151
|
β-catenin activation down-regulates cell-cell junction-related genes and induces epithelial-to-mesenchymal transition in colorectal cancers. Sci Rep 2019; 9:18440. [PMID: 31804558 PMCID: PMC6895046 DOI: 10.1038/s41598-019-54890-9] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 11/18/2019] [Indexed: 12/30/2022] Open
Abstract
WNT signaling activation in colorectal cancers (CRCs) occurs through APC inactivation or β-catenin mutations. Both processes promote β-catenin nuclear accumulation, which up-regulates epithelial-to-mesenchymal transition (EMT). We investigated β-catenin localization, transcriptome, and phenotypic differences of HCT116 cells containing a wild-type (HCT116-WT) or mutant β-catenin allele (HCT116-MT), or parental cells with both WT and mutant alleles (HCT116-P). We then analyzed β-catenin expression and associated phenotypes in CRC tissues. Wild-type β-catenin showed membranous localization, whereas mutant showed nuclear localization; both nuclear and non-nuclear localization were observed in HCT116-P. Microarray analysis revealed down-regulation of Claudin-7 and E-cadherin in HCT116-MT vs. HCT116-WT. Claudin-7 was also down-regulated in HCT116-P vs. HCT116-WT without E-cadherin dysregulation. We found that ZEB1 is a critical EMT factor for mutant β-catenin-mediated loss of E-cadherin and Claudin-7 in HCT116-P and HCT116-MT cells. We also demonstrated that E-cadherin binds to both WT and mutant β-catenin, and loss of E-cadherin releases β-catenin from the cell membrane and leads to its degradation. Alteration of Claudin-7, as well as both Claudin-7 and E-cadherin respectively caused tight junction (TJ) impairment in HCT116-P, and dual loss of TJs and adherens junctions (AJs) in HCT116-MT. TJ loss increased cell motility, and subsequent AJ loss further up-regulated that. Immunohistochemistry analysis of 101 CRCs revealed high (14.9%), low (52.5%), and undetectable (32.6%) β-catenin nuclear expression, and high β-catenin nuclear expression was significantly correlated with overall survival of CRC patients (P = 0.009). Our findings suggest that β-catenin activation induces EMT progression by modifying cell-cell junctions, and thereby contributes to CRC aggressiveness.
Collapse
|
152
|
Xia WX, Zhang LH, Liu YW. Weighted Gene Co-Expression Network Analysis Reveals Six Hub Genes Involved in and Tight Junction Function in Pancreatic Adenocarcinoma and their Potential Use in Prognosis. Genet Test Mol Biomarkers 2019; 23:829-836. [PMID: 31821092 DOI: 10.1089/gtmb.2019.0122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: Pancreatic adenocarcinoma (PAAD) is an aggressive and invasive tumor with poor prognosis. Identifying prognostic biomarkers of PAAD will provide crucial information for developing treatment plans. Methods: In this analysis, a gene-expression dataset, containing RNA-sequencing data recalculated into transcripts per million, was obtained from the UCSC Xena platform. Three thousand nine hundred and seventy six differentially expressed genes were obtained with analysis of variance. Using these data a co-expression network was constructed using weighted gene co-expression network analysis, from which we obtained eight modules. Results: The blue module included 497 genes and demonstrated significant negative correlation with overall survival. Furthermore, pathway analyses demonstrated the involvement of many of these genes in the tight junction pathway, which plays a critical role in PAAD. In addition, we identified six genes in common (i.e., ANXA2 [annexin A2], EPHA2 [erythropoietin-producing hepatocellular class A2], ITGB4 [integrin beta 4], KRT19 [keratin type I cytoskeletal 19], LGALS3 [galectin-3], and S100A14 [S100 calcium binding protein A14]) between the protein-protein interaction and gene co-expression networks that may have critical functions in PAAD. These hub genes were not only highly expressed at the RNA level but also exhibited high expression in the immunohistological data in the Human Protein Atlas Database. Conclusion: Thus, this research clarified the framework of co-expressed gene modules in PAAD and highlighted potential prognostic biomarkers for the clinical diagnosis of PAAD.
Collapse
Affiliation(s)
- Wang-Xiao Xia
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, China
| | - Lin-Heng Zhang
- Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing, China
| | - Yao-Wen Liu
- Shaanxi Key Laboratory of Brain Disorders, Institute of Basic Translational Medicine, Xi'an Medical University, Xi'an, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
153
|
Maity G, Chakraborty J, Ghosh A, Haque I, Banerjee S, Banerjee SK. Aspirin suppresses tumor cell-induced angiogenesis and their incongruity. J Cell Commun Signal 2019; 13:491-502. [PMID: 30610526 PMCID: PMC6946772 DOI: 10.1007/s12079-018-00499-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/26/2018] [Indexed: 01/10/2023] Open
Abstract
Tumor neovascularization/tumor angiogenesis is a pathophysiological process in which new blood vessels are formed from existing blood vessels in the primary tumors to supply adequate oxygen and nutrition to cancer cells for their proliferation and metastatic growth to the distant organs. Therefore, controlling tumor angiogenesis is an attractive target for cancer therapy. Structural abnormalities of the vasculature (i.e., leakiness due to the abnormal lining of pericytes on the microvessels) are one of the critical features of tumor angiogenesis that sensitizes vascular cells to cytokines and helps circulating tumor cells to metastasize to distant organs. Our goal is to repurpose the drugs that may prevent tumor angiogenesis or normalize the vessels by repairing leakiness via recruiting pericytes or both. In this study, we tested whether aspirin (ASA), which could block primary tumor growth, regulates tumor angiogenesis. We investigated the effects of low (1 mM) and high (2.5 mM) doses of ASA (direct effect), and ASA-treated or untreated triple negative breast cancer (TNBC) cells' conditioned media (indirect effect) on endothelial cell physiology. These include in vitro migration using modified Boyden chamber assay, in vitro capillary-like structure formation on Matrigel, interactions of pericytes-endothelial cells and cell permeability using in vitro endothelial permeability assay. We also examined the effect of ASA on various molecular factors associated with tumor angiogenesis. Finally, we found the outcome of ASA treatment on in vivo tumor angiogenesis. We found that ASA-treatment (direct or indirect) significantly blocks in vitro migration and capillary-like structure formation by endothelial cells. Besides, we found that ASA recruits pericytes from multipotent stem cells and helps in binding with endothelial cells, which is a hallmark of normalization of blood vessels, and decreases in vitro permeability through endothelial cell layer. The antiangiogenic effect of ASA was also documented in vivo assays. Mechanistically, ASA treatment blocks several angiogenic factors that are associated with tumor angiogenesis, and suggesting ASA blocks paracrine-autocrine signaling network between tumor cells and endothelial cells. Collectively, these studies implicate aspirin with proper dose may provide potential therapeutic for breast cancer via blocking as well as normalizing tumor angiogenesis.
Collapse
Affiliation(s)
- Gargi Maity
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jinia Chakraborty
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Blue Valley High School, 16200 Antioch Rd, Overland Park, KS, 66085, USA
| | - Arnab Ghosh
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Inamul Haque
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Snigdha Banerjee
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sushanta K Banerjee
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
154
|
Sun X, Cui S, Fu X, Liu C, Wang Z, Liu Y. MicroRNA-146-5p promotes proliferation, migration and invasion in lung cancer cells by targeting claudin-12. Cancer Biomark 2019; 25:89-99. [PMID: 31006666 DOI: 10.3233/cbm-182374] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
MicroRNAs (miRNAs) have been regarded as important regulators in different pathological processes of cells. Abnormal expression of miRNAs is frequently associated with cell proliferation, metastasis and apoptosis in various cancers. This study aimed to explore the effect of miR-146-5p on cell growth, metastasis and its mechanism in lung cancer cells. The expressions of miR-146-5p and claudin-12 in A549 and WI-38 cells were altered by transient transfection. Cisplatin was used to develop cells for regulation of cisplatin sensitivity. Cell viability, migration, invasion, and apoptosis were analyzed by CCK-8, Transwell and flow cytometry assays. The protein expressions of Wnt/β-catenin and PI3K/AKT/MAPK pathway-related factors were detected. miR-146-5p suppression inhibited cell viability, migration and invasion but promoted apoptosis in A549 cells. Moreover, overexpression of miR-146-5p reduced the sensitivity of A549 cells and WI-38 cells to cisplatin. In addition, claudin-12 was a direct target of miR-146-5p and was negatively regulated by miR-146-5p. Claudin-12 silence significantly reversed miR-146-5p suppression-mediated anti-tumor effects in A549 cells. Furthermore, miR-146-5p overexpression activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways via down-regulation of claudin-12. The results indicated that miR-146-5p promoted cell viability, migration and invasion, inhibited apoptosis and activated Wnt/β-catenin and PI3K/AKT/MAPK signal pathways by regulating claudin-12 expression in lung cancer cells.
Collapse
Affiliation(s)
- Xianghong Sun
- Department of Outpatient (Tumor), The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Shichao Cui
- Department of Respiratory Medicine, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Xiaofeng Fu
- Department of Outpatient, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| | - Chuan Liu
- Department of Surgery, Qingdao West Coast New District No.2 People's Hospital, Qingdao, Shandong, China
| | - Zhi Wang
- Department of Pharmacy, The Affiliated Hospital of Qingdao University (Shinan Branch), Qingdao, Shandong, China
| | - Yuanwei Liu
- Department of Cancer Comprehensive Therapy, The Affiliated Hospital of Qingdao University (West Coast Branch), Qingdao, Shandong, China
| |
Collapse
|
155
|
Kyuno T, Kyuno D, Kohno T, Konno T, Kikuchi S, Arimoto C, Yamaguchi H, Imamura M, Kimura Y, Kondoh M, Takemasa I, Kojima T. Tricellular tight junction protein LSR/angulin-1 contributes to the epithelial barrier and malignancy in human pancreatic cancer cell line. Histochem Cell Biol 2019; 153:5-16. [DOI: 10.1007/s00418-019-01821-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
|
156
|
Khan GJ, Sun L, Abbas M, Naveed M, Jamshaid T, Baig MMFA, Yuan S. In-vitro Pre-Treatment of Cancer Cells with TGF-β1: A Novel Approach of Tail Vein Lung Cancer Metastasis Mouse Model for Anti-Metastatic Studies. Curr Mol Pharmacol 2019; 12:249-260. [DOI: 10.2174/1874467212666190306165703] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/10/2019] [Accepted: 02/12/2019] [Indexed: 02/06/2023]
Abstract
Background:
Aggressive behavior of tumor metastasis comes from certain mutations,
changes in cellular metabolic and signaling pathways that are majorly altered by tumor microenvironment
(TME), its other components and growth factors like transforming growth factor-β1 (TGF-β1)
which is chiefly known for its epithelial to mesenchymal transformation (EMT). EMT is a critical step
of metastasis cascade in actual human lung cancer scenario.
Objective:
Our present study is focused on unveiling the in-vivo metastatic behavior of TGF-β1 treated
lung cancer cells that undergo EMT.
Methods:
The lung cancer epithelial A549 cells were treated in-vitro with TGF-β1 (3-5ng/ml for 72 h)
for EMT. After confirming the transformation of cells by phenotype modifications, wound healing and
cell migration assay and qRT-PCR analyses of EMT biomarkers including E. Cadherin, Vimentin,
Snail, Slug, MMP2 and MMP9; those TGF-β1 modified cells were probed with fluorescent trackers and
were injected into the tail vein of BALB/c nude mice for metastatic dissemination studies.
Results:
Our findings indicate that the distribution of TGF-β1 treated A549 cells as compared to W.T
A549 towards lungs is less in terms of total relative fluorescent cluster count, however, the difference is
insignificant (52±4, 60±5 respectively). Additionally, we show that TGF-β1 treated cells tend to metastasize
almost 2, 3, 1.5, 2 and 1.7 times more than W.T towards liver, brain, ovaries, bones and adrenal
gland, respectively, which is very much like human lung cancer metastasis.
Conclusion:
Conclusively, it is the first study ever reporting that a pre-treatment of cells with TGF-β1
for experimental lung cancer metastasis mouse model may portray a more precise approach for the
development of potential therapeutic treatments. Additional pre-treatment studies with the application
of other TME conditions like hypoxia and factors like NFκB, VEGF etc. may be a future prospect to
develop a better understanding.
Collapse
Affiliation(s)
- Ghulam Jilany Khan
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Li Sun
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| | - Muhammad Abbas
- State key Laboratory of Pharmaceutical Biotechnology, Nanjing University, 210023, Nanjing, China
| | - Muhammad Naveed
- Department of Clinical Pharmacology, School of Pharmacy, Nanjing Medical University, 211166, Nanjing, China
| | - Talha Jamshaid
- Department of Pharmaceutics, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | | | - Shengtao Yuan
- Jiangsu key laboratory of Drug Screening, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
157
|
Koohini Z, Koohini Z, Teimourian S. Slit/Robo Signaling Pathway in Cancer; a New Stand Point for Cancer Treatment. Pathol Oncol Res 2019; 25:1285-1293. [PMID: 30610466 DOI: 10.1007/s12253-018-00568-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Accepted: 12/17/2018] [Indexed: 10/27/2022]
Abstract
Angiogenesis and metastasis are two critical steps for cancer cells survival and migration. The microenvironment of tumor sphere induces new blood vessels formation for enhancing tumor mass. Preexisting capillaries and postcapillary venules in tumors bring about new blood vessels. ROBO1-ROBO4 are transmembrane receptors family which act as guidance molecules of the nervous system. The SLITs family is secreted glycoproteins that bind to these receptors. SLIT-ROBO signaling pathway plays an important role in neurogenesis and immune response. Linkage between ROBOs and their ligands (SLITs) induce chemorepllent signal for regulation of axon guidance and leukocyte cell migration, recent finding shows that it is also involved in endothelial cell migration and angiogenesis in various type of cancers. In this article we review recent finding of SLIT-ROBO pathway in angiogenesis and metastasis.
