1
|
Chen S, Zhao C, Liu R, Jiao W. A bibliometric analysis of malignant pleural mesothelioma from 2010 to 2023. J Thorac Dis 2025; 17:2014-2027. [PMID: 40400943 PMCID: PMC12090152 DOI: 10.21037/jtd-24-1778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Accepted: 03/07/2025] [Indexed: 05/23/2025]
Abstract
Background Malignant pleural mesothelioma (MPM) is an aggressive tumor originating from the mesothelial lining of the pleural cavity. It is characterized by extensive nodular pleural thickening and has a propensity to invade the pleural adipose tissue and adjacent chest structures. The prognosis is poor, with a median survival time rarely exceeding 12 months following diagnosis. Methods This bibliometric analysis systematically assessed global trends in MPM research from 2010 to 2023 using 6,487 publications indexed in PubMed. Quantitative evaluations of publication metrics, international collaboration, and keyword co-occurrence networks were conducted using R software with the bibliometrix package. Network construction and thematic mapping were employed to analyze the temporal evolution of research topics. Results The United States and Europe have played pivotal roles in this research, while contributions from China and Japan have been steadily increasing. Traditional treatment approaches and etiological studies are relatively well-established. Meanwhile, immunotherapy has emerged as a prominent focus of recent research. Conclusions Future global collaboration in this field should be enhanced, as precision medicine related to immunology and genetics has the potential to transform the treatment landscape of MPM.
Collapse
Affiliation(s)
- Sheng Chen
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ce Zhao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruiqi Liu
- Department of Radiology, The First People’s Hospital of Fuyang, Fuyang, China
| | - Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
2
|
Zang MX, Zhang G, Zhang Y, Wang SS, Zhai XW, Zhao N, Ge W, Xie JW, Shen W, Cheng SF. mTORC1 regulates the proliferation of SOX9 + porcine skin-derived stem cells (pSDSCs) by promoting S6K phosphorylation. Histochem Cell Biol 2025; 163:25. [PMID: 39833550 DOI: 10.1007/s00418-025-02354-9] [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] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Skin-derived stem cells (SDSCs) are a subtype of adult stem cells (ASCs) that are widely harvested and exempt from ethical restrictions in clinical applications. These cells possess capabilities for self-renewal, proliferation, and multi-lineage differentiation. Compared to model animals like rats and mice, pigs exhibit greater physiological similarity to humans. Porcine skin has very similar histological and physiological characteristics to human skin. Therefore, porcine skin is becoming increasingly significant as an in vitro model for research. In this study, porcine skin-derived stem cells (pSDSCs) were isolated and cultured in vitro for experiments. The expression of stemness-related gene SOX9 was detected. RNA sequencing (RNA-seq) results found that the mammalian target of rapamycin (mTOR) signaling pathway was significantly enriched in SOX9+ pSDSCs. To investigate the role of the mTOR signaling pathway, we added rapamycin (RAPA), an inhibitor of the mTOR complex 1 (mTORC1), and found that the proliferation rate of SOX9+ pSDSCs decreased significantly during culture. In addition, western blotting (WB) results demonstrated that mTORC1 promoted proliferation by phosphorylating S6 kinase (S6K) and then activating cyclin D1(CCND1) in SOX9+ pSDSCs. These findings provide insights into the mechanisms of adult stem cell proliferation.
Collapse
Affiliation(s)
- Ming-Xin Zang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Geng Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Ying Zhang
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Sha-Sha Wang
- Qingdao Animal Husbandry and Veterinary Institute, Qingdao, 266000, Shandong, China
| | - Xiang-Wei Zhai
- Animal Husbandry General Station of Shandong Province, Jinan, 250010, China
| | - Na Zhao
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Wei Ge
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Jin-Wen Xie
- Shandong Binzhou Animal Science and Veterinary Medicine Academy, Binzhou, 256600, China
| | - Wei Shen
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China
| | - Shun-Feng Cheng
- College of Life Sciences, Key Laboratory of Animal Reproduction and Biotechnology in Universities of Shandong, Qingdao Agricultural University, Qingdao, 266109, China.
| |
Collapse
|
3
|
Niclosamide Suppresses Migration and Invasion of Human Osteosarcoma Cells by Repressing TGFBI Expression via the ERK Signaling Pathway. Int J Mol Sci 2022; 23:ijms23010484. [PMID: 35008910 PMCID: PMC8745393 DOI: 10.3390/ijms23010484] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 12/30/2021] [Accepted: 12/31/2021] [Indexed: 02/06/2023] Open
Abstract
Osteosarcoma is a highly common malignant bone tumor. Its highly metastatic properties are the leading cause of mortality for cancer. Niclosamide, a salicylanilide derivative, is an oral antihelminthic drug of known anticancer potential. However, the effect of niclosamide on osteosarcoma cell migration, invasion and the mechanisms underlying have not been fully clarified. Therefore, this study investigated niclosamide’s underlying pathways and antimetastatic effects on osteosarcoma. In this study, U2OS and HOS osteosarcoma cell lines were treated with niclosamide and then subjected to assays for determining cell migration ability. The results indicated that niclosamide, at concentrations of up to 200 nM, inhibited the migration and invasion of human osteosarcoma U2OS and HOS cells and repressed the transforming growth factor beta-induced protein (TGFBI) expression of U2OS cells, without cytotoxicity. After TGFBI knockdown occurred, cellular migration and invasion behaviors of U2OS cells were significantly reduced. Moreover, niclosamide significantly decreased the phosphorylation of ERK1/2 in U2OS cells and the combination treatment of the MEK inhibitor (U0126) and niclosamide resulted in the intensive inhibition of the TGFBI expression and the migratory ability in U2OS cells. Therefore, TGFBI derived from osteosarcoma cells via the ERK pathway contributed to cellular migration and invasion and niclosamide inhibited these processes. These findings indicate that niclosamide may be a powerful preventive agent against the development and metastasis of osteosarcoma.
