51
|
Vengoji R, Macha MA, Nimmakayala RK, Rachagani S, Siddiqui JA, Mallya K, Gorantla S, Jain M, Ponnusamy MP, Batra SK, Shonka N. Afatinib and Temozolomide combination inhibits tumorigenesis by targeting EGFRvIII-cMet signaling in glioblastoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:266. [PMID: 31215502 PMCID: PMC6582495 DOI: 10.1186/s13046-019-1264-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/03/2019] [Indexed: 12/30/2022]
Abstract
Background Glioblastoma (GBM) is an aggressive brain tumor with universal recurrence and poor prognosis. The recurrence is largely driven by chemoradiation resistant cancer stem cells (CSCs). Epidermal growth factor receptor (EGFR) and its mutant EGFRvIII are amplified in ~ 60% and ~ 30% of GBM patients, respectively; however, therapies targeting EGFR have failed to improve disease outcome. EGFRvIII-mediated cross-activation of tyrosine kinase receptor, cMET, regulates GBM CSC maintenance and promote tumor recurrence. Here, we evaluated the efficacy of pan-EGFR inhibitor afatinib and Temozolomide (TMZ) combination on GBM in vitro and in vivo. Methods We analyzed the effect of afatinib and temozolomide (TMZ) combination on GBM cells U87MG and U251 engineered to express wild type (WT) EGFR, EGFRvIII or EGFRvIII dead kinase, CSCs isolated from U87 and U87EGFRvIII in vitro. The therapeutic utility of the drug combination was investigated on tumor growth and progression using intracranially injected U87EGFRvIII GBM xenografts. Results Afatinib and TMZ combination synergistically inhibited the proliferation, clonogenic survival, motility, invasion and induced senescence of GBM cells compared to monotherapy. Mechanistically, afatinib decreased U87EGFRvIII GBM cell proliferation and motility/invasion by inhibiting EGFRvIII/AKT, EGFRvIII/JAK2/STAT3, and focal adhesion kinase (FAK) signaling pathways respectively. Interestingly, afatinib specifically inhibited EGFRvIII-cMET crosstalk in CSCs, resulting in decreased expression of Nanog and Oct3/4, and in combination with TMZ significantly decreased their self-renewal property in vitro. More interestingly, afatinib and TMZ combination significantly decreased the xenograft growth and progression compared to single drug alone. Conclusion Our study demonstrated significant inhibition of GBM tumorigenicity, CSC maintenance in vitro, and delayed tumor growth and progression in vivo by combination of afatinib and TMZ. Our results warrant evaluation of this drug combination in EGFR and EGFRvIII amplified GBM patients. Electronic supplementary material The online version of this article (10.1186/s13046-019-1264-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Muzafar A Macha
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Department of Otolaryngology/Head and Neck Surgery, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Satyanarayana Rachagani
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Jawed A Siddiqui
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Santhi Gorantla
- Department of Pharmacology & Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Nicole Shonka
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, 68198, USA. .,Department of Internal Medicine, Division of Oncology and Hematology, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
52
|
Dong C, Sun J, Ma S, Zhang G. K-ras-ERK1/2 down-regulates H2A.X Y142ph through WSTF to promote the progress of gastric cancer. BMC Cancer 2019; 19:530. [PMID: 31151422 PMCID: PMC6545063 DOI: 10.1186/s12885-019-5750-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 05/24/2019] [Indexed: 12/19/2022] Open
Abstract
Background Histone H2AX phosphorylation at the site of Tyr-142 can participates in multiple biological progressions, which is including DNA repair. Ras pathway is closely involved in human cancers. Our study investigated the effects of Ras pathway via regulating H2AX.Y142ph. Methods Gastric cancer cell line SNU-16 and MKN1 cells were transfected with Ras for G12D and T35S site mutation. The phosphorylation of H2A.XY142 and ERK1/2, WSTF and MDM2 was detected by western blot. Cell viability, cell colonies and migration was analyzed by MTT assay, soft-agar colony formation assay, and Transwell assay, respectively. The expression of Ras pathway related downstream factors, EYA3 and WSTF was detected by qRT-PCR. The relationship between Ras and downstream factors were detected by ChIP. The cell cycle progression was measured by flow cytometry. Results RasG12D/T35V transection decreased the phosphorylation of H2A.XY142 and activated phosphorylation of ERK-1/2. H2A.XY142 inhibited cell viability, colonies and migration. H2A.XY142ph altered the expression of Ras downstream factors. CHIP assay revealed that RasG12D/T35V could bind to the promoters of these Ras pathway downstream factors. Silence of EYA3 increased H2A.XY142ph and inhibited cell viability, migration and percent cells in S stage. Furthermore, silence of EYA3 also changed the downstream factors expression. WSTF and H2A.XY142ph revealed the similar trend and MDM2 on the opposite. Conclusion Ras/ERK signal pathway decreased H2A.XY142ph and promoted cell growth and metastasis. This Ras regulation process was down-regulated by the cascade of MDM2-WSTF-EYA3 to decrease H2A.XY142ph in SNU-16 cells.
Collapse
Affiliation(s)
- Chao Dong
- Department of Clinical Medicine, Qujing Medical College, Qujing, 655000, Yunnan, China
| | - Jing Sun
- Department of Pharmacy, Qujing Medical College, Sanjiang Avenue, Economic Development Zone, Qilin District, Qujing, 655000, Yunnan, China
| | - Sha Ma
- Department of Pharmacy, Qujing Medical College, Sanjiang Avenue, Economic Development Zone, Qilin District, Qujing, 655000, Yunnan, China
| | - Guoying Zhang
- Department of Pharmacy, Qujing Medical College, Sanjiang Avenue, Economic Development Zone, Qilin District, Qujing, 655000, Yunnan, China.
| |
Collapse
|
53
|
Merve A, Zhang X, Pomella N, Acquati S, Hoeck JD, Dumas A, Rosser G, Li Y, Jeyapalan J, Vicenzi S, Fan Q, Yang ZJ, Sabò A, Sheer D, Behrens A, Marino S. c-MYC overexpression induces choroid plexus papillomas through a T-cell mediated inflammatory mechanism. Acta Neuropathol Commun 2019; 7:95. [PMID: 31142360 PMCID: PMC6540455 DOI: 10.1186/s40478-019-0739-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 05/14/2019] [Indexed: 12/26/2022] Open
Abstract
Choroid plexus tumours (CPTs) account for 2–5% of brain tumours in children. They can spread along the neuraxis and can recur after treatment. Little is known about the molecular mechanisms underlying their formation and only few high fidelity mouse models of p53-deficient malignant CPTs are available. We show here that c-MYC overexpression in the choroid plexus epithelium induces T-cell inflammation-dependent choroid plexus papillomas in a mouse model. We demonstrate that c-MYC is expressed in a substantial proportion of human choroid plexus tumours and that this subgroup of tumours is characterised by an inflammatory transcriptome and significant inflammatory infiltrates. In compound mutant mice, overexpression of c-MYC in an immunodeficient background led to a decreased incidence of CPP and reduced tumour bulk. Finally, reduced tumour size was also observed upon T-cell depletion in CPP-bearing mice. Our data raise the possibility that benign choroid plexus tumours expressing c-MYC could be amenable to medical therapy with anti-inflammatory drugs.
Collapse
|
54
|
Song KH, Trudeau T, Kar A, Borden MA, Gutierrez-Hartmann A. Ultrasound-mediated delivery of siESE complexed with microbubbles attenuates HER2+/- cell line proliferation and tumor growth in rodent models of breast cancer. Nanotheranostics 2019; 3:212-222. [PMID: 31183315 PMCID: PMC6536781 DOI: 10.7150/ntno.31827] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/02/2019] [Indexed: 12/18/2022] Open
Abstract
The highly tunable, noninvasive and spatially targeted nature of microbubble-enhanced, ultrasound-guided (MB+US) drug delivery makes it desirable for a wide variety of therapies. In breast cancer, both HER2+ and HER2- type neoplasms pose significant challenges to conventional therapeutics in greater than 40% of breast cancer patients, even with the widespread application of biologics such as trastuzumab. To address this therapeutic challenge, we examined the novel combination of tumor-injected microbubble-bound siRNA complexes and monodisperse size-isolated microbubbles (4-µm diameter) to attenuate tumor growth in vivo, as well as MB+US-facilitated shRNA and siRNA knockdown of ESE-1, an effector linked to dysregulated HER2 expression in HER2+/- cell line propagation. We first screened six variants of siESE and shESE for efficient knockdown of ESE in breast cancer cell lines. We demonstrated efficient reduction of BT-474 (PR+, ER+, HER2+; luminal B) and MDA-MB-468 (PR-, ER-, HER2-; triple-negative) clonogenicity and non-adherent growth after knockdown of ESE-1. A significant reduction in proliferative potential was seen for both cell lines using MB+US to deliver shESE and siESE. We then demonstrated significant attenuation of BT-474 xenograft tumor growth in Nod/SCID female mice using direct injection of microbubble-adsorbed siESE to the tumor and subsequent sonication. Our results suggest a positive effect on drug delivery from MB+US, and highlights the feasibility of using RNAi and MB+US for breast cancer pathologies. RNAi coupled with MB+US may also be an effective theranostic approach to treat other acoustically accessible tumors, such as melanoma, thyroid, parotid and skin cancer.