Collapse
Affiliation(s)
- Zahra Koohini
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zohreh Koohini
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
158
|
Pranda MA, Gray KM, DeCastro AJL, Dawson GM, Jung JW, Stroka KM. Tumor Cell Mechanosensing During Incorporation into the Brain Microvascular Endothelium. Cell Mol Bioeng 2019; 12:455-480. [PMID: 31719927 DOI: 10.1007/s12195-019-00591-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor metastasis to the brain occurs in approximately 20% of all cancer cases and often occurs due to tumor cells crossing the blood-brain barrier (BBB). The brain microenvironment is comprised of a soft hyaluronic acid (HA)-rich extracellular matrix with an elastic modulus of 0.1-1 kPa, whose crosslinking is often altered in disease states. Methods To explore the effects of HA crosslinking on breast tumor cell migration, we developed a biomimetic model of the human brain endothelium, consisting of brain microvascular endothelial cell (HBMEC) monolayers on HA and gelatin (HA/gelatin) films with different degrees of crosslinking, as established by varying the concentration of the crosslinker Extralink. Results and Discussion Metastatic breast tumor cell migration speed, diffusion coefficient, spreading area, and aspect ratio increased with decreasing HA crosslinking, a mechanosensing trend that correlated with tumor cell actin organization but not CD44 expression. Meanwhile, breast tumor cell incorporation into endothelial monolayers was independent of HA crosslinking density, suggesting that alterations in HA crosslinking density affect tumor cells only after they exit the vasculature. Tumor cells appeared to exploit both the paracellular and transcellular routes of trans-endothelial migration. Quantitative phenotyping of HBMEC junctions via a novel Python software revealed a VEGF-dependent decrease in punctate VE-cadherin junctions and an increase in continuous and perpendicular junctions when HBMECs were treated with tumor cell-secreted factors. Conclusions Overall, our quantitative results suggest that a combination of biochemical and physical factors promote tumor cell migration through the BBB.
Collapse
Affiliation(s)
- Marina A Pranda
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kelsey M Gray
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Ariana Joy L DeCastro
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Gregory M Dawson
- Department of Biology, University of Maryland, College Park, College Park, MD 20742 USA
| | - Jae W Jung
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA
| | - Kimberly M Stroka
- Fischell Department of Bioengineering, University of Maryland, College Park, College Park, MD 20742 USA.,Biophysics Program, University of Maryland, College Park, College Park, MD 20742 USA.,Center for Stem Cell Biology and Regenerative Medicine, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland - Baltimore, Baltimore, MD 21201 USA.,Fischell Department of Bioengineering, University of Maryland, College Park, 3110 A. James Clark Hall, 8278 Paint Branch Drive, College Park, MD 20742 USA
| |
Collapse
|
159
|
Reduction of metastatic potential by inhibiting EGFR/Akt/p38/ERK signaling pathway and epithelial-mesenchymal transition after carbon ion exposure is potentiated by PARP-1 inhibition in non-small-cell lung cancer. BMC Cancer 2019; 19:829. [PMID: 31438892 PMCID: PMC6704719 DOI: 10.1186/s12885-019-6015-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 08/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Carbon ion (12C) radiotherapy is becoming very promising to kill highly metastatic cancer cells keeping adjacent normal cells least affected. Our previous study shows that combined PARP-1 inhibition with 12C ion reduces MMP-2,-9 synergistically in HeLa cells but detailed mechanism are not clear. To understand this mechanism and the rationale of using PARP-1 inhibitor with 12C ion radiotherapy for better outcome in controlling metastasis, we investigated metastatic potential in two non-small cell lung cancer (NSCLC) A549 and H1299 (p53-deficient) cells exposed with 12C ion in presence and absence of PARP-1 inhibition using siRNA or olaparib. METHODS We monitored cell proliferation, in-vitro cell migration, wound healing, expression and activity of MMP-2, - 9 in A549 and p53-deficient H1299 cell lines exposed with 12C ion with and without PARP-1 inhibitor olaparib/DPQ. Expression and phosphorylation of NF-kB, EGFR, Akt, p38, ERK was also observed in A549 and H1299 cells exposed with 12C ion with and without PARP-1 inhibition using siRNA or olaparib. We also checked expression of few marker genes involved in epithelial-mesenchymal transition (EMT) pathways like N-cadherin, vimentin, anillin, claudin-1, - 2 in both NSCLC. To determine the generalized effect of 12C ion and olaparib in inhibition of cell's metastatic potential, wound healing and activity of MMP-2, - 9 was also studied in HeLa and MCF7 cell lines after 12C ion exposure and in combination with PARP-1 inhibitor olaparib. RESULTS Our experiments show that 12C ion and PARP-1 inhibition separately reduces cell proliferation, cell migration, wound healing, phosphorylation of EGFR, Akt, p38, ERK resulting inactivation of NF-kB. Combined treatment abolishes NF-kB expression and hence synergistically reduces MMP-2, - 9 expressions. Each single treatment reduces N-cadherin, vimentin, anillin but increases claudin-1, - 2 leading to suppression of EMT process. However, combined treatment synergistically alters these proteins to suppress EMT pathways significantly. CONCLUSION The activation pathways of transcription of MMP-2,-9 via NF-kB and key marker proteins in EMT pathways are targeted by both 12C ion and olaparib/siRNA. Hence, 12C ion radiotherapy could potentially be combined with olaparib as chemotherapeutic agent for better control of cancer metastasis.
Collapse
|
160
|
Kohno T, Konno T, Kojima T. Role of Tricellular Tight Junction Protein Lipolysis-Stimulated Lipoprotein Receptor (LSR) in Cancer Cells. Int J Mol Sci 2019; 20:E3555. [PMID: 31330820 PMCID: PMC6679224 DOI: 10.3390/ijms20143555] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 02/07/2023] Open
Abstract
Maintaining a robust epithelial barrier requires the accumulation of tight junction proteins, LSR/angulin-1 and tricellulin, at the tricellular contacts. Alterations in the localization of these proteins temporarily cause epithelial barrier dysfunction, which is closely associated with not only physiological differentiation but also cancer progression and metastasis. In normal human endometrial tissues, the endometrial cells undergo repeated proliferation and differentiation under physiological conditions. Recent observations have revealed that the localization and expression of LSR/angulin-1 and tricellulin are altered in a menstrual cycle-dependent manner. Moreover, it has been shown that endometrial cancer progression affects these alterations. This review highlights the differences in the localization and expression of tight junction proteins in normal endometrial cells and endometrial cancers and how they cause functional changes in cells.
Collapse
Affiliation(s)
- Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo 060-8556, Japan.
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| |
Collapse
|
161
|
Cordero A, Kanojia D, Miska J, Panek WK, Xiao A, Han Y, Bonamici N, Zhou W, Xiao T, Wu M, Ahmed AU, Lesniak MS. FABP7 is a key metabolic regulator in HER2+ breast cancer brain metastasis. Oncogene 2019; 38:6445-6460. [PMID: 31324889 PMCID: PMC6742563 DOI: 10.1038/s41388-019-0893-4] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 05/08/2019] [Accepted: 05/08/2019] [Indexed: 11/09/2022]
Abstract
Overexpression of human epidermal growth factor receptor 2 (HER2) in breast cancer patients is associated with increased incidence of breast cancer brain metastases (BCBM), but the mechanisms underlying this phenomenon remain unclear. Here, to identify brain-predominant genes critical for the establishment of BCBM, we conducted an in silico screening analysis and identified that increased levels of fatty acid-binding protein 7 (FABP7) correlate with a lower survival and higher incidence of brain metastases in breast cancer patients. We validated these findings using HER2+ BCBM cells compared with parental breast cancer cells. Importantly, through knockdown and overexpression assays, we characterized the role of FABP7 in the BCBM process in vitro and in vivo. Our results uncover a key role of FABP7 in metabolic reprogramming of HER2 + breast cancer cells, supporting a glycolytic phenotype and storage of lipid droplets that enable their adaptation and survival in the brain microenvironment. In addition, FABP7 is shown to be required for upregulation of key metastatic genes and pathways, such as integrins-Src and VEGFA, and for the growth of HER2+ breast cancer cells in the brain microenvironment in vivo. Together, our results support FABP7 as a potential target for the treatment of HER2+ BCBM.
Collapse
Affiliation(s)
- Alex Cordero
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Deepak Kanojia
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Jason Miska
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Wojciech K Panek
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Annie Xiao
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Yu Han
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Nicolas Bonamici
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Weidong Zhou
- Center for Applied Proteomics and Molecular Medicine, George Mason University, Manassas, VA, 20110, USA
| | - Ting Xiao
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Meijing Wu
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Atique U Ahmed
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Maciej S Lesniak
- Department of Neurological Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA.
| |
Collapse
|
162
|
Song ZY, Chao F, Zhuo Z, Ma Z, Li W, Chen G. Identification of hub genes in prostate cancer using robust rank aggregation and weighted gene co-expression network analysis. Aging (Albany NY) 2019; 11:4736-4756. [PMID: 31306099 PMCID: PMC6660050 DOI: 10.18632/aging.102087] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 07/04/2019] [Indexed: 12/20/2022]
Abstract
The pathogenic mechanisms of prostate cancer (PCa) remain to be defined. In this study, we utilized the Robust Rank Aggregation (RRA) method to integrate 10 eligible PCa microarray datasets from the GEO and identified a set of significant differentially expressed genes (DEGs) between tumor samples and normal, matched specimens. To explore potential associations between gene sets and PCa clinical features and to identify hub genes, we utilized WGCNA to construct gene co-expression networks incorporating the DEGs screened with the use of RRA. From the key module, we selected LMNB1, TK1, ZWINT, and RACGAP1 for validation. We found that these genes were up-regulated in PCa samples, and higher expression levels were associated with higher Gleason scores and tumor grades. Moreover, ROC and K-M plots indicated these genes had good diagnostic and prognostic value for PCa. On the other hand, methylation analyses suggested that the abnormal up-regulation of these four genes likely resulted from hypomethylation, while GSEA and GSVA for single hub gene revealed they all had a close association with proliferation of PCa cells. These findings provide new insight into PCa pathogenesis, and identify LMNB1, TK1, RACGAP1 and ZWINT as candidate biomarkers for diagnosis and prognosis of PCa.
Collapse
Affiliation(s)
- Zhen-yu Song
- Department of Urology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Fan Chao
- Department of Urology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhiyuan Zhuo
- Department of Urology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Zhe Ma
- Department of Urology, Jinshan Hospital of Fudan University, Shanghai, China
| | - Wenzhi Li
- Department of Urology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Gang Chen
- Department of Urology, Jinshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
163
|
Zhao B, Che D, Adams S, Guo N, Han R, Zhang C, Qin G, Farouk MH, Jiang H. N-Acetyl-d-galactosamine prevents soya bean agglutinin-induced intestinal barrier dysfunction in intestinal porcine epithelial cells. J Anim Physiol Anim Nutr (Berl) 2019; 103:1198-1206. [PMID: 30934149 DOI: 10.1111/jpn.13091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 02/25/2019] [Accepted: 02/25/2019] [Indexed: 12/23/2022]
Abstract
Soya bean agglutinin (SBA) is a glycoprotein and the main anti-nutritional component in most soya bean feedstuffs. It is mainly a non-fibre carbohydrate-based protein and represents about 10% of soya bean-based anti-nutritional effects. In this study, we sought to determine the effects of N-Acetyl-D-galactosamine (GalNAc or D-GalNAc) on the damage induced by SBA on the membrane permeability and tight junction proteins of piglet intestinal epithelium (IPEC-J2) cells. The IPEC-J2 cells were pre-cultured with 0, 0.125 × 10-4 , 0.25 × 10-4 , 0.5 × 10-4 , 1.0 × 10-4 and 2.0 × 10-4 mmol/L GalNAc at different time period (1, 2, 4 and 8 hr) before being exposed to 0.5 mg/ml SBA for 24 hr. The results indicate that pre-incubation with GalNAc mitigates the mechanical barrier injury as reflected by a significant increase in trans-epithelial electric resistance (TEER) value and a decrease in alkaline phosphatase (ALP) activity in cell culture medium pre-treated with GalNAc before incubation with SBA as both indicate a reduction in cellular membrane permeability. In addition, mRNA levels of the tight junction proteins occludin and claudin-3 were lower in the SBA-treated groups without pre-treatment with GalNAc. The mRNA expression of occludin was reduced by 17.3% and claudin-3 by 42% (p < 0.01). Moreover, the corresponding protein expression levels were lowered by 17.8% and 43.5% (p < 0.05) respectively. However, in the GalNAc pre-treated groups, occludin and claudin-3 mRNAs were reduced by 1.6% (p > 0.05) and 2.7% (p < 0.01), respectively, while the corresponding proteins were reduced by 4.3% and 7.2% (p < 0.05). In conclusion, GalNAc may prevent the effect of SBA on membrane permeability and tight junction proteins on IPEC-J2s.