Collapse
|
4
|
Chen Y, Zhao H, Feng Y, Ye Q, Hu J, Guo Y, Feng Y. Pan-Cancer Analysis of the Associations of TGFBI Expression With Prognosis and Immune Characteristics. Front Mol Biosci 2021; 8:745649. [PMID: 34671645 PMCID: PMC8521171 DOI: 10.3389/fmolb.2021.745649] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 09/17/2021] [Indexed: 01/25/2023] Open
Abstract
Transforming growth factor-beta-induced (TGFBI) protein has important roles in tumor growth, metastasis, and immunity. However, there is currently no pan-cancer evidence regarding TGFBI. In this study, we conducted a pan-cancer analysis of TGFBI mRNA and protein expression and prognoses of various cancer types using public databases. We also investigated the associations of TGFBI expression with tumor microenvironment (TME) components, immune cell infiltration, tumor mutational burden (TMB), and microsatellite instability (MSI), along with the TGFBI genetic alteration types. The results showed that TGFBI expression varied among different cancer types, and it was positively or negatively related to prognosis in various cancers. TGFBI expression was also significantly correlated with TME components, TMB, MSI, immune cell infiltration, and immunoinhibitory and immunostimulatory gene subsets. These findings indicate that TGFBI participates in various immune responses and it may function as a prognostic marker in various cancers. The findings may be useful for developing immunotherapies that target TGFBI.
Collapse
Affiliation(s)
- Yun Chen
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Han Zhao
- Department of Ophthalmology, Eye, Ear, Nose, and Throat Hospital of Fudan University, Shanghai, China
- Laboratory of Myopia, NHC Key Laboratory of Myopia (Fudan University), Chinese Academy of Medical Sciences, Shanghai, China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Fudan University, Shanghai, China
| | - Yao Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Qin Ye
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Jing Hu
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yue Guo
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yunzhi Feng
- Department of Stomatology, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
5
|
Kim HJ, Ahn D, Park TI, Jeong JY. TGFBI Expression Predicts the Survival of Patients With Oropharyngeal Squamous Cell Carcinoma. In Vivo 2021; 34:3005-3012. [PMID: 32871844 DOI: 10.21873/invivo.12132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM This study was conducted to investigate transforming growth factor beta-induced protein (TGFBI) expression and analyze the clinical and prognostic significance of TGFBI in oropharyngeal squamous cell carcinoma (OPSCC). PATIENTS AND METHODS We evaluated TGFBI expression by immunohistochemistry in 94 patients with OPSCC. For comprehensive analysis, TGFBI expression was subdivided into tumor cell score (T), stroma score (S), and the sum of two scores (TS) calculated using H-score. Clinicopathological features and survival outcomes were compared between groups of high expression and low expression of TGFBI in each area. RESULTS Overall, 12 patients (12.8%) showed high T score, and 41 patients (43.6%) revealed high S score. Although T score showed no significant difference both in overall survival (OS) (p=0.080) and recurrence free survival (RFS) (p=0.272), high S score patients had significantly worse OS (p=0.003) and worse RFS (p=0.043). High TS score also showed significant association with worse OS (p=0.011) and worse RFS (p=0.021). High S score was an independent prognostic factor predicting shorter OS (HR=6.352, 95%CI=1.206-40.050, p=0.029) and RFS (HR=18.843, 95%CI=1.030-344.799, p=0.048) in the multivariate analysis. CONCLUSION High S score of TGFBI was a significant predictor of poor prognosis in OPSCC. TGFBI could be a useful new predictive and prognostic biomarker in OPSCC.
Collapse
Affiliation(s)
- Ha-Jeong Kim
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Dongbin Ahn
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Tae-In Park
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Ji Yun Jeong
- Department of Pathology, School of Medicine, Kyungpook National University, Daegu, Republic of Korea .,Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu, Republic of Korea
| |
Collapse
|
6
|
Ning XJ, Lu XH, Luo JC, Chen C, Gao Q, Li ZY, Wang H. Molecular mechanism of microRNA-21 promoting Schwann cell proliferation and axon regeneration during injured nerve repair. RNA Biol 2020; 17:1508-1519. [PMID: 32507001 DOI: 10.1080/15476286.2020.1777767] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
At present, the functional recovery after nerve injury is not satisfactory in clinical practice. The aim of this study was to explore the molecular mechanism of miR-21 promoting Schwann cells (SC) proliferation and axon regeneration after peripheral nerve injury, providing a theoretical basis for injured nerve repair. Nerve injury models were constructed to determine the expression of miR-21 in the injured nerve by Quantitative Real-Time PCR (qRT-PCR). After miR-21 over-expression SC (mimic-miR-21) group, control SC (control-miR-21) group and blank SC (RSC96) group were constructed, SC proliferation was determined by CCK-8, cell cycle was analysed by flow cytometry, dorsal root ganglion neuron (DRGn) axon regeneration was observed after DRGn was cultured with SCs for 7 days, the expressions of TGFβI, TIMP3, EPHA4 as well as apoptosis-related proteins caspase-3 and caspase-9 were detected by qRT-PCR and Western blot in the three groups, respectively. Target genes were confirmed by dual-luciferase reporter gene assay. The expressions of TGFβI, TIMP3 and EPHA4 were assessed by immunofluorescence in vivo. qRT-PCR indicated that miR-21 expression was significantly higher in the model group than in the sham operation and blank groups. SC proliferation index (PI) was significantly higher, the apoptosis rate was significantly lower, the axon was significantly longer, and mRNA and protein expressions of TGFβI, TIMP3, EPHA4 as well as apoptosis-related proteins caspase-3 and caspase-9 were significantly lower in the mimic-miR-21 group than in the control-miR-21 and RSC96 groups. The double luciferase assay confirmed that TGFβI, TIMP3 and EPHA4 were potential target genes of miR-21. In vivo immunofluorescence also indicated that expressions of TGFβI, TIMP3, EPHA4 were lower in the mimic-miR-21 group than in the control-miR-21 and RSC96 groups. We conclude that during injured peripheral nerve repair, miRNA-21 plays an important role in promoting SC proliferation and axon regeneration by regulating TGFβI, TIMP3 and EPHA4 target genes.
