1
|
Du J, Wang J, Ge F, Ma H, Zhu H, Du J, Yan F, He Q, Yang B, Yuan T, Zhu H. JOSD2 promotes breast cancer metastasis by deubiquitinating and stabilizing SMAD4. Biochem Pharmacol 2025; 232:116748. [PMID: 39793716 DOI: 10.1016/j.bcp.2025.116748] [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: 09/30/2024] [Revised: 12/03/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Breast cancer is one of the most common malignant tumors among women worldwide, and its high degree of metastasis significantly impacts treatment effectiveness leading to poor prognosis. The potential molecular mechanisms underlying breast cancer metastasis remain to be further elucidated. In this study, via database analysis, we revealed that the deubiquitinase josephin domain containing 2 (JOSD2) was abnormally amplified in patients with metastatic breast cancer, and was significantly negatively correlated with patient prognosis. By integrating data from the Gene Expression Omnibus (GEO) database and Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathway enrichment analysis, we found that the transforming growth factor beta (TGF-β) signaling pathway was significantly activated in breast cancer patients with increased JOSD2 expression. Further studies revealed that JOSD2 interacted with and stabilized SMAD family member 4 (SMAD4) by removing polyubiquitin chains. Inhibition of JOSD2 by RNA interference effectively inhibited the metastasis of breast cancer cells both in vitro and in vivo. In conclusion, our study not only reveals the role of JOSD2 in promoting breast cancer metastasis for the first time, but also indicates promising directions for the future development of deubiquitinase inhibitors, which could yield significant therapeutic benefits. Nevertheless, extensive research and development are required to fully realize this potential.
Collapse
Affiliation(s)
- Jiamin Du
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiao Wang
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fujing Ge
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongrui Ma
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Hongdao Zhu
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiangxia Du
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, Hangzhou, Zhejiang, China
| | - Fangjie Yan
- Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, Hangzhou, Zhejiang, China
| | - Bo Yang
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Tao Yuan
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Hong Zhu
- Institute of Pharmacology & Toxicology Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China; Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
2
|
Tian Y, Guo J, Mao L, Chen Z, Zhang X, Li Y, Zhang Y, Zha X, Luo OJ. Single-cell dissection reveals promotive role of ENO1 in leukemia stem cell self-renewal and chemoresistance in acute myeloid leukemia. Stem Cell Res Ther 2024; 15:347. [PMID: 39380054 PMCID: PMC11463110 DOI: 10.1186/s13287-024-03969-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/30/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND Quiescent self-renewal of leukemia stem cells (LSCs) and resistance to conventional chemotherapy are the main factors leading to relapse of acute myeloid leukemia (AML). Alpha-enolase (ENO1), a key glycolytic enzyme, has been shown to regulate embryonic stem cell differentiation and promote self-renewal and malignant phenotypes in various cancer stem cells. Here, we sought to test whether and how ENO1 influences LSCs renewal and chemoresistance within the context of AML. METHODS We analyzed single-cell RNA sequencing data from bone marrow samples of 8 relapsed/refractory AML patients and 4 healthy controls using bioinformatics and machine learning algorithms. In addition, we compared ENO1 expression levels in the AML cohort with those in 37 control subjects and conducted survival analyses to correlate ENO1 expression with clinical outcomes. Furthermore, we performed functional studies involving ENO1 knockdown and inhibition in AML cell line. RESULTS We used machine learning to model and infer malignant cells in AML, finding more primitive malignant cells in the non-response (NR) group. The differentiation capacity of LSCs and progenitor malignant cells exhibited an inverse correlation with glycolysis levels. Trajectory analysis indicated delayed myeloid cell differentiation in NR group, with high ENO1-expressing LSCs at the initial stages of differentiation being preserved post-treatment. Simultaneously, ENO1 and stemness-related genes were upregulated and co-expressed in malignant cells during early differentiation. ENO1 level in our AML cohort was significantly higher than the controls, with higher levels in NR compared to those in complete remission. Knockdown of ENO1 in AML cell line resulted in the activation of LSCs, promoting cell differentiation and apoptosis, and inhibited proliferation. ENO1 inhibitor can impede the proliferation of AML cells. Furthermore, survival analyses associated higher ENO1 expression with poorer outcome in AML patients. CONCLUSIONS Our findings underscore the critical role of ENO1 as a plausible driver of LSC self-renewal, a potential target for AML target therapy and a biomarker for AML prognosis.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/genetics
- Phosphopyruvate Hydratase/metabolism
- Phosphopyruvate Hydratase/genetics
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Tumor Suppressor Proteins/metabolism
- Tumor Suppressor Proteins/genetics
- Female
- Drug Resistance, Neoplasm
- Single-Cell Analysis
- DNA-Binding Proteins/metabolism
- DNA-Binding Proteins/genetics
- Male
- Middle Aged
- Cell Self Renewal
- Adult
- Cell Line, Tumor
- Cell Differentiation
- Aged
- Biomarkers, Tumor
Collapse
Affiliation(s)
- Yun Tian
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jiafan Guo
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Lipeng Mao
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Zhixi Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xingwei Zhang
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yikai Zhang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, Jinan University, Guangzhou, 510632, China.
- Department of Hematology, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
- Guangdong Provincial Key Laboratory of Spine and Spinal Cord Reconstruction, The Fifth Affiliated Hospital of Jinan University (Heyuan Shenhe People's Hospital), Jinan University, Heyuan, 517000, China.
| | - Xianfeng Zha
- Department of Clinical Laboratory, First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
3
|
Mohammadi Azad Z, Moosazadeh Moghaddam M, Fasihi-Ramandi M, Haghighat S, Mirnejad R. Evaluation of the effect of Helicobacter pylori -derived OMVs and released exosomes from stomach cells treated with OMVs on the expression of genes related to the TGF-β/SMAD signaling pathway in hepatocellular carcinoma. J Recept Signal Transduct Res 2024; 44:181-190. [PMID: 39628127 DOI: 10.1080/10799893.2024.2436461] [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: 07/25/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
OMVs derived from Helicobacter pylori can lead to cell transformation in gastric epithelium and cancer. Additionally, exosomes (Exos) released by host cells infected with H. pylori can significantly contribute to the development of diseases such as cancer. In this study, the effects of both Exos from AGS cells treated with H. pylori-derived OMVs on the expression of genes related to the TGF-β/SMAD signaling pathway in hepatocellular carcinoma (HCC) cells were investigated. The TGF-β/SMAD pathway is one of the most important pathways that regulate the development and progression of HCC. For this purpose, after treating HepG2 cells with H. pylori-derived OMVs (directly) and Exos from AGS cells treated with H. pylori-derived OMVs (indirectly), the expression levels of TGF-β, SMAD2, SMAD3, SMAD4, and ERK genes were analyzed using Real-time PCR. The findings showed that OMVs derived from H. pylori can significantly increase the expression of genes involved in the TGF-β signaling pathway, which can affect the aggressive behavior of HepG2 cells. Additionally, exosomes secreted from AGS cells or AGS cells treated with OMVs had no effect on changing the expression of the studied genes. Therefore, only the OMVs released from H. pylori can affect the TGF-β/SMAD signaling pathway in HCC cells.
Collapse
Affiliation(s)
- Zohreh Mohammadi Azad
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran Iran
| | - Mehrdad Moosazadeh Moghaddam
- Tissue Engineering and Regenerative Medicine Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Mahdi Fasihi-Ramandi
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Setareh Haghighat
- Department of Microbiology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran Iran
| | - Reza Mirnejad
- Molecular Biology Research Center, Biomedicine Technologies Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Luo B, Zheng R, Shi C, Chen D, Jin X, Hou J, Xu G, Hu B. DACT2 modulates atrial fibrillation through TGF/β and Wnt signaling pathways. Heliyon 2024; 10:e36050. [PMID: 39224277 PMCID: PMC11367123 DOI: 10.1016/j.heliyon.2024.e36050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia that seriously affects the quality of life of patients. Effective treatment and prevention are important to control the morbidity and mortality of AF. It has been found that cardiac fibrosis promotes the onset and progression of AF. It is now known that transforming growth factor β (TGF-β), an important fibrotic cytokine, plays an important role in cardiac fibrosis by inducing myofibroblast activation via the activation of classical (SMAD-based) and non-classical (non-SMAD-based) signaling pathways. In addition, specific activation of the Wnt/β-catenin pathway has been shown to promote the transformation of fibroblasts into myofibroblasts. In recent years, a new family of proteins, namely Disheveled-associated antagonist of beta-catenin (DACT) 2, can affect the Wnt/β-catenin and TGF-β signaling pathways by regulating the phosphorylation levels of these target proteins, which in turn affects the progression of fibrosis. The present study focuses on the effect of DACT2-guided β-catenin on atrial fibrosis. It is expected that the summarized information can be helpful in the treatment of AF.
Collapse
Affiliation(s)
- Bairu Luo
- Department of Clinical Pathology, Jiaxing University Master Degree Cultivation Base, Jiaxing Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Jiaxing, 314001, ZJ, China
| | - Rui Zheng
- Department of Clinical Pathology, The 3rd Clinical Medical College Affiliated to Zhejiang Chinese Medical University, Hangzhou, 310053, ZJ, China
| | - Chaoqun Shi
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Deqing Chen
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Xin Jin
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Jian Hou
- Department of Cardiology, The Affiliated Panyu Central Hospital of Guangzhou Medical University, Guangzhou, 510080, GD, China
| | - Guangtao Xu
- Department of Pathology, Institute of Forensic Science, Jiaxing University, Jiaxing, 314001, ZJ, China
| | - Bo Hu
- Department of Clinical Pathology, Jiaxing Hospital of Traditional Chinese Medicine, the 3rd Clinical Medical College Affiliated to Zhejiang Chinese Medical University, Jiaxing, 314001, ZJ, China
| |
Collapse
|
5
|
Babic T, Ugrin M, Jeremic S, Kojic M, Dinic J, Banovic Djeri B, Zoidakis J, Nikolic A. Dysregulation of transcripts SMAD4-209 and SMAD4-213 and their respective promoters in colon cancer cell lines. J Cancer 2024; 15:5118-5131. [PMID: 39132157 PMCID: PMC11310865 DOI: 10.7150/jca.98911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 07/18/2024] [Indexed: 08/13/2024] Open
Abstract
Background: The pervasive role of alternative promoters in context-specific isoform expression and the importance of promoter choice over its level of transcriptional activity have been recently implied based on pan-cancer in silico studies. We aimed to explore this phenomenon at the cellular level on the example of a major tumor suppressor SMAD4 in search of molecular mechanisms in colorectal cancer that could be exploited for novel biomarkers or therapeutic approaches. Methods: Multi-omics technologies, in silico tools and in vitro functional assays were applied to analyze the transcripts expression and the alternative promoters' function of the SMAD4 gene in colon cell lines HCEC-1CT, HCT116, DLD-1, SW480 and SW620. Results: High expression of the transcript SMAD4-213 emerged as a hallmark of colon cancer cells, while in silico tools point to its possible additional role and potential for sponging miRNAs. Based on the observed dysregulation of SMAD4-209 and SMAD4-213 in malignant vs. non-malignant colon cells, we propose that their expression ratio might be a solid biomarker candidate for colorectal cancer detection. Conclusions: A differential pattern of the respective promoters' activity was observed that corresponds to the expression of transcripts, confirming the role of alternative promoters in context-specific isoform expression. The investigated SMAD4 promoters and transcripts harbor translational potential that should be further investigated.
Collapse
Affiliation(s)
- Tamara Babic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| | - Milena Ugrin
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| | - Sanja Jeremic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| | - Milan Kojic
- Institute of Virology, Vaccines and Sera “Torlak”, 11152 Belgrade, Serbia
| | - Jelena Dinic
- Institute for Biological Research “Siniša Stanković” — National Institute of the Republic of Serbia, University of Belgrade, 11060 Belgrade, Serbia
| | - Bojana Banovic Djeri
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| | - Jerome Zoidakis
- Department of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
- Proteomics Laboratory, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece
| | - Aleksandra Nikolic
- Institute of Molecular Genetics and Genetic Engineering, University of Belgrade, 11042 Belgrade, Serbia
| |
Collapse
|
6
|
Niu B, Tian T, Wang L, Tian Y, Tian T, Guo Y, Zhou H, Zhang Z. CCL9/CCR1 axis-driven chemotactic nanovesicles for attenuating metastasis of SMAD4-deficient colorectal cancer by trapping TGF- β. Acta Pharm Sin B 2024; 14:3711-3729. [PMID: 39220887 PMCID: PMC11365421 DOI: 10.1016/j.apsb.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 04/09/2024] [Accepted: 04/15/2024] [Indexed: 09/04/2024] Open
Abstract
SMAD4 deficiency in colorectal cancer (CRC) is highly correlated with liver metastasis and high mortality, yet there are few effective precision therapies available. Here, we show that CCR1+-granulocytic myeloid-derived suppressor cells (G-MDSCs) are highly infiltrated in SMAD4-deficient CRC via CCL15/CCR1 and CCL9/CCR1 axis in clinical specimens and mouse models, respectively. The excessive TGF-β, secreted by tumor-infiltrated CCR1+-G-MDSCs, suppresses the immune response of cytotoxic T lymphocytes (CTLs), thus facilitating metastasis. Hereby, we develop engineered nanovesicles displaying CCR1 and TGFBR2 molecules (C/T-NVs) to chemotactically target the tumor driven by CCL9/CCR1 axis and trap TGF-β through TGF-β-TGFBR2 specific binding. Chemotactic C/T-NVs counteract CCR1+-G-MDSC infiltration through competitive responding CCL9/CCR1 axis. C/T-NVs-induced intratumoral TGF-β exhaustion alleviates the TGF-β-suppressed immune response of CTLs. Collectively, C/T-NVs attenuate liver metastasis of SMAD4-deficient CRC. In further exploration, high expression of programmed cell death ligand-1 (PD-L1) is observed in clinical specimens of SMAD4-deficient CRC. Combining C/T-NVs with anti-PD-L1 antibody (aPD-L1) induces tertiary lymphoid structure formation with sustained activation of CTLs, CXCL13+-CD4+ T, CXCR5+-CD20+ B cells, and enhanced secretion of cytotoxic cytokine interleukin-21 and IFN-γ around tumors, thus eradicating metastatic foci. Our strategy elicits pleiotropic antimetastatic immunity, paving the way for nanovesicle-mediated precision immunotherapy in SMAD4-deficient CRC.