Collapse
Affiliation(s)
- Bao Zhao
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Dongsheng Che
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Seidu Adams
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Nan Guo
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Rui Han
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Chun Zhang
- Department of Animal Science and Technology, Changchun University of science and technology, Changchun, China
| | - Guixin Qin
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| | - Mohammed Hamdy Farouk
- Department of Animal Production, Faculty of Agriculture, Al-Azhar University, Cairo, Egypt
| | - Hailong Jiang
- Jilin Provincial Key Laboratory of Animal Nutrition and Feed Science, Jilin Agricultural University, Changchun, China
- Department of Animal Science and Technology, Jilin Agricultural University, Changchun, China
| |
Collapse
|
164
|
Zhang Z, Ruan L, Lu M, Yao X. Analysis of key candidate genes and pathways of endometriosis pathophysiology by a genomics-bioinformatics approach. Gynecol Endocrinol 2019; 35:576-581. [PMID: 30798642 DOI: 10.1080/09513590.2019.1576609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Endometriosis is a common disease in women, but the signaling pathways and driven genes involved remain unclear. This study integrated four datasets to elucidate potential key candidate genes and pathways in endometriosis. Four expression profile datasets including 29 endometriosis lesions and 37 normal tissues were integrated and analyzed. Differentially expressed genes (DEGs) were sorted, and the gene ontology, pathway enrichment, and protein-protein interaction network of candidate genes were then analyzed. A total of 94 shared DEGs were identified from the four datasets. The DEGs were clustered based on functions and signaling pathways through the analysis of significant enrichment. Among the DEG protein-protein interaction network complex, 87 nodes/DEGs were identified. Furthermore, 18 central node genes were identified, and most of the corresponding genes were involved in the angiotensin system, smooth muscle contraction, cell junction organization, and lipoxin pathways. Through integrated bioinformatic analysis, we identified candidate genes and pathways in endometriosis, which could improve our understanding of endometriosis.
Collapse
Affiliation(s)
- Zhimin Zhang
- a The Department of Obstetrics , The Fourth Hospital of Shijiazhuang City , Shijiazhuang , China
| | - Lin Ruan
- b Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns , Hebei University of Chinese Medicine , Shijiazhuang , China
| | - Mingxuan Lu
- c College of Integrative Medicine , Hebei University of Chinese Medicine , Shijiazhuang , China
| | - Xiaoguang Yao
- b Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns , Hebei University of Chinese Medicine , Shijiazhuang , China
- c College of Integrative Medicine , Hebei University of Chinese Medicine , Shijiazhuang , China
| |
Collapse
|
165
|
Ivana B, Emina M, Marijana MK, Irena J, Zoran B, Radmila J. High expression of junctional adhesion molecule-A is associated with poor survival in patients with epithelial ovarian cancer. Int J Biol Markers 2019; 34:262-268. [PMID: 31190601 DOI: 10.1177/1724600819850178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE Aberrant expression of different tight junction proteins, including the junctional adhesion molecule-A (JAM-A), has been frequently reported in association with tumor progression of several malignancies. To our knowledge, this is the first study examining the clinical significance of JAM-A gene expression in epithelial ovarian cancer. METHODS JAM-A expression levels in 44 epithelial ovarian cancer and 12 benign formalin-fixed paraffin-embedded samples were determined by reverse transcription quantitative polymerase chain reaction. Receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic and prognostic potential of JAM-A. Associations between JAM-A expression and clinicopathological characteristics of epithelial ovarian cancer were analyzed using Fisher's exact test. The Kaplan-Meier method and univariate Cox regression analysis were used for the survival analysis. P ⩽ 0.05 was considered statistically significant. RESULTS ROC curve analyses showed that JAM-A gene expression exhibits both diagnostic and prognostic performance in epithelial ovarian cancer (area under the curve (AUC) 0.640, 95% confidence interval (CI) 0.488, 0.792, sensitivity 43.18%, specificity 100% and AUC 0.621, 95% CI 0.427, 0.816, sensitivity 52.63%, specificity 85%, respectively). JAM-A expression was significantly associated with International Federation of Gynecologists and Obstetricians (FIGO) stage (P =0.049) and the Kaplan-Meier method demonstrated that patients with high expression of JAM-A had significantly worse overall survival compared to patients with low JAM-A expression (P =0.004). Moreover, univariate Cox regression analysis showed that FIGO stage, peritoneal metastasis, residual tumor and JAM-A expression were significantly associated with reduced overall survival in epithelial ovarian cancer. CONCLUSIONS Our results indicate that high levels of JAM-A expression are associated with an advanced clinicopathological feature and may have diagnostic potential; also, it could be a predictor of poor overall survival in patients with epithelial ovarian cancer.
Collapse
Affiliation(s)
- Boljevic Ivana
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova, Belgrade, Serbia
| | - Malisic Emina
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova, Belgrade, Serbia
| | - Milovic-Kovacevic Marijana
- Department of Medical Oncology, Institute for Oncology and Radiology of Serbia, Pasterova, Belgrade, Serbia
| | - Jovanic Irena
- Department of Pathology, Institute for Oncology and Radiology of Serbia, Pasterova, Belgrade, Serbia
| | - Bukumiric Zoran
- Institute for Medical Statistics and Informatics, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jankovic Radmila
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova, Belgrade, Serbia
| |
Collapse
|
166
|
Mierke CT. The matrix environmental and cell mechanical properties regulate cell migration and contribute to the invasive phenotype of cancer cells. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2019; 82:064602. [PMID: 30947151 DOI: 10.1088/1361-6633/ab1628] [Citation(s) in RCA: 138] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
The minimal structural unit of a solid tumor is a single cell or a cellular compartment such as the nucleus. A closer look inside the cells reveals that there are functional compartments or even structural domains determining the overall properties of a cell such as the mechanical phenotype. The mechanical interaction of these living cells leads to the complex organization such as compartments, tissues and organs of organisms including mammals. In contrast to passive non-living materials, living cells actively respond to the mechanical perturbations occurring in their microenvironment during diseases such as fibrosis and cancer. The transformation of single cancer cells in highly aggressive and hence malignant cancer cells during malignant cancer progression encompasses the basement membrane crossing, the invasion of connective tissue, the stroma microenvironments and transbarrier migration, which all require the immediate interaction of the aggressive and invasive cancer cells with the surrounding extracellular matrix environment including normal embedded neighboring cells. All these steps of the metastatic pathway seem to involve mechanical interactions between cancer cells and their microenvironment. The pathology of cancer due to a broad heterogeneity of cancer types is still not fully understood. Hence it is necessary to reveal the signaling pathways such as mechanotransduction pathways that seem to be commonly involved in the development and establishment of the metastatic and mechanical phenotype in several carcinoma cells. We still do not know whether there exist distinct metastatic genes regulating the progression of tumors. These metastatic genes may then be activated either during the progression of cancer by themselves on their migration path or in earlier stages of oncogenesis through activated oncogenes or inactivated tumor suppressor genes, both of which promote the metastatic phenotype. In more detail, the adhesion of cancer cells to their surrounding stroma induces the generation of intracellular contraction forces that deform their microenvironments by alignment of fibers. The amplitude of these forces can adapt to the mechanical properties of the microenvironment. Moreover, the adhesion strength of cancer cells seems to determine whether a cancer cell is able to migrate through connective tissue or across barriers such as the basement membrane or endothelial cell linings of blood or lymph vessels in order to metastasize. In turn, exposure of adherent cancer cells to physical forces, such as shear flow in vessels or compression forces around tumors, reinforces cell adhesion, regulates cell contractility and restructures the ordering of the local stroma matrix that leads subsequently to secretion of crosslinking proteins or matrix degrading enzymes. Hence invasive cancer cells alter the mechanical properties of their microenvironment. From a mechanobiological point-of-view, the recognized physical signals are transduced into biochemical signaling events that guide cellular responses such as cancer progression after the malignant transition of cancer cells from an epithelial and non-motile phenotype to a mesenchymal and motile (invasive) phenotype providing cellular motility. This transition can also be described as the physical attempt to relate this cancer cell transitional behavior to a T1 phase transition such as the jamming to unjamming transition. During the invasion of cancer cells, cell adaptation occurs to mechanical alterations of the local stroma, such as enhanced stroma upon fibrosis, and therefore we need to uncover underlying mechano-coupling and mechano-regulating functional processes that reinforce the invasion of cancer cells. Moreover, these mechanisms may also be responsible for the awakening of dormant residual cancer cells within the microenvironment. Physicists were initially tempted to consider the steps of the cancer metastasis cascade as single events caused by a single mechanical alteration of the overall properties of the cancer cell. However, this general and simple view has been challenged by the finding that several mechanical properties of cancer cells and their microenvironment influence each other and continuously contribute to tumor growth and cancer progression. In addition, basement membrane crossing, cell invasion and transbarrier migration during cancer progression is explained in physical terms by applying physical principles on living cells regardless of their complexity and individual differences of cancer types. As a novel approach, the impact of the individual microenvironment surrounding cancer cells is also included. Moreover, new theories and models are still needed to understand why certain cancers are malignant and aggressive, while others stay still benign. However, due to the broad variety of cancer types, there may be various pathways solely suitable for specific cancer types and distinct steps in the process of cancer progression. In this review, physical concepts and hypotheses of cancer initiation and progression including cancer cell basement membrane crossing, invasion and transbarrier migration are presented and discussed from a biophysical point-of-view. In addition, the crosstalk between cancer cells and a chronically altered microenvironment, such as fibrosis, is discussed including the basic physical concepts of fibrosis and the cellular responses to mechanical stress caused by the mechanically altered microenvironment. Here, is highlighted how biophysical approaches, both experimentally and theoretically, have an impact on classical hallmarks of cancer and fibrosis and how they contribute to the understanding of the regulation of cancer and its progression by sensing and responding to the physical environmental properties through mechanotransduction processes. Finally, this review discusses various physical models of cell migration such as blebbing, nuclear piston, protrusive force and unjamming transition migration modes and how they contribute to cancer progression. Moreover, these cellular migration modes are influenced by microenvironmental perturbances such as fibrosis that can induce mechanical alterations in cancer cells, which in turn may impact the environment. Hence, the classical hallmarks of cancer need to be refined by including biomechanical properties of cells, cell clusters and tissues and their microenvironment to understand mechano-regulatory processes within cancer cells and the entire organism.
Collapse
|
167
|
Zhou S, Piao X, Wang C, Wang R, Song Z. Identification of claudin‑1, ‑3, ‑7 and ‑8 as prognostic markers in human laryngeal carcinoma. Mol Med Rep 2019; 20:393-400. [PMID: 31115553 PMCID: PMC6580001 DOI: 10.3892/mmr.2019.10265] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 02/08/2019] [Indexed: 12/18/2022] Open
Abstract
Various genomic and epigenetic modifications that occur during the development of cancer act as potential biomarkers for early diagnosis and treatment. Previous studies have demonstrated abnormal expression of the claudin (CLDN) tight junction (TJ) proteins in numerous types of human cancer. Reverse transcription-quantitative polymerase chain reaction and western blotting were employed to investigate variations in the expression of the CLDN TJ proteins in laryngeal non-neoplastic tissues and laryngeal squamous carcinoma tissues. It was revealed that CLDN2, CLDN4, CLDN5, CLDN6, CLDN9, CLDN11 and CLDN12 were undetectable in laryngeal squamous carcinoma tissues and laryngeal non-neoplastic tissues. Additionally, CLDN10 was expressed in laryngeal squamous carcinoma tissues and laryngeal non-neoplastic tissues; however, no significant difference was reported. Conversely, the expression levels of CLDN1 and CLDN7 mRNA and protein were downregulated in laryngeal squamous carcinoma tissues compared with in adjacent non-neoplastic tissues, whereas those of CLDN3 and CLDN8 were upregulated. A total of 80 samples of laryngeal squamous carcinoma and non-neoplastic tissues were analyzed for the expression of CLDN1, −3, −7 and −8 via streptavidin-peroxidase immunohistochemical staining. It was revealed that the expression levels of CLDN1 and CLDN7 were downregulated in laryngeal squamous carcinoma tissues compared with in non-neoplastic mucosal tissues, whereas those of CLDN3 and CLDN8 were upregulated. Furthermore, the associations between CLDN expression and the clinicopathological factors of patients were analyzed. The expression levels of CLDN3 and CLDN7 were reported to be associated with distant metastasis and serve as potential predictors of poor prognosis. In conclusion, the findings of the present study demonstrated that the expression levels of CLDN1, −3, −7 and −8 varied between laryngeal squamous carcinoma tissues and non-neoplastic tissues. The expression levels of these CLDNs may be useful molecular markers for the diagnosis of laryngeal carcinoma, and determining the metastasis and prognosis of this disease.
Collapse
Affiliation(s)
- Shu Zhou
- Department of Anesthesiology, Jilin Cancer Hospital, Changchun, Jilin 130021, P.R. China
| | - Xue Piao
- Department of Anesthesiology, Maternity Hospital of Changchun City, Changchun, Jilin 130021, P.R. China
| | - Chengyan Wang
- Department of Ultrasound, Jilin Cancer Hospital, Changchun, Jilin 130021, P.R. China
| | - Rui Wang
- Department of Ultrasound, Jilin Cancer Hospital, Changchun, Jilin 130021, P.R. China
| | - Zhimin Song
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
168
|
Kang L, Shen L, Lu L, Wang D, Zhao Y, Chen C, Du L, Gong J, Zhang Y, Mi X, Xiang R, Zhang M, Tan X. Asparaginyl endopeptidase induces endothelial permeability and tumor metastasis via downregulating zonula occludens protein ZO-1. Biochim Biophys Acta Mol Basis Dis 2019; 1865:2267-2275. [PMID: 31096007 DOI: 10.1016/j.bbadis.2019.05.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 04/22/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Zona occludens-1 (ZO-1) is a key component of tight junctions that govern the function of the endothelial barrier against tumor metastasis. Factors secreted by tumor cells contribute to the maintenance of tumor vascular networks. How tumor cell-derived protein signals regulate ZO-1 expression is unclear. Here, we explored the effect of tumor cell-secreted asparaginyl endopeptidase (AEP) on the permeability of endothelial cells in the tumor microenvironment. First, we confirmed the existence of AEP in conditioned medium (CM) from AEP-overexpressing MDA-MB-231 and 4T1 cells. Treatment with CM from AEP-overexpressing tumor cells increased the permeability and tumor cell transversal of an endothelial monolayer. Furthermore, CM from AEP-overexpressing tumor cells suppressed endothelial ZO-1 expression, as well as ZO-1-associated nucleic acid binding protein ZONAB. In addition, the level of phosphorylated STAT3 was increased by treatment with AEP-containing CM. A mutation of RGD or blocking integrin αvβ3 with antibody recovered the ZO-1 downregulation induced by AEP. In vivo, a lung metastatic mouse model showed increased endothelial permeability in the AEP-overexpressing group compared with the control group. An orthotopic tumor transplantation model was established using AEP-overexpression and compared with mice receiving control 4T1 cells. Compared with controls, overexpression of AEP increased lung metastatic foci and area, as well as vascular instability in primary tumors or lung metastatic sites. Moreover, endothelial ZO-1 was decreased in the AEP-overexpressing group. Taken together, our data show that tumor cell-derived AEP increases the permeability of endothelial barriers. Interactions between RGD and endothelial integrin αvβ3 mediate this effect by downregulating ZO-1.
