201
|
Liu X, Sun N, Mo N, Lu S, Song E, Ren C, Li Z. Quercetin inhibits kidney fibrosis and the epithelial to mesenchymal transition of the renal tubular system involving suppression of the Sonic Hedgehog signaling pathway. Food Funct 2019; 10:3782-3797. [PMID: 31180394 DOI: 10.1039/c9fo00373h] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Quercetin is the most ubiquitous flavonoid in fruits, herbs, vegetables and products made from them. It shows the potential to inhibit the progression of kidney fibrosis and the epithelial to mesenchymal transition (EMT) of the renal tubular system, but the molecular mechanism behind this is still not known. In our study, we explored the effect of quercetin treatment on extracellular matrix (ECM) deposition and stimulation of the EMT in vitro and in vivo and tried to deduce the mechanisms regulating these effects. In rats having unilateral ureter obstruction (UUO), quercetin treatment significantly prevented renal function decline. Quercetin reduced the TGF-β1 expression and inhibited the epithelial cell to mesenchymal cell phenotypic switch, as well as ECM deposition in rats with UUO. In cultured epithelial cells of the renal tubular region (NRK-52E), quercetin markedly ameliorated the EMT and ECM synthesis induced by TGF-β1. Activation of the Hedgehog pathway was closely related to EMT induction. Quercetin effectively suppressed the hyperactive Hedgehog pathway in NRK-52E cells treated with TGF-β1 and in kidney obstructed rats, which reduced the EMT, ECM deposition and cellular proliferation. Moreover, we examined certain transcriptional factors (slug, snail, ZEB-1 and twist) that govern the E-cadherin expression at the level of transcription. The results unveiled that the four transcriptional factors were highly repressed in NRK-52E cells treated with TGF-β1 and also in obstructed kidneys by quercetin-mediated inhibition. Therefore, these outcomes indicate that quercetin could alleviate fibrosis and the EMT in vitro and in vivo by inhibiting the activation of Hedgehog signaling and could act as a therapeutic agent for patients having several kinds of renal fibrotic diseases.
Collapse
Affiliation(s)
- Xianghua Liu
- Scientific Research and Experiment Center, Henan University of Chinese Medicine, China
| | | | | | | | | | | | | |
Collapse
|
202
|
Shakib H, Rajabi S, Dehghan MH, Mashayekhi FJ, Safari-Alighiarloo N, Hedayati M. Epithelial-to-mesenchymal transition in thyroid cancer: a comprehensive review. Endocrine 2019; 66:435-455. [PMID: 31378850 DOI: 10.1007/s12020-019-02030-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 07/19/2019] [Indexed: 12/12/2022]
Abstract
The Metastatic progression of solid tumors, such as thyroid cancer is a complex process which involves various factors. Current understanding on the role of epithelial-mesenchymal transition (EMT) in thyroid carcinomas suggests that EMT is implicated in the progression from follicular thyroid cancer (FTC) and papillary thyroid cancer (PTC) to poorly differentiated thyroid carcinoma (PDTC) and anaplastic thyroid cancer (ATC). According to the literature, the initiation of the EMT program in thyroid epithelial cells elevates the number of stem cells, which contribute to recurrent and metastatic diseases. The EMT process is orchestrated by a complex network of transcription factors, growth factors, signaling cascades, epigenetic modulations, and the tumor milieu. These factors have been shown to be dysregulated in thyroid carcinomas. Therefore, molecular interferences restoring the expression of tumor suppressors, or thwarting overexpressed oncogenes is a hopeful therapeutic method to improve the treatment of progressive diseases. In this review, we summarize the recent findings on EMT in thyroid cancer focusing on the main role-players and regulators of this process in thyroid tumors.
