1
|
Wareing N, Mills TW, Collum S, Wu M, Revercomb L, Girard R, Lyons M, Skaug B, Bi W, Ali MA, Koochak H, Flores AR, Yang Y, Zheng WJ, Swindell WR, Assassi S, Karmouty-Quintana H. Deletion of adipocyte Sine Oculis Homeobox Homolog 1 prevents lipolysis and attenuates skin fibrosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.22.595271. [PMID: 38826482 PMCID: PMC11142148 DOI: 10.1101/2024.05.22.595271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Dermal fibrosis is a cardinal feature of systemic sclerosis (SSc) for which there are limited treatment strategies. This is in part due to our fragmented understanding of how dermal white adipose tissue (DWAT) contributes to skin fibrosis. We identified elevated sine oculis homeobox homolog 1 (SIX1) expression in SSc skin samples from the GENISOS and PRESS cohorts, the expression of which correlated with adipose-associated genes and molecular pathways. SIX1 localization studies identified increased signals in the DWAT area in SSc and in experimental models of skin fibrosis. Global and adipocyte specific Six1 deletion abrogated end-stage fibrotic gene expression and dermal adipocyte shrinkage induced by SQ bleomycin treatment. Further studies revealed a link between elevated SIX1 and increased expression of SERPINE1 and its protein PAI-1 which are known pro-fibrotic mediators. However, SIX1 deletion did not appear to affect cellular trans differentiation. Taken together these results point at SIX1 as a potential target for dermal fibrosis in SSc.
Collapse
Affiliation(s)
- Nancy Wareing
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Tingting W Mills
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Scott Collum
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Minghua Wu
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | | | - Rene Girard
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Marka Lyons
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Brian Skaug
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Weizhen Bi
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
| | - Meer A. Ali
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Haniyeh Koochak
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Anthony R Flores
- Department of Pediatrics, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Yuntao Yang
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - W Jim Zheng
- D Bradley McWilliams School of Biomedical Informatics, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - William R Swindell
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX
| | - Shervin Assassi
- Division of Rheumatology, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| | - Harry Karmouty-Quintana
- Department of Biochemistry and Molecular Biology, McGovern Medical School, University of Texas Health Science Center at Houston (UTHealth Houston), TX, USA
- Divisions of Critical Care, Pulmonary and Sleep Medicine, Department of Internal Medicine, McGovern Medical School, UTHealth Houston, Houston TX, USA
| |
Collapse
|
2
|
Bian Z, Benjamin MM, Bialousow L, Tian Y, Hobbs GA, Karan D, Choo YM, Hamann MT, Wang X. Targeting sine oculis homeoprotein 1 (SIX1): A review of oncogenic roles and potential natural product therapeutics. Heliyon 2024; 10:e33204. [PMID: 39022099 PMCID: PMC11252760 DOI: 10.1016/j.heliyon.2024.e33204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 07/20/2024] Open
Abstract
Sine oculis homeoprotein 1 (SIX1), a prominent representative of the homeodomain transcription factors within the SIX family, has attracted significant interest owing to its role in tumorigenesis, cancer progression, and prognostic assessments. Initially recognized for its pivotal role in embryonic development, SIX1 has emerged as a resurgent factor across a diverse set of mammalian cancers. Over the past two decades, numerous investigations have emphasized SIX1's dual significance as a developmental regulator and central player in oncogenic processes. A mounting body of evidence links SIX1 to the initiation of diverse cancers, encompassing enhanced cellular metabolism and advancement. This review provides an overview of the multifaceted roles of SIX1 in both normal development and oncogenic processes, emphasizing its importance as a possible therapeutic target and prognostic marker. Additionally, this review discusses the natural product agents that inhibit various pro-oncogenic mechanisms associated with SIX1.
