1
|
Hana C, Thaw Dar NN, Galo Venegas M, Vulfovich M. Claudins in Cancer: A Current and Future Therapeutic Target. Int J Mol Sci 2024; 25:4634. [PMID: 38731853 PMCID: PMC11083183 DOI: 10.3390/ijms25094634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/16/2024] [Accepted: 04/18/2024] [Indexed: 05/13/2024] Open
Abstract
Claudins are a family of 27 proteins that have an important role in the formation of tight junctions. They also have an important function in ion exchange, cell mobility, and the epithelial-to-mesenchymal transition, the latter being very important in cancer invasion and metastasis. Therapeutic targeting of claudins has been investigated to improve cancer outcomes. Recent evidence shows improved outcomes when combining monoclonal antibodies against claudin 18.2 with chemotherapy for patients with gastroesophageal junction cancer. Currently, chimeric antigen receptor T-cells targeting claudin 18 are under investigation. In this review, we will discuss the major functions of claudins, their distribution in the normal as well as cancerous tissues, and their effect in cancer metastasis, with a special focus on the therapeutic targeting of claudins to improve cancer outcomes.
Collapse
Affiliation(s)
- Caroline Hana
- Hematology/Oncology Department, Memorial Healthcare System, Pembroke Pines, FL 33028, USA; (N.N.T.D.); (M.G.V.)
| | | | | | | |
Collapse
|
2
|
Gong L, Liu F, Liu J, Wang J. Dietary fiber (oligosaccharide and non-starch polysaccharide) in preventing and treating functional gastrointestinal disorders - Challenges and controversies: A review. Int J Biol Macromol 2024; 258:128835. [PMID: 38128805 DOI: 10.1016/j.ijbiomac.2023.128835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Functional gastrointestinal disorders (FGIDs) are a group of chronic or recurrent gastrointestinal functional diseases, including functional dyspepsia, irritable bowel syndrome, and functional constipation. A lack of safe and reliable treatments for abdominal pain-related FGIDs has prompted interest in new therapies. Evidence has shown that supplementation with dietary fiber may help treat FGIDs. Dietary fibers (DFs) have been demonstrated to have regulatory effects on the gut microbiota, microbiota metabolites, and gastrointestinal movement and have important implications for preventing and treating FGIDs. However, the adverse effects of some DFs, such as fermentable oligosaccharides, on FGIDs are unclear. This review provides an overview of the DFs physiological properties and functional characteristics that influence their use in management of FGIDs, with emphasis on structural modification technology to improve their therapeutic activities. The review highlights that the use of appropriate or novel fibers is a potential therapeutic approach for FGIDs.
Collapse
Affiliation(s)
- Lingxiao Gong
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Feiyue Liu
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Jie Liu
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China
| | - Jing Wang
- Key Laboratory of Geriatric Nutrition and Health (Beijing Technology and Business University), Ministry of Education, Key Laboratory of Special Food Supervision Technology for State Market Regulation, School of Food and Health, Beijing Technology & Business University (BTBU), Beijing 100048, China.
| |
Collapse
|
3
|
Enichen E, Adams RB, Demmig-Adams B. Physical Activity as an Adjunct Treatment for People Living with HIV? Am J Lifestyle Med 2023; 17:502-517. [PMID: 37426740 PMCID: PMC10328202 DOI: 10.1177/15598276221078222] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023] Open
Abstract
This review evaluates physical activity as a candidate for an adjunct treatment, in conjunction with antiretroviral therapy (ART), for people living with HIV (PLWH). Evidence is summarized that chronic, non-resolving inflammation (a principal feature of immune system dysfunction) and a dysfunctional state of the gut environment are key factors in HIV infection that persist despite treatment with ART. In addition, evidence is summarized that regular physical activity may restore normal function of both the immune system and the gut environment and may thereby ameliorate symptoms and non-resolving inflammation-associated comorbidities that burden PLWH. Physicians who care for PLWH could thus consider incorporating physical activity into treatment plans to complement ART. It is also discussed that different types of physical activity can have different effects on the gut environment and immune function, and that future research should establish more specific criteria for the design of exercise regimens tailored to PLWH.
Collapse
Affiliation(s)
- Elizabeth Enichen
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Robert B. Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| | - Barbara Demmig-Adams
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, USA, (EE, BDA); Physical Therapy of Boulder, Boulder, CO, USA, (RBA)
| |
Collapse
|
4
|
Yadav R, Kumar Y, Dahiya D, Bhatia A. Claudins: The Newly Emerging Targets in Breast Cancer. Clin Breast Cancer 2022; 22:737-752. [PMID: 36175290 DOI: 10.1016/j.clbc.2022.09.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2022] [Accepted: 09/04/2022] [Indexed: 01/25/2023]
Abstract
Claudin-low breast cancers are recently described entities showing low expression of certain claudins and cell adhesion molecules. Claudins constitute the backbone of tight junctions (TJs) formed between 2 cells. Their dysregulation plays a vital role in tumorigenesis. First part of the article focuses on the role of claudins in the TJ organization, their structural-functional characteristics, and post-transcriptional and translational modifications. The latter part of the review attempts to summarize existing knowledge regarding the status of claudins in breast cancer. The article also provides an overview of the effect of claudins on tumor progression, metastasis, stemness, chemotherapy resistance, and their crosstalk with relevant signaling pathways in breast cancer. Claudins can act as 2-edged swords in tumors. Some claudins have either tumor-suppressive/ promoting action, while others work as both in a context-dependent manner. Claudins regulate many important events in breast cancer. However, the intricacies involved in their activity are poorly understood. Post-translational modifications in claudins and their impact on TJ integrity, function, and tumor behavior are still unclear. Although their role in adverse events in breast cancer is recognized, their potential to serve as relevant targets for future therapeutics, especially for difficult-to-treat subtypes of the above malignancy, remains to be explored.
Collapse
Affiliation(s)
- Reena Yadav
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Yashwant Kumar
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
5
|
Resveratrol Downregulates miR-155-5p to Block the Malignant Behavior of Gastric Cancer Cells. BIOMED RESEARCH INTERNATIONAL 2022; 2022:6968641. [PMID: 35789645 PMCID: PMC9250436 DOI: 10.1155/2022/6968641] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022]
Abstract
Studies have shown that resveratrol (Res) exerts significant antiproliferative effects in cancer, and regulating the expression of microRNAs (miRNAs) is one the underlying mechanisms of these effects. Overexpression of miR-155-5p leads to oncogenesis. However, it is unclear whether Res exerts antitumor effects by regulating the expression of miR-155-5p, and its specific mechanism in gastric cancer remains unknown. In this study, qRT-PCR was performed to assess the expression of miR-155-5p in gastric cells and clinical tissues, and the MTT assay, plate clone formation test, cell scratch test, Transwell assay, and flow cytometry were performed to investigate the functions of Res on the growth of gastric cancer cells after treatment with miR-155-5p. Western blot analysis was performed to detect the expression of claudin 1, c-Myc, cyclin D1, Bcl-2, and caspase-3 proteins in gastric cancer cell lines after treatment with miR-155-5p and Res. We found that miR-155-5p was overexpressed in gastric cancer cells and clinical tissues, while Res inhibited gastric cancer cell growth by regulating miR-155-5p expression. The results of MTT assay, plate clone formation test, cell scratch test, Transwell test, and flow cytometry showed that miR-155-5p promoted the proliferation, invasion, and metastasis of gastric cancer cell lines and inhibited apoptosis, while Res addition inhibited this effect (
). When miR-155-5p was overexpressed, the expressions of claudin 1, c-Myc, cyclin D1, and Bcl-2 were upregulated and that of caspase-3 was downregulated. Collectively, these results suggest that miR-155-5p may be a therapeutic target in gastric cancer, and Res may be a potential therapeutic agent based on its regulation of miR-155-5p.
Collapse
|
6
|
Siti Sarah CO, Nur Husna SM, Md. Shukri N, Wong KK, Mohd Ashari NS. Zonula occludens-1 expression is reduced in nasal epithelial cells of allergic rhinitis patients. PeerJ 2022; 10:e13314. [PMID: 35480562 PMCID: PMC9037125 DOI: 10.7717/peerj.13314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/31/2022] [Indexed: 01/15/2023] Open
Abstract
Allergic rhinitis (AR) is a common allergic disease characterized by disruption of nasal epithelial barrier. In this study, we investigated the mRNA expression of zonula occludens-1 (ZO-1), ZO-2 and ZO-3 and histone deacetylase 1 (HDAC1) and HDAC2 in AR patients compared to healthy controls. RNA samples were extracted from nasal epithelial cells of house dust mites (HDMs)-sensitized AR patients and healthy controls (n = 28 in each group). The RNAs were reverse transcribed into cDNAs for measurement of ZO-1, ZO-2, ZO-3, HDAC1 and HDAC2 expression levels by quantitative PCR. The mRNA expression of ZO-1 was significantly decreased in AR patients compared to healthy controls (p = 0.010). No significant difference was observed in the expression levels of ZO-2, ZO-3, HDAC1 and HDAC2 in AR patients compared to healthy controls. We found significant associations of higher HDAC2 levels in AR patients with lower frequency of changing bedsheet (p = 0.043) and with AR patients sensitized to Dermatophagoides farinae (p = 0.041). Higher expression of ZO-2 was observed in AR patients who had pets (p = 0.007). In conclusion, our data indicated that ZO-1 expression was lower in AR patients contributing to decreased integrity of nasal epithelial barrier integrity, and HDAC2 may be involved in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Che Othman Siti Sarah
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Siti Muhamad Nur Husna
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Norasnieda Md. Shukri
- Department of Otorhinolaryngology, Head and Neck Surgery, School of Medical Sciences, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Kah Keng Wong
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Noor Suryani Mohd Ashari
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
7
|
Su R, Wu X, Tao L, Wang C. The role of epigenetic modifications in Colorectal Cancer Metastasis. Clin Exp Metastasis 2022; 39:521-539. [PMID: 35429301 PMCID: PMC9338907 DOI: 10.1007/s10585-022-10163-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 03/18/2022] [Indexed: 12/19/2022]
Abstract
Distant metastasis is the major contributor to the high mortality rate of colorectal cancer (CRC). To overcome the poor prognosis caused by distant metastasis, the mechanisms of CRC metastasis should be further explored. Epigenetic events are the main mediators of gene regulation and further affect tumor progression. Recent studies have found that some epigenetic enzymes are often dysregulated or mutated in multiple tumor types, which prompted us to study the roles of these enzymes in CRC metastasis. In this review, we summarized the alteration of enzymes related to various modifications, including histone modification, nonhistone modification, DNA methylation, and RNA methylation, and their epigenetic mechanisms during the progression of CRC metastasis. Existing data suggest that targeting epigenetic enzymes is a promising strategy for the treatment of CRC metastasis.
Collapse
Affiliation(s)
- Riya Su
- Department of pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xinlin Wu
- Department of General Surgery, the Affiliated Hospital of Inner Mongolia Medical University, Huhhot, China
| | - Liang Tao
- Department of pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Changshan Wang
- The State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, School of Life Sciences, Inner Mongolia University, Hohhot, China.
| |
Collapse
|
8
|
Ornelas A, Dowdell AS, Lee JS, Colgan SP. Microbial Metabolite Regulation of Epithelial Cell-Cell Interactions and Barrier Function. Cells 2022; 11:cells11060944. [PMID: 35326394 PMCID: PMC8946845 DOI: 10.3390/cells11060944] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/04/2022] [Accepted: 03/07/2022] [Indexed: 02/04/2023] Open
Abstract
Epithelial cells that line tissues such as the intestine serve as the primary barrier to the outside world. Epithelia provide selective permeability in the presence of a large constellation of microbes, termed the microbiota. Recent studies have revealed that the symbiotic relationship between the healthy host and the microbiota includes the regulation of cell–cell interactions at the level of epithelial tight junctions. The most recent findings have identified multiple microbial-derived metabolites that influence intracellular signaling pathways which elicit activities at the epithelial apical junction complex. Here, we review recent findings that place microbiota-derived metabolites as primary regulators of epithelial cell–cell interactions and ultimately mucosal permeability in health and disease.