Collapse
Affiliation(s)
- Kang-Ho Song
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, USA
| | - Tammy Trudeau
- Departments of Medicine and of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Adwitiya Kar
- Departments of Medicine and of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Mark A. Borden
- Department of Mechanical Engineering, University of Colorado, Boulder, CO 80309, USA
| | - Arthur Gutierrez-Hartmann
- Departments of Medicine and of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| |
Collapse
|
55
|
Liu W, Huai R, Zhang Y, Rao S, Xiong L, Ding R, Mao C, Zhao W, Hao T, Huang Q, Guo Z. Down-regulation expression of TGFB2-AS1 inhibits the proliferation, migration, invasion and induces apoptosis in HepG2 cells. Genes Genomics 2019; 41:951-959. [PMID: 31066006 DOI: 10.1007/s13258-019-00826-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 04/24/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the leading cause of cancer mortality and without effective prognosis. Previous study has been confirmed that the abnormal expression of long non-coding RNAs (lncRNAs) TGFB2-AS1 was involved in tumorigenesis. However, the biological functions of TGFB2-AS1 in hepatocellular carcinoma (HCC) remain largely unclear. OBJECTIVE We comprehensively assess the clinical significance of TGFB2-AS1 and investigate the biological functions of TGFB2-AS1 on HCC HepG2 cells. METHODS We firstly confirmed the expression of TGFB2-AS1 between tumor and normal tissues using public available transcriptome data. We analyzed the clinical significance of TGFB2-AS1 using the TCGA HCC datasets. The biological functions of TGFB2-AS1 on HCC HepG2 cells were explored by multiple in vitro assays. RESULTS We found that TGFB2-AS1 was remarkably increased in HCC tissues (P = 0.00148) and exhibited a potential predictive marker for HCC, with an area under curve (AUC) of 0.708 (P = 0.0034) using the fifty pairs of matched HCC tissues of TCGA. Besides, higher expression of TGFB2-AS1 in HCC tissues was identified as being positively associated with advanced tumor (P = 0.012) and disease stage (P = 0.009) in 355 HCC cases using independent sample nonparametric test. Downregulation of TGFB2-AS1 expression significantly restrained proliferation (P < 0.01) and impaired colony formation (P < 0.05). Furthermore, TGFB2-AS1 depletion remarkably promoted the apoptosis of HepG2 cells (P < 0.05) and inhibited migration and invasion (P < 0.01). CONCLUSION Taken together, these findings suggested that TGFB2-AS1 might serve as a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Wenrong Liu
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Ruiping Huai
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Yin Zhang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Shuquan Rao
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Lili Xiong
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Ruofan Ding
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Canquan Mao
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Wenqing Zhao
- Department of Medical Oncology, Datong Second People's Hospital, Datong, Shanxi, People's Republic of China
| | - Tao Hao
- Department of Medical Oncology, Datong Second People's Hospital, Datong, Shanxi, People's Republic of China
| | - Qingqing Huang
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China
| | - Zhiyun Guo
- School of Life Sciences and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
56
|
Isoginkgetin, a Natural Biflavonoid Proteasome Inhibitor, Sensitizes Cancer Cells to Apoptosis via Disruption of Lysosomal Homeostasis and Impaired Protein Clearance. Mol Cell Biol 2019; 39:MCB.00489-18. [PMID: 30910794 DOI: 10.1128/mcb.00489-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 02/28/2019] [Indexed: 11/20/2022] Open
Abstract
Protein degradation pathways are critical for maintaining proper protein dynamics within the cell, and considerable efforts have been made toward the development of therapeutics targeting these catabolic processes. We report here that isoginkgetin, a naturally derived biflavonoid, sensitized cells undergoing nutrient starvation to apoptosis, induced lysosomal stress, and activated the lysosome biogenesis gene TFEB Isoginkgetin treatment led to the accumulation of aggregates of polyubiquitinated proteins that colocalized strongly with the adaptor protein p62, the 20S proteasome, and the endoplasmic reticulum-associated degradation (ERAD) protein UFD1L. Isoginkgetin directly inhibited the chymotrypsin-like, trypsin-like, and caspase-like activities of the 20S proteasome and impaired NF-κB signaling, suggesting that the molecule may display its biological activity in part through proteasome inhibition. Importantly, isoginkgetin was effective at killing multiple myeloma (MM) cell lines in vitro and displayed a higher rate of cell death induction than the clinically approved proteasome inhibitor bortezomib. We propose that isoginkgetin disturbs protein homeostasis, leading to an excess of protein cargo that places a burden on the lysosomes/autophagic machinery, eventually leading to cancer cell death.
Collapse
|
57
|
Chang CC, Huang KH, Hsu SP, Lee YCG, Sue YM, Juan SH. Simvastatin reduces the carcinogenic effect of 3-methylcholanthrene in renal epithelial cells through histone deacetylase 1 inhibition and RhoA reactivation. Sci Rep 2019; 9:4606. [PMID: 30872677 PMCID: PMC6418087 DOI: 10.1038/s41598-019-40757-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/21/2018] [Indexed: 12/24/2022] Open
Abstract
The therapeutic effects of simvastatin for renal cell carcinoma (RCC) are controversial. In this study, the effects of simvastatin on the carcinogenic properties of 3-methylcholanthrene (3MC; an aryl-hydrocarbon receptor [AhR] agonist) in human renal epithelial cells (hRECs) were investigated. We exposed in vitro and in vivo models to 3MC to induce RCC onset. 3MC upregulated the epithelial-mesenchymal transition (EMT) and tumor biomarkers; the models exhibited the reciprocal expression of histone deacetylase 1 (HDAC1) and RhoA, namely increased HDAC1 and decreased RhoA expression, through hypoxia-inducible-factor (HIF)- and AhR-dependent mechanisms. In addition to inducing EMT biomarkers, 3MC decreased von Hippel-Lindau protein levels (a risk factor for RCC) and increased CD44 expression in hRECs, which were reversed by digoxin (a HIF inhibitor) and HDAC inhibitors (suberoylanilide hydroxamic acid and trichostatin A [TSA]). Simvastatin abolished the detrimental effects of 3MC by reducing HDAC1 expression, with resulting RhoA upregulation, and reactivating RhoA in vitro and in vivo. Notably, the protective effects of simvastatin were negated by an HDAC activator (ITSA) through TSA suppression. The crucial role of RhoA in RCC carcinogenesis was verified by the overexpression of constitutively active RhoA. Collectively, these results demonstrate that simvastatin restores RhoA function through HDAC1 inhibition; therefore, simvastatin might serve as adjunct therapy for RCC induced by 3MC.
Collapse
Affiliation(s)
- Chih-Cheng Chang
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kuo-How Huang
- National Taiwan University Hospital; Department of Urology, College of Medicine, National Taiwan University; and National Taiwan University Hospital, Taipei, Taiwan
| | - Sung-Po Hsu
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Chii G Lee
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Mou Sue
- Division of Nephrology, Department of Internal Medicine, School of Medicine, College of Medicine and Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Shu-Hui Juan
- Department of Physiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
58
|
Loss of MADD expression inhibits cellular growth and metastasis in anaplastic thyroid cancer. Cell Death Dis 2019; 10:145. [PMID: 30760700 PMCID: PMC6374448 DOI: 10.1038/s41419-019-1351-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/05/2018] [Accepted: 12/17/2018] [Indexed: 02/07/2023]
Abstract
Anaplastic Thyroid Cancer (ATC) is an aggressive malignancy with limited therapeutic options and dismal patient survival. We have previously shown MADD to be differentially overexpressed in multiple cancer histologies and to contribute to tumor cell growth and survival. Therefore, we targeted MADD by gene silencing, explored its effect on cellular proliferation and metastases and examined its therapeutic potential in an orthotopic ATC model in athymic nude mice. When compared to untreated control and scramble siRNA, MADD siRNA treatment inhibited the proliferative capacity of 8505C, C643 and HTH7 cells in vitro and 8505C-derived-orthotopic tumor growth in vivo. MADD ablation caused a significant reduction in cellular migration and invasion potential; clonogenic capacity; as well as, mitochondrial length and potential in vitro. This MADD siRNA-induced anti-migratory/invasive effect corresponded with inhibition of epithelial–mesenchymal transition (EMT) and Wnt signaling. Mechanistically, MADD siRNA inhibited TNFα induced activation of pERK, pGSK3β and β-catenin, suggesting that MADD knockdown might exert its anti-migratory/invasive effects, by blocking TNFα/ERK/GSK3β axis. MADD siRNA can inhibit β-catenin nuclear translocation and consequently, the expression of its target genes in ATC cells. In in vivo experiments, along with tumor regression, MADD siRNA treatment also decreased evidence of lung metastases. Immunohistochemically, MADD siRNA-treated tumor tissues exhibited a reduction in Ki67 and N-Cadherin expression, and an increase in E-Cadherin expression. In conclusion, we show the crucial role of MADD in ATC tumorigenesis and metastasis and its potential implications as a molecular target for ATC therapy.
