1
|
Wang H, Li Y, Wu N, Lv C, Wang Y. P4HB regulates the TGFβ/SMAD3 signaling pathway through PRMT1 to participate in high glucose-induced epithelial-mesenchymal transition and fibrosis of renal tubular epithelial cells. BMC Nephrol 2024; 25:297. [PMID: 39251943 PMCID: PMC11385120 DOI: 10.1186/s12882-024-03733-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 08/26/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a common complication of diabetes mellitus, and Prolyl 4-Hydroxylase Subunit Beta (P4HB) expression is increased in high glucose (HG)-induced renal tubular epithelial cells (TECs). But it's role in HG-induced TECs remains to be elucidated. METHODS The HK-2 cells were induced using HG and transfected with SiRNA-P4HB. DCFH-DA staining was utilized for the detection of cellular levels of ROS. WB and immunofluorescence were utilized to detect the expression of P4HB, epithelial-mesenchymal transition (EMT), fibrosis, and TGFβ/SMAD3-related proteins in HK-2 cells. Online databases were utilized for predicting the interaction target of P4HB, and immunoprecipitation (IP) experiments were employed to validate the binding of P4HB with the target. SiRNA and overexpression vectors of target gene were used to verify the mechanism of action of P4HB. RESULTS HG induced an increase in the expression of P4HB and TGFβ, p-SMAD3, and ROS in HK-2 cells. Furthermore, HG downregulated the expression of E-cadherin and upregulated the expression of N-cadherin, Vimentin, α-SMA, Fibronectin, Collagen IV, SNAIL, and SLUG in HK-2 cells. Interfering with P4HB significantly reversed the expression of these proteins. Database predictions and IP experiments showed that P4HB interacts with PRMT1, and the expression of PRMT1 was increased in HG-induced HK-2 cells. Interfering with PRMT1 inhibited the changes in expression of EMT and fibrosis related proteins induced by HG. However, overexpression of PRMT1 weakened the regulatory effect of P4HB interference on the EMT, fibrosis, and TGFβ/SMAD3-related proteins in HK-2 cells. CONCLUSION P4HB regulated the TGFβ/SMAD3 signaling pathway through PRMT1 and thus participates in HG-induced EMT and fibrosis in HK-2 cells.
Collapse
Affiliation(s)
- Haifeng Wang
- Department of nephrology, China-Japan Friendship Hospital, chaoyang District, 100029, Beijing, China
| | - Yang Li
- Comprehensive Internal Medicine Department, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping District, 102211, Beijing, China
| | - Na Wu
- Comprehensive Internal Medicine Department, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping District, 102211, Beijing, China
| | - Chunmei Lv
- Comprehensive Internal Medicine Department, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping District, 102211, Beijing, China
| | - Yishu Wang
- Comprehensive Internal Medicine Department, Beijing Xiaotangshan Hospital, Xiaotangshan Town, Changping District, 102211, Beijing, China.
| |
Collapse
|
2
|
Son DS, Done KA, Son J, Izban MG, Virgous C, Lee ES, Adunyah SE. Intermittent Fasting Attenuates Obesity-Induced Triple-Negative Breast Cancer Progression by Disrupting Cell Cycle, Epithelial-Mesenchymal Transition, Immune Contexture, and Proinflammatory Signature. Nutrients 2024; 16:2101. [PMID: 38999849 PMCID: PMC11243652 DOI: 10.3390/nu16132101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/22/2024] [Accepted: 06/25/2024] [Indexed: 07/14/2024] Open
Abstract
Obesity is associated with one-fifth of cancer deaths, and breast cancer is one of the obesity-related cancers. Triple-negative breast cancer (TNBC) lacks estrogen and progesterone receptors and human epidermal growth factor receptor 2, leading to the absence of these therapeutic targets, followed by poor overall survival. We investigated if obesity could hasten TNBC progression and intermittent fasting (IF) could attenuate the progression of obesity-related TNBC. Our meta-analysis of the TNBC outcomes literature showed that obesity led to poorer overall survival in TNBC patients. Fasting-mimicking media reduced cell proliferation disrupted the cell cycle, and decreased cell migration and invasion. IF decreased body weight in obese mice but no change in normal mice. Obese mice exhibited elevated plasma glucose and cholesterol levels, increased tumor volume and weight, and enhanced macrophage accumulation in tumors. The obesity-exacerbated TNBC progression was attenuated after IF, which decreased cyclin B1 and vimentin levels and reduced the proinflammatory signature in the obesity-associated tumor microenvironment. IF attenuated obesity-induced TNBC progression through reduced obesity and tumor burdens in cell and animal experiments, supporting the potential of a cost-effective adjuvant IF therapy for TNBC through lifestyle change. Further evidence is needed of these IF benefits in TNBC, including from human clinical trials.
Collapse
Affiliation(s)
- Deok-Soo Son
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| | - Kaitlyn A. Done
- Biochemistry Program, College of Arts and Sciences, Spelman College, Atlanta, GA 30314, USA
| | - Jubin Son
- Neuroscience Program, College of Arts and Sciences, The University of Tennessee, Knoxville, TN 37996, USA
| | - Michael G. Izban
- Pathology, Anatomy and Cell Biology, Meharry Medical College, Nashville, TN 37208, USA
| | - Carlos Virgous
- Animal Core Facility, Meharry Medical College, Nashville, TN 37208, USA
| | - Eun-Sook Lee
- Department of Pharmaceutical Sciences, College of Pharmacy, Florida A&M University, Tallahassee, FL 32301, USA;
| | - Samuel E. Adunyah
- Department of Biochemistry, Cancer Biology, Neuroscience and Pharmacology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA;
| |
Collapse
|
3
|
Meng M, Shi LL. Serum tumor markers expression (CA199, CA242, and CEA) and its clinical implications in type 2 diabetes mellitus. World J Diabetes 2024; 15:232-239. [PMID: 38464372 PMCID: PMC10921164 DOI: 10.4239/wjd.v15.i2.232] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 11/17/2023] [Accepted: 12/18/2023] [Indexed: 02/04/2024] Open
Abstract
BACKGROUND Glucose and lipid metabolic disorder in patients with type 2 diabetes mellitus (T2DM) is associated with the levels of serum tumor markers of the digestive tract, such as cancer antigen (CA)199. Therefore, tumor markers in T2DM are important. AIM To evaluate the expression of serum tumor markers [CA199, CA242, and car-cinoembryonic antigen (CEA)] and the clinical implications of the expression in T2DM. METHODS For this observational study conducted at Hefei BOE Hospital, China, we enrolled 82 patients with first-onset T2DM and 51 controls between April 2019 and December 2020. Levels of fasting blood glucose (FBG), tumor markers (CA199, CEA, and CA242), glycosylated hemoglobin (HbA1c), etc. were measured and group index levels were compared. Moreover, FBG and HbA1c levels were correlated with tumor marker levels. Tumor markers were tested for diagnostic accuracy in patients with > 9% HbA1c using the receiver operating curve (ROC) curve. RESULTS The T2DM group had high serum FBG, HbA1c, CA199, and CEA levels (P < 0.05). A comparative analysis of the two groups based on HbA1c levels (Group A: HbA1c ≤ 9%; Group B: HbA1c > 9%) revealed significant differences in CEA and CA199 levels (P < 0.05). The areas under the ROC curve for CEA and CA199 were 0.853 and 0.809, respectively. CA199, CEA, and CA242 levels positively correlated with HbA1c (r = 0.308, 0.426, and 0.551, respectively) and FBG levels (r = 0.236, 0.231, and 0.298, respectively). CONCLUSION As compared to controls, serum CEA and CA199 levels were higher in patients with T2DM. HbA1c and FBG levels correlated with CA199, CEA, and CA242 levels. Patients with poorly controlled blood sugar must be screened for tumor markers.