Collapse
Affiliation(s)
- Alfredo Ornelas
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave, Mailstop B146, Aurora, CO 80045, USA; (A.O.); (A.S.D.); (J.S.L.)
| | - Alexander S. Dowdell
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave, Mailstop B146, Aurora, CO 80045, USA; (A.O.); (A.S.D.); (J.S.L.)
| | - J. Scott Lee
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave, Mailstop B146, Aurora, CO 80045, USA; (A.O.); (A.S.D.); (J.S.L.)
| | - Sean P. Colgan
- Mucosal Inflammation Program, Department of Medicine, University of Colorado Anschutz Medical Campus, 12700 E. 19th Ave, Mailstop B146, Aurora, CO 80045, USA; (A.O.); (A.S.D.); (J.S.L.)
- Rocky Mountain Regional Veterans Affairs Medical Center, 1700 N. Wheeling St., Aurora, CO 80045, USA
- Correspondence:
| |
Collapse
|
9
|
Gava F, Pignolet J, Déjean S, Mondésert O, Morin R, Agossa J, Ducommun B, Lobjois V. Quantitative Analysis of Cell Aggregation Dynamics Identifies HDAC Inhibitors as Potential Regulators of Cancer Cell Clustering. Cancers (Basel) 2021; 13:cancers13225840. [PMID: 34830995 PMCID: PMC8616495 DOI: 10.3390/cancers13225840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/16/2021] [Accepted: 11/19/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Metastases formation involves the formation, circulation and seeding of cohesive group of tumor cells called circulating tumors cells clusters at distant organs from the primary tumor. These clusters have a much higher metastatic potential than individual circulating tumor cell, it is therefore important to understand the molecular mechanisms involved in their formation. To this aim, in this study, from the analysis of the relationship between in vitro aggregation quantitative characterization of 25 cancer cell lines and their expression data, we identified genes significantly associated with aggregation. Interestingly, we found that these genes were strongly correlated with the transcriptional signature induced by HDAC inhibitors treatment and we finally showed experimentally that two HDAC inhibitors inhibits tumor cells cluster formation in vitro. These results open new therapeutic perspectives to prevent metastasis formation. Abstract Characterization of the molecular mechanisms involved in tumor cell clustering could open the way to new therapeutic strategies. Towards this aim, we used an in vitro quantitative procedure to monitor the anchorage-independent cell aggregation kinetics in a panel of 25 cancer cell lines. The analysis of the relationship between selected aggregation dynamic parameters and the gene expression data for these cell lines from the CCLE database allowed identifying genes with expression significantly associated with aggregation parameter variations. Comparison of these transcripts with the perturbagen signatures from the Connectivity Map resource highlighted that they were strongly correlated with the transcriptional signature of most histone deacetylase (HDAC) inhibitors. Experimental evaluation of two HDAC inhibitors (SAHA and ISOX) showed that they inhibited the initial step of in vitro tumor cell aggregation. This validates our findings and reinforces the potential interest of HDCA inhibitors to prevent metastasis spreading.
Collapse
Affiliation(s)
- Fabien Gava
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
| | - Julie Pignolet
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
| | - Sébastien Déjean
- Institut de Mathématiques de Toulouse (IMT)-UMR5219, Université de Toulouse, CNRS, Université Paul Sabatier, 31062 Toulouse, France;
| | - Odile Mondésert
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
| | - Renaud Morin
- Imactiv-3D SAS, 1 Place Pierre POTIER, 31100 Toulouse, France;
| | - Joseph Agossa
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
- Institut de Mathématiques de Toulouse (IMT)-UMR5219, Université de Toulouse, CNRS, Université Paul Sabatier, 31062 Toulouse, France;
| | - Bernard Ducommun
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
- CHU de Toulouse, 31000 Toulouse, France
| | - Valérie Lobjois
- Institut des Technologies Avancées en Sciences du Vivant (ITAV)-USR3505, Université de Toulouse, CNRS, Université Paul Sabatier, 31100 Toulouse, France; (F.G.); (J.P.); (O.M.); (J.A.); (B.D.)
- Molecular, Cellular & Developmental Biology Unit (MCD)–UMR5577, Center for Integrative Biology (CBI), Université de Toulouse, CNRS, Université Paul Sabatier, 31062 Toulouse, France
- Correspondence:
| |
Collapse
|
10
|
Koshre GR, Shaji F, Mohanan NK, Mohan N, Ali J, Laishram RS. Star-PAP RNA Binding Landscape Reveals Novel Role of Star-PAP in mRNA Metabolism That Requires RBM10-RNA Association. Int J Mol Sci 2021; 22:9980. [PMID: 34576144 PMCID: PMC8469156 DOI: 10.3390/ijms22189980] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/08/2021] [Accepted: 08/19/2021] [Indexed: 11/17/2022] Open
Abstract
Star-PAP is a non-canonical poly(A) polymerase that selects mRNA targets for polyadenylation. Yet, genome-wide direct Star-PAP targets or the mechanism of specific mRNA recognition is still vague. Here, we employ HITS-CLIP to map the cellular Star-PAP binding landscape and the mechanism of global Star-PAP mRNA association. We show a transcriptome-wide association of Star-PAP that is diminished on Star-PAP depletion. Consistent with its role in the 3'-UTR processing, we observed a high association of Star-PAP at the 3'-UTR region. Strikingly, there is an enrichment of Star-PAP at the coding region exons (CDS) in 42% of target mRNAs. We demonstrate that Star-PAP binding de-stabilises these mRNAs indicating a new role of Star-PAP in mRNA metabolism. Comparison with earlier microarray data reveals that while UTR-associated transcripts are down-regulated, CDS-associated mRNAs are largely up-regulated on Star-PAP depletion. Strikingly, the knockdown of a Star-PAP coregulator RBM10 resulted in a global loss of Star-PAP association on target mRNAs. Consistently, RBM10 depletion compromises 3'-end processing of a set of Star-PAP target mRNAs, while regulating stability/turnover of a different set of mRNAs. Our results establish a global profile of Star-PAP mRNA association and a novel role of Star-PAP in the mRNA metabolism that requires RBM10-mRNA association in the cell.
Collapse
Affiliation(s)
- Ganesh R. Koshre
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, India; (G.R.K.); (F.S.); (N.K.M.); (N.M.)
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Feba Shaji
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, India; (G.R.K.); (F.S.); (N.K.M.); (N.M.)
- Regional Centre for Biotechnology, Faridabad 121001, India
| | - Neeraja K. Mohanan
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, India; (G.R.K.); (F.S.); (N.K.M.); (N.M.)
- Manipal Academy of Higher Education, Manipal 576104, India
| | - Nimmy Mohan
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, India; (G.R.K.); (F.S.); (N.K.M.); (N.M.)
| | - Jamshaid Ali
- Bioinformatics Facility, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695585, India;
| | - Rakesh S. Laishram
- Cardiovascular Diseases & Diabetes Biology, Rajiv Gandhi Centre for Biotechnology, Trivandrum 695014, India; (G.R.K.); (F.S.); (N.K.M.); (N.M.)
| |
Collapse
|
11
|
Abstract
Claudins are adhesion molecules located at the tight junctions between epithelial cells. A series of studies have now reported aberrant expression of claudin proteins in the context of neoplastic transformation, suggesting its role in tumorigenesis. However, the precise mechanisms are still not well understood. Studies on expression alterations of claudins have revealed a range of outcomes that reflect the complexity of claudins in terms of spatial localization, tumor type and stage of disease. The diverse and dynamic expression patterns of claudins in cancer are tightly controlled by a wide range of regulatory mechanisms, which are commonly modulated by oncogenic signaling pathways. The present review summarizes the recent knowledge describing the dysregulation of claudin expression in cancer and discusses the intrinsic and extrinsic determinants of the context-specific expression patterns of claudins.
Collapse
|
12
|
Shetty MG, Pai P, Deaver RE, Satyamoorthy K, Babitha KS. Histone deacetylase 2 selective inhibitors: A versatile therapeutic strategy as next generation drug target in cancer therapy. Pharmacol Res 2021; 170:105695. [PMID: 34082029 DOI: 10.1016/j.phrs.2021.105695] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 05/04/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Acetylation and deacetylation of histone and several non-histone proteins are the two important processes amongst the different modes of epigenetic modulation that are involved in regulating cancer initiation and development. Abnormal expression of histone deacetylases (HDACs) is often reported in various types of cancers. Few pan HDAC inhibitors have been approved for use as therapeutic interventions for cancer treatment including vorinostat, belinostat and panobinostat. However, not all the HDAC isoforms are abnormally expressed in certain cancers, such as in the case of, ovarian cancer where overexpression of HDAC1-3, lung cancer where overexpression of HDAC 1 and 3 and gastric cancer where overexpression of HDAC2 is seen. Therefore, pan-inhibition of HDAC is not an efficient way to combat cancer via HDAC inhibition. Hence, isoform-selective HDAC inhibition can be one of the best therapeutic strategies in the treatment of cancer. In this context since aberrant expression of HDAC2 largely contributes to cancer progression by silencing pro-apoptotic protein expressions such as NOXA and APAF1 (caspase 9-activating proteins) and inactivation of tumor suppressor p53, HDAC2 specific inhibitors may help to develop not only the direct targets but also indirect targets that are crucial for tumor development. However, to develop a HDAC2 specific and potent inhibitor, extensive knowledge of its structure and specific functions is essential. The present review updates details on the structural features, physiological functions, and roles of HDAC2 in different types of cancer, emphasizing the challenges and status of the development of HDAC2 selective inhibitors against various types of cancer.
Collapse
Affiliation(s)
| | - Padmini Pai
- Department of Biophysics, Manipal School of Life Sciences, MAHE, Manipal, India
| | - Renita Esther Deaver
- Department of Biotechnology, Manipal School of Life Sciences, MAHE, Manipal, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, MAHE, Manipal, India
| | | |
Collapse
|
13
|
Kakiuchi A, Kakuki T, Ohwada K, Kurose M, Kondoh A, Obata K, Nomura K, Miyata R, Kaneko Y, Konno T, Kohno T, Himi T, Takano KI, Kojima T. HDAC inhibitors suppress the proliferation, migration and invasiveness of human head and neck squamous cell carcinoma cells via p63‑mediated tight junction molecules and p21‑mediated growth arrest. Oncol Rep 2021; 45:46. [PMID: 33649777 PMCID: PMC7934225 DOI: 10.3892/or.2021.7997] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 04/07/2020] [Indexed: 12/27/2022] Open
Abstract
In human head and neck squamous cell carcinoma (HNSCC), the invasion and metastatic properties of cancer cells are promoted by junctional adhesion molecule-A (JAM-A) and claudin-1; these are epithelial tight junction molecules regulated by histone deacetylases (HDACs) and transcription factor p63. HDAC expression is reportedly upregulated in HNSCC, and HDAC inhibitors suppress cancer cell proliferation by initiating proliferative arrest or apoptosis. However, little is known of the anti-cancer mechanisms of HDAC inhibitors in HNSCC. Thus, in the present study, the HNSCC Detroit 562 cell line and primary cultured HNSCC cells were treated with HDAC inhibitors to investigate their effects in HNSCC. Higher expression of p63, HDAC1, JAM-A and claudin-1 was observed in HNSCC tissues compared with the adjacent dysplastic regions. In Detroit 562 cells, treatment with trichostatin A (TSA), an inhibitor of HDAC1 and 6, downregulated the expression of p63, JAM-A and claudin-1, and upregulated that of acetylated tubulin; conversely, p63 knockdown resulted in the downregulation of JAM-A and claudin-1. Collectively, inhibiting HDAC suppressed the migration and invasiveness of cancer cells. In addition, treatment with TSA suppressed cancer cell proliferation via G2/M arrest, as well as upregulating p21 and downregulating cyclin D1 expression. TSA also downregulated the expression of epidermal growth factor receptor (EGFR) and phospho-ERK1/2. p63 knockdown and treatment with an EGFR inhibitor induced G1 arrest and downregulated EGFR and phospho-ERK1/2 levels, respectively. HDAC inhibition also suppressed the migration and invasiveness of primary cultured HNSCC cells. Collectively, the results of the present study indicate that HDAC inhibitors suppress the proliferation, migration and invasiveness of HNSCC by downregulating the p63-mediated tight junction molecules JAM-A and claudin-1, and inducing p63 or p21-mediated growth arrest.