Collapse
Affiliation(s)
- Lichun Kang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Long Shen
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Liqing Lu
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Dekun Wang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yong Zhao
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Chuan'ai Chen
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Lingfang Du
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Junbo Gong
- Tianjin Key Laboratory of Modern Drug Delivery and High Efficiency, Tianjin University, Tianjin 300072, China
| | - Yuying Zhang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xue Mi
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Rong Xiang
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Mianzhi Zhang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing 100078, China.
| | - Xiaoyue Tan
- School of Medicine, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China.
| |
Collapse
|
169
|
Bregenzer ME, Horst EN, Mehta P, Novak CM, Raghavan S, Snyder CS, Mehta G. Integrated cancer tissue engineering models for precision medicine. PLoS One 2019; 14:e0216564. [PMID: 31075118 PMCID: PMC6510431 DOI: 10.1371/journal.pone.0216564] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tumors are not merely cancerous cells that undergo mindless proliferation. Rather, they are highly organized and interconnected organ systems. Tumor cells reside in complex microenvironments in which they are subjected to a variety of physical and chemical stimuli that influence cell behavior and ultimately the progression and maintenance of the tumor. As cancer bioengineers, it is our responsibility to create physiologic models that enable accurate understanding of the multi-dimensional structure, organization, and complex relationships in diverse tumor microenvironments. Such models can greatly expedite clinical discovery and translation by closely replicating the physiological conditions while maintaining high tunability and control of extrinsic factors. In this review, we discuss the current models that target key aspects of the tumor microenvironment and their role in cancer progression. In order to address sources of experimental variation and model limitations, we also make recommendations for methods to improve overall physiologic reproducibility, experimental repeatability, and rigor within the field. Improvements can be made through an enhanced emphasis on mathematical modeling, standardized in vitro model characterization, transparent reporting of methodologies, and designing experiments with physiological metrics. Taken together these considerations will enhance the relevance of in vitro tumor models, biological understanding, and accelerate treatment exploration ultimately leading to improved clinical outcomes. Moreover, the development of robust, user-friendly models that integrate important stimuli will allow for the in-depth study of tumors as they undergo progression from non-transformed primary cells to metastatic disease and facilitate translation to a wide variety of biological and clinical studies.
Collapse
Affiliation(s)
- Michael E. Bregenzer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Eric N. Horst
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Pooja Mehta
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Caymen M. Novak
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Shreya Raghavan
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Catherine S. Snyder
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Geeta Mehta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Department of Materials Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- Rogel Cancer Center, School of Medicine, University of Michigan, Ann Arbor, Michigan, United States of America
- Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
170
|
Ueno Y, Ozaki S, Umakoshi A, Yano H, Choudhury ME, Abe N, Sumida Y, Kuwabara J, Uchida R, Islam A, Ogawa K, Ishimaru K, Yorozuya T, Kunieda T, Watanabe Y, Takada Y, Tanaka J. Chloride intracellular channel protein 2 in cancer and non-cancer human tissues: relationship with tight junctions. Tissue Barriers 2019; 7:1593775. [PMID: 30929599 PMCID: PMC6592591 DOI: 10.1080/21688370.2019.1593775] [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] [Indexed: 12/31/2022] Open
Abstract
Chloride intracellular channel protein 2 (CLIC2) belongs to the CLIC family of conserved metazoan proteins. Although CLICs have been identified as chloride channels, they are currently considered multifunctional proteins. CLIC2 is the least studied family member. We investigated CLIC2 expression and localization in human hepatocellular carcinoma, metastatic colorectal cancer in the liver, and colorectal cancer. Significant expression of mRNAs encoding CLIC1, 2, 4, and 5 were found in the human tissues, but only CLIC2 was predominantly expressed in non-cancer tissues surrounding cancer masses. Fibrotic or dysfunctional (aspartate aminotransferase ≥40) non-cancer liver tissues and advanced stage HCC tissues expressed low levels of CLIC2. Endothelial cells lining blood vessels but not lymphatic vessels in non-cancer tissues expressed CLIC2 as well as high levels of the tight junction proteins claudins 1 and 5, occludin, and ZO-1. Most endothelial cells in blood vessels in cancer tissues had very low expressions of CLIC2 and tight junction proteins. CD31+/CD45− endothelial cells isolated from non-cancer tissues expressed mRNAs encoding CLIC2, claudin 1, occludin and ZO-1, while similar cell fractions from cancer tissues had very low expressions of these molecules. Knockdown of CLIC2 expression in human umbilical vein endothelial cells (HUVECs) allowed human cancer cells to transmigrate through a HUVEC monolayer. These results suggest that CLIC2 may be involved in the formation and/or maintenance of tight junctions and that cancer tissue vasculature lacks CLIC2 and tight junctions, which allows the intravasation of cancer cells necessary for hematogenous metastasis.
Collapse
Affiliation(s)
- Yoshitomo Ueno
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Saya Ozaki
- b Department of Neurosurgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Akihiro Umakoshi
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Hajime Yano
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Mohammed E Choudhury
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Naoki Abe
- d Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yutaro Sumida
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Jun Kuwabara
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Rina Uchida
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Afsana Islam
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Kohei Ogawa
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Kei Ishimaru
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Toshihiro Yorozuya
- d Department of Anesthesia and Perioperative Medicine, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Takeharu Kunieda
- b Department of Neurosurgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yuji Watanabe
- e Department of Gastrointestinal Surgery and Surgical Oncology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Yasutsugu Takada
- a Department of Hepato-biliary Pancreatic Surgery and Breast Surgery, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| | - Junya Tanaka
- c Department of Molecular and Cellular Physiology, Graduate School of Medicine , Ehime University , Toon , Ehime , Japan
| |
Collapse
|
171
|
Bhat AA, Uppada S, Achkar IW, Hashem S, Yadav SK, Shanmugakonar M, Al-Naemi HA, Haris M, Uddin S. Tight Junction Proteins and Signaling Pathways in Cancer and Inflammation: A Functional Crosstalk. Front Physiol 2019; 9:1942. [PMID: 30728783 PMCID: PMC6351700 DOI: 10.3389/fphys.2018.01942] [Citation(s) in RCA: 255] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 12/22/2018] [Indexed: 12/14/2022] Open
Abstract
The ability of epithelial cells to organize through cell–cell adhesion into a functioning epithelium serves the purpose of a tight epithelial protective barrier. Contacts between adjacent cells are made up of tight junctions (TJ), adherens junctions (AJ), and desmosomes with unique cellular functions and a complex molecular composition. These proteins mediate firm mechanical stability, serves as a gatekeeper for the paracellular pathway, and helps in preserving tissue homeostasis. TJ proteins are involved in maintaining cell polarity, in establishing organ-specific apical domains and also in recruiting signaling proteins involved in the regulation of various important cellular functions including proliferation, differentiation, and migration. As a vital component of the epithelial barrier, TJs are under a constant threat from proinflammatory mediators, pathogenic viruses and bacteria, aiding inflammation and the development of disease. Inflammatory bowel disease (IBD) patients reveal loss of TJ barrier function, increased levels of proinflammatory cytokines, and immune dysregulation; yet, the relationship between these events is partly understood. Although TJ barrier defects are inadequate to cause experimental IBD, mucosal immune activation is changed in response to augmented epithelial permeability. Thus, the current studies suggest that altered barrier function may predispose or increase disease progression and therapies targeted to specifically restore the barrier function may provide a substitute or supplement to immunologic-based therapies. This review provides a brief introduction about the TJs, AJs, structure and function of TJ proteins. The link between TJ proteins and key signaling pathways in cell proliferation, transformation, and metastasis is discussed thoroughly. We also discuss the compromised intestinal TJ integrity under inflammatory conditions, and the signaling mechanisms involved that bridge inflammation and cancer.
Collapse
Affiliation(s)
- Ajaz A Bhat
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Srijayaprakash Uppada
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Iman W Achkar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Sheema Hashem
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | - Santosh K Yadav
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar
| | | | - Hamda A Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.,Department of Biological and Environmental Sciences, Qatar University, Doha, Qatar
| | - Mohammad Haris
- Division of Translational Medicine, Research Branch, Sidra Medicine, Doha, Qatar.,Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
172
|
Wu JI, Wang LH. Emerging roles of gap junction proteins connexins in cancer metastasis, chemoresistance and clinical application. J Biomed Sci 2019; 26:8. [PMID: 30642339 PMCID: PMC6332853 DOI: 10.1186/s12929-019-0497-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/02/2019] [Indexed: 02/06/2023] Open
Abstract
Connexin, a four-pass transmembrane protein, contributes to assembly of gap junctions among neighboring cells and thus facilitates gap junctional intercellular communication (GJIC). Traditionally, the roles of connexins were thought to mediate formation of hemichannels and GJIC assembly for transportation of ions and small molecules. Many studies have observed loss of GJIC, due to reduced expression or altered cytoplasmic localization of connexins, in primary tumor cells. Connexins are generally considered tumor-suppressive. However, recent studies of clinical samples suggested a different role of connexins in that expression levels and membrane localization of connexins, including Connexin 43 (Cx43, GJA1) and Connexin 26 (Cx26, GJB2), were found to be enhanced in metastatic lesions of cancer patients. Cx43- and Cx26-mediated GJIC was found to promote cancer cell migration and adhesion to the pulmonary endothelium. Regulatory circuits involved in the induction of connexins and their functional effects have also been reported in various types of cancer. Connexins expressed in stromal cells were correlated with metastasis and were implicated in regulating metastatic behaviors of cancer cells. Recent studies have revealed that connexins can contribute to cellular phenotypes via multiple ways, namely 1) GJIC, 2) C-terminal tail-mediated signaling, and 3) cell-cell adhesion during gap junction formation. Both expression levels and the subcellular localization could participate determining the functional roles of connexins in cancer. Compounds targeting connexins were thus tested as potential therapeutics intervening metastasis or chemoresistance. This review focuses on the recent findings in the correlation between the expression of connexins and patients’ prognosis, their roles in metastasis and chemoresistance, as well as the implications and concerns of using connexin-targeting drugs as anti-metastatic therapeutics. Overall, connexins may serve as biomarkers for cancer prognosis and as therapeutic targets for intervening metastasis and chemoresistance.
Collapse
Affiliation(s)
- Jun-I Wu
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan.,Department of Life Sciences, National Central University, Taoyuan, Taiwan
| | - Lu-Hai Wang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli County, Taiwan. .,Department of Life Sciences, National Central University, Taoyuan, Taiwan. .,Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan. .,Chinese Medical Research Center, China Medical University, Taichung, Taiwan.
| |
Collapse
|
173
|
Shi F, Steuer A, Zhuang J, Kolb JF. Bioimpedance Analysis of Epithelial Monolayers after Exposure to Nanosecond Pulsed Electric Fields. IEEE Trans Biomed Eng 2018; 66:2010-2021. [PMID: 30452351 DOI: 10.1109/tbme.2018.2882299] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Exposures to pulsed electric fields (PEFs) are known to affect cell membranes and consequently also cell-cell interactions as well as associated characteristics. Bioimpedance analysis offers direct and non-invasive insights into structural and functional changes of cell membranes and extracellular matrices through a rigorous evaluation of electrical parameters. Accordingly, the multi-frequency impedance of confluent monolayers of rat liver epithelial WB-F344 cells was monitored in situ before and after exposure to nanosecond PEFs (nsPEFs). The results were fitted by two Cole models in series to obtain the Cole parameters for the monolayer. For an interpretation of the results, dielectric parameters, were correlated with changes of the TJ protein zonula occludens (ZO-1) and the paracellular permeability of the monolayer Cole parameters in general change as a function of pulse number and time. The findings demonstrate that impedance analysis is an effective method to monitor changes of TJs cell-cell contacts and paracellular permeability and relate them to exposure parameters.