Collapse
Affiliation(s)
- Zhiwei Bian
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Menny M. Benjamin
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lucas Bialousow
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Yintai Tian
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - G. Aaron Hobbs
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC, USA
| | - Dev Karan
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yeun-Mun Choo
- Chemistry Department, Faculty of Science, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Mark T. Hamann
- Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Xiaojuan Wang
- School of Pharmacy, Lanzhou University, Lanzhou, 730000, Gansu, China
| |
Collapse
|
3
|
Zhu S, Li W, Zhang H, Yan Y, Mei Q, Wu K. Retinal determination gene networks: from biological functions to therapeutic strategies. Biomark Res 2023; 11:18. [PMID: 36750914 PMCID: PMC9906957 DOI: 10.1186/s40364-023-00459-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/28/2023] [Indexed: 02/09/2023] Open
Abstract
The retinal determinant gene network (RDGN), originally discovered as a critical determinator in Drosophila eye specification, has become an important regulatory network in tumorigenesis and progression, as well as organogenesis. This network is not only associated with malignant biological behaviors of tumors, such as proliferation, and invasion, but also regulates the development of multiple mammalian organs. Three members of this conservative network have been extensively investigated, including DACH, SIX, and EYA. Dysregulated RDGN signaling is associated with the initiation and progression of tumors. In recent years, it has been found that the members of this network can be used as prognostic markers for cancer patients. Moreover, they are considered to be potential therapeutic targets for cancer. Here, we summarize the research progress of RDGN members from biological functions to signaling transduction, especially emphasizing their effects on tumors. Additionally, we discuss the roles of RDGN members in the development of organs and tissue as well as their correlations with the pathogenesis of chronic kidney disease and coronary heart disease. By summarizing the roles of RDGN members in human diseases, we hope to promote future investigations into RDGN and provide potential therapeutic strategies for patients.
Collapse
Affiliation(s)
- Shuangli Zhu
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Wanling Li
- grid.412793.a0000 0004 1799 5032Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China ,grid.470966.aCancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032 China
| | - Hao Zhang
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Yuheng Yan
- grid.412793.a0000 0004 1799 5032Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Qi Mei
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China. .,Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China. .,Cancer Center, Tongji hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
4
|
Abboodi F, Buckhaults P, Altomare D, Liu C, Hosseinipour M, Banister CE, Creek KE, Pirisi L. HPV-inactive cell populations arise from HPV16-transformed human keratinocytes after p53 knockout. Virology 2020; 554:9-16. [PMID: 33321328 DOI: 10.1016/j.virol.2020.12.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/24/2020] [Accepted: 12/08/2020] [Indexed: 10/22/2022]
Abstract
HPV-inactive head and neck and cervical cancers contain HPV DNA but do not express HPV E6/E7. HPV-positive primary head and neck tumors usually express E6/E7, however they may produce HPV-inactive metastases. These observations led to our hypothesis that HPV-inactive cancers begin as HPV-active lesions, losing dependence on E6/E7 expression during progression. Because HPV-inactive cervical cancers often have mutated p53, we investigated whether p53 loss may play a role in the genesis of HPV-inactive cancers. p53 knockout (p53-KO) by CRISPR-Cas9 resulted in a 5-fold reduction of E7 mRNA in differentiation-resistant HPV16 immortalized human keratinocytes (HKc/DR). E7 expression was restored by 5-Aza-2 deoxycytidine in p53 KO lines, suggesting a role of DNA methylation in this process. In-situ hybridization showed that p53 KO lines consist of mixed populations of E6/E7-positive and negative cells. Hence, loss of p53 predisposes HPV16 transformed cells to losing dependence on the continuous expression of HPV oncogenes for proliferation.
