151
|
Hypoxia-induced tumor exosomes promote M2-like macrophage polarization of infiltrating myeloid cells and microRNA-mediated metabolic shift. Oncogene 2019; 38:5158-5173. [PMID: 30872795 DOI: 10.1038/s41388-019-0782-x] [Citation(s) in RCA: 204] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Revised: 02/08/2019] [Accepted: 03/04/2019] [Indexed: 12/12/2022]
Abstract
Developing tumors rapidly outgrow their oxygen supply and are subject to hypoxia, which stimulates hypersecretion of tumor-derived exosomes that promote angiogenesis, metastasis, and immunosuppression, but the molecular mediators of these pathological effects remain poorly defined. Using quantitative proteomics, we identified that exosomes produced by hypoxic tumor cells are highly enriched in immunomodulatory proteins and chemokines including CSF-1, CCL2, FTH, FTL, and TGFβ. Modeling exosome effects on tumor-infiltrating immune cells, we observed a potent ability of these hypoxia-induced vesicles to influence macrophage recruitment and promote M2-like polarization both in vitro and in vivo. In addition, hypoxic, but not normoxic, tumor exosomes enhanced oxidative phosphorylation in bone marrow-derived macrophages via transfer of let-7a miRNA, resulting in suppression of the insulin-Akt-mTOR signaling pathway. Together, these data demonstrate that hypoxia promotes tumor secretion of biomolecule-loaded exosomes that can modify the immunometabolic profile of infiltrating monocyte-macrophages to better evade host immunity and enhance tumor progression.
Collapse
|
152
|
Immunoexpression of Transforming Growth Factor Beta 3 (TGFβ3) and Its Receptor Type III (TGFβRIII) in Basal Cell Carcinomas. CURRENT HEALTH SCIENCES JOURNAL 2019; 44:166-171. [PMID: 30746165 PMCID: PMC6320465 DOI: 10.12865/chsj.44.02.12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/27/2018] [Indexed: 11/18/2022]
Abstract
ABSTRACT: Basal cell carcinomas (BCCs) are malignant tumors with particular biological prognosis and behavior, and the biomolecular investigation of these lesions can provide important therapeutic targets for epithelial neoplasia. In this study we analyzed the immunoexpression of transforming growth factor beta 3 (TGFβ3) and its receptor type III (TGFβRIII) for 53 cases of BCC in relation to the main histopathological prognostic parameters. The results indicated statistically significant differences of TGFβ3 and TGFβRIII expression related to histological type and lesion stage, the both proteins being higher expressed in adenoid and morpheaform advanced stage tumors. In this study, TGFβ3 and TGFβRIII immunoexpression analysis indicated their utility for identifying aggressive BCCs with potential for tumor progression
Collapse
|
153
|
Ihling C, Naughton B, Zhang Y, Rolfe PA, Frick-Krieger E, Terracciano LM, Dussault I. Observational Study of PD-L1, TGF-β, and Immune Cell Infiltrates in Hepatocellular Carcinoma. Front Med (Lausanne) 2019; 6:15. [PMID: 30800658 PMCID: PMC6375852 DOI: 10.3389/fmed.2019.00015] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 01/17/2019] [Indexed: 12/14/2022] Open
Abstract
Introduction: Hepatocellular carcinoma (HCC) typically develops in cirrhotic livers, with increased programed death ligand 1 (PD-L1) and transforming growth factor beta (TGF-β) activity implicated in immunosuppression. Methods: In an observational study of HCC liver samples, we determined the incidence of PD-L1 and immune cell (IC) infiltrates, and signs of TGF-β activity. HCCs were characterized by the incidence and distribution of PD-L1+ cells, and CD8+, CD68+, and FoxP3+ infiltrating ICs in HCC and surrounding liver. Gene expression signatures (GESs) associated with TGF-β activity and ICs were evaluated by RNAseq. Results: In non-neoplastic cirrhotic and non-cirrhotic liver, PD-L1 occurred on sinusoidal lining cells (mostly Kupffer cells), endothelial cells and ICs. In HCC, PD-L1+ tumor cells were rare. Most PD-L1+ cells were identified as ICs. CD8+, CD68+, and FoxP3+ ICs were associated with HCC, particularly in the invasive margin. CD8+ cell incidence correlated with PD-L1+ cells, consistent with PD-L1 being upregulated in response to pre-existing cytotoxic T-lymphocyte activity. TGFB1 mRNA levels and TGF-β activation GES correlated with the strength of the tumor-associated macrophage GES. Conclusion: Inhibition of PD-L1+ ICs and TGF-β activity and their respective immunomodulatory pathways may contribute to antitumor effects in HCC.
Collapse
Affiliation(s)
- Christian Ihling
- Global Clinical Biomarkers & Companion Diagnostics, Merck KGaA, Darmstadt, Germany
| | - Bartholomew Naughton
- Quantitative Pharmacology and Drug Disposition, EMD Serono, Inc., Billerica, MA, United States
| | - Yue Zhang
- Clinical Biomarkers & Companion Diagnostics Biopharma, EMD Serono, Inc., Billerica, MA, United States
| | - P Alexander Rolfe
- Quantitative Pharmacology and Drug Disposition, EMD Serono, Inc., Billerica, MA, United States
| | | | - Luigi M Terracciano
- Molecular Pathology Division, Institute of Pathology, University Hospital, Basel, Switzerland
| | - Isabelle Dussault
- Clinical Biomarkers & Companion Diagnostics Biopharma, EMD Serono, Inc., Billerica, MA, United States
| |
Collapse
|
154
|
Joshi S, Durden DL. Combinatorial Approach to Improve Cancer Immunotherapy: Rational Drug Design Strategy to Simultaneously Hit Multiple Targets to Kill Tumor Cells and to Activate the Immune System. JOURNAL OF ONCOLOGY 2019; 2019:5245034. [PMID: 30853982 PMCID: PMC6377965 DOI: 10.1155/2019/5245034] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/15/2018] [Accepted: 01/01/2019] [Indexed: 02/07/2023]
Abstract
Cancer immunotherapy, including immune checkpoint blockade and adoptive CAR T-cell therapy, has clearly established itself as an important modality to treat melanoma and other malignancies. Despite the tremendous clinical success of immunotherapy over other cancer treatments, this approach has shown substantial benefit to only some of the patients while the rest of the patients have not responded due to immune evasion. In recent years, a combination of cancer immunotherapy together with existing anticancer treatments has gained significant attention and has been extensively investigated in preclinical or clinical studies. In this review, we discuss the therapeutic potential of novel regimens combining immune checkpoint inhibitors with therapeutic interventions that (1) increase tumor immunogenicity such as chemotherapy, radiotherapy, and epigenetic therapy; (2) reverse tumor immunosuppression such as TAMs, MDSCs, and Tregs targeted therapy; and (3) reduce tumor burden and increase the immune effector response with rationally designed dual or triple inhibitory chemotypes.
Collapse
Affiliation(s)
- Shweta Joshi
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Moores Cancer Center, University of California, San Diego, CA, USA
| | - Donald L. Durden
- Division of Pediatric Hematology-Oncology, Department of Pediatrics, Moores Cancer Center, University of California, San Diego, CA, USA
- SignalRx Pharmaceuticals, Inc., San Diego, CA, USA
| |
Collapse
|
155
|
Lee G, Han SB, Lee JH, Kim HW, Kim DH. Cancer Mechanobiology: Microenvironmental Sensing and Metastasis. ACS Biomater Sci Eng 2019; 5:3735-3752. [PMID: 33405888 DOI: 10.1021/acsbiomaterials.8b01230] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cellular microenvironment plays an important role in regulating cancer progress. Cancer can physically and chemically remodel its surrounding extracellular matrix (ECM). Critical cellular behaviors such as recognition of matrix geometry and rigidity, cell polarization and motility, cytoskeletal reorganization, and proliferation can be changed as a consequence of these ECM alternations. Here, we present an overview of cancer mechanobiology in detail, focusing on cancer microenvironmental sensing of exogenous cues and quantification of cancer-substrate interactions. In addition, mechanics of metastasis classified with tumor progression will be discussed. The mechanism underlying cancer mechanosensation and tumor progression may provide new insights into therapeutic strategies to alleviate cancer malignancy.
Collapse
Affiliation(s)
- GeonHui Lee
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, South Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, South Korea.,Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, South Korea.,Department of Nanobiomedical Science and BK21 PLUS Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, South Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, South Korea
| |
Collapse
|
156
|
Chemotherapy and Inflammatory Cytokine Signalling in Cancer Cells and the Tumour Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:173-215. [PMID: 31456184 DOI: 10.1007/978-3-030-20301-6_9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer is the result of a cell's acquisition of a variety of biological capabilities or 'hallmarks' as outlined by Hanahan and Weinberg. These include sustained proliferative signalling, the ability to evade growth suppressors, resisting cell death, enabling replicative immortality, inducing angiogenesis, and the ability to invade other tissue and metastasize. More recently, the ability to escape immune destruction has been recognized as another important hallmark of tumours. It is suggested that genome instability and inflammation accelerates the acquisition of a variety of the above hallmarks. Inflammation, is a product of the body's response to tissue damage or pathogen invasion. It is required for tissue repair and host defense, but prolonged inflammation can often be the cause for disease. In a cancer patient, it is often unclear whether inflammation plays a protective or deleterious role in disease progression. Chemotherapy drugs can suppress tumour growth but also induce pathways in tumour cells that have been shown experimentally to support tumour progression or, in other cases, encourage an anti-tumour immune response. Thus, with the goal of better understanding the context under which each of these possible outcomes occurs, recent progress exploring chemotherapy-induced inflammatory cytokine production and the effects of cytokines on drug efficacy in the tumour microenvironment will be reviewed. The implications of chemotherapy on host and tumour cytokine pathways and their effect on the treatment of cancer patients will also be discussed.
Collapse
|
157
|
Corrigendum to “The Dual Role of TGF β in Human Cancer: From Tumor Suppression to Cancer Metastasis”. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2018. [DOI: 10.1155/2018/1497218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
158
|
TGF-β downregulation-induced cancer cell death is finely regulated by the SAPK signaling cascade. Exp Mol Med 2018; 50:1-19. [PMID: 30523245 PMCID: PMC6283885 DOI: 10.1038/s12276-018-0189-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
Transforming growth factor (TGF)-β signaling is increasingly recognized as a key driver in cancer. In progressive cancer tissues, TGF-β promotes tumor formation, and its increased expression often correlates with cancer malignancy. In this study, we utilized adenoviruses expressing short hairpin RNAs against TGF-β1 and TGF-β2 to investigate the role of TGF-β downregulation in cancer cell death. We found that the downregulation of TGF-β increased the phosphorylation of several SAPKs, such as p38 and JNK. Moreover, reactive oxygen species (ROS) production was also increased by TGF-β downregulation, which triggered Akt inactivation and NOX4 increase-derived ROS in a cancer cell-type-specific manner. We also revealed the possibility of substantial gene fluctuation in response to TGF-β downregulation related to SAPKs. The expression levels of Trx and GSTM1, which encode inhibitory proteins that bind to ASK1, were reduced, likely a result of the altered translocation of Smad complex proteins rather than from ROS production. Instead, both ROS and ROS-mediated ER stress were responsible for the decrease in interactions between ASK1 and Trx or GSTM1. Through these pathways, ASK1 was activated and induced cytotoxic tumor cell death via p38/JNK activation and (or) induction of ER stress. Reducing the levels of the multifunctional protein transforming growth factor (TGF)-β in cancer cells prevents tumor growth in mice. Previous studies have shown that high levels of TGF-β in cancerous tissue are associated with accelerated disease progression. Hye Jin Choi and Jae J Song at Yonsei University in Seoul, South Korea, and colleagues infected cancer cells with genetically modified viruses that reduced the expression of the gene encoding TGF-β. The resulting decrease in TGF-β protein led to cell death by stimulating the production of reactive oxygen species and signaling through the apoptosis signal-regulating kinase 1 (ASK1) pathway. When tumor-bearing mice were infected with these modified viruses, their overall survival was improved. Further understanding the mechanisms through which TGF-β regulates cancer cell survival will contribute to the development of new approaches in cancer treatment.