Collapse
Affiliation(s)
- Mei Meng
- Department of Endocrinology, Hefei BOE Hospital, Hefei 230013, Anhui Province, China
| | - Li-Li Shi
- Department of Cadre Ward, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang Province, China
| |
Collapse
|
4
|
Chen L, Du S, Li YB, Su Q, Zhang J, Zhang H. Changes in serum tumor markers in type 2 diabetes mellitus with microalbuminuria. Front Endocrinol (Lausanne) 2023; 14:1247099. [PMID: 38130399 PMCID: PMC10733510 DOI: 10.3389/fendo.2023.1247099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 11/20/2023] [Indexed: 12/23/2023] Open
Abstract
Objectives The objective of this study was to investigate changes in serum tumor markers in type 2 diabetes mellitus (T2DM) with microalbuminuria and analyze the relationship between tumor markers and microalbuminuria. Methods A total of 956 T2DM patients aged 40-70 years hospitalized in the Department of Endocrinology, Xinhua Hospital, China, affiliated with Shanghai Jiaotong University School of Medicine, were enrolled from January 2018 to December 2020. The sample comprised 313 T2DM patients with microalbuminuria and 643 T2DM patients with normal urinary microalbumin levels. After assessing the changes in serum tumor markers in T2DM with microalbuminuria, we analyzed the risk of microalbuminuria by the serum tumor marker category using multiple logistic regression analysis. Results Serum CEA, CA199, CA125, CA153, CA211, SCC, CA242, and CA50 levels were significantly higher in T2DM patients with microalbuminuria than in those without microalbuminuria, while serum AFP levels were lower in the microalbuminuria group (P < 0.05). Following adjustment of confounders, serum CEA, CA211, and SCC were independently associated with microalbuminuria in T2DM. An ROC curve was used to estimate the cutoff point of tumor markers for microalbuminuria. Taking the values under the cutoff points as a reference, values for CEA, CA211, and SCC above the cutoff points indicated a significantly high risk of microalbuminuria. The OR of increased CEA for microalbuminuria was 2.006 (95%CI 1.456-2.765), the OR of increased CA211 for microalbuminuria was 1.505 (95%CI 1.092-2.074), and the OR of increased SCC for microalbuminuria was 1.958 (95%CI 1.407-2.724). Conclusion Several serum tumor markers were related to microalbuminuria in T2DM. Serum tumor markers such as CEA, SCC, and CA211 may indicate early diabetic nephropathy, particularly when elevated in combination.
Collapse
Affiliation(s)
- Lina Chen
- Department of Endocrinology, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Shichun Du
- Department of Endocrinology, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Bo Li
- Department of Endocrinology, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Qing Su
- Department of Endocrinology, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiangrong Zhang
- Health Management Center, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hongmei Zhang
- Department of Endocrinology, Xinhua Hospital Affiliated with Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
5
|
Berr AL, Wiese K, Dos Santos G, Koch CM, Anekalla KR, Kidd M, Davis JM, Cheng Y, Hu YS, Ridge KM. Vimentin is required for tumor progression and metastasis in a mouse model of non-small cell lung cancer. Oncogene 2023:10.1038/s41388-023-02703-9. [PMID: 37161053 DOI: 10.1038/s41388-023-02703-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 11/15/2022] [Accepted: 04/20/2023] [Indexed: 05/11/2023]
Abstract
Vimentin is highly expressed in metastatic cancers, and its expression correlates with poor patient prognoses. However, no causal in vivo studies linking vimentin and non-small cell lung cancer (NSCLC) progression existed until now. We use three complementary in vivo models to show that vimentin is required for the progression of NSCLC. First, we crossed LSL-KrasG12D; Tp53fl/fl mice (KPV+/+) with vimentin knockout mice (KPV-/-) to demonstrate that KPV-/- mice have attenuated tumor growth and improved survival compared with KPV+/+ mice. Next, we therapeutically treated KPV+/+ mice with withaferin A (WFA), an agent that disrupts vimentin intermediate filaments (IFs). We show that WFA suppresses tumor growth and reduces tumor burden in the lung. Finally, luciferase-expressing KPV+/+, KPV-/-, or KPVY117L cells were implanted into the flanks of athymic mice to track cancer metastasis to the lung. In KPVY117L cells, vimentin forms oligomers called unit-length filaments but cannot assemble into mature vimentin IFs. KPV-/- and KPVY117L cells fail to metastasize, suggesting that cell-autonomous metastasis requires mature vimentin IFs. Integrative metabolomic and transcriptomic analysis reveals that KPV-/- cells upregulate genes associated with ferroptosis, an iron-dependent form of regulated cell death. KPV-/- cells have reduced glutathione peroxidase 4 (GPX4) levels, resulting in the accumulation of toxic lipid peroxides and increased ferroptosis. Together, our results demonstrate that vimentin is required for rapid tumor growth, metastasis, and protection from ferroptosis in NSCLC.
Collapse
Affiliation(s)
- Alexandra L Berr
- Department of Biomedical Engineering, Northwestern University, Chicago, IL, USA
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Kristin Wiese
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Gimena Dos Santos
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Clarissa M Koch
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Kishore R Anekalla
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Martha Kidd
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Jennifer M Davis
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Yuan Cheng
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Yuan-Shih Hu
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Northwestern University, Chicago, IL, USA.
- Department of Cell and Molecular Biology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
6
|
Silva ATF, Rodrigues CM, Ferreira ICC, Santos LLD, Santos DW, Araújo TG, Canto PPL, Paiva CE, Goulart LR, Maia YCP. A Novel Detection Method of Breast Cancer through a Simple Panel of Biomarkers. Int J Mol Sci 2022; 23:ijms231911983. [PMID: 36233281 PMCID: PMC9570447 DOI: 10.3390/ijms231911983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/23/2022] Open
Abstract
Circulating tumor cells (CTCs) have been identified as responsible for the spread of tumors to other organs of the body. In this sense, the development of sensitive and specific assays for their detection is important to reduce the number of deaths due to metastases. Here, we assessed whether the detection of CTCs in peripheral blood can serve in the construction of a panel of diagnosis and monitoring treatments of breast cancer (BC), focusing on the expression of markers of epithelial-mesenchymal transition. Through analyzing the blood from women without breast alterations (control), women with benign alterations, women with breast cancer without chemotherapy, and women with breast cancer with chemotherapy, we identified the best markers by transcriptional levels and determined three profiles of CTCs (mesenchymal, intermediate, and epithelial) by flow cytometry which, combined, can be used for diagnosis and therapy monitoring with sensitivity and specificity between 80% and 100%. Therefore, we have developed a method for detecting breast cancer based on the analysis of CTC profiles by epithelial-mesenchymal transition markers which, combined, can be used for the diagnosis and monitoring of therapy.