Collapse
Affiliation(s)
- Fadi Abboodi
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, USA; Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA; Department of Pediatrics, Mosul Medical College, University of Mosul, Iraq.
| | - Phillip Buckhaults
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA
| | - Changlong Liu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA
| | - Maria Hosseinipour
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, USA
| | - Carolyn E Banister
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA
| | - Kim E Creek
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, USA
| | - Lucia Pirisi
- Department of Pathology, Microbiology, & Immunology, School of Medicine, University of South Carolina, USA.
| |
Collapse
|
5
|
Liu X, Xie S, Zhang J, Kang Y. Long Noncoding RNA XIST Contributes to Cervical Cancer Development Through Targeting miR-889-3p/SIX1 Axis. Cancer Biother Radiopharm 2020; 35:640-649. [PMID: 32191528 DOI: 10.1089/cbr.2019.3318] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background: Cervical cancer (CC) is one of the most common cancers among women in the world. Long noncoding RNAs and microRNAs were identified as important regulators in many physiological processes. The objective of this study was to illuminate the mechanism of X-inactive-specific transcript (XIST)/miR-889-3p/Sine oculis homeobox 1 (SIX1) axis in CC. Methods: The expression levels of XIST, miR-889-3p, and SIX1 were detected by quantitative real-time polymerase chain reaction. Cell proliferation was assessed by cell counting Kit 8 assay. Cell migration and invasion were evaluated by transwell assay. Cell apoptosis was detected by flow cytometry assay. Murine model was established using transfected Me180 cell. The interaction among XIST, miR-889-3p, and SIX1 was tested by dual-luciferase reporter and RNA immunoprecipitation assays. Protein level of SIX1 was measured by Western blot. Results: XIST was highly expressed in CC tissues and cells. Silenced XIST inhibited proliferation, migration, and invasion and induced apoptosis. Moreover, XIST silencing blocked tumor growth in vivo. XIST directly bound to miR-889-3p, and XIST promoted proliferation, migration, and invasion and hindered apoptosis by suppressing miR-889-3p expression. MiR-889-3p targeted SIX1 and negatively regulated SIX1 expression. Furthermore, miR-889-3p had a low expression and SIX1 had a high expression in CC tissues and cells. XIST knockdown reduced SIX1 level by targeting miR-889-3p. In addition, miR-889-3p inhibition abolished the effects of SIX silencing on proliferation, migration, invasion, and apoptosis. Conclusion: XIST knockdown restrained cell proliferation, migration, and invasion and promoted apoptosis by regulating miR-889-3p/SIX1 axis.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Shuangshuang Xie
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Jing Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| | - Yanhua Kang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Hebei North University, Zhangjiakou, China
| |
Collapse
|
6
|
Karan D, Dubey S, Pirisi L, Nagel A, Pina I, Choo YM, Hamann MT. The Marine Natural Product Manzamine A Inhibits Cervical Cancer by Targeting the SIX1 Protein. JOURNAL OF NATURAL PRODUCTS 2020; 83:286-295. [PMID: 32022559 PMCID: PMC7161578 DOI: 10.1021/acs.jnatprod.9b00577] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Indexed: 06/10/2023]
Abstract
Natural products remain an important source of drug leads covering unique chemical space and providing significant therapeutic value for the control of cancer and infectious diseases resistant to current drugs. Here, we determined the antiproliferative activity of a natural product manzamine A (1) from an Indo-Pacific sponge following various in vitro cellular assays targeting cervical cancer (C33A, HeLa, SiHa, and CaSki). Our data demonstrated the antiproliferative effects of 1 at relatively low and non-cytotoxic concentrations (up to 4 μM). Mechanistic investigations confirmed that 1 blocked cell cycle progression in SiHa and CaSki cells at G1/S phase and regulated cell cycle-related genes, including restoration of p21 and p53 expression. In apoptotic assays, HeLa cells showed the highest sensitivity to 1 as compared to other cell types (C33A, SiHa, and CaSki). Interestingly, 1 decreased the levels of the oncoprotein SIX1, which is associated with oncogenesis in cervical cancer. To further investigate the structure-activity relationship among manzamine A (1) class with potential antiproliferative activity, molecular networking facilitated the efficient identification, dereplication, and assignment of structures from the manzamine class and revealed the significant potential in the design of optimized molecules for the treatment of cervical cancer. These data suggest that this sponge-derived natural product class warrants further attention regarding the design and development of novel manzamine analogues, which may be efficacious for preventive and therapeutic treatment of cancer. Additionally, this study reveals the significance of protecting fragile marine ecosystems from climate change-induced loss of species diversity.