Collapse
|
159
|
Dasatinib sensitises triple negative breast cancer cells to chemotherapy by targeting breast cancer stem cells. Br J Cancer 2018; 119:1495-1507. [PMID: 30482914 PMCID: PMC6288167 DOI: 10.1038/s41416-018-0287-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/21/2018] [Accepted: 09/12/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Patients with triple negative breast cancer (TNBC) exhibit poor prognosis and are at high risk of tumour relapse, due to the resistance to chemotherapy. These aggressive phenotypes are in part attributed to the presence of breast cancer stem cells (BCSCs). Therefore, targeting BCSCs is a priority to overcoming chemotherapy failure in TNBCs. METHODS We generated paclitaxel (pac)-resistant TNBC cells which displayed higher sphere forming potential and percentage of BCSC subpopulations compared to the parental cells. A screen with various kinase inhibitors revealed dasatinib, a Src kinase family inhibitor, as a potent suppressor of BCSC expansion/sphere formation in pac-resistant TNBC cells. RESULTS We found dasatinib to block pac-induced BCSC enrichment and Src activation in both parental and pac-resistant TNBC cells. Interestingly, dasatinib induced an epithelial differentiation of the pac-resistant mesenchymal cells, resulting in their enhanced sensitivity to paclitaxel. The combination treatment of dasatinib and paclitaxel not only decreased the BCSCs numbers and their sphere forming capacity but also synergistically reduced cell viability of pac-resistant cells. Preclinical models of breast cancer further demonstrated the efficiency of the dasatinib/paclitaxel combination treatment in inhibiting tumour growth. CONCLUSIONS Dasatinib is a promising anti-BCSC drug that could be used in combination with paclitaxel to overcome chemoresistance in TNBC.
Collapse
|
160
|
Liu Y, Shao L, Chen K, Wang Z, Wang J, Jing W, Hu M. GDF11 restrains tumor growth by promoting apoptosis in pancreatic cancer. Onco Targets Ther 2018; 11:8371-8379. [PMID: 30568460 PMCID: PMC6267626 DOI: 10.2147/ott.s181792] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Growth differentiation factor (GDF) acted as a factor that regulated proliferation, apoptosis and differentiation in several tumors. However, the effects of growth differentiation factor (GDF11) in pancreatic cancer remain unclear. Purpose To investigate the expression and significance of GDF11 in pancreatic cancer. Patients and methods Pancreatic cancer and corresponding paracancerous tissues (n=28) were collected from the Department of Hepatobiliary and Pancreatic Surgical Oncology of Chinese PLA General Hospital. Tissue microarray was obtained from Outdo Biotech Co., Ltd. (Shanghai, People’s Republic of China). GDF11 mRNA and protein expressions in pancreatic cancer samples and cell lines were detected using qRT-PCR, Western-Blot and immunohistochemistry. Overexpression and knockdown of GDF11 were performed with lentiviral transduction system and siRNA technique in PANC-1 cell line and CFPAC-1 cell line. Proliferation, migration and invasion of pancreatic cancer cell lines were examinated by MTS and transwell assay, respectively. Flow cytometry was used for cell apoptosis analysis. Results The results of this study indicated that GDF11 was significantly down-regulated in pancreatic cancer tissues compared with adjacent tissues of pancreatic cancer. GDF11 was also associated with low expression in pancreatic cancer cell lines when compared with normal pancreatic cell line. In a cohort of 63 pancreatic cancer patients, high GDF11 expression levels was associated with favorable perineural invasion, T classification, N classification and overall survival (OS). Cox proportional hazards model revealed that high GDF11 expression was an independent predictor of favorable prognosis (HR: 0.496; 95% CI: 0.255–0.967; P=0.040). Overexpression of GDF11 in PANC-1 cells repressed the proliferation, migration and invasion abilities in vitro. Inhibition of GDF11 in CFPAC-1 showed inverse results. Furthermore, enhanced GDF11 expression promoted apoptosis and down-regulated GDF11 expression inhibited apoptosis in pancreatic cancer cell lines. Conclusion These findings suggested that GDF11 acted as a tumor suppressor gene for pancreatic cancer.
Collapse
Affiliation(s)
- Yanzhe Liu
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China,
| | - Lijuan Shao
- Translational Medicine Collaborative Innovation Center, The Second Clinical Medical College (Shenzhen People's Hospital), Jinan University, Shenzhen, People's Republic of China
| | - Kuang Chen
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China,
| | - Zizheng Wang
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China,
| | - Jin Wang
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China,
| | - Wei Jing
- Department of General Surgery, Changhai Hospital, The Second Military Medical University, Shanghai, People's Republic of China,
| | - Minggen Hu
- Department of Hepatobiliary and Pancreatic Surgical Oncology, Chinese PLA General Hospital, Beijing, People's Republic of China,
| |
Collapse
|
161
|
Elliott B, Zackery DL, Eaton VA, Jones RT, Abebe F, Ragin CC, Khan SA. Ethnic differences in TGFβ-signaling pathway may contribute to prostate cancer health disparity. Carcinogenesis 2018; 39:546-555. [PMID: 29474521 DOI: 10.1093/carcin/bgy020] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 02/02/2018] [Indexed: 11/13/2022] Open
Abstract
Epidemiological studies show that the incidence and mortality rates of prostate cancer (PCa) are significantly higher in African-American (AA) men when compared with Caucasian (CA) men in the United States. Transforming growth factor β (TGFβ) signaling pathway is linked to health disparities in AAs. Recent studies suggest a role of TGFβ3 in cancer metastases and its effect on the migratory and invasive behavior; however, its role in PCa in AA men has not been studied. We determined the circulating levels of TGFβ3 in AA and CA men diagnosed with PCa using ELISA. We analyzed serum samples from both AA and CA men diagnosed with and without PCa. We show that AA PCa patients had higher levels of TGFβ3 protein compared with AA controls and CA patients. In fact, TGFβ3 protein levels in serum were higher in AA men without PCa compared with the CA population, which may correlate with more aggressive disease seen in AA men. Studies on AA-derived PCa cell lines revealed that TGFβ3 protein levels were also higher in these cells compared with CA-derived PCa cell lines. Our studies also reveal that TGFβ does not inhibit cell proliferation in AA-derived PCa cell lines, but it does induce migration and invasion through activation of PI3K pathway. We suggest that increased TGFβ3 levels are responsible for development of aggressive PCa in AA patients as a consequence of development of resistance to inhibitory effects of TGFβ on cell proliferation and induction of invasive metastatic behavior.
Collapse
Affiliation(s)
- Bethtrice Elliott
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA
| | - DeAdra L Zackery
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA
| | - Vanessa A Eaton
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA
| | - Re'Josef T Jones
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA
| | - Fisseha Abebe
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA.,Department of Mathematical Sciences, Clark Atlanta University, Atlanta, GA, USA
| | - Camille C Ragin
- Fox Chase Cancer Center-Temple Health, Philadelphia, PA, USA
| | - Shafiq A Khan
- Center for Cancer Research and Therapeutic Development, Atlanta, GA, USA
| |
Collapse
|
162
|
Reese JM, Bruinsma ES, Nelson AW, Chernukhin I, Carroll JS, Li Y, Subramaniam M, Suman VJ, Negron V, Monroe DG, Ingle JN, Goetz MP, Hawse JR. ERβ-mediated induction of cystatins results in suppression of TGFβ signaling and inhibition of triple-negative breast cancer metastasis. Proc Natl Acad Sci U S A 2018; 115:E9580-E9589. [PMID: 30257941 PMCID: PMC6187171 DOI: 10.1073/pnas.1807751115] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for a disproportionately high number of deaths due to a lack of targeted therapies and an increased likelihood of distant recurrence. Estrogen receptor beta (ERβ), a well-characterized tumor suppressor, is expressed in 30% of TNBCs, and its expression is associated with improved patient outcomes. We demonstrate that therapeutic activation of ERβ elicits potent anticancer effects in TNBC through the induction of a family of secreted proteins known as the cystatins, which function to inhibit canonical TGFβ signaling and suppress metastatic phenotypes both in vitro and in vivo. These data reveal the involvement of cystatins in suppressing breast cancer progression and highlight the value of ERβ-targeted therapies for the treatment of TNBC patients.
Collapse
Affiliation(s)
- Jordan M Reese
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Elizabeth S Bruinsma
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905
| | - Adam W Nelson
- Cancer Research UK Cambridge Institute, University of Cambridge, CB2 1TN Cambridge, United Kingdom
| | - Igor Chernukhin
- Cancer Research UK Cambridge Institute, University of Cambridge, CB2 1TN Cambridge, United Kingdom
| | - Jason S Carroll
- Cancer Research UK Cambridge Institute, University of Cambridge, CB2 1TN Cambridge, United Kingdom
| | - Ying Li
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905
| | | | - Vera J Suman
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905
| | - Vivian Negron
- Department of Pathology, Mayo Clinic, Rochester, MN 55905
| | - David G Monroe
- Robert and Arlene Kogod Center on Aging and Endocrine Research Unit, Mayo Clinic, Rochester, MN 55905
| | - James N Ingle
- Department of Oncology, Mayo Clinic, Rochester, MN 55905
| | - Matthew P Goetz
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905
- Department of Oncology, Mayo Clinic, Rochester, MN 55905
| | - John R Hawse
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905;
| |
Collapse
|
163
|
Zheng X, Liu Q, Yi M, Qin S, Wu K. The regulation of cytokine signaling by retinal determination gene network pathway in cancer. Onco Targets Ther 2018; 11:6479-6487. [PMID: 30323623 PMCID: PMC6177397 DOI: 10.2147/ott.s176113] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Tumor environment plays a pivotal role in determining cancer biology characteristics. Cytokine factors, as a critical component in tumor milieu, execute distinct functions in the process of tumorigenesis and progression via the autocrine or paracrine manner. The retinal determination gene network (RDGN), which mainly comprised DACH, SIX, and EYA family members, is required for the organ development in mammalian species. While the aberrant expression of RDGN is involved in the proliferation, apoptosis, angiogenesis, and metastasis of tumors via interacting with different cytokine-related signals, such as CXCL8, IL-6, TGF-β, FGF, and VEGF, in a cell- or tissue-dependent manner. Thus, joint detection of this pathway might be used as a potential biomarker for the stratification of target therapy and for the precision prediction of the prognosis of cancer patients.