Collapse
Affiliation(s)
- Xin-Jie Ning
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Xin-Hua Lu
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Jun-Cheng Luo
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Qun Gao
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Zhang-Yu Li
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University , Guangzhou, China
| |
Collapse
|
7
|
Wang BJ, Chi KP, Shen RL, Zheng SW, Guo Y, Li JF, Fei J, He Y. TGFBI Promotes Tumor Growth and is Associated with Poor Prognosis in Oral Squamous Cell Carcinoma. J Cancer 2019; 10:4902-4912. [PMID: 31598162 PMCID: PMC6775518 DOI: 10.7150/jca.29958] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Purpose: In a previous study, we found that transforming growth factor beta-induced (TGFBI) is a hub gene strongly associated with oral squamous cell carcinoma (OSCC), using gene chip meta-analysis and PPI network analysis. Thus, the present study was established to explore the role of TGFBI in the pathogenesis of OSCC and to define the underlying mechanisms. Methods: The correlations between TGFBI expression and the clinicopathological features and prognosis of OSCC were analyzed. Then, TGFBI-knockout HSC-3 cell lines were constructed using the CRISPR/Cas9 system. Cell proliferation, migration, and invasion in vitro were determined by cell counting, CCK-8, colony formation, and Transwell assays. Moreover, a xenograft animal study was implemented to determine the tumorigenicity and metastatic ability associated with TGFBI in vivo. The genes and pathways differentially expressed after TGFBI knockout were determined using transcriptional sequencing and bioinformatics. Results: TGFBI expression was significantly higher in OSCC than in normal tissue. Its high expression was also correlated with high stage and was predictive of poor prognosis, as we expected. Knockout of TGFBI inhibited cell proliferation and clone formation, and enhanced cell migration and invasion in vitro. Besides, the xenograft animal study showed that TGFBI knockout suppressed tumor growth and metastasis in vivo. Furthermore, transcriptome sequencing revealed that genes associated with cell proliferation, metastasis, and inflammatory responses exhibited a change of expression upon TGFBI knockout. GO and KEGG analyses indicated that the function of TGFBI is related to responses to bacteria and inflammatory responses. Conclusions: TGFBI overexpression can promote OSCC and is associated with poor prognosis in OSCC patients. TGFBI knockout can inhibit cell proliferation and metastasis in vivo. TGFBI may alter cell responses to bacteria, which causes an imbalance in the immune inflammatory response and promotes the development of OSCC.
Collapse
Affiliation(s)
- Bing-Jie Wang
- Department of Oral Medicine, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China.,Department of Stomatology, Ningbo Yinzhou People's Hospital, Zhejiang 315040, China
| | - Kun-Ping Chi
- Department of Pathology, First people's Hospital of Yunnan Province, Yunnan, 650032, China
| | - Ru-Ling Shen
- Shanghai Laboratory Animal Research Center, Shanghai 201203, China
| | - Sai-Wei Zheng
- Department of Oral Medicine, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| | - Yang Guo
- School of Life Science and Technology, Tongji University, Shanghai 200082, China
| | - Jian-Feng Li
- School of Life Science and Technology, Tongji University, Shanghai 200082, China
| | - Jian Fei
- School of Life Science and Technology, Tongji University, Shanghai 200082, China
| | - Yuan He
- Department of Oral Medicine, School & Hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, 200072, China
| |
Collapse
|
8
|
Palomeras S, Diaz-Lagares Á, Viñas G, Setien F, Ferreira HJ, Oliveras G, Crujeiras AB, Hernández A, Lum DH, Welm AL, Esteller M, Puig T. Epigenetic silencing of TGFBI confers resistance to trastuzumab in human breast cancer. Breast Cancer Res 2019; 21:79. [PMID: 31277676 PMCID: PMC6612099 DOI: 10.1186/s13058-019-1160-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022] Open
Abstract
Background Acquired resistance to trastuzumab is a major clinical problem in the treatment of HER2-positive (HER2+) breast cancer patients. The selection of trastuzumab-resistant patients is a great challenge of precision oncology. The aim of this study was to identify novel epigenetic biomarkers associated to trastuzumab resistance in HER2+ BC patients. Methods We performed a genome-wide DNA methylation (450K array) and a transcriptomic analysis (RNA-Seq) comparing trastuzumab-sensitive (SK) and trastuzumab-resistant (SKTR) HER2+ human breast cancer cell models. The methylation and expression levels of candidate genes were validated by bisulfite pyrosequencing and qRT-PCR, respectively. Functional assays were conducted in the SK and SKTR models by gene silencing and overexpression. Methylation analysis in 24 HER2+ human BC samples with complete response or non-response to trastuzumab-based treatment was conducted by bisulfite pyrosequencing. Results Epigenomic and transcriptomic analysis revealed the consistent hypermethylation and downregulation of TGFBI, CXCL2, and SLC38A1 genes in association with trastuzumab resistance. The DNA methylation and expression levels of these genes were validated in both sensitive and resistant models analyzed. Of the genes, TGFBI presented the highest hypermethylation-associated silencing both at the transcriptional and protein level. Ectopic expression of TGFBI in the SKTR model suggest an increased sensitivity to trastuzumab treatment. In primary tumors, TGFBI hypermethylation was significantly associated with trastuzumab resistance in HER2+ breast cancer patients. Conclusions Our results suggest for the first time an association between the epigenetic silencing of TGFBI by DNA methylation and trastuzumab resistance in HER2+ cell models. These results provide the basis for further clinical studies to validate the hypermethylation of TGFBI promoter as a biomarker of trastuzumab resistance in HER2+ breast cancer patients. Electronic supplementary material The online version of this article (10.1186/s13058-019-1160-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sònia Palomeras
- New Therapeutics Targets Lab (TargetsLab), Department of Medical Sciences, University of Girona, E-17071, Girona, Catalonia, Spain
| | - Ángel Diaz-Lagares
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Catalonia, Spain.,Cancer Epigenomics, Translational Medical Oncology (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago(CHUS/SERGAS), CIBERONC, Santiago de Compostela, Spain
| | - Gemma Viñas
- New Therapeutics Targets Lab (TargetsLab), Department of Medical Sciences, University of Girona, E-17071, Girona, Catalonia, Spain.,Medical Oncology Department, Catalan Institute of Oncology (ICO), Girona, Catalonia, Spain.,Girona Biomedical Research Institute (IDIBGI), E-17071, Girona, Catalonia, Spain
| | - Fernando Setien
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Humberto J Ferreira
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Catalonia, Spain
| | - Glòria Oliveras
- New Therapeutics Targets Lab (TargetsLab), Department of Medical Sciences, University of Girona, E-17071, Girona, Catalonia, Spain.,Pathology Department, Dr. Josep Trueta Hospital and Catalan Institute of Health (ICS), E-17071, Girona, Catalonia, Spain
| | - Ana B Crujeiras
- Laboratory of Epigenomics in Endocrinology and Nutrition, Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago (CHUS/SERGAS), Santiago de Compostela, Spain.,CIBER Fisiopatologia de la Obesidad y Nutricion (CIBERobn), Santiago de Compostela, Spain
| | - Alejandro Hernández
- Medical Oncology Department, Catalan Institute of Oncology (ICO), Girona, Catalonia, Spain
| | - David H Lum
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - Alana L Welm
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, USA
| | - Manel Esteller
- Cancer Epigenetics and Biology Program (PEBC), Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Barcelona, Catalonia, Spain. .,Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain. .,Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain. .,Institucio Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, Spain. .,Josep Carreras Leukaemia Research Institute (IJC), Badalona, Barcelona, Catalonia, Spain.