Collapse
|
59
|
Kalailingam P, Tan HB, Pan JY, Tan SH, Thanabalu T. Overexpression of CDC42SE1 in A431 Cells Reduced Cell Proliferation by Inhibiting the Akt Pathway. Cells 2019; 8:cells8020117. [PMID: 30717410 PMCID: PMC6406378 DOI: 10.3390/cells8020117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/25/2019] [Accepted: 01/30/2019] [Indexed: 12/28/2022] Open
Abstract
Cell division cycle 42 (CDC42), a small Rho GTPase, plays a critical role in many cellular processes, including cell proliferation and survival. CDC42 interacts with the CRIB (Cdc42- and Rac-interactive binding) domain of CDC42SE1, a small effector protein of 9 kDa. We found that the expression of CDC42SE1 was reduced in human skin cancer samples relative to matched perilesional control. Exogenous expression of CDC42SE1 but not CDC42SE1H38A (mutation within CRIB domain) in A431 cells (A431SE1, A431SE1-H38A) reduced cell proliferation. Antibody microarray analysis of A431Ctrl and A431SE1 lysate suggested that reduced A431SE1 cells proliferation was due to inhibition of Akt pathway, which was confirmed by the reduced P-Akt and P-mTOR levels in A431SE1 cells compared to A431Ctrl cells. This suggests that CDC42SE1 modulates the CDC42-mediated Akt pathway by competing with other effector proteins to bind CDC42. A431SE1 cells formed smaller colonies in soft agar compared to A431Ctrl and A431SE1-H38A cells. These findings correlate with nude mice xenograft assays, where A431SE1 cells formed tumors with significantly-reduced volume compared to the tumors formed by A431Ctrl cells. Our results suggest that CDC42SE1 is downregulated in skin cancer to promote tumorigenesis, and thus CDC42SE1 might be an important marker of skin cancer progression.
Collapse
Affiliation(s)
- Pazhanichamy Kalailingam
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Hui Bing Tan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| | - Jiun Yit Pan
- National Skin Centre, Singapore 308205, Singapore.
| | | | - Thirumaran Thanabalu
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
60
|
Zare K, Shademan M, Ghahramani Seno MM, Dehghani H. CRISPR/Cas9 Knockout Strategies to Ablate CCAT1 lncRNA Gene in Cancer Cells. Biol Proced Online 2018; 20:21. [PMID: 30410426 PMCID: PMC6211572 DOI: 10.1186/s12575-018-0086-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 10/11/2018] [Indexed: 02/01/2023] Open
Abstract
Background With the increasing discovery of long noncoding RNAs (lncRNAs), the application of functional techniques that could have very specific, efficient, and robust effects and readouts is necessary. Here, we have applied and analyzed three gene knockout (KO) strategies to ablate the CCAT1 gene in different colorectal adenocarcinoma cell lines. We refer to these strategies as “CRISPR excision”, “CRISPR HDR”, and “CRISPR du-HITI”. Results In order to obstruct the transcription of lncRNA or to alter its structure, in these strategies either a significant segment of the gene is removed, or a transcription termination signal is inserted in the target gene. We use RT-qPCR, RNA-seq, MTT, and colony formation assay to confirm the functional effects of CCAT1 gene ablation in knockout colorectal adenocarcinoma cell lines. We applied three different CRISPR/Cas9 mediated knockout strategies to abolish the transcription of CCAT1 lncRNA. CCAT1 knockout cells displayed dysregulation of genes involved in several biological processes, and a significant reduction for anchorage-independent growth. The du-HITI strategy introduced in this study removes a gene segment and inserts a reporter and a transcription termination signal in each of the two target alleles. The preparation of donor vector for this strategy is much easier than that in “CRISPR HDR”, and the selection of cells in this strategy is also much more practical than that in “CRISPR excision”. In addition, use of this technique in the first attempt of transfection, generates single cell knockouts for both alleles. Conclusions The strategies applied and introduced in this study can be used for the generation of CCAT1 knockout cell lines and in principle can be applied to the deletion of other lncRNAs for the study of their function. Electronic supplementary material The online version of this article (10.1186/s12575-018-0086-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Khadijeh Zare
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| | - Milad Shademan
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| | - Mohammad M Ghahramani Seno
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran.,2Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| | - Hesam Dehghani
- 1Department of Basic Sciences, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran.,2Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran.,3Stem Cell Biology and Regenerative Medicine Research Group, Research Institute of Biotechnology, Ferdowsi University of Mashhad, Azadi Square, Mashhad, Iran
| |
Collapse
|
61
|
Cell-Based Methods for Determination of Efficacy for Candidate Therapeutics in the Clinical Management of Cancer. Diseases 2018; 6:diseases6040085. [PMID: 30249005 PMCID: PMC6313784 DOI: 10.3390/diseases6040085] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/12/2018] [Accepted: 09/20/2018] [Indexed: 12/11/2022] Open
Abstract
Determination of therapeutic efficacy is a major challenge in developing treatment options for cancer. Prior to in vivo studies, candidate therapeutics are evaluated using cell-based in vitro methods to assess their anti-cancer potential. This review describes the utility and limitations of evaluating therapeutic efficacy using human tumor-derived cell lines. Indicators for therapeutic efficacy using tumor-derived cell lines include cell viability, cell proliferation, colony formation, cytotoxicity, cytostasis, induction of apoptosis, and cell cycle arrest. Cell panel screens, 3D tumor spheroid models, drug-drug/drug-radiation combinatorial analysis, and invasion/migration assays reveal analogous in vitro information. In animal models, cellular assays can assess tumor micro-environment and therapeutic delivery. The utility of tumor-derived cell lines for efficacy determination is manifest in numerous commercially approved drugs that have been applied in clinical management of cancer. Studies reveal most tumor-derived cell lines preserve the genomic signature of the primary tumor source and cell line-based data is highly predictive of subsequent clinical studies. However, cell-based data often disregards natural system components, resulting in cell autonomous outcomes. While 3D cell culture platforms can counter such limitations, they require additional time and cost. Despite the limitations, cell-based methods remain essential in early stages of anti-cancer drug development.
Collapse
|
62
|
MiR-514 attenuates proliferation and increases chemoresistance by targeting ATP binding cassette subfamily in ovarian cancer. Mol Genet Genomics 2018; 293:1159-1167. [PMID: 29752546 DOI: 10.1007/s00438-018-1447-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Cisplatin is one of the most popular chemotherapeutic drugs in treating ovarian cancer. Resistance to cisplatin is a common clinical challenge that needs to be solved to increase its anti-tumor effects. The relation of miR-514 expression with prognosis in ovarian cancer patients was analyzed based on GSE73584 datasets. The regulation of miR-514 on proliferation and cisplatin chemosensitivity of ovarian cells was examined by MTT assay, colony-formation assay and soft-agar colony-formation assay. Dual luciferase assay was performed to detect the direct interaction of miR-514 with its downstream targets. Immunobloting and qRT-PCR were performed for target gene expression analysis. Low expression of miR-514 was related to poor prognosis in ovarian cancer patients. MiR-514 repressed proliferation and decreased cisplatin chemosensitivity in ovarian cancer cells by targeting ATP binding cassette subfamily. MiR-514 is of clinically significance in ovarian cancer by attenuating proliferation of ovarian cancer cells and decreasing chemoresistance of cisplatin by targeting ATP binding cassette subfamily.