Collapse
Affiliation(s)
- Akito Kakiuchi
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Takuya Kakuki
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Kizuku Ohwada
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Makoto Kurose
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Atsushi Kondoh
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Kazufumi Obata
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Kazuaki Nomura
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Ryo Miyata
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Yakuto Kaneko
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Takumi Konno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Takayuki Kohno
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Tetsuo Himi
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Ken-Ichi Takano
- Department of Otolaryngology, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| | - Takashi Kojima
- Department of Cell Science, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, Sapporo 060‑8556, Japan
| |
Collapse
|
14
|
Role of tight junctions in the epithelial-to-mesenchymal transition of cancer cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183503. [PMID: 33189716 DOI: 10.1016/j.bbamem.2020.183503] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/15/2022]
Abstract
The epithelial-mesenchymal transition (EMT) is an essential step in cancer progression. Epithelial cells possess several types of cell-cell junctions, and tight junctions are known to play important roles in maintaining the epithelial program. EMT is characterized by a loss of epithelial markers, including E-cadherin and tight junction proteins. Somewhat surprisingly, the evidence is accumulating that upregulated expression of tight junction proteins plays an important role in the EMT of cancer cells. Tight junctions have distinct tissue-specific and cancer-specific regulatory mechanisms, enabling them to play different roles in EMT. Tight junctions and related signaling pathways are attractive targets for cancer treatments; signal transduction inhibitors and monoclonal antibodies for tight junction proteins may be used to suppress EMT, invasion, and metastasis. Here we review the role of bicellular and tricellular tight junction proteins during EMT. Further investigation of regulatory mechanisms of tight junctions during EMT in cancer cells will inform the development of biomarkers for predicting prognosis as well as novel therapies.
Collapse
|
15
|
Sanaei M, Kavoosi F. Effect of Zebularine in Comparison to and in Combination with Trichostatin A on CIP/KIP Family (p21Cip1/Waf1/Sdi1, p27Kip1, and p57Kip2), DNMTs (DNMT1, DNMT3a, and DNMT3b), Class I HDACs (HDACs 1, 2, 3) and Class II HDACs (HDACs 4, 5, 6) Gene Expression, Cell Growth Inhibition and Apoptosis Induction in Colon Cancer LS 174T Cell Line. Asian Pac J Cancer Prev 2020; 21:2131-2139. [PMID: 32711442 PMCID: PMC7573409 DOI: 10.31557/apjcp.2020.21.7.2131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Indexed: 12/15/2022] Open
Abstract
Background: A pattern of epigenetic modifications and changes, DNA methylation and histone modification, is central to many human cancers. A variety of tumor suppressor genes (TSGs) have been demonstrated to be silenced because of histone deacetylation and DNA hypermethylation in several cancers. Recent in vitro studies have shown that two known mechanisms of epigenetic alteration consisting of methylation and histone deacetylation seem to be the best candidate mechanisms for inactivation of CIP/KIP family (p21Cip1/Waf1/Sdi1, and p27Kip1) in numerous cancers. Numerous investigations have indicated that DNA demethylating and histone deacetylase inhibitors (HDACIs) can restore the CIP/KIP family gene expression. Previously, we evaluated the effect of trichostatin A (TSA) and 5-aza-2′-deoxycytidine (5-AZA-CdR) on hepatocellular carcinoma (HCC). The present study was designed to investigate the effect of zebularine in comparison to and in combination with trichostatin A on p21Cip1/Waf1/Sdi1, p27Kip1, p57Kip2, DNMT1, DNMT3a and DNMT3b, Class I HDACs (HDACs 1, 2, 3) and Class II HDACs (HDACs 4, 5, 6) gene expression, cell growth inhibition and apoptosis induction in colon cancer LS 174T cell line. Materials and Methods: The colon cancer LS 174T cell line was cultured and treated with zebularine and TSA. To determine cell viability, apoptosis, and the relative expression level of the genes, MTT assay, cell apoptosis assay, and qRT-PCR were done respectively. Results: Both compounds significantly inhibited cell growth, and induced apoptosis. Furthermore, both compounds increased p21Cip1/Waf1/Sdi1, p27Kip1, and p57Kip2 significantly. Additionally, zebularine and TSA decreased DNMTs and HDACs gene expression respectively. Conclusion: The zebularine and trichostatin A can reactivate the CIP/KIP family through inhibition of DNMTs and HDACs genes activity.
Collapse
Affiliation(s)
- Masumeh Sanaei
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| | - Fraidoon Kavoosi
- Research Center for Non-Communicable Diseases, Jahrom University of Medical Sciences, Jahrom, Iran
| |
Collapse
|
16
|
Sudheesh AP, Mohan N, Francis N, Laishram RS, Anderson RA. Star-PAP controlled alternative polyadenylation coupled poly(A) tail length regulates protein expression in hypertrophic heart. Nucleic Acids Res 2020; 47:10771-10787. [PMID: 31598705 PMCID: PMC6847588 DOI: 10.1093/nar/gkz875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 09/08/2019] [Accepted: 10/05/2019] [Indexed: 12/31/2022] Open
Abstract
Alternative polyadenylation (APA)-mediated 3′-untranslated region (UTR) shortening is known to increase protein expression due to the loss of miRNA regulatory sites. Yet, mRNAs with longer 3′-UTR also show enhanced protein expression. Here, we identify a mechanism by which longer transcripts generated by the distal-most APA site leads to increased protein expression compared to the shorter transcripts and the longer transcripts are positioned to regulate heart failure (HF). A Star-PAP target gene, NQO1 has three poly(A) sites (PA-sites) at the terminal exon on the pre-mRNA. Star-PAP selects the distal-most site that results in the expression of the longest isoform. We show that the NQO1 distal-specific mRNA isoform accounts for the majority of cellular NQO1 protein. Star-PAP control of the distal-specific isoform is stimulated by oxidative stress and the toxin dioxin. The longest NQO1 transcript has increased poly(A) tail (PA-tail) length that accounts for the difference in translation potentials of the three NQO1 isoforms. This mechanism is involved in the regulation of cardiac hypertrophy (CH), an antecedent condition to HF where NQO1 downregulation stems from the loss of the distal-specific transcript. The loss of NQO1 during hypertrophy was rescued by ectopic expression of the distal- but not the proximal- or middle-specific NQO1 mRNA isoforms in the presence of Star-PAP expression, and reverses molecular events of hypertrophy in cardiomyocytes.
Collapse
Affiliation(s)
- A P Sudheesh
- Cardiovascular and Diabetes Biology Group, Rajiv Gandhi Centre for Biotechnology, Trivandrum-014, India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Nimmy Mohan
- Cardiovascular and Diabetes Biology Group, Rajiv Gandhi Centre for Biotechnology, Trivandrum-014, India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Nimmy Francis
- Cardiovascular and Diabetes Biology Group, Rajiv Gandhi Centre for Biotechnology, Trivandrum-014, India.,Manipal Academy of Higher Education, Manipal 576104, India
| | - Rakesh S Laishram
- Cardiovascular and Diabetes Biology Group, Rajiv Gandhi Centre for Biotechnology, Trivandrum-014, India
| | - Richard A Anderson
- School of Medicine and Public Health, University of Wisconsin, MD 53726, USA
| |
Collapse
|
17
|
Role of Claudin Proteins in Regulating Cancer Stem Cells and Chemoresistance-Potential Implication in Disease Prognosis and Therapy. Int J Mol Sci 2019; 21:ijms21010053. [PMID: 31861759 PMCID: PMC6982342 DOI: 10.3390/ijms21010053] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 12/11/2019] [Accepted: 12/13/2019] [Indexed: 12/11/2022] Open
Abstract
Claudins are cell–cell adhesion proteins, which are expressed in tight junctions (TJs), the most common apical cell-cell adhesion. Claudin proteins help to regulate defense and barrier functions, as well as differentiation and polarity in epithelial and endothelial cells. A series of studies have now reported dysregulation of claudin proteins in cancers. However, the precise mechanisms are still not well understood. Nonetheless, studies have clearly demonstrated a causal role of multiple claudins in the regulation of epithelial to mesenchymal transition (EMT), a key feature in the acquisition of a cancer stem cell phenotype in cancer cells. In addition, claudin proteins are known to modulate therapy resistance in cancer cells, a feature associated with cancer stem cells. In this review, we have focused primarily on highlighting the causal link between claudins, cancer stem cells, and therapy resistance. We have also contemplated the significance of claudins as novel targets in improving the efficacy of cancer therapy. Overall, this review provides a much-needed understanding of the emerging role of claudin proteins in cancer malignancy and therapeutic management.
Collapse
|
18
|
Torun A, Enayat S, Sheraj I, Tunçer S, Ülgen DH, Banerjee S. Butyrate mediated regulation of RNA binding proteins in the post-transcriptional regulation of inflammatory gene expression. Cell Signal 2019; 64:109410. [DOI: 10.1016/j.cellsig.2019.109410] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/28/2019] [Accepted: 08/31/2019] [Indexed: 12/16/2022]
|
19
|
Zeng H, Umar S, Rust B, Lazarova D, Bordonaro M. Secondary Bile Acids and Short Chain Fatty Acids in the Colon: A Focus on Colonic Microbiome, Cell Proliferation, Inflammation, and Cancer. Int J Mol Sci 2019; 20:ijms20051214. [PMID: 30862015 PMCID: PMC6429521 DOI: 10.3390/ijms20051214] [Citation(s) in RCA: 270] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 02/07/2023] Open
Abstract
Secondary bile acids (BAs) and short chain fatty acids (SCFAs), two major types of bacterial metabolites in the colon, cause opposing effects on colonic inflammation at chronically high physiological levels. Primary BAs play critical roles in cholesterol metabolism, lipid digestion, and host–microbe interaction. Although BAs are reabsorbed via enterohepatic circulation, primary BAs serve as substrates for bacterial biotransformation to secondary BAs in the colon. High-fat diets increase secondary BAs, such as deoxycholic acid (DCA) and lithocholic acid (LCA), which are risk factors for colonic inflammation and cancer. In contrast, increased dietary fiber intake is associated with anti-inflammatory and anticancer effects. These effects may be due to the increased production of the SCFAs acetate, propionate, and butyrate during dietary fiber fermentation in the colon. Elucidation of the molecular events by which secondary BAs and SCFAs regulate colonic cell proliferation and inflammation will lead to a better understanding of the anticancer potential of dietary fiber in the context of high-fat diet-related colon cancer. This article reviews the current knowledge concerning the effects of secondary BAs and SCFAs on the proliferation of colon epithelial cells, inflammation, cancer, and the associated microbiome.
