical cancer cells using Transwell and on cytoskeleton microfilaments using confocal microscopy after immunofluorescence staining. Cdc42 expression was gradually increased in the order of cervical tissues with CIN I or below, CIN II or above and cancer, showing significant difference (P<0.05), and was significantly higher in HeLa cells than in Crl-2614 cells (P<0.05). Migration ability of HeLa cells transfected with Cdc42 CA was significantly higher than that of non-transfected, as well as Cdc42 WT- or Cdc42 DN-transfected HeLa cells (P<0.05). Overexpression of Cdc42 CA can promote filopodia formation in HeLa cells. We concluded that Cdc42 overexpression significantly improved the ability of cervical cancer cells to migrate possibly due to improved pseudopodia formation.
Collapse
Affiliation(s)
- Hongnan Ye
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Youyi Zhang
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Ministry of Health, Beijing 100191, P.R. China
| | - Li Geng
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing 100191, P.R. China
| | - Zijian Li
- Institute of Vascular Medicine, Peking University Third Hospital and Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptide, Ministry of Health, Beijing 100191, P.R. China
| |
Collapse
|
30
|
Xu HG, Zhai YX, Chen J, Lu Y, Wang JW, Quan CS, Zhao RX, Xiao X, He Q, Werle KD, Kim HG, Lopez R, Cui R, Liang J, Li YL, Xu ZX. LKB1 reduces ROS-mediated cell damage via activation of p38. Oncogene 2014; 34:3848-59. [PMID: 25263448 PMCID: PMC4377312 DOI: 10.1038/onc.2014.315] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 08/05/2014] [Accepted: 08/06/2014] [Indexed: 12/27/2022]
Abstract
Liver kinase B1 (LKB1, also known as serine/threonine kinase 11, STK11) is a tumor suppressor mutated in Peutz-Jeghers syndrome and in a variety of sporadic cancers. Herein, we demonstrate that LKB1 controls the levels of intracellular reactive oxygen species (ROS) and protects the genome from oxidative damage. Cells lacking LKB1 exhibit markedly increased intracellular ROS levels, excessive oxidation of DNA, increased mutation rates, and accumulation of DNA damage, which are effectively prevented by ectopic expression of LKB1 and by incubation with antioxidant N-acetylcysteine (NAC). The role of LKB1 in suppressing ROS is independent of AMPK, a canonical substrate of LKB1. Instead, under the elevated ROS, LKB1 binds to and maintains the activity of cdc42-PAK1 (p21 activated kinase 1) complex, which triggers the activation of p38 and its downstream signaling targets, such as ATF-2, thereby enhancing the activity of SOD-2 and catalase, two antioxidant enzymes that protect the cells from ROS accumulation, DNA damage, and loss of viability. Our results provide a new paradigm for a non-canonical tumor suppressor function of LKB1 and highlight the importance of targeting ROS signaling as a potential therapeutic strategy for cancer cells lacking LKB1.
Collapse
Affiliation(s)
- H-G Xu
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Y-X Zhai
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - J Chen
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Y Lu
- Department of Endocrinology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - J-W Wang
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - C-S Quan
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - R-X Zhao
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - X Xiao
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Q He
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - K D Werle
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - H-G Kim
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - R Lopez
- Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - R Cui
- Department of Dermatology, Boston University, School of Medicine, Boston, MA, USA
| | - J Liang
- Department of Systems Biology, UT MD Anderson Cancer Center, Houston, TX, USA
| | - Y-L Li
- Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Z-X Xu
- 1] Division of Hematology and Oncology, Department of Medicine, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA [2] Key Laboratory of Pathobiology, Ministry of Education, Department of Pathology, Norman Bethune College of Medicine, Jilin University, Changchun, China
| |
Collapse
|
31
|
Okon IS, Coughlan KA, Zhang C, Moriasi C, Ding Y, Song P, Zhang W, Li G, Zou MH. Protein kinase LKB1 promotes RAB7-mediated neuropilin-1 degradation to inhibit angiogenesis. J Clin Invest 2014; 124:4590-602. [PMID: 25180605 DOI: 10.1172/jci75371] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 07/25/2014] [Indexed: 12/16/2022] Open
Abstract
After internalization, transmembrane receptors (TMRs) are typically recycled back to the cell surface or targeted for degradation. Efficient TMR trafficking is critical for regulation of several processes, including signal transduction pathways, development, and disease. Here, we determined that trafficking of the angiogenic receptor neuropilin-1 (NRP-1) is abrogated by the liver kinase B1 (LKB1), a serine-threonine kinase of the calcium calmodulin family. We found that aberrant NRP-1 expression in tumor cells from patients with lung adenocarcinoma is associated with decreased levels of LKB1. In cultured lung cells, LKB1 accentuated formation of a complex between NRP-1 and RAB7 in late endosomes. LKB1 specifically bound GTP-bound RAB7, but not a dominant-negative GDP-bound form of RAB7, promoting rapid transfer and lysosome degradation of NRP-1. siRNA-mediated depletion of RAB7 disrupted the transfer of NRP-1 to the lysosome, resulting in recovery of the receptor as well as increased tumor growth and angiogenesis. Together, our findings indicate that LKB1 functions as a RAB7 effector and suppresses angiogenesis by promoting the cellular trafficking of NRP-1 from RAB7 vesicles to the lysosome for degradation. Furthermore, these data suggest that LKB1 and NRP-1 have potential as therapeutic targets for limiting tumorigenesis.