Collapse
Affiliation(s)
- Dev Karan
- Department
of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin, United States
| | - Seema Dubey
- Department
of Pathology, MCW Cancer Center and Prostate Cancer Center of Excellence, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin, United States
| | - Lucia Pirisi
- Department
of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, South Carolina, United States
| | - Alexis Nagel
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Ivett Pina
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, United States
| | - Yeun-Mun Choo
- Department
of Chemistry, University of Malaya, Kuala Lumpur, Malaysia
| | - Mark T Hamann
- Department
of Drug Discovery and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina, United States
| |
Collapse
|
7
|
Kong D, Li A, Liu Y, Cui Q, Wang K, Zhang D, Tang J, Du Y, Liu Z, Wu G, Wu K. SIX1 Activates STAT3 Signaling to Promote the Proliferation of Thyroid Carcinoma via EYA1. Front Oncol 2019; 9:1450. [PMID: 31921695 PMCID: PMC6933607 DOI: 10.3389/fonc.2019.01450] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/04/2019] [Indexed: 11/17/2022] Open
Abstract
As a critical member of the Retinal Determination Gene Network (RDGN), SIX1 has been regarded as a tumor promoter in various types of cancer. However, its role in papillary thyroid carcinoma (PTC) has never been investigated. In this study, thyroid carcinoma tissue microarray staining was employed to identify the expression patterns of SIX1 and its co-activator EYA1. Papillary thyroid cancer cell lines, BCPAP, and TPC-1 cells were used to investigate the potential mechanism of SIX1 in vitro and in vivo. Flow cytometry analysis, MTT assay, the growth curve assay, colony formation assay, EdU incorporation and xenograft assay were performed to demonstrate the role of SIX1 in the malignant change of PTC cells. Western blot and Real-time PCR were used to detect the interaction among the SIX1, EYA1, and STAT3 signaling. In comparison with normal tissue, high expressions of SIX1 and EYA1 were associated with a malignant tumor. Importantly, SIX1 strongly correlated with EYA1 in thyroid carcinoma tissue microarray. Functional assays indicated SIX1 increased EYA1 expression by stabilizing EYA1 at the post-transcriptional level. Besides, SIX1 promoted the proliferation and invasion of thyroid carcinoma via activation of STAT3 signaling and its downstream targets in an EYA1-dependent manner. SIX1 can integrate with EYA1 to contribute to PTC development via activation of the classical STAT3 signaling. These data suggested targeting the abnormal activation of the SIX1/EYA1 complex may represent a novel therapeutic strategy for advanced PTC patients.
Collapse
Affiliation(s)
- Deguang Kong
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Anping Li
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Yu Liu
- Department of Geriatrics, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuxia Cui
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Wang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Zhang
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianing Tang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yaying Du
- Department of Thyroid and Breast Surgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhisu Liu
- Department of General Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Gaosong Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kongming Wu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| |
Collapse
|
8
|
Hosseinipour M, Wan F, Altomare D, Creek KE, Pirisi L. HPV16-transformed human keratinocytes depend on SIX1 expression for proliferation and HPV E6/E7 gene expression. Virology 2019; 537:20-30. [PMID: 31425971 DOI: 10.1016/j.virol.2019.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 12/11/2022]
Abstract
The homeodomain transcription factor SIX1 plays a critical role in embryogenesis, is not expressed in normal adult tissue, but is expressed in many malignancies, including cervical cancer. SIX1 drives the progression of HPV16-immortalized human keratinocytes (HKc/HPV16) toward malignancy: HKc/HPV16 express high levels of SIX1 mRNA and protein; overexpression of SIX1 in HKc/HPV16 produces pre-malignant, differentiation-resistant lines (HKc/DR); SIX1 overexpression in HKc/DR induces tumorigenicity. In this paper, we explore the consequences of inhibition of SIX1 expression in premalignant HKc/DR. Only partial inhibition of SIX1 expression could be obtained in HKc/DR by RNA interference. Decreased SIX1 expression (up to 80%) in HKc/DR resulted in slower proliferation, decreased HPV16-E6/E7 mRNA levels, and increased p53 protein levels. Gene expression changes induced in HKc/DR by anti-SIX1 shRNA were indicative of mesenchymal-epithelial transition (MET) and changes in TGF-beta signaling. We conclude that HPV16-transformed cells depend on SIX1 for survival, HPV16 E6/E7 gene expression and epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Maria Hosseinipour
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA
| | - Fang Wan
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA; Inner Mongolia Agricultural University, Hohhot, Inner Mongolia, China
| | - Diego Altomare
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, 29208, USA
| | - Kim E Creek
- Department of Drug Discovery and Biomedical Sciences, University of South Carolina College of Pharmacy, Columbia, SC, 29208, USA
| | - Lucia Pirisi
- Department of Pathology, Microbiology & Immunology, University of South Carolina School of Medicine at Columbia, Columbia, SC 29208, USA.