Collapse
|
63
|
Ledet MM, Anderson R, Harman R, Muth A, Thompson PR, Coonrod SA, Van de Walle GR. BB-Cl-Amidine as a novel therapeutic for canine and feline mammary cancer via activation of the endoplasmic reticulum stress pathway. BMC Cancer 2018; 18:412. [PMID: 29649984 PMCID: PMC5898062 DOI: 10.1186/s12885-018-4323-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 03/29/2018] [Indexed: 11/24/2022] Open
Abstract
Background Mammary cancer is highly prevalent in dogs and cats and results in a poor prognosis due to critically lacking viable treatment options. Recent human and mouse studies have suggested that inhibiting peptidyl arginine deiminase enzymes (PAD) may be a novel breast cancer therapy. Based on the similarities between human breast cancer and mammary cancer in dogs and cats, we hypothesized that PAD inhibitors would also be an effective treatment for mammary cancer in these animals. Methods Canine and feline mammary cancer cell lines were treated with BB-Cl-Amidine (BB-CLA) and evaluated for viability and tumorigenicity. Endoplasmic reticulum stress was tested by western blot, immunofluorescence, and quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Canine and feline mammary cancer xenograft models were created using NOD scid gamma (NSG) mice, and were treated with BB-CLA for two weeks. Results We found that BB-CLA reduced viability and tumorigenicity of canine and feline mammary cancer cell lines in vitro. Additionally, we demonstrated that BB-CLA activates the endoplasmic reticulum stress pathway in these cells by downregulating 78 kDa Glucose-regulated Protein (GRP78), a potential target in breast cancer for molecular therapy, and upregulating the downstream target gene DNA Damage Inducible Transcript 3 (DDIT3). Finally, we established a mouse xenograft model of both canine and feline mammary cancer in which we preliminarily tested the effects of BB-CLA in vivo. Conclusion We propose that our established mouse xenograft models will be useful for the study of mammary cancer in dogs and cats, and furthermore, that BB-CLA has potential as a novel therapeutic for mammary cancer in these species. Electronic supplementary material The online version of this article (10.1186/s12885-018-4323-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Melissa M Ledet
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Robyn Anderson
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Rebecca Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Aaron Muth
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Scott A Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
64
|
Nemmara VV, Subramanian V, Muth A, Mondal S, Salinger AJ, Maurais AJ, Tilvawala R, Weerapana E, Thompson PR. The Development of Benzimidazole-Based Clickable Probes for the Efficient Labeling of Cellular Protein Arginine Deiminases (PADs). ACS Chem Biol 2018; 13:712-722. [PMID: 29341591 DOI: 10.1021/acschembio.7b00957] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Citrullination is the post-translational hydrolysis of peptidyl-arginines to form peptidyl-citrulline, a reaction that is catalyzed by the protein arginine deiminases (PADs), a family of calcium-regulated enzymes. Aberrantly increased protein citrullination is associated with a slew of autoimmune diseases (e.g., rheumatoid arthritis (RA), multiple sclerosis, lupus, and ulcerative colitis) and certain cancers. Given the clear link between increased PAD activity and human disease, the PADs are therapeutically relevant targets. Herein, we report the development of next generation cell permeable and "clickable" probes (BB-Cl-Yne and BB-F-Yne) for covalent labeling of the PADs both in vitro and in cell-based systems. Using advanced chemoproteomic technologies, we also report the off targets of both BB-Cl-Yne and BB-F-Yne. The probes are highly specific for the PADs, with relatively few off targets, especially BB-F-Yne, suggesting the preferential use of the fluoroacetamidine warhead in next generation irreversible PAD inhibitors. Notably, these compounds can be used in a variety of modalities, including the identification of off targets of the parent compounds and as activity-based protein profiling probes in target engagement assays to demonstrate the efficacy of PAD inhibitors.
Collapse
Affiliation(s)
- Venkatesh V. Nemmara
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Venkataraman Subramanian
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Aaron Muth
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Santanu Mondal
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Ari J. Salinger
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Aaron J. Maurais
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Ronak Tilvawala
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| | - Eranthie Weerapana
- Department of Chemistry, Boston College, Chestnut Hill, Massachusetts 02467, United States
| | - Paul R. Thompson
- Department of Biochemistry and Molecular Pharmacology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
- Program in Chemical Biology, UMass Medical School, 364 Plantation Street, Worcester, Massachusetts 01605, United States
| |
Collapse
|
65
|
Chen Y, Zhou Q, Zhang L, Zhong Y, Fan G, Zhang Z, Wang R, Jin M, Qiu Y, Kong D. Stellettin B induces apoptosis in human chronic myeloid leukemia cells via targeting PI3K and Stat5. Oncotarget 2018; 8:28906-28921. [PMID: 28423649 PMCID: PMC5438702 DOI: 10.18632/oncotarget.15957] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 02/15/2017] [Indexed: 12/30/2022] Open
Abstract
Novel agents are still urgently expected for therapy of chronic myeloid leukemia (CML). The in vitro anti-leukemia activity of Stellettin B (Stel B), a triterpenoid we isolated from marine sponge Jaspis stellifera, on human CML K562 and KU812 cells was recently investigated. Stel B inhibited K562 and KU812 cell proliferation with IC50 as 0.035 μM and 0.95 μM respectively. While no obvious cell cycle arrest was observed, apoptosis was induced in K562 cells after Stel B treatment. The Stel B-induced apoptosis might be in mitochondrial pathway, with increase of Bad and Bax, decrease of Bcl-2 and activation of caspase-9. In addition, dose-dependent increase of reactive oxygen species (ROS) and loss of mitochondrial membrane potential (MMP) occurred. Meanwhile, Stel B inhibited phosphorylation of Stat5, expression of 4 PI3K catalytic isoforms, and phosphorylation of the downstream effectors including PDK1 and Akt, suggesting that inhibition against Stat5 and PI3K might be involved in the apoptosis-inducing effect. Combination of Stel B with Imatinib with ratio as IC50 Stel B : 5×IC50 Imatinib led to synergistic effect. Stel B might become a promising candidate for CML therapy alone or together with Imatinib.
Collapse
Affiliation(s)
- Yali Chen
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Qianxiang Zhou
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Lei Zhang
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Yuxu Zhong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China
| | - Guanwei Fan
- Institute of Traditional Chinese Medicine Research, State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Zhe Zhang
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Ran Wang
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Meihua Jin
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Yuling Qiu
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China
| | - Dexin Kong
- Department of Biopharmaceutical Sciences, Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.,Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| |
Collapse
|
66
|
Laosuwan K, Epasinghe DJ, Wu Z, Leung WK, Green DW, Jung HS. Comparison of biofilm formation and migration of Streptococcus mutans on tooth roots and titanium miniscrews. Clin Exp Dent Res 2018; 4:40-47. [PMID: 29744214 PMCID: PMC5893475 DOI: 10.1002/cre2.101] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 11/30/2022] Open
Abstract
Periodontitis and peri‐implantitis are inflammatory diseases caused by periodontal pathogenic bacteria leading to destruction of supporting periodontal/peri‐implant tissue. However, the progression of inflammatory process of these two diseases is different. The bacterial biofilm is the source of bacteria during the inflammatory process. As the bacteria migrate down the surface of tooth or titanium implant, the inflammation spreads along with it. Streptococcus mutans has an important role in oral bacterial biofilm formation in early stage biofilm before the microbiota shift to late stage and become more virulent. The other major difference is the existence of periodontal ligament (PDL) cells in normal teeth but not in peri‐implant tissue. This study aims to compare the S. mutans bacterial biofilm formation and migration on 2 different surfaces, tooth root and titanium miniscrew. The biofilm was grown with a flow cells system to imitate the oral dynamic system with PDL cells. The migration distances were measured, and the biofilm morphology was observed. Data showed that the biofilm formation on miniscrew was slower than those on tooth root at 24 hr. However, there were no difference in the morphology of the biofilm formed on the tooth root with those formed on the miniscrew at both 24 and 48 hr. The biofilm migration rate was significantly faster on miniscrew surface compare with those on tooth root when observe at 48 hr (p < .001). There are no significant differences in biofilm migration within miniscrew group and tooth root group despite the exiting of PDL cell (p > .05). The biofilm's migration rate differences on various surfaces could be one of the factors accounting for the different inflammatory progression between periodontitis and peri‐implantitis disease.
Collapse
Affiliation(s)
- Kittipong Laosuwan
- Applied Oral sciences, Faculty of Dentistry The University of Hong Kong Hong Kong
| | | | - Zhaoming Wu
- Applied Oral sciences, Faculty of Dentistry The University of Hong Kong Hong Kong
| | - Wai Keung Leung
- Periodontology, Faculty of Dentistry The University of Hong Kong Hong Kong
| | | | - Han Sung Jung
- Applied Oral sciences, Faculty of Dentistry The University of Hong Kong Hong Kong.,Dept of Oral Biology Yonsei University College of Dentistry Korea
| |
Collapse
|
67
|
Chen LY, Huang YC, Huang ST, Hsieh YC, Guan HH, Chen NC, Chuankhayan P, Yoshimura M, Tai MH, Chen CJ. Domain swapping and SMYD1 interactions with the PWWP domain of human hepatoma-derived growth factor. Sci Rep 2018; 8:287. [PMID: 29321480 PMCID: PMC5762634 DOI: 10.1038/s41598-017-18510-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 12/13/2017] [Indexed: 12/14/2022] Open
Abstract
The human hepatoma-derived growth factor (HDGF), containing the chromatin-associated N-terminal PWWP domain capable of binding the SMYD1 promoter, participates in various cellular processes and is involved in human cancers. We report the first crystal structures of the human HDGF PWWP domain (residues 1–100) in a complex with SMYD1 of 10 bp at 2.84 Å resolution and its apo form at 3.3 Å, respectively. The structure of the apo PWWP domain comprises mainly four β-strands and two α-helices. The PWWP domain undergoes domain swapping to dramatically transform its secondary structures, altering the overall conformation from monomeric globular folding into an extended dimeric structure upon DNA binding. The flexible loop2, as a hinge loop with the partially built structure in the apo PWWP domain, notably refolds into a visible and stable α-helix in the DNA complex. The swapped PWWP domain interacts with the minor grooves of the DNA through residues Lys19, Gly22, Arg79 and Lys80 in varied ways on loops 1 and 4 of the two chains, and the structure becomes more rigid than the apo form. These novel structural findings, together with physiological and activity assays of HDGF and the PWWP domain, provide new insights into the DNA-binding mechanism of HDGF during nucleosomal functions.