Collapse
|
32
|
Lu C, Xiong M, Luo Y, Li J, Zhang Y, Dong Y, Zhu Y, Niu T, Wang Z, Duan L. Genome-wide transcriptional analysis of apoptosis-related genes and pathways regulated by H2AX in lung cancer A549 cells. Apoptosis 2014; 18:1039-47. [PMID: 23793869 DOI: 10.1007/s10495-013-0875-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Histone H2AX is a novel tumor suppressor protein and plays an important role in apoptosis of cancer cells. However, the role of H2AX in lung cancer cells is unclear. The detailed mechanism and epigenetic regulation by H2AX remain elusive in cancer cells. We showed that H2AX was involved in apoptosis of lung cancer A549 cells as in other tumor cells. Knockdown of H2AX strongly suppressed apoptosis of A549 cells. We clarified the molecular mechanisms of apoptosis regulated by H2AX based on genome-wide transcriptional analysis. Microarray data analysis demonstrated that H2AX knockdown in A549 cells affected expression of 3,461 genes, including upregulation of 1,435 and downregulation of 2,026. These differentially expressed genes were subjected to bioinformatic analysis for exploring biological processes regulated by H2AX in lung cancer cells. Gene ontology analysis showed that H2AX affected expression of many genes, through which, many important functions including response to stimuli, gene expression, and apoptosis were involved in apoptotic regulation of lung cancer cells. Pathway analysis identified the mitogen-activated protein kinase signaling pathway and apoptosis as the most important pathways targeted by H2AX. Signal transduction pathway networks analysis and chromatin immunoprecipitation assay showed that two core genes, NFKB1 and JUN, were involved in apoptosis regulated by H2AX in lung cancer cells. Taken together, these data provide compelling clues for further exploration of H2AX function in cancer cells.
Collapse
Affiliation(s)
- Chengrong Lu
- Aviation Medicine Research Laboratory, Air Force General Hospital, PLA, Beijing, 100142, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Okon IS, Coughlan KA, Zou MH. Liver kinase B1 expression promotes phosphatase activity and abrogation of receptor tyrosine kinase phosphorylation in human cancer cells. J Biol Chem 2013; 289:1639-48. [PMID: 24285539 DOI: 10.1074/jbc.m113.500934] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aberrant receptor tyrosine kinase phosphorylation (pRTK) has been associated with diverse pathological conditions, including human neoplasms. In lung cancer, frequent liver kinase B1 (LKB1) mutations correlate with tumor progression, but potential links with pRTK remain unknown. Heightened and sustained receptor activation was demonstrated by LKB1-deficient A549 (lung) and HeLaS3 (cervical) cancer cell lines. Depletion (siRNA) of endogenous LKB1 expression in H1792 lung cancer cells also correlated with increased pRTK. However, ectopic LKB1 expression in A549 and HeLaS3 cell lines, as well as H1975 activating-EGF receptor mutant lung cancer cell resulted in dephosphorylation of several tumor-enhancing RTKs, including EGF receptor, ErbB2, hepatocyte growth factor receptor (c-Met), EphA2, rearranged during transfection (RET), and insulin-like growth factor I receptor. Receptor abrogation correlated with attenuation of phospho-Akt and increased apoptosis. Global phosphatase inhibition by orthovanadate or depletion of protein tyrosine phosphatases (PTPs) resulted in the recovery of receptor phosphorylation. Specifically, the activity of SHP-2, PTP-1β, and PTP-PEST was enhanced by LKB1-expressing cells. Our findings provide novel insight on how LKB1 loss of expression or function promotes aberrant RTK signaling and rapid growth of cancer cells.