Collapse
Affiliation(s)
- Boning Niu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tianyi Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yinmei Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tian Tian
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yuanyuan Guo
- Department of Pharmacy, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hu Zhou
- School of Pharmaceutical Sciences, Fujian Provincial Key Laboratory of Innovative Drug Target Research, High Throughput Drug Screening Platform, Xiamen University, Xiamen 361102, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
- National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Engineering Research Centre for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
7
|
Forte G, Buonadonna AL, Fasano C, Sanese P, Cariola F, Manghisi A, Guglielmi AF, Lepore Signorile M, De Marco K, Grossi V, Disciglio V, Simone C. Clinical and Molecular Characterization of SMAD4 Splicing Variants in Patients with Juvenile Polyposis Syndrome. Int J Mol Sci 2024; 25:7939. [PMID: 39063183 PMCID: PMC11276957 DOI: 10.3390/ijms25147939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Juvenile polyposis syndrome (JPS) is an inherited autosomal dominant condition that predisposes to the development of juvenile polyps throughout the gastrointestinal (GI) tract, and it poses an increased risk of GI malignancy. Germline causative variants were identified in the SMAD4 gene in a subset (20%) of JPS cases. Most SMAD4 germline genetic variants published to date are missense, nonsense, and frameshift mutations. SMAD4 germline alterations predicted to result in aberrant splicing have rarely been reported. Here, we report two unrelated Italian families harboring two different SMAD4 intronic variants, c.424+5G>A and c.425-9A>G, which are clinically associated with colorectal cancer and/or juvenile GI polyps. In silico prediction analysis, in vitro minigene assays, and RT-PCR showed that the identified variants lead to aberrant SMAD4 splicing via the exonization of intronic nucleotides, resulting in a premature stop codon. This is expected to cause the production of a truncated protein. This study expands the landscape of SMAD4 germline genetic variants associated with GI polyposis and/or cancer. Moreover, it emphasizes the importance of the functional characterization of SMAD4 splicing variants through RNA analysis, which can provide new insights into genetic disease variant interpretation, enabling tailored genetic counseling, management, and surveillance of patients with GI polyposis and/or cancer.
Collapse
Affiliation(s)
- Giovanna Forte
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Antonia Lucia Buonadonna
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Candida Fasano
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Paola Sanese
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Filomena Cariola
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Andrea Manghisi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Anna Filomena Guglielmi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Martina Lepore Signorile
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Katia De Marco
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Valentina Grossi
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Vittoria Disciglio
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
| | - Cristiano Simone
- Medical Genetics, National Institute of Gastroenterology-IRCCS “Saverio de Bellis” Research Hospital, 70013 Castellana Grotte, Italy; (G.F.); (A.L.B.); (C.F.); (P.S.); (F.C.); (A.M.); (A.F.G.); (M.L.S.); (K.D.M.); (V.G.)
- Medical Genetics, Department of Precision and Regenerative Medicine and Jonic Area (DiMePRe-J), University of Bari Aldo Moro, 70124 Bari, Italy
| |
Collapse
|
8
|
Zohud O, Midlej K, Lone IM, Nashef A, Abu-Elnaaj I, Iraqi FA. Studying the Effect of the Host Genetic Background of Juvenile Polyposis Development Using Collaborative Cross and Smad4 Knock-Out Mouse Models. Int J Mol Sci 2024; 25:5812. [PMID: 38891999 PMCID: PMC11172477 DOI: 10.3390/ijms25115812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/17/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
Juvenile polyposis syndrome (JPS) is a rare autosomal dominant disorder characterized by multiple juvenile polyps in the gastrointestinal tract, often associated with mutations in genes such as Smad4 and BMPR1A. This study explores the impact of Smad4 knock-out on the development of intestinal polyps using collaborative cross (CC) mice, a genetically diverse model. Our results reveal a significant increase in intestinal polyps in Smad4 knock-out mice across the entire population, emphasizing the broad influence of Smad4 on polyposis. Sex-specific analyses demonstrate higher polyp counts in knock-out males and females compared to their WT counterparts, with distinct correlation patterns. Line-specific effects highlight the nuanced response to Smad4 knock-out, underscoring the importance of genetic variability. Multimorbidity heat maps offer insights into complex relationships between polyp counts, locations, and sizes. Heritability analysis reveals a significant genetic basis for polyp counts and sizes, while machine learning models, including k-nearest neighbors and linear regression, identify key predictors, enhancing our understanding of juvenile polyposis genetics. Overall, this study provides new information on understanding the intricate genetic interplay in the context of Smad4 knock-out, offering valuable insights that could inform the identification of potential therapeutic targets for juvenile polyposis and related diseases.
Collapse
Affiliation(s)
- Osayd Zohud
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Kareem Midlej
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Iqbal M. Lone
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| | - Aysar Nashef
- Department of Oral and Maxillofacial Surgery, Baruch Padeh Medical Center, Poriya Tebaria 42310, Israel; (A.N.); (I.A.-E.)
- Department of Oral and Maxillofacial Surgery, Meir Medical Center, Faculty of Medicine and Health Sciences, Tel-Aviv University, Kfar-Saba 69978, Israel
| | - Imad Abu-Elnaaj
- Department of Oral and Maxillofacial Surgery, Baruch Padeh Medical Center, Poriya Tebaria 42310, Israel; (A.N.); (I.A.-E.)
- Azrieli Faculty of Medicine, Bar-Ilan University, Tsaft 33241, Israel
| | - Fuad A. Iraqi
- Department of Clinical Microbiology and Immunology, Faculty of Medicine and Health Sciences, Tel-Aviv University, Tel-Aviv 69978, Israel; (O.Z.); (K.M.); (I.M.L.)
| |
Collapse
|
9
|
Xu Y, Yang Y, Fan S. Research progress on the dermatomyositis specific autoantibodies and malignancy associated dermatomyositis. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2023; 48:1890-1898. [PMID: 38448383 PMCID: PMC10930743 DOI: 10.11817/j.issn.1672-7347.2023.220594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Indexed: 03/08/2024]
Abstract
Dermatomyositis (DM) is an autoimmune disease often complicated with malignant tumors. More than 50% of DM patients have myositis specific autoantibodies in their bodies. DM specific autoantibodies [including anti-migration inhibitory factor (Mi)-2 antibody, anti-nuclear matrix protein (NXP)-2 antibody, anti-transcription intermediary factor (TIF) 1-γ antibody, and anti-small ubiquitin like modifier activating enzyme (SAE) antibody] play important roles in the pathogenesis of malignancy associated DM. Revealing the role of DM specific autoantibodies in the development of malignant tumors in DM patients can provide important evidence for accurately assessing the risk of developing malignant tumors in DM patients, and also provide new ideas for clinical diagnosis of DM and precise treatment.
Collapse
Affiliation(s)
- Yue Xu
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| | - Yang Yang
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Songqing Fan
- Department of Pathology, Second Xiangya Hospital, Central South University, Changsha 410011, China.
| |
Collapse
|
10
|
Wang H, Su Y, Chen D, Li Q, Shi S, Huang X, Fang M, Yang M. Advances in the mechanisms and applications of inhibitory oligodeoxynucleotides against immune-mediated inflammatory diseases. Front Pharmacol 2023; 14:1119431. [PMID: 36825156 PMCID: PMC9941346 DOI: 10.3389/fphar.2023.1119431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/26/2023] [Indexed: 02/09/2023] Open
Abstract
Inhibitory oligodeoxynucleotides (ODNs) are short single-stranded DNA, which capable of folding into complex structures, enabling them to bind to a large variety of targets. With appropriate modifications, the inhibitory oligodeoxynucleotides exhibited many features of long half-life time, simple production, low toxicity and immunogenicity. In recent years, inhibitory oligodeoxynucleotides have received considerable attention for their potential therapeutic applications in immune-mediated inflammatory diseases (IMIDs). Inhibitory oligodeoxynucleotides could be divided into three categories according to its mechanisms and targets, including antisense ODNs (AS-ODNs), DNA aptamers and immunosuppressive ODNs (iSup ODNs). As a synthetic tool with immunomodulatory activity, it can target RNAs or proteins in a specific way, resulting in the reduction, increase or recovery of protein expression, and then regulate the state of immune activation. More importantly, inhibitory oligodeoxynucleotides have been used to treat immune-mediated inflammatory diseases, including inflammatory disorders and autoimmune diseases. Several inhibitory oligodeoxynucleotide drugs have been developed and approved on the market already. These drugs vary in their chemical structures, action mechanisms and cellular targets, but all of them could be capable of inhibiting excessive inflammatory responses. This review summarized their chemical modifications, action mechanisms and applications of the three kinds of inhibitory oligodeoxynucleotidesin the precise treatment of immune-mediated inflammatory diseases.
Collapse
Affiliation(s)
- Hongrui Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Yingying Su
- Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Duoduo Chen
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Qi Li
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Shuyou Shi
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Xin Huang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Mingli Fang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,*Correspondence: Mingli Fang, ; Ming Yang,
| | - Ming Yang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China,*Correspondence: Mingli Fang, ; Ming Yang,
| |
Collapse
|
11
|
Bauer AH, Basta DW, Hornick JL, Dong F. Loss of function SMAD4 nonstop mutations in human cancer. Histopathology 2023; 82:1098-1104. [PMID: 36740808 DOI: 10.1111/his.14880] [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: 11/08/2022] [Revised: 01/18/2023] [Accepted: 02/02/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND SMAD4 is a tumour suppressor gene that is mutated in a variety of cancers. SMAD4 nonstop mutations, which convert stop codons to sense codons that extend transcription, have been identified in genomic databases but have not been characterised in human pathology samples. The frequency of SMAD4 nonstop mutations and the consequences of nonstop mutations on SMAD4 protein expression are unknown. METHODS We retrospectively analysed our cancer sequencing database of 38,002 tumour specimens and evaluated the spectrum of SMAD4 mutations. SMAD4 protein expression was evaluated by immunohistochemistry in tumours with SMAD4 nonstop mutations. RESULTS In total, 1956 SMAD4 mutations were identified in 1822 tumours. SMAD4 mutations were most common in tumours of the gastrointestinal tract and included nonsense variants (n = 344), frameshift indels (n = 258), splice site variants (n = 104), and missense variants at codon R361 (n = 245). In a subset of cases with immunohistochemistry, SMAD4 expression was lost in 23 of 25 tumours (92%) with protein truncating variants and in 7 of 27 tumours (26%) with missense variants. Four cases harboured SMAD4 nonstop mutations. SMAD4 nonstop mutations were identified in two pancreatic adenocarcinomas, one colonic adenocarcinoma, and one non-small cell lung carcinoma. Immunohistochemistry demonstrated loss of SMAD4 protein expression in each of the four tumours with SMAD4 nonstop mutations. CONCLUSION SMAD4 nonstop mutations are associated with loss of SMAD4 protein expression in multiple tumour types. SMAD4 nonstop mutations should be clinically interpreted as pathogenic loss of function alterations when identified in cancer sequencing panels.
Collapse
Affiliation(s)
- Anna H Bauer
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,University of Missouri School of Medicine, Columbia, MO, USA
| | - David W Basta
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason L Hornick
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Fei Dong
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Nikolic A, Despotovic J, Babic T, Antic J, Markovic S, Krivokapic Z, Radojkovic D. SMAD4 Gene Analysis in Patients with Early Onset Colorectal Cancer: A Pilot Study. CYTOL GENET+ 2022. [DOI: 10.3103/s0095452722030082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
13
|
Song H, Huang Y, Jiang X. Mutation spectrum associated with metastasis of advanced cholangiocarcinoma. J Int Med Res 2022; 50:3000605221102080. [PMID: 35726602 PMCID: PMC9218467 DOI: 10.1177/03000605221102080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Background The mutations associated with metastasis in advanced-stage cholangiocarcinoma (CCA) have not been investigated. Objective To explore mutations in patients with advanced CCA and independent factors related to metastasis. Methods This retrospective study performed next-generation sequencing of tumor specimens from patients with advanced CCA treated between January 2017 and December 2019. Tumor mutational burden (TMB), microsatellite instability, and programmed cell death ligand (PD-L)1 positivity were determined. Factors independently associated with metastasis were explored via logistic regression. Results Ninety-one patients were included in this study. TP53 mutation frequencies were significantly higher in extrahepatic than intrahepatic CCA, while ARID1A mutations were significantly more frequent in intrahepatic CCA. Mutation frequencies in six selected genes did not differ according to patient age or sex. SMAD4 mutations were significantly less frequent in stage IV cancer; ARID1A and PBRM1 mutation frequencies were significantly higher in TMB >10 tumors. PBRM1 mutation frequencies were significantly higher in PD-L1-positive tumors, but lower in patients with metastasis. Multivariable analysis showed that a history of biliary surgery, SMAD4 mutations, and PBRM1 mutations were independently associated with CCA metastasis. Conclusions A history of biliary surgery and mutations in SMAD4 and PBRM1 are independent protective factors for metastasis in patients with advanced CCA.