| |
Collapse
|
9
|
Yang ZC, Qu ZH, Yi MJ, Shan YC, Ran N, Xu L, Liu XJ. MiR-448-5p inhibits TGF-β1-induced epithelial-mesenchymal transition and pulmonary fibrosis by targeting Six1 in asthma. J Cell Physiol 2018; 234:8804-8814. [PMID: 30362537 DOI: 10.1002/jcp.27540] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 09/13/2018] [Indexed: 12/28/2022]
Abstract
MicroRNAs (miRNAs) are small yet versatile gene tuners that regulate a variety of cellular processes, including cell growth and proliferation. The aim of this study was to explore how miR-448-5p affects airway remodeling and transforming growth factor-β1 (TGF-β1)-stimulated epithelial-mesenchymal transition (EMT) by targeting Sine oculis homeobox homolog 1 (Six1) in asthma. Asthmatic mice models with airway remodeling were induced with ovalbumin solution. MiRNA expression was evaluated using quantitative real-time polymerase chain reaction. Transfection studies of bronchial epithelial cells were performed to determine the target genes. A luciferase reporter assay system was applied to identify whether Six1 is a target gene of miR-448-5p. In the current study, we found that miR-448-5p was dramatically decreased in lung tissues of asthmatic mice and TGF-β1-stimulated bronchial epithelial cells. In addition, the decreased level of miR-448-5p was closely associated with the increased expression of Six1. Overexpression of miR-448-5p decreased Six1 expression and, in turn, suppressed TGF-β1-mediated EMT and fibrosis. Next, we predicted that Six1 was a potential target gene of miR-448-5p and demonstrated that miR-448-5p could directly target Six1. An SiRNA targeting Six1 was sufficient to suppress TGF-β1-induced EMT and fibrosis in 16HBE cells. Furthermore, the overexpression of Six1 partially reversed the protective effect of miR-448-5p on TGF-β1-mediated EMT and fibrosis in bronchial epithelial cells. Taken together, the miR-448-5p/TGF-β1/Six1 link may play roles in the progression of EMT and pulmonary fibrosis in asthma.