Collapse
Affiliation(s)
- Xinhua Zheng
- Department of Clinical Medicine, Medical School of Pingdingshan University, Pingdingshan, Henan 467000, China.,Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,
| | - Qian Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China,
| |
Collapse
|
164
|
Companioni O, Bonet C, García N, Ramírez-Lázaro MJ, Lario S, Mendoza J, Adrados MM, Poves E, Espinosa L, Pozo-Kreilinger JJ, Ortega L, Bujanda L, Cosme A, Ferrández A, Muñoz G, Cuatrecasas M, Elizalde I, Andreu V, Paules MJ, Madrigal B, Barrio J, Berdasco M, Calvet X, Sanz-Anquela JM, Gisbert JP, González CA, Sala N. Genetic variation analysis in a follow-up study of gastric cancer precursor lesions confirms the association of MUC2
variants with the evolution of the lesions and identifies a significant association with NFKB1
and CD14. Int J Cancer 2018; 143:2777-2786. [PMID: 30171605 DOI: 10.1002/ijc.31839] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/03/2018] [Accepted: 07/11/2018] [Indexed: 01/05/2023]
Affiliation(s)
- Osmel Companioni
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
| | - Catalina Bonet
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
| | - Nadia García
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
- Translational Research Laboratory; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
| | - María José Ramírez-Lázaro
- Departament of Medicine, Digestive Diseases Service; Institut Universitari Parc Taulí, Sabadell, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Spain
| | - Sergio Lario
- Departament of Medicine, Digestive Diseases Service; Institut Universitari Parc Taulí, Sabadell, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Spain
| | - Jorge Mendoza
- Department of Gastroenterology; Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and CIBEREHD; Madrid Spain
| | - Mª Magdalena Adrados
- Department of Pathology; Hospital Universitario de la Princesa, IIS-IP; Madrid, Spain
| | - Elvira Poves
- Department of Gastroenterology; Hospital Universitario Príncipe de Asturias; Alcalá de Henares Spain
| | - Laura Espinosa
- Department of Gastroenterology; Hospital Universitario Príncipe de Asturias; Alcalá de Henares Spain
| | | | - Luís Ortega
- Department of Pathology; Hospital Clínico San Carlos; Madrid Spain
| | - Luis Bujanda
- Department of Pathology and Hospital Donostia/Instituto Biodonostia; Universidad del País Vasco (UPV/EHU), and CIBEREHD; San Sebastián Spain
| | - Angel Cosme
- Department of Gastroenterology; Hospital Donostia/Instituto Biodonostia, Universidad del País Vasco (UPV/EHU), and CIBEREHD; San Sebastián Spain
| | - Angel Ferrández
- Department of Gastroenterology and Hospital Clínico Universitario Lozano Blesa Zaragoza, and CIBEREHD; Spain
| | - Guillermo Muñoz
- Department of Pathology; Hospital Clínico Universitario Lozano Blesa, Zaragoza, and CIBEREHD; Spain
| | - Miriam Cuatrecasas
- Department of Pathology; Hospital Clínic de Barcelona, IDIBAPS and CIBEREHD, and Universitat de Barcelona; Spain
| | - Ignasi Elizalde
- Department of Gastroenterology; Hospital Clínic de Barcelona, IDIBAPS and CIBEREHD; Spain
| | - Victoria Andreu
- Department of Gastroenterology; Hospital de Viladecans; Spain
| | - Mª José Paules
- Department of Pathology; Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat; Spain
| | - Beatriz Madrigal
- Department of Pathology; Hospital Universitario Río Hortega; Valladolid Spain
| | - Jesús Barrio
- Department of Gastroenterology; Hospital Universitario Río Hortega; Valladolid Spain
| | - María Berdasco
- Cancer Epigenetics and Biology Program, IDIBELL; Barcelona Spain
| | - Xavier Calvet
- Departament of Medicine, Digestive Diseases Service; Institut Universitari Parc Taulí, Sabadell, and Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD); Spain
| | - José Miguel Sanz-Anquela
- Department of Pathology; Hospital “Principe de Asturias” and University of Alcalá; Alcalá de Henares Spain
| | - Javier P. Gisbert
- Department of Gastroenterology; Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP) and CIBEREHD; Madrid Spain
| | - Carlos A. González
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
| | - Núria Sala
- Unit of Nutrition and Cancer, Cancer Epidemiology Research Program; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
- Translational Research Laboratory; Catalan Institute of Oncology (ICO)-IDIBELL; Barcelona Spain
| | | |
Collapse
|
165
|
van Dijk N, Funt SA, Blank CU, Powles T, Rosenberg JE, van der Heijden MS. The Cancer Immunogram as a Framework for Personalized Immunotherapy in Urothelial Cancer. Eur Urol 2018; 75:435-444. [PMID: 30274701 DOI: 10.1016/j.eururo.2018.09.022] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 09/12/2018] [Indexed: 01/05/2023]
Abstract
CONTEXT The abysmal outlook of urothelial cancer (UC) has changed with the introduction of immunotherapy. Still, many patients do not respond and distinctive biomarkers are currently lacking. The rise of this novel armamentarium of immunotherapy treatments, in combination with the complex biology of an immunological tumor response, warrants the development of a comprehensive framework that can provide an overview of important immunological processes at play in individual patients. OBJECTIVE To develop a comprehensive framework based on tumor- and host-specific parameters to understand immunotherapy response in UC. This framework can inform rational, biology-driven clinical trials and ultimately guide us toward individualized patient treatment. EVIDENCE ACQUISITION A literature review was conducted on UC immunotherapy, clinical trial data, and biomarkers of response to checkpoint inhibition. EVIDENCE SYNTHESIS Here, we propose a UC immunogram, based on currently available clinical and translational data. The UC immunogram describes several tumor- and host-specific parameters that are required for successful immunotherapy treatment. These seven parameters are tumor foreignness, immune cell infiltration, absence of inhibitory checkpoints, general performance and immune status, absence of soluble inhibitors, absence of inhibitory tumor metabolism, and tumor sensitivity to immune effectors. CONCLUSIONS Longitudinal integration of individual patient parameters may ultimately lead to personalized and dynamic immunotherapy, to adjust to the Darwinian forces that drive tumor evolution. Incorporating multiparameter biomarkers into quantitative predictive models will be a key challenge to integrate the immunogram into daily clinical practice. PATIENT SUMMARY Here, we propose the urothelial cancer immunogram, a novel way of describing important immunological characteristics of urothelial cancer patients and their tumors. Seven characteristics determine the chance of having an immunological tumor response. Using this immunogram, we aim to better understand why some patients respond to immunotherapy and some do not, to ultimately improve anticancer therapy.
Collapse
Affiliation(s)
- Nick van Dijk
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Samuel A Funt
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christian U Blank
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Thomas Powles
- Department of Medical Oncology, Barts Cancer Institute, London, UK
| | - Jonathan E Rosenberg
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | |
Collapse
|
166
|
Trugilo KP, Cebinelli GCM, Berti FCB, Okuyama NCM, Cezar-Dos-Santos F, Sena MM, Mangieri LFL, Watanabe MAE, de Oliveira KB. Polymorphisms in the TGFB1 signal peptide influence human papillomavirus infection and development of cervical lesions. Med Microbiol Immunol 2018; 208:49-58. [PMID: 30167873 DOI: 10.1007/s00430-018-0557-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/23/2018] [Indexed: 12/26/2022]
Abstract
The main purpose was to assess the effect of c.29C>T and c.74G>C polymorphisms in the TGFB1 signal peptide on HPV infection and development of cervical lesions. Cervical swabs and blood samples were obtained from 349 outpatient women, along with socio-demographic and sexual behavioral data. The study population was stratified by absence or presence of HPV DNA, as tested by PCR, as well as by lesion grade. TGFB1 signal peptide polymorphisms were genotyped using PCR-restriction fragment length polymorphism. HPV DNA was detected in 172 (49.3%) patients. c.74GC and the combined c.29CC+CT/c.74GC genotype were more frequent in infected patients (35.1 and 15.7%) than in uninfected women (6.2 and 14.7%). Accordingly, these genotypes were associated with a higher risk of HPV infection, with odds ratio and 95% confidence interval of 2.81 and 1.35-5.86 (P = 0.004) for c.74GC and 3.14 and 1.42-6.94 (P = 0.004) for the combined genotype, respectively. High-grade lesions were also 2.48 times more likely to occur in c.29CC patients than in c.29TT patients, with a 95% confidence interval of 1.01-6.08 (P = 0.047). The data demonstrate that c.74G>C and c.29C>T polymorphisms are significantly associated with risk of HPV infection and high-grade squamous intraepithelial lesions, respectively. Thus, TGFB1 signal peptide polymorphisms are potential susceptibility markers.
Collapse
Affiliation(s)
- Kleber Paiva Trugilo
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | | | - Fernanda Costa Brandão Berti
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Nádia Calvo Martins Okuyama
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Fernando Cezar-Dos-Santos
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Michelle Mota Sena
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Luis Fernando Lásaro Mangieri
- Department of Gynecology and Obstetrics, Health Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Maria Angelica Ehara Watanabe
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil
| | - Karen Brajão de Oliveira
- Department of Pathological Science, Biological Science Center, State University of Londrina, Londrina, Paraná, Brazil.
| |
Collapse
|
167
|
Hseu YC, Chang GR, Pan JY, Rajendran P, Mathew DC, Li ML, Liao JW, Chen WTL, Yang HL. Antrodia camphorata inhibits epithelial-to-mesenchymal transition by targeting multiple pathways in triple-negative breast cancers. J Cell Physiol 2018; 234:4125-4139. [PMID: 30146779 DOI: 10.1002/jcp.27222] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 07/16/2018] [Indexed: 12/28/2022]
Abstract
Antrodia camphorata (AC) exhibits potential for engendering cell-cycle arrest as well as prompting apoptosis and metastasis inhibition in triple-negative breast cancer (TNBC) cells. We performed the current study to explore the anti-epithelial-to-mesenchymal transition (EMT) properties of fermented AC broth in TNBC cells. Our results illustrated that noncytotoxic concentrations of AC (20-60 μg/ml) reversed the morphological changes (fibroblastic-to-epithelial phenotype) as well as the EMT by upregulating the observed E-cadherin expression. Furthermore, we discovered treatment with AC substantially inhibit the Twist expression in human TNBC (MDA-MB-231) cells as well as in those that were transfected with Twist. In addition, we determined AC to decrease the observed Wnt/β-catenin nuclear translocation through a pathway determined to be dependent on GSK3β. Notably, AC treatment consistently inhibited the EMT by downregulating mesenchymal marker proteins like N-cadherin, vimentin, Snail, ZEB-1, and fibronectin; at that same time upregulating epithelial marker proteins like occludin and ZO-1. Bioluminescence imaging that was executed in vivo demonstrated AC substantially suppressed breast cancer metastasis to the lungs. Notably, we found that western blot analysis confirmed that AC decreased lung metastasis as demonstrated by upregulation of E-cadherin expression in biopsied lung tissue. Together with our results support the anti-EMT activity of AC, indicating AC as having the potential for acting as an anticancer agent for the treatment of human TNBC treatment.
Collapse
Affiliation(s)
- You-Cheng Hseu
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan.,Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.,Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
| | - Geng-Ruei Chang
- Institute of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Jian-You Pan
- Institute of Biotechnology and Bioinformatics, Asia University, Taichung, Taiwan
| | - Peramaiyan Rajendran
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Dony Chacko Mathew
- Department of Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Mei-Ling Li
- Department of Nutrition, Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung, Taiwan
| | - William Tzu-Liang Chen
- Division of Colorectal Surgery, Department of Surgery, Center of Minimally Invasive Surgery, China Medical University Hospital, China Medical University, Taichung, Taiwan.,College of Medicine, China Medical University, Taichung, Taiwan
| | - Hsin-Ling Yang
- Department of Nutrition, Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung, Taiwan
| |
Collapse
|
168
|
Plou J, Juste-Lanas Y, Olivares V, Del Amo C, Borau C, García-Aznar JM. From individual to collective 3D cancer dissemination: roles of collagen concentration and TGF-β. Sci Rep 2018; 8:12723. [PMID: 30143683 PMCID: PMC6109049 DOI: 10.1038/s41598-018-30683-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/31/2018] [Indexed: 02/06/2023] Open
Abstract
Cancer cells have the ability to migrate from the primary (original) site to other places in the body. The extracellular matrix affects cancer cell migratory capacity and has been correlated with tissue-specific spreading patterns. However, how the matrix orchestrates these behaviors remains unclear. Here, we investigated how both higher collagen concentrations and TGF-β regulate the formation of H1299 cell (a non-small cell lung cancer cell line) spheroids within 3D collagen-based matrices and promote cancer cell invasive capacity. We show that at low collagen concentrations, tumor cells move individually and have moderate invasive capacity, whereas when the collagen concentration is increased, the formation of cell clusters is promoted. In addition, when the concentration of TGF-β in the microenvironment is lower, most of the clusters are aggregates of cancer cells with a spheroid-like morphology and poor migratory capacity. In contrast, higher concentrations of TGF-β induced the formation of clusters with a notably higher invasive capacity, resulting in clear strand-like collective cell migration. Our results show that the concentration of the extracellular matrix is a key regulator of the formation of tumor clusters that affects their development and growth. In addition, chemical factors create a microenvironment that promotes the transformation of idle tumor clusters into very active, invasive tumor structures. These results collectively demonstrate the relevant regulatory role of the mechano-chemical microenvironment in leading the preferential metastasis of tumor cells to specific tissues with high collagen concentrations and TFG-β activity.