Collapse
Affiliation(s)
- Alinne T. F. Silva
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Cláudia M. Rodrigues
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Izabella C. C. Ferreira
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Letícia L. D. Santos
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Donizeti W. Santos
- Obstetric Division, University Hospital, Federal University of Uberlandia, Av. Pará, 1720, Block 2H, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
| | - Thaise G. Araújo
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Paula P. L. Canto
- Department of Clinical Oncology, Clinics Hospital, Federal University of Uberlandia, Av. Pará, 1720, Oncology Sector, Room 9, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
| | - Carlos E. Paiva
- Department of Clinical Oncology, Graduate Program in Oncology, Palliative Care and Quality of Life Research Group (GPQual), Barretos Cancer Hospital, R. Antenor Duarte Vilela, 1331, Doutor Paulo Prata, Barretos 14784-400, Sao Paulo, Brazil
| | - Luiz R. Goulart
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
| | - Yara C. P. Maia
- Molecular Biology and Nutrition Research Group, School of Medicine, Graduate Program in Health Science, Av. Amazonas sn, Block 2E, 2º Floor, Room 210, Campus Umuarama, Uberlandia 38405-320, Minas Gerais, Brazil
- Laboratory of Nanobiotechnology Prof. Dr. Luiz Ricardo Goulart Filho, Institute of Biotechnology, Federal University of Uberlandia, Av. Amazonas sn, Block 2E, 2º Floor, Room 248, Campus Umuarama, Uberlandia 38405-302, Minas Gerais, Brazil
- Correspondence: ; Tel.: +34-3225-8628
| |
Collapse
|
7
|
Qi L, Knifley T, Chen M, O'Connor KL. Integrin α6β4 requires plectin and vimentin for adhesion complex distribution and invasive growth. J Cell Sci 2022; 135:273711. [PMID: 34897465 PMCID: PMC8917354 DOI: 10.1242/jcs.258471] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 12/01/2021] [Indexed: 01/30/2023] Open
Abstract
Integrin α6β4 binds plectin to associate with vimentin; however, the biological function remains unclear. Here, we utilized various integrin β4 mutants and CRISPR-Cas9 editing to investigate this association. Upon laminin binding, integrin α6β4 distinctly distributed peripherally as well as centrally, proximal to the nucleus. Upon fibronectin addition, integrin α6β4 was centrally recruited to large focal adhesions (FAs) and enhanced Fak (also known as PTK2) phosphorylation. Integrin β4 plectin-binding mutants or genetic deletion of plectin inhibited β4 recruitment to FAs and integrin α6β4-enhanced cell spreading, migration and three-dimensional invasive growth. Loss of the β4 signaling domain (but retaining plectin binding) blocked migration and invasiveness but not cell spreading, recruitment to FAs or colony growth. Immunostaining revealed that integrin α6β4 redistributed vimentin perinuclearly, where it colocalized with plectin and FAs. Depletion of vimentin completely blocked integrin β4-enhanced invasive growth, Fak phosphorylation and proliferation in three dimensions but not two dimensions. In summary, we demonstrate the essential roles of plectin and vimentin in promoting an invasive phenotype downstream of integrin α6β4. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Lei Qi
- Markey Cancer Center, University of Kentucky, Lexington 40506-0509, USA,Departments of Molecular and Cellular Biochemistry, University of Kentucky, Lexington 40506-0509, USA
| | - Teresa Knifley
- Markey Cancer Center, University of Kentucky, Lexington 40506-0509, USA,Departments of Molecular and Cellular Biochemistry, University of Kentucky, Lexington 40506-0509, USA
| | - Min Chen
- Markey Cancer Center, University of Kentucky, Lexington 40506-0509, USA,Toxicology and Cancer Biology, University of Kentucky, Lexington 40506-0509, USA
| | - Kathleen L. O'Connor
- Markey Cancer Center, University of Kentucky, Lexington 40506-0509, USA,Departments of Molecular and Cellular Biochemistry, University of Kentucky, Lexington 40506-0509, USA,Author for correspondence ()
| |
Collapse
|
8
|
TMEM176B Regulates AKT/mTOR Signaling and Tumor Growth in Triple-Negative Breast Cancer. Cells 2021; 10:cells10123430. [PMID: 34943938 PMCID: PMC8700203 DOI: 10.3390/cells10123430] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/30/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
TMEM176B is a member of the membrane spanning 4-domains (MS4) family of transmembrane proteins, and a putative ion channel that is expressed in immune cells and certain cancers. We aimed to understand the role of TMEM176B in cancer cell signaling, gene expression, cell proliferation, and migration in vitro, as well as tumor growth in vivo. We generated breast cancer cell lines with overexpressed and silenced TMEM176B, and a therapeutic antibody targeting TMEM176B. Proliferation and migration assays were performed in vitro, and tumor growth was evaluated in vivo. We performed gene expression and Western blot analyses to identify the most differentially regulated genes and signaling pathways in cells with TMEM176B overexpression and silencing. Silencing TMEM176B or inhibiting it with a therapeutic antibody impaired cell proliferation, while overexpression increased proliferation in vitro. Syngeneic and xenograft tumor studies revealed the attenuated growth of tumors with TMEM176B gene silencing compared with controls. We found that the AKT/mTOR signaling pathway was activated or repressed in cells overexpressing or silenced for TMEM176B, respectively. Overall, our results suggest that TMEM176B expression in breast cancer cells regulates key signaling pathways and genes that contribute to cancer cell growth and progression, and is a potential target for therapeutic antibodies.
Collapse
|
9
|
Vimentin Promotes the Aggressiveness of Triple Negative Breast Cancer Cells Surviving Chemotherapeutic Treatment. Cells 2021; 10:cells10061504. [PMID: 34203746 PMCID: PMC8232646 DOI: 10.3390/cells10061504] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/08/2021] [Accepted: 06/10/2021] [Indexed: 11/24/2022] Open
Abstract
Tremendous data have been accumulated in the effort to understand chemoresistance of triple negative breast cancer (TNBC). However, modifications in cancer cells surviving combined and sequential treatment still remain poorly described. In order to mimic clinical neoadjuvant treatment, we first treated MDA-MB-231 and SUM159-PT TNBC cell lines with epirubicin and cyclophosphamide for 2 days, and then with paclitaxel for another 2 days. After 4 days of recovery, persistent cells surviving the treatment were characterized at both cellular and molecular level. Persistent cells exhibited increased growth and were more invasive in vitro and in zebrafish model. Persistent cells were enriched for vimentinhigh sub-population, vimentin knockdown using siRNA approach decreased the invasive and sphere forming capacities as well as Akt phosphorylation in persistent cells, indicating that vimentin is involved in chemotherapeutic treatment-induced enhancement of TNBC aggressiveness. Interestingly, ectopic vimentin overexpression in native cells increased cell invasion and sphere formation as well as Akt phosphorylation. Furthermore, vimentin overexpression alone rendered the native cells resistant to the drugs, while vimentin knockdown rendered them more sensitive to the drugs. Together, our data suggest that vimentin could be considered as a new targetable player in the ever-elusive status of drug resistance and recurrence of TNBC.
Collapse
|
10
|
Meng X, Xu Y, Ning X. Tumor microenvironment acidity modulates ROR1 to promote epithelial-mesenchymal transition and hepatocarcinoma metastasis. J Cell Sci 2021; 134:237804. [PMID: 33648935 DOI: 10.1242/jcs.255349] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/18/2021] [Indexed: 11/20/2022] Open
Abstract
The tendency of hepatocarcinoma to metastasize results in a high rate of mortality, making it a hot research topic in cancer studies. Although an acidic tumor microenvironment has been proven to promote cancer metastasis, the underlying regulatory mechanisms remain poorly defined. Here, we found that acidic conditions significantly enhanced cell migration and invasion ability in hepatocellular carcinoma, and the expression of receptor tyrosine kinase-like orphan receptor 1 (ROR1) was distinctly upregulated in acid-treated cells. In addition, siRNA-mediated knockdown of ROR1 could effectively inhibit acid-induced cell migration, invasion and epithelial-mesenchymal transition (EMT). Importantly, neutralization of acidic environments with NaHCO3 could downregulate acid-stimulated ROR1 expression, thereby retarding cell metastatic potential. Notably, the formation of metastatic nodules was significantly increased after intrapulmonary injection of acid-stimulated cancer cells, and this was inhibited by pretreating with NaHCO3. In summary, we reveal that an acidic tumor microenvironment modulates ROR1 expression to promote tumor metastasis, providing not only a better understanding of molecular mechanisms related to metastasis, but also a promising target for tumor management.
Collapse
Affiliation(s)
- Xia Meng
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials , Nanjing University, Nanjing 210093, China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials , Nanjing University, Nanjing 210093, China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials , Nanjing University, Nanjing 210093, China
| |
Collapse
|
11
|
Tong W, Gao H, Wei X, Mao D, Zhang L, Chen Q, Zhang Z, Li Y. Correlation of serum CA199 levels with glycemic control and microvascular complications in patients with type 2 diabetes mellitus. Am J Transl Res 2021; 13:3302-3308. [PMID: 34017502 PMCID: PMC8129216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/07/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE This study aimed to explore the correlation between glycemic control, microvascular complications and serum glycogen antigen (CA199) in patients with type 2 diabetes mellitus (T2DM). METHODS 519 patients with T2DM admitted to our hospital were included. All patients had CA199 levels measured. Patients were divided into low glycation (LH) group (HbA1C <7.5%), Hyperglycemia (HH) group (HbA1C ≥7.5%), fasting glucose compliance (SF) group (FBG <7.0 mmol/L), high fasting glucose (HF) group (FBG ≥7.0 mmol/L), postprandial glucose compliance (SP) group (PBG <10.0 mmol/L) and high postprandial glucose (HP) group (PBG ≥10.0 mmol/L) and with microvascular complications group, and no microvascular complications group. Division was according to levels of glycated hemoglobin (HbA1C), fasting blood glucose (FBG), 2-hour postprandial blood glucose (2hPBG), and diabetic microvascular complications. RESULTS CA199 levels were significantly higher in the HH and HF groups than in the LH and SF groups (P<0.05); HbA1C and FBG were positively correlated with CA199; CA199 levels were not significantly different between SP and HP groups (P>0.05), and PBG was not significantly correlated with CA199 levels. CA199 levels were significantly higher in the group with microvascular complications than in the group without microvascular complications (P<0.05); HbA1C was an independent risk factor for elevated CA199. CONCLUSION Patients with T2DM and higher CA199 levels need to be evaluated for glycemic status and the presence of microvascular complications. HbA1C is a major risk factor for elevated CA199 levels.