Collapse
Affiliation(s)
- Zhao-Chuan Yang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China.,Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zheng-Hai Qu
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ming-Ji Yi
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan-Chun Shan
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ni Ran
- Department of Child Health Care, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Xu
- Department of Pediatrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xin-Jie Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
10
|
Stem Cell Properties of Normal Human Keratinocytes Determine Transformation Responses to Human Papillomavirus 16 DNA. J Virol 2018; 92:JVI.00331-18. [PMID: 29593030 DOI: 10.1128/jvi.00331-18] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 03/17/2018] [Indexed: 01/22/2023] Open
Abstract
Human papillomavirus (HPV) infection of the genital tract is common; however, only about 10 to 15% of infections persist, and approximately 10 to 15% of these persistent infections result in cancer. Basal epidermal stem cells are the presumed target cells for HPV infection, providing a reservoir of latently infected cells that persist over time and initiate lesions. However, it is not known whether stem cell density has any influence on transformation of human keratinocytes by HPV. We explored the relationship between stem cell properties of normal human keratinocytes and their susceptibility to transformation by HPV16 DNA. Normal human keratinocyte isolates (NHKc) derived from different donors were cultured in three-dimensional anchorage-free suspension to assess their spheroid-forming ability. NHKc spheroids were then plated back into plastic monolayer culture and transfected with full-length HPV16 DNA, which we have previously shown to integrate into the host cell genome upon transfection. Spheroid-derived NHKc (SD-NHKc) and fluorescence-activated cell sorting-purified populations of basal stem-like keratinocytes, expressing low levels of epidermal growth factor receptor and high levels of integrin alpha 6 (EGFRlo/ITGα6hi), responded to transfection with HPV16 DNA with more vigorous proliferation, greater immortalization efficiency, and faster progression to differentiation resistance than autologous mass-cultured cells. Conversely, cells committed to terminal differentiation (EGFRhi/ITGα6lo) grew slowly after transfection with HPV16 and failed to generate immortalized or DR clones. HPV16 DNA induced stem cell properties in mass-cultured NHKc. We conclude that HPV16 preferentially immortalizes basal keratinocytes with stem cell properties and that these cells readily achieve a differentiation-resistant phenotype upon immortalization by HPV16.IMPORTANCE This paper explores the relationship between the stem cell properties of normal human epidermal cells in culture and these cells' susceptibility to transformation by HPV16 DNA, the HPV type present in about 50% of cervical cancers. We report variable susceptibilities to HPV16-mediated transformation among different keratinocyte isolates derived from neonatal foreskin. Our findings provide strong experimental evidence that HPV16 preferentially transforms basal keratinocytes with stem cell properties. Insights gained from these studies increase our understanding of the host cell-specific factors influencing individual susceptibility to HPV-driven transformation and the contributing factors leading to preneoplastic and neoplastic progression of HPV-positive lesions.
Collapse
|
11
|
Zacapala-Gómez AE, Navarro-Tito N, Alarcón-Romero LDC, Ortuño-Pineda C, Illades-Aguiar B, Castañeda-Saucedo E, Ortiz-Ortiz J, Garibay-Cerdenares OL, Jiménez-López MA, Mendoza-Catalán MA. Ezrin and E-cadherin expression profile in cervical cytology: a prognostic marker for tumor progression in cervical cancer. BMC Cancer 2018; 18:349. [PMID: 29587669 PMCID: PMC5872531 DOI: 10.1186/s12885-018-4243-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 03/16/2018] [Indexed: 01/26/2023] Open
Abstract