Collapse
|
174
|
Zheng L, Jiang WD, Feng L, Wu P, Tang L, Kuang SY, Zeng YY, Zhou XQ, Liu Y. Selenium deficiency impaired structural integrity of the head kidney, spleen and skin in young grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2018; 82:408-420. [PMID: 30142391 DOI: 10.1016/j.fsi.2018.08.038] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 08/06/2018] [Accepted: 08/17/2018] [Indexed: 06/08/2023]
Abstract
This study focused on the effects of dietary selenium deficiency on structural integrity of the head kidney, spleen and skin in young grass carp (Ctenopharyngodon idella). A total of 540 healthy grass carp (mean weight 226.48 ± 0.68 g) were randomly divided into six groups and fed six separate diets with graded dietary levels of selenium (0.025-1.049 mg/kg diet) for 80 days. Results showed that selenium deficiency (1) caused oxidative damage in part by reducing the activities of antioxidant enzymes (such as SOD, CAT, GPx, GST and GR) and glutathione (GSH) content, down-regulating the transcript abundances of antioxidant enzymes (except GSTp1) partly related to Kelch-like-ECH-associated protein 1a (Keap1a)/NF-E2-related factor 2 (Nrf2) signalling; (2) aggravated apoptosis in part by up-regulating the mRNA levels of caspase-2, -3, -7, -8 and -9, which were partially related to p38MAPK/FasL/caspase-8 signalling and JNK/(BAX, Bcl-2, Mcl-1b, IAP)/(Apaf1, caspase-9) signalling; (3) damaged the tight junctions in part by down-regulating the mRNA levels of ZO-1 (except spleen), ZO-2 (except spleen), claudin-c, -f, -7, -11 and claudin-15, and up-regulating the mRNA levels of claudin-12, which were partially related to myosin light chain kinase (MLCK) signalling. Interesting, selenium deficiency failed to affect the expression of GSTp1, Keap1a, occludin, claudin-b, claudin-3c, ZO-1 (spleen only) and ZO-2 (spleen only) in the head kidney, spleen and skin of grass carp. Finally, based on the activities of glutathione peroxidase (GPx) and reactive oxygen species (ROS) content in the head kidney, spleen and skin, the dietary selenium requirements for young grass carp were estimated to be 0.558-0.588 mg/kg diet.
Collapse
Affiliation(s)
- Lin Zheng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Ling Tang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Sheng-Yao Kuang
- Animal Nutrition Institute, Sichuan Academy of Animal Science, Chengdu 610066, China
| | - Yun-Yun Zeng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, China; Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, China; Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Sichuan Agricultural University, Chengdu 611130, China.
| |
Collapse
|
175
|
Cao X, He GZ. Knockdown of CLDN6 inhibits cell proliferation and migration via PI3K/AKT/mTOR signaling pathway in endometrial carcinoma cell line HEC-1-B. Onco Targets Ther 2018; 11:6351-6360. [PMID: 30319275 PMCID: PMC6171518 DOI: 10.2147/ott.s174618] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUD Dysregulation of claudin-6 (CLDN6) expression in cancers has been widely documented. However, no study has reported a complete mechanistic understanding of CLDN6 regulation and function in endometrial carcinoma (EC) progression. In the current study, we aimed to assess the expression and biological functions of CLDN6 in EC. METHODS Firstly, the expression level of CLDN6 in EC was measured based on The Cancer Genome Atlas (TCGA) database. Then, qRT-PCR and western blotting were implemented to detect the expression levels of CLDN6 in 82 pairs of EC tissues and corresponding non-tumor tissues, as well as EC cell line HEC-1B. After knockdown of CLDN6, with the attempt to assess whether CLDN6 reduction had positive effects on the cell proliferation, clone formation, invasion and migration abilities of HLC-1Bs, cell counting kit-8 (CCK-8) assay (24, 48, 72 and 96 hours post-transfection), clone experiment, and invasion and migration assays were conducted. Through western blotting analysis, CLDN6-mediated phosphatidylinositol 3-kinase (PI3K) pathway was evaluated. RESULTS Based on the data of TCGA database, clinical patients and cell line HEC-1B, CLDN6 was up-regulated in EC compared with normal. Univariate as well as multivariate COX analysis indicated that CLDN6 expression can act as an independent prognostic factor for overall survival of EC. Further, knockdown of CLDN6 significantly inhibited HEC-1B cell proliferation, suppressed the colony numbers of HEC-1-B cells, and restrained the invasive and migratory ability of HEC-1-B cells. Importantly, through western blot analysis, we found that inhibition of CLDN6 remarkably decreased p-AKT, p-PI3K, and mTOR expression level in EC HEC-1B cell line. CONCLUSION Our data underscore the significance of CLDN6 in EC progression, and CLDN6 is a new candidate oncogene in EC. Our findings propose that targeting CLDN6 might offer future clinical utility in EC.
Collapse
Affiliation(s)
- Xia Cao
- Department of Gynaecology, Danyang People's Hospital of Jiangsu Province, Danyang 212300, Jiangsu Province, China,
| | - Guo-Zhao He
- Department of Gynaecology, Danyang People's Hospital of Jiangsu Province, Danyang 212300, Jiangsu Province, China,
| |
Collapse
|
176
|
Sapich S, Hittinger M, Hendrix-Jastrzebski R, Repnik U, Griffiths G, May T, Wirth D, Bals R, Schneider-Daum N, Lehr CM. Murine alveolar epithelial cells and their lentivirus-mediated immortalisation. Altern Lab Anim 2018; 46:73-89. [PMID: 29856645 DOI: 10.1177/026119291804600207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
In this study, we describe the isolation and immortalisation of primary murine alveolar epithelial cells (mAEpC), as well as their epithelial differentiation and barrier properties when grown on Transwell® inserts. Like human alveolar epithelial cells (hAEpC), mAEpC transdifferentiate in vitro from an alveolar type II (ATII) phenotype to an ATI-like phenotype and exhibit features of the air-blood barrier, such as the establishment of a thin monolayer with functional tight junctions (TJs). This is demonstrated by the expression of TJ proteins (ZO-1 and occludin) and the development of high transepithelial electrical resistance (TEER), peaking at 1800Ω ·cm². Transport across the air-blood barrier, for general toxicity assessments or preclinical drug development, is typically studied in mice. The aim of this work was the generation of novel immortalised murine lung cell lines, to help meet Three Rs requirements in experimental testing and research. To achieve this goal, we lentivirally transduced mAEpC of two different mouse strains with a library of 33 proliferation-promoting genes. With this immortalisation approach, we obtained two murine alveolar epithelial lentivirus-immortalised (mAELVi) cell lines. Both showed similar TJ protein localisation, but exhibited less prominent barrier properties (TEERmax ~250Ω·cm²) when compared to their primary counterparts. While mAEpC demonstrated their suitability for use in the assessment of paracellular transport rates, mAELVi cells could potentially replace mice for the prediction of acute inhalation toxicity during early ADMET studies.
Collapse
Affiliation(s)
- Sandra Sapich
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI ), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | | | - Remi Hendrix-Jastrzebski
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI ), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | | | - Dagmar Wirth
- Research Group Model Systems for Infection and Immunity (MSYS), Helmholtz-Centre for Infection Research (HZI), Braunschweig, Germany; Institute of Experimental Haematology, Medical School Hannover, Hannover, Germany
| | - Robert Bals
- Department of Internal Medicine V - Pulmonology, Allergology and Respiratory Critical Care Medicine, Saarland University, Homburg (Saar), Germany
| | - Nicole Schneider-Daum
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI ), Saarbrücken, Germany
| | - Claus-Michael Lehr
- Department of Drug Delivery (DDEL), Helmholtz-Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz-Centre for Infection Research (HZI ), Saarbrücken, Germany; Department of Pharmacy, Saarland University, Saarbrücken, Germany; PharmBioTec GmbH, Saarbrücken, Germany
| |
Collapse
|
177
|
Kim A, Ma JY. Rhaponticin decreases the metastatic and angiogenic abilities of cancer cells via suppression of the HIF‑1α pathway. Int J Oncol 2018; 53:1160-1170. [PMID: 30015877 PMCID: PMC6065401 DOI: 10.3892/ijo.2018.4479] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 06/12/2018] [Indexed: 12/15/2022] Open
Abstract
Rhaponticin (RA; 3′5-dihydroxy-4′-methoxystilbene 3-O-β-D-glucopyranoside) is a component isolated from various medicinal herbs including Rheum undulatum L. RA has been reported to be an effective treatment for allergy, diabetes, thrombosis, liver steatosis, lung fibrosis and colitis. In addition, RA effectively inhibits tumor growth and induces apoptosis; however, the effects of RA, at non-cytotoxic doses, on the metastasis and angiogenesis of malignant cancer cells have, to be the best of our knowledge, not been identified. In the present study, it was identified that RA suppressed the metastatic potential of MDA-MB231 breast cancer cells, including colony formation, migration and invasion. Human umbilical vein endothelial cells (HUVECs) treated with RA exhibited a decreased ability to form tube-like networks and to migrate across a Transwell membrane, when compared with RA-untreated HUVECs. Using the chick chorioallantoic membrane assay, RA treatment significantly suppressed spontaneous and vascular endothelial growth factor (VEGF)-induced angiogenesis. Furthermore, RA inhibited the production of pro-angiogenic factors, including matrix metalloproteinase (MMP)-9, pentraxin-3, interleukin-8, VEGF and placental growth factor under normoxic and hypoxic conditions, and suppressed the phorbol 12-myristate 13-acetate-induced increase in the gelatinolytic MMP-9 activity and MMP-9 expression in HT1080 cells. RA also significantly inhibited the hypoxia-inducible factor (HIF)-1α pathway, leading to decreased HIF-1α accumulation and HIF-1α nuclear expression under hypoxia. These results indicated that RA exhibits potent anti-metastatic and anti-angiogenic activities with no cytotoxicity via suppression of the HIF-1α signaling pathway. Thus, RA may control malignant cancer cells by inhibiting the spread from primary tumors and expansion to distant organs.
Collapse
Affiliation(s)
- Aeyung Kim
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| | - Jin Yeul Ma
- Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine (KIOM), Daegu 701‑300, Republic of Korea
| |
Collapse
|
178
|
Liu M, Yang J, Zhang Y, Zhou Z, Cui X, Zhang L, Fung KM, Zheng W, Allard FD, Yee EU, Ding K, Wu H, Liang Z, Zheng L, Fernandez-Zapico ME, Li YP, Bronze MS, Morris KT, Postier RG, Houchen CW, Yang J, Li M. ZIP4 Promotes Pancreatic Cancer Progression by Repressing ZO-1 and Claudin-1 through a ZEB1-Dependent Transcriptional Mechanism. Clin Cancer Res 2018; 24:3186-3196. [PMID: 29615456 PMCID: PMC7006048 DOI: 10.1158/1078-0432.ccr-18-0263] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/08/2018] [Accepted: 03/22/2018] [Indexed: 12/20/2022]
Abstract
Purpose: ZIP4 is overexpressed in human pancreatic cancer and promotes tumor growth. However, little is known about the role of ZIP4 in advanced stages of this dismal neoplasm. Our goal is to study the underlying mechanism and define a novel signaling pathway controlled by ZIP4-modulating pancreatic tumor metastasis.Experimental Design: The expression of ZIP4, ZO-1, claudin-1, and ZEB1 in human pancreatic cancer tissues, genetically engineered mouse model, xenograft tumor model, and pancreatic cancer cell lines were examined, and the correlations between ZIP4 and those markers were also analyzed. Functional analysis of ZO-1, claudin-1, and ZEB1 was investigated in pancreatic cancer cell lines and orthotopic xenografts.Results: Genetic inactivation of ZIP4 inhibited migration and invasion in pancreatic cancer and increased the expression of ZO-1 and claudin-1. Conversely, overexpression of ZIP4 promoted migration and invasion and increased the expression of ZEB1 and downregulation of the aforementioned epithelial genes. ZIP4 downregulation of ZO-1 and claudin-1 requires the transcriptional repressor ZEB1. Further analysis demonstrated that ZIP4-mediated repression of ZO-1 and claudin-1 leads to upregulation of their targets FAK and Paxillin. Silencing of ZIP4 caused reduced phosphorylation of FAK and Paxillin, which was rescued by simultaneous blocking of ZO-1 or claudin-1. Clinically, we demonstrated that ZIP4 positively correlates with the levels of ZEB1 and inversely associates with the expression of ZO-1 and claudin-1.Conclusions: These findings suggest a novel pathway activated by ZIP4-controlling pancreatic cancer invasiveness and metastasis, which could serve as a new therapeutic target for this devastating disease. Clin Cancer Res; 24(13); 3186-96. ©2018 AACR.
Collapse
Affiliation(s)
- Mingyang Liu
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jingxuan Yang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Yuqing Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Zhijun Zhou
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Xiaobo Cui
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Liyang Zhang
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kar-Ming Fung
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wei Zheng
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Felicia D Allard
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Eric U Yee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Kai Ding
- Department of Biostatistics and Epidemiology, College of Public Health, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Huanwen Wu
- Department of Pathology, Peking Union Hospital, Peking Union Medical College, Beijing, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Hospital, Peking Union Medical College, Beijing, China
| | - Lei Zheng
- The Sidney Kimmel Comprehensive Cancer Center and Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin E Fernandez-Zapico
- Department of Oncology, Mayo Clinic, Rochester, Minnesota
- Schulze Center for Novel Therapeutics, Mayo Clinic, Rochester, Minnesota
| | - Yi-Ping Li
- Department of Integrative Biology and Pharmacology, the University of Texas Medical School at Houston, Houston, Texas
| | - Michael S Bronze
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Katherine T Morris
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Russell G Postier
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Courtney W Houchen
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Jing Yang
- Department of Pharmacology and Pediatrics, University of California at San Diego, La Jolla, California
| | - Min Li
- Department of Medicine, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma.