Collapse
Affiliation(s)
- J Plou
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain.
| | - Y Juste-Lanas
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - V Olivares
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - C Del Amo
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - C Borau
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain
| | - J M García-Aznar
- Multiscale in Mechanical and Biological Engineering, Aragon Institute of Engineering Research (I3A), Department of Mechanical Engineering, University of Zaragoza, 50018, Zaragoza, Spain.
| |
Collapse
|
169
|
Kordaß T, Weber CEM, Eisel D, Pane AA, Osen W, Eichmüller SB. miR-193b and miR-30c-1 * inhibit, whereas miR-576-5p enhances melanoma cell invasion in vitro. Oncotarget 2018; 9:32507-32522. [PMID: 30197759 PMCID: PMC6126698 DOI: 10.18632/oncotarget.25986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 07/29/2018] [Indexed: 01/01/2023] Open
Abstract
In cancer cells, microRNAs (miRNAs) are often aberrantly expressed resulting in impaired mRNA translation. In this study we show that miR-193b and miR-30c-1* inhibit, whereas miR-576-5p accelerates invasion of various human melanoma cell lines. Using Boyden chamber invasion assays the effect of selected miRNAs on the invasive capacity of various human melanoma cell lines was analyzed. Upon gene expression profiling performed on transfected A375 cells, CTGF, THBS1, STMN1, BCL9, RAC1 and MCL1 were identified as potential targets. For target validation, qPCR, Western blot analyses or luciferase reporter assays were applied. This study reveals opposed effects of miR-193b / miR-30c-1* and miR-576-5p, respectively, on melanoma cell invasion and on expression of BCL9 and MCL1, possibly accounting for the contrasting invasive phenotypes observed in A375 cells transfected with these miRNAs. The miRNAs studied and their targets identified fit well into a model proposed by us explaining the regulation of invasion associated genes and the observed opposed phenotypes as a result of networked direct and indirect miRNA / target interactions. The results of this study suggest miR-193b and miR-30c-1* as tumor-suppressive miRNAs, whereas miR-576-5p appears as potential tumor-promoting oncomiR. Thus, miR-193b and miR-30c-1* mimics as well as antagomiRs directed against miR-576-5p might become useful tools in future therapy approaches against advanced melanoma.
Collapse
Affiliation(s)
- Theresa Kordaß
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - Claudia E M Weber
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - David Eisel
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - Antonino A Pane
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - Wolfram Osen
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- GMP and T Cell Therapy Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
170
|
Chanda A, Sarkar A, Bonni S. The SUMO System and TGFβ Signaling Interplay in Regulation of Epithelial-Mesenchymal Transition: Implications for Cancer Progression. Cancers (Basel) 2018; 10:cancers10080264. [PMID: 30096838 PMCID: PMC6115711 DOI: 10.3390/cancers10080264] [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] [Received: 07/09/2018] [Revised: 08/06/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023] Open
Abstract
Protein post-translational modification by the small ubiquitin-like modifier (SUMO), or SUMOylation, can regulate the stability, subcellular localization or interactome of a protein substrate with key consequences for cellular processes including the Epithelial-Mesenchymal Transition (EMT). The secreted protein Transforming Growth Factor beta (TGFβ) is a potent inducer of EMT in development and homeostasis. Importantly, the ability of TGFβ to induce EMT has been implicated in promoting cancer invasion and metastasis, resistance to chemo/radio therapy, and maintenance of cancer stem cells. Interestingly, TGFβ-induced EMT and the SUMO system intersect with important implications for cancer formation and progression, and novel therapeutics identification.
Collapse
Affiliation(s)
- Ayan Chanda
- Department of Biochemistry and Molecular Biology, The Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Anusi Sarkar
- Department of Biochemistry and Molecular Biology, The Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| | - Shirin Bonni
- Department of Biochemistry and Molecular Biology, The Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
171
|
Emon B, Bauer J, Jain Y, Jung B, Saif T. Biophysics of Tumor Microenvironment and Cancer Metastasis - A Mini Review. Comput Struct Biotechnol J 2018; 16:279-287. [PMID: 30128085 PMCID: PMC6097544 DOI: 10.1016/j.csbj.2018.07.003] [Citation(s) in RCA: 163] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 07/20/2018] [Accepted: 07/21/2018] [Indexed: 02/07/2023] Open
Abstract
The role of tumor microenvironment in cancer progression is gaining significant attention. It is realized that cancer cells and the corresponding stroma co-evolve with time. Cancer cells recruit and transform the stromal cells, which in turn remodel the extra cellular matrix of the stroma. This complex interaction between the stroma and the cancer cells results in a dynamic feed-forward/feed-back loop with biochemical and biophysical cues that assist metastatic transition of the cancer cells. Although biochemistry has long been studied for the understanding of cancer progression, biophysical signaling is emerging as a critical paradigm determining cancer metastasis. In this mini review, we discuss the role of one of the biophysical cues, mostly the mechanical stiffness of tumor microenvironment, in cancer progression and its clinical implications.
Collapse
Key Words
- ADAMs, Adamalysins
- ANGPT2, Angiopoietin 2
- Activin/TGFβ
- CAF, Cancer associated fibroblast
- CSF-1, Colony stimulating factor 1
- CTGF, Connective tissue growth factor
- CYR61/CCN1, Cysteine-rich angiogenic inducer 61/CCN family member 1
- Cancer
- ECM stiffness
- ECM, Extracellular matrix
- EGF, Epidermal growth factor
- EMT, Epithelial to mesenchymal transition
- FGF, Fibroblast growth factor
- Growth factors
- HGF/SF, Hepatocyte growth factor/Scatter factor
- IGFs, Insulin-like growth factors
- IL-13, Interleukin-13
- IL-33, Interleukin-33
- IL-6, Interleukin-6
- KGF, Keratinocyte growth factor, also FGF7
- LOX, Lysyl Oxidase
- MMPs, Matrix metalloproteinases
- Metastasis
- NO, Nitric oxide
- SDF-1/CXCL12, Stromal cell-derived factor 1/C-X-C motif chemokine 12
- TACs, Tumor-associated collagen signatures
- TGFβ, Transforming growth factor β
- TNF-α, Tumor necrosis factor-α
- Tumor biophysics
- VEGF, Vascular endothelial growth factor
- α-SMA, α-Smooth muscle actin
Collapse
Affiliation(s)
- Bashar Emon
- Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, United States
| | - Jessica Bauer
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, United States
| | - Yasna Jain
- Department of Architecture, BRAC University, Dhaka
| | - Barbara Jung
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, United States
| | - Taher Saif
- Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, United States
- Bioengineering, University of Illinois at Urbana-Champaign, United States
| |
Collapse
|
172
|
Velapasamy S, Dawson CW, Young LS, Paterson IC, Yap LF. The Dynamic Roles of TGF-β Signalling in EBV-Associated Cancers. Cancers (Basel) 2018; 10:E247. [PMID: 30060514 PMCID: PMC6115974 DOI: 10.3390/cancers10080247] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
The transforming growth factor-β (TGF-β) signalling pathway plays a critical role in carcinogenesis. It has a biphasic action by initially suppressing tumorigenesis but promoting tumour progression in the later stages of disease. Consequently, the functional outcome of TGF-β signalling is strongly context-dependent and is influenced by various factors including cell, tissue and cancer type. Disruption of this pathway can be caused by various means, including genetic and environmental factors. A number of human viruses have been shown to modulate TGF-β signalling during tumorigenesis. In this review, we describe how this pathway is perturbed in Epstein-Barr virus (EBV)-associated cancers and how EBV interferes with TGF-β signal transduction. The role of TGF-β in regulating the EBV life cycle in tumour cells is also discussed.
Collapse
Affiliation(s)
- Sharmila Velapasamy
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Christopher W Dawson
- Institute of Cancer and Genomic Medicine, University of Birmingham, Birmingham B15 2TT, UK.
| | - Lawrence S Young
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK.
| | - Ian C Paterson
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Oral Cancer Research and Coordinating Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Lee Fah Yap
- Department of Oral & Craniofacial Sciences, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
- Oral Cancer Research and Coordinating Centre, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
173
|
Xie J, Li R. False discovery rate control for high dimensional networks of quantile associations conditioning on covariates. J R Stat Soc Series B Stat Methodol 2018; 80:1015-1034. [PMID: 31057329 DOI: 10.1111/rssb.12288] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Motivated by gene coexpression pattern analysis, we propose a novel sample quantile contingency (SQUAC) statistic to infer quantile associations conditioning on covariates. It features enhanced flexibility in handling variables with both arbitrary distributions and complex association patterns conditioning on covariates. We first derive its asymptotic null distribution, and then develop a multiple-testing procedure based on the SQUAC statistic to test simultaneously the independence between one pair of variables conditioning on covariates for all p(p-1)/2 pairs. Here, p is the length of the outcomes and could exceed the sample size. The testing procedure does not require resampling or perturbation and thus is computationally efficient. We prove by theory and numerical experiments that this testing method asymptotically controls the false discovery rate. It outperforms all alternative methods when the complex association patterns exist. Applied to a gastric cancer data set, this testing method successfully inferred the gene coexpression networks of early and late stage patients. It identified more changes in the networks which are associated with cancer survivals. We extend our method to the case that both the length of the outcomes and the length of covariates exceed the sample size, and show that the asymptotic theory still holds.
Collapse
Affiliation(s)
- Jichun Xie
- Duke University School of Medicine, Durham, USA
| | - Ruosha Li
- University of Texas Health Science Center at Houston, USA
| |
Collapse
|
174
|
Watt K, Newsted D, Voorand E, Gooding RJ, Majewski A, Truesdell P, Yao B, Tuschl T, Renwick N, Craig AW. MicroRNA-206 suppresses TGF-β signalling to limit tumor growth and metastasis in lung adenocarcinoma. Cell Signal 2018; 50:25-36. [PMID: 29935234 DOI: 10.1016/j.cellsig.2018.06.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 06/18/2018] [Accepted: 06/18/2018] [Indexed: 12/19/2022]
Abstract
MicroRNA-206 (miR-206) has demonstrated tumor suppressive effects in a variety of cancers. Numerous studies have identified aberrantly expressed targets of miR-206 that contribute to tumor progression and metastasis, however, the broader gene-networks and pathways regulated by miR-206 remain poorly defined. Here, we have ectopically expressed miR-206 in lung adenocarcinoma cell lines and tumors to identify differentially expressed genes, and study the effects on tumor growth and metastasis. In H1299 tumor xenograft assays, stable expression of miR-206 suppressed both tumor growth and metastasis in mice. Profiling of xenograft tumors using small RNA sequencing and a targeted panel of tumor progression and metastasis-related genes revealed a network of genes involved in TGF-β signalling that were regulated by miR-206. Among these were the TGFB1 ligand, as well as direct transcriptional targets of Smad3. Other differentially expressed genes included components of the extracellular matrix involved in TGF-β activation and signalling, including Thrombospondin-1, which is responsible for the activation of latent TGF-β in the stroma. In cultured lung adenocarcinoma cells treated with recombinant TGF-β, ectopic expression of miR-206 impaired canonical signalling, and expression of TGF-β target genes linked to epithelial-mesenchymal transition. This was due at least in part to the suppression of Smad3 protein levels in lung adenocarcinoma cells with ectopic miR-206 expression. Together, these findings indicate that miR-206 can suppress tumor progression and metastasis by limiting autocrine production of TGF-β, and highlight the potential utility of TGF-β inhibitors for the treatment of lung adenocarcinomas.
Collapse
Affiliation(s)
- Kathleen Watt
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Daniel Newsted
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Elena Voorand
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Robert J Gooding
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Physics, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Adrianna Majewski
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada
| | - Peter Truesdell
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Binchen Yao
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada
| | - Thomas Tuschl
- HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Neil Renwick
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Canada; HHMI Laboratory of RNA Molecular Biology, The Rockefeller University, New York, USA
| | - Andrew W Craig
- Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Canada.
| |
Collapse
|
175
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
176
|
Deregulation of Negative Controls on TGF-β1 Signaling in Tumor Progression. Cancers (Basel) 2018; 10:cancers10060159. [PMID: 29799477 PMCID: PMC6025439 DOI: 10.3390/cancers10060159] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 05/22/2018] [Accepted: 05/23/2018] [Indexed: 12/19/2022] Open
Abstract
The multi-functional cytokine transforming growth factor-β1 (TGF-β1) has growth inhibitory and anti-inflammatory roles during homeostasis and the early stages of cancer. Aberrant TGF-β activation in the late-stages of tumorigenesis, however, promotes development of aggressive growth characteristics and metastatic spread. Given the critical importance of this growth factor in fibrotic and neoplastic disorders, the TGF-β1 network is subject to extensive, multi-level negative controls that impact receptor function, mothers against decapentaplegic homolog 2/3 (SMAD2/3) activation, intracellular signal bifurcation into canonical and non-canonical pathways and target gene promotor engagement. Such negative regulators include phosphatase and tensin homologue (PTEN), protein phosphatase magnesium 1A (PPM1A), Klotho, bone morphogenic protein 7 (BMP7), SMAD7, Sloan-Kettering Institute proto-oncogene/ Ski related novel gene (Ski/SnoN), and bone morphogenetic protein and activin membrane-bound Inhibitor (BAMBI). The progression of certain cancers is accompanied by loss of expression, overexpression, mislocalization, mutation or deletion of several endogenous repressors of the TGF-β1 cascade, further modulating signal duration/intensity and phenotypic reprogramming. This review addresses how their aberrant regulation contributes to cellular plasticity, tumor progression/metastasis and reversal of cell cycle arrest and discusses the unexplored therapeutic value of restoring the expression and/or function of these factors as a novel approach to cancer treatment.