Collapse
|
34
|
Kline ER, Shupe J, Gilbert-Ross M, Zhou W, Marcus AI. LKB1 represses focal adhesion kinase (FAK) signaling via a FAK-LKB1 complex to regulate FAK site maturation and directional persistence. J Biol Chem 2013; 288:17663-74. [PMID: 23637231 DOI: 10.1074/jbc.m112.444620] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Liver kinase β1 (LKB1, also known as STK11) is a serine/threonine kinase that has multiple cellular functions including the regulation of cell polarity and motility. Murine proteomic studies show that LKB1 loss causes aberrant adhesion signaling; however, the mechanistic underpinnings of this relationship are unknown. We show that cells stably depleted of LKB1 or its co-activator STRADα have increased phosphorylation of focal adhesion kinase (FAK) at Tyr(397)/Tyr(861) and enhanced adhesion to fibronectin. LKB1 associates in a complex with FAK and LKB1 accumulation at the cellular leading edge is mutually excluded from regions of activated Tyr(397)-FAK. LKB1-compromised cells lack directional persistence compared with wild-type cells, but this is restored through subsequent pharmacological FAK inhibition or depletion, showing that cell directionality is mediated through LKB1-FAK signaling. Live cell confocal imaging reveals that LKB1-compromised cells lack normal FAK site maturation and turnover, suggesting that defects in adhesion and directional persistence are caused by aberrant adhesion dynamics. Furthermore, re-expression of full-length wild-type or the LKB1 N-terminal domain repressed FAK activity, whereas the kinase domain or C-terminal domain alone did not, indicating that FAK suppression is potentially regulated through the LKB1 N-terminal domain. Based upon these results, we conclude that LKB1 serves as a FAK repressor to stabilize focal adhesion sites, and when LKB1 function is compromised, aberrant FAK signaling ensues, resulting in rapid FAK site maturation and poor directional persistence.
Collapse
Affiliation(s)
- Erik R Kline
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
35
|
Jain P, Baranwal S, Dong S, Struckhoff AP, Worthylake RA, Alahari SK. Integrin-binding protein nischarin interacts with tumor suppressor liver kinase B1 (LKB1) to regulate cell migration of breast epithelial cells. J Biol Chem 2013; 288:15495-509. [PMID: 23572524 DOI: 10.1074/jbc.m112.418103] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Biallelic inactivation of LKB1, a serine/threonine kinase, has been detected in 30% of lung adenocarcinomas, and inhibition of breast tumor growth has been demonstrated. We have identified the tumor suppressor, Nischarin, as a novel binding partner of LKB1. Our mapping analysis shows that the N terminus of Nischarin interacts with amino acids 44-436 of LKB1. Time lapse microscopy and Transwell migration data show that the absence of both Nischarin and LKB1 from an invasive breast cancer cell line (MDA-MB-231) enhances migration as measured by increased distance and speed of migrating cells. Our data suggest that this is a result of elevated PAK1 and LIMK1 phosphorylation. Moreover, the absence of Nischarin and LKB1 increased tumor growth in vivo. Consistent with this, the percentage of S phase cells was increased, as demonstrated by flow cytometry and enhanced cyclin D1. The absence of Nischarin and LKB1 also led to a dramatic increase in the formation of lung metastases. Our studies, for the first time, demonstrate functional interaction between LKB1 and Nischarin to inhibit cell migration and breast tumor progression. Mechanistically, we show that these two proteins together regulate PAK-LIMK-Cofilin and cyclin D1/CDK4 pathways.
Collapse
Affiliation(s)
- Prachi Jain
- Department of Biochemistry and Molecular Biology, School of Medicine, Louisiana State University Health Science Center, New Orleans, Louisiana 70112, USA
| | | | | | | | | | | |
Collapse
|
36
|
Pugnaloni A, Giantomassi F, Lucarini G, Capella S, Bloise A, Di Primio R, Belluso E. Cytotoxicity induced by exposure to natural and synthetic tremolite asbestos: an in vitro pilot study. Acta Histochem 2013; 115:100-12. [PMID: 22578742 DOI: 10.1016/j.acthis.2012.04.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2012] [Revised: 04/13/2012] [Accepted: 04/16/2012] [Indexed: 01/13/2023]
Abstract
Mineral fibers are potential carcinogens to humans. In order to help clarify the etiology of the pathological effects of asbestos, cellular reactions to natural and synthetic asbestos fibers were compared using a lung alveolar cancer cell line (A549 epithelial cells), considered the first target of inhaled micro-environmental contaminants. Natural asbestos tremolite (NAT) fibers were collected from rocks in NW Italy. Synthetic asbestos tremolite (SAT) was iron-free and therefore considered as standard tremolite. Both fibers, subjected to mineralogical characterization by X-ray powder diffractometry, electron microscopy and energy dispersive spectrometry, fell within the definition of respirable and potentially carcinogenic fibers. Several signs of functional and structural cell damage were found after treatment with both fibers, documented by viability, motility, and morphological perturbations. Phalloidin labeling showed irregular distribution of cytoskeletal F-actin, whereas immunohistochemical investigations showed abnormal expression of VEGF, Cdc42, β-catenin, assessed as risks indicators for cancer development. Both fibers caused significant loss of viability, even compared to UICC crocidolite, but, while SAT fibers exerted a more direct cytotoxic effect, survival of damaged cells expressing high VEGF levels was detected after NAT contact. This in vitro pilot study outlines potential health risks of NAT fibers in vivo related to their iron content, which could trigger signaling networks connected with cell proliferation and neoplastic transformation.
