1
|
Sun Y, Xiao Z, Yang S, Hao C, Zhao H, An Y. Advances and insights for DKK3 in non-cancerous diseases: a systematic review. PeerJ 2025; 13:e18935. [PMID: 39959827 PMCID: PMC11830365 DOI: 10.7717/peerj.18935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025] Open
Abstract
This review delves into the role of Dickkopf-3 (DKK3), a secreted glycoprotein and member of the Dickkopf family, in non-malignant diseases. DKK3 is particularly known for its regulatory effects on the Wnt signaling pathway, a critical mediator in various biological processes including cell proliferation, differentiation, and migration. Our review highlights DKK3's influence in disorders of the cardiovascular, respiratory, renal, and muscular systems, where it contributes to disease progression by modulating these key biological processes. As an emerging biomarker, DKK3's levels have been found to correlate with various disease states, underscoring its potential diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Yao Sun
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Zengli Xiao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Shuguang Yang
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Chenxiao Hao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Huiying Zhao
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| | - Youzhong An
- Intensive Care Unit, Peking University People’s Hospital, Beijing, China
| |
Collapse
|
2
|
Mourtada J, Thibaudeau C, Wasylyk B, Jung AC. The Multifaceted Role of Human Dickkopf-3 (DKK-3) in Development, Immune Modulation and Cancer. Cells 2023; 13:75. [PMID: 38201279 PMCID: PMC10778571 DOI: 10.3390/cells13010075] [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: 11/14/2023] [Revised: 12/22/2023] [Accepted: 12/26/2023] [Indexed: 01/12/2024] Open
Abstract
The human Dickkopf (DKK) family includes four main secreted proteins, DKK-1, DKK-2, DKK-3, and DKK-4, as well as the DKK-3 related protein soggy (Sgy-1 or DKKL1). These glycoproteins play crucial roles in various biological processes, and especially modulation of the Wnt signaling pathway. DKK-3 is distinct, with its multifaceted roles in development, stem cell differentiation and tissue homeostasis. Intriguingly, DKK-3 appears to have immunomodulatory functions and a complex role in cancer, acting as either a tumor suppressor or an oncogene, depending on the context. DKK-3 is a promising diagnostic and therapeutic target that can be modulated by epigenetic reactivation, gene therapy and DKK-3-blocking agents. However, further research is needed to optimize DKK-3-based therapies. In this review, we comprehensively describe the known functions of DKK-3 and highlight the importance of context in understanding and exploiting its roles in health and disease.
Collapse
Affiliation(s)
- Jana Mourtada
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France; (J.M.); (C.T.)
- Laboratoire STREINTH (Stress Response and Innovative Therapies), INSERM U1113 IRFAC, Université de Strasbourg, 67200 Strasbourg, France
| | - Chloé Thibaudeau
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France; (J.M.); (C.T.)
- Laboratoire STREINTH (Stress Response and Innovative Therapies), INSERM U1113 IRFAC, Université de Strasbourg, 67200 Strasbourg, France
| | - Bohdan Wasylyk
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), 67404 Illkirch Graffenstaden, France;
- Institut National de la Santé et de la Recherche Médicale (INSERM), U 1258, 67404 Illkirch Graffenstaden, France
- Centre Nationale de la Recherche Scientifique (CNRS), UMR 7104, 67404 Illkirch Graffenstaden, France
- Université de Strasbourg, 67000 Strasbourg, France
| | - Alain C. Jung
- Laboratoire de Biologie Tumorale, Institut de Cancérologie Strasbourg Europe, 67200 Strasbourg, France; (J.M.); (C.T.)
- Laboratoire STREINTH (Stress Response and Innovative Therapies), INSERM U1113 IRFAC, Université de Strasbourg, 67200 Strasbourg, France
| |
Collapse
|
3
|
Wen B, Hu S, Yin J, Wu J, Guo W. Molecular Evolution and Protein Structure Variation of Dkk Family. Genes (Basel) 2023; 14:1863. [PMID: 37895211 PMCID: PMC10606412 DOI: 10.3390/genes14101863] [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: 08/24/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/29/2023] Open
Abstract
Dkks have inhibitory effects on the Wnt signaling pathway, which is involved in the development of skin and its appendages and the regulation of hair growth. The nucleotide sequences were compared and analyzed to further investigate the relationship between the structure and function of the Dkk gene family and vertebrate epidermal hair. The analysis of the molecular evolution of the Dkk family revealed that the evolution rate of the genes changed significantly after speciation, with the Aves and Reptilia branches showing accelerated evolution. Additionally, positive selection was observed at specific sites. The tertiary structure of the protein was also predicted. The analysis of the functional divergence of the Dkk family revealed that the functional divergence coefficient of each gene was greater than 0, with most of the functional divergence sites were located in the Cys-2 domain and a few in the Cys-1 domain. This suggests that the amino acid and functional divergence sites may play a role in regulating the binding of the Dkk family to LRP5/6, and thus affect the inhibition of Wnt signaling, leading to different functions of Dkk1, Dkk2, and Dkk4 in the development of skin hair follicles. In addition, the Dkk families of Aves and Reptilia may have undergone adaptive evolution and functional divergence.
Collapse
Affiliation(s)
- Binhong Wen
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Sile Hu
- College of Life Science, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Jun Yin
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China;
| | - Jianghong Wu
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Wenrui Guo
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
4
|
Park MH, Shin JH, Bothwell AL, Chae WJ. Dickkopf proteins in pathological inflammatory diseases. J Leukoc Biol 2022; 111:893-901. [PMID: 34890067 PMCID: PMC9889104 DOI: 10.1002/jlb.3ri0721-385r] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/01/2021] [Accepted: 11/17/2021] [Indexed: 02/02/2023] Open
Abstract
The human body encounters various challenges. Tissue repair and regeneration processes are augmented after tissue injury to reinstate tissue homeostasis. The Wnt pathway plays a crucial role in tissue repair since it induces target genes required for cell proliferation and differentiation. Since tissue injury causes inflammatory immune responses, it has become increasingly clear that the Wnt ligands can function as immunomodulators while critical for tissue homeostasis. The Wnt pathway and Wnt ligands have been studied extensively in cancer biology and developmental biology. While the Wnt ligands are being studied actively, how the Wnt antagonists and their regulatory mechanisms can modulate immune responses during chronic pathological inflammation remain elusive. This review summarizes DKK family proteins as immunomodulators, aiming to provide an overarching picture for tissue injury and repair. To this end, we first review the Wnt pathway components and DKK family proteins. Next, we will review DKK family proteins (DKK1, 2, and 3) as a new class of immunomodulatory protein in cancer and other chronic inflammatory diseases. Taken together, DKK family proteins and their immunomodulatory functions in chronic inflammatory disorders provide novel insights to understand immune diseases and make them attractive molecular targets for therapeutic intervention.