Collapse
|
177
|
Jaiswal RK, Kumar P, Kumar M, Yadava PK. hTERT promotes tumor progression by enhancing TSPAN13 expression in osteosarcoma cells. Mol Carcinog 2018; 57:1038-1054. [PMID: 29722072 DOI: 10.1002/mc.22824] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/06/2018] [Accepted: 04/14/2018] [Indexed: 01/11/2023]
Abstract
Telomerase complex maintains the length of the telome, cbre, and protects erosion of the physical ends of the eukaryotic chromosome in all actively dividing cells including cancer cells. Telomerase activation extends the lifespan of cells in culture by maintaining the length of the telomere. Compared to terminally differentiated somatic cells, telomerase activity remains high in over 90% of cancer cells. It has now become clear that the role of telomerase is much more complex than just telomere lengthening. The remaining 10% of cancers deploy ALT (alternative lengthening of telomeres) pathway to maintain telomere length. Telomerase inhibitors offer a good therapeutic option. Also, telomerase-associated molecules can be targeted provided their roles are clearly established. In any case, it is necessary to understand the major role of telomerase in cancer cells. Many studies have already been done to explore gene profiling of a telomerase positive cell by knocking down expression of hTERT (telomerase reverse transcriptase). To complement these studies, we performed global gene profiling of a telomerase negative cell by ectopically expressing hTERT and studied changes in the global gene expression patterns. Analysis of microarray data for telomerase negative cells ectopically expressing telomerase showed 76 differentially regulated genes, out of which 39 genes were upregulated, and 37 were downregulated. Three upregulated genes such as TSPAN13, HMGCS2, DLX5, and three downregulated genes like DHRS2, CRYAB, and PDLIM1 were validated by real-time PCR. Knocking down of TSAPN13 in hTERT overexpressing U2OS cells enhanced the apoptosis of the cells. TSPAN13 knockdown in these cells suppressed mesenchymal properties and enhanced epithelial character.
Collapse
Affiliation(s)
- Rishi K Jaiswal
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pramod Kumar
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India.,Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Manoj Kumar
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Pramod K Yadava
- Applied Molecular Biology Laboratory, School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
178
|
Townsend MH, Felsted AM, Piccolo SR, Robison RA, O'Neill KL. Metastatic colon adenocarcinoma has a significantly elevated expression of IL-10 compared with primary colon adenocarcinoma tumors. Cancer Biol Ther 2018; 19:913-920. [PMID: 28812429 PMCID: PMC6300346 DOI: 10.1080/15384047.2017.1360453] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Classical anti-inflammatory cytokines are known to play a role in both cancer progression as well as cancer elimination. We evaluated the anti-inflammatory cytokines IL-10 and TGF-β in patients with colon adenocarcinoma and metastatic colon adenocarcinoma using immunohistochemical assays to determine the expression of the cytokines between various malignant tissues. We found tissues stained with TGF-β showed no significant upregulation within malignant tumors when compared with normal tissue controls. We observed high levels of TGF-β presence in most tissues similar to GAPDH expression. Within both colon adenocarcinoma and metastatic carcinomas there was a significant variability among patients in the expression of IL-10. While some patients experienced insignificant increases in the cytokine compared with controls, other patients had a clear upregulation of the protein within their tissue. In addition, there was an increase in the number of patients positive for IL-10 upregulation within metastatic tumors when compared with primary tumors. These data indicate that there is substantial variability between patients in regards to IL-10 expression, which may further aid in characterizing tumors and evaluating metastatic potential.
Collapse
Affiliation(s)
- Michelle H Townsend
- a Department of Microbiology and Molecular Biology , Brigham Young University , Provo UT
| | - Abigail M Felsted
- a Department of Microbiology and Molecular Biology , Brigham Young University , Provo UT
| | - Stephen R Piccolo
- b Department of Biology , Brigham Young University , Provo UT.,c Department of Biomedical Informatics , University of Utah , Salt Lake City UT
| | - Richard A Robison
- a Department of Microbiology and Molecular Biology , Brigham Young University , Provo UT
| | - Kim L O'Neill
- a Department of Microbiology and Molecular Biology , Brigham Young University , Provo UT
| |
Collapse
|
179
|
Yang R, Liang J, Xu GX, Ding LM, Huang HM, Su QZ, Yan J, Li YC. Human cytomegalovirus glycoprotein B inhibits migration of breast cancer MDA-MB-231 cells and impairs TGF-β/Smad2/3 expression. Oncol Lett 2018; 15:7730-7738. [PMID: 29849800 PMCID: PMC5962863 DOI: 10.3892/ol.2018.8344] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 12/06/2017] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is a leading cause of cancer-associated mortality in females worldwide and evidence suggests that human cytomegalovirus (HCMV) infection may be implicated in the progress of breast cancer. HCMV glycoprotein B (gB) is the most abundant envelope protein and serves an important role in host cell entry. The present study aimed to clarify the role of HCMV gB in breast cancer cells. A HCMV gB construct (UL55) was generated and stable vUL55 gene lentivirus-transfected MDA-MB-231 cells were established. Subsequently, the effect of HCMV gB on the apoptosis and proliferation of MDA-MB-231 cells was measured by flow cytometry and Cell Counting Kit-8 assay. Furthermore, whether HCMV gB may modulate MDA-MB-231 cell migration was examined using Transwell and cell scratch assays. In addition, alterations in HCMV gB-modulated protein levels of transforming growth factor-β (TGF-β) and Mothers against decapentaplegic homologs 2/3 (Smad2/3) were detected using western blot analysis. The results indicated that UL55 cDNA was stably transfected into MDA-MB-231 cells, and that HCMV gB protein was stably expressed. No significant differences in cell apoptosis and proliferation between transfected (231-GB-OE) and negative control (231-NC) cells were observed, while the rate of cell migration was significantly decreased in the 231-GB-OE cells compared with the 231-NC cells. Additionally, the expression level of TGF-β and phosphorylation level of Smad2/3 were also decreased in 231-GB-OE cells compared with the 231-NC cells. Although certain previous studies indicated that HCMV infection was associated with breast carcinogenesis, the results of the present study indicate that the envelope protein HCMV gB exhibits no effect on cell apoptosis and proliferation, but inhibits breast cancer cell migration. This may be due to downregulated TGF-β/Smad signaling. Taken together, these studies may assist in developing anti-TGF-β agents that contribute to tumor suppression.
Collapse
Affiliation(s)
- Rui Yang
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Jie Liang
- Institute of Immunology, University of Heidelberg, Heidelberg D-69120, Germany
| | - Guo-Xiong Xu
- Center Laboratory, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Liu-Mei Ding
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Hong-Mei Huang
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Qi-Zhu Su
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Jing Yan
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| | - Yun-Chun Li
- Department of Laboratory Medicine, Jinshan Hospital, Fudan University, Shanghai 201508, P.R. China
| |
Collapse
|
180
|
Li Y, Liu Q, McGrail DJ, Dai H, Li K, Lin SY. CHD4 mutations promote endometrial cancer stemness by activating TGF-beta signaling. Am J Cancer Res 2018; 8:903-914. [PMID: 29888111 PMCID: PMC5992509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 04/08/2018] [Indexed: 06/08/2023] Open
Abstract
Endometrial cancer is one of the most common cancers of the female reproductive system. CHD4, a core subunit of the nucleosome remodeling and deacetylation (NuRD) complex, is frequently mutated in these patients. However the role it plays in promoting endometrial tumorigenesis is poorly understood. Here, we use genetic engineering approaches to modulate CHD4 expression levels and functionally evaluate hot spot mutations R975H and R1162W. We find that CHD4 depletion induces up-regulation of the cancer stem cell (CSC) marker CD133. This CSC phenotype was verified functionally by invasion ability, spheroid formation, and tumorigenicity in vivo. While cells expressing mutated CHD4 did not display impaired CHD4 DNA recruitment or NuRD complex formation, the mutations did reduce the stability of CHD4 protein to phenocopy CHD4 depletion. Consistently, patients with mutant CHD4 showed overexpression of CD133. Network analysis indicated activation of the TGFβ signaling pathway may drive this CSC phenotype, and chemical blockade of TGFβ abrogated the ability CHD4 knockdown cells to form spheroids. Taken together, these results indicate that mutations in CHD4 can promote endometrial tumorigenesis by increasing CSC character through TGFβ signaling pathway.
Collapse
Affiliation(s)
- Yang Li
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Qingxin Liu
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Daniel J McGrail
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Hui Dai
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| | - Kaiyi Li
- The Michael E. DeBakey Department of Surgery, Baylor College of MedicineHouston, Texas 77030, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer CenterHouston, Texas 77030, USA
| |
Collapse
|
181
|
O'Bryant D, Wang Z. The essential role of WD repeat domain 77 in prostate tumor initiation induced by Pten loss. Oncogene 2018; 37:4151-4163. [PMID: 29706654 DOI: 10.1038/s41388-018-0254-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 02/06/2018] [Accepted: 03/09/2018] [Indexed: 12/30/2022]
Abstract
Prostate cancer is the most commonly diagnosed malignancy among men, but few genetic factors that drive prostate cancer initiation have been identified. The WD repeat domain 77 (Wdr77) protein is essential for cellular proliferation when localizes in the cytoplasm of epithelial cells at the early stage of prostate development. In the adult prostate, it is transported into the nucleus and functions as a co-regulator of the androgen receptor to promote cellular differentiation and prostate function. This developmental process is reversed during prostate tumorigenesis, i.e., Wdr77 is translocated from the nucleus into the cytoplasm to drive proliferation of prostate cancer cells. In this study, we used in vivo genetic studies to further investigate the role of Wdr77 in prostate tumorigenesis. We found that prostate-specific deletion of Wdr77 abolished prostate tumor initiation induced by loss of the tumor suppressor Pten. Mechanistically, Wdr77 ablation inhibited E2F3 activation and enhanced TGFβ signaling, leading to attenuated cellular proliferation induced by loss of Pten. These findings establish a critical role of Wdr77 for prostate tumor initiation.
Collapse
Affiliation(s)
- Deon O'Bryant
- Department of Biological Sciences, The Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA, 30314, USA
| | - Zhengxin Wang
- Department of Biological Sciences, The Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA, 30314, USA.
| |
Collapse
|
182
|
Dual Roles of Serine-Threonine Kinase Receptor-Associated Protein (STRAP) in Redox-Sensitive Signaling Pathways Related to Cancer Development. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:5241524. [PMID: 29849900 PMCID: PMC5933018 DOI: 10.1155/2018/5241524] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 03/08/2018] [Indexed: 02/07/2023]
Abstract
Serine-threonine kinase receptor-associated protein (STRAP) is a transforming growth factor β (TGF-β) receptor-interacting protein that has been implicated in both cell proliferation and cell death in response to various stresses. However, the precise roles of STRAP in these cellular processes are still unclear. The mechanisms by which STRAP controls both cell proliferation and cell death are now beginning to be unraveled. In addition to its biological roles, this review also focuses on the dual functions of STRAP in cancers displaying redox dysregulation, where it can behave as a tumor suppressor or an oncogene (i.e., it can either inhibit or promote tumor formation), depending on the cellular context. Further studies are needed to define the functions of STRAP and the redox-sensitive intracellular signaling pathways that enhance either cell proliferation or cell death in human cancer tissues, which may help in the development of effective treatments for cancer.