- Department of Surgery, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
179
|
Mashaly AH, Anwar R, Ebrahim MA, Eissa LA, El Shishtawy MM. Diagnostic and Prognostic Value of Talin-1 and Midkine as Tumor Markers in Hepatocellular Carcinoma in Egyptian Patients. Asian Pac J Cancer Prev 2018; 19:1503-1508. [PMID: 29936723 PMCID: PMC6103586 DOI: 10.22034/apjcp.2018.19.6.1503] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Hepatocellular carcinoma (HCC) is a main cause of cancer death all over the world. Treatment and outcome of HCC based on its early diagnosis. This study was conducted to estimate the role of talin-1 and midkine in combination with total antioxidant capacity (TAC) as tumor markers in HCC patients. Methods: Serum levels of talin-1 and midkine were measured in 90 Egyptian subjects including 44 patients with HCC, 31patients with cirrhosis and 15 healthy controls using enzyme-linked immunosorbent assay (ELISA) technique. While a colorimetric method was used for measurement of TAC. Results: Serum talin-1 in HCC patients was significantly lower than that in patients with cirrhosis (P<0.001) and normal control (P<0.001). In addition, increased invasion and metastasis correlated with reduced talin-1 level. Serum midkine in HCC patients was significantly higher compared to cirrhotic patients (P<0.001) and normal control (P<0.001). Midkine at a cut off value of 1683 pg/ml showed a sensitivity of (81.82%) and specificity of (83.87%). While alpha-fetoprotein (AFP) at a cut off value of 200 ng/ml had a sensitivity of (52.27%), while specificity was (96.77%). Midkine was positive in 80.9% of HCC patients with negative AFP. Serum TAC was significantly decreased in HCC patients when compared with control group (P<0.001). Conclusion: Talin-1 may be implicated in the carcinogenesis and metastasis of HCC and can be used as a useful tumor marker for HCC. Midkine may be a potential diagnostic marker for HCC and may be used in addition to AFP to increase the sensitivity of HCC detection.
Collapse
Affiliation(s)
- Aya H Mashaly
- Department of Biochemistry, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt. ,
| | | | | | | | | |
Collapse
|
180
|
Yang L, Sun X, Meng X. Differences in the expression profiles of claudin proteins in human gastric carcinoma compared with non‑neoplastic mucosa. Mol Med Rep 2018; 18:1271-1278. [PMID: 29901188 PMCID: PMC6072183 DOI: 10.3892/mmr.2018.9122] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2017] [Accepted: 03/01/2018] [Indexed: 01/12/2023] Open
Abstract
Numerous genetic alterations associated with cancer progression have the potential to serve as biomarkers for the early diagnosis of cancer. Numerous studies have suggested that claudin proteins, which are the primary components of tight junction structures, are associated with the regulation of cell polarity and cell differentiation. To investigate the expression profiles of the tight junction proteins claudin-2, −5, −7 and −8 in gastric carcinoma, immunohistochemical analysis, western blotting and reverse transcription-quantitative polymerase chain reaction analysis was used to detect the expression profiles of these claudin proteins in gastric carcinoma tissues and in homologous non-neoplastic mucosal tissues. According to the present study, the expression levels of claudin-7 and claudin-8 were downregulated, while the expression of claudin-5 was upregulated in gastric carcinoma tissues compared with in non-neoplastic mucosal tissues. Additionally, no notable difference was observed between claudin-2 expression in gastric carcinoma tissues and non-neoplastic mucosae. Correlations between claudin-7 and −8 expression and lymphatic metastasis in gastric carcinoma tissues were additionally reported. In summary, the present study revealed the distinct expression profiles of claudin-5, −7 and −8 in non-neoplastic mucosal tissues and gastric carcinoma tissues. Furthermore, the expression of these claudin proteins was highly associated with metastatic progression and prognosis in patients with gastric carcinoma, and had predictive value for the metastasis and survival of patients with gastric carcinoma.
Collapse
Affiliation(s)
- Luoluo Yang
- Department of Gastroenterology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xun Sun
- Department of Pathology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiangwei Meng
- Department of Gastroenterology, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
181
|
Sabry D, Mostafa A, Hassouna A. Breast carcinoma is a multifactorial disease involving FOXN3, SINA3 and NEAT through repression of GATA3 and TJP. J Thorac Dis 2018; 10:1167-1171. [PMID: 29707264 DOI: 10.21037/jtd.2018.02.65] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Dina Sabry
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Abeer Mostafa
- Medical Biochemistry and Molecular Biology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Amira Hassouna
- School of Interprofessional Health Studies, Faculty of Health and Environmental Sciences, AUT University, Auckland, New Zealand
| |
Collapse
|
182
|
Salehi B, Zucca P, Sharifi-Rad M, Pezzani R, Rajabi S, Setzer WN, Varoni EM, Iriti M, Kobarfard F, Sharifi-Rad J. Phytotherapeutics in cancer invasion and metastasis. Phytother Res 2018; 32:1425-1449. [DOI: 10.1002/ptr.6087] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/11/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Affiliation(s)
- Bahare Salehi
- Medical Ethics and Law Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Paolo Zucca
- Department of Biomedical Sciences; University of Cagliari; Cagliari Italy
| | - Mehdi Sharifi-Rad
- Department of Medical Parasitology; Zabol University of Medical Sciences; Zabol 61663-335 Iran
| | - Raffaele Pezzani
- OU Endocrinology, Dept. Medicine (DIMED); University of Padova; via Ospedale 105 Padova 35128 Italy
- AIROB, Associazione Italiana per la Ricerca Oncologica di Base; Padova Italy
| | - Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - William N. Setzer
- Department of Chemistry; University of Alabama in Huntsville; Huntsville AL 35899 USA
| | - Elena Maria Varoni
- Department of Biomedical, Surgical and Dental Sciences; Milan State University; Milan Italy
| | - Marcello Iriti
- Department of Agricultural and Environmental Sciences; Milan State University; Milan Italy
| | - Farzad Kobarfard
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Medicinal Chemistry, School of Pharmacy; Shahid Beheshti University of Medical Sciences; Tehran Iran
| | - Javad Sharifi-Rad
- Phytochemistry Research Center; Shahid Beheshti University of Medical Sciences; Tehran Iran
- Department of Chemistry, Richardson College for the Environmental Science Complex; The University of Winnipeg; Winnipeg MB Canada
| |
Collapse
|
183
|
Funamoto K, Yoshino D, Matsubara K, Zervantonakis IK, Funamoto K, Nakayama M, Masamune J, Kimura Y, Kamm RD. Endothelial monolayer permeability under controlled oxygen tension. Integr Biol (Camb) 2018; 9:529-538. [PMID: 28488717 DOI: 10.1039/c7ib00068e] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Endothelial permeability has been extensively investigated in the context of pathologies such as cancer and also in studies of drug delivery from the circulation. Hypoxia is a critical regulator of endothelial cell (EC) behavior and affects the barrier function of endothelial linings, yet its role has been little studied. This paper reveals the effect of hypoxia on the permeability of an EC monolayer by cellular experiments using a microfluidic device and a conventional cell culture dish. Human umbilical vein endothelial cells (HUVECs) were seeded into one microfluidic channel, creating an EC monolayer on each vertical surface of a collagen gel confined to a central chamber. Oxygen tension was regulated to produce normoxic (21% O2) or hypoxic (3% O2) conditions by the supply of gas mixtures of oxygen, carbon dioxide, and nitrogen at predefined ratios into channels fabricated into the device. Permeability of the EC monolayer quantified by analyzing diffusion of fluorescence-labelled dextrans into the collagen gel increases with barrier function loss by 6 hour hypoxic exposure, showing 11-fold and 4-fold increases for 70 kDa and 10 kDa dextrans, respectively, on average. Consistent with this, subsequent immunofluorescent staining and separate western blot analysis of HUVECs on a culture dish demonstrate loose cell-cell adhesion resulting from internalization of VE-cadherin under hypoxia. Thus, hypoxic stress increases endothelial permeability by altering cell-cell junction integrity.
Collapse
Affiliation(s)
- Kenichi Funamoto
- Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai 980-8578, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
184
|
Jo Y, Choi N, Kim HN, Choi J. Probing characteristics of cancer cells cultured on engineered platforms simulating different microenvironments. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018. [DOI: 10.1080/21691401.2018.1446970] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yeonho Jo
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Nakwon Choi
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Hong Nam Kim
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Jonghoon Choi
- School of Integrative Engineering, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
185
|
Talib WH. Melatonin and Cancer Hallmarks. Molecules 2018; 23:molecules23030518. [PMID: 29495398 PMCID: PMC6017729 DOI: 10.3390/molecules23030518] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/09/2018] [Accepted: 02/19/2018] [Indexed: 02/07/2023] Open
Abstract
Melatonin is a natural indoleamine produced by the pineal gland that has many functions, including regulation of the circadian rhythm. Many studies have reported the anticancer effect of melatonin against a myriad of cancer types. Cancer hallmarks include sustained proliferation, evading growth suppressors, metastasis, replicative immortality, angiogenesis, resisting cell death, altered cellular energetics, and immune evasion. Melatonin anticancer activity is mediated by interfering with various cancer hallmarks. This review summarizes the anticancer role of melatonin in each cancer hallmark. The studies discussed in this review should serve as a solid foundation for researchers and physicians to support basic and clinical studies on melatonin as a promising anticancer agent.
Collapse
Affiliation(s)
- Wamidh H Talib
- Department of Clinical Pharmacy and Therapeutics, Applied Science Private University, Amman 11931-166, Jordan.
| |
Collapse
|
186
|
Liu Y, Chang K, Fu K, Dong X, Chen X, Liu J, Cui N, Ni J. DNA demethylation of claudin-4 suppresses migration and invasion in laryngeal squamous carcinoma cells. Hum Pathol 2018; 75:71-80. [PMID: 29447921 DOI: 10.1016/j.humpath.2018.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Revised: 01/28/2018] [Accepted: 02/01/2018] [Indexed: 11/24/2022]
Abstract
Claudin-4 (CLDN4) is a member of the claudin transmembrane protein family, which consists of integral membrane proteins that are components of the epithelial cell tight junctions; these tight junctions regulate movement of solutes and ions through the paracellular space. CLDN4 is also a differentiation marker and is believed to indicate an epithelial phenotype. However, the role of CLDN4 in laryngeal squamous carcinoma is still unclear. Here, we showed that CLDN4 expression was down-regulated in laryngeal squamous carcinoma tissues and negatively correlated with methyl-CpG-binding protein 2. In addition, CLDN4 was hypermethylated in HEp-2 cells. DNA demethylation of CLDN4 by 5-aza-2'-deoxycytidine suppressed migration and invasion of HEp-2 cells, whereas CLDN4 silencing restored the migration and invasion of HEp-2 cells. Therefore, CLDN4 plays a key role in laryngeal squamous carcinoma progression.
Collapse
Affiliation(s)
- Yafang Liu
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Kai Chang
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China; Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, Sichuan 610000, China.
| | - Kexin Fu
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Xinjie Dong
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Xiaoshuai Chen
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Jixuan Liu
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| | - Ni Cui
- Department of Gastrointestinal Colorectal and Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin 130031, China.
| | - Jinsong Ni
- Department of Pathology, the First Bethune Hospital of Jilin University, Changchun, Jilin 130021, China.
| |
Collapse
|
187
|
Lachowicz-Scroggins ME, Gordon ED, Wesolowska-Andersen A, Jackson ND, MacLeod HJ, Sharp LZ, Sun M, Seibold MA, Fahy JV. Cadherin-26 (CDH26) regulates airway epithelial cell cytoskeletal structure and polarity. Cell Discov 2018; 4:7. [PMID: 29449961 PMCID: PMC5809386 DOI: 10.1038/s41421-017-0006-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 12/06/2017] [Accepted: 12/15/2017] [Indexed: 01/27/2023] Open
Abstract
Polarization of the airway epithelial cells (AECs) in the airway lumen is critical to the proper function of the mucociliary escalator and maintenance of lung health, but the cellular requirements for polarization of AECs are poorly understood. Using human AECs and cell lines, we demonstrate that cadherin-26 (CDH26) is abundantly expressed in differentiated AECs, localizes to the cell apices near ciliary membranes, and has functional cadherin domains with homotypic binding. We find a unique and non-redundant role for CDH26, previously uncharacterized in AECs, in regulation of cell-cell contact and cell integrity through maintaining cytoskeletal structures. Overexpression of CDH26 in cells with a fibroblastoid phenotype increases contact inhibition and promotes monolayer formation and cortical actin structures. CDH26 expression is also important for localization of planar cell polarity proteins. Knockdown of CDH26 in AECs results in loss of cortical actin and disruption of CRB3 and other proteins associated with apical polarity. Together, our findings uncover previously unrecognized functions for CDH26 in the maintenance of actin cytoskeleton and apicobasal polarity of AECs.