Collapse
Affiliation(s)
- Min Hee Park
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, 401 College Street., Richmond, VA 23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, 401 College Street., Richmond, VA 23298
| | - Jae Hun Shin
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520
| | - Alfred L.M. Bothwell
- Department of Immunobiology, Yale University School of Medicine, 300 Cedar Street, New Haven, CT 06520
| | - Wook-Jin Chae
- Department of Microbiology and Immunology, Virginia Commonwealth University School of Medicine, 401 College Street., Richmond, VA 23298
- Massey Cancer Center, Virginia Commonwealth University School of Medicine, 401 College Street., Richmond, VA 23298
| |
Collapse
|
5
|
The role of the WNT signaling pathway in the maxillary sinus squamous cell carcinoma. Med Oncol 2022; 39:42. [PMID: 35092507 DOI: 10.1007/s12032-021-01640-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
Abstract
Paranasal sinus tumors are a rare type of cancer. Most of these tumors are of epithelial origin and 80% of them are maxillary sinus squamous cell carcinoma. The WNT signaling pathway is an essential embryonic regulatory pathway known to play an important role in many cancers, including head and neck cancers. However, the effect of this pathway in maxillary sinus tumors has not been studied before. The aim of the study was to determine the changes in the regulatory genes of the WNT signaling pathway in maxillary sinus tumors. For this purpose, total RNA was isolated from the pathological preparations of 85 patients who had previously been operated on for squamous cell maxillary sinus tumor, and gene expression changes were evaluated by real-time RT-qPCR. The interactions among proteins encoded by genes, whose expression levels were found to be decreased and increased, were determined by protein-protein interaction (PPI) network analysis using string database, and signaling pathways that they are involved in were examined by Reactome database. A significant decrease in the expression of 28 genes compared to the control (fold change < 2.00 and p-value < 0.05) and a significant increase in the expression of 23 genes (fold change < 2.00 and p-value < 0.05) were detected. According to in silico analysis results, Signal Transduction (REACTOME:R-HSA-162582) and Signaling by WNT (REACTOME:R-HSA-195721) pathways were determined as most regulated pathways and FZD4-LRP5 and BCL9-CTNNB1 were determined as the strongest interactions. The current study contributes to illuminating the genetic regulation of maxillary sinus carcinoma in which genetic knowledge is limited. Our findings take attention to the dysregulations of the WNT signaling pathway that may support maxillary sinus carcinogenesis. The results will pave the way for further studies that investigate the therapy target potential of the WNT signaling pathway in this rare cancer.
Collapse
|
6
|
Molecular epigenetic dynamics in breast carcinogenesis. Arch Pharm Res 2021; 44:741-763. [PMID: 34392501 DOI: 10.1007/s12272-021-01348-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer has become one of the most common dreadful diseases that target women across the globe. The most obvious reasons we associate with it are either genetic mutations or dysregulation of pathways. However, there is yet another domain that has a significant role in influencing the genetic mutations and pathways. Epigenetic mechanisms influence these pathways either independently or in association with genetic mutations, thereby expediting the process of breast carcinogenesis. Breast cancer is governed by various transduction pathways such as PI3K/AKT/mTOR, NOTCH, β Catenin, NF-kB, Hedgehog, etc. There are many proteins as well that serve to be tumor suppressors but somehow lose their ability to function. This may be because of either genetic mutation or a process that represses their function. Apart from these, there are a lot of individual factors like puberty, breastfeeding, abortion, parity, circadian rhythm, alcohol consumption, pollutants, and obesity that drive these mutations and hence alter the pathways. Epigenetic mechanisms like DNA methylation, histone modifications, and lncRNAs directly or indirectly bring alterations in the proteins that are involved in the pathways. They do this by either promoting the transcription of genes or by repressing it at the ground genetic level that advances breast carcinogenesis. Epigenetics precedes genetic mutation in driving carcinogenesis and so, it needs to be explored further to diversify the possibilities of target specific treatments. In this review, the general role of DNA methylation, histone modification, and lncRNAs in breast cancer and their role in influencing the oncogenic signaling pathways along with the various factors governing them have been discussed for a better understanding of the role of epigenetics in breast carcinogenesis.
Collapse
|
7
|
Zhou AX, Mondal T, Tabish AM, Abadpour S, Ericson E, Smith DM, Knöll R, Scholz H, Kanduri C, Tyrberg B, Althage M. The long noncoding RNA TUNAR modulates Wnt signaling and regulates human β-cell proliferation. Am J Physiol Endocrinol Metab 2021; 320:E846-E857. [PMID: 33682459 DOI: 10.1152/ajpendo.00335.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Many long noncoding RNAs (lncRNAs) are enriched in pancreatic islets and several lncRNAs are linked to type 2 diabetes (T2D). Although they have emerged as potential players in β-cell biology and T2D, little is known about their functions and mechanisms in human β-cells. We identified an islet-enriched lncRNA, TUNAR (TCL1 upstream neural differentiation-associated RNA), which was upregulated in β-cells of patients with T2D and promoted human β-cell proliferation via fine-tuning of the Wnt pathway. TUNAR was upregulated following Wnt agonism by a glycogen synthase kinase-3 (GSK3) inhibitor in human β-cells. Reciprocally, TUNAR repressed a Wnt antagonist Dickkopf-related protein 3 (DKK3) and stimulated Wnt pathway signaling. DKK3 was aberrantly expressed in β-cells of patients with T2D and displayed a synchronized regulatory pattern with TUNAR at the single cell level. Mechanistically, DKK3 expression was suppressed by the repressive histone modifier enhancer of zeste homolog 2 (EZH2). TUNAR interacted with EZH2 in β-cells and facilitated EZH2-mediated suppression of DKK3. These findings reveal a novel cell-specific epigenetic mechanism via islet-enriched lncRNA that fine-tunes the Wnt pathway and subsequently human β-cell proliferation.NEW & NOTEWORTHY The discovery that long noncoding RNA TUNAR regulates β-cell proliferation may be important in designing new treatments for diabetes.