Collapse
|
37
|
Inhibition of AMP-activated protein kinase accentuates lipopolysaccharide-induced lung endothelial barrier dysfunction and lung injury in vivo. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1021-30. [PMID: 23306156 DOI: 10.1016/j.ajpath.2012.11.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 11/09/2012] [Accepted: 11/21/2012] [Indexed: 01/24/2023]
Abstract
The aim of this study was to determine the role of AMP-activated protein kinase (AMPK) in lipopolysaccharide (LPS)-induced lung endothelial barrier dysfunction and lung injury in vivo. Both cultured human pulmonary artery endothelial cells (HPAECs) and experimental animals [AMPK subunit α-deficient mice and wild-type (WT) control mice (C57BL/6J)] were used. In cultured HPAECs, LPS increased endothelial permeability in parallel with a decrease in AMPK activity. Consistent with this observation, AMPK activation with the potent AMPK activator 5-aminoimidazole-4-carboxamide-1-d-ribofuranoside (AICAR) attenuated LPS-induced endothelial hyperpermeability in vitro. Intratracheal administration of LPS (1 mg/kg) in WT mice reduced AMPK phosphorylation at Thr172 in lung tissue extracts, increased protein content and cell count in bronchial alveolar lavage fluid, and increased Evans Blue dye infiltration into the lung. These same attributes were similarly enhanced in AMPKα-knockout mice, compared with WT mice. Pretreatment with AICAR reduced these lung injury indicators in LPS-treated WT mice. AMPK activation with AICAR attenuated LPS-induced endothelial hyperpermeability by activating the Rac/Cdc42/PAK pathway, with concomitant inhibition of the Rho pathway, and decreased VE-cadherin phosphorylation at Tyr658. We conclude that AMPK activity supports normal endothelial barrier function and that LPS exposure inhibits AMPK, thereby contributing to endothelial barrier dysfunction and lung injury.
Collapse
|
38
|
Korsse SE, Peppelenbosch MP, van Veelen W. Targeting LKB1 signaling in cancer. Biochim Biophys Acta Rev Cancer 2012; 1835:194-210. [PMID: 23287572 DOI: 10.1016/j.bbcan.2012.12.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 12/18/2012] [Accepted: 12/20/2012] [Indexed: 12/13/2022]
Abstract
The serine/threonine kinase LKB1 is a master kinase involved in cellular responses such as energy metabolism, cell polarity and cell growth. LKB1 regulates these crucial cellular responses mainly via AMPK/mTOR signaling. Germ-line mutations in LKB1 are associated with the predisposition of the Peutz-Jeghers syndrome in which patients develop gastrointestinal hamartomas and have an enormously increased risk for developing gastrointestinal, breast and gynecological cancers. In addition, somatic inactivation of LKB1 has been associated with sporadic cancers such as lung cancer. The exact mechanisms of LKB1-mediated tumor suppression remain so far unidentified; however, the inability to activate AMPK and the resulting mTOR hyperactivation has been detected in PJS-associated lesions. Therefore, targeting LKB1 in cancer is now mainly focusing on the activation of AMPK and inactivation of mTOR. Preclinical in vitro and in vivo studies show encouraging results regarding these approaches, which have even progressed to the initiation of a few clinical trials. In this review, we describe the functions, regulation and downstream signaling of LKB1, and its role in hereditary and sporadic cancers. In addition, we provide an overview of several AMPK activators, mTOR inhibitors and additional mechanisms to target LKB1 signaling, and describe the effect of these compounds on cancer cells. Overall, we will explain the current strategies attempting to find a way of treating LKB1-associated cancer.