Collapse
Affiliation(s)
- Marrah E. Lachowicz-Scroggins
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143 USA
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Erin D. Gordon
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143 USA
| | | | - Nathan D. Jackson
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206 USA
| | - Hannah J. MacLeod
- Johns Hopkins Bloomberg School of Public Health, W. Harry Feinstone Department of Molecular Microbiology and Immunology, Baltimore, MD 21205 USA
| | - Louis Z. Sharp
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Matthew Sun
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143 USA
| | - Max A. Seibold
- Center for Genes, Environment, and Health, National Jewish Health, Denver, CO 80206 USA
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO 80045 USA
| | - John V. Fahy
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94143 USA
- Division of Pulmonary and Critical Care Medicine, University of California, San Francisco, San Francisco, CA 94143 USA
| |
Collapse
|
188
|
Zhang X, Wang H, Li Q, Liu Y, Zhao P, Li T. Differences in the expression profiles of claudin proteins in human nasopharyngeal carcinoma compared with non-neoplastic mucosa. Diagn Pathol 2018; 13:11. [PMID: 29402318 PMCID: PMC5800018 DOI: 10.1186/s13000-018-0685-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 01/15/2018] [Indexed: 11/18/2022] Open
Abstract
Background Several studies have suggested that claudin proteins, which are the main components of tight junction structures, are related to the regulation of cell polarity and cell differentiation. Method To explore the expression profiles of the tight junction proteins claudin-2, − 5, − 8 and − 9 in nasopharyngeal carcinoma, IHC (immunohistochemical analysis), Western blot and real-time PCR were used to detect the expression profiles of these claudin proteins in nasopharyngeal carcinoma tissues and in non-neoplastic mucosal tissues. Results According to our study, the expression levels of claudin-2 and claudin-5 were reduced, while the expression of claudin-8 was increased in nasopharyngeal carcinoma tissues in comparison with non-neoplastic mucosal tissues. Correlations between claudin-2 and -5 expression and metastatic progression in nasopharyngeal carcinoma patients were also found. Conclusion In summary, our research reveals distinct expression profiles of claudin-2, − 5 and − 8 in non-neoplastic mucosal tissues and nasopharyngeal carcinoma tissues. In addition, the expression of these claudin proteins was highly correlated with metastatic progression and prognosis in patients with nasopharyngeal carcinoma and had predictive value for the metastasis and survival of nasopharyngeal carcinoma patients.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Center for Translational Medicine; Department of Spinal Surgery, Central Hospital of Zibo, Affiliated with Shandong University, Gongqingtuan Road 54Hao, Zibo, Shandong Province, China
| | - Haiming Wang
- Department of General Surgery, People's Hospital of Linzi District, Affiliated with Binzhou Medical College, Shandong Province, China
| | - Qian Li
- Department of Spinal Surgery, Central Hospital of Zibo, Affiliated with Shandong University, Zibo, Shandong Province, China
| | - Yunpeng Liu
- Department of Thoracic Surgery, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Peiqing Zhao
- Center for Translational Medicine; Department of Spinal Surgery, Central Hospital of Zibo, Affiliated with Shandong University, Gongqingtuan Road 54Hao, Zibo, Shandong Province, China
| | - Tao Li
- Center for Translational Medicine; Department of Spinal Surgery, Central Hospital of Zibo, Affiliated with Shandong University, Gongqingtuan Road 54Hao, Zibo, Shandong Province, China.
| |
Collapse
|
189
|
Pearce SC, Al-Jawadi A, Kishida K, Yu S, Hu M, Fritzky LF, Edelblum KL, Gao N, Ferraris RP. Marked differences in tight junction composition and macromolecular permeability among different intestinal cell types. BMC Biol 2018; 16:19. [PMID: 29391007 PMCID: PMC5793346 DOI: 10.1186/s12915-018-0481-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 01/03/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mammalian small intestinal tight junctions (TJ) link epithelial cells to one another and function as a permselective barrier, strictly modulating the passage of ions and macromolecules through the pore and leak pathways, respectively, thereby preventing the absorption of harmful compounds and microbes while allowing regulated transport of nutrients and electrolytes. Small intestinal epithelial permeability is ascribed primarily to the properties of TJs between adjoining enterocytes (ENTs), because there is almost no information on TJ composition and the paracellular permeability of nonenterocyte cell types that constitute a small but significant fraction of the intestinal epithelia. RESULTS Here we directed murine intestinal crypts to form specialized organoids highly enriched in intestinal stem cells (ISCs), absorptive ENTs, secretory goblet cells, or Paneth cells. The morphological and morphometric characteristics of these cells in organoids were similar to those in vivo. The expression of certain TJ proteins varied with cell type: occludin and tricellulin levels were high in both ISCs and Paneth cells, while claudin-1, -2, and -7 expression was greatest in Paneth cells, ISCs, and ENTs, respectively. In contrast, the distribution of claudin-15, zonula occludens 1 (ZO-1), and E-cadherin was relatively homogeneous. E-cadherin and claudin-7 marked mainly the basolateral membrane, while claudin-2, ZO-1, and occludin resided in the apical membrane. Remarkably, organoids enriched in ENTs or goblet cells were over threefold more permeable to 4 and 10 kDa dextran compared to those containing stem and Paneth cells. The TJ-regulator larazotide prevented the approximately tenfold increases in dextran flux induced by the TJ-disrupter AT1002 into organoids of different cell types, indicating that this ZO toxin nonselectively increases permeability. Forced dedifferentiation of mature ENTs results in the reacquisition of ISC-like characteristics in TJ composition and dextran permeability, suggesting that the post-differentiation properties of TJs are not hardwired. CONCLUSIONS Differentiation of adult intestinal stem cells into mature secretory and absorptive cell types causes marked, but potentially reversible, changes in TJ composition, resulting in enhanced macromolecular permeability of the TJ leak pathway between ENTs and between goblet cells. This work advances our understanding of how cell differentiation affects the paracellular pathway of epithelia.
Collapse
Affiliation(s)
- Sarah C Pearce
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
- Present address: Performance Nutrition Team, Combat Feeding Directorate, US Army, 15 General Greene Ave, Natick, MA, 01760-5018, USA
| | - Arwa Al-Jawadi
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Kunihiro Kishida
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
- Present address: Department of Science and Technology on Food Safety, Kindai University, Wakayama, 649-6493, Japan
| | - Shiyan Yu
- Department of Biological Sciences, Rutgers University, Life Science Center, 225 University Avenue, Newark, NJ, 07102, USA
| | - Madeleine Hu
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Luke F Fritzky
- Advanced Microscopic Imaging Core Facility, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Karen L Edelblum
- Department of Pathology & Laboratory Medicine, Center for Inflammation and Immunity, Rutgers New Jersey Medical School, Newark, NJ, 07103, USA
| | - Nan Gao
- Department of Biological Sciences, Rutgers University, Life Science Center, 225 University Avenue, Newark, NJ, 07102, USA
| | - Ronaldo P Ferraris
- Department of Pharmacology, Physiology and Neurosciences, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA.
| |
Collapse
|
190
|
Abramczyk H, Imiela A. The biochemical, nanomechanical and chemometric signatures of brain cancer. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2018; 188:8-19. [PMID: 28688336 DOI: 10.1016/j.saa.2017.06.037] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 05/09/2023]
Abstract
Raman spectroscopy and imaging combined with AFM topography and mechanical indentation by AFM have been shown to be an effective tool for analysis and discrimination of human brain tumors from normal structures. Raman methods have potential to be applied in clinical practice as they allow for identification of tumor margins during surgery. In this study, we investigate medulloblastoma (grade IV WHO) (n=5) and the tissue from the negative margins used as normal controls. We compare a high grade medulloblastoma (IV grade), and non-tumor samples from human central nervous system (CNS) tissue. Based on the properties of the Raman vibrational spectra and Raman images we provide a real-time feedback that is label-free method to monitor tumor metabolism that reveals reprogramming of biosynthesis of lipids, and proteins. We have found that the high-grade tumors of central nervous system (medulloblastoma) exhibit enhanced level of β-sheet conformation and down-regulated level of α-helix conformation when comparing against normal tissue. We have shown that the ratio of Raman intensities I2930/I2845 at 2930 and 2845cm-1 is a good source of information on the ratio of lipid and protein contents. We have found that the ratio reflects the lipid and protein contents of tumorous brain tissue compared to the non-tumor tissue. Almost all brain tumors have the Raman intensity ratios significantly higher (1.99±0.026) than that found in non-tumor brain tissue, which is 1.456±0.02, and indicates that the relative amount of lipids compared to proteins is significantly higher in the normal brain tissue. Mechanical indentation using AFM on sliced human brain tissues (medulloblastoma, grade IV) revealed that the mechanical properties of this tissue are strongly heterogeneous, between 1.8 and 75.7kPa, and the mean of 27.16kPa. The sensitivity and specificity obtained directly from PLSDA and cross validation gives a sensitivity and specificity of 98.5% and 96% and 96.3% and 92% for cross-validation, respectively. The high sensitivity and specificity demonstrates usefulness for a proper decision for a Raman diagnostic test on biochemical alterations monitored by Raman spectroscopy related to brain cancer development.
Collapse
Affiliation(s)
- Halina Abramczyk
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland.
| | - Anna Imiela
- Lodz University of Technology, Faculty of Chemistry, Institute of Applied Radiation Chemistry, Laboratory of Laser Molecular Spectroscopy, Wroblewskiego 15, 93-590 Lodz, Poland
| |
Collapse
|
191
|
Si M, Jiao X, Li Y, Chen H, He P, Jiang F. The role of cytokines and chemokines in the microenvironment of the blood-brain barrier in leukemia central nervous system metastasis. Cancer Manag Res 2018; 10:305-313. [PMID: 29483784 PMCID: PMC5815469 DOI: 10.2147/cmar.s152419] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM Central nervous system (CNS) metastasis is a major obstacle in the treatment of leukemia, and the underlying mechanisms of leukemia CNS metastasis are not fully understood. The present study is an investigation of the role of the CNS microenvironment in leukemia CNS metastasis. METHODS Analog blood-brain barrier (BBB) was set by coculturing human brain microvascular endothelial cells (HBMVECs) and leukemia cells (U937 and IL-60), as well as HBMVECs and sera from leukemia patients, in vitro. The permeability of the HBMVEC monolayer and the levels of tight junction proteins, cytokines and chemokines (C&Ckines) were measured. RESULTS The permeability of HBMVECs increased when cocultured with leukemia sera. The expression of C&Ckines was significantly upregulated in HBMVECs cocultured with leukemia sera or leukemia cells, compared to the normal sera (P<0.05, respectively). Specifically, significantly higher levels of vascular endothelial growth factor A (VEGF-A) and matrix metalloprotease 9 (MMP-9) were found in HBMVECs and leukemia cells/sera coculturing systems. CONCLUSION Both leukemia cells and the molecules in leukemia sera play an important role in leukemia CNS metastasis. VEGF-A and MMPs may be the main factors resulting in the degradation of the BBB and inducing the CNS migration of leukemia cells.
Collapse
Affiliation(s)
- Mengya Si
- The First Affiliated Hospital of Shantou University Medical College
| | - Xiaoyang Jiao
- Cell Biology and Genetics Department, Shantou University Medical College, Shantou, People’s Republic of China
| | - Yazhen Li
- Cell Biology and Genetics Department, Shantou University Medical College, Shantou, People’s Republic of China
| | - Huanzhu Chen
- Cell Biology and Genetics Department, Shantou University Medical College, Shantou, People’s Republic of China
| | - Ping He
- Cell Biology and Genetics Department, Shantou University Medical College, Shantou, People’s Republic of China
| | - Fang Jiang
- The First Affiliated Hospital of Shantou University Medical College
- Correspondence Fang Jiang, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, Guangdong 515041, People’s Republic of China, Email
| |
Collapse
|
192
|
Shimada H, Satohisa S, Kohno T, Takahashi S, Hatakeyama T, Konno T, Tsujiwaki M, Saito T, Kojima T. The roles of tricellular tight junction protein lipolysis-stimulated lipoprotein receptor in malignancy of human endometrial cancer cells. Oncotarget 2017; 7:27735-52. [PMID: 27036040 PMCID: PMC5053684 DOI: 10.18632/oncotarget.8408] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 03/16/2016] [Indexed: 02/07/2023] Open
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) has been identified as a novel molecular constituent of tricellular contacts that have a barrier function for the cellular sheet. LSR recruits tricellulin (TRIC), which is the first molecular component of tricellular tight junctions. Knockdown of LSR increases cell motility and invasion of certain cancer cells. However, the behavior and the roles of LSR in endometrial cancer remain unknown. In the present study, we investigated the behavior and roles of LSR in normal and endometrial cancer cells in vivo and in vitro. In endometriosis and endometrial cancer, LSR was observed not only in the subapical region but also throughout the lateral region as well as in normal endometrial epithelial cells in the secretory phase, and LSR in the cancer was reduced in correlation with the malignancy. Knockdown of LSR by the siRNA in cells of the endometrial cancer cell line Sawano, induced cell migration, invasion and proliferation, while TRIC relocalized from the tricellular region to the bicellular region at the membrane. In Sawano cells and normal HEEs, a decrease of LSR induced by leptin and an increase of LSR induced by adiponectin and the drugs for type 2 diabetes metformin and berberine were observed via distinct signaling pathways including JAK2/STAT. In Sawano cells, metformin and berberine prevented cell migration and invasion induced by downregulation of LSR by the siRNA and leptin treatment. The dissection of the mechanism in the downregulation of endometrial LSR during obesity is important in developing new diagnostic and therapy for endometrial cancer.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Seiro Satohisa
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Syunta Takahashi
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsubasa Hatakeyama
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Mitsuhiro Tsujiwaki
- Department of Pathology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
193
|
Gallik KL, Treffy RW, Nacke LM, Ahsan K, Rocha M, Green-Saxena A, Saxena A. Neural crest and cancer: Divergent travelers on similar paths. Mech Dev 2017; 148:89-99. [PMID: 28888421 PMCID: PMC5811199 DOI: 10.1016/j.mod.2017.08.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Revised: 08/20/2017] [Accepted: 08/24/2017] [Indexed: 12/29/2022]
Abstract
Neural crest cells are multipotent progenitors that dynamically interpret diverse microenvironments to migrate significant distances as a loosely associated collective and contribute to many tissues in the developing vertebrate embryo. Uncovering details of neural crest migration has helped to inform a general understanding of collective cell migration, including that which occurs during cancer metastasis. Here, we discuss several commonalities and differences of neural crest and cancer cell migration and behavior. First, we focus on some of the molecular pathways required for the initial specification and potency of neural crest cells and the roles of many of these pathways in cancer progression. We also describe epithelial-to-mesenchymal transition, which plays a critical role in initiating both neural crest migration and cancer metastasis. Finally, we evaluate studies that demonstrate myriad forms of cell-cell and cell-environment communication during neural crest and cancer collective migration to highlight the remarkable similarities in their molecular and cell biological regulation.