| | - Teresa Puig
- New Therapeutics Targets Lab (TargetsLab), Department of Medical Sciences, University of Girona, E-17071, Girona, Catalonia, Spain.
| |
Collapse
|
9
|
Klamer SE, Dorland YL, Kleijer M, Geerts D, Lento WE, van der Schoot CE, von Lindern M, Voermans C. TGFBI Expressed by Bone Marrow Niche Cells and Hematopoietic Stem and Progenitor Cells Regulates Hematopoiesis. Stem Cells Dev 2018; 27:1494-1506. [PMID: 30084753 PMCID: PMC6209430 DOI: 10.1089/scd.2018.0124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The interactions of hematopoietic stem and progenitor cells (HSPCs) with extracellular matrix (ECM) components and cells from the bone marrow (BM) microenvironment control their homeostasis. Regenerative BM conditions can induce expression of the ECM protein transforming growth factor beta-induced gene H3 (TGFBI or BIGH3) in murine HSPCs. In this study, we examined how increased or reduced TGFBI expression in human HSPCs and BM mesenchymal stromal cells (MSCs) affects HSPC maintenance, differentiation, and migration. HSPCs that overexpressed TGFBI showed accelerated megakaryopoiesis, whereas granulocyte differentiation and proliferation of granulocyte, erythrocyte, and monocyte cultures were reduced. In addition, both upregulation and downregulation of TGFBI expression impaired HSPC colony-forming capacity of HSPCs. Interestingly, the colony-forming capacity of HSPCs with reduced TGFBI levels was increased after long-term co-culture with MSCs, as measured by long-term culture-colony forming cell (LTC-CFC) formation. Moreover, TGFBI downregulation in HSPCs resulted in increased cobblestone area-forming cell (CAFC) frequency, a measure for hematopoietic stem cell (HSC) capacity. Concordantly, TGFBI upregulation in HSPCs resulted in a decrease of CAFC and LTC-CFC frequency. These results indicate that reduced TGFBI levels in HSPCs enhanced HSC maintenance, but only in the presence of MSCs. In addition, reduced levels of TGFBI in MSCs affected MSC/HSPC interaction, as observed by an increased migration of HSPCs under the stromal layer. In conclusion, tight regulation of TGFBI expression in the BM niche is essential for balanced HSPC proliferation and differentiation.
Collapse
Affiliation(s)
- Sofieke E Klamer
- 1 Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| | - Yvonne L Dorland
- 2 Sanquin Research and Landsteiner Laboratory, Department of Molecular and Cellular Hemostasis, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| | - Marion Kleijer
- 1 Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| | - Dirk Geerts
- 3 Department of Medical Biology, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| | - William E Lento
- 4 Department of Pharmacology, Duke University , Durham, North Carolina
| | - C Ellen van der Schoot
- 5 Sanquin Research and Landsteiner Laboratory, Department of Experimental Immunohematology, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands .,6 Department of Hematology, Academic Medical Center , Amsterdam, the Netherlands
| | - Marieke von Lindern
- 1 Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| | - Carlijn Voermans
- 1 Sanquin Research and Landsteiner Laboratory, Department of Hematopoiesis, Academic Medical Center, University of Amsterdam , Amsterdam, the Netherlands
| |
Collapse
|
10
|
Bissey PA, Law JH, Bruce JP, Shi W, Renoult A, Chua MLK, Yip KW, Liu FF. Dysregulation of the MiR-449b target TGFBI alters the TGFβ pathway to induce cisplatin resistance in nasopharyngeal carcinoma. Oncogenesis 2018; 7:40. [PMID: 29795279 PMCID: PMC5966388 DOI: 10.1038/s41389-018-0050-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 02/26/2018] [Accepted: 04/11/2018] [Indexed: 01/07/2023] Open
Abstract
Despite the improvement in locoregional control of nasopharyngeal carcinoma (NPC), distant metastasis (DM), and chemoresistance persist as major causes of mortality. This study identified a novel role for miR-449b, an overexpressed gene in a validated four-miRNA signature for NPC DM, leading to chemoresistance via the direct targeting of transforming growth factor beta-induced (TGFBI). In vitro shRNA-mediated downregulation of TGFBI induced phosphorylation of PTEN and AKT, increasing cisplatin resistance. Conversely, the overexpression of TGFBI sensitized the NPC cells to cisplatin. In NPC patients treated with concurrent chemoradiotherapy (CRT), the overall survival (OS) was significantly inversely correlated with miR-449b, and directly correlated with both TGFBI mRNA and protein expression, as assessed by RNA sequencing and immunohistochemistry (IHC). Mechanistically, co-immunoprecipitation demonstrated that TGFBI competes with pro-TGFβ1 for integrin receptor binding. Decreased TGFBI led to increased pro-TGFβ1 activation and TGFβ1 canonical/noncanonical pathway-induced cisplatin resistance. Thus, overexpression of miR-449b decreases TGFBI, thereby altering the balance between TGFBI and pro-TGFβ1, revealing a novel mechanism of chemoresistance in NPC.