Collapse
Affiliation(s)
- S E Korsse
- Dept. of Gastroenterology and Hepatology, Erasmus Medical University Center, Rotterdam, The Netherlands
| | | | | |
Collapse
|
39
|
Perturbing microtubule integrity blocks AMP-activated protein kinase-induced meiotic resumption in cultured mouse oocytes. ZYGOTE 2012. [PMID: 23199370 DOI: 10.1017/s0967199412000457] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The oocyte meiotic spindle is comprised of microtubules (MT) that bind chromatin and regulate both metaphase plate formation and karyokinesis during meiotic maturation; however, little information is known about their role in meiosis reinitiation. This study was conducted to determine if microtubule integrity is required for meiotic induction and to ascertain how it affects activation of AMP-activated protein kinase (AMPK), an important participant in the meiotic induction process. Treatment with microtubule-disrupting agents nocodazole and vinblastine suppressed meiotic resumption in a dose-dependent manner in both arrested cumulus cell-enclosed oocytes (CEO) stimulated with follicle-stimulating hormone (FSH) and arrested denuded oocytes (DO) stimulated with the AMPK activator, 5-aminoimidazole-4-carboxamide-1-beta-4-ribofuranoside (AICAR). This effect coincided with suppression of AMPK activation as determined by western blotting and germinal vesicle immunostaining. Treatment with the MT stabilizer paclitaxel also suppressed meiotic induction. Targeting actin filament polymerization had only a marginal effect on meiotic induction. Immunolocalization experiments revealed that active AMPK colocalized with γ-tubulin during metaphase I and II stages, while it localized at the spindle midzone during anaphase. This discrete localization pattern was dependent on MT integrity. Treatment with nocodazole led to disruption of proper spindle pole localization of active AMPK, while paclitaxel induced excessive polymerization of spindle MT and formation of ectopic asters with accentuated AMPK colocalization. Although stimulation of AMPK increased the rate of germinal vesicle breakdown (GVB), spindle formation and polar body (PB) extrusion, the kinase had no effect on peripheral movement of the spindle. These data suggest that the meiosis-inducing action and localization of AMPK are regulated by MT spindle integrity during mouse oocyte maturation.
Collapse
|
40
|
Hepatitis C viral protein NS5A induces EMT and participates in oncogenic transformation of primary hepatocyte precursors. J Hepatol 2012; 57:1021-8. [PMID: 22750466 DOI: 10.1016/j.jhep.2012.06.027] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2012] [Revised: 06/07/2012] [Accepted: 06/21/2012] [Indexed: 01/22/2023]
Abstract
BACKGROUND & AIMS Apicobasal polarity, which is essential for epithelial structure and function, is targeted by several tumour-related pathogens and is generally perturbed in the course of carcinogenesis. Hepatitis C virus (HCV) infection is associated with a strong risk of hepatocellular carcinoma, typically preceded by dysplastic alterations of cell morphology. We investigated the molecular mechanisms and the functional consequences of HCV-driven perturbations of epithelial polarity. METHODS We used biochemical, genetic, and cell biology approaches to assess the impact of hepatitis C viral protein NS5A on the polarity and function of hepatocytes and hepatic progenitors. Transgenic animals and xenograft models served for in vivo validation of the results obtained in cell culture. RESULTS We found that expression of HCV-NS5A in primary hepatic precursors and in immortalized hepatocyte cell lines gave rise to profound modifications of cell polarity, leading to epithelial to mesenchymal transition (EMT). NS5A, either alone or in the context of the full complement of viral proteins in the course of infection, acted through activating Twist2, a transcriptional regulator of EMT. The effects of NS5A were additive to those of TGF-β, a cytokine abundant in diseased liver and highly relevant to HCV-related pathology. Moreover, NS5A cooperates with oncogenic Ras, giving rise to transformed, invasive cells that are highly tumorigenic in vivo. CONCLUSIONS Our data suggest that in the context of HCV infection, NS5A favors formation of preneoplastic lesions by disrupting cell polarity and additional oncogenic events cooperate with the viral protein to give rise to motile and invasive tumour cells.
Collapse
|
41
|
AMPK regulates mitotic spindle orientation through phosphorylation of myosin regulatory light chain. Mol Cell Biol 2012; 32:3203-17. [PMID: 22688514 DOI: 10.1128/mcb.00418-12] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The proper orientation of the mitotic spindle is essential for mitosis; however, how these events unfold at the molecular level is not well understood. AMP-activated protein kinase (AMPK) regulates energy homeostasis in eukaryotes, and AMPK-null Drosophila mutants have spindle defects. We show that threonine(172) phosphorylated AMPK localizes to the mitotic spindle poles and increases when cells enter mitosis. AMPK depletion causes a mitotic delay with misoriented spindles relative to the normal division plane and a reduced number and length of astral microtubules. AMPK-depleted cells contain mitotic actin bundles, which prevent astral microtubule-actin cortex attachments. Since myosin regulatory light chain (MRLC) is an AMPK downstream target and mediates actin function, we investigated whether AMPK signals through MRLC to control spindle orientation. Mitotic levels of serine(19) phosphorylated MRLC (pMRLC(ser19)) and spindle pole-associated pMRLC(ser19) are abolished when AMPK function is compromised, indicating that AMPK is essential for pMRLC(ser19) spindle pole activity. Phosphorylation of AMPK and MRLC in the mitotic spindle is dependent upon calcium/calmodulin-dependent protein kinase kinase (CamKK) activity in LKB1-deficient cells, suggesting that CamKK regulates this pathway when LKB1 function is compromised. Taken together, these data indicate that AMPK mediates spindle pole-associated pMRLC(ser19) to control spindle orientation via regulation of actin cortex-astral microtubule attachments.