Collapse
Affiliation(s)
- Alex-Xianghua Zhou
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Tanmoy Mondal
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
- Department of Clinical Chemistry and Transfusion Medicine, Sahlgrenska University Hospital Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Ali Mustafa Tabish
- Integrated Cardio Metabolic Centre, Karolinska Institute, Stockholm, Sweden
| | - Shadab Abadpour
- Department of Transplant Medicine, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
- Hybrid Technology Hub, Centre of Excellence, University of Oslo, Oslo, Norway
| | - Elke Ericson
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - David M Smith
- Emerging Innovations Unit, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Ralph Knöll
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Integrated Cardio Metabolic Centre, Karolinska Institute, Stockholm, Sweden
| | - Hanne Scholz
- Department of Transplant Medicine, Institute for Surgical Research, Oslo University Hospital, Oslo, Norway
| | - Chandrasekhar Kanduri
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Björn Tyrberg
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Althage
- Research and Early Development Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
8
|
Ramezani A, Zakeri A, Mard-Soltani M, Mohammadian A, Hashemi ZS, Mohammadpour H, Jahangiri A, Khalili S, Rasaee MJ. Structure Based Screening for Inhibitory Therapeutics of CTLA-4 Unveiled New Insights About Biology of ACTH. Int J Pept Res Ther 2020. [DOI: 10.1007/s10989-019-09891-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Yang Y, Xu W, Zheng Z, Cao Z. LINC00459 sponging miR-218 to elevate DKK3 inhibits proliferation and invasion in melanoma. Sci Rep 2019; 9:19139. [PMID: 31844121 PMCID: PMC6914790 DOI: 10.1038/s41598-019-55701-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 11/27/2019] [Indexed: 12/22/2022] Open
Abstract
The lncRNA biomarkers in melanoma remain to be further explored. The lncRNAs with different expression levels in melanoma tissue were identified by microarray analysis. To investigate the biological functions of target lncRNA, several in-vivo and in-vitro studies were performed. Potential mechanisms of competitive endogenous RNAs (ceRNAs) were predicted by using bioinformatics analysis and explored by western blot assay, fluorescence in situ hybridization assay, real-time quantitative PCR (RT-qPCR) array, RNA pull-down analysis, AGO2-RIP assay, and dual-luciferase reporter assay. The results demonstrated decreased LINC00459 in melanoma cell lines and tissues. According to the in-vitro and in-vivo experiments, up-regulated LINC00459 had inhibitory effect on cell proliferation and invasion. Bioinformatics analyses suggested that miR-218 could be a direct target of LINC00459. In addition, miR-218 was proved to be able to directly target the dickkopf-related protein 3 (DKK3) gene. In conclusion, our analysis suggested that the LINC00459 could sponge miR-218 and increase the expression of DKK3 gene, thus inhibiting the invasion and proliferation of melanoma cells, which indicated that the LINC00459 could be an effective biomarker for melanoma and its potential as the therapeutic target.
Collapse
Affiliation(s)
- Yuhua Yang
- Department of Dermatology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Wenxian Xu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhuojun Zheng
- Department of Hematology, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| | - Zhihai Cao
- Department of Emergency, The Third Affiliated Hospital of Soochow University, Changzhou, China.
| |
Collapse
|
10
|
Bonakdar A, Sahebazzamani F, Rasaee MJ, Hosseinkhani S, Rahbarizadeh F, Mahboudi F, Ganjali MR. In silico design and in vitro characterization of a recombinant antigen for specific recognition of NMP22. Int J Biol Macromol 2019; 140:69-77. [PMID: 31404598 DOI: 10.1016/j.ijbiomac.2019.08.065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 08/06/2019] [Accepted: 08/07/2019] [Indexed: 01/22/2023]
Abstract
Although urine cytology and cystoscopy are current gold standard methods in diagnosis and surveillance of Bladder cancer (BC), they have some limitations which necessitates novel diagnostic approaches to compensate their drawbacks. In this regard, Nuclear Matrix Protein 22 (NMP22) is introduced as a potential tumor biomarker for BC detection (FDA approved). NMP22 determination mainly occurs through immunoassay platforms, raising a proper antibody against its antigen. Hence, development of such immunoassays seems crucial. Various bioinformatic tools were harnessed to select a region with lowest variability, highest density for linear and conformational epitopes, lowest post translational modifications, highest antigenicity, best physicochemical properties and reliable transcriptional properties. Subsequently, E. coli BL21 (DE3) and P. pastoris GS115 were applied for exogenous expression. Ultimately, protein purification and quantification was followed by ELISA test for antibody analyses. Both host successfully expressed the antigen, while the E. coli expression was with higher yield. The commercial anti-NMP22 antibodies showed relatively equal detection results. However, the slight better detection for the antigen with P. pastoris origin could be deduced as better structural properties for P. pastoris. These results indicate higher expression yields and lower costs for over-expression of this eukaryotic antigen.
Collapse
Affiliation(s)
- Alireza Bonakdar
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Sahebazzamani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Mohammad Reza Ganjali
- Center of Excellence in Electrochemistry, University of Tehran, Tehran, Iran; Biosensor Research Center, Endocrinology & Metabolism Molecular - Cellular Sciences Institute, Iran
| |
Collapse
|
11
|
Bhattacharyya S, Feferman L, Tobacman JK. Dihydrotestosterone inhibits arylsulfatase B and Dickkopf Wnt signaling pathway inhibitor (DKK)-3 leading to enhanced Wnt signaling in prostate epithelium in response to stromal Wnt3A. Prostate 2019; 79:689-700. [PMID: 30801800 DOI: 10.1002/pros.23776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/23/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND In tissue microarrays, immunostaining of the enzyme arylsulfatase B (ARSB; N-acetylgalactosamine-4-sulfatase) was less in recurrent prostate cancers and in cancers with higher Gleason scores. In cultured prostate stem cells, decline in ARSB increased Wnt signaling through effects on Dickkopf Wnt Signaling Pathway Inhibitor (DKK)3. The effects of androgen exposure on ARSB and the impact of decline in ARSB on Wnt signaling in prostate tissue were unknown. METHODS Epithelial and stromal tissues from malignant and normal human prostate were obtained by laser capture microdissection. mRNA expression of ARSB, galactose-6-sulfate-sulfatase (GALNS) and Wnt-signaling targets was determined by QPCR. Non-malignant human epithelial and stromal prostate cells were grown in tissue culture, including two-cell layer cultures. ARSB was silenced by specific siRNA, and epithelial cells were treated with stromal spent media following treatment with IWP-2, an inhibitor of Wnt secretion, and by exogenous recombinant human Wnt3A. Promoter methylation was detected using specific DKK3 and ARSB promoter primers. The effects of DHT and of ARSB overexpression on DKK expression were determined. Cell proliferation was assessed by BrdU incorporation. RESULTS Normal stroma showed higher expression of vimentin, ARSB, and Wnt3A than epithelium. Normal epithelium had higher expression of E-cadherin, galactose 6-sulfate-sulfatase (GALNS), and DKK3 than stroma. In malignant epithelium, expression of ARSB and DKK3 declined, and expression of GALNS and Wnt signaling targets increased. In cultured prostate epithelial cells, Wnt-mediated signaling was greatest when ARSB was silenced and cells were exposed to exogenous Wnt3A. Exposure to 5α-dihydrotestosterone (DHT) increased ARSB and DKK3 promoter rmethylation, and effects of DHT on DKK3 expression were reversed when ARSB was overexpressed. CONCLUSIONS Androgen-induced declines in ARSB and DKK3 may contribute to prostate carcinogenesis by sustained activation of Wnt signaling in prostate epithelium in response to stromal Wnt3A.