Collapse
Affiliation(s)
- Hao Song
- Department of Organ Transplantation, Eastern Hepatobiliary Surgery Hospital Affiliated to Shanghai Second Military Medical University, Shanghai, China
| | - Yao Huang
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Xiaoqing Jiang
- Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital Affiliated to Shanghai Second Military Medical University, Shanghai, China
| |
Collapse
|
14
|
Rajabi S, Alix-Panabières C, Alaei AS, Abooshahab R, Shakib H, Ashrafi MR. Looking at Thyroid Cancer from the Tumor-Suppressor Genes Point of View. Cancers (Basel) 2022; 14:2461. [PMID: 35626065 PMCID: PMC9139614 DOI: 10.3390/cancers14102461] [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: 05/03/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 11/17/2022] Open
Abstract
Thyroid cancer is the most frequent endocrine malignancy and accounts for approximately 1% of all diagnosed cancers. A variety of mechanisms are involved in the transformation of a normal tissue into a malignant one. Loss of tumor-suppressor gene (TSG) function is one of these mechanisms. The normal functions of TSGs include cell proliferation and differentiation control, genomic integrity maintenance, DNA damage repair, and signaling pathway regulation. TSGs are generally classified into three subclasses: (i) gatekeepers that encode proteins involved in cell cycle and apoptosis control; (ii) caretakers that produce proteins implicated in the genomic stability maintenance; and (iii) landscapers that, when mutated, create a suitable environment for malignant cell growth. Several possible mechanisms have been implicated in TSG inactivation. Reviewing the various TSG alteration types detected in thyroid cancers may help researchers to better understand the TSG defects implicated in the development/progression of this cancer type and to find potential targets for prognostic, predictive, diagnostic, and therapeutic purposes. Hence, the main purposes of this review article are to describe the various TSG inactivation mechanisms and alterations in human thyroid cancer, and the current therapeutic options for targeting TSGs in thyroid cancer.
Collapse
Affiliation(s)
- Sadegh Rajabi
- Traditional Medicine and Materia Medica Research Center, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran;
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, CEDEX 5, 34093 Montpellier, France
- Centre for Ecological and Evolutionary Cancer Research (CREEC), Unité Mixte de Recherches, Institut de Recherche pour le Développement (IRD) 224–Centre National de Recherche Scientifique (CNRS) 5290–University of Montpellier, 34000 Montpellier, France
| | - Arshia Sharbatdar Alaei
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran 19839-63113, Iran;
| | | | - Heewa Shakib
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran 19857-17443, Iran;
| | - Mohammad Reza Ashrafi
- Department of Biochemistry, Afzalipoor Faculty of Medicine, Kerman University of Medical Sciences, Kerman 76169-13555, Iran;
| |
Collapse
|
15
|
HNF4A-AS1-encoded small peptide promotes self-renewal and aggressiveness of neuroblastoma stem cells via eEF1A1-repressed SMAD4 transactivation. Oncogene 2022; 41:2505-2519. [PMID: 35318442 DOI: 10.1038/s41388-022-02271-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 02/19/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023]
Abstract
Cancer stem cells play crucial roles in tumorigenesis and aggressiveness, while regulatory mechanisms in neuroblastoma (NB), a pediatric extracranial malignancy with highest incidence, are still unknown. Herein, a small 51-amino acid peptide (sPEP1) encoded by hepatocyte nuclear factor 4 alpha antisense RNA 1 (HNF4A-AS1) was identified in tumor tissues and cells, which facilitated self-renewal and aggressiveness of NB stem cells. MiRNA-409-5p interacted with HNF4A-AS1 to facilitate sPEP1 translation via recruiting eukaryotic translation initiation factor 3 subunit G, while sPEP1 repressed serum deprivation-induced senescence and promoted sphere formation, growth, or metastasis of NB stem cells. Mechanistically, sPEP1 directly interacted with eukaryotic translation elongation factor 1 alpha 1 (eEF1A1) to facilitate its binding to SMAD family member 4 (SMAD4), resulting in repression of SMAD4 transactivation and transcriptional upregulation of stem cell genes associated with tumor progression. Rescue experiments revealed that sPEP1 exerted oncogenic roles via facilitating physical interaction between eEF1A1 and SMAD4. Notably, knockdown of sPEP1 significantly repressed the self-renewal and metastasis of NB stem cells in vivo. High sPEP1 or eEF1A1 levels in clinical NB tissues were linked to poor patients' survival. These findings suggest that HNF4A-AS1-encoded sPEP1 promotes self-renewal and aggressive features of NB stem cells by eEF1A1-repressed SMAD4 transactivation.
Collapse
|
16
|
SMAD4-201 transcript as a putative biomarker in colorectal cancer. BMC Cancer 2022; 22:72. [PMID: 35034624 PMCID: PMC8762975 DOI: 10.1186/s12885-022-09186-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 12/30/2021] [Indexed: 11/28/2022] Open
Abstract
Background Transcripts with alternative 5′-untranslated regions (UTRs) result from the activity of alternative promoters and they can determine gene expression by influencing its stability and translational efficiency, thus executing complex regulation of developmental, physiological and pathological processes. Transcriptional regulation of human SMAD4, a key tumor suppressor deregulated in most gastrointestinal cancers, entails four alternative promoters. These promoters and alternative transcripts they generate remain unexplored as contributors to the SMAD4 deregulation in cancer. The aim of this study was to investigate the relative abundance of the transcript SMAD4–201 in colorectal cell lines and tissues in order to establish if its fluctuations may be associated with colorectal cancer (CRC). Methods Relative abundance of SMAD4–201 in total SMAD4 mRNA was analyzed using quantitative PCR in a set of permanent human colon cell lines and tumor and corresponding healthy tissue samples from patients with CRC. Results The relative abundance of SMAD4–201 in analyzed cell lines varied between 16 and 47%. A similar relative abundance of SMAD4–201 transcript was found in the majority of analyzed human tumor tissue samples, and it was averagely 20% lower in non-malignant in comparison to malignant tissue samples (p = 0.001). Transcript SMAD4–202 was not detectable in any of the analyzed samples, so the observed fluctuations in the composition of SMAD4 transcripts can be attributed to transcripts other than SMAD4–201 and SMAD4–202. Conclusion The expression profile of SMAD4–201 in human tumor and non-tumor tissue samples may indicate the translational potential of this molecule in CRC, but further research is needed to clarify its usability as a potential biomarker for early diagnosis. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-022-09186-z.
Collapse
|
17
|
Wu M, Wang H, Kong D, Shao J, Song C, Yang T, Zhang Y. miR-452-3p inhibited osteoblast differentiation by targeting Smad4. PeerJ 2021; 9:e12228. [PMID: 34692253 PMCID: PMC8485836 DOI: 10.7717/peerj.12228] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/08/2021] [Indexed: 12/28/2022] Open
Abstract
Osteoblast differentiation is a complex process that is essential for normal bone formation. A growing number of studies have shown that microRNAs (miRNAs) are key regulators in a variety of physiological and pathological processes, including osteogenesis. In this study, BMP2 was used to induce MC3T3-E1 cells to construct osteoblast differentiation cell model. Then, we investigated the effect of miR-452-3p on osteoblast differentiation and the related molecular mechanism by RT-PCR analysis, Western blot analysis, ALP activity, and Alizarin Red Staining. We found that miR-452-3p was significantly downregulated in osteoblast differentiation. Overexpression miR-452-3p (miR-452-3p mimic) significantly inhibited the expression of osteoblast marker genes RUNX2, osteopontin (OPN), and collagen type 1 a1 chain (Col1A1), and decreased the number of calcium nodules and ALP activity. In contrast, knockdown miR-452-3p (miR-452-3p inhibitor) produced the opposite effect. In terms of mechanism, we found that Smad4 may be the target of miR-452-3p, and knockdown Smad4 (si-Smad4) partially inhibited the osteoblast differentiation enhanced by miR-452-3p. Our results suggested that miR-452-3p plays an important role in osteoblast differentiation by targeting Smad4. Therefore, miR-452-3p is expected to be used in the treatment of bone formation and regeneration.
Collapse
Affiliation(s)
- Ming Wu
- Postgraduate Training Base in Shanghai Gongli Hospital, Ningxia Medical University, Shanghai, China
| | - Hongyan Wang
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| | - Dece Kong
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| | - Jin Shao
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| | - Chao Song
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| | - Tieyi Yang
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| | - Yan Zhang
- Department of Orthopaedics, Gongli Hospital of Pudong New Area, Shanghai, China
| |
Collapse
|
18
|
Tong K, Kothari OA, Haro KS, Panda A, Bandari MM, Carrick JN, Hur JJ, Zhang L, Chan CS, Xing J, Gatza ML, Ganesan S, Verzi MP. SMAD4 is critical in suppression of BRAF-V600E serrated tumorigenesis. Oncogene 2021; 40:6034-6048. [PMID: 34453124 PMCID: PMC8559887 DOI: 10.1038/s41388-021-01997-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/04/2021] [Accepted: 08/17/2021] [Indexed: 02/07/2023]
Abstract
BRAF-driven colorectal cancer is among the poorest prognosis subtypes of colon cancer. Previous studies suggest that BRAF-mutant serrated cancers frequently exhibit Microsatellite Instability (MSI) and elevated levels of WNT signaling. The loss of tumor-suppressor Smad4 in oncogenic BRAF-V600E mouse models promotes rapid serrated tumor development and progression, and SMAD4 mutations co-occur in human patient tumors with BRAF-V600E mutations. This study assesses the role of SMAD4 in early-stage serrated tumorigenesis. SMAD4 loss promotes microsatellite stable (MSS) serrated tumors in an oncogenic BRAF-V600E context, providing a model for MSS serrated cancers. Inactivation of Msh2 in these mice accelerated tumor formation, and whole-exome sequencing of both MSS and MSI serrated tumors derived from these mouse models revealed that all serrated tumors developed oncogenic WNT mutations, predominantly in the WNT-effector gene Ctnnb1 (β-catenin). Mouse models mimicking the oncogenic β-catenin mutation show that the combination of three oncogenic mutations (Ctnnb1, Braf, and Smad4) are critical to drive rapid serrated dysplasia formation. Re-analysis of human tumor data reveals BRAF-V600E mutations co-occur with oncogenic mutations in both WNT and SMAD4/TGFβ pathways. These findings identify SMAD4 as a critical factor in early-stage serrated cancers and helps broaden the knowledge of this rare but aggressive subset of colorectal cancer.
Collapse
Affiliation(s)
- Kevin Tong
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Om A. Kothari
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Katherine S. Haro
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Anshuman Panda
- Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Manisha M. Bandari
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Jillian N. Carrick
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Joseph J. Hur
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Lanjing Zhang
- Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA,Department of Pathology, Penn Medicine Princeton Medical Center, Plainsboro, NJ, USA
| | - Chang S. Chan
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Jinchuan Xing
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA
| | - Michael L. Gatza
- Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA,Department of Radiation Oncology, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Michael P. Verzi
- Department of Genetics, Human Genetics Institute of New Jersey (HGINJ), Rutgers University, 145 Bevier Road, Piscataway, NJ 08854, USA,Rutgers Cancer Institute of New Jersey (CINJ), 195 Little Albany Street, New Brunswick, NJ 08903, USA,Corresponding Author: Michael P. Verzi,
| |
Collapse
|
19
|
Chen N, Zheng Q, Wan G, Guo F, Zeng X, Shi P. Impact of posttranslational modifications in pancreatic carcinogenesis and treatments. Cancer Metastasis Rev 2021; 40:739-759. [PMID: 34342796 DOI: 10.1007/s10555-021-09980-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/06/2021] [Indexed: 01/22/2023]
Abstract
Pancreatic cancer (PC) is a highly aggressive cancer, with a 9% 5-year survival rate and a high risk of recurrence. In part, this is because PC is composed of heterogeneous subgroups with different biological and functional characteristics and personalized anticancer treatments are required. Posttranslational modifications (PTMs) play an important role in modifying protein functions/roles and are required for the maintenance of cell viability and biological processes; thus, their dysregulation can lead to disease. Different types of PTMs increase the functional diversity of the proteome, which subsequently influences most aspects of normal cell biology or pathogenesis. This review primarily focuses on ubiquitination, SUMOylation, and NEDDylation, as well as the current understanding of their roles and molecular mechanisms in pancreatic carcinogenesis. Additionally, we briefly summarize studies and clinical trials on PC treatments to advance our knowledge of drugs available to target the ubiquitination, SUMOylation, and NEDDylation PTM types. Further investigation of PTMs could be a critical field of study in relation to PC, as they have been implicated in the initiation and progression of many other types of cancer.
Collapse
Affiliation(s)
- Nianhong Chen
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital, 2Nd Clinical Medical College, Jinan University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School, Guangdong Province, Shenzhen University, Shenzhen, 518037, People's Republic of China.
- Department of Cell Biology & University of Pittsburgh Cancer Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15213, USA.