Collapse
Affiliation(s)
- Heewa Shakib
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sadegh Rajabi
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Nahid Safari-Alighiarloo
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Hedayati
- Cellular and Molecular Endocrine Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
203
|
Zhang X, Lu H, Xie S, Wu C, Guo Y, Xiao Y, Zheng S, Zhu H, Zhang Y, Bai Y. Resveratrol suppresses the myofibroblastic phenotype and fibrosis formation in kidneys via proliferation-related signalling pathways. Br J Pharmacol 2019; 176:4745-4759. [PMID: 31454852 PMCID: PMC6965682 DOI: 10.1111/bph.14842] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE Renal fibrosis acts as the common pathway leading to the development of end-stage renal disease. Previous studies have shown that resveratrol has anti-fibrotic activity, but its potential molecular mechanisms of action are not well understood. EXPERIMENTAL APPROACH The anti-fibrotic effects of resveratrol were assayed in a rat model of unilateral ureteral obstruction (UUO) in vivo and in fibroblasts and tubular epithelial cells (TECs) stimulated by TGF-β1 in vitro. Gene and protein expression levels were analysed by PCR, Western blotting, and immunohistochemical staining. KEY RESULTS Resveratrol inhibits the myofibroblastic phenotype and fibrosis formation in UUO kidneys by targeting fibroblast-myofibroblast differentiation (FMD) and epithelial-mesenchymal transition (EMT). The anti-fibrotic effects of resveratrol correlated with decreased proliferation of TECs in the interstitium and tubules, resulting in suppressed activity of the proliferation-related signalling pathways, including that of the MAPK, PI3K/Akt, Wnt/β-catenin, and JAK2/STAT3 pathways. Resveratrol treatment suppressed TGF-β1-induced FMD and the expression of the myofibroblastic phenotype in fibroblasts in vitro by antagonizing the activation of proliferation-related signalling. Similarly, TGF-β1-mediated overactivation of the proliferation-related signalling in TECs induced EMT, and the myofibroblastic phenotype was suppressed by resveratrol. The anti-fibrotic and anti-proliferative effects of resveratrol were associated with the inactivation of Smad2/3 signalling and resulted in a partial reversal of FMD and EMT and the inhibition of the myofibroblastic phenotype. CONCLUSIONS AND IMPLICATIONS Resveratrol suppresses the myofibroblastic phenotype and fibrosis formation in vivo and in vitro via proliferation-related pathways, making it a potential therapeutic agent for preventing renal fibrosis.
Collapse
Affiliation(s)
- Xing Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Zhejiang University School of MedicineHangzhouChina
| | - Hong Lu
- Department of Laboratory MedicineThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | | | - Cunzao Wu
- Department of TransplantationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yangyang Guo
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yanyi Xiao
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Shizhang Zheng
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Hengyue Zhu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Yan Zhang
- Department of TransplantationThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Institute of Kidney Health, Center for Health AssessmentWenzhou Medical UniversityWenzhouChina
| | - Yongheng Bai
- Key Laboratory of Diagnosis and Treatment of Severe Hepato‐Pancreatic Diseases of Zhejiang ProvinceThe First Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
- Institute of Kidney Health, Center for Health AssessmentWenzhou Medical UniversityWenzhouChina
| |
Collapse
|
204
|
Gasior K, Wagner NJ, Cores J, Caspar R, Wilson A, Bhattacharya S, Hauck ML. The role of cellular contact and TGF-beta signaling in the activation of the epithelial mesenchymal transition (EMT). Cell Adh Migr 2019; 13:63-75. [PMID: 30296203 PMCID: PMC6527395 DOI: 10.1080/19336918.2018.1526597] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 07/31/2018] [Accepted: 09/07/2018] [Indexed: 01/06/2023] Open
Abstract
The epithelial mesenchymal transition (EMT) is one step in the process through which carcinoma cells metastasize by gaining the cellular mobility associated with mesenchymal cells. This work examines the dual influence of the TGF-β pathway and intercellular contact on the activation of EMT in colon (SW480) and breast (MCF7) carcinoma cells. While the SW480 population revealed an intermediate state between the epithelial and mesenchymal states, the MC7 cells exhibited highly adhesive behavior. However, for both cell lines, an exogenous TGF-β signal and a reduction in cellular confluence can push a subgroup of the population towards the mesenchymal phenotype. Together, these results highlight that, while EMT is induced by the synergy of multiple signals, this activation varies across cell types.