Collapse
Affiliation(s)
- Sumit Bhattacharyya
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| | - Leo Feferman
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| | - Joanne K Tobacman
- Department of Medicine, The University of Illinois at Chicago and Jesse Brown VAMC, Chicago, Illinois
| |
Collapse
|
12
|
MiR-92a modulates proliferation, apoptosis, migration, and invasion of osteosarcoma cell lines by targeting Dickkopf-related protein 3. Biosci Rep 2019; 39:BSR20190410. [PMID: 30926679 PMCID: PMC6487267 DOI: 10.1042/bsr20190410] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2019] [Revised: 03/21/2019] [Accepted: 03/26/2019] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma (OS) is recognized as a common malignant tumor with a high trend of metastasis and diffusion. Despite the progresses that have been made in surgery, chemotherapy, and radiotherapy in the recent decades, the prognosis of patients with OS still remains poor. MiRNAs are being increasingly considered as new therapeutic targets for OS treatment. Our research aims to investigate the regulatory impact of miR-92a in the development of OS. Quantitative real-time PCR (qRT-PCR) results revealed that the expression of miR-92a was aberrantly overexpressed in human OS cell lines. By using cell counting kit-8 (CCK-8) assays, colony formation assays, flow cytometric analyses and Transwell assays, our data suggested that up-regulation of miR-92a promoted the proliferation, migration, and invasion of MNNG and U2OS cells, while inhibiting their apoptosis. In contrast, the knockdown of miR-92a effectively reversed these cellular biological behaviors. Furthermore, bioinformatics analysis indicated that Dickkopf-related protein 3 (DKK3) was a possible target of miR-92a. Subsequently, negative regulation of miR-92a on DKK3 was observed, which further supported the direct binding between them. In addition, silencing DKK3 rescued the inhibitory effect of miR-92a inhibitor on the development of OS. To sum up, our study revealed that miR-92a played a carcinogenic role in the growth of OS by promoting the tumorigenesis of OS cells via targeting of DKK3, thus revealing a new therapeutic target for OS.
Collapse
|
13
|
Igbinigie E, Guo F, Jiang SW, Kelley C, Li J. Dkk1 involvement and its potential as a biomarker in pancreatic ductal adenocarcinoma. Clin Chim Acta 2019; 488:226-234. [PMID: 30452897 DOI: 10.1016/j.cca.2018.11.023] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 11/12/2018] [Accepted: 11/14/2018] [Indexed: 02/05/2023]
Abstract
Dickkopf-1 (Dkk1)'s dysregulation has been implicated in the pathogenesis of a variety of cancers. It is part of the Dkk family of proteins that includes Dkk2, Dkk3 and Dkk4. This family of secreted proteins shares similar conserved cysteine domains and inhibits the Wnt/b-catenin pathway by causing proteasomal B-catenin degradation, inducing apoptosis, and preventing cell proliferation. Pancreatic ductal adenocarcinoma (PDAC) is the 4th leading cause of cancer mortality in the United States due to the late stage of diagnosis and the limited effectiveness of current therapy. Dkk1 is found increased in PADC patients' specimens and serum. Dkk1 can be a promising biomarker specific to PDAC, which has the potential to increase PDAC survival rates through improving early stage detection and monitoring progression compared to current biomarker gold standards. In addition, recent studies suggest that Dkk1 could be an excellent target for cancer immunotherapy. Interestingly, Dkk1-CKAP4-PI3K/AKT signal pathway also plays role in pancreatic cancer cell proliferation. In this review, we present the multiple mechanisms of Dkk1 in PDAC studied thus far and explore its function, regulation, and clinical applications in gynecological cancers including pancreatic ductal adenocarcinoma (PDAC), breast, ovarian, cervical, and endometrial cancer. Further research into Dkk1's mechanism and use as a diagnostic tool, alone or in combination with other biomarkers, could prove clinically useful for better understanding the pathology of PDAC and improving its early detection and treatment.
Collapse
Affiliation(s)
- Eseosaserea Igbinigie
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Fengbiao Guo
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA; Department of Histology and Embryology, Shantou University Medical College, Shantou 515000, China.
| | - Shi-Wen Jiang
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Cullen Kelley
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA.
| | - Jinping Li
- Department of Biomedical Sciences, Mercer University School of Medicine, Savannah, GA 31404, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic, Florida Campus, Jacksonville, FL 32224, USA.
| |
Collapse
|
14
|
Zhu X, Li W, Li H. miR-214 ameliorates acute kidney injury via targeting DKK3 and activating of Wnt/β-catenin signaling pathway. Biol Res 2018; 51:31. [PMID: 30180910 PMCID: PMC6122444 DOI: 10.1186/s40659-018-0179-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022] Open
Abstract
Background miR-214 was demonstrated to be upregulated in models of renal disease and promoted fibrosis in renal injury independent of TGF-β signaling in vivo. However, the detailed role of miR-214 in acute kidney injury (AKI) and its underlying mechanism are still largely unknown. Methods In this study, an I/R-induced rat AKI model and a hypoxia-induced NRK-52E cell model were used to study AKI. The concentrations of kidney injury markers serum creatinine, blood urea nitrogen, and kidney injury molecule-1 were measured. The expressions of miR-214, tumor necrosis factor-α, interleukin (IL)-1β, IL-6, were detected by RT-qPCR. The protein levels of Bcl-2, Bax, Dickkopf-related protein 3, β-catenin, c-myc, and cyclinD1 were determined by western blot. Cell apoptosis and caspase 3 activity were evaluated by flow cytometry analysis and caspase 3 activity assay, respectively. Luciferase reporter assay was used to confirm the interaction between miR-214 and Dkk3. Results miR-214 expression was induced in ischemia–reperfusion (I/R)-induced AKI rat and hypoxic incubation of NRK-52E cells. Overexpression of miR-214 alleviated hypoxia-induced NRK-52E cell apoptosis while inhibition of miR-214 expression exerted the opposite effect. Dkk3 was identified as a target of miR-214. Anti-miR-214 abolished the inhibitory effects of DKK3 knockdown on hypoxia-induced NRK-52E cell apoptosis by inactivation of Wnt/β-catenin signaling. Moreover, miR-214 ameliorated AKI in vivo by inhibiting apoptosis and fibrosis through targeting Dkk3 and activating Wnt/β-catenin pathway. Conclusion miR-214 ameliorates AKI by inhibiting apoptosis through targeting Dkk3 and activating Wnt/β-catenin signaling pathway, offering the possibility of miR-214 in the therapy of ischemic AKI. Electronic supplementary material The online version of this article (10.1186/s40659-018-0179-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoguang Zhu
- Department of Nephrology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China.