- Laboratory of Signal Transduction, Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA.
| | - Qiaoqiao Zheng
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Guoqing Wan
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital, 2Nd Clinical Medical College, Jinan University, Guangzhou, People's Republic of China
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School, Guangdong Province, Shenzhen University, Shenzhen, 518037, People's Republic of China
| | - Feng Guo
- Department of Medicine, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Xiaobin Zeng
- Center Lab of Longhua Branch and Department of Infectious Disease, Shenzhen People's Hospital, 2Nd Clinical Medical College, Jinan University, Guangzhou, People's Republic of China.
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Medicine School, Guangdong Province, Shenzhen University, Shenzhen, 518037, People's Republic of China.
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China.
| |
Collapse
|
20
|
Zhytnik L, Peters M, Tilk K, Simm K, Tõnisson N, Reimand T, Maasalu K, Acharya G, Krjutškov K, Salumets A. From late fatherhood to prenatal screening of monogenic disorders: evidence and ethical concerns. Hum Reprod Update 2021; 27:1056-1085. [PMID: 34329448 DOI: 10.1093/humupd/dmab023] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 06/27/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND With the help of ART, an advanced parental age is not considered to be a serious obstacle for reproduction anymore. However, significant health risks for future offspring hide behind the success of reproductive medicine for the treatment of reduced fertility associated with late parenthood. Although an advanced maternal age is a well-known risk factor for poor reproductive outcomes, understanding the impact of an advanced paternal age on offspring is yet to be elucidated. De novo monogenic disorders (MDs) are highly associated with late fatherhood. MDs are one of the major sources of paediatric morbidity and mortality, causing significant socioeconomic and psychological burdens to society. Although individually rare, the combined prevalence of these disorders is as high as that of chromosomal aneuploidies, indicating the increasing need for prenatal screening. With the help of advanced reproductive technologies, families with late paternity have the option of non-invasive prenatal testing (NIPT) for multiple MDs (MD-NIPT), which has a sensitivity and specificity of almost 100%. OBJECTIVE AND RATIONALE The main aims of the current review were to examine the effect of late paternity on the origin and nature of MDs, to highlight the role of NIPT for the detection of a variety of paternal age-associated MDs, to describe clinical experiences and to reflect on the ethical concerns surrounding the topic of late paternity and MD-NIPT. SEARCH METHODS An extensive search of peer-reviewed publications (1980-2021) in English from the PubMed and Google Scholar databases was based on key words in different combinations: late paternity, paternal age, spermatogenesis, selfish spermatogonial selection, paternal age effect, de novo mutations (DNMs), MDs, NIPT, ethics of late fatherhood, prenatal testing and paternal rights. OUTCOMES An advanced paternal age provokes the accumulation of DNMs, which arise in continuously dividing germline cells. A subset of DNMs, owing to their effect on the rat sarcoma virus protein-mitogen-activated protein kinase signalling pathway, becomes beneficial for spermatogonia, causing selfish spermatogonial selection and outgrowth, and in some rare cases may lead to spermatocytic seminoma later in life. In the offspring, these selfish DNMs cause paternal age effect (PAE) disorders with a severe and even life-threatening phenotype. The increasing tendency for late paternity and the subsequent high risk of PAE disorders indicate an increased need for a safe and reliable detection procedure, such as MD-NIPT. The MD-NIPT approach has the capacity to provide safe screening for pregnancies at risk of PAE disorders and MDs, which constitute up to 20% of all pregnancies. The primary risks include pregnancies with a paternal age over 40 years, a previous history of an affected pregnancy/child, and/or congenital anomalies detected by routine ultrasonography. The implementation of NIPT-based screening would support the early diagnosis and management needed in cases of affected pregnancy. However, the benefits of MD-NIPT need to be balanced with the ethical challenges associated with the introduction of such an approach into routine clinical practice, namely concerns regarding reproductive autonomy, informed consent, potential disability discrimination, paternal rights and PAE-associated issues, equity and justice in accessing services, and counselling. WIDER IMPLICATIONS Considering the increasing parental age and risks of MDs, combined NIPT for chromosomal aneuploidies and microdeletion syndromes as well as tests for MDs might become a part of routine pregnancy management in the near future. Moreover, the ethical challenges associated with the introduction of MD-NIPT into routine clinical practice need to be carefully evaluated. Furthermore, more focus and attention should be directed towards the ethics of late paternity, paternal rights and paternal genetic guilt associated with pregnancies affected with PAE MDs.
Collapse
Affiliation(s)
- Lidiia Zhytnik
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Maire Peters
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Kadi Tilk
- Competence Centre on Health Technologies, Tartu, Estonia
| | - Kadri Simm
- Institute of Philosophy and Semiotics, Faculty of Arts and Humanities, University of Tartu, Tartu, Estonia.,Centre of Ethics, University of Tartu, Tartu, Estonia
| | - Neeme Tõnisson
- Institute of Genomics, University of Tartu, Tartu, Estonia.,Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia.,Department of Reproductive Medicine, West Tallinn Central Hospital, Tallinn, Estonia
| | - Tiia Reimand
- Department of Clinical Genetics, United Laboratories, Tartu University Hospital, Tartu, Estonia.,Department of Clinical Genetics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Katre Maasalu
- Clinic of Traumatology and Orthopaedics, Tartu University Hospital, Tartu, Estonia.,Department of Traumatology and Orthopaedics, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Ganesh Acharya
- Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Kaarel Krjutškov
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia
| | - Andres Salumets
- Competence Centre on Health Technologies, Tartu, Estonia.,Department of Obstetrics and Gynaecology, Institute of Clinical Medicine, University of Tartu, Tartu, Estonia.,Institute of Genomics, University of Tartu, Tartu, Estonia.,Division of Obstetrics and Gynaecology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
21
|
Sommariva S, Caviglia G, Piana M. Gain and loss of function mutations in biological chemical reaction networks: a mathematical model with application to colorectal cancer cells. J Math Biol 2021; 82:55. [PMID: 33945019 PMCID: PMC8096774 DOI: 10.1007/s00285-021-01607-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 03/20/2021] [Accepted: 04/13/2021] [Indexed: 12/19/2022]
Abstract
This paper studies a system of Ordinary Differential Equations modeling a chemical reaction network and derives from it a simulation tool mimicking Loss of Function and Gain of Function mutations found in cancer cells. More specifically, from a theoretical perspective, our approach focuses on the determination of moiety conservation laws for the system and their relation with the corresponding stoichiometric surfaces. Then we show that Loss of Function mutations can be implemented in the model via modification of the initial conditions in the system, while Gain of Function mutations can be implemented by eliminating specific reactions. Finally, the model is utilized to examine in detail the G1-S phase of a colorectal cancer cell.
Collapse
Affiliation(s)
- Sara Sommariva
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy
| | - Giacomo Caviglia
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy
| | - Michele Piana
- Dipartimento di Matematica, Universitá di Genova, Via Dodecaneso, 35 16146, Genoa, Italy. .,CNR - SPIN GENOVA, Via Dodecaneso, 35 16146, Genoa, Italy.
| |
Collapse
|
22
|
Bao D, Li M, Zhou D, Zhuang C, Ge Z, Wei Q, Zhang L. miR-130b-3p is high-expressed in polycystic ovarian syndrome and promotes granulosa cell proliferation by targeting SMAD4. J Steroid Biochem Mol Biol 2021; 209:105844. [PMID: 33582305 DOI: 10.1016/j.jsbmb.2021.105844] [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] [Received: 11/22/2020] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 01/01/2023]
Abstract
BACKGROUND Being one of the most prevalent metabolic and endocrine disorders, Polycystic Ovary Syndrome (PCOS) has been proven to be associated with microRNA-130b-3p (miR-130b-3p). However, the exact role played by miR-130b-3p in the pathogenesis and progression of PCOS remains unknown. Thus, this article is focused on elucidating the function of miR-130b-3p in the pathogenesis of PCOS. METHODS The expression levels of miR-130b-3p and SMAD4 in tissues and cells responsible for the development of PCOS were determined by RT-qPCR and western blot. A miR-130b-3p mimic/inhibitor or si-SMAD4 were transfected into KGN cells. The cell viability was detected by CCK-8 and EDU methods. The activity of caspase-3 was measured by caspase-3 analysis. Subsequently, apoptosis and the cell cycle were measured via flow cytometry. The correlation between SMAD4 and miR-130b-3p was confirmed using an RNA pull-down assay and a dual luciferase reporter system assay. RESULTS MiR-130b-3p was upregulated in the KGN cells and ovarian granulosa cells (GCs) of PCOS patients. It was found that miR-130b-3p overexpression or SMAD4 silencing can promote KGN cell proliferation and positive EDU rates, induce S phase arrest, inhibit apoptosis and caspase-3 activity. On the other hand, miR-130b-3p inhibitors reduce KGN cell proliferation, inhibit apoptosis and reverse the effect of si-SMAD4. CONCLUSION MiR-130b-3p directly interacts with SMAD4 to induce KGN cell proliferation, inhibit apoptosis, suggesting that miR-130b-3p expression is positively correlated with the development of PCOS. This may serve as new evidence for the abnormal proliferation of GCs in PCOS.
Collapse
Affiliation(s)
- Dongqin Bao
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China.
| | - Mingan Li
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Dongxia Zhou
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Chaohui Zhuang
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Zhijuan Ge
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Qian Wei
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| | - Limin Zhang
- Center for Reproductive Medicine, The Affiliated Shuyang Hospital of Xuzhou Medical University, Suqian City, Jiangsu Province, China
| |
Collapse
|
23
|
Mojsilovic S, Mojsilovic SS, Bjelica S, Santibanez JF. Transforming growth factor-beta1 and myeloid-derived suppressor cells: A cancerous partnership. Dev Dyn 2021; 251:105-124. [PMID: 33797140 DOI: 10.1002/dvdy.339] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/15/2021] [Accepted: 03/25/2021] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta1 (TGF-β1) plays a crucial role in tumor progression. It can inhibit early cancer stages but promotes tumor growth and development at the late stages of tumorigenesis. TGF-β1 has a potent immunosuppressive function within the tumor microenvironment that largely contributes to tumor cells' immune escape and reduction in cancer immunotherapy responses. Likewise, myeloid-derived suppressor cells (MDSCs) have been postulated as leading tumor promoters and a hallmark of cancer immune evasion mechanisms. This review attempts to analyze the prominent roles of both TGF-β1 and MDSCs and their interplay in cancer immunity. Furthermore, therapies against either TGF-β1 or MDSCs, and their potential synergistic combination with immunotherapies are discussed. Simultaneous TGF-β1 and MDSCs inhibition suggest a potential improvement in immunotherapy or subverted tumor immune resistance.
Collapse
Affiliation(s)
- Slavko Mojsilovic
- Laboratory of Experimental Hematology and Stem Cells, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Sonja S Mojsilovic
- Laboratory for Immunochemistry, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Bjelica
- Department of Hematology, Clinical Hospital Centre Dragisa Misovic, Belgrade, Serbia
| | - Juan F Santibanez
- Molecular oncology group, Institute for Medical Research, University of Belgrade, Republic of Serbia.,Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
24
|
Zhao Y, Liu S, Li X, Xu Z, Hao L, Cui Z, Bi K, Zhang Y, Liu Z. Cross-talk of Signaling Pathways in the Pathogenesis of Allergic Asthma and Cataract. Protein Pept Lett 2021; 27:810-822. [PMID: 32031062 DOI: 10.2174/0929866527666200207113439] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/27/2019] [Accepted: 11/30/2019] [Indexed: 12/13/2022]
Abstract
Allergic asthma is a chronic inflammatory disease, which involves many cellular and cellular components. Cataract is a condition that affects the transparency of the lens, which the opacity of the lens caused by any innate or acquired factor degrades its transparency or changes in color. Both of them belong to diseases induced by immune disorders or inflammation. We want to confirm the signaling pathways involved in the regulation of asthma and cataract simultaneously, and provide reference for the later related experiments. So we conducted a scoping review of many databases and searched for studies (Academic research published in Wiley, Springer and Bentham from 2000 to 2019) about the possible relationship between asthma and cataract. It was found that during the onset of asthma and cataract, Rho/Rock signaling pathway, Notch signaling pathway, Wnt/β-catenin signaling pathway, PI3K/AKT signaling pathway, JAK/STAT signaling pathway, MAPK signaling pathway, TGF-β1/Smad signaling pathway and NF-κB signaling pathway are all active, so they may have a certain correlation in pathogenesis. Asthma may be associated with cataract through the eight signaling pathways, causing inflammation or immune imbalance based on allergy that can lead to cataract. According to these studies, we speculated that the three most likely signaling pathways are PI3K/AKT, MAPK and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Yang Zhao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Sumei Liu
- Department of Stomatology, No. 2 Hospital of Baoding, Baoding 071002, China
| | - Xiangsheng Li
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Zhenzhen Xu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Lifang Hao
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Zhe Cui
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Kewei Bi
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| | - Yanfen Zhang
- Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China,Offices of Science and Technology, Hebei University, Baoding 071002, China
| | - Zhongcheng Liu
- College of Pharmaceutical Sciences, Hebei University, Baoding 071002, China.,Key Laboratory of Pharmaceutical Quality Control of Hebei Province, Baoding 071002, China
| |
Collapse
|
25
|
Galka-Marciniak P, Urbanek-Trzeciak M, Nawrocka P, Kozlowski P. A pan-cancer atlas of somatic mutations in miRNA biogenesis genes. Nucleic Acids Res 2021; 49:601-620. [PMID: 33406242 PMCID: PMC7826265 DOI: 10.1093/nar/gkaa1223] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/28/2020] [Accepted: 12/04/2020] [Indexed: 02/07/2023] Open
Abstract
It is a well-known and intensively studied phenomenon that the levels of many miRNAs are differentiated in cancer. miRNA biogenesis and functional expression are complex processes orchestrated by many proteins cumulatively called miRNA biogenesis proteins. To characterize cancer somatic mutations in the miRNA biogenesis genes and investigate their potential impact on the levels of miRNAs, we analyzed whole-exome sequencing datasets of over 10 000 cancer/normal sample pairs deposited within the TCGA repository. We identified and characterized over 3600 somatic mutations in 29 miRNA biogenesis genes and showed that some of the genes are overmutated in specific cancers and/or have recurrent hotspot mutations (e.g. SMAD4 in PAAD, COAD and READ; DICER1 in UCEC; PRKRA in OV and LIN28B in SKCM). We identified a list of miRNAs whose level is affected by particular types of mutations in either SMAD4, SMAD2 or DICER1 and showed that hotspot mutations in the RNase domains in DICER1 not only decrease the level of 5p-miRNAs but also increase the level of 3p-miRNAs, including many well-known cancer-related miRNAs. We also showed an association of the mutations with patient survival. Eventually, we created an atlas/compendium of miRNA biogenesis alterations providing a useful resource for different aspects of biomedical research.