Collapse
Affiliation(s)
| | - Jacqueline H Law
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
| | - Jeff P Bruce
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Wei Shi
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Aline Renoult
- LabEx DEVweCAN, Université de Lyon, F-69000, Lyon, France
| | - Melvin L K Chua
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.,Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.,Division of Radiation Oncology, National Cancer Centre, Singapore, Singapore.,Duke-NUS Graduate School, Singapore, Singapore
| | - Kenneth W Yip
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Fei-Fei Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada. .,Department of Radiation Oncology, Princess Margaret Cancer Centre and University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
11
|
Carbone M, Kanodia S, Chao A, Miller A, Wali A, Weissman D, Adjei A, Baumann F, Boffetta P, Buck B, de Perrot M, Dogan AU, Gavett S, Gualtieri A, Hassan R, Hesdorffer M, Hirsch FR, Larson D, Mao W, Masten S, Pass HI, Peto J, Pira E, Steele I, Tsao A, Woodard GA, Yang H, Malik S. Consensus Report of the 2015 Weinman International Conference on Mesothelioma. J Thorac Oncol 2017; 11:1246-1262. [PMID: 27453164 PMCID: PMC5551435 DOI: 10.1016/j.jtho.2016.04.028] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/31/2016] [Accepted: 04/26/2016] [Indexed: 01/31/2023]
Abstract
On November 9 and 10, 2015, the International Conference on Mesothelioma in Populations Exposed to Naturally Occurring Asbestiform Fibers was held at the University of Hawaii Cancer Center in Honolulu, Hawaii. The meeting was cosponsored by the International Association for the Study of Lung Cancer, and the agenda was designed with significant input from staff at the U.S. National Cancer Institute and National Institute of Environmental Health Sciences. A multidisciplinary group of participants presented updates reflecting a range of disciplinary perspectives, including mineralogy, geology, epidemiology, toxicology, biochemistry, molecular biology, genetics, public health, and clinical oncology. The group identified knowledge gaps that are barriers to preventing and treating malignant mesothelioma (MM) and the required next steps to address barriers. This manuscript reports the group’s efforts and focus on strategies to limit risk to the population and reduce the incidence of MM. Four main topics were explored: genetic risk, environmental exposure, biomarkers, and clinical interventions. Genetics plays a critical role in MM when the disease occurs in carriers of germline BRCA1 associated protein 1 mutations. Moreover, it appears likely that, in addition to BRCA1 associated protein 1, other yet unknown genetic variants may also influence the individual risk for development of MM, especially after exposure to asbestos and related mineral fibers. MM is an almost entirely preventable malignancy as it is most often caused by exposure to commercial asbestos or mineral fibers with asbestos-like health effects, such as erionite. In the past in North America and in Europe, the most prominent source of exposure was related to occupation. Present regulations have reduced occupational exposure in these countries; however, some people continue to be exposed to previously installed asbestos in older construction and other settings. Moreover, an increasing number of people are being exposed in rural areas that contain noncommercial asbestos, erionite, and other mineral fibers in soil or rock (termed naturally occurring asbestos [NOA]) and are being developed. Public health authorities, scientists, residents, and other affected groups must work together in the areas where exposure to asbestos, including NOA, has been documented in the environment to mitigate or reduce this exposure. Although a blood biomarker validated to be effective for use in screening and identifying MM at an early stage in asbestos/ NOA-exposed populations is not currently available, novel biomarkers presented at the meeting, such as high mobility group box 1 and fibulin-3, are promising. There was general agreement that current treatment for MM, which is based on surgery and standard chemotherapy, has a modest effect on the overall survival (OS), which remains dismal. Additionally, although much needed novel therapeutic approaches for MM are being developed and explored in clinical trials, there is a critical need to invest in prevention research, in which there is a great opportunity to reduce the incidence and mortality from MM.
Collapse
Affiliation(s)
- Michele Carbone
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii.
| | - Shreya Kanodia
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii; Samuel Oschin Comprehensive Cancer Institute and Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Ann Chao
- Center for Global Health, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Aubrey Miller
- National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Anil Wali
- Center to Reduce Cancer Health Disparities, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - David Weissman
- Respiratory Health Division, National Institute for Occupational Safety and Health, Centers for Disease Control and Prevention, Morgantown, West Virginia
| | | | | | - Paolo Boffetta
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Brenda Buck
- Department of Geoscience, University of Nevada Las Vegas, Las Vegas, Nevada
| | - Marc de Perrot
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - A Umran Dogan
- Chemical and Biochemical Engineering Department and Center for Global and Regional Environmental Research, University of Iowa, Iowa City, Iowa
| | - Steve Gavett
- Office of Research and Development, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina
| | | | - Raffit Hassan
- Thoracic Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland
| | - Mary Hesdorffer
- Mesothelioma Applied Research Foundation, Alexandria, Virginia
| | - Fred R Hirsch
- University of Colorado Cancer Center, Denver, Colorado
| | - David Larson
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Weimin Mao
- Cancer Research Institute, Zhejiang Cancer Hospital and Key Laboratory of Diagnosis and Treatment Technology on Thoracic Oncology of Zhejiang, Hangzhou, People's Republic of China
| | - Scott Masten
- National Toxicology Program, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina
| | - Harvey I Pass
- Cardiothoracic Surgery, New York University Langone Medical Center, New York, New York
| | - Julian Peto
- Cancer Research UK, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Enrico Pira
- Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Ian Steele
- Notre Dame Integrated Imaging Facility, Notre Dame University, Notre Dame, Indiana
| | - Anne Tsao
- Department of Thoracic and Head and Neck Medical Oncology, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Gavitt Alida Woodard
- Thoracic Surgery, University of California at San Francisco, San Francisco, California
| | - Haining Yang
- Thoracic Oncology, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - Shakun Malik
- Cancer Therapy Evaluation Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
12
|
Bigh3 silencing increases retinoblastoma tumor growth in the murine SV40-TAg-Rb model. Oncotarget 2017; 8:15490-15506. [PMID: 28099942 PMCID: PMC5362501 DOI: 10.18632/oncotarget.14659] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 12/24/2016] [Indexed: 11/25/2022] Open
Abstract
BIGH3, a secreted protein of the extracellular matrix interacts with collagen and integrins on the cell surface. BIGH3 can have opposing functions in cancer, acting either as tumor suppressor or promoter by enhancing tumor progression and angiogenesis. In the eye, BIGH3 is expressed in the cornea and the retinal pigment epithelium and could impact on the development of retinoblastoma, the most common paediatric intraocular neoplasm. Retinoblastoma initiation requires the inactivation of both alleles of the RB1 tumor suppressor gene in the developing retina and tumor progression involves additional genomic changes. To determine whether BIGH3 affects retinoblastoma development, we generated a retinoblastoma mouse model with disruption of the Bigh3 genomic locus. Bigh3 silencing in these mice resulted in enhanced tumor development in the retina. A decrease in apoptosis is involved in the initial events of tumorigenesis, followed by an increased activity of the pro-survival ERK pathway as well as an upregulation of cyclin-dependent kinases (CDKs). Taken together, these data suggest that BIGH3 acts as a tumor suppressor in the retina.