Collapse
Affiliation(s)
- Kelsey Gasior
- Biomathematics Program, North Carolina State University, Raleigh, NC, USA
| | - Nikki J. Wagner
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Jhon Cores
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC, USA
| | - Rose Caspar
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| | - Alyson Wilson
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology & Toxicology, Michigan State University, East Lansing, MI, USA
- Center for Research on Ingredient Safety, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Marlene L. Hauck
- College of Veterinary Medicine, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
205
|
Xu Z, Ding W, Deng X. PM 2.5, Fine Particulate Matter: A Novel Player in the Epithelial-Mesenchymal Transition? Front Physiol 2019; 10:1404. [PMID: 31849690 PMCID: PMC6896848 DOI: 10.3389/fphys.2019.01404] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 10/31/2019] [Indexed: 12/12/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) refers to the conversion of epithelial cells to mesenchymal phenotype, which endows the epithelial cells with enhanced migration, invasion, and extracellular matrix production abilities. These characteristics link EMT with the pathogenesis of organ fibrosis and cancer progression. Recent studies have preliminarily established that fine particulate matter with an aerodynamic diameter of less than 2.5 μm (PM2.5) is correlated with EMT initiation. In this pathological process, PM2.5 particles, excessive reactive oxygen species (ROS) derived from PM2.5, and certain components in PM2.5, such as ions and polyaromatic hydrocarbons (PAHs), have been implicated as potential EMT mediators that are linked to the activation of transforming growth factor β (TGF-β)/SMADs, NF-κB, growth factor (GF)/extracellular signal-regulated protein kinase (ERK), GF/phosphatidylinositol 3-kinase (PI3K)/Akt, wingless/integrated (Wnt)/β-catenin, Notch, Hedgehog, high mobility group box B1 (HMGB1)-receptor for advanced glycation end-products (RAGE), and aryl hydrocarbon receptor (AHR) signaling cascades and to cytoskeleton rearrangement. These pathways directly and indirectly transduce pro-EMT signals that regulate EMT-related gene expression in epithelial cells, finally inducing the characteristic alterations in morphology and functions of epithelia. In addition, novel associations between autophagy, ATP citrate lyase (ACLY), and exosomes with PM2.5-induced EMT have also been summarized. However, some debates and paradoxes remain to be consolidated. This review discusses the potential molecular mechanisms underlying PM2.5-induced EMT, which might account for the latent role of PM2.5 in cancer progression and fibrogenesis.
Collapse
Affiliation(s)
- Zihan Xu
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenjun Ding
- Laboratory of Environment and Health, College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiaobei Deng
- Faculty of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
206
|
Liu C, Zhang L, Cui W, Du J, Li Z, Pang Y, Liu Q, Shang H, Meng L, Li W, Song L, Wang P, Xie Y, Wang Y, Liu Y, Hu J, Zhang W, Li F. Epigenetically upregulated GEFT-derived invasion and metastasis of rhabdomyosarcoma via epithelial mesenchymal transition promoted by the Rac1/Cdc42-PAK signalling pathway. EBioMedicine 2019; 50:122-134. [PMID: 31761617 PMCID: PMC6921210 DOI: 10.1016/j.ebiom.2019.10.060] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/02/2019] [Accepted: 10/31/2019] [Indexed: 01/12/2023] Open
Abstract
Background Metastasis of rhabdomyosarcoma (RMS) is the primary cause of tumour-related deaths. Previous studies have shown that overexpression of the guanine nucleotide exchange factor T (GEFT) is correlated with a poorer RMS prognosis, but the mechanism remains largely unexplored. Methods We focused on determining the influence of the GEFT-Rho-GTPase signalling pathway and the epithelial–mesenchymal transition (EMT) or mesenchymal–epithelial transition (MET) on RMS progression and metastasis by using RMS cell lines, BALB/c nude mice and cells and molecular biology techniques. Findings GEFT promotes RMS cell viability, migration, and invasion; GEFT also inhibits the apoptosis of RMS cells and accelerates the growth and lung metastasis of RMS by activating the Rac1/Cdc42 pathways. Interestingly, GEFT upregulates the expression levels of N-cadherin, Snail, Slug, Twist, Zeb1, and Zeb2 and reduces expression level of E-cadherin. Thus, GEFT influences the expression of markers for EMT and MET in RMS cells via the Rac1/Cdc42-PAK1 pathways. We also found that the level of GEFT gene promoter methylation in RMS is lower than that in normal striated muscle tissue. Significant differences were observed in the level of GEFT gene methylation in different histological subtypes of RMS. Interpretation These findings suggest that GEFT accelerates the tumourigenicity and metastasis of RMS by activating Rac1/Cdc42-PAK signalling pathway-induced EMT; thus, it may serve as a novel therapeutic target. Fund This work was supported by grants from the National Natural Science Foundation of China (81660441, 81460404, and 81160322) and Shihezi University Initiative Research Projects for Senior Fellows (RCZX201447). Funders had no role in the design of the study, data collection, data analysis, interpretation, or the writing of this report.
Collapse
Affiliation(s)
- Chunxia Liu
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China.
| | - Liang Zhang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Wenwen Cui
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Juan Du
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Zhenzhen Li
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Yuwen Pang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Qianqian Liu
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Hao Shang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Lian Meng
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Wanyu Li
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Lingxie Song
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Ping Wang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Yuwen Xie
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Yuanyuan Wang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Yang Liu
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Jianming Hu
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Wenjie Zhang
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China
| | - Feng Li
- Department of Pathology, Shihezi University School of Medicine and The Key Laboratories for Xinjiang Endemic and Ethnic Diseases, Chinese Ministry of Education, Shihezi 832002, Xinjiang, PR China; Department of Pathology and Medical Research Center, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China.