Collapse
Affiliation(s)
| | | | | | - Piotr Kozlowski
- Institute of Bioorganic Chemistry, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
26
|
Liu Y, Jia W, Li J, Zhu H, Yu J. Identification of Survival-Associated Alternative Splicing Signatures in Lung Squamous Cell Carcinoma. Front Oncol 2020; 10:587343. [PMID: 33117720 PMCID: PMC7561379 DOI: 10.3389/fonc.2020.587343] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023] Open
Abstract
Purpose: Alternative splicing (AS) is a post-transcriptional process that plays a significant role in enhancing the diversity of transcription and protein. Accumulating evidences have demonstrated that dysregulation of AS is associated with oncogenic processes. However, AS signature specifically in lung squamous cell carcinoma (LUSC) remains unknown. This study aimed to evaluate the prognostic values of AS events in LUSC patients. Methods: The RNA-seq data, AS events data and corresponding clinical information were obtained from The Cancer Genome Atlas (TCGA) database. Univariate Cox regression analysis was performed to identify survival-related AS events and survival-related parent genes were subjected to Gene Ontology enrichment analysis and gene network analysis. The least absolute shrinkage and selection operator (LASSO) method and multivariate Cox regression analysis were used to construct prognostic prediction models, and their predictive values were assessed by Kaplan-Meier analysis and receiver operating characteristic (ROC) curves. Then a nomogram was established to predict the survival of LUSC patients. And the interaction network of splicing factors (SFs) and survival-related AS events was constructed by Spearman correlation analysis and visualized by Cytoscape. Results: Totally, 467 LUSC patients were included in this study and 1,991 survival-related AS events within 1,433 genes were identified. SMAD4, FOS, POLR2L, and RNPS1 were the hub genes in the gene interaction network. Eight prognostic prediction models (seven types of AS and all AS) were constructed and all exhibited high efficiency in distinguishing good or poor survival of LUSC patients. The final integrated prediction model including all types of AS events exhibited the best prognostic power with the maximum AUC values of 0.778, 0.816, 0.814 in 1, 3, 5 years ROC curves, respectively. Meanwhile, the nomogram performed well in predicting the 1-, 3-, and 5-year survival of LUSC patients. In addition, the SF-AS regulatory network uncovered a significant correlation between SFs and survival-related AS events. Conclusion: This is the first comprehensive study to analyze the role of AS events in LUSC specifically, which improves our understanding of the prognostic value of survival-related AS events for LUSC. And these survival-related AS events might serve as novel prognostic biomarkers and drug therapeutic targets for LUSC.
Collapse
Affiliation(s)
- Yang Liu
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wenxiao Jia
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Ji Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
27
|
Stanley S, Balic Z, Hubmacher D. Acromelic dysplasias: how rare musculoskeletal disorders reveal biological functions of extracellular matrix proteins. Ann N Y Acad Sci 2020; 1490:57-76. [PMID: 32880985 DOI: 10.1111/nyas.14465] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/16/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022]
Abstract
Acromelic dysplasias are a group of rare musculoskeletal disorders that collectively present with short stature, pseudomuscular build, stiff joints, and tight skin. Acromelic dysplasias are caused by mutations in genes (FBN1, ADAMTSL2, ADAMTS10, ADAMTS17, LTBP2, and LTBP3) that encode secreted extracellular matrix proteins, and in SMAD4, an intracellular coregulator of transforming growth factor-β (TGF-β) signaling. The shared musculoskeletal presentations in acromelic dysplasias suggest that these proteins cooperate in a biological pathway, but also fulfill distinct roles in specific tissues that are affected in individual disorders of the acromelic dysplasia group. In addition, most of the affected proteins directly interact with fibrillin microfibrils in the extracellular matrix and have been linked to the regulation of TGF-β signaling. Together with recently developed knockout mouse models targeting the affected genes, novel insights into molecular mechanisms of how these proteins regulate musculoskeletal development and homeostasis have emerged. Here, we summarize the current knowledge highlighting pathogenic mechanisms of the different disorders that compose acromelic dysplasias and provide an overview of the emerging biological roles of the individual proteins that are compromised. Finally, we develop a conceptual model of how these proteins may interact and form an "acromelic dysplasia complex" on fibrillin microfibrils in connective tissues of the musculoskeletal system.
Collapse
Affiliation(s)
- Sarah Stanley
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Zerina Balic
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Dirk Hubmacher
- Leni & Peter W. May Department of Orthopedics, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
28
|
Knyazeva M, Korobkina E, Karizky A, Sorokin M, Buzdin A, Vorobyev S, Malek A. Reciprocal Dysregulation of MiR-146b and MiR-451 Contributes in Malignant Phenotype of Follicular Thyroid Tumor. Int J Mol Sci 2020; 21:E5950. [PMID: 32824921 PMCID: PMC7503510 DOI: 10.3390/ijms21175950] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 08/15/2020] [Accepted: 08/17/2020] [Indexed: 01/08/2023] Open
Abstract
Over the last few years, incidental thyroid nodules are being diagnosed with increasing frequency with the use of highly sensitive imaging techniques. The ultrasound thyroid gland examination, followed by the fine-needle aspiration cytology is the standard diagnostic approach. However, in cases of the follicular nature of nodules, cytological diagnosis is not enough. Analysis of miRNAs in the biopsy presents a promising approach. Increasing our knowledge of miRNA's role in follicular carcinogenesis, and development of the appropriate the miRNA analytical technologies are required to implement miRNA-based tests in clinical practice. We used material from follicular thyroid nodes (n.84), grouped in accordance with their invasive properties. The invasion-associated miRNAs expression alterations were assayed. Expression data were confirmed by highly sensitive two-tailed RT-qPCR. Reciprocally dysregulated miRNAs pair concentration ratios were explored as a diagnostic parameter using receiver operation curve (ROC) analysis. A new bioinformatics method (MiRImpact) was applied to evaluate the biological significance of the observed expression alterations. Coupled experimental and computational approaches identified reciprocal dysregulation of miR-146b and miR-451 as important attributes of follicular cell malignant transformation and follicular thyroid cancer progression. Thus, evaluation of combined dysregulation of miRNAs relevant to invasion and metastasis can help to distinguish truly malignant follicular thyroid cancer from indolent follicular adenoma.
Collapse
Affiliation(s)
- Margarita Knyazeva
- Subcellular technology Lab., N. N. Petrov National Medical Center of Oncology, 197758 Saint Petersburg, Russia; (M.K.); (E.K.)
- Oncosystem Company Limited, 121205 Moscow, Russia
- Institute of Biomedical Systems and Biotechnologies, Peter the Great Saint. Petersburg Polytechnic University (SPbPU), 195251 Saint Petersburg, Russia
| | - Ekaterina Korobkina
- Subcellular technology Lab., N. N. Petrov National Medical Center of Oncology, 197758 Saint Petersburg, Russia; (M.K.); (E.K.)
- Oncosystem Company Limited, 121205 Moscow, Russia
| | - Alexey Karizky
- Information Technologies and Programming Faculty, Information Technologies, Mechanics and Optics (ITMO) University, 197101 Saint-Petersburg, Russia;
| | - Maxim Sorokin
- Institute of Personalized Medicine, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.S.); (A.B.)
- Omicsway Corporation, Walnut, CA 91789, USA
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Anton Buzdin
- Institute of Personalized Medicine, I.M. Sechenov First Moscow State Medical University, 119048 Moscow, Russia; (M.S.); (A.B.)
- Omicsway Corporation, Walnut, CA 91789, USA
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Sergey Vorobyev
- National Center of Clinical Morphological Diagnostics, 192283 Saint Petersburg, Russia;
| | - Anastasia Malek
- Subcellular technology Lab., N. N. Petrov National Medical Center of Oncology, 197758 Saint Petersburg, Russia; (M.K.); (E.K.)
- Oncosystem Company Limited, 121205 Moscow, Russia
| |
Collapse
|
29
|
N6-(2-hydroxyethyl)-Adenosine Induces Apoptosis via ER Stress and Autophagy of Gastric Carcinoma Cells In Vitro and In Vivo. Int J Mol Sci 2020; 21:ijms21165815. [PMID: 32823628 PMCID: PMC7461581 DOI: 10.3390/ijms21165815] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 01/03/2023] Open
Abstract
Gastric cancer is the most common malignant tumor of the digestive tract and is great challenge in clinical treatment. N6-(2-Hydroxyethyl)-adenosine (HEA), widely present in various fungi, is a natural adenosine derivative with many biological and pharmacological activities. Here, we assessed the antineoplastic effect of HEA on gastric carcinoma. HEA exerted cytotoxic effects against gastric carcinoma cells (SGC-7901 and AGS) in a dose and time-dependent manner. Additionally, we found that HEA induced reactive oxygen species production and mitochondrial membrane potential depolarization. Moreover, it could trigger caspase-dependent apoptosis, promoting intracellular Ca2+-related endoplasmic reticulum (ER) stress and autophagy. On the other hand, HEA could significantly inhibit the growth of transplanted tumors in nude mice and induce apoptosis of tumor tissues cells in vivo. In conclusion, HEA induced apoptosis of gastric carcinoma cells in vitro and in vivo, demonstrating that HEA is a potential chemotherapeutic agent for gastric carcinoma.
Collapse
|
30
|
Par-4 mediated Smad4 induction in PDAC cells restores canonical TGF-β/ Smad4 axis driving the cells towards lethal EMT. Eur J Cell Biol 2020; 99:151076. [PMID: 32439219 DOI: 10.1016/j.ejcb.2020.151076] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 12/22/2022] Open
Abstract
Deregulation of TGF-β signaling is intricately engrossed in the pathophysiology of pancreatic adenocarcinomas (PDACs). The role of TGF-β all through pancreatic cancer initiation and progression is multifarious and somewhat paradoxical. TGF-β plays a tumor suppressive role in early-stage pancreatic cancer by promoting apoptosis and inhibiting epithelial cell cycle progression, but incites tumor promotion in late-stage by modulating genomic instability, neo-angiogenesis, immune evasion, cell motility, and metastasis. Here, we provide evidences that Par-4 acts as one of the vital mediators to regulate TGF-β/Smad4 pathway, wherein, Par-4 induction/over-expression induced EMT which was later culminated in to apoptosis in presence of TGF-β via positive regulation of Smad4. Intriguingly, Par-4-/- cells were devoid of significant Smad4 induction compared to Par-4+/+ cells in presence of TGF-β and ectopic Par-4 steadily augmented Smad4 expression by restoring TGF-β/Smad4 axis in Panc-1 cells. Further, our FACS and western blotting results unveiled that Par-4 dragged the PDAC cells to G1 arrest in presence of TGF-β byelevating p21 and p27 levels while attenuating Cyclin E and A levels and augmenting caspase 3 cleavage triggering lethal EMT. Through restoration of Smad4, we further establish that in BxPC3 cell line (Smad4-/-), Smad4 is essential for Par-4 to indulge TGF-β dependent lethal EMT program. The mechanistic relevance of Par-4 mediated Smad4 activation was additionally validated by co-immunoprecipitation wherein disruption of NM23H1-STRAP interaction by Par-4 rescues TGF-β/Smad4 pathway in PDAC and mediates the tumor suppressive role of TGF-β, therefore serving as a vital cog to restore the apoptotic functions of TGF-β pathway.