| | - Wenwen Li
- Department of Cardiology, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Huicong Li
- Department of Nephrology, Huaihe Hospital of Henan University, No. 8, Baobei Road, Gulou District, Kaifeng, 475000, China
| |
Collapse
|
15
|
Han SW, Kim YY, Kang WJ, Kim HC, Ku SY, Kang BC, Yun JW. The Use of Normal Stem Cells and Cancer Stem Cells for Potential Anti-Cancer Therapeutic Strategy. Tissue Eng Regen Med 2018; 15:365-380. [PMID: 30603561 PMCID: PMC6171655 DOI: 10.1007/s13770-018-0128-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 05/08/2018] [Accepted: 05/24/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Despite recent advance in conventional cancer therapies including surgery, radiotherapy, chemotherapy, and immunotherapy to reduce tumor size, unfortunately cancer mortality and metastatic cancer incidence remain high. Along with a deeper understanding of stem cell biology, cancer stem cell (CSC) is important in targeted cancer therapy. Herein, we review representative patents using not only normal stem cells as therapeutics themselves or delivery vehicles, but also CSCs as targets for anti-cancer strategy. METHODS Relevant patent literatures published between 2005 and 2017 are discussed to present developmental status and experimental results on using normal stem cells and CSCs for cancer therapy and explore potential future directions in this field. RESULTS Stem cells have been considered as important element of regenerative therapy by promoting tissue regeneration. Particularly, there is a growing trend to use stem cells as a target drug-delivery system to reduce undesirable side effects in non-target tissues. Noteworthy, studies on CSC-specific markers for distinguishing CSCs from normal stem cells and mature cancer cells have been conducted as a selective anti-cancer therapy with few side effects. Many researchers have also reported the development of various substances with anticancer effects by targeting CSCs from cancer tissues. CONCLUSION There has been a continuing increase in the number of studies on therapeutic stem cells and CSC-specific markers for selective diagnosis and therapy of cancer. This review focuses on the current status in the use of normal stem cells and CSCs for targeted cancer therapy. Future direction is also proposed.
Collapse
Affiliation(s)
- Seung-Woo Han
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| | - Yoon Young Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Woo-Ju Kang
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| | - Hyoung-Chin Kim
- Laboratory Animal Resource Center, Korea Research Institute of Bioscience and Biotechnology, 30 Yeongudanji-ro, Ochang-eup, Cheongwon-gu, Cheongju-si, Chungcheongbuk-do 28116 Republic of Korea
| | - Seung-Yup Ku
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
| | - Byeong-Cheol Kang
- Department of Experimental Animal Research, Biomedical Research Institute, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Biomedical Center for Animal Resource and Development, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Graduate School of Translational Medicine, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080 Republic of Korea
- Designed Animal and Transplantation Research Institute, Institute of GreenBio Science Technology, Seoul National University, 1447 Pyeongchang-daero, Daehwa-myeon, Pyeongchang-gun, Gangwon-do 25354 Republic of Korea
| | - Jun-Won Yun
- Department of Biotechnology, The Catholic University of Korea, 43 Jibongro, Bucheon, 14662 Republic of Korea
| |
Collapse
|
16
|
Mohammadpour H, Du W, O'Neill R, Khalili S, Qiu J, Repasky EA, McCarthy PL, Cao X. Host-Derived Serine Protease Inhibitor 6 Provides Granzyme B-Independent Protection of Intestinal Epithelial Cells in Murine Graft-versus-Host Disease. Biol Blood Marrow Transplant 2018; 24:2397-2408. [PMID: 30006303 DOI: 10.1016/j.bbmt.2018.07.003] [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: 01/11/2018] [Accepted: 07/03/2018] [Indexed: 01/03/2023]
Abstract
Graft-versus-host disease (GVHD) is a serious complication after allogeneic hematopoietic cell transplantation (allo-HCT) that limits the therapeutic potential of this treatment. Host antigen-presenting cells (APCs) play a vital role in activating donor T cells that subsequently use granzyme B (GzmB) and other cytotoxic molecules to damage host normal tissues. Serine protease inhibitor 6 (Spi6), known as the sole endogenous inhibitor of GzmB, has been implicated in protecting T cells and APCs against GzmB-inflicted damage. In this study we used murine models to examine the previously unknown role of host-derived Spi6 in GVHD pathogenesis. Our results indicated that host Spi6 deficiency exacerbated GVHD as evidenced by significantly increased lethality and clinical and histopathologic scores. Using bone marrow chimera system, we found that Spi6 in nonhematopoietic tissue played a dominant role in protecting against GVHD and was significantly upregulated in intestinal epithelial cells after allo-HCT, whereas Spi6 in hematopoietic APCs surprisingly suppressed alloreactive T cell response. Interestingly, the protective effect of Spi6 and its expression in intestinal epithelial cells appeared to be independent of donor-derived GzmB. We used in silico modeling to explore potential targets of Spi6. Interaction tested in silico demonstrated that Spi6 could inhibit caspase-3 and caspase-8 with the same functional loop that inhibits GzmB but was not capable of forming stable interaction with caspase-1 or granzyme A. Using an in vitro co-culture system, we further identified that donor T cell-derived IFN-γ was important for inducing Spi6 expression in an intestinal epithelial cell line. Altogether, our data indicate that host Spi6 plays a novel, GzmB-independent role in regulating alloreactive T cell response and protecting intestinal epithelial cells. Therefore, enhancing host-derived Spi6 function has the potential to reduce GVHD.
Collapse
Affiliation(s)
- Hemn Mohammadpour
- Department of Immunology; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Wei Du
- Department of Immunology; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Rachel O'Neill
- Department of Immunology; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Saeed Khalili
- Department of Biology Sciences, Faculty of Sciences, Shahid Rajee Teacher Training University, Tehran, Iran
| | - Jingxin Qiu
- Department of Pathology; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Elizabeth A Repasky
- Department of Immunology; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Philip L McCarthy
- Department of Medicine; Roswell Park Comprehensive Cancer Center, Buffalo, New York
| | - Xuefang Cao
- Department of Immunology; Roswell Park Comprehensive Cancer Center, Buffalo, New York.