Collapse
Affiliation(s)
- Weishuang Tong
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Huashan Gao
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Xiaoya Wei
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Dongxue Mao
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Lei Zhang
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Qiu Chen
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Zhilu Zhang
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| | - Yanjiao Li
- College of Medicine, Henan Province Key Laboratory of Germplasm Innovation and Utilization of Eco-economic Woody Plant, Pingdingshan University Pingdingshan 467000, Henan, China
| |
Collapse
|
12
|
Shah H, Hacker A, Langburt D, Dewar M, McFadden MJ, Zhang H, Kuzmanov U, Zhou YQ, Hussain B, Ehsan F, Hinz B, Gramolini AO, Heximer SP. Myocardial Infarction Induces Cardiac Fibroblast Transformation within Injured and Noninjured Regions of the Mouse Heart. J Proteome Res 2021; 20:2867-2881. [PMID: 33789425 DOI: 10.1021/acs.jproteome.1c00098] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Heart failure (HF) is associated with pathological remodeling of the myocardium, including the initiation of fibrosis and scar formation by activated cardiac fibroblasts (CFs). Although early CF-dependent scar formation helps prevent cardiac rupture by maintaining the heart's structural integrity, ongoing deposition of the extracellular matrix in the remote and infarct regions can reduce tissue compliance, impair cardiac function, and accelerate progression to HF. In our study, we conducted mass spectrometry (MS) analysis to identify differentially altered proteins and signaling pathways between CFs isolated from 7 day sham and infarcted murine hearts. Surprisingly, CFs from both the remote and infarct regions of injured hearts had a wide number of similarly altered proteins and signaling pathways that were consistent with fibrosis and activation into pathological myofibroblasts. Specifically, proteins enriched in CFs isolated from MI hearts were involved in pathways pertaining to cell-cell and cell-matrix adhesion, chaperone-mediated protein folding, and collagen fibril organization. These results, together with principal component analyses, provided evidence of global CF activation postinjury. Interestingly, however, direct comparisons between CFs from the remote and infarct regions of injured hearts identified 15 differentially expressed proteins between MI remote and MI infarct CFs. Eleven of these proteins (Gpc1, Cthrc1, Vmac, Nexn, Znf185, Sprr1a, Specc1, Emb, Limd2, Pawr, and Mcam) were higher in MI infarct CFs, whereas four proteins (Gstt1, Gstm1, Tceal3, and Inmt) were higher in MI remote CFs. Collectively, our study shows that MI injury induced global changes to the CF proteome, with the magnitude of change reflecting their relative proximity to the site of injury.
Collapse
Affiliation(s)
- Haisam Shah
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Alison Hacker
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Dylan Langburt
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Michael Dewar
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Meghan J McFadden
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Hangjun Zhang
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Uros Kuzmanov
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Yu-Qing Zhou
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Bilal Hussain
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1
| | - Fahad Ehsan
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Ontario, Canada M5G 1G6
| | - Anthony O Gramolini
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Scott P Heximer
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, 661 University Avenue, Toronto, Ontario, Canada M5G 1M1.,Department of Physiology, Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
13
|
Scully T, Ettela A, LeRoith D, Gallagher EJ. Obesity, Type 2 Diabetes, and Cancer Risk. Front Oncol 2021; 10:615375. [PMID: 33604295 PMCID: PMC7884814 DOI: 10.3389/fonc.2020.615375] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 12/09/2020] [Indexed: 12/12/2022] Open
Abstract
Obesity and type 2 diabetes have both been associated with increased cancer risk and are becoming increasingly prevalent. Metabolic abnormalities such as insulin resistance and dyslipidemia are associated with both obesity and type 2 diabetes and have been implicated in the obesity-cancer relationship. Multiple mechanisms have been proposed to link obesity and diabetes with cancer progression, including an increase in insulin/IGF-1 signaling, lipid and glucose uptake and metabolism, alterations in the profile of cytokines, chemokines, and adipokines, as well as changes in the adipose tissue directly adjacent to the cancer sites. This review aims to summarize and provide an update on the epidemiological and mechanistic evidence linking obesity and type 2 diabetes with cancer, focusing on the roles of insulin, lipids, and adipose tissue.
Collapse
Affiliation(s)
- Tiffany Scully
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Abora Ettela
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Derek LeRoith
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Emily Jane Gallagher
- Division of Endocrinology, Diabetes and Bone Disease, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
- Tisch Cancer Institute at Mount Sinai, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| |
Collapse
|
14
|
Abstract
Simple Summary Cell migration is an essential process from embryogenesis to cell death. This is tightly regulated by numerous proteins that help in proper functioning of the cell. In diseases like cancer, this process is deregulated and helps in the dissemination of tumor cells from the primary site to secondary sites initiating the process of metastasis. For metastasis to be efficient, cytoskeletal components like actin, myosin, and intermediate filaments and their associated proteins should co-ordinate in an orderly fashion leading to the formation of many cellular protrusions-like lamellipodia and filopodia and invadopodia. Knowledge of this process is the key to control metastasis of cancer cells that leads to death in 90% of the patients. The focus of this review is giving an overall understanding of these process, concentrating on the changes in protein association and regulation and how the tumor cells use it to their advantage. Since the expression of cytoskeletal proteins can be directly related to the degree of malignancy, knowledge about these proteins will provide powerful tools to improve both cancer prognosis and treatment. Abstract Successful metastasis depends on cell invasion, migration, host immune escape, extravasation, and angiogenesis. The process of cell invasion and migration relies on the dynamic changes taking place in the cytoskeletal components; actin, tubulin and intermediate filaments. This is possible due to the plasticity of the cytoskeleton and coordinated action of all the three, is crucial for the process of metastasis from the primary site. Changes in cellular architecture by internal clues will affect the cell functions leading to the formation of different protrusions like lamellipodia, filopodia, and invadopodia that help in cell migration eventually leading to metastasis, which is life threatening than the formation of neoplasms. Understanding the signaling mechanisms involved, will give a better insight of the changes during metastasis, which will eventually help targeting proteins for treatment resulting in reduced mortality and longer survival.
Collapse
|
15
|
Dai J, Wang Y, Gong J, Yao Y. Biointerface anisotropy modulates migration of breast cancer cell. Colloids Surf B Biointerfaces 2020; 190:110973. [PMID: 32199258 DOI: 10.1016/j.colsurfb.2020.110973] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 02/24/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
Migration of cancer cell is a cyclic process, which involves dynamic interaction between extracellular biointerface and cellular responds. In tumors, collagen as extracellular matrix reorganizes biointerface from curl and isotropic fibers to straightened and anisotropic fibers during tumorigenesis, yet how cell migration respond to topography of biointerface is unknown. In this research, we introduced a facile fabrication method on nanofibers of varying topography, which was mimicking the alignment of extracellular nanofibers, to examine the change of cytoskeleton during cell migration. We took advantage of breast carcinoma cell line (MDA-MB-231) for time-lapse imaging analysis. We found that biointerface anisotropy modulated morphology of cell and mediated the pattern of migration. Morphologically, cells on anisotropic nanofiber showed extending spindle shape. The trajectories of migration templated the topographic pattern on biointerface. Besides, aligned nanofiber induced caterpillar-like model of migration through protrusion - retraction cycle, which was indicated by periodical variation of aspect ratio and velocity of cells. The biointerface anisotropy triggered vimentin filaments and microtubule networks preferentially oriented along the alignment of nanofibers. And the velocity of cell mobility by vimentin, β-catenin or CDC42 knockdown was significantly enhanced on aligned nanofibers. Thus, we implied that biointerface anisotropy modulated migration of breast cancer cell and it associated with reorganization of cytoskeleton filaments.