Collapse
|
31
|
Huang W, Wu Y, Cheng D, He Z. Mechanism of epithelial‑mesenchymal transition inhibited by miR‑203 in non‑small cell lung cancer. Oncol Rep 2019; 43:437-446. [PMID: 31894278 PMCID: PMC6967097 DOI: 10.3892/or.2019.7433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 10/25/2019] [Indexed: 01/05/2023] Open
Abstract
The aim of the present study was to investigate whether miR-203 can inhibit transforming growth factor-β (TGF-β)-induced epithelial-mesenchymal transition (EMT), and the migration and invasion ability of non-small cell lung cancer (NSCLC) cells by targeting SMAD3. In the present study, the expression levels of miR-203, SMAD3 mRNA and protein in NSCLC tissues were examined, as well as their corresponding paracancerous samples. The miR-203 mimics and miR-203 inhibitor were transfected into the H226 cell line. RT-qPCR was used to assess the expression levels of E-cadherin, Snail, N-cadherin and vimentin mRNA, and western blotting was performed to detect the expression levels of p-SMAD2, SMAD2, p-SMAD3, SMAD3 and SMAD4. The cell migration and invasion abilities were detected by Transwell assays. The target site of SMAD3 was predicted by the combined action between miR-203 and dual luciferase. The results revealed that the RNA levels of miR-203, compared with paracancerous tissues, were decreased in NSCLC tissues, while SMAD3 mRNA and protein levels were upregulated, and miR-203 inhibited SMAD3 expression. Induction of TGF-β led to decreased E-cadherin mRNA levels, upregulation of Snail, N-cadherin and vimentin mRNA levels (P<0.05), and significant increase in cell migration and invasion, whereas transfection of miR-203 mimics reversed the aforementioned results (P<0.05). Conversely, miR-203 inhibitor could further aggravate the aforementioned results (P<0.05). Western blot results revealed that transfection of miR-203 mimics significantly reduced the protein expression of SMAD3 and p-SMAD3 (P<0.05). Furthermore, the results of the Dual-Luciferase assay revealed that miR-203 inhibited SMAD3 expression by interacting with specific regions of its 3′-UTR. Overall, a novel mechanism is revealed, in which, miR-203 can inhibit SMAD3 by interacting with specific regions of the 3′-UTR of SMAD3, thereby restraining TGF-β-induced EMT progression and migration and invasion of NSCLC cells.
Collapse
Affiliation(s)
- Weicong Huang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Yuanbo Wu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Dezhi Cheng
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhifeng He
- Department of Thoracic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
32
|
Are Ski and SnoN Involved in the Tumorigenesis of Oral Squamous Cell Carcinoma Through Smad4? Appl Immunohistochem Mol Morphol 2019; 27:626-630. [DOI: 10.1097/pai.0000000000000667] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
33
|
Chen J, Deng Y, Ao L, Song Y, Xu Y, Wang CC, Choy KW, Tony Chung KH, Du Q, Sui Y, Yang T, Yang J, Li H, Zou C, Tang T. The high-risk HPV oncogene E7 upregulates miR-182 expression through the TGF-β/Smad pathway in cervical cancer. Cancer Lett 2019; 460:75-85. [PMID: 31247272 DOI: 10.1016/j.canlet.2019.06.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/20/2019] [Accepted: 06/21/2019] [Indexed: 01/08/2023]
Abstract
Accumulating experimental evidence has shown that the aberrant expression of microRNAs (miRNAs) is involved in the development and progression of human cervical cancer. Previously, we identified miR-182 as an oncomiRNA in cervical cancer. However, the mechanism by which miR-182 is regulated and the interaction between human papillomavirus (HPV) and miR-182 in cervical cancer development remains unknown. In the present study, we explored the link between HPV E7 and miR-182 and verified that high-risk HPV E7 upregulated miR-182 expression through TGF-β/Smad4 signaling pathway in cervical cancer. By contrast, low-risk HPV E7 did not affect the expression of TGF-β and miR-182. Mechanistically, as high-risk HPV E7 bound to pRb, E2F was released from the complex and bound to the TGF-β promoter region, resulting in TGF-β overexpression. Furthermore, the Smad4 signaling pathway was activated upon TGF-β overexpression, which led to an interaction between Smad4 and the miR-182 promoter region, subsequently inducing the upregulation of miR-182 in both cervical cancer cells and the surrounding normal cells. In conclusion, this newly identified high-risk HPV E7/TGF-β/miR-182 regulatory network might inform the development of specific therapeutic strategies for cervical cancer.
Collapse
Affiliation(s)
- Jiao Chen
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Deng
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Liangfei Ao
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Song
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yan Xu
- Department of Obstetrics & Gynaecology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Chi Chiu Wang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kwong Wai Choy
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Kwok Hung Tony Chung
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Quan Du
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking, University, Beijing, China
| | - Yi Sui
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Department of Nutrition, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Tao Yang
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong, Medical Center, Shanghai, China
| | - Jing Yang
- Reproductive Medicine Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hu Li
- Department of Obstetrics & Gynaecology, Guangzhou Panyu Central Hospital, Guangzhou, China
| | - Chang Zou
- Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University, Shenzhen People's Hospital, Shenzhen, 518020, China; Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, 518020, China.
| | - Tao Tang
- Department of Obstetrics & Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong; Department of Obstetrics & Gynaecology, Guangzhou Panyu Central Hospital, Guangzhou, China.
| |
Collapse
|
34
|
Xu S, Liu C, Ji H. Concise Review: Therapeutic Potential of the Mesenchymal Stem Cell Derived Secretome and Extracellular Vesicles for Radiation-Induced Lung Injury: Progress and Hypotheses. Stem Cells Transl Med 2019; 8:344-354. [PMID: 30618085 PMCID: PMC6431606 DOI: 10.1002/sctm.18-0038] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 11/27/2018] [Indexed: 12/15/2022] Open
Abstract
Radiation-induced lung injury (RILI) is a common complication in radiotherapy of thoracic tumors and limits the therapeutic dose of radiation that can be given to effectively control tumors. RILI develops through a complex pathological process, resulting in induction and activation of various cytokines, infiltration by inflammatory cells, cytokine-induced activation of fibroblasts, and subsequent tissue remodeling by activated fibroblasts, ultimately leading to impaired lung function and respiratory failure. Increasing evidence shows that mesenchymal stem cells (MSCs) may play a main role in modulating inflammation and immune responses, promoting survival and repair of damaged resident cells and enhancing regeneration of damaged tissue through soluble paracrine factors and therapeutic extracellular vesicles. Therefore, the use of the MSC-derived secretome and exosomes holds promising potential for RILI therapy. Here, we review recent progress on the potential mechanisms of MSC therapy for RILI, with an emphasis on soluble paracrine factors of MSCs. Hypotheses on how MSC derived exosomes or MSC-released exosomal miRNAs could attenuate RILI are also proposed. Problems and translational challenges of the therapies based on the MSC-derived secretome and exosomes are further summarized and underline the need for caution on rapid clinical translation. Stem Cells Translational Medicine 2019;8:344-354.
Collapse
Affiliation(s)
- Siguang Xu
- Institute of Lung and Molecular TherapyXinxiang Medical UniversityXinxiangHenanPeople's Republic of China
| | - Cong Liu
- Institute of Lung and Molecular TherapyXinxiang Medical UniversityXinxiangHenanPeople's Republic of China
| | - Hong‐Long Ji
- Department of Cellular and Molecular BiologyUniversity of Texas Health Science Center at TylerTylerTexasUSA
- Texas Lung Injury InstituteUniversity of Texas Health Science Center at TylerTylerTexasUSA
| |
Collapse
|
35
|
Luo J, Chen XQ, Li P. The Role of TGF-β and Its Receptors in Gastrointestinal Cancers. Transl Oncol 2019; 12:475-484. [PMID: 30594036 PMCID: PMC6314240 DOI: 10.1016/j.tranon.2018.11.010] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 11/20/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
Early detection of gastrointestinal tumors improves patient survival. However, patients with these tumors are typically diagnosed at an advanced stage and have poor prognosis. The incidence and mortality of gastrointestinal cancers, including esophageal, gastric, liver, colorectal, and pancreatic cancers, are increasing worldwide. Novel diagnostic and therapeutic agents are required to improve patient survival and quality of life. The tumor microenvironment, which contains nontumor cells, signaling molecules such as growth factors and cytokines, and extracellular matrix proteins, plays a critical role in cancer cell proliferation, invasion, and metastasis. Transforming growth factor beta (TGF-β) signaling has dual roles in gastrointestinal tumor development and progression as both a tumor suppressor and tumor promoter. Here, we review the dynamic roles of TGF-β and its receptors in gastrointestinal tumors and provide evidence that targeting TGF-β signaling may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Jingwen Luo
- Oncology Department, West China Hospital of Medicine, Sichuan University, Chengdu, Sichuan, 610041, P.R. China
| | - Xu-Qiao Chen
- Department of Neurosciences, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Ping Li
- Oncology Department, West China Hospital of Medicine, Sichuan University, Chengdu, Sichuan, 610041, P.R. China.
| |
Collapse
|
36
|
Cheng Y, Li Z, Xie J, Wang P, Zhu J, Li Y, Wang Y. MiRNA-224-5p inhibits autophagy in breast cancer cells via targeting Smad4. Biochem Biophys Res Commun 2018; 506:793-798. [PMID: 30389135 DOI: 10.1016/j.bbrc.2018.10.150] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 10/23/2018] [Indexed: 12/23/2022]
Abstract
BACKGROUND/AIMS Autophagy is known as a protective intracellular procedure, which can be regulated by several factors. MiRNA has been suggested as a potential element to mediate autophagy pathway in carcinomas. Our study was aim to investigate the role of autophagy in breast cancer cells and identify the involved molecular mechanism METHODS: The expression of LC3I/II, SQSTM1 and Smad4 were detected by western blot. The mRNA level were quantified by real-time PCR. MDC staining was used to directly visualize autophagosome formation. Target Scan 7.2 was used to predict biological targets of miR-224-5p RESULTS: MiR-224 -5p expression was upregulated in metastatic breast cancer and non-metastatic breast cancer cells compare with control. Moreover, miR-224-5p inhibition enhanced cellular autophagy levels in breast cancer cells. MiR-224-5p could suppress Smad4 expression in MDA-MB-231 cells, which indicated that Smad4 was identified as a target of miR-224-5p in breast cancer cells with high metastatic potential CONCLUSIONS: Our study revealed that miR-224-5p inhibited autophagy by targeting Smad4 in MDA-MB-231 cells. The results indicated that miR-224-5p/Smad4 regulating autophagy might be a novel regulatory network contributing to metastasis of breast cancer. MiR-224-5p and Smad4 is involved in breast tumorigenesis, which is possibly a novel target for breast cancer therapy.
Collapse
Affiliation(s)
- You Cheng
- Department of Clinical Laboratory Medicine, TaiZhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China; School of Medical Laboratory, Tianjin Medical University, No.1 Guangdong Road, Hexi District, Tianjin, 300203, China
| | - Zhaoyun Li
- Department of Clinical Laboratory Medicine, TaiZhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Jiaogui Xie
- Department of Urology, The Fifteenth Military Hospital of China, Wusu, Xinjiang, 833000, China
| | - Pan Wang
- Department of Clinical Laboratory Medicine, TaiZhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Jie Zhu
- Department of Clinical Laboratory Medicine, TaiZhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China
| | - Yueguo Li
- Department of Laboratory, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
| | - Yichao Wang
- Department of Clinical Laboratory Medicine, TaiZhou Central Hospital (Taizhou University Hospital), No.999 Donghai Road, Jiaojiang District, Taizhou, Zhejiang, 318000, China; School of Medical Laboratory, Tianjin Medical University, No.1 Guangdong Road, Hexi District, Tianjin, 300203, China.
| |
Collapse
|
37
|
Ullah I, Sun W, Tang L, Feng J. Roles of Smads Family and Alternative Splicing Variants of Smad4 in Different Cancers. J Cancer 2018; 9:4018-4028. [PMID: 30410607 PMCID: PMC6218760 DOI: 10.7150/jca.20906] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2017] [Accepted: 08/20/2018] [Indexed: 12/15/2022] Open
Abstract
Transforming Growth Factor β (TGF-β) is one of the most common secretory proteins which are recognized by membrane receptors joined to transcription regulatory factor. TGF-β signals are transduced by the Smads family that regulate differentiation, proliferation, early growth, apoptosis, homeostasis, and tumor development. Functional study of TGF-β signaling pathway and Smads role is vital for certain diseases such as cancer. Alternative splicing produces a diverse range of protein isoforms with unique function and the ability to react differently with various pharmaceutical products. This review organizes to describe the general study of Smads family, the process of alternative splicing, the general aspect of alternative splicing of Smad4 in cancer and the possible use of spliceoforms for the diagnosis and therapeutic purpose. The main aim and objective of this article are to highlight some particular mechanisms involving in alternatives splicing of cancer and also to demonstrate new evidence about alternative splicing in different steps given cancer initiation and progression.