| |
Collapse
|
207
|
Deng M, Cai X, Long L, Xie L, Ma H, Zhou Y, Liu S, Zeng C. CD36 promotes the epithelial-mesenchymal transition and metastasis in cervical cancer by interacting with TGF-β. J Transl Med 2019; 17:352. [PMID: 31655604 PMCID: PMC6815430 DOI: 10.1186/s12967-019-2098-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/11/2019] [Indexed: 01/02/2023] Open
Abstract
Background Accumulating evidence indicates that CD36 initiates metastasis and correlates with an unfavorable prognosis in cancers. However, there are few reports regarding the roles of CD36 in initiation and metastasis of cervical cancer. Methods Using immunohistochemistry, we analyzed 133 cervical cancer samples for CD36 protein expression levels, and then investigated the correlation between changes in its expression and clinicopathologic parameters. The effect of CD36 expression on the epithelial–mesenchymal transition (EMT) in cervical cancer cells was evaluated by Western immunoblotting analysis. In vitro invasion and in vivo metastasis assays were also used to evaluate the role of CD36 in cervical cancer metastasis. Results In the present study, we confirmed that CD36 was highly expressed in cervical cancer samples relative to normal cervical tissues. Moreover, overexpression of CD36 promoted invasiveness and metastasis of cervical cancer cells in vitro and in vivo, while CD36 knockdown suppressed proliferation, migration, and invasiveness. We demonstrated that TGF-β treatment attenuated E-cadherin expression and enhanced the expression levels of CD36, vimentin, slug, snail, and twist in si-SiHa, si-HeLa, and C33a–CD36 cells, suggesting that TGF-β synergized with CD36 on EMT via active CD36 expression. We also observed that the expression levels of TGF-β in si-SiHa cells and si-HeLa cells were down-regulated, whereas the expression levels of TGF-β were up-regulated in C33a–CD36 cells. These results imply that CD36 and TGF-β interact with each other to promote the EMT in cervical cancer. Conclusions Our findings suggest that CD36 is likely to be an effective target for guiding individualized clinical therapy of cervical cancer.
Collapse
Affiliation(s)
- Min Deng
- Affliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, Guangdong, China
| | - Xiaodong Cai
- Department of Neurology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, Guangdong, China
| | - Ling Long
- Department of Neurology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510600, Guangdong, China
| | - Linying Xie
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Futian District, Shenzhen, 518033, Guangdong, China
| | - Hongmei Ma
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Futian District, Shenzhen, 518033, Guangdong, China
| | - Youjian Zhou
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Futian District, Shenzhen, 518033, Guangdong, China
| | - Shuguang Liu
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Futian District, Shenzhen, 518033, Guangdong, China.
| | - Chao Zeng
- Department of Pathology, The Eighth Affiliated Hospital, Sun Yat-sen University, Futian District, Shenzhen, 518033, Guangdong, China. .,Department of Pathology, Guangdong Medical University, Dongguan, 523808, Guangdong, China.
| |
Collapse
|
208
|
Pasanisi E, Ciavarella C, Valente S, Ricci F, Pasquinelli G. Differentiation and plasticity of human vascular wall mesenchymal stem cells, dermal fibroblasts and myofibroblasts: a critical comparison including ultrastructural evaluation of osteogenic potential. Ultrastruct Pathol 2019; 43:261-272. [DOI: 10.1080/01913123.2019.1673863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Emanuela Pasanisi
- Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, University Hospital, S. Orsola, Bologna, Italy
| | - Carmen Ciavarella
- Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, University Hospital, S. Orsola, Bologna, Italy
| | - Sabrina Valente
- Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, University Hospital, S. Orsola, Bologna, Italy
| | - Francesca Ricci
- Immunohaematology and Transfusion Medicine Service, University Hospital, S. Orsola, Bologna, Italy
| | - Gianandrea Pasquinelli
- Clinical Pathology, Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, University Hospital, S. Orsola, Bologna, Italy
- National Institute for Cardiovascular Research (INRC), University of Bologna, Italy
- National Institute of Biostructures and Biosystems (INBB), Rome, Italy
| |
Collapse
|
209
|
Krzysiek-Maczka G, Wrobel T, Targosz A, Szczyrk U, Strzalka M, Ptak-Belowska A, Czyz J, Brzozowski T. Helicobacter pylori-activated gastric fibroblasts induce epithelial-mesenchymal transition of gastric epithelial cells in vitro in a TGF-β-dependent manner. Helicobacter 2019; 24:e12653. [PMID: 31411795 DOI: 10.1111/hel.12653] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/03/2019] [Accepted: 07/06/2019] [Indexed: 12/16/2022]
Abstract