Collapse
|
42
|
EMT as the ultimate survival mechanism of cancer cells. Semin Cancer Biol 2012; 22:194-207. [DOI: 10.1016/j.semcancer.2012.02.013] [Citation(s) in RCA: 354] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2011] [Revised: 02/27/2012] [Accepted: 02/28/2012] [Indexed: 12/24/2022]
|
43
|
Wildenberg ME, Vos ACW, Wolfkamp SCS, Duijvestein M, Verhaar AP, Te Velde AA, van den Brink GR, Hommes DW. Autophagy attenuates the adaptive immune response by destabilizing the immunologic synapse. Gastroenterology 2012; 142:1493-503.e6. [PMID: 22370477 DOI: 10.1053/j.gastro.2012.02.034] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 01/25/2012] [Accepted: 02/15/2012] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Variants in the genes ATG16L1 and IRGM affect autophagy and are associated with the development of Crohn's disease. It is not clear how autophagy is linked to loss of immune tolerance in the intestine. We investigated the involvement of the immunologic synapse-the site of contact between dendritic cells (DCs) and T cells, which contains molecules involved in antigen recognition and regulates immune response. METHODS DC autophagy was reduced using small interfering RNAs or pharmacologic inhibitors. DC phenotype and function were analyzed by confocal microscopy, time-lapse microscopy, and flow cytometry. We also examined DCs isolated from patients with Crohn's disease who carried the ATG16L1 risk allele. RESULTS Immunologic synapse formation induced formation of autophagosomes in DCs; the autophagosomes were oriented toward the immunologic synapse and contained synaptic components. Knockdown of ATG16L1 and IRGM with small interfering RNAs in DCs resulted in hyperstable interactions between DCs and T cells, increased activation of T cells, and activation of a T-helper 17 cell response. LKB1 was recruited to the immunologic synapse, and induction of autophagy in DC required inhibition of mammalian target of rapamycine signaling by the LKB1-AMP activated protein kinase (AMPK) pathway. DCs from patients with Crohn's disease who had an ATG16L1 risk allele had a similar hyperstability of the immunologic synapse. CONCLUSIONS Autophagy is induced upon formation of the immunologic synapse and negatively regulates T-cell activation. This mechanism might increase adaptive immunity in patients with Crohn's disease who carry ATG16L1 risk alleles.
Collapse
Affiliation(s)
- Manon E Wildenberg
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Eggers CM, Kline ER, Zhong D, Zhou W, Marcus AI. STE20-related kinase adaptor protein α (STRADα) regulates cell polarity and invasion through PAK1 signaling in LKB1-null cells. J Biol Chem 2012; 287:18758-68. [PMID: 22493453 DOI: 10.1074/jbc.m111.316422] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
LKB1 is a Ser/Thr kinase, and its activity is regulated by the pseudokinase, STE20-related adaptor α (STRADα). The STRADα-LKB1 pathway plays critical roles in epithelial cell polarity, neuronal polarity, and cancer metastasis. Though much attention is given to the STRADα-LKB1 pathway, the function of STRADα itself, including a role outside of the LKB1 pathway, has not been well-studied. Data in Caenorhabditis elegans suggest that STRADα has an LKB1-independent role in regulating cell polarity, and therefore we tested the hypothesis that STRADα regulates cancer cell polarity and motility when wild-type LKB1 is absent. These results show that STRADα protein is reduced in LKB1-null cell lines (mutation or homozygous deletion) and this partial degradation occurs through the Hsp90-dependent proteasome pathway. The remaining STRADα participates in cell polarity and invasion, such that STRADα depletion results in misaligned lamellipodia, improper Golgi positioning, and reduced invasion. To probe the molecular basis of this defect, we show that STRADα associates in a complex with PAK1, and STRADα loss disrupts PAK1 activity via Thr(423) PAK1 phosphorylation. When STRADα is depleted, PAK1-induced invasion could not occur, suggesting that STRADα is necessary for PAK1 to drive motility. Furthermore, STRADα overexpression caused increased activity of the PAK1-activating protein, rac1, and a constitutively active rac1 mutant (Q61L) rescued pPAK(Thr423) and STRADα invasion defects. Taken together, these results show that a STRADα-rac1-PAK1 pathway regulates cell polarity and invasion in LKB1-null cells. It also suggests that while the function of LKB1 and STRADα undoubtedly overlap, they may also have mutually exclusive roles.