Collapse
Affiliation(s)
- Jing Dai
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Ceramics, Chinese Academy of Sciences, 1295 Dingxi Road, Changning, Shanghai, 200050, China; University of Chinese Academy of Sciences, 19 Yuquan Road, Shijingshan, Beijing, 100049, China
| | - Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China; Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, 345 Lingling Road, Shanghai, 200032, China
| | - Jinkang Gong
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai, 201210, China.
| |
Collapse
|
16
|
Wang Y, Yao Y. Nanofiber Alignment Mediates the Pattern of Single Cell Migration. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2129-2135. [PMID: 32040329 DOI: 10.1021/acs.langmuir.9b03314] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In natural tissue, nanofibrils in extracellular matrix (ECM), such as collagen fibrils, direct cell migration through contacting guidance. The contacting nanofibers on cell-ECM interface are reorganized from curl fibers to straightened fibers. However, how these nanofibers regulate single cell migration remains obscure. To investigate this issue, we fabricated collagen/polymer based biomimetic nanofiber sheets of varying topography. And we selected tumorigenic cell KGN and nontumorigenic cell 293T for comparison. We found KGN showed higher sensitivity to the nanofiber alignment rather than the nontumorigenic cell 293T, in morphological change, trajectory adaptation, and velocity variation. We also found aligned nanofibers shaped both KGN and 293T into elongated spindle morphology. Comparatively, KGN had greater perimeter and lower roundness than 293T. To study the dynamics of single cell migration of KGN and 293T, we conducted trajectory tracking and siRNA validation on regulatory proteins. We found nanofibers of varying topography regulated the patterns of single cell migration differently. For KGN cell, β-catenin, Rac1, and Cdc42 participated in its directional migration, but it was impervious to vimentin. Comparatively, epithelial cell 293T involved vimentin in its directional migration.
Collapse
Affiliation(s)
- Yiqun Wang
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| | - Yuan Yao
- School of Physical Science and Technology, ShanghaiTech University, 393 Middle Huaxia Road, Pudong, Shanghai 201210, China
| |
Collapse
|
17
|
Upregulation of Epithelial-To-Mesenchymal Transition Markers and P2X7 Receptors Is Associated to Increased Invasiveness Caused by P2X7 Receptor Stimulation in Human Glioblastoma Stem Cells. Cells 2019; 9:cells9010085. [PMID: 31905754 PMCID: PMC7016938 DOI: 10.3390/cells9010085] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 12/23/2019] [Accepted: 12/25/2019] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma (GBM) stem cells (GSCs), which contribute to GBM unfavorable prognosis, show high expression levels of ATP/P2X7 receptors (P2X7R). Here, we reported that cells exposure to 2’(3’)-O-(4-benzoylbenzoyl)-ATP (BzATP), a P2X7R agonist, up-regulated the expression of markers associated to epithelial-to-mesenchymal transition (EMT), a process likely contributing to GSC malignancy, and increased GSC migration/invasiveness like the known EMT inducer, Transforming Growth Factor β1 (TGFβ1). These effects were coupled to phosphorylation of SMAD2, a downstream effector in the TGFβ pathway, suggesting its involvement in P2X7R-mediated activity in GSCs. All BzATP effects, including a decrease in the caspase 3/7 activity in GSC medium, were mostly counteracted by the P2X7R antagonist A438079. Finally, BzATP increased the subunit expression of two main human P2X7R splice variants, the full-length P2X7A and the truncated P2X7B, lacking the carboxylic tail, which have different functional properties depending on their arrangement. Since up-regulation of A/B subunits might favor their assembly into a heterotrimeric P2X7R with great sensitivity towards agonists and cell energy support, this is in line with increased EMT markers expression, cell migration/invasion and GSC survival observed following P2X7R stimulation. As in GBM microenvironment extracellular ATP levels may activate P2X7R, our data suggest a P2X7R role in GBM recurrence/invasiveness.
Collapse
|
18
|
Luo C, Wang Y, Wei C, Chen Y, Ji Z. The anti-migration and anti-invasion effects of Bruceine D in human triple-negative breast cancer MDA-MB-231 cells. Exp Ther Med 2019; 19:273-279. [PMID: 31853299 PMCID: PMC6909795 DOI: 10.3892/etm.2019.8187] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 10/15/2019] [Indexed: 12/11/2022] Open
Abstract
Brucein D (BD) is a naturally occurring major active quassinoid extracted from the Chinese medicinal herb Brucea javanica, which has been previously demonstrated to exhibit anticancer activities. The present study aimed to investigate the anticancer effects of BD on MDA-MB-231 cells, a human triple-negative breast cancer (TNBC) cell line. An MTT assay was performed to assess cell viability, whilst wound healing and Transwell assay were applied to measure cell migration and invasion, respectively. Western blot analysis was performed to assess the expression of E-cadherin, vimentin and β-catenin, which are proteins associated with epithelial-mesenchymal transformation (EMT), and PI3K, AKT and p-AKT, which are key components of the PI3K/AKT signaling pathway. BD was indicated to reduce cell viability in a dose- and time-dependent manner, whilst cell invasion and migration were also significantly inhibited in a dose-dependent manner. Western blot analysis demonstrated that BD treatment significantly upregulated the expression of E-cadherin and downregulated the expression of vimentin and β-catenin. Additionally, BD downregulated the expression of PI3K and reduced AKT phosphorylation. In conclusion, BD can inhibit MDA-MB-231 cell viability, migration and invasion, suggesting the potential use of BD for the treatment of TNBC.
Collapse
Affiliation(s)
- Can Luo
- Department of Oncology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Yu Wang
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Cheng Wei
- Department of Oncology, Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| | - Yuxin Chen
- Department of Oncology, Gaoyou People's Hospital, Gaoyou Hospital Affiliated to Soochow University, Gaoyou, Jiangsu 225600, P.R. China
| | - Zhaoning Ji
- Department of Oncology, Yijishan Hospital of Wannan Medical College, Wuhu, Anhui 241001, P.R. China
| |
Collapse
|
19
|
Todenhöfer T, Pantel K, Stenzl A, Werner S. Pathophysiology of Tumor Cell Release into the Circulation and Characterization of CTC. Recent Results Cancer Res 2019; 215:3-24. [PMID: 31605221 DOI: 10.1007/978-3-030-26439-0_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The traditional model of metastatic progression postulates that the ability to form distant metastases is driven by random mutations in cells of the primary tumor.
Collapse
Affiliation(s)
- Tilman Todenhöfer
- Department of Urology, Eberhard-Karls-University, Tuebingen, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Arnulf Stenzl
- Department of Urology, Eberhard-Karls-University, Tuebingen, Germany
| | - Stefan Werner
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
20
|
Intermediate Filaments as Effectors of Cancer Development and Metastasis: A Focus on Keratins, Vimentin, and Nestin. Cells 2019; 8:cells8050497. [PMID: 31126068 PMCID: PMC6562751 DOI: 10.3390/cells8050497] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 05/16/2019] [Accepted: 05/22/2019] [Indexed: 02/08/2023] Open
Abstract
Intermediate filament (IF) proteins make up the largest family of cytoskeletal proteins in metazoans, and are traditionally known for their roles in fostering structural integrity in cells and tissues. Remarkably, individual IF genes are tightly regulated in a fashion that reflects the type of tissue, its developmental and differentiation stages, and biological context. In cancer, IF proteins serve as diagnostic markers, as tumor cells partially retain their original signature expression of IF proteins. However, there are also characteristic alterations in IF gene expression and protein regulation. The use of high throughput analytics suggests that tumor-associated alterations in IF gene expression have prognostic value. Parallel research is also showing that IF proteins directly and significantly impact several key cellular properties, including proliferation, death, migration, and invasiveness, with a demonstrated impact on the development, progression, and characteristics of various tumors. In this review, we draw from recent studies focused on three IF proteins most associated with cancer (keratins, vimentin, and nestin) to highlight how several “hallmarks of cancer” described by Hanahan and Weinberg are impacted by IF proteins. The evidence already in hand establishes that IF proteins function beyond their classical roles as markers and serve as effectors of tumorigenesis.
Collapse
|
21
|
Abstract
The vimentin gene (
VIM) encodes one of the 71 human intermediate filament (IF) proteins, which are the building blocks of highly ordered, dynamic, and cell type-specific fiber networks. Vimentin is a multi-functional 466 amino acid protein with a high degree of evolutionary conservation among vertebrates.