BACKGROUND Colonization of the gastric mucosa with Helicobacter pylori (Hp) leads to the cascade of pathologic events including local inflammation, gastric ulceration, and adenocarcinoma formation. Paracrine loops between tissue cells and Hp contribute to the formation of gastric cancerous loci; however, the specific mechanisms underlying existence of these loops remain unknown. We determined the phenotypic properties of gastric fibroblasts exposed to Hp (cagA+vacA+) infection and their influence on normal epithelial RGM-1 cells. MATERIALS AND METHODS RGM-1 cells were cultured in the media conditioned with Hp-activated gastric fibroblasts. Their morphology and phenotypical changes associated with epithelial-mesenchymal transition (EMT) were assessed by Nomarski and fluorescence microscopy and Western blot analysis. Motility pattern of RGM-1 cells was examined by time-lapse video microscopy and transwell migration assay. The content of TGF-β in Hp-activated fibroblast-conditioned media was determined by ELISA. RESULTS The supernatant from Hp-activated gastric fibroblasts caused the EMT-like phenotypic diversification of RGM-1 cells. The formation of fibroblastoid cell sub-populations, the disappearance of their collective migration, an increase in transmigration potential with downregulation of E-cadherin and upregulation of N-cadherin proteins, prominent stress fibers, and decreased proliferation were observed. The fibroblast (CAF)-like transition was manifested by increased secretome TGF-β level, α-SMA protein expression, and its incorporation into stress fibers, and the TGF-βR1 kinase inhibitor reduced the rise in Snail, Twist, and E-cadherin mRNA and increased E-cadherin expression induced by CAFs. CONCLUSION Gastric fibroblasts which are one of the main targets for Hp infection contribute to the paracrine interactions between Hp, gastric fibroblasts, and epithelial cells. TGF-β secreted by Hp-activated gastric fibroblasts prompting their differentiation toward CAF-like phenotype promotes the EMT-related phenotypic shifts in normal gastric epithelial cell populations. This mechanism may serve as the prerequisite for GC development.
Collapse
Affiliation(s)
- Gracjana Krzysiek-Maczka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Tomasz Wrobel
- Department of Cell Biology, The Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Aneta Targosz
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Urszula Szczyrk
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Malgorzata Strzalka
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Agata Ptak-Belowska
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| | - Jaroslaw Czyz
- Department of Cell Biology, The Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Cracow, Poland
| | - Tomasz Brzozowski
- Department of Physiology, The Faculty of Medicine, Jagiellonian University Medical College, Cracow, Poland
| |
Collapse
|
210
|
Zou Y, Song W, Zhou L, Mao Y, Hong W. House dust mite induces Sonic hedgehog signaling that mediates epithelial‑mesenchymal transition in human bronchial epithelial cells. Mol Med Rep 2019; 20:4674-4682. [PMID: 31702025 PMCID: PMC6797970 DOI: 10.3892/mmr.2019.10707] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/09/2019] [Indexed: 12/20/2022] Open
Abstract
Epithelial‑mesenchymal transition (EMT) provides a valuable source of fibroblasts that produce extracellular matrix in airway walls. The Sonic hedgehog (SHH) signaling pathway plays an essential role in regulating tissue turnover and homeostasis. SHH is strikingly upregulated in the bronchial epithelia during asthma. Snail1 is a major target of SHH signaling, which regulates EMT and fibroblast motility. The present study was designed to ascertain whether the combination of house dust mite (HDM) and transforming growth factor β1 (TGF‑β1) could induce EMT via the SHH signaling pathway in human bronchial epithelial cells (HBECs). HBEC cultures were treated with HDM/TGF‑β1 for different periods of time. The involvement of SHH signaling and EMT biomarkers was evaluated by quantitative real‑time PCR, western blotting and immunofluorescence staining. Small‑interfering RNA (siRNA) for glioma‑associated antigen‑1 (Gli1) or cyclopamine was used to inhibit SHH signaling in HBECs. HBECs stimulated by HDM/TGF‑β1 exhibited morphological features of EMT. E‑cadherin (an epithelial marker) was decreased after a 72‑h exposure to HDM/TGF‑β1 compared to that in the control cells, and the expression of type I collagen and FSP1 (mesenchymal markers) was increased. HDM/TGF‑β1 activated the SHH signaling pathway in HBECs, which led to Gli1 nuclear translocation and the transcriptional activation of Snail1 expression. Moreover, gene silencing or the pharmacological inhibition of Gli1 ameliorated EMT. In summary, these findings suggest that HDM/TGF‑β1 may induce EMT in HBECs via an SHH signaling mechanism. Inhibition of SHH signaling may be a novel therapeutic method for preventing airway remodeling in asthma.