Background Cervical cancer (CC) is the fourth cause of mortality by neoplasia in women worldwide. The use of immunomarkers is an alternative tool to complement currently used algorithms for detection of cancer, and to improve selection of therapeutic schemes. Aberrant expression of Ezrin and E-cadherin play an important role in tumor invasion. In this study we analyzed Ezrin and E-cadherin expression in liquid-based cervical cytology samples, and evaluated their potential use as prognostic immunomarkers. Methods Immunocytochemical staining of Ezrin and E-cadherin was performed in cervical samples of 125 patients. The cytological or histological diagnostic was performed by Papanicolaou staining or H&E staining, respectively. HPV genotyping was determined using INNO-LIPA Genotyping Extra kit and the HPV physical status by in situ hybridization. Ezrin expression in HaCaT, HeLa and SiHa cell lines was determined by immunocytochemistry, immunofluorescence and Western blot. Results High Ezrin expression was observed in cervical cancer samples (70%), samples with multiple infection by HR-HPV (43%), and samples with integrated viral genome (47%). High Ezrin expression was associated with degree of SIL, viral genotype and physical status. In contrast, low E-cadherin expression was found in cervical cancer samples (95%), samples with multiple infection by HR-HPV/LR-HPV (87%) and integrated viral genome (72%). Low E-cadherin expression was associated with degree of SIL and viral genotype. Interestingly, Ezrin nuclear staining was associated with degree of SIL and viral genotype. High Ezrin expression, high percent of nuclear Ezrin and low E-cadherin expression behaved as risk factors for progression to HSIL and cervical cancer. Conclusions Ezrin and E-cadherin expression profile in cervical cytology samples could be a potential prognostic marker, useful for identifying cervical lesions with a high-risk of progression to cervical cancer. Electronic supplementary material The online version of this article (10.1186/s12885-018-4243-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ana E Zacapala-Gómez
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lazaro Cardenas s/n, Ciudad Universitaria, CP, 39090, Chilpancingo, Guerrero, Mexico
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Luz Del C Alarcón-Romero
- Laboratorio de Citopatología e Histoquímica, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Carlos Ortuño-Pineda
- Laboratorio de Ácidos nucleicos y proteínas, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lazaro Cardenas s/n, Ciudad Universitaria, CP, 39090, Chilpancingo, Guerrero, Mexico
| | - Eduardo Castañeda-Saucedo
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Julio Ortiz-Ortiz
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lazaro Cardenas s/n, Ciudad Universitaria, CP, 39090, Chilpancingo, Guerrero, Mexico
| | - Olga L Garibay-Cerdenares
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lazaro Cardenas s/n, Ciudad Universitaria, CP, 39090, Chilpancingo, Guerrero, Mexico
| | | | - Miguel A Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lazaro Cardenas s/n, Ciudad Universitaria, CP, 39090, Chilpancingo, Guerrero, Mexico.
| |
Collapse
|
12
|
Yu M, Han G, Qi B, Wu X. Cx32 reverses epithelial-mesenchymal transition in doxorubicin-resistant hepatocellular carcinoma. Oncol Rep 2017; 37:2121-2128. [PMID: 28260043 DOI: 10.3892/or.2017.5462] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/30/2016] [Indexed: 11/06/2022] Open
Abstract
Recently, epithelial-mesenchymal transition (EMT) has been reported to be an important mechanism of drug resistance in numerous types of cancer cells, including hepatocellular carcinoma (HCC). However, the underlying mechanisms remain to be fully elucidated. Connexin (Cx)32 plays a crucial role in hepatocarcinogenesis. The present study investigated the role of Cx32 in the regulation of chemotherapy-induced EMT in HCC. We found that the expression levels of Cx32 and E-cadherin were clearly decreased in HCC tissues compared with the corresponding paracancerous tissues, while the expression level of vimentin was significantly enhanced in HCC tissues. The expression of Cx32 had a strong correlation with the expression of E-cadherin and vimentin. In an in vitro study, a doxorubicin (DOX)-resistant liver cell line HepG2/DOX was established from parental HepG2 cells. The results showed that HepG2/DOX cells acquired EMT characteristics, with a decreased expression level of E-cadherin and an enhanced expression level of vimentin, and possessed high migratory abilities and invasiveness. Meanwhile, Cx32 was significantly decreased in the HepG2/DOX cells. Knockdown of Cx32 by shRNA in HepG2 cells induced EMT, while overexpression of Cx32 converted EMT to mesenchymal-epithelial transition (MET) in the HepG2/DOX cells. These results suggest that Cx32 is an important regulator of DOX-induced EMT in HCC. Cx32 could be considered as a novel target to reverse DOX resistance in HCC.