Collapse
|
183
|
Zabkiewicz C, Resaul J, Hargest R, Jiang WG, Ye L. Increased Expression of Follistatin in Breast Cancer Reduces Invasiveness and Clinically Correlates with Better Survival. Cancer Genomics Proteomics 2018. [PMID: 28647698 DOI: 10.21873/cgp.20035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND/AIM Activin and its antagonist follistatin (FST) have been implicated in several solid tumours. This study investigated the role of FST in breast cancer. MATERIALS AND METHODS FST expression was examined using reverse transcription polymerase chain reaction (RT-PCR), real-time quantitative polymerase chain reaction (qPCR) and immunohistochemistry in a cohort of breast cancer samples. Expression was correlated to pathological and prognostic parameters in our patient cohort. FST was overexpressed in MCF-7 cells and assays for growth and invasion were performed. RESULTS FST is expressed in breast tissue, in the cytoplasm of mammary epithelial cells. Expression was decreased in breast cancer tissue in comparison to normal mammary tissue. Over-expression of FST in vitro led to significantly increased growth rate and reduced invasion. Higher FST associates with lower-grade tumours and better survival. CONCLUSION Our results suggest a role for FST as a suppressor of invasion and metastasis in breast cancer.
Collapse
Affiliation(s)
- Catherine Zabkiewicz
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, U.K
| | - Jeyna Resaul
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, U.K
| | - Rachel Hargest
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, U.K
| | - Wen Guo Jiang
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, U.K.
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff, U.K.
| |
Collapse
|
184
|
Park S, Yang KM, Park Y, Hong E, Hong CP, Park J, Pang K, Lee J, Park B, Lee S, An H, Kwak MK, Kim J, Kang JM, Kim P, Xiao Y, Nie G, Ooshima A, Kim SJ. Identification of Epithelial-Mesenchymal Transition-related Target Genes Induced by the Mutation of Smad3 Linker Phosphorylation. J Cancer Prev 2018; 23:1-9. [PMID: 29629343 PMCID: PMC5886489 DOI: 10.15430/jcp.2018.23.1.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/02/2022] Open
Abstract
Background Smad3 linker phosphorylation plays essential roles in tumor progression and metastasis. We have previously reported that the mutation of Smad3 linker phosphorylation sites (Smad3-Erk/Pro-directed kinase site mutant constructs [EPSM]) markedly reduced the tumor progression while increasing the lung metastasis in breast cancer. Methods We performed high-throughput RNA-Sequencing of the human prostate cancer cell lines infected with adenoviral Smad3-EPSM to identify the genes regulated by Smad3-EPSM. Results In this study, we identified genes which are differentially regulated in the presence of Smad3-EPSM. We first confirmed that Smad3-EPSM strongly enhanced a capability of cell motility and invasiveness as well as the expression of epithelial-mesenchymal transition marker genes, CDH2, SNAI1, and ZEB1 in response to TGF-β1 in human pancreatic and prostate cancer cell lines. We identified GADD45B, CTGF, and JUNB genes in the expression profiles associated with cell motility and invasiveness induced by the Smad3-EPSM. Conclusions These results suggested that inhibition of Smad3 linker phosphorylation may enhance cell motility and invasiveness by inducing expression of GADD45B, CTGF, and JUNB genes in various cancers.
Collapse
Affiliation(s)
- Sujin Park
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Kyung-Min Yang
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Yuna Park
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biomedical Science, College of Life Science, CHA University, CHA Bio Complex, Seongnam, Korea
| | - Eunji Hong
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | | | - Jinah Park
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Kyoungwha Pang
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biomedical Science, College of Life Science, CHA University, CHA Bio Complex, Seongnam, Korea
| | - Jihee Lee
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biomedical Science, College of Life Science, CHA University, CHA Bio Complex, Seongnam, Korea
| | - Bora Park
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Siyoung Lee
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Haein An
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biological Sciences, Sungkyunkwan University, Suwon, Korea
| | - Mi-Kyung Kwak
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Junil Kim
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Jin Muk Kang
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Pyunggang Kim
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Department of Biomedical Science, College of Life Science, CHA University, CHA Bio Complex, Seongnam, Korea
| | - Yang Xiao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing, China
| | - Akira Ooshima
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea
| | - Seong-Jin Kim
- Precision Medicine Research Center, Advanced Institutes of Convergence Technology, Suwon, Korea.,Theragen Etex Bio Institute, Suwon, Korea.,Department of Transdisciplinary Studies, Graduate School of Convergence Science and Technology, Seoul National University, Suwon, Korea
| |
Collapse
|
185
|
Li K, Zhang TT, Wang F, Cui B, Zhao CX, Yu JJ, Lv XX, Zhang XW, Yang ZN, Huang B, Li X, Hua F, Hu ZW. Metformin suppresses melanoma progression by inhibiting KAT5-mediated SMAD3 acetylation, transcriptional activity and TRIB3 expression. Oncogene 2018. [PMID: 29520103 DOI: 10.1038/s41388-018-0172-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Metformin has beneficial effects of preventing and treating cancers on type 2 diabetic patients. However, the role of metformin in non-diabetic cancer patients and the precise molecular mechanisms against cancer have not yet been sufficiently elucidated. We recently reported that the pseudokinase protein TRIB3 acts as a stress sensor linking metabolic stressors to cancer promotion by inhibiting autophagy and ubiquitin-proteasomal degradation systems; genetically abrogating of TRIB3 expression reduces tumourigenesis and cancer progression. Thus, TRIB3 is a potential therapeutic target for diverse cancers. In this study, we found that metformin attenuates melanoma growth and metastasis by reducing TRIB3 expression in non-diabetic C57BL/6 mice and diabetic KK-Ay mice; overexpression of TRIB3 protects metformin from the activation of autophagic flux, the clearance of accumulated tumour-promoting factors and the attenuation of tumour progression. We further elucidated that TRIB3 acts as an adaptor to recruit lysine acetyltransferase 5 (KAT5) to SMAD3 and induce a phosphorylation-dependent K333 acetylation of SMAD3, which sustains transcriptional activity of SMAD3 and subsequently enhances TRIB3 transcription. Metformin suppresses SMAD3 phosphorylation and decreases the KAT5/SMAD3 interaction, to attenuate the KAT5-mediated K333 acetylation of SMAD3, reduce the SMAD3 transcriptional activity and subsequent TRIB3 expression, thereby antagonizes melanoma progression. Together, our study not only defines a molecular mechanism by which metformin protects against melanoma progression by disturbing the KAT5/TRIB3/SMAD3 positive feedback loop in diabetes and non-diabetes mice, but also suggests a candidate diverse utility of metformin in tumour prevention and therapy because of suppressing stress protein TRIB3 expression.
Collapse
Affiliation(s)
- Ke Li
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.,Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | | | - Feng Wang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Bing Cui
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Chen-Xi Zhao
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiao-Jiao Yu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiao-Xi Lv
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xiao-Wei Zhang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Zhao-Na Yang
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Bo Huang
- Institute of Basic Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Xia Li
- Shandong University, Weihai, 264209, China
| | - Fang Hua
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zhuo-Wei Hu
- Immunology and Cancer Pharmacology Group, State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
186
|
Wilson RB. Hypoxia, cytokines and stromal recruitment: parallels between pathophysiology of encapsulating peritoneal sclerosis, endometriosis and peritoneal metastasis. Pleura Peritoneum 2018; 3:20180103. [PMID: 30911653 PMCID: PMC6405013 DOI: 10.1515/pp-2018-0103] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
Peritoneal response to various kinds of injury involves loss of peritoneal mesothelial cells (PMC), danger signalling, epithelial-mesenchymal transition and mesothelial-mesenchymal transition (MMT). Encapsulating peritoneal sclerosis (EPS), endometriosis (EM) and peritoneal metastasis (PM) are all characterized by hypoxia and formation of a vascularized connective tissue stroma mediated by vascular endothelial growth factor (VEGF). Transforming growth factor-β1 (TGF-β1) is constitutively expressed by the PMC and plays a major role in the maintenance of a transformed, inflammatory micro-environment in PM, but also in EPS and EM. Persistently high levels of TGF-β1 or stimulation by inflammatory cytokines (interleukin-6 (IL-6)) induce peritoneal MMT, adhesion formation and fibrosis. TGF-β1 enhances hypoxia inducible factor-1α expression, which drives cell growth, extracellular matrix production and cell migration. Disruption of the peritoneal glycocalyx and exposure of the basement membrane release low molecular weight hyaluronan, which initiates a cascade of pro-inflammatory mediators, including peritoneal cytokines (TNF-α, IL-1, IL-6, prostaglandins), growth factors (TGF-α, TGF-β, platelet-derived growth factor, VEGF, epidermal growth factor) and the fibrin/coagulation cascade (thrombin, Tissue factor, plasminogen activator inhibitor [PAI]-1/2). Chronic inflammation and cellular transformation are mediated by damage-associated molecular patterns, pattern recognition receptors, AGE-RAGE, extracellular lactate, pro-inflammatory cytokines, reactive oxygen species, increased glycolysis, metabolomic reprogramming and cancer-associated fibroblasts. The pathogenesis of EPS, EM and PM shows similarities to the cellular transformation and stromal recruitment of wound healing.
Collapse
Affiliation(s)
- Robert Beaumont Wilson
- Upper GI Surgery Department, Liverpool Hospital, Elizabeth St, Liverpool, 2170, NSW, Australia
| |
Collapse
|
187
|
Purnamawati P, Pawitan JA, Rachman A, Wanandi SI. Effects of umbilical cord- and adipose-derived stem cell secretomes on ALDH1A3 expression and autocrine TGF-β1 signaling in human breast cancer stem cells. F1000Res 2018. [DOI: 10.12688/f1000research.13609.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background: Nowadays, umbilical cord- and adipose-derived stem cells (UCSCs and ASCs) are the most common sources of mesenchymal stem cells (MSCs). As part of the tumor microenvironment, MSCs are known to communicate with cancer cells via their secretomes. Increased activity of aldehyde dehydrogenase-1 (ALDH1) has been widely used as a common intrinsic stemness marker in normal and cancer stem cells. Our study aimed to elaborate on the effect of UCSC and ASC secretomes on the expression of ALDH1A3, as one of the important variants of ALDH1, TGF-β1 and TGF-β receptor type I (TβRI) in human breast cancer stem cells (BCSCs). Methods: UCSCs and ASCs were cultured in serum-free α-MEM media under standard conditions for 24 hours. The conditioned medium (CM) containing secretomes of UCSCs and ASCs were collected and added 50% (v/v) to the cultured of human BCSCs for 72 hours. The mRNA expressions of ALDH1A3, TGF-β1, and TβRI were determined using quantitative Reverse Transcriptase Polymerase Chain Reaction (q-RT-PCR). Results: We found that CM of UCSCs significantly increased the ALDH1A3 expression of BCSCs in parallel with the increase of TGF-β1 and TβRI expressions. Conversely, CM of ASCs had no significant effect on the ALDH1A3 expression, but significantly decreased TGF-β1 and TβRI expressions of BCSCs. These results contradict our published data on ALDH1A1, which is another important variant of ALDH1, as well as data of the pluripotency markers OCT4 and SOX2 expressions. Conclusions: UCSC and ASC secretomes have different regulation on ALDH1A3 expression in human BCSCs, which may be related to the autocrine TGF-β1 signaling in modulating cell proliferation and stemness of BCSCs. Further studies are required to evaluate factors involved in the differential effects of UCSC and ASC secretomes that regulate ALDH1A3 expression in relation to autocrine TGF-β1 signaling and aggressiveness of human BCSCs.