Collapse
Affiliation(s)
- Li-Ying Chen
- Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan.,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan.,Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Yen-Chieh Huang
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Shih-Tsung Huang
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, Academia Sinica, Taipei, 11529, Taiwan
| | - Yin-Cheng Hsieh
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Hong-Hsiang Guan
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Nai-Chi Chen
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Phimonphan Chuankhayan
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Masato Yoshimura
- Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan
| | - Ming-Hong Tai
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, 804, Taiwan.
| | - Chun-Jung Chen
- Institute of Biotechnology, National Cheng Kung University, Tainan, 701, Taiwan. .,Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan, 701, Taiwan. .,Life Science Group, Scientific Research Division, National Synchrotron Radiation Research Center, Hsinchu, 30076, Taiwan. .,Department of Physics, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
68
|
Development of Activity-Based Proteomic Probes for Protein Citrullination. Curr Top Microbiol Immunol 2018; 420:233-251. [PMID: 30203394 DOI: 10.1007/82_2018_132] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Protein arginine deiminases (PADs) catalyze the post-translational deimination of peptidyl arginine to form peptidyl citrulline. This modification is increased in multiple inflammatory diseases and in certain cancers. PADs regulate a variety of signaling pathways including apoptosis, terminal differentiation, and transcriptional regulation. Activity-based protein profiling (ABPP) probes have been developed to understand the role of the PADs in vivo and to investigate the effect of protein citrullination in various pathological conditions. Furthermore, these ABPPs have been utilized as a platform for high-throughput inhibitor discovery. This review will showcase the development of ABPPs targeting the PADs. In addition, it provides a brief overview of PAD structure and function along with recent advances in PAD inhibitor development.
Collapse
|
69
|
Liu XR, Zhou LH, Hu JX, Liu LM, Wan HP, Zhang XQ. UNC0638, a G9a inhibitor, suppresses epithelial‑mesenchymal transition‑mediated cellular migration and invasion in triple negative breast cancer. Mol Med Rep 2017; 17:2239-2244. [PMID: 29207160 PMCID: PMC5783465 DOI: 10.3892/mmr.2017.8190] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/05/2017] [Indexed: 01/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with an aggressive clinical history, high risk of recurrence and metastasis, and shorter patient survival due to lack of targeted therapy. In the present study, UNC0638, a chemical G9a inhibitor, was identified to suppress TNBC cell invasion and migration in vitro at a non-cytotoxic concentration. In addition, UNC0638 reduced the size and number of the tumorsphere and decreased anchor-independent colony formation in the two TNBC cell lines. Evaluation of the underlying mechanism revealed that the suppressive effect of UNC0638 is associated with modulation of epithelial-mesenchymal transition through enhancing E-cadherin promoter activities and restoring its expression. Thus, the current data indicates that UNC0638 may be developed as a chemotherapeutic agent to effectively treat metastatic cancers, including TNBC.
Collapse
Affiliation(s)
- Xiang-Rong Liu
- Department of Pharmacy, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Hua Zhou
- Department of Pharmacy, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Jian-Xin Hu
- Department of Pharmacy, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Li-Min Liu
- Department of Pharmacy, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Hui-Ping Wan
- Department of Tumor, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| | - Xi-Quan Zhang
- Department of Tumor, The People's Hospital of Jiangxi Province, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|
70
|
Li W, Zhou J, Chen Y, Zhang G, Jiang P, Hong L, Shen Y, Wang X, Gong X. Cigarette smoke enhances initiation and progression of lung cancer by mutating Notch1/2 and dysregulating downstream signaling molecules. Oncotarget 2017; 8:115128-115139. [PMID: 29383147 PMCID: PMC5777759 DOI: 10.18632/oncotarget.22924] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 06/19/2017] [Indexed: 02/07/2023] Open
Abstract
Lung cancer is the primary cause of cancer related deaths in the western world and smoking significantly increases the risk of developing lung cancer. Smoking enhances lung cancer initiation and progression. The effects of cigarette smoke on lung cancer are mediated by the presence of highly mutagenic substances, including nicotine, leading to mutations in oncogenes and tumor suppressor genes. An emerging pathway in cancer is the Notch signaling pathway which is essential for embryonic lung development and tissue homeostasis. The role of Notch signaling in lung cancer remains controversial and no studies have directly linked cigarette exposure to mutations in Notch. Therefore, we investigated the direct effect of Notch signaling pathways on cigarette-induced lung tumors and the correlation between smoking and mutations in Notch leading to altered downstream signaling. Human cell lines, mouse models and clinical lung cancer samples were utilized in this study. Cigarette-induced in vitro human lung cancer models and in vivo mouse models demonstrated strong effects of cigarette exposure on the Notch signaling pathway. Immunohistochemistry (IHC) of 50 clinical samples collected from smokers and non-smokers with and without lung cancer also demonstrated a link between smoking and changes in Notch signaling. Finally, 34 lung cancer samples analyzed through direct sequencing indicated smoking significantly increased small nucleotide polymorphisms (SNPs) in Notch 1 and 2 and specific SNPs significantly modulated expression levels of downstream signaling pathway molecules. Taken together, these results demonstrate a direct effect of smoking on the Notch signaling pathway leading to lung cancer initiation and progression.