Vim
−/− mice, though viable, exhibit systemic defects related to development and wound repair, which may have implications for understanding human disease pathogenesis. Vimentin IFs are required for the plasticity of mesenchymal cells under normal physiological conditions and for the migration of cancer cells that have undergone epithelial–mesenchymal transition. Although it was observed years ago that vimentin promotes cell migration, the molecular mechanisms were not completely understood. Recent advances in microscopic techniques, combined with computational image analysis, have helped illuminate vimentin dynamics and function in migrating cells on a precise scale. This review includes a brief historical account of early studies that unveiled vimentin as a unique component of the cell cytoskeleton followed by an overview of the physiological vimentin functions documented in studies on
Vim
−/− mice. The primary focus of the discussion is on novel mechanisms related to how vimentin coordinates cell migration. The current hypothesis is that vimentin promotes cell migration by integrating mechanical input from the environment and modulating the dynamics of microtubules and the actomyosin network. These new findings undoubtedly will open up multiple avenues to study the broader function of vimentin and other IF proteins in cell biology and will lead to critical insights into the relevance of different vimentin levels for the invasive behaviors of metastatic cancer cells.
Collapse
Affiliation(s)
- Rachel A Battaglia
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Samed Delic
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| | - Harald Herrmann
- Division of Molecular Genetics, German Cancer Research Center, Heidelberg, Germany.,Institute of Neuropathology, University Hospital Erlangen, Erlangen, Germany
| | - Natasha T Snider
- Department of Cell Biology and Physiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Ghosh A, Sarkar S, Banerjee S, Behbod F, Tawfik O, McGregor D, Graff S, Banerjee SK. MIND model for triple-negative breast cancer in syngeneic mice for quick and sequential progression analysis of lung metastasis. PLoS One 2018; 13:e0198143. [PMID: 29813119 PMCID: PMC5973560 DOI: 10.1371/journal.pone.0198143] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/14/2018] [Indexed: 12/25/2022] Open
Abstract
Mouse models of breast cancer with specific molecular subtypes (e.g., ER or HER2 positive) in an immunocompetent or an immunocompromised environment significantly contribute to our understanding of cancer biology, despite some limitations, and they give insight into targeted therapies. However, an ideal triple-negative breast cancer (TNBC) mouse model is lacking. What has been missing in the TNBC mouse model is a sequential progression of the disease in an essential native microenvironment. This notion inspired us to develop a TNBC-model in syngeneic mice using a mammary intraductal (MIND) method. To achieve this goal, Mvt-1and 4T1 TNBC mouse cell lines were injected into the mammary ducts via nipples of FVB/N mice and BALB/c wild-type immunocompetent mice, respectively. We established that the TNBC-MIND model in syngeneic mice could epitomize all breast cancer progression stages and metastasis into the lungs via lymphatic or hematogenous dissemination within four weeks. Collectively, the syngeneic mouse-TNBC-MIND model may serve as a unique platform for further investigation of the underlying mechanisms of TNBC growth and therapies.
Collapse
Affiliation(s)
- Arnab Ghosh
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Sandipto Sarkar
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Snigdha Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Fariba Behbod
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
| | - Ossama Tawfik
- Saint Luke’s Hospital of Kansas City, Kansas City, Missouri, United States of America
| | - Douglas McGregor
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Pathology Department, VA Medical Center, Kansas City, Missouri, United States of America
| | - Stephanie Graff
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Sarah Cannon Cancer Center at HCA Midwest Health, Overland Park, Kansas, United States of America
| | - Sushanta K. Banerjee
- Cancer Research Unit, VA Medical Center, Kansas City, Missouri, United States of America
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States of America
- * E-mail: ,
| |
Collapse
|
23
|
Takai K, Drain AP, Lawson DA, Littlepage LE, Karpuj M, Kessenbrock K, Le A, Inoue K, Weaver VM, Werb Z. Discoidin domain receptor 1 (DDR1) ablation promotes tissue fibrosis and hypoxia to induce aggressive basal-like breast cancers. Genes Dev 2018; 32:244-257. [PMID: 29483153 PMCID: PMC5859966 DOI: 10.1101/gad.301366.117] [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: 05/02/2017] [Accepted: 01/24/2018] [Indexed: 12/17/2022]
Abstract
Here, Takai et al. researched the function of discoidin domain receptor 1 (DDR1), a member of the subfamily of receptor tyrosine kinases activated by collagens that is overexpressed in breast and other carcinoma cells. Using bioinformatics analysis, breast cancer cell lines, and knockout mice, they demonstrate that DDR1 ablation leads to aggressive breast cancer, and their findings suggest that the absence of DDR1 provides a growth and adhesion advantage that favors the expansion of basal cells, potentiates fibrosis, and enhances necrosis/hypoxia and basal differentiation of transformed cells to increase their aggression and metastatic potential. The discoidin domain receptor 1 (DDR1) is overexpressed in breast carcinoma cells. Low DDR1 expression is associated with worse relapse-free survival, reflecting its controversial role in cancer progression. We detected DDR1 on luminal cells but not on myoepithelial cells of DDR1+/+ mice. We found that DDR1 loss compromises cell adhesion, consistent with data that older DDR1−/− mammary glands had more basal/myoepithelial cells. Basal cells isolated from older mice exerted higher traction forces than the luminal cells, in agreement with increased mammary branches observed in older DDR1−/− mice and higher branching by their isolated organoids. When we crossed DDR1−/− mice with MMTV-PyMT mice, the PyMT/DDR1−/− mammary tumors grew faster and had increased epithelial tension and matricellular fibrosis with a more basal phenotype and increased lung metastases. DDR1 deletion induced basal differentiation of CD90+CD24+ cancer cells, and the increase in basal cells correlated with tumor cell mitoses. K14+ basal cells, including K8+K14+ cells, were increased adjacent to necrotic fields. These data suggest that the absence of DDR1 provides a growth and adhesion advantage that favors the expansion of basal cells, potentiates fibrosis, and enhances necrosis/hypoxia and basal differentiation of transformed cells to increase their aggression and metastatic potential.
Collapse
Affiliation(s)
- Ken Takai
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA.,Division of Breast Oncology, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Allison P Drain
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA
| | - Devon A Lawson
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Laurie E Littlepage
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Marcela Karpuj
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Kai Kessenbrock
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Annie Le
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| | - Kenichi Inoue
- Division of Breast Oncology, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Valerie M Weaver
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California at San Francisco, San Francisco, California 94143, USA.,Department of Bioengineering and Therapeutic Sciences, University of California at San Francisco, San Francisco, California 94143, USA.,Department of Radiation Oncology, University of California at San Francisco, San Francisco, California 94143, USA
| | - Zena Werb
- Department of Anatomy, University of California at San Francisco, San Francisco, California 94143, USA
| |
Collapse
|
24
|
Wang T, Zheng L, Wang Q, Hu YW. Emerging roles and mechanisms of FOXC2 in cancer. Clin Chim Acta 2018; 479:84-93. [PMID: 29341903 DOI: 10.1016/j.cca.2018.01.019] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 01/12/2018] [Accepted: 01/12/2018] [Indexed: 12/20/2022]
Abstract
Forkhead box protein C2 (FOXC2), a transcription factor of the forkhead/winged-helix family, is required for embryonic and prenatal development. FOXC2 acts as a crucial modulator during both angiogenesis and lymphangiogenesis via multiple angiogenic and lymphangiogenic pathways, respectively. Although recent studies have shed light on the emerging role of FOXC2 in cancer, very little is known about the precise underlying mechanisms. The purpose of this review is to summarize the current understanding of FOXC2 and provide potential mechanistic explanations of the relationship between FOXC2 and cancer, as well as discuss the prospect for future research in the promising prognostic value of FOXC2 in cancer.
Collapse
Affiliation(s)
- Teng Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lei Zheng
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Qian Wang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yan-Wei Hu
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
25
|
High content image analysis reveals function of miR-124 upstream of Vimentin in regulating motor neuron mitochondria. Sci Rep 2018; 8:59. [PMID: 29311649 PMCID: PMC5758812 DOI: 10.1038/s41598-017-17878-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 12/02/2017] [Indexed: 01/24/2023] Open
Abstract
microRNAs (miRNAs) are critical for neuronal function and their dysregulation is repeatedly observed in neurodegenerative diseases. Here, we implemented high content image analysis for investigating the impact of several miRNAs in mouse primary motor neurons. This survey directed our attention to the neuron-specific miR-124, which controls axonal morphology. By performing next generation sequencing analysis and molecular studies, we characterized novel roles for miR-124 in control of mitochondria localization and function. We further demonstrated that the intermediate filament Vimentin is a key target of miR-124 in this system. Our data establishes a new pathway for control of mitochondria function in motor neurons, revealing the value of a neuron-specific miRNA gene as a mechanism for the re-shaping of otherwise ubiquitously-expressed intermediate filament network, upstream of mitochondria activity and cellular metabolism.