Collapse
|
188
|
Knudson KM, Hicks KC, Luo X, Chen JQ, Schlom J, Gameiro SR. M7824, a novel bifunctional anti-PD-L1/TGFβ Trap fusion protein, promotes anti-tumor efficacy as monotherapy and in combination with vaccine. Oncoimmunology 2018; 7:e1426519. [PMID: 29721396 PMCID: PMC5927523 DOI: 10.1080/2162402x.2018.1426519] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/04/2018] [Accepted: 01/07/2018] [Indexed: 02/07/2023] Open
Abstract
Tumors evade host immune surveillance through multiple mechanisms, including the generation of a tumor microenvironment that suppresses immune effector function. Secretion of TGFβ and upregulation of immune checkpoint programmed cell death ligand-1 (PD-L1) are two main contributors to immune evasion and tumor progression. Here, we examined the efficacy of a first-in-class bifunctional checkpoint inhibitor, the fusion protein M7824, comprising the extracellular domain of human TGFβRII (TGFβ Trap) linked to the C-terminus of human anti-PD-L1 heavy chain (αPD-L1). We demonstrate that M7824 reduces plasma TGFβ1, binds to PD-L1 in the tumor, and decreases TGFβ-induced signaling in the tumor microenvironment in mice. In murine breast and colon carcinoma models, M7824 decreased tumor burden and increased overall survival as compared to targeting TGFβ alone. M7824 treatment promoted CD8+ T cell and NK cell activation, and both of these immune populations were required for optimal M7824-mediated tumor control. M7824 was superior to TGFβ- or αPD-L1-targeted therapies when in combination with a therapeutic cancer vaccine. These findings demonstrate the value of using M7824 to simultaneously target TGFβ and PD-L1/PD-1 immunosuppressive pathways to promote anti-tumor responses and efficacy. The studies also support the potential clinical use of M7824 as a monotherapy or in combination with other immunotherapies, such as therapeutic cancer vaccines, including for patients who have progressed on αPD-L1/αPD-1 checkpoint blockade therapies.
Collapse
Affiliation(s)
- Karin M Knudson
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristin C Hicks
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiaoling Luo
- Collaborative Protein Technology Resource (CPTR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jin-Qiu Chen
- Collaborative Protein Technology Resource (CPTR), Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey Schlom
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sofia R Gameiro
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
189
|
Zhang YH, Pan LH, Pang Y, Yang JX, Lv MJ, Liu F, Qu XF, Chen XX, Gong HJ, Liu D, Wei Y. GDF11/BMP11 as a novel tumor marker for liver cancer. Exp Ther Med 2018; 15:3495-3500. [PMID: 29545874 DOI: 10.3892/etm.2018.5861] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/08/2017] [Indexed: 12/16/2022] Open
Abstract
Growth differentiation factor 11 (GDF11), also known as bone morphogenetic protein 11, a member of the transforming growth factor-β superfamily, has been reported to be involved in colorectal cancer. However, the roles of GDF11 in Chinese patients with liver cancer and the underlying mechanisms have remained elusive. The present study assessed the expression of GDF11 in 10 paired samples of cancerous and normal tissues from Chinese liver cancer patients. The results indicated that the expression of GDF11 was significantly lower in cancerous tissues than in normal tissues. In vitro, the expression of GDF11 was reduced in a panel of liver cancer cell lines compared with that in a normal liver cell line at the mRNA and protein level. Treatment with GDF11 reduced the viability of HepG2 for up to 72 h and GDF11 treatment reduced the viability of SMMC-7721 after 48 and 72 h. Furthermore, GDF11 activated Smad2/3 signaling in HepG2 cells. In conclusion, GDF11 has a tumor suppressor role in liver cancer, exerts its effects through Smad2/3 signaling and may serve as a novel tumor marker in liver cancer diagnosis.
Collapse
Affiliation(s)
- Yong-Hui Zhang
- Department of Basic Medical Science, Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China.,Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Lian-Hong Pan
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Yi Pang
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Jin-Xin Yang
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Meng-Jia Lv
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Feng Liu
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Xue-Feng Qu
- Department of Nutrition and Food Hygiene, Zhejiang Academy of Medical Sciences, Hangzhou, Zhejiang 310013, P.R. China
| | - Xin-Xin Chen
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Hua-Jun Gong
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Dan Liu
- Department of Basic Medical Science, Chongqing Three Gorges Medical College, Chongqing 404120, P.R. China.,Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing 404120, P.R. China
| | - Yong Wei
- Key Laboratory of Intelligent Information Processing and Control, College of Electronic and Information Engineering, Chongqing Three Gorges University, Chongqing 404110, P.R. China
| |
Collapse
|
190
|
Wang Y, Liu Z, Han Y, Xu J, Huang W, Li Z. Medium Chain Triglycerides enhances exercise endurance through the increased mitochondrial biogenesis and metabolism. PLoS One 2018; 13:e0191182. [PMID: 29420554 PMCID: PMC5805166 DOI: 10.1371/journal.pone.0191182] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 12/31/2017] [Indexed: 11/18/2022] Open
Abstract
Medium Chain Triglycerides (MCT) is a dietary supplement and usually used along with medications for treating food absorption disorders including diarrhea, steatorrhea and liver disease. It has been shown that MCT plays a role in lowering weight, and decreasing metabolic syndrome, abdominal obesity and inflammation. However, it is still unknown whether MCT enhances exercise endurance. Here, we demonstrated that MCT containing diet improves high temperature induced exercise performance impairment. We found that MCT up-regulates the expression and protein levels of genes involved in mitochondrial biogenesis and metabolism. Further investigation demonstrated that the increased mitochondrial biogenesis and metabolism is mediated through the activation of Akt and AMPK signaling pathways and inhibition of TGF-β signaling pathway. Collectively, our findings indicate a beneficial effect of dietary MCT in exercise performance through the increase of mitochondrial biogenesis and metabolism.
Collapse
Affiliation(s)
- Ying Wang
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
| | - Zhenzhen Liu
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
| | - Yi Han
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
| | - Jiping Xu
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
| | - Wen Huang
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
- * E-mail:
| | - Zhaoshen Li
- Changhai Hospital,Second Military Medical University, Yangpu District, Shanghai, China
| |
Collapse
|
191
|
Mehta S, Lo Cascio C. Developmentally regulated signaling pathways in glioma invasion. Cell Mol Life Sci 2018; 75:385-402. [PMID: 28821904 PMCID: PMC5765207 DOI: 10.1007/s00018-017-2608-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 07/18/2017] [Accepted: 08/03/2017] [Indexed: 01/06/2023]
Abstract
Malignant gliomas are the most common, infiltrative, and lethal primary brain tumors affecting the adult population. The grim prognosis for this disease is due to a combination of the presence of highly invasive tumor cells that escape surgical resection and the presence of a population of therapy-resistant cancer stem cells found within these tumors. Several studies suggest that glioma cells have cleverly hijacked the normal developmental program of neural progenitor cells, including their transcriptional programs, to enhance gliomagenesis. In this review, we summarize the role of developmentally regulated signaling pathways that have been found to facilitate glioma growth and invasion. Furthermore, we discuss how the microenvironment and treatment-induced perturbations of these highly interconnected signaling networks can trigger a shift in cellular phenotype and tumor subtype.
Collapse
Affiliation(s)
- Shwetal Mehta
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA.
| | - Costanza Lo Cascio
- Division of Neurobiology, Barrow Brain Tumor Research Center, Barrow Neurological Institute, Phoenix, AZ, 85013, USA
| |
Collapse
|
192
|
Mikuła-Pietrasik J, Uruski P, Tykarski A, Książek K. The peritoneal "soil" for a cancerous "seed": a comprehensive review of the pathogenesis of intraperitoneal cancer metastases. Cell Mol Life Sci 2018; 75:509-525. [PMID: 28956065 PMCID: PMC5765197 DOI: 10.1007/s00018-017-2663-1] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 08/29/2017] [Accepted: 09/20/2017] [Indexed: 01/02/2023]
Abstract
Various types of tumors, particularly those originating from the ovary and gastrointestinal tract, display a strong predilection for the peritoneal cavity as the site of metastasis. The intraperitoneal spread of a malignancy is orchestrated by a reciprocal interplay between invading cancer cells and resident normal peritoneal cells. In this review, we address the current state-of-art regarding colonization of the peritoneal cavity by ovarian, colorectal, pancreatic, and gastric tumors. Particular attention is paid to the pro-tumoral role of various kinds of peritoneal cells, including mesothelial cells, fibroblasts, adipocytes, macrophages, the vascular endothelium, and hospicells. Anatomo-histological considerations on the pro-metastatic environment of the peritoneal cavity are presented in the broader context of organ-specific development of distal metastases in accordance with Paget's "seed and soil" theory of tumorigenesis. The activity of normal peritoneal cells during pivotal elements of cancer progression, i.e., adhesion, migration, invasion, proliferation, EMT, and angiogenesis, is discussed from the perspective of well-defined general knowledge on a hospitable tumor microenvironment created by the cellular elements of reactive stroma, such as cancer-associated fibroblasts and macrophages. Finally, the paper addresses the unique features of the peritoneal cavity that predispose this body compartment to be a niche for cancer metastases, presents issues that are topics of an ongoing debate, and points to areas that still require further in-depth investigations.
Collapse
Affiliation(s)
- Justyna Mikuła-Pietrasik
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Paweł Uruski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Hypertensiology, Angiology and Internal Medicine, Poznań University of Medical Sciences, Długa 1/2 Str., 61-848, Poznan, Poland.
| |
Collapse
|
193
|
Grover P, Nath S, Nye MD, Zhou R, Ahmad M, Mukherjee P. SMAD4-independent activation of TGF-β signaling by MUC1 in a human pancreatic cancer cell line. Oncotarget 2018; 9:6897-6910. [PMID: 29467938 PMCID: PMC5805524 DOI: 10.18632/oncotarget.23966] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 12/18/2017] [Indexed: 12/18/2022] Open
Abstract
Pancreatic Ductal Adenocarcinoma (PDA) has a mortality rate that nearly matches its incidence rate. Transforming Growth Factor Beta (TGF-β) is a cytokine with a dual role in tumor development switching from a tumor suppressor to a tumor promoter. There is limited knowledge of how TGF-β function switches during tumorigenesis. Mucin 1 (MUC1) is an aberrantly glycosylated, membrane-bound, glycoprotein that is overexpressed in >80% of PDA cases and is associated with poor prognosis. In PDA, MUC1 promotes tumor progression and metastasis via signaling through its cytoplasmic tail (MUC1-CT) and interacting with other oncogenic signaling molecules. We hypothesize that high levels of MUC1 in PDA may be partly responsible for the TGF-β functional switch during oncogenesis. We report that overexpression of MUC1 in BxPC3 human PDA cells (BxPC3.MUC1) enhances the induction of epithelial to mesenchymal transition leading to increased invasiveness in response to exogenous TGF-β1. Simultaneously, these cells resist TGF-β induced apoptosis by downregulating levels of cleaved caspases. We show that mutating the tyrosines in MUC1-CT to phenylalanine reverses the TGF-β induced invasiveness. This suggests that the tyrosine residues in MUC1-CT are required for TGF-β induced invasion. Some of these tyrosines are phosphorylated by the tyrosine kinase c-Src. Thus, treatment of BxPC3.MUC1 cells with a c-Src inhibitor (PP2) significantly reduces TGF-β induced invasiveness. Similar observations were confirmed in the Chinese hamster ovarian (CHO) cell line. Data strongly suggests that MUC1 may regulate TGF-β function in PDA cells and thus have potential clinical relevance in the use of TGF-β inhibitors in clinical trials.