| |
Collapse
|
17
|
Xiang L, Rong G, Zhao J, Wang Z, Shi F. Identification of candidate genes associated with tubal origin of high-grade serous ovarian cancer. Oncol Lett 2018; 15:7769-7775. [PMID: 29731902 PMCID: PMC5920857 DOI: 10.3892/ol.2018.8346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 01/23/2018] [Indexed: 01/24/2023] Open
Abstract
Evidence indicates that high-grade serous ovarian carcinoma arises from the fallopian tube, rather than ovarian surface epithelium. This is termed the 'tubal origin' theory. The aim of the present study was to compare the immunophenotype and gene expression profiling among high-grade serous ovarian carcinoma (HGSOC), fallopian tube epithelium (FTE) and ovarian surface epithelium (OSE) based on tubal origin theory, and identify the differential genes associated with ovarian carcinogenesis. A total of 61 cases of fresh tissue samples including 21 cases of HGSOC, 20 cases of OSE, and 20 cases of FTE were obtained following surgical resection. Immunostaining was performed to detect the expression of PAX8, which has been considered as a potential immunophenotype marker of Müllerian origin. Illumina BeadChip was applied for gene expression profiling. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was performed to confirm the differential expression of candidate genes between HGSOC and FTE. The results of the present study demonstrated that PAX8 was highly expressed in HGSOC (19/21, 90.4%) and FTE (20/20, 100%), but not in OSE (3/20, 14.3%). A dendrogram generated by cluster analysis indicated a higher similarity of gene expression profile between HGSOC and FTE than OSE. A total of 2,412 differentially expressed genes were identified (absolute fold change >2) between HGSOC and FTE, including 822 upregulated genes in cancer and 1,590 downregulated genes. S100 calcium binding protein P, Ras-interacting protein 1, Wnt family member 5A, tumor-associated calcium signal transducer 2, Dickkopf Wnt signaling pathway inhibitor 3 and tumor suppressor candidate 3 genes were identified as candidate markers, of which the differential gene expression in HGSOC and FTE was confirmed by RT-qPCR (P<0.05). The results indicate the presence of a greater similarity in the immunophenotype and gene expression profile of HGSOC and FTE, when compared with OSE, which was consistent with the tubal origin theory of HGSOC.
Collapse
Affiliation(s)
- Li Xiang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Guohua Rong
- Department of Breast Surgery, Qingdao Municipal Hospital (Group), Qingdao, Shandong 266000, P.R. China
| | - Jing Zhao
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Zhenyan Wang
- Department of Gynecology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| | - Fengfeng Shi
- Department of Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, P.R. China
| |
Collapse
|
18
|
Jahangiri A, Rasooli I, Owlia P, Fooladi AAI, Salimian J. An integrative in silico approach to the structure of Omp33-36 in Acinetobacter baumannii. Comput Biol Chem 2018; 72:77-86. [DOI: 10.1016/j.compbiolchem.2018.01.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 01/07/2018] [Accepted: 01/10/2018] [Indexed: 01/01/2023]
|
19
|
Jahangiri A, Rasooli I, Owlia P, Imani Fooladi AA, Salimian J. Highly conserved exposed immunogenic peptides of Omp34 against Acinetobacter baumannii: An innovative approach. J Microbiol Methods 2018; 144:79-85. [DOI: 10.1016/j.mimet.2017.11.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 11/06/2017] [Accepted: 11/09/2017] [Indexed: 11/16/2022]
|
20
|
Karamariti E, Zhai C, Yu B, Qiao L, Wang Z, Potter CMF, Wong MM, Simpson RML, Zhang Z, Wang X, Del Barco Barrantes I, Niehrs C, Kong D, Zhao Q, Zhang Y, Hu Y, Zhang C, Xu Q. DKK3 (Dickkopf 3) Alters Atherosclerotic Plaque Phenotype Involving Vascular Progenitor and Fibroblast Differentiation Into Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2017; 38:425-437. [PMID: 29284609 DOI: 10.1161/atvbaha.117.310079] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Accepted: 12/13/2017] [Indexed: 01/31/2023]
Abstract
OBJECTIVE DKK3 (dickkopf 3), a 36-kD secreted glycoprotein, has been shown to be involved in the differentiation of partially reprogrammed cells and embryonic stem cells to smooth muscle cells (SMCs), but little is known about its involvement in vascular disease. This study aims to assess the effects of DKK3 on atherosclerotic plaque composition. APPROACH AND RESULTS In the present study, we used a murine model of atherosclerosis (ApoE-/-) in conjunction with DKK3-/- and performed tandem stenosis of the carotid artery to evaluate atherosclerotic plaque development. We found that the absence of DKK3 leads to vulnerable atherosclerotic plaques, because of a reduced number of SMCs and reduced matrix protein deposition, as well as increased hemorrhage and macrophage infiltration. Further in vitro studies revealed that DKK3 can induce differentiation of Sca1+ (stem cells antigen 1) vascular progenitors and fibroblasts into SMCs via activation of the TGF-β (transforming growth factor-β)/ATF6 (activating transcription factor 6) and Wnt signaling pathways. Finally, we assessed the therapeutic potential of DKK3 in mouse and rabbit models and found that DKK3 altered the atherosclerotic plaque content via increasing SMC numbers and reducing vascular inflammation. CONCLUSIONS Cumulatively, we provide the first evidence that DKK3 is a potent SMC differentiation factor, which might have a therapeutic effect in reducing intraplaque hemorrhage related to atherosclerotic plaque phenotype.
Collapse
Affiliation(s)
- Eirini Karamariti
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Chungang Zhai
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Baoqi Yu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Lei Qiao
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Zhihong Wang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Claire M F Potter
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Mei Mei Wong
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Russell M L Simpson
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Zhongyi Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Xiaocong Wang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Ivan Del Barco Barrantes
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Christof Niehrs
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Deling Kong
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Qiang Zhao
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Yun Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Yanhua Hu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.)
| | - Cheng Zhang
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.).
| | - Qingbo Xu
- From the School of Cardiovascular Medicine & Sciences, King's College London BHF Centre, United Kingdom (E.K., B.Y., C.M.F.P., M.M.W., R.M.L.S., Z.Z., X.W., Y.H., Q.X.); The Key Laboratory of Cardiovascular Remodelling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Shandong University, Jinan, China (C. Zhai, L.Q., Y.Z., C. Zhang); State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin, China (Z.W., D.K., Q.Z.); Division of Molecular Embryology, DKFZ-ZMBH Alliance, Heidelberg, Germany (I.d.B.B., C.N.); and Institute of Molecular Biology (IMB), Mainz, Germany (C.N.).