Collapse
|
26
|
Privat M, Rudewicz J, Sonnier N, Tamisier C, Ponelle-Chachuat F, Bignon YJ. Antioxydation And Cell Migration Genes Are Identified as Potential Therapeutic Targets in Basal-Like and BRCA1 Mutated Breast Cancer Cell Lines. Int J Med Sci 2018; 15:46-58. [PMID: 29333087 PMCID: PMC5765739 DOI: 10.7150/ijms.20508] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 10/11/2017] [Indexed: 12/26/2022] Open
Abstract
Basal-like breast cancers are among the most aggressive cancers and effective targeted therapies are still missing. In order to identify new therapeutic targets, we performed Methyl-Seq and RNA-Seq of 10 breast cancer cell lines with different phenotypes. We confirmed that breast cancer subtypes cluster the RNA-Seq data but not the Methyl-Seq data. Basal-like tumor hypermethylated phenotype was not confirmed in our study but RNA-Seq analysis allowed to identify 77 genes significantly overexpressed in basal-like breast cancer cell lines. Among them, 48 were overexpressed in triple negative breast cancers of TCGA data. Some molecular functions were overrepresented in this candidate gene list. Genes involved in antioxydation, such as SOD1, MGST3 and PRDX or cadherin-binding genes, such as PFN1, ITGB1 and ANXA1, could thus be considered as basal like breast cancer biomarkers. We then sought if these genes were linked to BRCA1, since this gene is often inactivated in basal-like breast cancers. Nine genes were identified overexpressed in both basal-like breast cancer cells and BRCA1 mutated cells. Amongst them, at least 3 genes code for proteins implicated in epithelial cell migration and epithelial to mesenchymal transition (VIM, ITGB1 and RhoA). Our study provided several potential therapeutic targets for triple negative and BRCA1 mutated breast cancers. It seems that migration and mesenchymal properties acquisition of basal-like breast cancer cells is a key functional pathway in these tumors with a high metastatic potential.
Collapse
Affiliation(s)
- Maud Privat
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Justine Rudewicz
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Nicolas Sonnier
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
- Biological Resources Center BB-0033-00075, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Christelle Tamisier
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Flora Ponelle-Chachuat
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| | - Yves-Jean Bignon
- Université Clermont Auvergne, Centre Jean Perrin, INSERM, U1240 Imagerie Moléculaire et Stratégies Théranostiques, F-63000 Clermont Ferrand, France
- Département d'Oncogénétique, Centre Jean Perrin, F-63000 Clermont Ferrand, France
- Biological Resources Center BB-0033-00075, Centre Jean Perrin, F-63000 Clermont Ferrand, France
| |
Collapse
|
27
|
Peuhu E, Virtakoivu R, Mai A, Wärri A, Ivaska J. Epithelial vimentin plays a functional role in mammary gland development. Development 2017; 144:4103-4113. [PMID: 28947532 DOI: 10.1242/dev.154229] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 09/19/2017] [Indexed: 01/01/2023]
Abstract
In the mammary gland, vimentin intermediate filaments are expressed in stromal cells and in basal epithelial cell populations, including gland-reconstituting mammary stem cells, with largely undefined functions. Here, we have studied how vimentin deficiency affects mouse mammary gland development. We find that, in adult vimentin knockout mice (Vim-/- ), mammary ductal outgrowth is delayed. The adult Vim-/- glands display dilated ducts and a reduced basal-to-luminal mouse mammary epithelial cell (MMEC) ratio indicative of altered progenitor cell activity. Accordingly, isolated Vim-/- MMECs form fewer mammospheres and basal-like organoids in vitro than their wild-type counterparts. Importantly, reduced basal MMEC number translates into defects in Vim-/- mammary gland regeneration in vivo Global gene expression profiling of basal MMECs reveals that lack of vimentin alters multiple pathways, including adhesion, cancer and Wnt signalling. Furthermore, vimentin contributes to stem-like cell properties in MDA-MB-231 breast cancer cells, wherein vimentin depletion reduces tumoursphere formation and attenuates expression of breast cancer stem cell-associated surface markers. Together, our findings identify vimentin as a positive regulator of stemness in the developing mouse mammary gland and in breast cancer cells.
Collapse
Affiliation(s)
- Emilia Peuhu
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | | | - Anja Mai
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | - Anni Wärri
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland
| | - Johanna Ivaska
- Centre for Biotechnology, University of Turku, 20520 Turku, Finland .,Department of Biochemistry and Food Chemistry, University of Turku, 20520 Turku, Finland
| |
Collapse
|
28
|
Elevated tumor LDLR expression accelerates LDL cholesterol-mediated breast cancer growth in mouse models of hyperlipidemia. Oncogene 2017; 36:6462-6471. [PMID: 28759039 PMCID: PMC5690879 DOI: 10.1038/onc.2017.247] [Citation(s) in RCA: 146] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 06/14/2017] [Accepted: 06/16/2017] [Indexed: 12/11/2022]
Abstract
Obesity is associated with an increase in cancer-specific mortality in women with breast cancer. Elevated cholesterol, particularly low-density lipoprotein cholesterol (LDL-C) is frequently seen in obese women. Here, we aimed to determine the importance of elevated circulating LDL, and LDL receptor (LDLR) expression in tumor cells, on the growth of breast cancer using mouse models of hyperlipidemia. We describe two novel immunodeficient mouse models of hyperlipidemia (Rag1−/−/LDLR−/− and Rag1−/−/ApoE (apolipoprotein E)−/− mice), in addition to established immunocompetent LDLR−/− and ApoE−/− mice. The mice were used to study the effects of elevated LDL-C in human triple negative (MDA-MB-231) and mouse Her2/Neu overexpressing (MCNeuA) breast cancers. Tumors derived from MCNeuA and MDA-MB-231 cells had high LDLR expression and formed larger tumors in mice with high circulating LDL-C concentrations than in mice with lower LDL-C. Silencing the LDLR in the tumor cells led to decreased growth of Her2Neu overexpressing tumors in LDLR−/− and ApoE−/− mice, with increased Caspase 3 cleavage. Additionally, in vitro, silencing the LDLR led to decreased cell survival in serum-starved conditions, associated with Caspase 3 cleavage. Examining publically available human datasets, we found that high LDLR expression in human breast cancers was associated with decreased recurrence-free survival, particularly in patients treated with systemic therapies. Overall, our results highlight the importance of the LDLR in the growth of triple negative and HER2 overexpressing breast cancers in the setting of elevated circulating LDL-C, which may be important contributing factors to the increased recurrence and mortality in obese women with breast cancer.
Collapse
|
29
|
Wang W, Yi M, Zhang R, Li J, Chen S, Cai J, Zeng Z, Li X, Xiong W, Wang L, Li G, Xiang B. Vimentin is a crucial target for anti-metastasis therapy of nasopharyngeal carcinoma. Mol Cell Biochem 2017; 438:47-57. [PMID: 28744809 DOI: 10.1007/s11010-017-3112-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/01/2017] [Indexed: 12/23/2022]
Abstract
Nasopharyngeal carcinoma (NPC) is a unique subtype of head and neck cancer, with tendency to spread to regional lymph nodes and distant organs at early stage. Vimentin, a major cytoskeletal protein constituent of the intermediate filament, plays a critical role in the epithelial-mesenchymal transition. Overexpression of vimentin is considered to be a critical prerequisite for metastasis in numerous human cancers. Therefore, targeting vimentin for cancer therapy has gained a lot of interest. In the present study, we detected vimentin expression in NPC tissues and found that overexpression of vimentin is associated with poor prognosis of NPC patients. Silencing of vimentin in NPC CNE2 cells by RNAi suppresses cells migration and invasion in vitro. However, blocking vimentin did not affect cell proliferation of CNE2 cells. In addition, the in vivo metastatic potential of CNE2 cells transfected with Vimentin shRNA was suppressed in a nude mouse model of pulmonary metastasis. Silencing of Vimentin in CNE2 cells leads to a decrease of microvessel density and VEGF, CD31, MMP2, and MMP9 expressions in pulmonary metastatic tumors. Importantly, we found that it is easier for the tumor cells from the high vimentin-expressing pulmonary metastatic tumors to reinvade the microvessel and to form stable tumor plaques attached to the endothelial cells, which resemble the resource of circulating tumor cells and are very hard to eliminate. However, depletion of vimentin inhibits the formation of vascular tumor plaques. Our findings suggest that RNAi-based vimentin silencing may be a potential and promising anti-metastatic therapeutic strategy for NPC.