Collapse
Affiliation(s)
- Carrie M Eggers
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
45
|
Abstract
To elucidate the effect of cigarette smoke on developing lung cancer among individuals, numerous genetic and epigenetic factors related to cigarette smoke-induced lung cancers have been widely investigated and a various genes, loci and pathways have been identified as candidates to date. However, the importance of these molecular alterations in the initiation and progression of lung cancer still remains imprecise and different molecules altered in lung cancer are being used for stratification of patients for targeted therapy. There are a number of molecular pathways involved in the development of lung cancer, and environmental factors related to these alterations are still unclear. Furthermore, various genetic alterations determined by candidate gene approach have not been re-evaluated for their functional significance together with epigenetic alterations in the same population. Accumulated evidence suggested that lung cancer in ever smokers and never smokers follow distinct molecular pathways and may therefore respond to distinct therapy. Therefore, additional studies will be essential to re-evaluate the individual risk of developing lung cancer based on the combination of genetic and epigenetic alterations and to set up a guideline to assess the individual risk for lung cancer and for its prevention.
Collapse
Affiliation(s)
- Shahnaz Begum
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA.
| |
Collapse
|
46
|
Mian I, Pierre-Louis WS, Dole N, Gilberti RM, Dodge-Kafka K, Tirnauer JS. LKB1 destabilizes microtubules in myoblasts and contributes to myoblast differentiation. PLoS One 2012; 7:e31583. [PMID: 22348111 PMCID: PMC3279410 DOI: 10.1371/journal.pone.0031583] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Accepted: 01/09/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Skeletal muscle myoblast differentiation and fusion into multinucleate myotubes is associated with dramatic cytoskeletal changes. We find that microtubules in differentiated myotubes are highly stabilized, but premature microtubule stabilization blocks differentiation. Factors responsible for microtubule destabilization in myoblasts have not been identified. FINDINGS We find that a transient decrease in microtubule stabilization early during myoblast differentiation precedes the ultimate microtubule stabilization seen in differentiated myotubes. We report a role for the serine-threonine kinase LKB1 in both microtubule destabilization and myoblast differentiation. LKB1 overexpression reduced microtubule elongation in a Nocodazole washout assay, and LKB1 RNAi increased it, showing LKB1 destabilizes microtubule assembly in myoblasts. LKB1 levels and activity increased during myoblast differentiation, along with activation of the known LKB1 substrates AMP-activated protein kinase (AMPK) and microtubule affinity regulating kinases (MARKs). LKB1 overexpression accelerated differentiation, whereas RNAi impaired it. CONCLUSIONS Reduced microtubule stability precedes myoblast differentiation and the associated ultimate microtubule stabilization seen in myotubes. LKB1 plays a positive role in microtubule destabilization in myoblasts and in myoblast differentiation. This work suggests a model by which LKB1-induced microtubule destabilization facilitates the cytoskeletal changes required for differentiation. Transient destabilization of microtubules might be a useful strategy for enhancing and/or synchronizing myoblast differentiation.
Collapse
Affiliation(s)
- Isma Mian
- Center for Molecular Medicine and University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Willythssa Stéphie Pierre-Louis
- Center for Molecular Medicine and University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Neha Dole
- Center for Molecular Medicine and University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Renée M. Gilberti
- Center for Molecular Medicine and University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Kimberly Dodge-Kafka
- Calhoun Center for Cardiology, University of Connecticut Health Center, Farmington, Connecticut, United States of America
| | - Jennifer S. Tirnauer
- Center for Molecular Medicine and University of Connecticut Health Center, Farmington, Connecticut, United States of America
| |
Collapse
|
47
|
Jain M, Bhat GP, Vijayraghavan K, Inamdar MS. Rudhira/BCAS3 is a cytoskeletal protein that controls Cdc42 activation and directional cell migration during angiogenesis. Exp Cell Res 2012; 318:753-67. [PMID: 22300583 DOI: 10.1016/j.yexcr.2012.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 01/11/2012] [Accepted: 01/15/2012] [Indexed: 12/19/2022]
Abstract
Cell migration is a common cellular process in angiogenesis and tumor metastasis. Rudhira/BCAS3 (Breast Cancer Amplified Sequence 3) is a conserved protein expressed in the embryonic vasculature and malignant tumors. Here, we show for the first time that Rudhira plays an active role in directional cell migration. Rudhira depletion in endothelial cells inhibits Matrigel-induced tube formation and retards healing of wounded cell monolayers. We demonstrate that during wound healing, Rudhira rapidly re-localizes and promotes Cdc42 activation and recruitment to the leading edge of migrating cells. Rudhira deficient cells show impaired downstream signaling of Cdc42 leading to dramatic changes in actin organization and classic cell polarity defects such as loss of microtubule organizing center (MTOC) and Golgi re-orientation. Biochemical assays and co-localization studies show that Rudhira interacts with microtubules as well as intermediate filaments. Thus, Rudhira could control directional cell migration and angiogenesis by facilitating crosstalk between cytoskeletal elements.