Collapse
|
13
|
Wang J, Zhang G, Wang J, Wang L, Huang X, Cheng Y. The role of cancer-associated fibroblasts in esophageal cancer. J Transl Med 2016; 14:30. [PMID: 26822225 PMCID: PMC4732002 DOI: 10.1186/s12967-016-0788-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 01/17/2016] [Indexed: 01/04/2023] Open
Abstract
Fibroblasts are known as critical stromal cells in wound healing by synthesizing extracellular matrix and collagen. A subpopulation of them is called cancer-associated fibroblasts (CAFs), because their production of proteins participated in various biological activities including tumor cell proliferation, invasion and metastasis. Currently some studies shed light on their role in esophageal cancer which was an aggressive cancer with a dismal survival and high rate of metastasis. Thus, to find cures for it relies on elucidating the epithelial-fibroblasts crosstalk. Herein, we reviewed the present knowledge of the CAFs’ role in esophageal premalignant condition, cancer initiation, progression, metastasis and prognosis prediction and further provided some insights into its clinical application.
Collapse
Affiliation(s)
- Jiangfeng Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Guangyu Zhang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Jianbo Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Lu Wang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Xiaochen Huang
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| | - Yufeng Cheng
- Department of Radiation Oncology, Qilu Hospital of Shandong University, No 107 West Wenhua Road, Jinan, 250012, People's Republic of China.
| |
Collapse
|
14
|
Bai L, Liang R, Yang Y, Hou X, Wang Z, Zhu S, Wang C, Tang Z, Li K. MicroRNA-21 Regulates PI3K/Akt/mTOR Signaling by Targeting TGFβI during Skeletal Muscle Development in Pigs. PLoS One 2015; 10:e0119396. [PMID: 25950587 PMCID: PMC4423774 DOI: 10.1371/journal.pone.0119396] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 01/12/2015] [Indexed: 01/05/2023] Open
Abstract
MicroRNAs (miRNAs), which are short (22–24 base pairs), non-coding RNAs, play critical roles in myogenesis. Using Solexa deep sequencing, we detected the expression levels of 229 and 209 miRNAs in swine skeletal muscle at 90 days post-coitus (E90) and 100 days postnatal (D100), respectively. A total of 138 miRNAs were up-regulated on E90, and 31 were up-regulated on D100. Of these, 9 miRNAs were selected for the validation of the small RNA libraries by quantitative RT-PCR (RT-qPCR). We found that miRNA-21 was down-regulated by 17-fold on D100 (P<0.001). Bioinformatics analysis suggested that the transforming growth factor beta-induced (TGFβI) gene was a potential target of miRNA-21. Both dual luciferase reporter assays and western blotting demonstrated that the TGFβI gene was regulated by miRNA-21. Co-expression analysis revealed that the mRNA expression levels of miRNA-21 and TGFβI were negatively correlated (r = -0.421, P = 0.026) in skeletal muscle during the 28 developmental stages. Our results revealed that more miRNAs are expressed in prenatal than in postnatal skeletal muscle. The miRNA-21 is a novel myogenic miRNA that is involved in skeletal muscle development and regulates PI3K/Akt/mTOR signaling by targeting the TGFβI gene.
Collapse
Affiliation(s)
- Lijing Bai
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Ruyi Liang
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Yalan Yang
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Xinhua Hou
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Zishuai Wang
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shiyun Zhu
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Chuduan Wang
- National Engineering Laboratory for Animal Breeding, Key Laboratory of Agricultural Animal Genetics and Breeding, Department of Animal Breeding and Genetics, College of Animal Sciences and Technology, China Agricultural University, Beijing, China
| | - Zhonglin Tang
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
- Agricultural Genome Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
- * E-mail: ,
| | - Kui Li
- State Key Laboratory for Animal Nutrition, Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture of China, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
15
|
Han B, Cai H, Chen Y, Hu B, Luo H, Wu Y, Wu J. The role of TGFBI (βig-H3) in gastrointestinal tract tumorigenesis. Mol Cancer 2015; 14:64. [PMID: 25889002 PMCID: PMC4435624 DOI: 10.1186/s12943-015-0335-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2014] [Accepted: 03/09/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND TGFβ-induced (TGFBI/βig-H3) is a protein inducible by TGFβ1 and secreted by many types of cells. It binds to collagen, forms part of the extracellular matrix (ECM), and interacts with integrins on cell surfaces. In this study, we investigated the role of TGFBI in tumorigenesis and the underlying mechanisms. METHODS Patient serum TGFBI levels were determined by ELISA. TGFBI transgenic and gene knockout mice and TGFBI-overexpressing liver cells were used for mechanistic studies. RESULTS We demonstrated that patients with cholangiocarcinomas, hepatic carcinomas or gastric carcinomas presented significantly elevated serum TGFBI levels, and the excess TGFBI was derived from the tumor masses. TGFBI overexpression in mice resulted in increased incidence of spontaneous tumors and N,N-diethylnitrosamine (DEN)-induced liver tumor nodules, compared to that in wild type (WT) mice, while TGFBI knockout mice were comparable to WT controls in these 2 aspects. TGFBI promoted the survival of Aml-12 liver cells with DNA damage after irradiation, and augmented their post-irradiation proliferation. It activated the FAK/AKT/AKT1S1/PRS6/EIF4EBP pathway, which is known to modulate cell survival and proliferation. CONCLUSIONS Our data suggest that TGFBI functions as a promoter of certain gastrointestinal tract cancers. It provides a survival advantage to cells with DNA damage. Over a long time span, this advantage could translate into increased tumor risks.