| |
Collapse
|
21
|
Antigenic Properties of Iron Regulated Proteins in Acinetobacter baumannii: An In Silico Approach. Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9665-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
22
|
Platonov ME, Borovjagin AV, Kaverina N, Xiao T, Kadagidze Z, Lesniak M, Baryshnikova M, Ulasov IV. KISS1 tumor suppressor restricts angiogenesis of breast cancer brain metastases and sensitizes them to oncolytic virotherapy in vitro. Cancer Lett 2017; 417:75-88. [PMID: 29269086 DOI: 10.1016/j.canlet.2017.12.024] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/13/2017] [Accepted: 12/14/2017] [Indexed: 12/12/2022]
Abstract
KISS1 tumor suppressor protein regulates cancer cell invasion via MMP9 metalloproteinase. Downregulation of KISS1 gene expression promotes progression of breast cancer and melanoma, resulting in the development of distant metastases. In the current study, we investigated whether restoration of KISS1 expression in KISS1-deficient human metastatic breast cancer cells holds potential as an advanced anticancer strategy. To this end we engineered an infectivity-enhanced conditionally-replicative human adenovirus type 5 encoding KISS1 as an "arming" transgene in the Ad5 E3 region for an ectopic KISS1 expression in transduced cancer cells. The oncolytic potential of the vector was examined using brain-invading metastatic clones of CN34 and MDA-MB-231 breast cancer cells, which supported high levels of AdKISS1 replication, correlating with a robust CRAd-mediated cytotoxicity. Secretion of cellular factors responsible for tumor angiogenesis, cell-to-cell communication and anti-tumoral immune responses upon KISS1 expression in breast cancer cells was analyzed by a RayBiotech Kiloplex Quantibody array. Overall, our results indicate that KISS1 transgene expression provides an important benefit for CRAd-mediated cytotoxicity in breast cancer cells and holds potential as an anticancer treatment in conjunction with oncolytic virotherapy of breast and other metastatic cancers.
Collapse
Affiliation(s)
- Mikhail E Platonov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia
| | - Anton V Borovjagin
- Institute of Oral Health Research, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Natalya Kaverina
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Ting Xiao
- Department of Neurological Surgery, Northwestern University, Chicago, 60611, USA
| | - Zaira Kadagidze
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Maciej Lesniak
- Department of Neurological Surgery, Northwestern University, Chicago, 60611, USA
| | - Marya Baryshnikova
- N.N. Blokhin Cancer Research Center, RAMN, Kashirskoe Shosse 23, Moscow, 115478, Russia
| | - Ilya V Ulasov
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| |
Collapse
|
23
|
In Silico Analysis for Determination and Validation of Human CD20 Antigen 3D Structure. Int J Pept Res Ther 2017. [DOI: 10.1007/s10989-017-9654-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
24
|
Singla H, Ludhiadch A, Kaur RP, Chander H, Kumar V, Munshi A. Recent advances in HER2 positive breast cancer epigenetics: Susceptibility and therapeutic strategies. Eur J Med Chem 2017; 142:316-327. [DOI: 10.1016/j.ejmech.2017.07.075] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/27/2017] [Accepted: 07/31/2017] [Indexed: 12/31/2022]
|
25
|
Asadi-Ghalehni M, Rasaee MJ, RajabiBazl M, Khosravani M, Motaghinejad M, Javanmardi M, Khalili S, Modjtahedi H, Sadroddiny E. A novel recombinant anti-epidermal growth factor receptor peptide vaccine capable of active immunization and reduction of tumor volume in a mouse model. Microbiol Immunol 2017; 61:531-538. [PMID: 29023946 DOI: 10.1111/1348-0421.12547] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 09/28/2017] [Accepted: 10/08/2017] [Indexed: 02/06/2023]
Abstract
Over-expression of epidermal growth factor receptor (EGFR) has been reported in a number of human malignancies. Strong expression of this receptor has been associated with poor survival in many such patients. Active immunizations that elicit antibodies of the desired type could be an appealing alternative to conventional passive immunization. In this regard, a novel recombinant peptide vaccine capable of prophylactic and therapeutic effects was constructed. A novel fusion recombinant peptide base vaccine consisting of L2 domain of murine extra-cellular domain-EGFR and EGFR mimotope (EM-L2) was constructed and its prophylactic and therapeutic effects in a Lewis lung carcinoma mouse (C57/BL6) model evaluated. Constructed recombinant peptide vaccine is capable of reacting with anti-EGFR antibodies. Immunization of mice with EM-L2 peptide resulted in antibody production against EM-L2. The constructed recombinant peptide vaccine reduced tumor growth and increased the survival rate. Designing effective peptide vaccines could be an encouraging strategy in contemporary cancer immunotherapy. Investigating the efficacy of such cancer immunotherapy approaches may open exciting possibilities concerning hyperimmunization, leading to more promising effects on tumor regression and proliferation.
Collapse
Affiliation(s)
- Majid Asadi-Ghalehni
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Javad Rasaee
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Masoumeh RajabiBazl
- Department of Clinical Biochemistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masood Khosravani
- Department of Nanomedicine, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Motaghinejad
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Masoud Javanmardi
- Department of Medical Biotechnology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saeed Khalili
- Department of Laboratory Sciences, School of Paramedical, Dezful University of Medical Sciences, Dezful, Iran
| | - Helmout Modjtahedi
- Department of Life Sciences, Faculty of Science, Engineering and Computing, Kingston University, London, UK
| | - Esmaeil Sadroddiny
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Luo S, Li N, Yu S, Chen L, Liu C, Rong J. MicroRNA-92a promotes cell viability and invasion in cervical cancer via directly targeting Dickkopf-related protein 3. Exp Ther Med 2017; 14:1227-1234. [PMID: 28810582 DOI: 10.3892/etm.2017.4586] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 03/10/2017] [Indexed: 12/18/2022] Open
Abstract
MicroRNA-92a (miR-92a) was recently reported to have an oncogenic role in cervical cancer; however, the underlying mechanism remains largely unclear. The present study aimed to investigate the expression, clinical significance and regulatory mechanism of miR-92a in cervical cancer. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) data indicated that miR-92a was significantly upregulated in cervical cancer tissues compared with matched adjacent non-tumor tissues (P<0.01). High expression of miR-92a was significantly associated with poor differentiation (P=0.031), advanced clinical stage (P=0.011) and lymph node metastasis (P=0.014), but not associated with age, tumor size and distant metastasis. Knockdown of miR-92a significantly inhibited the viability and invasion of cervical cancer HeLa cells, while overexpression of miR-92a significantly enhanced HeLa cell viability and invasion (P<0.01). Luciferase reporter assay identified Dickkopf-related protein 3 (DKK3) as a target gene of miR-92a, and the protein expression of DKK3 was negatively regulated by miR-92a in HeLa cells. Furthermore, overexpression of DKK3 significantly eliminated the stimulative effects of miR-92a on HeLa cell viability and invasion (P<0.01). Additionally, DKK3 was significantly downregulated in cervical cancer tissues compared with adjacent non-tumor tissues (P<0.01), inversely correlated to the miR-92a levels in cervical cancer tissues (P<0.01). In summary, the present study indicated that miR-92a promotes cell viability and invasion in cervical cancer, partly at least, via inhibiting the protein expression of DKK3. Therefore, the present study highlights the clinical significance of the miR-92a/DKK3 axis in cervical cancer.