Collapse
Affiliation(s)
- Wei Li
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Provincial Key Laboratory of Respiratory Disease in Anhui, Bengbu 233004, China
| | - Jihong Zhou
- Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu 233004, China
| | - Yuqing Chen
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Provincial Key Laboratory of Respiratory Disease in Anhui, Bengbu 233004, China
| | - Gengyan Zhang
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu 233004, China
| | - Peng Jiang
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Department of Biochemistry and Molecular Biology, Bengbu Medical College, Bengbu 233004, China
| | - Lei Hong
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Provincial Key Laboratory of Respiratory Disease in Anhui, Bengbu 233004, China
| | - Yuangbing Shen
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Provincial Key Laboratory of Respiratory Disease in Anhui, Bengbu 233004, China
| | - Xiaojing Wang
- Department of Respiratory Disease, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China.,Provincial Key Laboratory of Respiratory Disease in Anhui, Bengbu 233004, China
| | - Xiaomeng Gong
- Department of Pathology, The First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| |
Collapse
|
71
|
Sandoval-Bórquez A, Polakovicova I, Carrasco-Véliz N, Lobos-González L, Riquelme I, Carrasco-Avino G, Bizama C, Norero E, Owen GI, Roa JC, Corvalán AH. MicroRNA-335-5p is a potential suppressor of metastasis and invasion in gastric cancer. Clin Epigenetics 2017; 9:114. [PMID: 29075357 PMCID: PMC5645854 DOI: 10.1186/s13148-017-0413-8] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 10/02/2017] [Indexed: 12/13/2022] Open
Abstract
Background Multiple aberrant microRNA expression has been reported in gastric cancer. Among them, microRNA-335-5p (miR-335), a microRNA regulated by DNA methylation, has been reported to possess both tumor suppressor and tumor promoter activities. Results Herein, we show that miR-335 levels are reduced in gastric cancer and significantly associate with lymph node metastasis, depth of tumor invasion, and ultimately poor patient survival in a cohort of Amerindian/Hispanic patients. In two gastric cancer cell lines AGS and, Hs 746T the exogenous miR-335 decreases migration, invasion, viability, and anchorage-independent cell growth capacities. Performing a PCR array on cells transfected with miR-335, 19 (30.6%) out of 62 genes involved in metastasis and tumor invasion showed decreased transcription levels. Network enrichment analysis narrowed these genes to nine (PLAUR, CDH11, COL4A2, CTGF, CTSK, MMP7, PDGFA, TIMP1, and TIMP2). Elevated levels of PLAUR, a validated target gene, and CDH11 were confirmed in tumors with low expression of miR-335. The 3′UTR of CDH11 was identified to be directly targeted by miR-335. Downregulation of miR-335 was also demonstrated in plasma samples from gastric cancer patients and inversely correlated with DNA methylation of promoter region (Z = 1.96, p = 0.029). DNA methylation, evaluated by methylation-specific PCR assay, was found in plasma from 23 (56.1%) out of 41 gastric cancer patients but in only 9 (30%) out of 30 healthy donors (p = 0.029, Pearson’s correlation). Taken in consideration, our results of the association with depth of invasion, lymph node metastasis, and poor prognosis together with functional assays on cell migration, invasion, and tumorigenicity are in accordance with the downregulation of miR-335 in gastric cancer. Conclusions Comprehensive evaluation of metastasis and invasion pathway identified a subset of associated genes and confirmed PLAUR and CDH11, both targets of miR-335, to be overexpressed in gastric cancer tissues. DNA methylation of miR-335 may be a promissory strategy for non-invasive approach to gastric cancer. Electronic supplementary material The online version of this article (10.1186/s13148-017-0413-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alejandra Sandoval-Bórquez
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Molecular Pathology, Department of Pathology, School of Medicine, BIOREN-CEGIN, and Graduate Program in Applied Cell and Molecular Biology, Universidad de La Frontera, Temuco, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Iva Polakovicova
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás Carrasco-Véliz
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Instituto de Química, Faculty of Science, Pontificia Universidad Católica de Valparaíso, Valparaiso, Chile
| | - Lorena Lobos-González
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile.,Fundación Ciencia y Vida, Parque Biotecnológico, Santiago, Chile
| | - Ismael Riquelme
- Laboratory of Molecular Pathology, Department of Pathology, School of Medicine, BIOREN-CEGIN, and Graduate Program in Applied Cell and Molecular Biology, Universidad de La Frontera, Temuco, Chile
| | - Gonzalo Carrasco-Avino
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Pathology, Faculty of Medicine, Hospital Clínico Universidad de Chile, Santiago, Chile
| | - Carolina Bizama
- Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Enrique Norero
- Esophagogastric Surgery Unit, Hospital Dr. Sótero del Río, Santiago, Chile.,Digestive Surgery Department, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gareth I Owen
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Juan C Roa
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Laboratory of Molecular Pathology, Department of Pathology, School of Medicine, BIOREN-CEGIN, and Graduate Program in Applied Cell and Molecular Biology, Universidad de La Frontera, Temuco, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Pathology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alejandro H Corvalán
- Advanced Center for Chronic Diseases (ACCDiS), Pontificia Universidad Católica de Chile, Santiago, Chile.,Center UC for Investigational in Oncology (CITO), Pontificia Universidad Católica de Chile, Santiago, Chile.,Department of Hematology-Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
72
|
Segovia-Mendoza M, Díaz L, Prado-Garcia H, Reginato MJ, Larrea F, García-Becerra R. The addition of calcitriol or its synthetic analog EB1089 to lapatinib and neratinib treatment inhibits cell growth and promotes apoptosis in breast cancer cells. Am J Cancer Res 2017; 7:1486-1500. [PMID: 28744399 PMCID: PMC5523030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/01/2017] [Indexed: 06/07/2023] Open
Abstract
In breast cancer the use of small molecule inhibitors of tyrosine kinase activity of the ERBB family members improves survival thus represents a valuable therapeutic strategy. The addition of calcitriol, the most active metabolite of vitamin D, or some of its analogs, to conventional anticancer drugs, including tyrosine kinase inhibitors (TKIs), has shown an increased effect on the inhibition of cancer cell growth. In this work, we have evaluated the effects and the mechanism of action of the combination of calcitriol or its analog EB1089 with lapatinib or neratinib on EGFR and/or HER2 positive breast cancer cell lines. Lapatinib, neratinib, calcitriol and EB1089 inhibited breast cancer cell proliferation in a concentration-dependent manner. Addition of calcitriol or EB1089 to TKIs treatment induced more effective inhibiting effect on cell growth and AKT and MAPK phosphorylation than all compounds alone. The combined treatments incremented also the expression of active caspase 3 and induced cell death in two and three-dimensional cell culture and significantly inhibited anchorage-independent colony formation. Our results suggest that the addition of calcitriol or its analog EB1089 to conventional targeted therapies, including lapatinib or neratinib might be of benefit to patients with breast cancer, particularly those with an EGFR and/or HER2 positive phenotype.
Collapse
Affiliation(s)
- Mariana Segovia-Mendoza
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránVasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, México, Ciudad de México
| | - Lorenza Díaz
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránVasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, México, Ciudad de México
| | - Heriberto Prado-Garcia
- Departamento de Enfermedades Crónico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosío Villegas”Calzada de Tlalpan 4502, Belisario Domínguez Sección XVI, Tlalpan 14080, México, Ciudad de México
| | - Mauricio J Reginato
- Department of Biochemistry and Molecular Biology, College of Medicine, Drexel UniversityPhiladelphia, PA, USA
| | - Fernando Larrea
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránVasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, México, Ciudad de México
| | - Rocío García-Becerra
- Departamento de Biología de la Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador ZubiránVasco de Quiroga No. 15, Belisario Domínguez Sección XVI, Tlalpan 14080, México, Ciudad de México
| |
Collapse
|
73
|
Horibata S, Rogers KE, Sadegh D, Anguish LJ, McElwee JL, Shah P, Thompson PR, Coonrod SA. Role of peptidylarginine deiminase 2 (PAD2) in mammary carcinoma cell migration. BMC Cancer 2017; 17:378. [PMID: 28549415 PMCID: PMC5446677 DOI: 10.1186/s12885-017-3354-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 05/15/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Penetration of the mammary gland basement membrane by cancer cells is a crucial first step in tumor invasion. Using a mouse model of ductal carcinoma in situ, we previously found that inhibition of peptidylarginine deiminase 2 (PAD2, aka PADI2) activity appears to maintain basement membrane integrity in xenograft tumors. The goal of this investigation was to gain insight into the mechanisms by which PAD2 mediates this process. METHODS For our study, we modulated PAD2 activity in mammary ductal carcinoma cells by lentiviral shRNA-mediated depletion, lentiviral-mediated PAD2 overexpression, or PAD inhibition and explored the effects of these treatments on changes in cell migration and cell morphology. We also used these PAD2-modulated cells to test whether PAD2 may be required for EGF-induced cell migration. To determine how PAD2 might promote tumor cell migration in vivo, we tested the effects of PAD2 inhibition on the expression of several cell migration mediators in MCF10DCIS.com xenograft tumors. In addition, we tested the effect of PAD2 inhibition on EGF-induced ductal invasion and elongation in primary mouse mammary organoids. Lastly, using a transgenic mouse model, we investigated the effects of PAD2 overexpression on mammary gland development. RESULTS Our results indicate that PAD2 depletion or inhibition suppresses cell migration and alters the morphology of MCF10DCIS.com cells. In addition, we found that PAD2 depletion suppresses the expression of the cytoskeletal regulatory proteins RhoA, Rac1, and Cdc42 and also promotes a mesenchymal to epithelial-like transition in tumor cells with an associated increase in the cell adhesion marker, E-cadherin. Our mammary gland organoid study found that inhibition of PAD2 activity suppresses EGF-induced ductal invasion. In vivo, we found that PAD2 overexpression causes hyperbranching in the developing mammary gland. CONCLUSION Together, these results suggest that PAD2 plays a critical role in breast cancer cell migration. Our findings that EGF treatment increases protein citrullination and that PAD2 inhibition blocks EGF-induced cell migration suggest that PAD2 likely functions within the EGF signaling pathway to mediate cell migration.
Collapse
Affiliation(s)
- Sachi Horibata
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA.,Department of Molecular Medicine, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Katherine E Rogers
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - David Sadegh
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Lynne J Anguish
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - John L McElwee
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Pragya Shah
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Scott A Coonrod
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14850, USA.
| |
Collapse
|
74
|
Matak D, Brodaczewska KK, Lipiec M, Szymanski Ł, Szczylik C, Czarnecka AM. Colony, hanging drop, and methylcellulose three dimensional hypoxic growth optimization of renal cell carcinoma cell lines. Cytotechnology 2017; 69:565-578. [PMID: 28321776 PMCID: PMC5507837 DOI: 10.1007/s10616-016-0063-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/28/2016] [Indexed: 12/20/2022] Open
Abstract
Renal cell carcinoma (RCC) is the most lethal of the common urologic malignancies, comprising 3% of all human neoplasms, and the incidence of kidney cancer is rising annually. We need new approaches to target tumor cells that are resistant to current therapies and that give rise to recurrence and treatment failure. In this study, we focused on low oxygen tension and three-dimensional (3D) cell culture incorporation to develop a new RCC growth model. We used the hanging drop and colony formation methods, which are common in 3D culture, as well as a unique methylcellulose (MC) method. For the experiments, we used human primary RCC cell lines, metastatic RCC cell lines, human kidney cancer stem cells, and human healthy epithelial cells. In the hanging drop assay, we verified the potential of various cell lines to create solid aggregates in hypoxic and normoxic conditions. With the semi-soft agar method, we also determined the ability of various cell lines to create colonies under different oxygen conditions. Different cell behavior observed in the MC method versus the hanging drop and colony formation assays suggests that these three assays may be useful to test various cell properties. However, MC seems to be a particularly valuable alternative for 3D cell culture, as its higher efficiency of aggregate formation and serum independency are of interest in different areas of cancer biology.