Collapse
Affiliation(s)
- Bing Han
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| | - Haolei Cai
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Ying Chen
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Bing Hu
- Anatomic Pathology, AmeriPath Central Florida, 8150 Chancellor Dr, Orlando, FL, USA.
| | - Hongyu Luo
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| | - Yulian Wu
- Department of Surgery, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Hangzhou, China.
| | - Jiangping Wu
- Laboratory of Immunology and Cardiovascular Research, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
- Nephrology Service, Research Centre, Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Stain-Denis Street, Montreal, Quebec, Canada.
| |
Collapse
|
16
|
Rudra-Ganguly N, Lowe C, Mattie M, Chang MS, Satpayev D, Verlinsky A, An Z, Hu L, Yang P, Challita-Eid P, Stover DR, Pereira DS. Discoidin domain receptor 1 contributes to tumorigenesis through modulation of TGFBI expression. PLoS One 2014; 9:e111515. [PMID: 25369402 PMCID: PMC4219757 DOI: 10.1371/journal.pone.0111515] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Accepted: 09/26/2014] [Indexed: 12/18/2022] Open
Abstract
Discoidin domain receptor 1 (DDR1) is a member of the receptor tyrosine kinase family. The receptor is activated upon binding to its ligand, collagen, and plays a crucial role in many fundamental processes such as cell differentiation, adhesion, migration and invasion. Although DDR1 is expressed in many normal tissues, upregulated expression of DDR1 in a variety of human cancers such as lung, colon and brain cancers is known to be associated with poor prognosis. Using shRNA silencing, we assessed the oncogenic potential of DDR1. DDR1 knockdown impaired tumor cell proliferation and migration in vitro and tumor growth in vivo. Microarray analysis of tumor cells demonstrated upregulation of TGFBI expression upon DDR1 knockdown, which was subsequently confirmed at the protein level. TGFBI is a TGFβ-induced extracellular matrix protein secreted by the tumor cells and is known to act either as a tumor promoter or tumor suppressor, depending on the tumor environment. Here, we show that exogenous addition of recombinant TGFBI to BXPC3 tumor cells inhibited clonogenic growth and migration, thus recapitulating the phenotypic effect observed from DDR1 silencing. BXPC3 tumor xenografts demonstrated reduced growth with DDR1 knockdown, and the same xenograft tumors exhibited an increase in TGFBI expression level. Together, these data suggest that DDR1 expression level influences tumor growth in part via modulation of TGFBI expression. The reciprocal expression of DDR1 and TGFBI may help to elucidate the contribution of DDR1 in tumorigenesis and TGFBI may also be used as a biomarker for the therapeutic development of DDR1 specific inhibitors.
Collapse
Affiliation(s)
- Nandini Rudra-Ganguly
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
- * E-mail:
| | - Christine Lowe
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Michael Mattie
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Mi Sook Chang
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | | | - Alla Verlinsky
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Zili An
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Liping Hu
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Peng Yang
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Pia Challita-Eid
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - David R. Stover
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| | - Daniel S. Pereira
- Agensys Inc., an affiliate of Astellas Pharma Inc, Santa Monica, CA, United States of America
| |
Collapse
|
17
|
Zhao T, Sun Q, del Rincon SV, Lovato A, Marques M, Witcher M. Gallotannin imposes S phase arrest in breast cancer cells and suppresses the growth of triple-negative tumors in vivo. PLoS One 2014; 9:e92853. [PMID: 24658335 PMCID: PMC3962455 DOI: 10.1371/journal.pone.0092853] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2013] [Accepted: 02/27/2014] [Indexed: 12/18/2022] Open
Abstract
Triple-negative breast cancers are associated with poor clinical outcomes and new therapeutic strategies are clearly needed. Gallotannin (Gltn) has been previously demonstrated to have potent anti-tumor properties against cholangiocarcinoma in mice, but little is known regarding its capacity to suppress tumor outgrowth in breast cancer models. We tested Gltn for potential growth inhibitory properties against a variety of breast cancer cell lines in vitro. In particular, triple-negative breast cancer cells display higher levels of sensitivity to Gltn. The loss of proliferative capacity in Gltn exposed cells is associated with slowed cell cycle progression and S phase arrest, dependent on Chk2 phosphorylation and further characterized by changes to proliferation related genes, such as cyclin D1 (CcnD1) as determined by Nanostring technology. Importantly, Gltn administered orally or via intraperitoneal (IP) injections greatly reduced tumor outgrowth of triple-negative breast cells from mammary fat pads without signs of toxicity. In conclusion, these data strongly suggest that Gltn represents a novel approach to treat triple-negative breast carcinomas.