Collapse
Affiliation(s)
- Shengtian Luo
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| | - Na Li
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| | - Shaohua Yu
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| | - Lichun Chen
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| | - Chunying Liu
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| | - Jiawei Rong
- Department of Obstetrics and Gynecology, General Hospital of Daqing Oil Field, Daqing, Heilongjiang 163001, P.R. China
| |
Collapse
|
27
|
Jahangiri A, Rasooli I, Owlia P, Fooladi AAI, Salimian J. In silico design of an immunogen against Acinetobacter baumannii based on a novel model for native structure of Outer membrane protein A. Microb Pathog 2017; 105:201-210. [DOI: 10.1016/j.micpath.2017.02.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 02/05/2017] [Accepted: 02/20/2017] [Indexed: 11/17/2022]
|
28
|
Poorebrahim M, Sadeghi S, Rahimi H, Karimipoor M, Azadmanesh K, Mazlomi MA, Teimoori-Toolabi L. Rational design of DKK3 structure-based small peptides as antagonists of Wnt signaling pathway and in silico evaluation of their efficiency. PLoS One 2017; 12:e0172217. [PMID: 28234935 PMCID: PMC5325476 DOI: 10.1371/journal.pone.0172217] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022] Open
Abstract
Dysregulated Wnt signaling pathway is highly associated with the pathogenesis of several human cancers. Dickkopf proteins (DKKs) are thought to inhibit Wnt signaling pathway through binding to lipoprotein receptor-related protein (LRP) 5/6. In this study, based on the 3-dimensional (3D) structure of DKK3 Cys-rich domain 2 (CRD2), we have designed and developed several peptide inhibitors of Wnt signaling pathway. Modeller 9.15 package was used to predict 3D structure of CRD2 based on the Homology modeling (HM) protocol. After refinement and minimization with GalaxyRefine and NOMAD-REF servers, the quality of selected models was evaluated utilizing VADAR, SAVES and ProSA servers. Molecular docking studies as well as literature-based information revealed two distinct boxes located at CRD2 which are actively involved in the DKK3-LRP5/6 interaction. A peptide library was constructed conducting the backrub sequence tolerance scanning protocol in Rosetta3.5 according to the DKK3-LRP5/6 binding sites. Seven tolerated peptides were chosen and their binding affinity and stability were improved by some logical amino acid substitutions. Molecular dynamics (MD) simulations of peptide-LRP5/6 complexes were carried out using GROMACS package. After evaluation of binding free energies, stability, electrostatic potential and some physicochemical properties utilizing computational approaches, three peptides (PEP-I1, PEP-I3 and PEP-II2) demonstrated desirable features. However, all seven improved peptides could sufficiently block the Wnt-binding site of LRP6 in silico. In conclusion, we have designed and improved several small peptides based on the LRP6-binding site of CRD2 of DKK3. These peptides are highly capable of binding to LRP6 in silico, and may prevent the formation of active Wnt-LRP6-Fz complex.
Collapse
Affiliation(s)
- Mansour Poorebrahim
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Solmaz Sadeghi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | - Hamzeh Rahimi
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| | | | | | - Mohammad Ali Mazlomi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ladan Teimoori-Toolabi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Molecular Medicine Department, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
29
|
Structural pierce into molecular mechanism underlying Clostridium perfringens Epsilon toxin function. Toxicon 2017; 127:90-99. [PMID: 28089770 DOI: 10.1016/j.toxicon.2017.01.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 01/09/2017] [Accepted: 01/10/2017] [Indexed: 12/17/2022]
Abstract
Epsilon toxin of the Clostridium perfringens garnered a lot of attention due to its potential for toxicity in humans, extreme potency for cytotoxicity in mice and lack of any approved therapeutics prescribed for human. However, the intricacies of the Epsilon toxin action mechanism are yet to be understood. In this regard, various in silico tools have been exploited to model and refine the 3D structure of the toxin and its two receptors. The receptor proteins were embedded into designed lipid membranes within an aqueous and ionized environment. Thereafter, the modeled structures subjected to series of consecutive molecular dynamics runs to achieve the most natural like coordination for each model. Ultimately, protein-protein interaction analyses were performed to understand the probable action mechanism. The obtained results successfully confirmed the accuracy of employed methods to achieve high quality models for the toxin and its receptors within their lipid bilayers. Molecular dynamics analyses lead the structures to a more native like coordination. Moreover, the results of previous empirical studies were confirmed, while new insights for action mechanisms including the detailed roles of Hepatitis A virus cellular receptor 1 (HAVCR1) and Myelin and lymphocyte protein (MAL) proteins were achieved. In light of previous and our observations, we suggested novel models which elucidated the existing interplay between potential players of Epsilon toxin action mechanism with detailed structural evidences. These models would pave the way to have more robust understanding of the Epsilon toxin biology, more precise vaccine construction and more successful drug (inhibitor) design.
Collapse
|
30
|
Irradiation enhances susceptibility of tumor cells to the antitumor effects of TNF-α activated adipose derived mesenchymal stem cells in breast cancer model. Sci Rep 2016; 6:28433. [PMID: 27329316 PMCID: PMC4916474 DOI: 10.1038/srep28433] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 06/03/2016] [Indexed: 12/14/2022] Open
Abstract
Gene modified or cytokine activated mesenchymal stem cells (MSCs) have been used as a treatment in various types of cancer. Moreover, irradiation is usually applied as either a standard primary or adjuvant therapy. Here, we showed that the expression of TNF related apoptosis-inducing ligand (TRAIL) and Dickouf-3 (Dkk-3), the promising anticancer proteins, increased in murine adipose-derived mesenchymal stromal cells (AD-MSCs) following activation with TNF-α, resulting in the induction of apoptosis in cancer cells. Also, anticancer effects of TNF-α activated AD-MSCs were intensified with irradiation. In vivo results showed that TNF-α preactivated AD-MSCs combined with irradiation decreased tumor size and increased survival rate in tumor bearing mice. On the other hands, both TNF-α preactivated AD-MSCs with or without irradiation prevented metastasis in ling and liver, and increased apoptosis in tumor mass. Finally, flowcytometry assay demonstrated that naïve AD-MSCs combined with irradiation but not TNF-α activated MSCs with irradiation increased Treg population in lymph node and spleen. Altogether, obtained results suggest that TNF-α activated MSCs combined with irradiation therapy can serve as new strategy in breast cancer therapy.
Collapse
|
31
|
Mohammadpour H, Fekrazad R. Antitumor effect of combined Dkk-3 and 5-ALA mediated photodynamic therapy in breast cancer cell’s colony. Photodiagnosis Photodyn Ther 2016; 14:200-3. [DOI: 10.1016/j.pdpdt.2016.04.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 03/29/2016] [Accepted: 04/05/2016] [Indexed: 11/15/2022]
|