Collapse
Affiliation(s)
- Mamta Jain
- Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | | | | | | |
Collapse
|
48
|
Abstract
Initially identified as the Caenorhabditis elegans PAR-4 homologue, the serine threonine kinase LKB1 is conserved throughout evolution and ubiquitously expressed. In humans, LKB1 is causally linked to the Peutz-Jeghers syndrome and is one of the most commonly mutated genes in several cancers like lung and cervical carcinomas. These observations have led to classify LKB1 as tumour suppressor gene. Although, considerable dark zones remain, an impressive leap in the understanding of LKB1 functions has been done during the last decade. Role of LKB1 as a major actor of the AMPK/mTOR pathway connecting cellular metabolism, cell growth and tumorigenesis has been extensively studied probably to the detriment of other functions of equal importance. This review will discuss about LKB1 activity regulation, its effectors and clues on their involvement in cell polarity.
Collapse
|
49
|
Abstract
In humans, the LKB1 gene is located on the short arm of chromosome 19, which is frequently deleted in lung tumors. Unlike most cancers of sporadic origin, in non-small cell lung cancer (NSCLC) nearly half of the tumors harbor somatic and homozygous inactivating mutations in LKB1. In NSCLC, LKB1 inactivation strongly predominates in adenocarcinomas from smokers and coexists with mutations at other important cancer genes, including KRAS and TP53. Remarkably, LKB1 alterations frequently occur simultaneously with inactivation at another important tumor suppressor gene, BRG1 (also called SMARCA4), which is also located on chromosome 19p. The present review considers the frequency and pattern of LKB1 mutations in lung cancer and the distinct biological pathways in which the LKB1 protein is involved in the development of this type of cancer. Finally, the possible clinical applications in cancer management, especially in lung cancer treatment, associated with the presence of absence of LKB1 are discussed.
Collapse
Affiliation(s)
- Montse Sanchez-Cespedes
- Programa Epigenetica i Biologia del Cancer-PEBC, Institut Investigacions Biomediques Bellvitge, Hospital Duran i Reynals, Av. Gran Via de l'Hospitalet, 199-203, 08907, Hospitalet de Llobregat-Barcelona, Spain.
| |
Collapse
|
50
|
Khoury HJ, Garcia-Manero G, Borthakur G, Kadia T, Foudray MC, Arellano M, Langston A, Bethelmie-Bryan B, Rush S, Litwiler K, Karan S, Simmons H, Marcus AI, Ptaszynski M, Kantarjian H. A phase 1 dose-escalation study of ARRY-520, a kinesin spindle protein inhibitor, in patients with advanced myeloid leukemias. Cancer 2011; 118:3556-64. [PMID: 22139909 DOI: 10.1002/cncr.26664] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/19/2011] [Indexed: 11/12/2022]
Abstract
BACKGROUND ARRY-520 selectively inhibits the mitotic kinesin spindle protein (KSP), which leads to abnormal monopolar spindle formation and apoptosis. METHODS A phase 1 trial was conducted to establish the safety and the maximum tolerated dose (MTD) of ARRY-520 given as a 1-hour infusion in either a single dose or on a day 1, 3, and 5 divided-dose schedule per cycle in patients with advanced or refractory myeloid leukemias. Additional objectives were to characterize pharmacokinetics, assess preliminary clinical activity, and explore biomarkers of KSP inhibition with ARRY-520. A total of 36 patients with acute myelogenous leukemia (n = 34) or myelodysplastic syndromes (n = 2) with a median age of 66 years (range, 21-88 years) were enrolled: 15 in the single-dose schedule (dose levels: 2.5, 3.75, 4.5, and 5.6 mg/m(2)) and 21 in the divided-dose schedule (dose levels: 0.8, 1.2, 1.5, and 1.8 mg/m(2)/day). RESULTS The MTD was 4.5 mg/m(2) total dose per cycle for both dose schedules. Dose-limiting toxicities included mucositis, exfoliative rash, hand-foot syndrome, and hyperbilirubinemia. Grades 3 or 4 reversible drug-related myelosuppression were observed in 33 of 36 patients. Plasma pharmacokinetic analyses revealed low clearance of ARRY-520 (~3 L/hour), a volume of distribution of ~450 L, and a median terminal half-life of >90 hours. Monopolar spindles were observed in blood mononuclear cells, through use of 4',6-diamidino-2-phenylindole nucleic acid stain and antitubulin antibodies. CONCLUSIONS On the basis of the relative lack of clinical activity, further development of ARRY-520 as an antileukemic agent was halted. (Clinicaltrials.gov identifier NCT00637052).
Collapse
Affiliation(s)
- Hanna Jean Khoury
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|