Collapse
Affiliation(s)
- Wei Wang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Mei Yi
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Renya Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining, 272029, Shandong, China
| | - Junjun Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Shengnan Chen
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Jing Cai
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Zhaoyang Zeng
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiaoling Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Wei Xiong
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Li Wang
- Department of Thoracic Surgery, The Second Xiangya Hospital of Central South University, Changsha, 410008, Hunan, China
| | - Guiyuan Li
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China.,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Bo Xiang
- The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha, 410078, Hunan, China. .,Cancer Research Institute, Xiangya School of Medicine, Central South University, Changsha, 410078, Hunan, China.
| |
Collapse
|
30
|
Xu Z, Bian H, Zhang F, Mi R, Wang Q, Lu Y, Zheng Q, Gu J. URI promotes the migration and invasion of human cervical cancer cells potentially via upregulation of vimentin expression. Am J Transl Res 2017; 9:3037-3047. [PMID: 28670391 PMCID: PMC5489903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 05/19/2017] [Indexed: 06/07/2023]
Abstract
URI is known to act as an oncoprotein in several tumors. Our previous studies have shown that URI is associated with the migration process in cervical and gastric cancer cells, but the mechanisms remain to be determined. Given the fact that URI positively regulates vimentin expression, we therefore investigated how URI regulated vimentin expression affects the migration and invasion of cells from two human cervical cancer cell lines HeLa and C33A, which differentially express URI. We have shown that knock-down of URI in HeLa cells using URI siRNA caused decreased vimentin mRNA and protein levels along with attenuated cell motility. Meanwhile, overexpression of URI by transfection of PCMV6-URI in C33A cells resulted in increased vimentin expression and enhanced cell migration and invasion. We have also used TGF-β to induce vimentin expression, which enhanced the cell migration and invasion abilities affected by URI, while inhibition of vimentin by siRNA attenuated URI's effect on cell migration and invasion. In addition, we have performed luciferase reporter and ChIP assays, and the results support that URI indirectly enhances the activity of vimentin promoter. Taken together, our results suggest that URI plays essential roles in the migration and invasion of human cervical cancer cells, possibly via targeting vimentin expression.
Collapse
Affiliation(s)
- Zhonghai Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Huiqin Bian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Fei Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Rui Mi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Qian Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Yaojuan Lu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Qiping Zheng
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| | - Junxia Gu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Department of Hematology and Hematological Laboratory Science, School of Medicine, Jiangsu UniversityZhenjiang 212013, China
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Adipocytes have adapted to store energy in the form of lipid and also secrete circulating factors called adipokines that signal to other tissues to coordinate energy homeostasis. These functions are disrupted in the setting of obesity, promoting the development of diseases such as diabetes, cardiovascular disease, and cancer. RECENT FINDINGS Obesity is linked to an increased risk of many types of cancer and increased cancer-related mortality. The basis for the striking association between obesity and cancer is not well understood. Here, we review the cellular and molecular pathways that appear to be involved in obesity-driven cancer. We also describe possible therapeutic considerations and highlight important unanswered questions in the field.
Collapse
Affiliation(s)
- Sarah E Ackerman
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Olivia A Blackburn
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
- Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - François Marchildon
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA
| | - Paul Cohen
- Laboratory of Molecular Metabolism, The Rockefeller University, New York, NY, USA.
- The Rockefeller University, 1230 York Avenue, Box 223, New York, NY, 10065, USA.
| |
Collapse
|
32
|
Werner S, Stenzl A, Pantel K, Todenhöfer T. Expression of Epithelial Mesenchymal Transition and Cancer Stem Cell Markers in Circulating Tumor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 994:205-228. [DOI: 10.1007/978-3-319-55947-6_11] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
33
|
Mei Y, Yang JP, Qian CN. For robust big data analyses: a collection of 150 important pro-metastatic genes. CHINESE JOURNAL OF CANCER 2017; 36:16. [PMID: 28109319 PMCID: PMC5251273 DOI: 10.1186/s40880-016-0178-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 11/03/2016] [Indexed: 02/08/2023]
Abstract
Metastasis is the greatest contributor to cancer-related death. In the era of precision medicine, it is essential to predict and to prevent the spread of cancer cells to significantly improve patient survival. Thanks to the application of a variety of high-throughput technologies, accumulating big data enables researchers and clinicians to identify aggressive tumors as well as patients with a high risk of cancer metastasis. However, there have been few large-scale gene collection studies to enable metastasis-related analyses. In the last several years, emerging efforts have identified pro-metastatic genes in a variety of cancers, providing us the ability to generate a pro-metastatic gene cluster for big data analyses. We carefully selected 285 genes with in vivo evidence of promoting metastasis reported in the literature. These genes have been investigated in different tumor types. We used two datasets downloaded from The Cancer Genome Atlas database, specifically, datasets of clear cell renal cell carcinoma and hepatocellular carcinoma, for validation tests, and excluded any genes for which elevated expression level correlated with longer overall survival in any of the datasets. Ultimately, 150 pro-metastatic genes remained in our analyses. We believe this collection of pro-metastatic genes will be helpful for big data analyses, and eventually will accelerate anti-metastasis research and clinical intervention.
Collapse
Affiliation(s)
- Yan Mei
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Jun-Ping Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China
| | - Chao-Nan Qian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China. .,Department of Nasopharyngeal Carcinoma, Sun Yat-sen University Cancer Center, Guangzhou, 510060, Guangdong, P. R. China.
| |
Collapse
|
34
|
Ben-Shmuel S, Rashed R, Rostoker R, Isakov E, Shen-Orr Z, LeRoith D. Activating Transcription Factor-5 Knockdown Reduces Aggressiveness of Mammary Tumor Cells and Attenuates Mammary Tumor Growth. Front Endocrinol (Lausanne) 2017; 8:173. [PMID: 28785242 PMCID: PMC5519529 DOI: 10.3389/fendo.2017.00173] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/05/2017] [Indexed: 12/13/2022] Open
Abstract
Activating transcription factor-5 (ATF5) is an anti-apoptotic factor and has been implicated in enhancing the survival of cancer cells under stress and in regulating the autophagy process. Targeting ATF5 in anticancer therapy may be particularly attractive because of its differential role in cancer cells than in non-transformed cells, thus allowing specificity of the treatment. Using the delivery of short hairpin RNA vectors into the Mvt1 and Met1 cell lines, we tested the role of ATF5 in the development of mammary tumors in vivo and in regulating proliferation and migration of these cells in vitro. In this study, we demonstrate that knockdown of ATF5 (ATF5-KD) in both cell lines results in a decreased tumor volume and weight, as well as in a reduced proliferation rate and migratory potential of the cells. In addition, ATF5-KD led to an increased autophagy flux and a shift in the sub-populations comprising Mvt1 cells from the aggressive CD24-positive cells toward less aggressive CD24-negative cells. Taken together, these findings suggest that ATF5 plays an important role in enhancing mammary tumor cells overall aggressiveness and in promoting mammary tumor growth and emphasize the possible benefit of anti-ATF5 therapy in breast cancer patients, particularly, against tumors characterized with the positive expression of cell surface CD24.
Collapse
Affiliation(s)
- Sarit Ben-Shmuel
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
| | - Rola Rashed
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | - Ran Rostoker
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
| | - Elina Isakov
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
| | - Zila Shen-Orr
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
| | - Derek LeRoith
- Clinical Research Institute at Rambam (CRIR), Diabetes and Metabolism Clinical Research Center of Excellence, Rambam Medical Center, Haifa, Israel
- Division of Endocrinology, Diabetes and Bone Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Derek LeRoith,
| |
Collapse
|