Collapse
Affiliation(s)
- Yimin Zou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wenjuan Song
- Department of Economics, School of Economics and Management, Zhejiang Sci‑Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | - Lingxiao Zhou
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Yanxiong Mao
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, P.R. China
| | - Wei Hong
- Biological Laboratory Center, Guangzhou Institute of Biomedicine and Health Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong 510030, P.R. China
| |
Collapse
|
211
|
Bai J, Kwok WC, Thiery JP. Traditional Chinese Medicine and regulatory roles on epithelial-mesenchymal transitions. Chin Med 2019; 14:34. [PMID: 31558913 PMCID: PMC6755703 DOI: 10.1186/s13020-019-0257-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 09/11/2019] [Indexed: 02/06/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is a critical biological process allowing epithelial cells to de-differentiate into mesenchymal cells. Orchestrated signaling pathways cooperatively induce EMT and effect physiological, sometimes pathological outcomes. Traditional Chinese Medicine (TCM) has been clinically prescribed for thousands of years and recent studies have found that TCM therapies can participate in EMT regulation. In this review, the historical discovery of EMT will be introduced, followed by a brief overview of its major roles in development and diseases. The second section will focus on EMT in organ fibrosis and tissue regeneration. The third section discusses EMT-induced cancer metastasis, and details how EMT contribute to distant dissemination. Finally, new EMT players are described, namely microRNA, epigenetic modifications, and alternative splicing. TCM drugs that affect EMT proven through an evidence-based research approach will be presented in each section.
Collapse
Affiliation(s)
- Jing Bai
- 1Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, USA
| | - Wee Chiew Kwok
- 2Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jean-Paul Thiery
- Guangzhou Regenerative Medicine and Health, Guangdong Laboratory, Guangzhou, China
| |
Collapse
|
212
|
Presence of Stromal Cells Enhances Epithelial-to-Mesenchymal Transition (EMT) Induction in Lung Bronchial Epithelium after Protracted Exposure to Oxidative Stress of Gamma Radiation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4120379. [PMID: 31583039 PMCID: PMC6754954 DOI: 10.1155/2019/4120379] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/19/2019] [Accepted: 07/23/2019] [Indexed: 02/03/2023]
Abstract
The aim of the study was to investigate the role of a microenvironment in the induction of epithelial-to-mesenchymal transition (EMT) as a sign of early stages of carcinogenesis in human lung epithelial cell lines after protracted low-dose rate γ-radiation exposures. BEAS-2B and HBEC-3KT lung cell lines were irradiated with low-dose rate γ-rays (137Cs, 1.4 or 14 mGy/h) to 0.1 or 1 Gy with or without adding TGF-β. TGF-β-treated samples were applied as positive EMT controls and tested in parallel to find out if the radiation has a potentiating effect on the EMT induction. To evaluate the effect of the stromal component, the epithelial cells were irradiated in cocultures with stromal MRC-9 lung fibroblasts. On day 3 post treatment, the EMT markers: α-SMA, vimentin, fibronectin, and E-cadherin, were analyzed. The oxidative stress levels were evaluated by 8-oxo-dG analysis in both epithelial and fibroblast cells. The protracted exposure to low Linear Energy Transfer (LET) radiation at the total absorbed dose of 1 Gy was able to induce changes suggestive of EMT. The results show that the presence of the stromal component and its signaling (TGF-β) in the cocultures enhances the EMT. Radiation had a minor cumulative effect on the TGF-β-induced EMT with both doses. The oxidative stress levels were higher than the background in both epithelial and stromal cells post chronic irradiation (0.1 and 1 Gy); as for the BEAS-2B cell line, the increase was statistically significant. We suggest that the induction of EMT in bronchial epithelial cells by radiation requires more than single acute exposure and the presence of stromal component might enhance the effect through free radical production and accumulation.