Collapse
Affiliation(s)
- Huawei Zeng
- U. S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA.
| | - Shahid Umar
- Department of Surgery and University of Kansas Cancer Center, Kansas City, KS 66160, USA.
| | - Bret Rust
- U. S. Department of Agriculture, Agricultural Research Service, Grand Forks Human Nutrition Research Center, Grand Forks, ND 58203, USA.
| | - Darina Lazarova
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA.
| | - Michael Bordonaro
- Department of Medical Education, Geisinger Commonwealth School of Medicine, Scranton, PA 18509, USA.
| |
Collapse
|
20
|
Zeisel MB, Dhawan P, Baumert TF. Tight junction proteins in gastrointestinal and liver disease. Gut 2019; 68:547-561. [PMID: 30297438 PMCID: PMC6453741 DOI: 10.1136/gutjnl-2018-316906] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/16/2018] [Accepted: 08/19/2018] [Indexed: 12/11/2022]
Abstract
Over the past two decades a growing body of evidence has demonstrated an important role of tight junction (TJ) proteins in the physiology and disease biology of GI and liver disease. On one side, TJ proteins exert their functional role as integral proteins of TJs in forming barriers in the gut and the liver. Furthermore, TJ proteins can also be expressed outside TJs where they play important functional roles in signalling, trafficking and regulation of gene expression. A hallmark of TJ proteins in disease biology is their functional role in epithelial-to-mesenchymal transition. A causative role of TJ proteins has been established in the pathogenesis of colorectal cancer and gastric cancer. Among the best characterised roles of TJ proteins in liver disease biology is their function as cell entry receptors for HCV-one of the most common causes of hepatocellular carcinoma. At the same time TJ proteins are emerging as targets for novel therapeutic approaches for GI and liver disease. Here we review our current knowledge of the role of TJ proteins in the pathogenesis of GI and liver disease biology and discuss their potential as therapeutic targets.
Collapse
Affiliation(s)
- Mirjam B. Zeisel
- Inserm U1052, CNRS UMR 5286, Cancer Research Center of Lyon (CRCL), Université de Lyon (UCBL), Lyon, France
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE
- Buffet Cancer Center, University of Nebraska Medical Center, Omaha, NE
- VA Nebraska-Western Iowa Health Care System, Omaha, NE
| | - Thomas F. Baumert
- Inserm, U1110, Institut de Recherche sur les Maladies Virales et Hépatiques, Strasbourg, France
- Université de Strasbourg, Strasbourg, France
- Institut Hospitalo-Universitaire, Pôle hépato-digestif, Nouvel Hôpital Civil, Strasbourg, France
| |
Collapse
|
21
|
Legrand N, Dixon DA, Sobolewski C. AU-rich element-binding proteins in colorectal cancer. World J Gastrointest Oncol 2019; 11:71-90. [PMID: 30788036 PMCID: PMC6379757 DOI: 10.4251/wjgo.v11.i2.71] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 12/11/2018] [Accepted: 01/01/2019] [Indexed: 02/05/2023] Open
Abstract
Trans-acting factors controlling mRNA fate are critical for the post-transcriptional regulation of inflammation-related genes, as well as for oncogene and tumor suppressor expression in human cancers. Among them, a group of RNA-binding proteins called “Adenylate-Uridylate-rich elements binding proteins” (AUBPs) control mRNA stability or translation through their binding to AU-rich elements enriched in the 3’UTRs of inflammation- and cancer-associated mRNA transcripts. AUBPs play a central role in the recruitment of target mRNAs into small cytoplasmic foci called Processing-bodies and stress granules (also known as P-body/SG). Alterations in the expression and activities of AUBPs and P-body/SG assembly have been observed to occur with colorectal cancer (CRC) progression, indicating the significant role AUBP-dependent post-transcriptional regulation plays in controlling gene expression during CRC tumorigenesis. Accordingly, these alterations contribute to the pathological expression of many early-response genes involved in prostaglandin biosynthesis and inflammation, along with key oncogenic pathways. In this review, we summarize the current role of these proteins in CRC development. CRC remains a major cause of cancer mortality worldwide and, therefore, targeting these AUBPs to restore efficient post-transcriptional regulation of gene expression may represent an appealing therapeutic strategy.
Collapse
Affiliation(s)
- Noémie Legrand
- Department of Microbiology, Faculty of Medicine, University of Geneva, Geneva CH-1211, Switzerland
| | - Dan A Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, and University of Kansas Cancer Center, Kansas City, KS 66045, United States
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva CH-1211, Switzerland
| |
Collapse
|
22
|
Cherradi S, Martineau P, Gongora C, Del Rio M. Claudin gene expression profiles and clinical value in colorectal tumors classified according to their molecular subtype. Cancer Manag Res 2019; 11:1337-1348. [PMID: 30863148 PMCID: PMC6389001 DOI: 10.2147/cmar.s188192] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Purpose Colorectal cancer (CRC) is a heterogeneous disease that can be classified into distinct molecular subtypes. The aims of this study were 1) to compare claudin (CLDN) gene expression in CRC samples and normal colon mucosa, and then in the different CRC molecular subtypes, and 2) to assess their prognostic value. Patients and methods CLDN expression in CRC samples was analyzed using gene expression data for a cohort of 143 primary CRC samples, and compared in the same CRC samples classified into different molecular subtypes (C1 to C6 according to the Marisa's classification, and CMS1 to CMS4 of the consensus classification). Comparison of CLDN expression in normal and tumor colon samples was also made on a smaller number of samples. Then, the relationship between CLDN expression profiles and overall survival (OS) and progression-free survival was examined. Results Compared with normal mucosa, CLDN1 and CLDN2 were upregulated, whereas CLDN5, 7, 8, and 23 were downregulated in CRC samples. Variations in CLDN expression profiles were observed mainly in the CMS2/C1 and CMS4/C4 subtypes. Overall, expression of CLDN2 or CLDN4 alone had a strong prognostic value that increased when they were associated. In the CMS4/C4 subtypes, lower expressions of CLDN11, CLDN12, and CLDN23 were associated with longer OS. Conversely, in the CMS2 and C1 subtypes, low CLDN23 expression was associated with shorter OS and progression-free survival, suggesting a dual role for CLDN23 as a tumor suppressor/promoter in CRC. CLDN6 and CLDN11 had a prognostic value in the CMS2 and C4 subtypes, respectively. Conclusion This analysis of CLDN gene expression profiles and prognostic value in CRC samples classified according to their molecular subtype shows that CRC heterogeneity must be taken into account when assessing CLDN potential value as prognostic markers or therapeutic targets.
Collapse
Affiliation(s)
- Sara Cherradi
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France,
| | - Pierre Martineau
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France,
| | - Céline Gongora
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France,
| | - Maguy Del Rio
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut régional du Cancer de Montpellier, Montpellier F-34298, France,
| |
Collapse
|
23
|
Bai Y, Chen Y, Chen X, Jiang J, Wang X, Wang L, Wang J, Zhang J, Gao L. Trichostatin A activates FOXO1 and induces autophagy in osteosarcoma. Arch Med Sci 2019; 15:204-213. [PMID: 30697272 PMCID: PMC6348367 DOI: 10.5114/aoms.2018.73860] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 12/25/2017] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Histone deacetylase inhibitors (HDACIs) inhibit human osteosarcoma growth and cause apoptosis. Previously, we reported that HDACIs induce autophagy via the FOXO1 pathway. Whether there is involvement of autophagy in anti-osteosarcoma activity of HDACIs is still unknown. MATERIAL AND METHODS Confocal microscopy was performed to determine the formation of GFP-LC3 puncta. Western blotting was conducted to measure FOXO1, and autophagy-related protein levels. Small interference RNA (siRNA) specific for FOXO1 was transfected into U2OS cells to knock down FOXO1 expression level. Flow cytometry was performed to quantify cell death. RESULTS In this study, we first observed that trichostatin A (TSA) induces autophagy in human osteosarcoma cells. Moreover, we found that TSA treatment inhibits the mammalian target of rapamycin (mTOR) signaling pathway and enhances forkhead box O1 (FOXO1) transcriptional activity, which is responsible for the increased autophagy level, while suppression of FOXO1 function by siRNA knockdown markedly decreases TSA-induced autophagy. CONCLUSIONS We found that inhibition of autophagy, either by autophagy inhibitors or ATG gene knockdown, markedly enhances TSA-caused cell death. Taken together, our studies reveal the function of autophagy in HDACI-caused osteosarcoma cell death and thus support the development of a novel therapeutic strategy by combining HDACIs and autophagy inhibitors in osteosarcoma treatment.
Collapse
Affiliation(s)
- Yunjuan Bai
- Department of Emergency, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yun Chen
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiaochen Chen
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Jiukun Jiang
- Department of Emergency, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiao Wang
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liping Wang
- Department of Emergency, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jigang Wang
- Department of Pharmacology, National University of Singapore, Singapore
| | - Jianbin Zhang
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Liang Gao
- Department of Oncology, Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
24
|
Kim CH. Immune regulation by microbiome metabolites. Immunology 2018; 154:220-229. [PMID: 29569377 PMCID: PMC5980225 DOI: 10.1111/imm.12930] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/18/2018] [Accepted: 03/06/2018] [Indexed: 02/06/2023] Open
Abstract
Commensal microbes and the host immune system have been co-evolved for mutual regulation. Microbes regulate the host immune system, in part, by producing metabolites. A mounting body of evidence indicates that diverse microbial metabolites profoundly regulate the immune system via host receptors and other target molecules. Immune cells express metabolite-specific receptors such as P2X7 , GPR41, GPR43, GPR109A, aryl hydrocarbon receptor precursor (AhR), pregnane X receptor (PXR), farnesoid X receptor (FXR), TGR5 and other molecular targets. Microbial metabolites and their receptors form an extensive array of signals to respond to changes in nutrition, health and immunological status. As a consequence, microbial metabolite signals contribute to nutrient harvest from diet, and regulate host metabolism and the immune system. Importantly, microbial metabolites bidirectionally function to promote both tolerance and immunity to effectively fight infection without developing inflammatory diseases. In pathogenic conditions, adverse effects of microbial metabolites have been observed as well. Key immune-regulatory functions of the metabolites, generated from carbohydrates, proteins and bile acids, are reviewed in this article.
Collapse
Affiliation(s)
- Chang H. Kim
- Department of Pathology and Mary H. Weiser Food Allergy CenterUniversity of Michigan Medical SchoolAnn ArborMIUSA
| |
Collapse
|
25
|
Induction of colon and cervical cancer cell death by cinnamic acid derivatives is mediated through the inhibition of Histone Deacetylases (HDAC). PLoS One 2017; 12:e0186208. [PMID: 29190639 PMCID: PMC5708809 DOI: 10.1371/journal.pone.0186208] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 09/27/2017] [Indexed: 12/16/2022] Open
Abstract
Recent studies from our group and many others have shown the ability of histone deacetylase (HDAC) inhibitors for retarding the growth of carcinomas of cervix, colon and rectum in vitro. A search for naturally occurring HDAC inhibitors continues due to the adverse effects associated with known HDAC inhibitors like SAHA and TSA. Therefore in the current study, naturally occurring cinnamic acids derivatives were screened for HDAC inhibitory effect using in silico docking method which identified cinnamic acids as potential candidates. Cinnamic acids (CA) are naturally occurring phenolic compounds known to exhibit anticancer properties. However, it is not clearly known whether the anticancer properties of CA derivatives are due to the inhibition of oncogenic HDACs, if so how the efficacy varies among various CA derivatives. Hence, the HDAC inhibitory potential of CA derivatives containing increasing number of hydroxylic groups or methoxy moieties was determined using Discovery Studio software and the most potent CA derivatives tested ex vivo (biochemical assay) as well as in vitro (using cell based assay). Among CA derivatives tested, dihydroxy cinnamic acid (DHCA, commonly known as caffeic acid) exhibited better interactions with HDAC2 (compared to other isoforms) in silico and inhibited its activity ex vivo as well as in vitro. Targeted reduction of HDAC activity using DHCA induced death of cancer cells by (a) generating reactive oxygen species, (b) arresting cells in S and G2/M phases; and (c) induction of caspase-3 mediated apoptosis. In conclusion, we demonstrated that DHCA inhibited cancer cell growth by binding to HDAC followed by the induction of apoptosis.