Collapse
Affiliation(s)
- Tiejun Zhao
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Qiang Sun
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Sonia V. del Rincon
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Amanda Lovato
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Maud Marques
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Witcher
- The Lady Davis Institute and Segal Cancer Center of the Jewish General Hospital, McGill University, Montreal, Quebec, Canada
- * E-mail:
| |
Collapse
|
18
|
Choi SI, Kim BY, Dadakhujaev S, Oh JY, Kim TI, Kim JY, Kim EK. Impaired autophagy and delayed autophagic clearance of transforming growth factor β-induced protein (TGFBI) in granular corneal dystrophy type 2. Autophagy 2012; 8:1782-97. [PMID: 22995918 DOI: 10.4161/auto.22067] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Granular corneal dystrophy type 2 (GCD2) is an autosomal dominant disease characterized by a progressive age-dependent extracellular accumulation of transforming growth factor β-induced protein (TGFBI). Corneal fibroblasts from GCD2 patients also have progressive degenerative features, but the mechanism underlying this degeneration remains unknown. Here we observed that TGFBI was degraded by autophagy, but not by the ubiquitin/proteasome-dependent pathway. We also found that GCD2 homozygous corneal fibroblasts displayed a greater number of fragmented mitochondria. Most notably, mutant TGFBI (mut-TGFBI) extensively colocalized with microtubule-associated protein 1 light chain 3β (MAP1LC3B, hereafter referred to as LC3)-enriched cytosolic vesicles and CTSD in primary cultured GCD2 corneal fibroblasts. Levels of LC3-II, a marker of autophagy activation, were significantly increased in GCD2 corneal fibroblasts. Nevertheless, levels of SQSTM1/p62 and of polyubiquitinated protein were also significantly increased in GCD2 corneal fibroblasts compared with wild-type (WT) cells. However, LC3-II levels did not differ significantly between WT and GCD2 cells, as assessed by the presence of bafilomycin A 1, the fusion blocker of autophagosomes and lysosomes. Likewise, bafilomycin A 1 caused a similar change in levels of SQSTM1. Thus, the increase in autophagosomes containing mut-TGFBI may be due to inefficient fusion between autophagosomes and lysosomes. Rapamycin, an autophagy activator, decreased mut-TGFBI, whereas inhibition of autophagy increased active caspase-3, poly (ADP-ribose) polymerase 1 (PARP1) and reduced the viability of GCD2 corneal fibroblasts compared with WT controls. These data suggest that defective autophagy may play a critical role in the pathogenesis of GCD2.
Collapse
Affiliation(s)
- Seung-Il Choi
- Corneal Dystrophy Research Institute and Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
19
|
Transforming growth Factor-Beta-Induced Protein (TGFBI)/(βig-H3): a matrix protein with dual functions in ovarian cancer. Int J Mol Sci 2012; 13:10461-10477. [PMID: 22949874 PMCID: PMC3431872 DOI: 10.3390/ijms130810461] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/03/2012] [Accepted: 08/16/2012] [Indexed: 02/04/2023] Open
Abstract
Transforming growth factor-beta-induced protein (TGFBI, also known as βig-H3 and keratoepithelin) is an extracellular matrix protein that plays a role in a wide range of physiological and pathological conditions including diabetes, corneal dystrophy and tumorigenesis. Many reports indicate that βig-H3 functions as a tumor suppressor. Loss of βig-H3 expression has been described in several cancers including ovarian cancer and promoter hypermethylation has been identified as an important mechanism for the silencing of the TGFBI gene. Our recent findings that βig-H3 is down-regulated in ovarian cancer and that high concentrations of βig-H3 can induce ovarian cancer cell death support a tumor suppressor role. However, there is also convincing data in the literature reporting a tumor-promoting role for βig-H3. We have shown βig-H3 to be abundantly expressed by peritoneal cells and increase the metastatic potential of ovarian cancer cells by promoting cell motility, invasion, and adhesion to peritoneal cells. Our findings suggest that βig-H3 has dual functions and can act both as a tumor suppressor or tumor promoter depending on the tumor microenvironment. This article reviews the current understanding of βig-H3 function in cancer cells with particular focus on ovarian cancer.
Collapse
|
20
|
Li B, Wen G, Zhao Y, Tong J, Hei TK. The role of TGFBI in mesothelioma and breast cancer: association with tumor suppression. BMC Cancer 2012; 12:239. [PMID: 22695319 PMCID: PMC3480943 DOI: 10.1186/1471-2407-12-239] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Accepted: 05/21/2012] [Indexed: 11/17/2022] Open
Abstract
Background Transforming growth factor β induced (TGFBI) product, an extracellular matrix (ECM) protein, has been implicated as a putative tumor suppressor in recent studies. Our previous findings revealed that expression of TGFBI gene is down-regulated in a variety of cancer cell lines and clinical tissue samples. In this study, ectopic expression of TGFBI was used to ascertain its role as a tumor suppressor and to determine the underlying mechanism of mesothelioma and breast cancer. Methods Cells were stably transfected with pRc/CMV2-TGFBI and pRc/CMV2-empty vector with Lipofectamine Plus. Ectopic expression of TGFBI was quantified by using quantitative PCR and Western-blotting. Characterization of cell viability was assessed using growth curve, clonogenic survival and soft agar growth. The potential of tumor formation was evaluated by an in vivo mouse model. Cell cycle was analyzed via flow cytometry. Expressions of p21, p53, p16 and p14 were examined using Western-blotting. Senescent cells were sorted by using a Senescence β-Galactosidase Staining Kit. Telomerase activity was measured using quantitative telomerase detection kit. Results In this study, an ectopic expression of TGFBI in two types of cancer cell lines, a mesothelioma cell line NCI-H28 and a breast cancer cell line MDA-MB-231 was found to have reduced the cellular growth, plating efficiency, and anchorage-independent growth. The tumorigenicity of these cancer cell lines as determined by subcutaneous inoculation in nude mice was similarly suppressed by TGFBI expression. Likewise, TGFBI expression reduced the proportion of S-phase while increased the proportion of G1 phase in these cells. The redistribution of cell cycle phase after re-expression of TGFBI was correspondent with transiently elevated expression of p21 and p53. The activities of senescence-associated β-galactosidase and telomerase were enhanced in TGFBI-transfected cells. Conclusion Collectively, these results imply that TGFBI plays a suppressive role in the development of mesothelioma and breast cancer cells, possibly through inhibitions of cell proliferation, delaying of G1-S phase transition, and induction of senescence.
Collapse
Affiliation(s)
- Bingyan Li
- School of Radiation Medicine and Public Health, Soochow University, Suzhou, China
| | | | | | | | | |
Collapse
|