Collapse
|
213
|
Park YJ, Bang IJ, Jeong MH, Kim HR, Lee DE, Kwak JH, Chung KH. Effects of β-Sitosterol from Corn Silk on TGF-β1-Induced Epithelial-Mesenchymal Transition in Lung Alveolar Epithelial Cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:9789-9795. [PMID: 31373816 DOI: 10.1021/acs.jafc.9b02730] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Pulmonary fibrosis is a chronic lung disease characterized by abnormal accumulation of the extracellular matrix (ECM). Chronic damage of the alveolar epithelium leads to a process called "epithelial-mesenchymal transition" (EMT) and increases synthesis and deposition of ECM proteins. Therefore, inhibition of EMT might be a promising therapeutic approach for the treatment of pulmonary fibrosis. β-Sitosterol is one of the most abundant phytosterols in the plant kingdom and the major constituent in corn silk, which is derived from the stigma and style of maize (Zea mays). In this study, we elucidated that β-sitosterol inhibited transforming growth factor-β1 (TGF-β1)-induced EMT and consequently had an antifibrotic effect. β-Sitosterol (1-10 μg/mL) significantly downregulated the TGF-β1-induced fibrotic proteins, such as collagen, fibronectin, and α-smooth muscle actin in human alveolar epithelial cells (p < 0.01). After 24 h, relative wound density (RWD) was increased in TGF-β1 treated group (82.16 ± 5.70) compare to the control group (64.63 ± 2.21), but RWD was decreased in β-sitosterol cotreated group (10 μg/mL: 71.54 ± 7.39; 20 μg/mL: 65.69 ± 6.42). In addition, the changes of the TGF-β1-induced morphological shape and protein expression of EMT markers, N-cadherin, vimentin, and E-cadherin, were significantly blocked by β-sitosterol treatment (p < 0.01). The effects of β-sitosterol on EMT were found to be associated with the TGF-β1/Snail pathway, which is regulated by Smad and non-Smad signaling pathways. Taken together, these findings suggest that β-sitosterol can be used to attenuate pulmonary fibrosis through suppression of EMT by inhibiting the TGF-β1/Snail pathway.
Collapse
Affiliation(s)
- Yong Joo Park
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| | - In Jae Bang
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| | - Mi Ho Jeong
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| | - Ha Ryong Kim
- College of Pharmacy , Daegu Catholic University , Gyeongsan 38430 , Republic of Korea
| | - Da Eun Lee
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| | - Jong Hwan Kwak
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| | - Kyu Hyuck Chung
- School of Pharmacy , Sungkyunkwan University , Suwon 16419 , Republic of Korea
| |
Collapse
|
214
|
The Use of Genetically Engineered Mouse Models for Studying the Function of Mutated Driver Genes in Pancreatic Cancer. J Clin Med 2019; 8:jcm8091369. [PMID: 31480737 PMCID: PMC6780401 DOI: 10.3390/jcm8091369] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/26/2019] [Accepted: 08/27/2019] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is often treatment-resistant, with the emerging standard of care, gemcitabine, affording only a few months of incrementally-deteriorating survival. Reflecting on the history of failed clinical trials, genetically engineered mouse models (GEMMs) in oncology research provides the inspiration to discover new treatments for pancreatic cancer that come from better knowledge of pathogenesis mechanisms, not only of the derangements in and consequently acquired capabilities of the cancer cells, but also in the aberrant microenvironment that becomes established to support, sustain, and enhance neoplastic progression. On the other hand, the existing mutational profile of pancreatic cancer guides our understanding of the disease, but leaves many important questions of pancreatic cancer biology unanswered. Over the past decade, a series of transgenic and gene knockout mouse modes have been produced that develop pancreatic cancers with features reflective of metastatic pancreatic ductal adenocarcinoma (PDAC) in humans. Animal models of PDAC are likely to be essential to understanding the genetics and biology of the disease and may provide the foundation for advances in early diagnosis and treatment.
Collapse
|
215
|
Caruso S, Calatayud AL, Pilet J, La Bella T, Rekik S, Imbeaud S, Letouzé E, Meunier L, Bayard Q, Rohr-Udilova N, Péneau C, Grasl-Kraupp B, de Koning L, Ouine B, Bioulac-Sage P, Couchy G, Calderaro J, Nault JC, Zucman-Rossi J, Rebouissou S. Analysis of Liver Cancer Cell Lines Identifies Agents With Likely Efficacy Against Hepatocellular Carcinoma and Markers of Response. Gastroenterology 2019; 157:760-776. [PMID: 31063779 DOI: 10.1053/j.gastro.2019.05.001] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/19/2019] [Accepted: 05/01/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND AND AIMS Hepatocellular carcinomas (HCCs) are heterogeneous aggressive tumors with low rates of response to treatment at advanced stages. We screened a large panel of liver cancer cell lines (LCCLs) to identify agents that might be effective against HCC and markers of therapeutic response. METHODS We performed whole-exome RNA and microRNA sequencing and quantification of 126 proteins in 34 LCCLs. We screened 31 anticancer agents for their ability to decrease cell viability. We compared genetic, RNA, and protein profiles of LCCLs with those of primary HCC samples and searched for markers of response. RESULTS The protein, RNA and mutational signatures of the LCCLs were similar to those of the proliferation class of HCC, which is the most aggressive tumor type. Cell lines with alterations in genes encoding members of the Ras-MAPK signaling pathway and that required fibroblast growth factor (FGF)19 signaling via FGF receptor 4 for survival were more sensitive to trametinib than to FGF receptor 4 inhibitors. Amplification of FGF19 resulted in increased activity of FGF19 only in tumor cells that kept a gene expression pattern of hepatocyte differentiation. We identified single agents and combinations of agents that reduced viability of cells with features of the progenitor subclass of HCC. LCCLs with inactivating mutations in TSC1 and TSC2 were sensitive to the mammalian target of rapamycin inhibitor rapamycin, and cells with inactivating mutations in TP53 were sensitive to the Aurora kinase A inhibitor alisertib. Amplification of MET was associated with hypersensitivity to cabozantinib and the combination of sorafenib and inhibitors of MAP kinase 1 and MAP kinase2 had a synergistic antiproliferative effect. CONCLUSION LCCLs can be screened for drugs and agents that might be effective for treatment of HCC. We identified genetic alterations and gene expression patterns associated with response to these agents. This information might be used to select patients for clinical trials.