Collapse
|
26
|
van der Beek CM, Dejong CHC, Troost FJ, Masclee AAM, Lenaerts K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr Rev 2017; 75:286-305. [PMID: 28402523 DOI: 10.1093/nutrit/nuw067] [Citation(s) in RCA: 224] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Short-chain fatty acids (SCFAs), mainly acetate, propionate, and butyrate, produced by microbial fermentation of undigested food substances are believed to play a beneficial role in human gut health. Short-chain fatty acids influence colonic health through various mechanisms. In vitro and ex vivo studies show that SCFAs have anti-inflammatory and anticarcinogenic effects, play an important role in maintaining metabolic homeostasis in colonocytes, and protect colonocytes from external harm. Animal studies have found substantial positive effects of SCFAs or dietary fiber on colonic disease, but convincing evidence in humans is lacking. Most human intervention trials have been conducted in the context of inflammatory bowel disease. Only a limited number of those trials are of high quality, showing little or no favorable effect of SCFA treatment over placebo. Opportunities for future research include exploring the use of combination therapies with anti-inflammatory drugs, prebiotics, or probiotics; the use of prodrugs in the setting of carcinogenesis; or the direct application of SCFAs to improve mucosal healing after colonic surgery.
Collapse
Affiliation(s)
- Christina M van der Beek
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Cornelis H C Dejong
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Freddy J Troost
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Ad A M Masclee
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Kaatje Lenaerts
- C.M. van der Beek, C.H.C. Dejong, F.J. Troost, A.A.M. Masclee, and K. Lenaerts are with Top Institute Food and Nutrition, Wageningen, the Netherlands. C.M. van der Beek, C.H.C. Dejong, and K. Lenaerts are with the Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands. C.H.C. Dejong is with the School for Oncology and Developmental Biology (GROW), Maastricht University Medical Center+, Maastricht, the Netherlands. F.J. Troost and A.A.M. Masclee are with the Department of Internal Medicine, Division of Gastroenterology-Hepatology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center+, Maastricht, the Netherlands
| |
Collapse
|
27
|
Byun SK, An TH, Son MJ, Lee DS, Kang HS, Lee EW, Han BS, Kim WK, Bae KH, Oh KJ, Lee SC. HDAC11 Inhibits Myoblast Differentiation through Repression of MyoD-Dependent Transcription. Mol Cells 2017; 40:667-676. [PMID: 28927261 PMCID: PMC5638774 DOI: 10.14348/molcells.2017.0116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 12/12/2022] Open
Abstract
Abnormal differentiation of muscle is closely associated with aging (sarcopenia) and diseases such as cancer and type II diabetes. Thus, understanding the mechanisms that regulate muscle differentiation will be useful in the treatment and prevention of these conditions. Protein lysine acetylation and methylation are major post-translational modification mechanisms that regulate key cellular processes. In this study, to elucidate the relationship between myogenic differentiation and protein lysine acetylation/methylation, we performed a PCR array of enzymes related to protein lysine acetylation/methylation during C2C12 myoblast differentiation. Our results indicated that the expression pattern of HDAC11 was substantially increased during myoblast differentiation. Furthermore, ectopic expression of HDAC11 completely inhibited myoblast differentiation, concomitant with reduced expression of key myogenic transcription factors. However, the catalytically inactive mutant of HDAC11 (H142/143A) did not impede myoblast differentiation. In addition, wild-type HDAC11, but not the inactive HDAC11 mutant, suppressed MyoD-induced promoter activities of MEF2C and MYOG (Myogenin), and reduced histone acetylation near the E-boxes, the MyoD binding site, of the MEF2C and MYOG promoters. Collectively, our results indicate that HDAC11 would suppress myoblast differentiation via regulation of MyoD-dependent transcription. These findings suggest that HDAC11 is a novel critical target for controlling myoblast differentiation.
Collapse
Affiliation(s)
- Sang Kyung Byun
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Tae Hyeon An
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Min Jeong Son
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
| | - Da Som Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
| | - Hyun Sup Kang
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Eun-Woo Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Won Kon Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Kyoung-Jin Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| | - Sang Chul Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141,
Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34141,
Korea
| |
Collapse
|
28
|
Ahmad R, Kumar B, Pan K, Dhawan P, Singh AB. HDAC-4 regulates claudin-2 expression in EGFR-ERK1/2 dependent manner to regulate colonic epithelial cell differentiation. Oncotarget 2017; 8:87718-87736. [PMID: 29152115 PMCID: PMC5675667 DOI: 10.18632/oncotarget.21190] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 08/23/2017] [Indexed: 01/13/2023] Open
Abstract
In normal colon, claudin-2 expression is restricted to the crypt bottom containing the undifferentiated and proliferative colonocytes. Claudin-2 expression is also upregulated in colorectal cancer (CRC) and promotes carcinogenesis. However, cellular mechanism/s regulated by increased claudin-2 expression during the CRC and mechanism/s regulating this increase remain poorly understood. Epigenetic mechanisms help regulate expression of cancer-associated genes and inhibition of Histone Deacetylases (HDACs) induces cell cycle arrest and differentiation. Accordingly, based on a comprehensive in vitro and in vivo analysis we here report that Histone Deacetylases regulate claudin-2 expression in causal association with colonocyte dedifferentiation to promote CRC. Detailed differentiation analyses using colon cancer cells demonstrated inverse association between claudin-2 expression and epithelial differentiation. Genetic manipulation studies revealed the causal role of HDAC-4 in regulating claudin-2 expression during this process. Further analysis identified transcriptional regulation as the underlying mechanism, which was dependent on HDAC-4 dependent modulation of the EGFR-ERK1/2 signaling. Accordingly, colon tumors demonstrated marked upregulation of the HDAC-4/ERK1/2/Claudin-2 signaling. Taken together, we demonstrate a novel role for HDAC-4/EGFR/ERK1/2 signaling in regulating claudin-2 expression to modulate colonocyte differentiation. These findings are of clinical significance and highlight epigenetic regulation as potential mechanism to regulate claudin-2 expression during mucosal pathologies including CRC.
Collapse
Affiliation(s)
- Rizwan Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Balawant Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kaichao Pan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - Amar B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
29
|
Kim YE, Cho N, Cheon S, Kim KK. Bortezomib, a proteasome inhibitor, alleviates atopic dermatitis by increasing claudin 1 protein expression. Biochem Biophys Res Commun 2017; 493:744-750. [PMID: 28859979 DOI: 10.1016/j.bbrc.2017.08.120] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/28/2017] [Indexed: 12/25/2022]
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease. Many studies investigating AD pathogenesis and its therapy have been conducted but none have been successful. One of the causes of AD is dysfunction of tight junctions through reduction of claudin 1 expression in the epidermal barrier of the skin. In the present study, we investigated the role of bortezomib (BTZ) in the restoration of the reduced expression of claudin 1. Immunoblot and immunofluorescence analyses revealed that BTZ increased the protein expression level of claudin 1 in the human keratinocyte cell line HaCaT, thereby forming paracellular barriers. Furthermore, repeated application of BTZ alleviated atopic symptoms on the backs and ears of 2, 4-dinitrochlorobenzene (DNCB)-induced AD mice, and led to the formation of normal tight junctions in the epidermal barrier of DNCB-induced mice skin. Taken together, these results demonstrate that BTZ-induced claudin 1 expression may be a valuable therapeutic approach for AD.
Collapse
Affiliation(s)
- Yong-Eun Kim
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Namjoon Cho
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seonghye Cheon
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Kee K Kim
- Department of Biochemistry, Chungnam National University, Daejeon, 34134, Republic of Korea.
| |
Collapse
|
30
|
Zhu BY, Shang BY, Du Y, Li Y, Li L, Xu XD, Zhen YS. A new HDAC inhibitor cinnamoylphenazine shows antitumor activity in association with intensive macropinocytosis. Oncotarget 2017; 8:14748-14758. [PMID: 28107195 PMCID: PMC5362440 DOI: 10.18632/oncotarget.14714] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 01/08/2017] [Indexed: 02/06/2023] Open
Abstract
Previous studies have shown that intensive macropinocytosis occurs in cancer cells and neutral red (NR) is noted for its capability to enter into the cell massively through a process mimetic to macropinocytosis. In addition, trans-cinnamic acid (tCA) has been found to be an inhibitor of histone deacetylase (HDAC). In the present study, cinnamoylphenazine (CA-PZ) that consists of NR and tCA moieties was synthesized and evaluated. As shown, CA-PZ massively entered into colon carcinoma HT-29 cells and pancreatic carcinoma MIA PaCa-2 cells and this entry was blocked by 5-(N-ethyl-N-isopropyl) amiloride (EIPA, an inhibitor of macropinocytosis), indicating a macropinocytosis-mediated uptake. Furthermore, CA-PZ markedly increased the protein expression levels of acetyl-H3, acetyl-H4 and p21 in HT-29 cells and MIA PaCa-2 cells. CA-PZ significantly inhibited the growth of colon carcinoma HT-29 and pancreatic carcinoma MIA PaCa-2 xenografts. By in vivo imaging, CA-PZ displayed prominent accumulation in the tumor xenografts. The study indicates that the newly synthesized CA-PZ acts as an HDAC inhibitor in association with intensive macropinocytosis-mediated intracellular delivery in cancer cells. The use of neutral red for preparation of chimeric molecules with the attribute of macropinocytosis-mediated intracellular delivery might open an alternative way for development of HDAC inhibitors.
Collapse
Affiliation(s)
- Bing-Yan Zhu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Bo-Yang Shang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yue Du
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Liang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xian-Dong Xu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| |
Collapse
|
31
|
Ahmad R, Kumar B, Chen Z, Chen X, Müller D, Lele SM, Washington MK, Batra SK, Dhawan P, Singh AB. Loss of claudin-3 expression induces IL6/gp130/Stat3 signaling to promote colon cancer malignancy by hyperactivating Wnt/β-catenin signaling. Oncogene 2017; 36:6592-6604. [PMID: 28783170 PMCID: PMC6512312 DOI: 10.1038/onc.2017.259] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 04/28/2017] [Accepted: 06/19/2017] [Indexed: 12/12/2022]
Abstract
The Hyperactivated Wnt/β-catenin signaling acts as a switch to induce EMT and promote colorectal cancer. However, due to its essential role in gut homeostasis, therapeutic targeting of this pathway has proven challenging. Additionally, IL-6/Stat-3 signaling, activated by microbial translocation through the dysregulated mucosal barrier in colon adenomas, facilitates the adenoma to adenocarcinomas transition. However, inter-dependence between these signaling pathways and key mucosal barrier components in regulating colon tumorigenesis and cancer progression remains unclear. In current study, we have discovered, using a comprehensive investigative regimen, a novel and tissue specific role of claudin-3, a tight junction integral protein, in inhibiting colon cancer progression by serving as the common rheostat of Stat-3 and Wnt-signaling activation. Loss of claudin-3 also predicted poor patient survival. These findings however contrasted an upregulated claudin-3 expression in other cancer types and implicated role of the epigenetic regulation. Claudin-3−/− mice revealed dedifferentiated and leaky colonic epithelium, and developed invasive adenocarcinoma when subjected to colon cancer. Wnt-signaling hyperactivation, albeit in GSK-3β independent manner, differentiated colon cancer in claudin-3−/− mice versus WT-mice. Claudin-3 loss also upregulated the gp130/IL6/Stat3 signaling in colonic epithelium potentially assisted by infiltrating immune components. Genetic and pharmacological studies confirmed that claudin-3 loss induces Wnt/β-catenin activation, which is further exacerbated by Stat-3-activation and help promote colon cancer. Overall, these novel findings identify claudin-3 as a therapeutic target for inhibiting overactivation of Wnt-signaling to prevent CRC malignancy.