Collapse
Affiliation(s)
- Damian Matak
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia K Brodaczewska
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Monika Lipiec
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,Faculty of Pharmacy with Laboratory Medicine Division, Medical University of Warsaw, Warsaw, Poland
| | - Łukasz Szymanski
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.,Institute of Genetics and Biotechnology, Faculty of Biology, University of Warsaw, Warsaw, Poland.,Department of Microwave Safety, Military Institute of Hygiene and Epidemiology, Warsaw, Poland
| | - Cezary Szczylik
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland
| | - Anna M Czarnecka
- Laboratory of Molecular Oncology, Department of Oncology, Military Institute of Medicine, Szaserow 128, 04-141, Warsaw, Poland.
| |
Collapse
|
75
|
Young CH, Rothfuss HM, Gard PF, Muth A, Thompson PR, Ashley RL, Cherrington BD. Citrullination regulates the expression of insulin-like growth factor-binding protein 1 (IGFBP1) in ovine uterine luminal epithelial cells. Reproduction 2016; 153:1-10. [PMID: 29565015 DOI: 10.1530/rep-16-0494] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 09/28/2016] [Accepted: 10/10/2016] [Indexed: 01/19/2023]
Abstract
There are five peptidylarginine deiminase (PAD) isozymes designated as PADs 1, 2, 3, 4 and 6, and many are expressed in female reproductive tissues. These enzymes post-translationally convert positively charged arginine amino acids into neutral citrulline residues. Targets for PAD-catalyzed citrullination include arginine residues on histone tails, which results in chromatin decondensation and changes in gene expression. Some of the first studies examining PADs found that they are localized to rodent uterine epithelial cells. Despite these findings, the function of PAD-catalyzed citrullination in uterine epithelial cells is still unknown. To address this, we first examined PAD expression in uterine cross-sections from pregnant ewes on gestation day 25 (d25). Immunohistochemistry revealed that the levels of PADs 2 and 4 are robust in luminal and glandular epithelia compared with those of PADs 1 and 3. As PADs 2 and 4 have well-characterized roles in histone citrullination, we next hypothesized that PADs citrullinate histones in these uterine cells. Examination of caruncle lysates from pregnant ewes on gestation d25 and an ovine luminal epithelial (OLE) cell line shows that histone H3 arginine residues 2, 8, 17 and 26 are citrullinated, but histone H4 arginine 3 is not. Using a pan-PAD inhibitor, we next attenuated histone citrullination in OLE cells, which resulted in a significant decrease in the expression of insulin-like growth factor-binding protein 1 (IGFBP1) mRNA. As IGFBP1 is important for the migration and attachment of the trophectoderm to uterine endometrium, our results suggest that PAD-catalyzed citrullination may be an important post-translational mechanism for the establishment of pregnancy in ewes.
Collapse
Affiliation(s)
- Coleman H Young
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Heather M Rothfuss
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Philip F Gard
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| | - Aaron Muth
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Paul R Thompson
- Department of Biochemistry and Molecular PharmacologyUniversity of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Ryan L Ashley
- Department of Animal and Range SciencesNew Mexico State University, Las Cruces, New Mexico, USA
| | - Brian D Cherrington
- Department of Zoology and PhysiologyUniversity of Wyoming, Laramie, Wyoming, USA
| |
Collapse
|
76
|
Xu E, Yu X, Zeng Q, Qiao G, Xuan Y, Tang Y, Zhu Q, He J. Functional role of lncRNA DB327252 in lung cancer. J Thorac Dis 2016; 8:2793-2802. [PMID: 27867555 DOI: 10.21037/jtd.2016.10.44] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Lung cancer becomes a concerning health issue and is considered one of the most deadly cancers in the worldwide. Most recently, long non-coding RNAs (lncRNAs) are newfound non-coding RNAs that are thought as one of the major players in a range of biological processes of human diseases. Although lncRNAs are involved in numerous cancer types, the precise understandings of lncRNAs' functional roles and mechanisms in lung cancer are limited. In this study, we looked for lung cancer related lncRNAs. METHODS The quantitative reverse transcriptase polymerase chain reaction (qRT-PCR) technique was utilized to investigate the lncRNA DB327252 expression in 91 paired clinical lung cancer tissues and related cell lines. Moreover, its biological functions were also evaluated in the development of lung cancer. RESULTS The results indicated that the expression of DB327252 was up-regulated in lung cancer tissues compared to the cancer-adjacent normal tissues (P<0.05); and the up-regulated expression is likely to relate to those with bigger tumor size, adenocarcinoma and advanced TNM stage (P<0.05). In addition, the knockdown of DB327252 inhibited the growth and proliferation of tumor cell in vitro and in vivo. According to the observation from our study, we found that the knockdown of the DB327252 expression, led to G0/G1 phase cell-cycle arrested, colony formation suppressed in vitro, and tumor growth inhibited in a nude mouse xenograft model. Our experimental results also suggest that lncRNA DB327252 may be a lncRNA related to lung cancer and acts an important role in A549 and 16HBE-T cancer cells, which provides evidence that DB327252 has an oncogene-like function in lung cancer. CONCLUSIONS The lncRNA DB327252 is up-regulated in lung cancer, and its expression implies that it was probable related to biologic functions of lung cancer.
Collapse
Affiliation(s)
- Enwu Xu
- Southern Medical University, Guangzhou 510515, China;; Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China;; Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Xuanwei Yu
- Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 510182, China
| | - Qian Zeng
- Southern Medical University, Guangzhou 510515, China
| | - Guibin Qiao
- Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China
| | - Yiwen Xuan
- Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China
| | - Yong Tang
- Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China
| | - Qihang Zhu
- Department of Thoracic Surgery, General Hospital of Guangzhou Military Command of Chinese People's Liberation Army, Guangzhou 510010, China
| | - Jianxing He
- Southern Medical University, Guangzhou 510515, China;; Department of Thoracic Surgery, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| |
Collapse
|
77
|
Khan SA, Edwards BS, Muth A, Thompson PR, Cherrington BD, Navratil AM. GnRH Stimulates Peptidylarginine Deiminase Catalyzed Histone Citrullination in Gonadotrope Cells. Mol Endocrinol 2016; 30:1081-1091. [PMID: 27603413 DOI: 10.1210/me.2016-1085] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Peptidylarginine deiminase (PAD) enzymes convert histone tail arginine residues to citrulline resulting in chromatin decondensation. Our previous work found that PAD isoforms are expressed in female reproductive tissues in an estrous cycle-dependent fashion, but their role in the anterior pituitary gland is unknown. Thus, we investigated PAD expression and function in gonadotrope cells. The gonadotrope-derived LβT2 cell line strongly expresses PAD2 at the protein level compared with other PAD isoforms. Consistent with this, PAD2 protein expression is highest during the estrous phase of the estrous cycle and colocalizes with the LH β-subunit in the mouse pituitary. Using the GnRH agonist buserelin (GnRHa), studies in LβT2 and mouse primary gonadotrope cells revealed that 30 minutes of stimulation caused distinct puncta of PAD2 to localize in the nucleus. Once in the nucleus, GnRHa stimulated PAD2 citrullinates histone H3 tail arginine residues at sites 2, 8, and 17 within 30 minutes; however, this effect and PAD2 nuclear localization was blunted by incubation of the cells with the pan-PAD inhibitor, biphenyl-benzimidazole-Cl-amidine. Given that PAD2 citrullinates histones in gonadotropes, we next analyzed the functional consequence of PAD2 inhibition on gene expression. Our results show that GnRHa stimulates an increase in LHβ and FSHβ mRNA and that this response is significantly reduced in the presence of the PAD inhibitor biphenyl-benzimidazole-Cl-amidine. Overall, our data suggest that GnRHa stimulates PAD2-catalyzed histone citrullination in gonadotropes to epigenetically regulate gonadotropin gene expression.