Collapse
Affiliation(s)
- Irfan Ullah
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Weichao Sun
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jianguo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, China
| |
Collapse
|
38
|
Pellatt AJ, Mullany LE, Herrick JS, Sakoda LC, Wolff RK, Samowitz WS, Slattery ML. The TGFβ-signaling pathway and colorectal cancer: associations between dysregulated genes and miRNAs. J Transl Med 2018; 16:191. [PMID: 29986714 PMCID: PMC6038278 DOI: 10.1186/s12967-018-1566-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/29/2018] [Indexed: 12/19/2022] Open
Abstract
Background The TGFβ-signaling pathway plays an important role in the pathogenesis of colorectal cancer (CRC). Loss of function of several genes within this pathway, such as bone morphogenetic proteins (BMPs) have been seen as key events in CRC progression. Methods In this study we comprehensively evaluate differential gene expression (RNASeq) of 81 genes in the TGFβ-signaling pathway and evaluate how dysregulated genes are associated with miRNA expression (Agilent Human miRNA Microarray V19.0). We utilize paired carcinoma and normal tissue from 217 CRC cases. We evaluate the associations between differentially expressed genes and miRNAs and sex, age, disease stage, and survival months. Results Thirteen genes were significantly downregulated and 14 were significantly upregulated after considering fold change (FC) of > 1.50 or < 0.67 and multiple comparison adjustment. Bone morphogenetic protein genes BMP5, BMP6, and BMP2 and growth differentiation factor GDF7 were downregulated. BMP4, BMP7, INHBA (Inhibin beta A), TGFBR1, TGFB2, TGIF1, TGIF2, and TFDP1 were upregulated. In general, genes with the greatest dysregulation, such as BMP5 (FC 0.17, BMP6 (FC 0.25), BMP2 (FC 0.32), CDKN2B (FC 0.32), MYC (FC 3.70), BMP7 (FC 4.17), and INHBA (FC 9.34) showed dysregulation in the majority of the population (84.3, 77.4, 81.1, 80.2, 82.0, 51.2, and 75.1% respectively). Four genes, TGFBR2, ID4, ID1, and PITX2, were un-associated or slightly upregulated in microsatellite-stable (MSS) tumors while downregulated in microsatellite-unstable (MSI) tumors. Eight dysregulated genes were associated with miRNA differential expression. E2F5 and THBS1 were associated with one or two miRNAs; RBL1, TGFBR1, TGIF2, and INHBA were associated with seven or more miRNAs with multiple seed-region matches. Evaluation of the joint effects of mRNA:miRNA identified interactions that were stronger in more advanced disease stages and varied by survival months. Conclusion These data support an interaction between miRNAs and genes in the TGFβ-signaling pathway in association with CRC risk. These interactions are associated with unique clinical characteristics that may provide targets for further investigations. Electronic supplementary material The online version of this article (10.1186/s12967-018-1566-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Lila E Mullany
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Jennifer S Herrick
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, CA, USA
| | - Roger K Wolff
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA
| | - Wade S Samowitz
- Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Martha L Slattery
- Department of Medicine, University of Utah, 383 Colorow, Salt Lake City, UT, 84108, USA.
| |
Collapse
|
39
|
Sinkala M, Mulder N, Martin DP. Integrative landscape of dysregulated signaling pathways of clinically distinct pancreatic cancer subtypes. Oncotarget 2018; 9:29123-29139. [PMID: 30018740 PMCID: PMC6044387 DOI: 10.18632/oncotarget.25632] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 06/04/2018] [Indexed: 12/29/2022] Open
Abstract
Despite modern therapeutic advances, the survival prospects of pancreatic cancer patients have remained poor. Besides being highly metastatic, pancreatic cancer is challenging to treat because it is caused by a heterogeneous array of somatic mutations that impact a variety of signaling pathways and cellular regulatory systems. Here we use publicly available transcriptomic, copy number alteration and mutation profiling datasets from pancreatic cancer patients together with data on disease outcomes to show that the three major pancreatic cancer subtypes each display distinctive aberrations in cell signaling and metabolic pathways. Importantly, patients afflicted with these different pancreatic cancer subtypes also exhibit distinctive survival profiles. Within these patients, we find that dysregulation of the phosphoinositide 3-kinase and mitogen-activated protein kinase pathways, and p53 mediated disruptions of cell cycle processes are apparently drivers of disease. Further, we identify for the first time the molecular perturbations of signalling networks that are likely the primary causes of oncogenesis in each of the three pancreatic cancer subtypes.
Collapse
Affiliation(s)
- Musalula Sinkala
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| | - Nicola Mulder
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| | - Darren Patrick Martin
- University of Cape Town, School of Health Sciences, Department of Integrative Biomedical Sciences, Computational Biology Division, Observatory, 7925, South Africa
| |
Collapse
|
40
|
Signaling Crosstalk of TGF-β/ALK5 and PAR2/PAR1: A Complex Regulatory Network Controlling Fibrosis and Cancer. Int J Mol Sci 2018; 19:ijms19061568. [PMID: 29795022 PMCID: PMC6032192 DOI: 10.3390/ijms19061568] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 05/09/2018] [Accepted: 05/14/2018] [Indexed: 02/07/2023] Open
Abstract
Both signaling by transforming growth factor-β (TGF-β) and agonists of the G Protein-coupled receptors proteinase-activated receptor-1 (PAR1) and -2 (PAR2) have been linked to tissue fibrosis and cancer. Intriguingly, TGF-β and PAR signaling either converge on the regulation of certain matrix genes overexpressed in these pathologies or display mutual regulation of their signaling components, which is mediated in part through sphingosine kinases and sphingosine-1-phosphate and indicative of an intimate signaling crosstalk between the two pathways. In the first part of this review, we summarize the various regulatory interactions that have been discovered so far according to the organ/tissue in which they were described. In the second part, we highlight the types of signaling crosstalk between TGF-β on the one hand and PAR2/PAR1 on the other hand. Both ligand–receptor systems interact at various levels and by several mechanisms including mutual regulation of ligand–ligand, ligand–receptor, and receptor–receptor at the transcriptional, post-transcriptional, and receptor transactivation levels. These mutual interactions between PAR2/PAR1 and TGF-β signaling components eventually result in feed-forward loops/vicious cycles of matrix deposition and malignant traits that exacerbate fibrosis and oncogenesis, respectively. Given the crucial role of PAR2 and PAR1 in controlling TGF-β receptor activation, signaling, TGF-β synthesis and bioactivation, combining PAR inhibitors with TGF-β blocking agents may turn out to be more efficient than targeting TGF-β alone in alleviating unwanted TGF-β-dependent responses but retaining the beneficial ones.
Collapse
|
41
|
Shen K, Johnson DW, Vesey DA, McGuckin MA, Gobe GC. Role of the unfolded protein response in determining the fate of tumor cells and the promise of multi-targeted therapies. Cell Stress Chaperones 2018; 23:317-334. [PMID: 28952072 PMCID: PMC5904077 DOI: 10.1007/s12192-017-0844-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 09/13/2017] [Indexed: 02/06/2023] Open
Abstract
Although there have been advances in our understanding of carcinogenesis and development of new treatments, cancer remains a common cause of death. Many regulatory pathways are incompletely understood in cancer development and progression, with a prime example being those related to the endoplasmic reticulum (ER). The pathological sequelae that arise from disruption of ER homeostasis are not well defined. The ER is an organelle that is responsible for secretory protein biosynthesis and the quality control of protein folding. The ER triggers an unfolded protein response (UPR) when misfolded proteins accumulate, and while the UPR acts to restore protein folding and ER homeostasis, this response can work as a switch to determine the death or survival of cells. The treatment of cancer with agents that target the UPR has shown promising outcomes. The UPR has wide crosstalk with other signaling pathways. Multi-targeted cancer therapies which target the intersections within signaling networks have shown synergistic tumoricidal effects. In the present review, the basic cellular and signaling pathways of the ER and UPR are introduced; then the crosstalk between the ER and other signaling pathways is summarized; and ultimately, the evidence that the UPR is a potential target for cancer therapy is discussed. Regulation of the UPR downstream signaling is a common therapeutic target for different tumor types. Tumoricidal effects achieved from modulating the UPR downstream signaling could be enhanced by phosphodiesterase 5 (PDE5) inhibitors. Largely untapped by Western medicine for cancer therapies are Chinese herbal medicines. This review explores and discusses the value of some Chinese herbal extracts as PDE5 inhibitors.
Collapse
Affiliation(s)
- Kunyu Shen
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
| | - David W Johnson
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - David A Vesey
- Department of Nephrology, Princess Alexandra Hospital, Woolloongabba, Brisbane, Australia
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Michael A McGuckin
- Mucosal Disease Inflammatory Disease Biology and Therapeutics Group, UQ Mater Research Institute, Translational Research Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Glenda C Gobe
- Kidney Disease Research Group, UQ Diamantina Institute, Translational Research Institute, The University of Queensland, 37 Kent Street, Woolloongabba, Brisbane, 4102, Australia.
- Centre for Health Services Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
42
|
Pu W, Shang Y, Shao Q, Yuan X. miR-146a promotes cell migration and invasion in melanoma by directly targeting SMAD4. Oncol Lett 2018; 15:7111-7117. [PMID: 29731876 PMCID: PMC5921230 DOI: 10.3892/ol.2018.8172] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 01/23/2018] [Indexed: 01/27/2023] Open
Abstract
Previous studies have explored the functions of microRNA (miR)-146a in different types of cancer through mediating different targets. However, the roles of miR-146a in malignant melanoma (MM) cell migration and invasion remain largely elusive. In the present study, the potential molecular function of miR-146a in MM was investigated. Reverse transcription-quantitative polymerase chain reaction was utilized to detect miR-146a expression in MM tissues and cell lines. A Transwell assay was performed to confirm the ability of migration and invasion. A luciferase assay and biological analysis were used to predict and determine the targets of miR-146a. The expression of miR-146a was upregulated in melanoma tissues and cell lines. Clinicopathological analysis indicated that the miR-146a level was negatively correlated with the progression of melanoma. Abnormal expression of miR-146a promoted cell migration and invasion in MM cells. Additionally, it was also observed that Mothers against decapentaplegic homolog 4 (SMAD4) was a novel target of miR-146a in MM. SMAD4 was negatively associated with miR-146a in MM and abnormal expression of SMAD4 attenuated miR-146a-mediated promotion of cell migration and invasion. In conclusion, miR-146a functioned as an oncogene by directly targeting SMAD4 and it may be a novel diagnostic and therapeutic marker of MM.
Collapse
Affiliation(s)
- Wei Pu
- Department of Dermatology, The Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| | - Yongming Shang
- Department of Dermatology, Zibo Traditional Chinese Medicine Hospital, Zibo, Shandong 255300, P.R. China
| | - Qiang Shao
- Department of Dermatology, The Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| | - Xinpeng Yuan
- Department of Dermatology, The Central Hospital of Zibo, Zibo, Shandong 255000, P.R. China
| |
Collapse
|
43
|
Weidle UH, Birzele F, Kollmorgen G, Nopora A. Potential microRNA-related Targets for Therapeutic Intervention with Ovarian Cancer Metastasis. Cancer Genomics Proteomics 2018; 15:1-15. [PMID: 29275359 PMCID: PMC5822180 DOI: 10.21873/cgp.20061] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 10/09/2017] [Accepted: 10/17/2017] [Indexed: 02/06/2023] Open
Abstract
Treatment of disseminated epithelial ovarian cancer (EOC) is an unmet medical need. Therefore, the identification along with preclinical and clinical validation of new targets is an issue of high importance. In this review we focus on microRNAs that mediate metastasis of EOC. We summarize up-regulated metastasis-promoting and down-regulated metastasis-suppressing microRNAs. We focus on preclinical in vitro and in vivo functions as well as their metastasis-related clinical correlations. Finally, we outline modalities for therapeutic intervention and critical issues of microRNA-based therapeutics in the context of metastatic EOC.
Collapse
Affiliation(s)
- Ulrich H Weidle
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hofman La Roche, Basel, Switzerland
| | - Gwen Kollmorgen
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| | - Adam Nopora
- Roche Innovation Center Munich, Roche Diagnostics GmbH, Penzberg, Germany
| |
Collapse
|
44
|
Ungefroren H, Witte D, Rauch BH, Settmacher U, Lehnert H, Gieseler F, Kaufmann R. Proteinase-Activated Receptor 2 May Drive Cancer Progression by Facilitating TGF-β Signaling. Int J Mol Sci 2017; 18:E2494. [PMID: 29165389 PMCID: PMC5713460 DOI: 10.3390/ijms18112494] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 11/16/2017] [Accepted: 11/20/2017] [Indexed: 12/23/2022] Open
Abstract
The G protein-coupled receptor proteinase-activated receptor 2 (PAR2) has been implicated in various aspects of cellular physiology including inflammation, obesity and cancer. In cancer, it usually acts as a driver of cancer progression in various tumor types by promoting invasion and metastasis in response to activation by serine proteinases. Recently, we discovered another mode through which PAR2 may enhance tumorigenesis: crosstalk with transforming growth factor-β (TGF-β) signaling to promote TGF-β1-induced cell migration/invasion and invasion-associated gene expression in ductal pancreatic adenocarcinoma (PDAC) cells. In this chapter, we review what is known about the cellular TGF-β responses and signaling pathways affected by PAR2 expression, the signaling activities of PAR2 required for promoting TGF-β signaling, and the potential molecular mechanism(s) that underlie(s) the TGF-β signaling-promoting effect. Since PAR2 is activated through various serine proteinases and biased agonists, it may couple TGF-β signaling to a diverse range of other physiological processes that may or may not predispose cells to cancer development such as local inflammation, systemic coagulation and pathogen infection.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany.