Collapse
Affiliation(s)
- Priyanka Grover
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| | - Sritama Nath
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| | - Monica D. Nye
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| | - Ru Zhou
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| | - Mohammad Ahmad
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| | - Pinku Mukherjee
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, North Carolina 28223-0001, USA
| |
Collapse
|
194
|
Deryugina EI, Zajac E, Zilberberg L, Muramatsu T, Joshi G, Dabovic B, Rifkin D, Quigley JP. LTBP3 promotes early metastatic events during cancer cell dissemination. Oncogene 2018; 37:1815-1829. [PMID: 29348457 PMCID: PMC5889352 DOI: 10.1038/s41388-017-0075-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/09/2017] [Accepted: 11/10/2017] [Indexed: 02/04/2023]
Abstract
Latent Transforming Growth Factor β (TGFβ) Binding Proteins (LTBPs) are important for the secretion, activation and function of mature TGFβ, especially so in cancer cell physiology. However, specific roles of the LTBPs remain understudied in the context of the primary tumor microenvironment. Herein, we investigated the role of LTBP-3 in the distinct processes involved in cancer metastasis. By using three human tumor cell lines of different tissue origin (epidermoid HEp-3 and prostate PC-3 carcinomas and HT-1080 fibrosarcoma) and several metastasis models conducted in both mammalian and avian settings, we show that LTBP-3 is involved in the early dissemination of primary cancer cells, namely in the intravasation step of the metastatic cascade. Knockdown of LTBP-3 in all tested cell lines led to significant inhibition of tumor cell intravasation, but did not affect primary tumor growth. LTBP-3 was dispensable in the late steps of carcinoma cell metastasis that follow tumor cell intravasation, including vascular arrest, extravasation and tissue colonization. However, LTBP-3 depletion diminished the angiogenesis-inducing potential of HEp-3 cells in vivo, which was restorable by exogenous delivery of LTBP-3 protein. A similar compensatory approach rescued the dampened intravasation of LTBP-3-deficient HEp-3 cells, suggesting that LTBP-3 regulates the induction of the intravasation-supporting angiogenic vasculature within developing primary tumors. Using our recently developed microtumor model, we confirmed that LTBP-3 loss resulted in the development of intratumoral vessels with an abnormal microarchitecture incompatible with efficient intravasation of HEp-3 carcinoma cells. Collectively, these findings demonstrate that LTBP-3 represents a novel oncotarget that has distinctive functions in the regulation of angiogenesis-dependent tumor cell intravasation, a critical process during early cancer dissemination. Our experimental data are also consistent with the survival prognostic value of LTBP3 expression in early stage head and neck squamous cell carcinomas, further indicating a specific role for LTBP-3 in cancer progression towards metastatic disease.
Collapse
Affiliation(s)
| | - Ewa Zajac
- The Scripps Research Institute, La Jolla, CA, USA
| | - Lior Zilberberg
- The New York University School of Medicine, New York, NY, USA
| | | | - Grishma Joshi
- The New York University School of Medicine, New York, NY, USA
| | - Branka Dabovic
- The New York University School of Medicine, New York, NY, USA
| | - Daniel Rifkin
- The New York University School of Medicine, New York, NY, USA
| | | |
Collapse
|
195
|
Chae YK, Choi WM, Bae WH, Anker J, Davis AA, Agte S, Iams WT, Cruz M, Matsangou M, Giles FJ. Overexpression of adhesion molecules and barrier molecules is associated with differential infiltration of immune cells in non-small cell lung cancer. Sci Rep 2018; 8:1023. [PMID: 29348685 PMCID: PMC5773521 DOI: 10.1038/s41598-018-19454-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 12/12/2017] [Indexed: 12/26/2022] Open
Abstract
Immunotherapy is emerging as a promising option for lung cancer treatment. Various endothelial adhesion molecules, such as integrin and selectin, as well as various cellular barrier molecules such as desmosome and tight junctions, regulate T-cell infiltration in the tumor microenvironment. However, little is known regarding how these molecules affect immune cells in patients with lung cancer. We demonstrated for the first time that overexpression of endothelial adhesion molecules and cellular barrier molecule genes was linked to differential infiltration of particular immune cells in non-small cell lung cancer. Overexpression of endothelial adhesion molecule genes is associated with significantly lower infiltration of activated CD4 and CD8 T-cells, but higher infiltration of activated B-cells and regulatory T-cells. In contrast, overexpression of desmosome genes was correlated with significantly higher infiltration of activated CD4 and CD8 T-cells, but lower infiltration of activated B-cells and regulatory T-cells in lung adenocarcinoma. This inverse relation of immune cells aligns with previous studies of tumor-infiltrating B-cells inhibiting T-cell activation. Although overexpression of endothelial adhesion molecule or cellular barrier molecule genes alone was not predictive of overall survival in our sample, these genetic signatures may serve as biomarkers of immune exclusion, or resistance to T-cell mediated immunotherapy.
Collapse
Affiliation(s)
- Young Kwang Chae
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, 60611, USA. .,Northwestern University Feinberg School of Medicine, Chicago, 60611, USA.
| | - Wooyoung M. Choi
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - William H. Bae
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Jonathan Anker
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Andrew A. Davis
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Sarita Agte
- 0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, 60611 USA
| | - Wade T. Iams
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Marcelo Cruz
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Maria Matsangou
- 0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| | - Francis J. Giles
- 0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, Chicago, 60611 USA
| |
Collapse
|
196
|
Conditional deletion of RB1 in the Tie2 lineage leads to aortic valve regurgitation. PLoS One 2018; 13:e0190623. [PMID: 29304157 PMCID: PMC5755794 DOI: 10.1371/journal.pone.0190623] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2023] Open
Abstract
Objective Aortic valve disease is a complex process characterized by valve interstitial cell activation, disruption of the extracellular matrix culminating in valve mineralization occurring over many years. We explored the function of the retinoblastoma protein (pRb) in aortic valve disease, given its critical role in mesenchymal cell differentiation including bone development and mineralization. Approach and results We generated a mouse model of conditional pRb knockout (cKO) in the aortic valve regulated by Tie2-Cre-mediated excision of floxed RB1 alleles. Aged pRb cKO animals showed significantly more aortic valve regurgitation by echocardiography compared to pRb het control animals. The pRb cKO aortic valves had increased leaflet thickness without increased cellular proliferation. Histologic studies demonstrated intense α-SMA expression in pRb cKO leaflets associated with disorganized extracellular matrix and increased leaflet stiffness. The pRb cKO mice also showed increased circulating cytokine levels. Conclusions Our studies demonstrate that pRb loss in the Tie2-lineage that includes aortic valve interstitial cells is sufficient to cause age-dependent aortic valve dysfunction.
Collapse
|
197
|
Kumari K, Keshari S, Sengupta D, Sabat SC, Mishra SK. Transcriptome analysis of genes associated with breast cancer cell motility in response to Artemisinin treatment. BMC Cancer 2017; 17:858. [PMID: 29246124 PMCID: PMC5732364 DOI: 10.1186/s12885-017-3863-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022] Open
Abstract
Background Well-known anti-malarial drug artemisinin exhibits potent anti-cancerous activities. In-vivo and in-vitro studies showed its anti-tumor and immunomodulatory properties signifying it as a potent drug candidate for study. The studies of mechanisms of cell movement are relevant which can be understood by knowing the involvement of genes in an effect of a drug. Although cytotoxicity and anti-proliferative activity of artemisinin is evident, the genes participating in its anti-migratory and reduced invasive effect are not well studied. The present study reports the alteration in the expression of 84 genes involved in cell motility upon artemisinin treatment in MCF-7 breast cancer cells using pathway focused gene expression PCR array. In addition, the effect of artemisinin on epigenetic modifier HDACs is studied. Methods We checked the functional stimulus of artemisinin on cell viability, migration, invasion and apoptosis in breast cancerous cell lines. Using qRT-PCR and western blot, we validated the altered expression of relevant genes associated with proliferation, migration, invasion, apoptosis and mammary gland development. Results Artemisinin inhibited cell proliferation of estrogen receptor negative breast cancer cells with fewer efficacies in comparison to estrogen receptor positive ones. At the same time, cell viability and proliferation of normal breast epithelial MCF10A cells was un-affected. Artemisinin strongly inhibited cancer cell migration and invasion. Along with orphan nuclear receptors (ERRα, ERRβ and ERRγ), artemisinin altered the ERα/ERβ/PR/Her expression status of MCF-7 cells. The expression of genes involved in the signaling pathways associated with proliferation, migration, invasion and apoptosis was significantly altered which cooperatively resulted into reduced growth promoting activities of breast cancer cells. Interestingly, artemisinin exhibited inhibitory effect on histone deacetylases (HDACs). Conclusions Upregulated expression of tumor suppressor genes along with reduced expression of oncogenes significantly associated with growth stimulating signaling pathways in response to artemisinin treatment suggests its efficacy as an effective drug in breast cancer treatment.
Collapse
Affiliation(s)
- Kanchan Kumari
- Cancer Biology Laboratory, Institute of Life Sciences (Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India
| | | | | | - Surendra C Sabat
- Molecular biology of abiotic stress, Institute of Life Sciences, Nalco Square, Bhubaneswar, Odisha, 751023, India
| | - Sandip K Mishra
- Cancer Biology Laboratory, Institute of Life Sciences (Government of India), Nalco Square, Bhubaneswar, Odisha, 751023, India.
| |
Collapse
|
198
|
Toward Biological Subtyping of Papillary Renal Cell Carcinoma With Clinical Implications Through Histologic, Immunohistochemical, and Molecular Analysis. Am J Surg Pathol 2017; 41:1618-1629. [DOI: 10.1097/pas.0000000000000962] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
199
|
Bai J, Gao Z, Li X, Dong L, Han W, Nie J. Regulation of PD-1/PD-L1 pathway and resistance to PD-1/PD-L1 blockade. Oncotarget 2017; 8:110693-110707. [PMID: 29299180 PMCID: PMC5746415 DOI: 10.18632/oncotarget.22690] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Accepted: 11/08/2017] [Indexed: 12/31/2022] Open
Abstract
Immune checkpoint blockades, such as inhibitors against programmed death 1 (PD-1) and its ligand (PD-L1), have received extensive attention in the past decade because of their dramatic clinical outcomes in advanced malignancies. However, both primary and acquired resistance becomes one of the major obstacles, which greatly limits the long-lasting effects and wide application of PD-1/PD-L1 blockade therapy. PD-1/PD-L1 both regulates and is regulated by cellular signaling pathways and epigenetic modification, thus inhibiting the proliferation and effector function of T and B cells. The lack of tumor antigens and effective antigen presentation, aberrant activation of oncogenic pathways, mutations in IFN-γ signaling, immunosuppressive tumor microenvironment such as regulatory T cells, myeloid-derived suppressor cells, M2 macrophages, and immunoinhibitory cytokines can lead to resistance to PD-1/PD-L1 blockade. In this review, we describe PD-1 related signaling pathways, essential factors contributing to the resistance of PD-1 blockade, and discuss strategies to increase the efficacy of immunotherapy. Furthermore, we discuss the possibility of combined epigenetic therapy with PD-1 blockade as a potential promising approach for cancer treatment.
Collapse
Affiliation(s)
- Jie Bai
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Zhitao Gao
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Xiang Li
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Liang Dong
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Weidong Han
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Nie
- Department of Molecular Biology and Bio-Therapeutic, School of Life Science, Chinese PLA General Hospital, Beijing 100853, China
| |
Collapse
|
200
|
FBXO32 promotes microenvironment underlying epithelial-mesenchymal transition via CtBP1 during tumour metastasis and brain development. Nat Commun 2017; 8:1523. [PMID: 29142217 PMCID: PMC5688138 DOI: 10.1038/s41467-017-01366-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 09/12/2017] [Indexed: 12/12/2022] Open
Abstract
The set of events that convert adherent epithelial cells into migratory cells are collectively known as epithelial–mesenchymal transition (EMT). EMT is involved during development, for example, in triggering neural crest migration, and in pathogenesis such as metastasis. Here we discover FBXO32, an E3 ubiquitin ligase, to be critical for hallmark gene expression and phenotypic changes underlying EMT. Interestingly, FBXO32 directly ubiquitinates CtBP1, which is required for its stability and nuclear retention. This is essential for epigenetic remodeling and transcriptional induction of CtBP1 target genes, which create a suitable microenvironment for EMT progression. FBXO32 is also amplified in metastatic cancers and its depletion in a NSG mouse xenograft model inhibits tumor growth and metastasis. In addition, FBXO32 is essential for neuronal EMT during brain development. Together, these findings establish that FBXO32 acts as an upstream regulator of EMT by governing the gene expression program underlying this process during development and disease. Epithelial-to-mesenchymal transition (EMT) regulates both processes of organism development and changes in cell state causing disease. Here, the authors show that an E3 ubiquitin ligase, FBXO32, regulates EMT via CtBP1 and the transcriptional program, and also mediates cancer metastatic burden and neurogenesis.
Collapse
|