Collapse
Affiliation(s)
- Shaihla A Khan
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Brian S Edwards
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Aaron Muth
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Paul R Thompson
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Brian D Cherrington
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | - Amy M Navratil
- Department of Zoology and Physiology (S.A.K., B.S.E., B.D.C., A.M.N.), University of Wyoming, Laramie, Wyoming 82071; and Department of Biochemistry and Molecular Pharmacology (A.M., P.R.T.), University of Massachusetts Medical School, Worcester, Massachusetts 01605
| |
Collapse
|
78
|
Regev M, Sabanay H, Kartvelishvily E, Kam Z, Bershadsky AD. Involvement of Rho GAP GRAF1 in maintenance of epithelial phenotype. Cell Adh Migr 2016; 11:367-383. [PMID: 27588930 PMCID: PMC5569970 DOI: 10.1080/19336918.2016.1227910] [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] [Indexed: 12/15/2022] Open
Abstract
Adhesion of epithelial cell to each other and to extracellular matrix, as well as cell migration ability and cytoskeleton organization undergo significant alterations in the course of neoplastic transformation, but regulatory mechanisms involved in these processes are not fully understood. Here, we studied the role of a Rho GAP protein GRAF1 (GTPase Regulator Associated with Focal adhesion kinase-1) in the regulation of the epithelial phenotype in cells of breast derived, non-malignant, MCF10A cell line. GRAF1 was shown to be localized to cell-cell junctions, and its depletion resulted in accelerated cell migration velocity, elongation of the cells and cell colonies, impaired monolayer integrity and significant disruption of desmosomes with a loss of associated keratin filaments. These processes were accompanied by formation of larger focal adhesions, an increased number of contractile actin stress fibers, reduction in epithelial markers and increase in mesenchymal markers such as epithelial-mesenchymal transition (EMT)-specific transcription factors Snail-1 and Snail-2, as well as N-cadherin, and vimentin. Moreover, unlike control cells, GRAF1 knocked-down cells demonstrated anchorage-independent growth in soft agar. GRAF1 expression in several highly invasive breast cancer cell lines was low, as compared to the non-malignant MCF10A cells, while overexpressing of GRAF1 in the malignant BT-549 cell line led to a decrease of mesenchymal markers, especially the Snail-1 and 2. Altogether, our analysis suggests that GRAF1 plays a role in the maintenance of normal epithelial phenotype and its depletion leads to an EMT-like process that might be involved in neoplastic transformation.
Collapse
Affiliation(s)
- Miriam Regev
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel.,b The Chaim Sheba Medical Center, Tel-Hashomer Hospital , Israel
| | - Helena Sabanay
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Elena Kartvelishvily
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Zvi Kam
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel
| | - Alexander D Bershadsky
- a Department of Molecular Cell Biology , Weizmann Institute of Science , Rehovot , Israel.,c Mechanobiology Institute, National University of Singapore , Singapore
| |
Collapse
|
79
|
Zhou Q, Chai W. Suppression of STN1 enhances the cytotoxicity of chemotherapeutic agents in cancer cells by elevating DNA damage. Oncol Lett 2016; 12:800-808. [PMID: 27446354 PMCID: PMC4950739 DOI: 10.3892/ol.2016.4676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 04/22/2016] [Indexed: 11/06/2022] Open
Abstract
DNA damage-inducing agents are among the most effective treatment regimens in clinical chemotherapy. However, drug resistance and severe side effects caused by these agents greatly limit their efficacy. Sensitizing malignant cells to chemotherapeutic agents has long been a goal of chemotherapy. In the present study, suppression of STN1, a gene important for safeguarding genome stability, potentiated the anticancer effect of chemotherapeutic agents in tumor cells. Using multiple cancer cells from a variety of origins, it was observed that downregulation of STN1 resulted in a significant decrease in the half maximal inhibitory concentration values of several conventional anticancer agents. When cells are treated with anticancer agents, STN1 suppression leads to a decline in colony formation and diminished anchorage-independent growth. Furthermore, it was additionally observed that STN1 knockdown augmented the levels of DNA damage caused by damage-inducing agents. The present study concluded that suppression of STN1 enhances the cytotoxicity of damage-inducing chemotherapeutic agents by increasing DNA damage in cancer cells.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Biomedical Sciences, Elson S. Floyd College of Medical Sciences, Washington State University, Spokane, WA 99210, USA
| | - Weihang Chai
- Department of Biomedical Sciences, Elson S. Floyd College of Medical Sciences, Washington State University, Spokane, WA 99210, USA; School of Molecular Biosciences, Washington State University, Spokane, WA 99210, USA
| |
Collapse
|
80
|
Vishnoi M, Peddibhotla S, Yin W, T Scamardo A, George GC, Hong DS, Marchetti D. The isolation and characterization of CTC subsets related to breast cancer dormancy. Sci Rep 2015; 5:17533. [PMID: 26631983 PMCID: PMC4668355 DOI: 10.1038/srep17533] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 10/30/2015] [Indexed: 02/08/2023] Open
Abstract
Uncovering CTCs phenotypes offer the promise to dissect their heterogeneity related to metastatic competence. CTC survival rates are highly variable and this can lead to many questions as yet unexplored properties of CTCs responsible for invasion and metastasis vs dormancy. We isolated CTC subsets from peripheral blood of patients diagnosed with or without breast cancer brain metastasis. CTC subsets were selected for EpCAM negativity but positivity for CD44(+)/CD24(-) stem cell signature; along with combinatorial expression of uPAR and int β1, two markers directly implicated in breast cancer dormancy mechanisms. CTC subsets were cultured in vitro generating 3D CTC tumorspheres which were interrogated for biomarker profiling and biological characteristics. We identified proliferative and invasive properties of 3D CTC tumorspheres distinctive upon uPAR/int β1 combinatorial expression. The molecular characterization of uPAR/int β1 CTC subsets may enhance abilities to prospectively identify patients who may be at high risk of developing BCBM.
Collapse
Affiliation(s)
- Monika Vishnoi
- Biomarker Research Program Center, Houston Methodist Research Institute, Houston, TX
| | - Sirisha Peddibhotla
- Department of Pathology &Immunology, Baylor College of Medicine, Houston, TX
| | - Wei Yin
- Biomarker Research Program Center, Houston Methodist Research Institute, Houston, TX
| | - Antonio T Scamardo
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Goldy C George
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Dario Marchetti
- Biomarker Research Program Center, Houston Methodist Research Institute, Houston, TX.,Department of Molecular &Cellular Biology and The Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| |
Collapse
|
81
|
Kashyap R, Roucourt B, Lembo F, Fares J, Carcavilla AM, Restouin A, Zimmermann P, Ghossoub R. Syntenin controls migration, growth, proliferation, and cell cycle progression in cancer cells. Front Pharmacol 2015; 6:241. [PMID: 26539120 PMCID: PMC4612656 DOI: 10.3389/fphar.2015.00241] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 10/07/2015] [Indexed: 11/25/2022] Open
Abstract
The scaffold protein syntenin abounds during fetal life where it is important for developmental movements. In human adulthood, syntenin gain-of-function is increasingly associated with various cancers and poor prognosis. Depending on the cancer model analyzed, syntenin affects various signaling pathways. We previously have shown that syntenin allows syndecan heparan sulfate proteoglycans to escape degradation. This indicates that syntenin has the potential to support sustained signaling of a plethora of growth factors and adhesion molecules. Here, we aim to clarify the impact of syntenin loss-of-function on cancer cell migration, growth, and proliferation, using cells from various cancer types and syntenin shRNA and siRNA silencing approaches. We observed decreased migration, growth, and proliferation of the mouse melanoma cell line B16F10, the human colon cancer cell line HT29 and the human breast cancer cell line MCF7. We further documented that syntenin controls the presence of active β1 integrin at the cell membrane and G1/S cell cycle transition as well as the expression levels of CDK4, Cyclin D2, and Retinoblastoma proteins. These data confirm that syntenin supports the migration and growth of tumor cells, independently of their origin, and further highlight the attractiveness of syntenin as potential therapeutic target.
Collapse
Affiliation(s)
- Rudra Kashyap
- Laboratory for Signal Integration in Cell Fate Decision, Department of Human Genetics, KU Leuven Leuven, Belgium ; Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| | - Bart Roucourt
- Laboratory for Signal Integration in Cell Fate Decision, Department of Human Genetics, KU Leuven Leuven, Belgium
| | - Frederique Lembo
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| | - Joanna Fares
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| | - Ane Marcos Carcavilla
- Laboratory for Signal Integration in Cell Fate Decision, Department of Human Genetics, KU Leuven Leuven, Belgium
| | - Audrey Restouin
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| | - Pascale Zimmermann
- Laboratory for Signal Integration in Cell Fate Decision, Department of Human Genetics, KU Leuven Leuven, Belgium ; Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| | - Rania Ghossoub
- Centre de Recherche en Cancérologie de Marseille, Aix-Marseille Université Marseille, France ; Inserm U1068, Institut Paoli-Calmettes Marseille, France ; Centre National de la Recherche Scientifique, UMR7258 Marseille, France
| |
Collapse
|