Collapse
Affiliation(s)
- Meiling Yu
- Department of Pharmacy, The First Affiliated Hospital of Bengbu Medical College, Anhui, Bengbu 233004, P.R. China
| | - Guangshu Han
- Faculty of Pharmacy, Bengbu Medical College, Anhui, Bengbu 233030, P.R. China
| | - Benquan Qi
- Department of Emergency Internal Medicine, The First Affiliated Hospital of Bengbu Medical College, Anhui, Bengbu 233004, P.R. China
| | - Xiaoxiang Wu
- Department of Pharmacy, The Second Affiliated Hospital of Bengbu Medical College, Anhui, Bengbu 233004, P.R. China
| |
Collapse
|
13
|
Xu H, Zhang Y, Peña MM, Pirisi L, Creek KE. Six1 promotes colorectal cancer growth and metastasis by stimulating angiogenesis and recruiting tumor-associated macrophages. Carcinogenesis 2017; 38:281-292. [PMID: 28199476 PMCID: PMC5862328 DOI: 10.1093/carcin/bgw121] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 10/06/2016] [Accepted: 11/09/2016] [Indexed: 01/05/2023] Open
Abstract
The homeoprotein Six1 is overexpressed in many human cancers and is associated with increased tumor progression and metastasis. Recent studies have shown that Six1 is associated with poorer overall survival in advanced-stage colorectal cancer (CRC). In the current study, we explored the functional changes and molecular events associated with Six1 overexpression in a mouse model of CRC. An orthotopic model and a splenic injection metastasis model were used to investigate the role of Six1 in CRC tumor growth and metastasis using mouse colon adenocarcinoma MC38 cells overexpressing Six1. We found that overexpression of Six1 dramatically promotes CRC tumor growth and metastasis in vivo. Six1 overexpression in MC38 increased protein levels of aldehyde dehydrogenase-1 and expanded CD44+/CD166+ populations, indicating Six1 increased features of cancer stem cells. In addition, Six1 overexpression stimulated angiogenesis by upregulating the expression of vascular endothelial growth factor (VEGF). Six1-overexpressing tumor cells recruited tumor-associated macrophages (TAM) by increasing the expression of macrophage-specific colony stimulating factor, chemokine (C-C motif) ligand 2/5 and VEGF, further facilitating CRC tumor growth and metastasis. Furthermore, we determined that Six1 activated mitogen-activated protein kinase (MAPK) signaling in CRC cells. In summary, our studies strongly suggest that Six1 overexpression promotes CRC growth and metastasis and remodels tumor stroma by stimulating angiogenesis and recruiting TAM. MAPK activation may be a pivotal event in Six1-associated tumor progression, which may provide opportunities for pharmacologic intervention.
Collapse
Affiliation(s)
- Hanwen Xu
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Columbia, SC 29208, USA
| | - Yu Zhang
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
| | - Maria M Peña
- Department of Biological Sciences, University of South Carolina, Columbia, SC 29208, USA
- Center for Colon Cancer Research, University of South Carolina, Columbia, SC 29208, USA
| | - Lucia Pirisi
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | - Kim E Creek
- Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, Columbia, SC 29208, USA
| |
Collapse
|
14
|
Chow LT. Model systems to study the life cycle of human papillomaviruses and HPV-associated cancers. Virol Sin 2015; 30:92-100. [PMID: 25924993 DOI: 10.1007/s12250-015-3600-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 04/21/2015] [Indexed: 12/21/2022] Open
Abstract
The prevalent human papillomaviruses (HPVs) infect either cutaneous or mucosal epithelium. Active Infections lead to epithelial hyperprolifeation and are usually cleared in healthy individuals within a year. Persistent infections in the anogenital tracts by certain high-risk genotypes such as HPV-16, HPV-18 and closely related types, can progress to high grade dysplasias and carcinomas in women and men, including cervical, vulva, penile and anal cancers. A significant fraction of the head and neck cancers are also caused by HPV-16. The viral oncogenes responsible for neoplastic conversion are E6 and E7 that disrupt the pathways controlled by the two major tumor suppressor genes, p53 and members of pRB family. Because HPV cannot be propagated in conventional submerged monolayer cell cultures, organotypic epithelial raft cultures that generate a stratified and differentiated epithelium have been used to study the viral life cycle. This article describes several systems to examine aspects of the viral productive phase, along with the advantages and limitations. Animal model systems of HPV carcinogenesis are also briefly described.
Collapse
Affiliation(s)
- Louise T Chow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, 35294-0005, USA,
| |
Collapse
|