Collapse
Affiliation(s)
- R Ahmad
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - B Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Z Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - X Chen
- Division of Biostatistics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - D Müller
- Department of Pediatric Nephrology, Charité, and Berlin Institute of Health, Berlin, Germany
| | - S M Lele
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - M K Washington
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - P Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| | - A B Singh
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA.,VA Nebraska-Western Iowa Health Care System, Omaha, NE, USA
| |
Collapse
|
32
|
Histone deacetylase inhibition prevents cell death induced by loss of tricellular tight junction proteins in temperature-sensitive mouse cochlear cells. PLoS One 2017; 12:e0182291. [PMID: 28767685 PMCID: PMC5540400 DOI: 10.1371/journal.pone.0182291] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/14/2017] [Indexed: 01/16/2023] Open
Abstract
Tricellular tight junctions (tTJs) are specialized structures that occur where the corners of three cells meet to seal adjacent intercellular space. The molecular components of tTJs include tricellulin (TRIC) and lipolysis-stimulated lipoprotein receptor (LSR) which recruits TRIC, are required for normal hearing. Although loss of TRIC causes hearing loss with degeneration of cochlear cells, the detailed mechanisms remains unclear. In the present study, by using temperature-sensitive mouse cochlear cells, US/VOT-E36 cell line, we investigated the changes of TRIC and LSR during cochlear cell differentiation and the effects of histone deacetylase (HDAC) inhibitors against cell degeneration induced by loss of TRIC and LSR. During cell differentiation induced by the temperature change, expression of TRIC and LSR were clearly induced. Treatment with metformin enhanced expression TRIC and LSR via AMPK during cell differentiation. Loss of TRIC and LSR by the siRNAs induced cell death in differentiated cells. Treatment with HDAC inhibitors trichostatin A and HDAC6 inhibitor prevented the cell death induced by loss of TRIC and LSR. Collectively, these findings suggest that both tTJ proteins TRIC and LSR have crucial roles for the differentiated cochlear cell survival, and that HDAC inhibitors may be potential therapeutic agents to prevent hearing loss.
Collapse
|
33
|
Rashed HE, Hussein S, Mosaad H, Abdelwahab MM, Abdelhamid MI, Mohamed SY, Mohamed AM, Fayed A. Prognostic significance of the genetic and the immunohistochemical expression of epithelial-mesenchymal-related markers in colon cancer. Cancer Biomark 2017; 20:107-122. [PMID: 28759954 DOI: 10.3233/cbm-170034] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Hayam E. Rashed
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Samia Hussein
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Hala Mosaad
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Mai M. Abdelwahab
- Pathology Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | | | - Salem Y. Mohamed
- Internal Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Abdel Motaleb Mohamed
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| | - Alaa Fayed
- Clinical Oncology and Nuclear Medicine Department, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
34
|
RBFOX3 regulates Claudin-1 expression in human lung tissue via attenuation of proteasomal degradation. Biosci Rep 2017; 37:BSR20160623. [PMID: 28126724 PMCID: PMC5322747 DOI: 10.1042/bsr20160623] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Revised: 01/21/2017] [Accepted: 01/26/2017] [Indexed: 01/30/2023] Open
Abstract
RBFOX3, a nuclear RNA-binding protein, is well known as a regulator of alternative pre-mRNA splicing during neuronal development. However, other functions of RBFOX3 are poorly understood. Here, we investigated the function of RBFOX3 in the cytoplasm with respect to regulation of Claudin-1 expression. In human lung tissue, Claudin-1 is higher in RBFOX3-positive cells than in RBFOX3-negative cells. Immunostaining and mRNA quantification revealed that protein levels, but not mRNA levels, of Claudin-1 are increased by RBFOX3. In addition, cycloheximide treatment of human lung cancer cells revealed that RBFOX3 increases the stability of Claudin-1 through attenuation of its ubiquitination. Our study provides insights into the molecular mechanisms by which RBFOX3 regulates Claudin-1 expression in human lung tissue.
Collapse
|
35
|
Effect of Fusarium-Derived Metabolites on the Barrier Integrity of Differentiated Intestinal Porcine Epithelial Cells (IPEC-J2). Toxins (Basel) 2016; 8:toxins8110345. [PMID: 27869761 PMCID: PMC5127141 DOI: 10.3390/toxins8110345] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 10/20/2016] [Accepted: 11/15/2016] [Indexed: 12/20/2022] Open
Abstract
The human, animal and plant pathogen Fusarium, which contaminates agricultural commodities worldwide, produces numerous secondary metabolites. An example is the thoroughly-investigated deoxynivalenol (DON), which severely impairs gastrointestinal barrier integrity. However, to date, the toxicological profile of other Fusarium-derived metabolites, such as enniatins, beauvericin, moniliformin, apicidin, aurofusarin, rubrofusarin, equisetin and bikaverin, are poorly characterized. Thus we examined their effects—as metabolites alone and as metabolites in combination with DON—on the intestinal barrier function of differentiated intestinal porcine epithelial cells (IPEC-J2) over 72 h. Transepithelial electrical resistance (TEER) was measured at 24-h intervals, followed by evaluation of cell viability using neutral red (NR) assay. Enniatins A, A1, B and B1, apicidin, aurofusarin and beauvericin significantly reduced TEER. Moniliformin, equisetin, bikaverin and rubrofusarin had no effect on TEER. In the case of apicidin, aurofusarin and beauvericin, TEER reductions were further substantiated by the addition of otherwise no-effect DON concentrations. In all cases, viability was unaffected, confirming that TEER reductions were not due to compromised viability. Considering the prevalence of mycotoxin contamination and the diseases associated with intestinal barrier disruption, consumption of contaminated food or feed may have substantial health implications.
Collapse
|
36
|
Osanai M, Takasawa A, Murata M, Sawada N. Claudins in cancer: bench to bedside. Pflugers Arch 2016; 469:55-67. [PMID: 27624415 DOI: 10.1007/s00424-016-1877-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 08/31/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023]
Abstract
The claudin family, in mammals, encoded by at least 27 members of a single ancestral gene, CLDN, is the main constituent as integral membrane proteins of tight junctions. It has been shown that the expression levels of claudins are often decreased or that their expressions are absent in human neoplasias. These findings are consistent with the well-accepted concept that carcinogenesis is accompanied by the disruption or loss of functional tight junctions. In contrast, accumulating data have showed elevated or aberrant expression of claudins in various cancers, indicating specific roles of claudins in tumorigenesis. Importantly, dysregulated claudins play an oncogenic role or conversely have a tumor-suppressive effect depending on target tissues or cell types, and thus, they contribute to tumor development and progression. Although tight junctions are intercellular structures in epithelial cells, specific roles of claudins in cancer are supported by the evidence that TJs are not simple static constituents for establishing cell adhesion structures but are also cell signaling components that have functions in receiving environmental cues and transmitting signals inside cells. Since the expression profile of claudins is associated with patients' outcome and prognosis in several cancer types, an understanding of the expression pattern and subcellular localization of claudins in various pathologies will lead to the establishment of claudins as useful biomarkers for the detection and diagnosis of cancers.
Collapse
Affiliation(s)
- Makoto Osanai
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan.
| | - Akira Takasawa
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Masaki Murata
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| | - Norimasa Sawada
- Department of Pathology, Sapporo Medical University School of Medicine, South-1, West-17, Chuo-ku, Sapporo, 060-8556, Japan
| |
Collapse
|
37
|
Xie C, Wu B, Chen B, Shi Q, Guo J, Fan Z, Huang Y. Histone deacetylase inhibitor sodium butyrate suppresses proliferation and promotes apoptosis in osteosarcoma cells by regulation of the MDM2-p53 signaling. Onco Targets Ther 2016; 9:4005-13. [PMID: 27445491 PMCID: PMC4938147 DOI: 10.2147/ott.s105418] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Histone deacetylase inhibitors have been reported to induce tumor cell growth arrest, differentiation, and apoptosis. This study aimed to investigate the effects of one histone deacetylase inhibitor - sodium butyrate (SB) - on osteosarcoma (OS) cell proliferation and apoptosis and also the molecular mechanisms by which SB exerts regulatory effects on OS cells. U2OS and MG63 cells were treated with SB at various concentrations. Then, cell proliferation and apoptosis were determined by 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide and flow cytometry assays, respectively; the expression of Ki67, Bax, Bcl-2, MDM2, and p53 proteins was determined by using Western blot assay. The results showed that SB suppressed proliferation in a concentration-dependent manner and promoted apoptosis of OS cells. In addition, SB enhanced p53 expression and decreased MDM2 expression, indicating that SB can regulate MDM2-p53 feedback loop. p53 inhibited proliferation and promoted apoptosis, whereas MDM2 promoted proliferation and suppressed apoptosis, which indicated that functional effect of SB on OS cell lines at least in part depended on the MDM2-p53 signaling. We also explored the effect of SB on OS cells in vivo and found that SB suppressed the growth of OS cells with no noticeable effect on activity and body weight of mice in vivo. These findings will offer new clues for OS development and progression and offer SB as a potent targeted agent for OS treatment.
Collapse
Affiliation(s)
- Chuhai Xie
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Boyi Wu
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Binwei Chen
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Qunwei Shi
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianhong Guo
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Ziwen Fan
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Yan Huang
- Department of Orthopedics, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
38
|
Sun BS, Yao YQ, Pei BX, Zhang ZF, Wang CL. Claudin-1 correlates with poor prognosis in lung adenocarcinoma. Thorac Cancer 2016; 7:556-563. [PMID: 27766775 PMCID: PMC5130200 DOI: 10.1111/1759-7714.12368] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 05/01/2016] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND This study was conducted to investigate the clinical significance of claudin-1 (CLDN1) expression in patients with lung adenocarcinoma. METHODS We examined CLDN1 protein expression by immunohistochemistry in a tissue microarray from 258 patients with lung adenocarcinoma. We investigated messenger ribonucleic acid (mRNA) expression in H358 (formerly bronchioloalveolar carcinoma) and lung adenocarcinoma cell lines (A549) by real-time reverse transcriptase-polymerase chain reaction. RESULTS Multivariate analysis showed that prognostic factors for lung adenocarcinoma were histologic type, CLDN1, T stage and N stage. Patients with positive CLDN1 expression had a poorer prognosis than patients with negative CLDN1 expression. CLDN1 expression was correlated with Ras and epidermal growth factor receptor (EGFR) expression. Patients with positive expressions of both CLDN1 and Ras/EGFR had a poorer prognosis than patients with CLDN1 (+) Ras/EGFR(-) or CLDN1 (-) Ras/EGFR(+) and patients with negative expressions of both CLDN1 and Ras/EGFR. CLDN1 mRNA expression was lower in the H358 compared with the lung adenocarcinoma cell line (A549). CONCLUSION The combination of CLDN1 and Ras/EGFR is a valuable independent prognostic predictor for lung adenocarcinoma.