Collapse
Affiliation(s)
- Kristin L Gallik
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Randall W Treffy
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Lynne M Nacke
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Kamil Ahsan
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| | - Manuel Rocha
- Department of Organismal Biology and Anatomy, University of Chicago, Chicago, IL 60637, USA
| | - Abigail Green-Saxena
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Ankur Saxena
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL 60607, USA.
| |
Collapse
|
194
|
Chuffa LGDA, Reiter RJ, Lupi LA. Melatonin as a promising agent to treat ovarian cancer: molecular mechanisms. Carcinogenesis 2017; 38:945-952. [PMID: 28575150 DOI: 10.1093/carcin/bgx054] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 06/01/2017] [Indexed: 12/15/2022] Open
Abstract
Ovarian cancer (OC) has the highest mortality rate of all gynecological cancers, and most patients develop chemoresistance after first-line treatments. Despite recent advances, the 5-year relative survival is ~45% for all OC subtypes, and invasive epithelial OC has only a 17% survival rate when diagnosed at a late stage. Identification of new efficacious molecules or biomarkers represents important opportunities in the treatment of OC. The pharmacological and physiological properties of melatonin indicate this agent could be useful against OC progression and metastasis. In normal cells, melatonin has potent antioxidant and anti-apoptotic actions. Conversely, melatonin has pro-oxidant as well as anti-proliferative, anti-angiogenic and immunomodulatory properties in many cancer types including hormone-dependent cancers. Although melatonin receptors have been identified in OC cells, the exact mechanism by which melatonin induces anticancer activities remains incompletely understood. Clinical studies have reported negative correlation between aggressiveness of OC and serum levels of melatonin, reinforcing the idea that melatonin may be a critical factor determining OC development. In vitro and in vivo studies suggest melatonin differentially regulates multiple signaling pathways in OC cells. This focused review explores the potential mechanisms of action of melatonin on cultured OC cells and in experimental models of OC in an attempt to clarify how melatonin modulates the signaling pathways involved in cancer cell apoptosis, survival, inflammation, proliferation and metabolic processes. Based on the evidence presented, we feel that melatonin, as an agent that controls cellular signals associated with malignancy, may be beneficial in combination with other therapeutics for OC treatment.
Collapse
Affiliation(s)
- Luiz Gustavo de Almeida Chuffa
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| | - Russel J Reiter
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| | - Luiz Antonio Lupi
- Department of Anatomy, Institute of Biosciences, UNESP - Universidade Estadual Paulista, Botucatu-SP, Brazil and Department of Cellular and Structural Biology, UTHSCSA, San Antonio, TX 78229, USA
| |
Collapse
|
195
|
Tokuhara Y, Morinishi T, Matsunaga T, Sakai M, Sakai T, Ohsaki H, Kadota K, Kushida Y, Haba R, Hirakawa E. Nuclear expression of claudin-3 in human colorectal adenocarcinoma cell lines and tissues. Oncol Lett 2017; 15:99-108. [PMID: 29285188 DOI: 10.3892/ol.2017.7281] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 08/10/2017] [Indexed: 12/21/2022] Open
Abstract
Claudins are members of a large family of transmembrane proteins, which are essential for the formation of tight junctions and have a significant effect on the biological behavior of tumor progression. Previous studies have demonstrated that several claudins show aberrant expression patterns in numerous types of cancer. The present study investigated the expression and localization of claudin-3 and claudin-7 in human colorectal adenocarcinoma cell lines and tissues. The protein expression levels of claudin-3 and claudin-7 were determined using immunocytochemical and immunohistochemical staining. Claudin-3, but not claudin-7, exhibited nuclear localization in the human colorectal adenocarcinoma Caco-2 and SW620 cell lines. Surgically resected colorectal adenocarcinoma tissue specimens were obtained, and the associations between the expression of claudin-3 or claudin-7 and various clinicopathological parameters were analyzed. The membranous expression rates of claudin-3 and claudin-7 were 58.0 and 50.0%, while their nuclear expression rates were 22.0 and 2.0%, respectively. The membranous expression of claudin-3 and claudin-7 was not associated with any clinicopathological factors, whereas the nuclear expression of claudin-3 was associated with histological type and was significantly increased in colorectal mucinous adenocarcinomas compared with that in well- to moderately-differentiated colorectal adenocarcinomas (P<0.01). However, no associations were observed between the nuclear expression of claudin-7 and any clinicopathological parameter. In conclusion, the nuclear expression of claudin-3 in colorectal mucinous adenocarcinoma may be involved in the biological transformation of tumors. The results from the present study indicated that claudin-3 is an important protein associated with histological type and has potential as a prognostic marker. Although the mechanisms underlying the nuclear localization of claudin-3 in tumorigenesis have not yet been elucidated in detail, the present results indicated the potential of claudin-3 as a histopathological biomarker for colorectal adenocarcinomas.
Collapse
Affiliation(s)
- Yasunori Tokuhara
- Laboratory of Pathology, Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Takamatsu, Kagawa 761-0123, Japan.,Department of Medical Technology, Ehime Prefectural University of Health Sciences, Tobe, Ehime 791-2101, Japan
| | - Tatsuya Morinishi
- Laboratory of Pathology, Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Takamatsu, Kagawa 761-0123, Japan
| | - Toru Matsunaga
- Department of Diagnostic Pathology, University Hospital, Faculty of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Manabu Sakai
- Department of Clinical Laboratory, Osaka University Dental Hospital, Osaka 565-0871, Japan
| | - Takayoshi Sakai
- Department of Oral-Facial Disorders, Osaka University Graduate School of Dentistry, Osaka 565-0871, Japan
| | - Hiroyuki Ohsaki
- Laboratory of Pathology, Department of Medical Biophysics, Kobe University Graduate School of Health Sciences, Kobe, Hyogo 654-0142, Japan
| | - Kyuichi Kadota
- Department of Diagnostic Pathology, University Hospital, Faculty of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Yoshio Kushida
- Department of Diagnostic Pathology, University Hospital, Faculty of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Reiji Haba
- Department of Diagnostic Pathology, University Hospital, Faculty of Medicine, Kagawa University, Miki, Kagawa 761-0793, Japan
| | - Eiichiro Hirakawa
- Laboratory of Pathology, Department of Medical Technology, Kagawa Prefectural University of Health Sciences, Takamatsu, Kagawa 761-0123, Japan
| |
Collapse
|
196
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Molecular targets and anticancer potential of sanguinarine-a benzophenanthridine alkaloid. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 34:143-153. [PMID: 28899497 DOI: 10.1016/j.phymed.2017.08.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 07/06/2017] [Accepted: 08/06/2017] [Indexed: 05/03/2023]
Abstract
BACKGROUND Cancer is an enormous global health burden, and should be effectively addressed with better therapeutic strategies. Currently, over 60% of the clinically approved anticancer agents are either directly isolated from natural sources or are modified from natural lead molecules. Sanguinarine (SNG), a quaternary benzophenanthridine alkaloid has gained increasing attention in recent years as a potential anticancer agent. PURPOSE There is a large untapped source of phytochemical-based anticancer agents remaining to be explored. This review article aims to recapitulate different anticancer properties of SNG, and describes some of the molecular targets involved in exerting its effect. It also depicts the pharmacokinetic and toxicological properties of SNG, two parameters important in determining the druggability of a molecule. METHODS Numerous in vivo and in vitro published studies have signified the anticancer properties of SNG. In order to collate and decipher these properties, an extensive literature search was conducted in PubMed, ScienceDirect, and Scopus using keywords followed by the evaluation of the relevant articles where the relevant reports are integrated and analyzed. RESULTS Apart from inducing cell death, SNG inhibits pro-tumorigenic processes such as invasion, angiogenesis, and metastasis in different cancers. Moreover, SNG has been shown to synergistically enhance the sensitivity of several chemotherapeutic agents and is effective against a variety of multi-drug resistant cancers.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Anees Rahman
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Death Signaling Laboratory, Division of Science (Biology), Experimental Research Building, New York University Abu Dhabi, PO Box 129188, Saadiyat Island Campus, Abu Dhabi, UAE.
| |
Collapse
|
197
|
Mendes M, Peláez-García A, López-Lucendo M, Bartolomé RA, Calviño E, Barderas R, Casal JI. Mapping the Spatial Proteome of Metastatic Cells in Colorectal Cancer. Proteomics 2017; 17. [DOI: 10.1002/pmic.201700094] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/28/2017] [Indexed: 12/27/2022]
Affiliation(s)
- Marta Mendes
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Alberto Peláez-García
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - María López-Lucendo
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Rubén A. Bartolomé
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Eva Calviño
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| | - Rodrigo Barderas
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
- Instituto de Salud Carlos III.; Majadahonda Spain
| | - J. Ignacio Casal
- Department of Cellular and Molecular Medicine; Centro de Investigaciones Biológicas (CIB-CSIC); Madrid Spain
| |
Collapse
|
198
|
Mbele M, Hull R, Dlamini Z. African medicinal plants and their derivatives: Current efforts towards potential anti-cancer drugs. Exp Mol Pathol 2017; 103:121-134. [DOI: 10.1016/j.yexmp.2017.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 07/19/2017] [Accepted: 08/07/2017] [Indexed: 12/29/2022]
|
199
|
Shimada H, Satohisa S, Kohno T, Konno T, Takano KI, Takahashi S, Hatakeyama T, Arimoto C, Saito T, Kojima T. Downregulation of lipolysis-stimulated lipoprotein receptor promotes cell invasion via claudin-1-mediated matrix metalloproteinases in human endometrial cancer. Oncol Lett 2017; 14:6776-6782. [PMID: 29151917 DOI: 10.3892/ol.2017.7038] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/05/2017] [Indexed: 12/30/2022] Open
Abstract
Lipolysis-stimulated lipoprotein receptor (LSR) is a novel molecule present at tricellular contacts which recruits tricellulin (TRIC), a molecular component of tricellular tight junctions (tTJs). LSR and TRIC are colocalized with the bicellular tight junction (bTJ) protein claudin (CLDN)-1-based tight junction strands at tricellular corners. Knockdown of LSR in normal epithelial cells affects tTJ formation and the epithelial barrier function. In cancer cells knockdown of LSR has been demonstrated to increase cell invasion. However, the detailed mechanisms of how the downregulation of LSR enhances cell invasion in cancer remain unclear. In the present study, knockdown of LSR by small interfering RNA (siRNA) in Sawano human endometrial adenocarcinoma cells induced cell invasion. In LSR-knockdown Sawano cells, upregulation of CLDN-1 protein, which contributes to the cell invasion via matrix metalloproteinases (MMPs), was observed compared with the control group by western blotting and immunostaining. Knockdown of LSR significantly induced Sp1 transcription factor activity in the CLDN-1 promoter region. In LSR-knockdown Sawano cells, DNA microarray analysis demonstrated that MMP-1, MMP-2 and MMP-10 mRNA levels were increased, and the protein levels of membrane-type 1-MMP, MMP-2, MMP-9 and MMP-10 were shown to be increased on western blots. Knockdown of CLDN-1 with siRNA prevented the upregulation of cell invasion induced by the knockdown of LSR in Sawano cells. On the invasive front of human endometrial carcinoma tissue samples, a decrease in LSR and increase in CLDN-1 protein levels were observed using immunohistochemical methods. In conclusion, the results indicate that the downregulation of LSR promotes cell invasion of human endometrial cancer via CLDN-1 mediation of MMPs. This mechanism is important for studying the association of tTJs with the cellular invasion of cancer.
Collapse
Affiliation(s)
- Hiroshi Shimada
- Department of Obstetrics and Gynecology, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan.,Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Seiro Satohisa
- Department of Obstetrics and Gynecology, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Ken-Ichi Takano
- Department of Otolaryngology, Sapporo Medical University School of Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Syunta Takahashi
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Tsubasa Hatakeyama
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Chihiro Arimoto
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Tsuyoshi Saito
- Department of Obstetrics and Gynecology, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo, Hokkaido 060-8556, Japan
| |
Collapse
|
200
|
Naz A, Obaid A, Awan FM, Ikram A, Ahmad J, Ali A. Petri Net-Based Model of Helicobacter pylori Mediated Disruption of Tight Junction Proteins in Stomach Lining during Gastric Carcinoma. Front Microbiol 2017; 8:1682. [PMID: 28932213 PMCID: PMC5592237 DOI: 10.3389/fmicb.2017.01682] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022] Open
Abstract
Tight junctions help prevent the passage of digestive enzymes and microorganisms through the space between adjacent epithelial cells lining. However, Helicobacter pylori encoded virulence factors negatively regulate these tight junctions and contribute to dysfunction of gastric mucosa. Here, we have predicted the regulation of important tight junction proteins, such as Zonula occludens-1, Claudin-2 and Connexin32 in the presence of pathogenic proteins. Molecular events such as post translational modifications and crosstalk between phosphorylation, O-glycosylation, palmitoylation and methylation are explored which may compromise the integrity of these tight junction proteins. Furthermore, the signaling pathways disrupted by dysregulated kinases, proteins and post-translational modifications are reviewed to design an abstracted computational model showing the situation-dependent dynamic behaviors of these biological processes and entities. A qualitative hybrid Petri Net model is therefore constructed showing the altered host pathways in the presence of virulence factor cytotoxin-associated gene A, leading to the disruption of tight junction proteins. The model is qualitative logic-based, which does not depend on any kinetic parameter and quantitative data and depends on knowledge derived from experiments. The designed model provides insights into the tight junction disruption and disease progression. Model is then verified by the available experimental data, nevertheless formal in vitro experimentation is a promising way to ensure its validation. The major findings propose that H. pylori activated kinases are responsible to trigger specific post translational modifications within tight junction proteins, at specific sites. These modifications may favor alterations in gastric barrier and provide a route to bacterial invasion into host cells.
Collapse
Affiliation(s)
- Anam Naz
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and TechnologyIslamabad, Pakistan
| | - Ayesha Obaid
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and TechnologyIslamabad, Pakistan
| | - Faryal M. Awan
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and TechnologyIslamabad, Pakistan
| | - Aqsa Ikram
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and TechnologyIslamabad, Pakistan
| | - Jamil Ahmad
- Research Center for Modeling & Simulation, National University of Sciences and TechnologyIslamabad, Pakistan
| | - Amjad Ali
- Atta-ur-Rahman School of Applied Biosciences, National University of Sciences and TechnologyIslamabad, Pakistan
| |
Collapse
|