- Department of General and Thoracic Surgery, University Hospital Schleswig-Holstein, D-24105 Kiel, Germany.
| | - David Witte
- First Department of Medicine, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany.
| | - Bernhard H Rauch
- Department of General Pharmacology, Institute of Pharmacology, University Medicine Greifswald, D-17487 Greifswald, Germany.
| | - Utz Settmacher
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, D-07747 Jena, Germany.
| | - Hendrik Lehnert
- First Department of Medicine, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany.
| | - Frank Gieseler
- First Department of Medicine, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany.
| | - Roland Kaufmann
- Department of General, Visceral and Vascular Surgery, Jena University Hospital, D-07747 Jena, Germany.
| |
Collapse
|
45
|
Liu Z, Huo X, Zhao S, Yang J, Shi W, Jing L, Li W, Li Y, Ma L, Gao Y, Diao A. Low density lipoprotein receptor class A domain containing 4 (LDLRAD4) promotes tumorigenesis of hepatic cancer cells. Exp Cell Res 2017; 360:189-198. [PMID: 28888937 DOI: 10.1016/j.yexcr.2017.09.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 08/03/2017] [Accepted: 09/02/2017] [Indexed: 02/08/2023]
Abstract
LDLRAD4 was previously identified and shown to be connected with psychiatric disorders. The structure of LDLRAD4 protein is similar to that of TMEPAI protein, which is overexpressed in many tumors. However, it is still unknown whether LDLRAD4 is involved in tumorigenesis. In this study, the potential role of LDLRAD4 in tumorigenesis was investigated. LDLRAD4 is elevated in hepatic cancer cells and tumor tissues, and expression of LDLRAD4 promotes hepatic cancer cell HepG2 and SMMC-7721 proliferation and migration. LDLRAD4 interacts Nedd4 to promote cell proliferation and migration and negatively regulates the TGF-β signaling. Furthermore, immunofluorescence microscopy analysis indicates that LDLRAD4 is localized to the lysosome and association with Nedd4 is necessary for its intracellular transport to the lysosome. In addition, depletion of LDLRAD4 in HepG2 liver cancer cells inhibited tumorigenesis in nude mice. These results reveal an oncogenic role of LDLRAD4 in tumorigenesis through its association with Nedd4.
Collapse
Affiliation(s)
- Zhenxing Liu
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Xin Huo
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Shuang Zhao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Jingjing Yang
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Wenxia Shi
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, No. 83, Jintang Road, Hedong District, Tianjin 300170, China
| | - Lei Jing
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Wei Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Yuyin Li
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Long Ma
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China
| | - Yingtang Gao
- Tianjin Key Laboratory of Artificial Cell, Tianjin Institute of Hepatobiliary Disease, Artificial Cell Engineering Technology Research Center of Public Health Ministry, Third Central Hospital of Tianjin, No. 83, Jintang Road, Hedong District, Tianjin 300170, China.
| | - Aipo Diao
- School of Biotechnology, Tianjin University of Science and Technology, Key Lab of Industrial Fermentation Microbiology of the Ministry of Education, No. 29, 13th Avenue, TEDA District, Tianjin 300457, China.
| |
Collapse
|
46
|
Jia X, Shanmugam C, Paluri RK, Jhala NC, Behring MP, Katkoori VR, Sugandha SP, Bae S, Samuel T, Manne U. Prognostic value of loss of heterozygosity and sub-cellular localization of SMAD4 varies with tumor stage in colorectal cancer. Oncotarget 2017; 8:20198-20212. [PMID: 28423626 PMCID: PMC5386755 DOI: 10.18632/oncotarget.15560] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 01/24/2017] [Indexed: 12/24/2022] Open
Abstract
Background Although loss of heterozygosity (LOH) at chromosome location 18q21 and decreased expression of SMAD4 in invasive colorectal cancers (CRCs) correlate with poor patient survival, the prognostic value of LOH at 18q21 and sub-cellular localization of SMAD4 have not been evaluated in relation to tumor stage. Methods Genomic DNA samples from 209 formalin-fixed, paraffin-embedded sporadic CRC tissues and their matching controls were analyzed for 18q21 LOH, and corresponding tissue sections were evaluated by immunohistochemistry for expression of SMAD4 and assessed for its sub-cellular localization (nuclear vs. cytoplasmic). In addition, 53 frozen CRCs and their matching control tissues were analyzed for their mutational status and mRNA expression of SMAD4. The phenotypic expression pattern and LOH status were evaluated for correlation with patient survival by the use of Kaplan-Meier and Cox regression models. Results LOH of 18q21 was detected in 61% of the informative cases. In 8% of the cases, missense point mutations were detected in Smad4. In CRCs, relative to controls, there was increased SMAD4 staining in the cytoplasm (74%) and decreased staining in the nuclei (37%). LOH of 18q21 and high cytoplasmic localization of SMAD4 were associated with shortened overall survival of Stage II patients, whereas low nuclear expression of SMAD4 was associated with worse survival, but only for patients with Stage III CRCs. Conclusions LOH of 18q21 and high cytoplasmic localization of SMAD4 in Stage II CRCs and low nuclear SMAD4 in Stage III CRCs are predictors of shortened patient survival.
Collapse
Affiliation(s)
- Xu Jia
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chandrakumar Shanmugam
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Current address: Department of Pathology, ESIC Medical College and Hospital, Sanathnagar, Hyderabad, Telangana, India
| | - Ravi K Paluri
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nirag C Jhala
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Current address: Pathology & Laboratory Medicine, Temple University, Philadelphia, PA, USA
| | - Michael P Behring
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Venkat R Katkoori
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Current address: Department of Surgery, Michigan State University, College of Human Medicine, Lansing, MI, USA
| | - Shajan P Sugandha
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sejong Bae
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Temesgen Samuel
- College of Veterinary Medicine, Tuskegee University, Tuskegee, AL, USA
| | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.,Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
47
|
Slattery ML, Trivellas A, Pellatt AJ, Mullany LE, Stevens JR, Wolff RK, Herrick JS. Genetic variants in the TGFβ-signaling pathway influence expression of miRNAs in colon and rectal normal mucosa and tumor tissue. Oncotarget 2017; 8:16765-16783. [PMID: 28061442 PMCID: PMC5370000 DOI: 10.18632/oncotarget.14508] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 12/16/2016] [Indexed: 01/04/2023] Open
Abstract
The TGF-β signaling pathway is involved in regulation of cell growth, angiogenesis, and metastasis. We test the hypothesis that genetic variation in the TGF-β signaling pathway alters miRNA expression.We use data from 1188 colorectal cancer cases to evaluate associations between 80 SNPs in 21 genes.Seven variants eIF4E rs12498533, NFκB1 rs230510, TGFB1 rs4803455, TGFBR1 rs1571590 and rs6478974, SMAD3 rs3743343, and RUNX1 rs8134179 were associated with expression level of miRNAs in normal colorectal mucosa. RUNX2 rs12333172 and BMPR1B rs13134042 were associated with miRNAs in normal colon mucosa; eIF4EBP3 rs250425, SMAD3 rs12904944, SMAD7 rs3736242, and PTEN rs532678 were associated with miRNA expression in normal rectal mucosa. Evaluation of the differential expression between carcinoma and normal mucosa showed that SMAD3 rs12708491 and rs2414937, NFκB1 rs230510 and rs3821958, and RUNX3 rs6672420 were associated with several miRNAs for colorectal carcinoma. Evaluation of site-specific differential miRNA expression showed that BMPR1B rs2120834, BMPR2 rs2228545, and eIF4EBP3 rs250425 were associated with differential miRNA expression in colon tissue and SMAD3 rs12901071, rs1498506, and rs2414937, BMPR2 rs2228545, and RUNX2 rs2819854, altered differential miRNA expression in rectal tissue.These data support the importance of the TGF-β signaling pathway to the carcinogenic process, possibly through their influence on miRNA expression levels.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | | | | | - Lila E Mullany
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - John R Stevens
- Department of Mathematics and Statistics, Utah State University, Logan, Utah, USA
| | - Roger K Wolff
- Department of Medicine, University of Utah, Salt Lake City, Utah, USA
| | | |
Collapse
|
48
|
Song C, Liu W, Li J. USP17 is upregulated in osteosarcoma and promotes cell proliferation, metastasis, and epithelial–mesenchymal transition through stabilizing SMAD4. Tumour Biol 2017; 39:1010428317717138. [PMID: 28670958 DOI: 10.1177/1010428317717138] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
USP17 is upregulated in several cancers, indicating that USP17 might play essential functions in tumor development. However, the function of USP17 in osteosarcoma is still unknown. Our work aimed to investigate the function of USP17 in osteosarcoma. We found that the expression of USP17 was upregulated in osteosarcoma tissues and cell lines, including MG-63 and U2OS. Several functional experiments, such as colony formation analysis, Cell Counting Kit-8 assay, wound healing analysis, and transwell assay, showed that USP17 promoted cell proliferation, migration, and invasion. Moreover, we found that USP17 facilitated migration and invasion through promoting epithelial–mesenchymal transition. SMAD4 has been found to regulate epithelial–mesenchymal transition, co-immunopurification, and glutathione S-transferase pull-down analysis demonstrated that USP17 interacted with SMAD4. Furthermore, USP17 stabilized SMAD4 through its deubiquitinase activity. In conclusion, this study shows that USP17 enhances osteosarcoma cell proliferation and invasion through stabilizing SMAD4.
Collapse
Affiliation(s)
- Chenyang Song
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Wenge Liu
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jiandong Li
- Department of Orthopedics, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
49
|
Ruan C, Lu J, Wang H, Ge Z, Zhang C, Xu M. miR-26b-5p regulates hypoxia-induced phenotypic switching of vascular smooth muscle cells via the TGF-β/Smad4 signaling pathway. Mol Med Rep 2017; 15:4185-4190. [PMID: 28487943 DOI: 10.3892/mmr.2017.6509] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 07/14/2016] [Indexed: 11/06/2022] Open
Abstract
Hypoxia contributes to the phenotypic switch of vascular smooth muscle cells (VSMCs). Various microRNAs (miRNAs) participate in this process as post‑transcriptional regulators, however the mechanism remains unclear. In the present study, mouse VSMCs (mVSMCs) harvested from aortas were cultured in normoxic and hypoxic conditions, and the mRNA levels of miR-26b-5p, desmin, H‑caldesmon and smoothelin were quantified using reverse transcription‑quantitative polymerase chain reaction. Following treatment with a miR‑26b‑5p antagonist (agomir) or non‑targeting control (scramble), the cell areas of normoxic and hypoxic mVSMCs were analyzed by immunofluorescence staining. In addition, the protein expression levels of collagen Iα, Smad2/phosphorylated (p)‑Smad2, Smad3/p‑Smad3 and Smad4 were determined by western blotting. Potential miRNA26b‑5p binding sequences in the 3'‑untranslated region (UTR) of Smad4 were investigated, and the distribution of Smad4 in mVSMCs was visualized using immunofluorescence methods. Hypoxic mVSMCs exhibited a significant downregulation miR‑26b‑5p, upregulation of hypoxia inducible factor‑1α mRNA and suppression of desmin, H‑caldesmon and smoothelin mRNA levels. Additionally, miR‑26b‑5p agomir reduced the cell area and decreased collagen Iα expression levels in hypoxic mVSMCs compared with normoxic mVSMCs transfected with agomir, and the area was comparable with those of normoxic mVSMCs transfected with agomir or scramble. Furthermore, miR‑26b‑5p suppressed Smad4 expression in hypoxic mVSMCs, but did not change the expression levels of Smad2 and Smad3, p‑Smad2 and p‑Smad3, however p‑Smad2 and p‑Smad3 levels were upregulated in response to hypoxic stimuli. Additionally, the miR‑26b‑5p agomir caused weak immunoreactivity with Smad4 in hypoxic mVSMCs. The binding motif of miR‑26b‑5p in the Smad4 3'‑UTR was identified as UACUUGA at position 978-984. These findings suggest that miR‑26b‑5p regulates hypoxia‑induced phenotypic switching of VSMCs via the transforming growth factor β/Smad4 signaling pathway.
Collapse
Affiliation(s)
- Changwu Ruan
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| | - Jide Lu
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| | - Hairong Wang
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| | - Zhiru Ge
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| | - Chenjun Zhang
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| | - Maochun Xu
- Department of Cardiology, Gongli Hospital, Shanghai 200135, P.R. China
| |
Collapse
|
50
|
Zhao H, Zhang J, Shao H, Liu J, Jin M, Chen J, Huang Y. Transforming Growth Factor β1/Smad4 Signaling Affects Osteoclast Differentiation via Regulation of miR-155 Expression. Mol Cells 2017; 40:211-221. [PMID: 28359146 PMCID: PMC5386959 DOI: 10.14348/molcells.2017.2303] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 02/20/2017] [Accepted: 02/22/2017] [Indexed: 01/02/2023] Open
Abstract
Transforming growth factor β1 (TGFβ1)/Smad4 signaling plays a pivotal role in maintenance of the dynamic balance between bone formation and resorption. The microRNA miR-155 has been reported to exert a significant role in the differentiation of macrophage and dendritic cells. The goal of this study was to determine whether miR-155 regulates osteoclast differentiation through TGFβ1/Smad4 signaling. Here, we present that TGFβ1 elevated miR-155 levels during osteoclast differentiation through the stimulation of M-CSF and RANKL. Additionally, we found that silencing Smad4 attenuated the upregulation of miR-155 induced by TGFβ1. The results of luciferase reporter experiments and ChIP assays demonstrated that TGFβ1 promoted the binding of Smad4 to the miR-155 promoter at a site located in 454 bp from the transcription start site in vivo, further verifying that miR-155 is a transcriptional target of the TGFβ1/Smad4 pathway. Subsequently, TRAP staining and qRT-PCR analysis revealed that silencing Smad4 impaired the TGFβ1-mediated inhibition on osteoclast differentiation. Finally, we found that miR-155 may target SOCS1 and MITF to suppress osteoclast differentiation. Taken together, we provide the first evidence that TGFβ1/Smad4 signaling affects osteoclast differentiation by regulation of miR-155 expression and the use of miR-155 as a potential therapeutic target for osteoclast-related diseases shows great promise.
Collapse
Affiliation(s)
- Hongying Zhao
- Department of Pharmacy, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Jun Zhang
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Haiyu Shao
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Jianwen Liu
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Mengran Jin
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Jinping Chen
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| | - Yazeng Huang
- Department of Orthopedics, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang,
China
| |
Collapse
|