Collapse
Affiliation(s)
- Bing-Sheng Sun
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University, Tianjin, China
| | - Yi-Qun Yao
- Department of Neurosurgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bao-Xiang Pei
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University, Tianjin, China
| | - Zhen-Fa Zhang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University, Tianjin, China
| | - Chang-Li Wang
- Department of Lung Cancer, Tianjin Lung Cancer Center, Tianjin Cancer Institute and Hospital, Key Laboratory of Cancer Prevention and Therapy of Tianjin, Tianjin Medical University, Tianjin, China
| |
Collapse
|
39
|
Vad-Nielsen J, Jakobsen KR, Daugaard TF, Thomsen R, Brügmann A, Sørensen BS, Nielsen AL. Regulatory dissection of the CBX5 and hnRNPA1 bi-directional promoter in human breast cancer cells reveals novel transcript variants differentially associated with HP1α down-regulation in metastatic cells. BMC Cancer 2016; 16:32. [PMID: 26791953 PMCID: PMC4721113 DOI: 10.1186/s12885-016-2059-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 01/10/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The three members of the human heterochromatin protein 1 (HP1) family of proteins, HP1α, HP1β, and HPγ, are involved in chromatin packing and epigenetic gene regulation. HP1α is encoded from the CBX5 gene and is a suppressor of metastasis. CBX5 is down-regulated at the transcriptional and protein level in metastatic compared to non-metastatic breast cancer. CBX5 shares a bi-directional promoter structure with the hnRNPA1 gene. But whereas CBX5 expression is down-regulated in metastatic cells, hnRNAP1 expression is constant. Here, we address the regulation of CBX5 in human breast cancer. METHODS Transient transfection and transposon mediated integration of dual-reporter mini-genes containing the bi-directional hnRNPA1 and CBX5 promoter was performed to investigate transcriptional regulation in breast cancer cell lines. Bioinformatics and functional analysis were performed to characterize transcriptional events specifically regulating CBX5 expression. TSA treatment and Chromatin Immunoprecipitation (ChIP) were performed to investigate the chromatin structure along CBX5 in breast cancer cells. Finally, expression of hnRNPA1 and CBX5 mRNA isoforms were measured by quantitative reverse transcriptase PCR (qRT-PCR) in breast cancer tissue samples. RESULTS We demonstrate that an hnRNPA1 and CBX5 bi-directional core promoter fragment does not comprise intrinsic capacity for specific CBX5 down-regulation in metastatic cells. Characterization of transcriptional events in the 20 kb CBX5 intron 1 revealed existence of several novel CBX5 transcripts. Two of these encode consensus HP1α protein but used autonomous promoters in intron 1 by which HP1α expression could be de-coupled from the bi-directional promoter. In addition, another CBX5 transcriptional isoform, STET, was discovered. This transcript includes CBX5 exon 1 and part of intron 1 sequences but lacks inclusion of HP1α encoding exons. Inverse correlation between STET and HP1α coding CBX5 mRNA expression was observed in breast cancer cell lines and tissue samples from breast cancer patients. CONCLUSION We find that HP1α is down-regulated in a mechanism involving CBX5 promoter downstream sequences and that regulation through alternative polyadenylation and splicing generates a transcript, STET, with potential importance in carcinogenesis.
Collapse
Affiliation(s)
- Johan Vad-Nielsen
- Department of Biomedicine, The Bartholin building, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Kristine Raaby Jakobsen
- Department of Biomedicine, The Bartholin building, Aarhus University, DK-8000, Aarhus C, Denmark.,Department of Clinical-Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Tina Fuglsang Daugaard
- Department of Biomedicine, The Bartholin building, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Rune Thomsen
- Department of Biomedicine, The Bartholin building, Aarhus University, DK-8000, Aarhus C, Denmark
| | - Anja Brügmann
- Department of Pathology, Aalborg University Hospital, Aalborg, Denmark
| | - Boe Sandahl Sørensen
- Department of Clinical-Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anders Lade Nielsen
- Department of Biomedicine, The Bartholin building, Aarhus University, DK-8000, Aarhus C, Denmark.
| |
Collapse
|
40
|
Zheng XT, Xiao XQ, Dai JJ. Sodium butyrate down-regulates tristetraprolin-mediated cyclin B1 expression independent of the formation of processing bodies. Int J Biochem Cell Biol 2015; 69:241-8. [PMID: 26555753 DOI: 10.1016/j.biocel.2015.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 10/10/2015] [Accepted: 11/02/2015] [Indexed: 01/18/2023]
Abstract
Butyrate regulates multiple host cellular events including the cell cycle; however, little is known about the molecular mechanism by which butyrate induces a global down-regulation of the expression of genes associated with the cell cycle. Here, we demonstrate that treating HEK293T cells and the non-small-cell lung cancer cell line A549 with a high concentration of sodium butyrate reduces cyclin B1 expression. The underlying mechanism is related to the destabilization of its mRNA by tristetraprolin, which is up-regulated in response to sodium butyrate. Specifically, the sodium butyrate stimulation reduces the mRNA and protein expression of cyclin B1 and, conversely, upregulates tristetraprolin expression. Importantly, the overexpression of tristetraprolin in HEK293T decreases the mRNA and protein expression of cyclin B1; in contrast, knockdown of tristetraprolin mediated by small interfering RNA increases its expression in response to sodium butyrate treatment for both HEK293T and A549 cells. Furthermore, results from luciferase reporter assays and RNA immunoprecipitation indicate that sodium butyrate accelerates 3' UTR-dependent cyclin B1 decay by enhancing the binding of tristetraprolin to the 3' untranslated region of cyclin B1. Surprisingly, the overexpression of tristetraprolin prevents the formation of processing bodies, and the siRNA-mediated silencing of EDC4 does not restore the sodium butyrate-induced reduction of cyclin B1 expression. Thus, we confirm that NaBu regulates ZFP36-mediated cyclin B1 expression in a manner that is independent of the formation of P-bodies. The above findings disclose a novel mechanism of sodium butyrate-mediated gene expression regulation and might benefit its application in tumor treatment.
Collapse
Affiliation(s)
- Xiang-Tao Zheng
- Department of Vascular Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou Medical University, Wenzhou, People's Republic of China
| | - Xiao-Qiang Xiao
- The Institute of Genomic Medicine, Wenzhou Medical University, Wenzhou, People's Republic of China.
| | | |
Collapse
|
41
|
The effects of a novel aliphatic-chain hydroxamate derivative WMJ-S-001 in HCT116 colorectal cancer cell death. Sci Rep 2015; 5:15900. [PMID: 26510776 PMCID: PMC4625135 DOI: 10.1038/srep15900] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 10/05/2015] [Indexed: 01/03/2023] Open
Abstract
Hydroxamate derivatives have attracted considerable attention due to their broad pharmacological properties and have been extensively investigated. We recently demonstrated that WMJ-S-001, a novel aliphatic hydroxamate derivative, exhibits anti-inflammatory and anti-angiogenic activities. In this study, we explored the underlying mechanisms by which WMJ-S-001 induces HCT116 colorectal cancer cell death. WMJ-S-001 inhibited cell proliferation and induced cell apoptosis in HCT116 cells. These actions were associated with AMP-activated protein kinase (AMPK) and p38 mitogen-activated protein kinase (MAPK) activation, p53 phosphorylation and acetylation, as well as the modulation of p21(cip/Waf1), cyclin D1, survivin and Bax. AMPK-p38MAPK signaling blockade reduced WMJ-S-001-induced p53 phosphorylation. Transfection with AMPK dominant negative mutant (DN) reduced WMJ-S-001's effects on p53 and Sp1 binding to the survivn promoter region. Transfection with HDAC3-Flag or HDAC4-Flag also abrogated WMJ-S-001's enhancing effect on p53 acetylation. WMJ-S-001's actions on p21(cip/Waf1), cyclin D1, survivin, Bax were reduced in p53-null HCT116 cells. Furthermore, WMJ-S-001 was shown to suppress the growth of subcutaneous xenografts of HCT116 cells in vivo. In summary, the death of HCT116 colorectal cancer cells exposed to WMJ-S-001 may involve AMPK-p38MAPK-p53-survivin cascade. These results support the role of WMJ-S-001 as a potential drug candidate and warrant the clinical development in the treatment of cancer.
Collapse
|
42
|
Leech AO, Cruz RGB, Hill ADK, Hopkins AM. Paradigms lost-an emerging role for over-expression of tight junction adhesion proteins in cancer pathogenesis. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:184. [PMID: 26366401 DOI: 10.3978/j.issn.2305-5839.2015.08.01] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 07/23/2015] [Indexed: 12/31/2022]
Abstract
Tight junctions (TJ) are multi-protein complexes located at the apicalmost tip of the lateral membrane in polarised epithelial and endothelial cells. Their principal function is in mediating intercellular adhesion and polarity. Accordingly, it has long been a paradigm that loss of TJ proteins and consequent deficits in cell-cell adhesion are required for tumour cell dissemination in the early stages of the invasive/metastatic cascade. However it is becoming increasingly apparent that TJ proteins play important roles in not just adhesion but also intracellular signalling events, activation of which can contribute to, or even drive, tumour progression and metastasis. In this review, we shall therefore highlight cases wherein the gain of TJ proteins has been associated with signals promoting tumour progression. We will also discuss the potential of overexpressed TJ proteins to act as therapeutic targets in cancer treatment. The overall purpose of this review is not to disprove the fact that loss of TJ-based adhesion contributes to the progression of several cancers, but rather to introduce the growing body of evidence that gain of TJ proteins may have adhesion-independent consequences for promoting progression in other cancers.
Collapse
Affiliation(s)
- Astrid O Leech
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Rodrigo G B Cruz
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Arnold D K Hill
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ann M Hopkins
- Department of Surgery, Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
43
|
Claudins and cancer: Fall of the soldiers entrusted to protect the gate and keep the barrier intact. Semin Cell Dev Biol 2015; 42:58-65. [PMID: 26025580 DOI: 10.1016/j.semcdb.2015.05.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/04/2015] [Accepted: 05/06/2015] [Indexed: 12/11/2022]
Abstract
The role of the tight junctions (TJ) in controlling paracellular traffic of ions and molecules, through the regulation of claudin proteins, is now established. However, it has also become increasingly evident that claudin proteins, as integral components of the TJs, play crucial role in maintaining the cell-cell integrity. In conformity, deregulation of claudin expression and cellular distribution in cancer tissues has been widely documented and correlated with cancer progression and metastasis. However, this correlation is not unidirectional and rather suggests tissue specific regulations. Irrespective, if the widely described correlations between altered claudin expression and cancer initiation/progression could be established, they may serve as important markers for prognostic purposes and potential therapeutic targets. In this review, we summarize data from screening of the cancer tissues, manipulation of claudin expression in cells and animals subjected to cancer models, and how claudins are regulated in cancer. The focus of this article remains analysis of the association between cancer and the claudins and to decipher clinical relevance.
Collapse
|
44
|
Tissue invasion and metastasis: Molecular, biological and clinical perspectives. Semin Cancer Biol 2015; 35 Suppl:S244-S275. [PMID: 25865774 DOI: 10.1016/j.semcancer.2015.03.008] [Citation(s) in RCA: 336] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 03/17/2015] [Accepted: 03/18/2015] [Indexed: 12/12/2022]
Abstract
Cancer is a key health issue across the world, causing substantial patient morbidity and mortality. Patient prognosis is tightly linked with metastatic dissemination of the disease to distant sites, with metastatic diseases accounting for a vast percentage of cancer patient mortality. While advances in this area have been made, the process of cancer metastasis and the factors governing cancer spread and establishment at secondary locations is still poorly understood. The current article summarizes recent progress in this area of research, both in the understanding of the underlying biological processes and in the therapeutic strategies for the management of metastasis. This review lists the disruption of E-cadherin and tight junctions, key signaling pathways, including urokinase type plasminogen activator (uPA), phosphatidylinositol 3-kinase/v-akt murine thymoma viral oncogene (PI3K/AKT), focal adhesion kinase (FAK), β-catenin/zinc finger E-box binding homeobox 1 (ZEB-1) and transforming growth factor beta (TGF-β), together with inactivation of activator protein-1 (AP-1) and suppression of matrix metalloproteinase-9 (MMP-9) activity as key targets and the use of phytochemicals, or natural products, such as those from Agaricus blazei, Albatrellus confluens, Cordyceps militaris, Ganoderma lucidum, Poria cocos and Silybum marianum, together with diet derived fatty acids gamma linolenic acid (GLA) and eicosapentanoic acid (EPA) and inhibitory compounds as useful approaches to target tissue invasion and metastasis as well as other hallmark areas of cancer. Together, these strategies could represent new, inexpensive, low toxicity strategies to aid in the management of cancer metastasis as well as having holistic effects against other cancer hallmarks.
Collapse
|
45
|
Wang N, Chen Y, Yang X, Jiang Y. Selenium-binding protein 1 is associated with the degree of colorectal cancer differentiation and is regulated by histone modification. Oncol Rep 2014; 31:2506-14. [PMID: 24737289 DOI: 10.3892/or.2014.3141] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/24/2014] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to examine the regulation of selenium binding protein 1 (SELENBP1) expression in colorectal cancer (CRC). Samples of cancer tissue and adjacent normal mucosa were collected from 83 CRC patients, and analyzed for SELENBP1 expression by 2D-DIGE, immunoblotting, RT-PCR and immunostaining. Expression levels of SELENBP1, carcinoembryonic antigen (CEA) and alkaline phosphatase (AKP) were determined in cultures of human colon cancer cell lines (SW480, SW620 and HT29) folllowing treatment with i) sodium butyrate (NaB, 2 mM), a differentiation inducer; ii) Trichostatin A (TSA, 0.3 µM), a histone deacetylase inhibitor; or iii) 5'-aza-2'-deoxycytidine (5-Aza-dC, 5 µM), a DNA methylation inhibitor. SELENBP1 expression was found to be downregulated (2.54-fold) in the CRC samples as determined by 2D-DIGE and confirmed by immunoblotting and RT-PCR. SELENBP1 expression was correlated with the degree of differentiation, but not with TNM stage or lymph node metastasis, and was higher in benign polyps (1.97±0.57) than in CRC tissues (0.96±0.59). In the CRC cell lines, NaB treatment led to the upregulation of SELENBP1, CEA and AKP when compared with the untreated cells (2.24- to 4.82-fold). SELENBP1 was also upregulated in cells treated with TSA alone (1.25- to 3.64-fold), or in combination with 5-Aza-dC (1.32- to 4.13-fold). In CRC, the downregulated SELENBP1 expression was reactivated by inducing differentiation. Therefore, SELENBP1 is a potential pharmacological target for individualized CRC treatment.
Collapse
Affiliation(s)
- Ning Wang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yang Chen
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Xinghua Yang
- Department of General Surgery, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Yi Jiang
- Central Experimental Laboratory, The First Hospital, China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
46
|
Huang J, Li J, Qu Y, Zhang J, Zhang L, Chen X, Liu B, Zhu Z. The expression of claudin 1 correlates with β-catenin and is a prognostic factor of poor outcome in gastric cancer. Int J Oncol 2014; 44:1293-301. [PMID: 24535143 DOI: 10.3892/ijo.2014.2298] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/30/2013] [Indexed: 11/06/2022] Open
Abstract
Claudin 1 is one of the tight junction proteins, which are critical in the maintenance of epithelial integrity. Aberrant regulation of CLDN1 and its correlation with β-catenin have been discovered in malignant tumors. The present study aimed to investigate the expression profile and clinical relevance of CLDN1 and β-catenin. The protein levels of CLDN1 and β-catenin were examined using immunohistochemical staining. The characteristics of expression profile and prognostic value were analyzed using Pearson's χ² test and Kaplan-Meier analysis, respectively. β-catenin overexpression and knockdown were used to investigate its role in regulating CLDN1 expression. We showed that CLDN1 was overexpressed in intestinal-type, presence of lymph node metastasis, higher TNM stage in gastric cancer patients and correlated with decreased overall survival. The characteristics of CLDN1 expression were associated with that of β-catenin. CLDN1 and β-catenin showed similar prognostic value in intestinal-type gastric cancers. β-catenin knockdown and overexpression in cell models revealed a positive relation between CLDN1 and β-catenin. Our study demonstrated that CLDN1 is a biomarker for intestinal-type gastric cancer with shorter survival. The expression of CLDN1 was strongly associated with β-catenin in gastric cancer patients and a gastric cancer cell model.
Collapse
Affiliation(s)
- Jie Huang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Jianfang Li
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Ying Qu
- Department of Surgery, Department of Obstetrics and Gynecology, Women's Cancer Institute, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Jianian Zhang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Li Zhang
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Xuehua Chen
- Department of Surgery, Department of Obstetrics and Gynecology, Women's Cancer Institute, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Bingya Liu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| | - Zhenggang Zhu
- Department of Surgery, Shanghai Institute of Digestive Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P.R. China
| |
Collapse
|
47
|
Kral AM, Ozerova N, Close J, Jung J, Chenard M, Fleming J, Haines BB, Harrington P, Maclean J, Miller TA, Secrist P, Wang H, Heidebrecht RW. Divergent Kinetics Differentiate the Mechanism of Action of Two HDAC Inhibitors. Biochemistry 2014; 53:725-34. [DOI: 10.1021/bi400936h] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Astrid M. Kral
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Nicole Ozerova
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Joshua Close
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Joon Jung
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Melissa Chenard
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Judith Fleming
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Brian B. Haines
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Paul Harrington
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - John Maclean
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Thomas A. Miller
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Paul Secrist
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | - Hongmei Wang
- Merck & Co., Inc., 33 Avenue Louis Pasteur, Boston, Massachusetts 02115, United States
| | | |
Collapse
|
48
|
Stache C, Hölsken A, Fahlbusch R, Flitsch J, Schlaffer SM, Buchfelder M, Buslei R. Tight junction protein claudin-1 is differentially expressed in craniopharyngioma subtypes and indicates invasive tumor growth. Neuro Oncol 2013; 16:256-64. [PMID: 24305709 DOI: 10.1093/neuonc/not195] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Claudins are tight junction proteins expressed in epithelial tissues that play important roles in cell polarity and adhesion. Altered distribution of claudin-1(CLDN1) affects cell mobility and tumor invasiveness. Craniopharyngiomas (CPs) represent epithelial tumors of the sellar region, consisting of adamantinomatous (adaCP) and papillary (papCP) variants. Their tendency to infiltrate surrounding brain structures complicates successful surgery. Reliable markers are required to predict tumor behavior and to establish individualized treatment protocols. METHODS We describe the distribution pattern of CLDN1 in a large cohort of 66 adaCPs, 21 papCPs, and 24 Rathke`s cleft cyst (RCC) cases using immunohistochemistry. CLDN1 mRNA levels were analyzed with qRT-PCR in 33 CP samples. The impact on the migration potential was studied in primary adaCP cell cultures (n = 11) treated with small interfering RNA (siRNA) for CLDN1. Furthermore, CLDN1 distribution patterns and expression levels were compared between invasive (n = 16) and noninvasive (n = 17) tumor groups. RESULTS PapCPs and RCCs exhibited a distinct homogenous and membranous expression pattern, whereas CLDN1 immunoreactivity appeared weaker and more heterogeneous in adaCPs. In the latter cases, whirl-like cell clusters showed complete absence of CLDN1. mRNA analysis confirmed reduced CLDN1 levels in adaCPs versus papCPs. Interestingly, invasive tumors exhibited significantly lower CLDN1 expression compared with noninvasive counterparts regardless of CP subtype. Accordingly, siRNA treatment for CLDN1 altered tumor cell migration in vitro. CONCLUSION CLDN1 represents a novel marker in the differential diagnosis of CP variants and RCCs. Low CLDN1 expression levels correlate with an invasive CP growth pattern and may serve as a prognostic marker.
Collapse
Affiliation(s)
- Christina Stache
- Corresponding author: Rolf Buslei, MD, University Hospital Erlangen, Department of Neuropathology, Schwabachanlage 6, 91054 Erlangen, Germany.
| | | | | | | | | | | | | |
Collapse
|
49
|
Hrabeta J, Stiborova M, Adam V, Kizek R, Eckschlager T. Histone deacetylase inhibitors in cancer therapy. A review. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2013; 158:161-9. [PMID: 24263215 DOI: 10.5507/bp.2013.085] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 11/12/2013] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Despite recent success toward discovery of more effective anticancer drugs, chemoresistance remains a major cause of treatment failure. There is emerging evidence that epigenetics plays a key role in the development of the resistance. Epigenetic regulators such as histone acetyltransferases (HATs) and histone deacetylases (HDACs) play an important role in gene expression. The latter are found to be commonly linked with many types of cancers and influence cancer development. Overall, histone acetylation is being investigated as a therapeutic target because of its importance in regulating gene expression. This review summarizes mechanisms of the anticancer effects of histone deacetylase (HDAC) inhibitors and the results of clinical studies. RESULTS Different HDAC inhibitors induce cancer cell death by different mechanisms that include changes in gene expression and alteration of both histone and non-histone proteins. Enhanced histone acetylation in tumors results in modification of expression of genes involved in cell signaling. Inhibition of HDACs causes changed expression in 2-10 % of genes involved in important biological processes. The results of experiments and clinical studies demonstrate that combination of HDAC inhibitors with some anticancer drugs have synergistic or additive effects. CONCLUSIONS Even though many biological effects of HDAC inhibitors have been found, most of the mechanisms of their action remain unclear. In addition, their use in combination with other drugs and the combination regime need to be investigated. The discovery of predictive factors is also necessary. Finally, a key question is whether the pan-HDAC inhibitors or the selective inhibitors will be more efficient for different types of cancers.
Collapse
Affiliation(s)
- Jan Hrabeta
- Department of Pediatric Hematology and Oncology, 2nd Faculty of Medicine, Charles University in Prague and University Hospital Motol, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
50
|
Sharma A, Bhat AA, Krishnan M, Singh AB, Dhawan P. Trichostatin-A modulates claudin-1 mRNA stability through the modulation of Hu antigen R and tristetraprolin in colon cancer cells. Carcinogenesis 2013; 34:2610-21. [PMID: 23880304 PMCID: PMC3810835 DOI: 10.1093/carcin/bgt207] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 04/26/2013] [Accepted: 05/18/2013] [Indexed: 02/06/2023] Open
Abstract
Expression of claudin-1, a tight junction protein, is highly upregulated in colon cancer. We have reported that claudin-1 expression in colon cancer cells is epigenetically regulated as histone deacetylase (HDAC) inhibitors decrease claudin-1 messenger RNA (mRNA) stability and thus expression. In this regard, our data suggested a role of the 3'-untranslated region (UTR) in the regulation of HDAC-dependent regulation of claudin-1 mRNA stability. In the current study, we demonstrate, based on our continued investigation, that the ELAV-like RNA-binding proteins (RBPs), human antigen R (HuR) and tristetraprolin (TTP) associate with the 3'-UTR of claudin-1 mRNA to modulate the latter's stability. Ribonomic and site-directed mutagenesis approaches were used to confirm the binding of HuR and TTP to the 3'-UTR of claudin-1. We further confirmed their roles in the stabilization of claudin-1 mRNA, under conditions of HDAC inhibition. In summary, we report that HuR and TTP are the critical regulators of the posttranscriptional regulation of claudin-1 expression in colon cancer cells. We also demonstrate that inhibition of HDACs by trichostatin treatment decreased the binding of HuR while increasing the binding of TTP to the 3'-UTR of claudin-1. Additionally, we provide data showing transcriptional regulation of claudin-1 expression, through the regulation of transcription factor Sp1. Taken together, we demonstrate epigenetic regulation of claudin-1 expression in colon cancer cells at the transcriptional and posttranscriptional levels.
Collapse
Affiliation(s)
- Ashok Sharma
- Department of Surgery, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Ajaz A. Bhat
- Department of Surgery, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
| | - Moorthy Krishnan
- Department of Surgery, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside CA 92521, USA
| | - Amar B. Singh
- Department of Surgery, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
- Department of Medicine and
| | - Punita Dhawan
- Department of Surgery, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, MCN, B-2211, Nashville, TN 37232, USA
| |
Collapse
|