Collapse
Affiliation(s)
- Stefano Caruso
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Anna-Line Calatayud
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Jill Pilet
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Tiziana La Bella
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Samia Rekik
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Sandrine Imbeaud
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Eric Letouzé
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Léa Meunier
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Quentin Bayard
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Nataliya Rohr-Udilova
- Division of Gastroenterology and Hepatology, Department of Internal, Medicine III, Medical University of Vienna, Vienna, Austria
| | - Camille Péneau
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Bettina Grasl-Kraupp
- Department of Medicine I, Division: Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Leanne de Koning
- RPPA Platform, Curie Institute, PSL Research University, Paris, France
| | - Bérengère Ouine
- RPPA Platform, Curie Institute, PSL Research University, Paris, France
| | - Paulette Bioulac-Sage
- Bariton INSERM, UMR-1053, Bordeaux, France; Department of Pathology, Pellegrin Hospital, Hospital of Bordeaux, Bordeaux, France
| | - Gabrielle Couchy
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France
| | - Julien Calderaro
- Anathomopathology Department, Henri Mondor Hospital, Créteil, University of Paris Est Créteil, Inserm U955, Team 18, Mondor Institute of Biomedical Research, Créteil, France
| | - Jean-Charles Nault
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France; Liver unit, Jean Verdier Hospital, University Hospitals Paris-Seine-Saint-Denis, AP-HP, Bondy, France; Training and Research Unit of Health Medicine and Human Biology, University of Paris 13, Community of Universities and Institutions Sorbonne Paris Cité, Paris, France
| | - Jessica Zucman-Rossi
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France; European Hospital Georges Pompidou, AP-HP, F-75015, Paris, France.
| | - Sandra Rebouissou
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, UMRS-1138, F-75006 Paris, France; Functional Genomics of Solid Tumors, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris 13, Labex Immuno-Oncology, équipe labellisée Ligue Contre le Cancer, F-75000 Paris, France.
| |
Collapse
|
216
|
Bisceglia F, Battistelli C, Noce V, Montaldo C, Zammataro A, Strippoli R, Tripodi M, Amicone L, Marchetti A. TGFβ Impairs HNF1α Functional Activity in Epithelial-to-Mesenchymal Transition Interfering With the Recruitment of CBP/p300 Acetyltransferases. Front Pharmacol 2019; 10:942. [PMID: 31543815 PMCID: PMC6728925 DOI: 10.3389/fphar.2019.00942] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/24/2019] [Indexed: 12/24/2022] Open
Abstract
The cytokine transforming growth factor β (TGFβ) plays a crucial role in the induction of both epithelial-to-mesenchymal transition (EMT) program and fibro-cirrhotic process in the liver, where it contributes also to organ inflammation following several chronic injuries. All these pathological situations greatly increase the risk of hepatocellular carcinoma (HCC) and contribute to tumor progression. In particular, late-stage HCCs are characterized by constitutive activation of TGFβ pathway and by an EMT molecular signature leading to the acquisition of invasive and metastatic properties. In these pathological conditions, the cytokine has been shown to induce the transcriptional downregulation of HNF1α, a master regulator of the epithelial/hepatocyte differentiation and of the EMT reverse process, the mesenchymal-to-epithelial transition (MET). Therefore, the restoration of HNF1α expression/activity has been proposed as targeted therapeutic strategy for liver fibro-cirrhosis and late-stage HCCs. In this study, TGFβ is found to trigger an early functional inactivation of HNF1α during EMT process that anticipates the effects of the transcriptional downregulation of its own gene. Mechanistically, the cytokine, while not affecting the HNF1α DNA-binding capacity, impaired its ability to recruit CBP/p300 acetyltransferases on target gene promoters and, consequently, its transactivating function. The loss of HNF1α capacity to bind to CBP/p300 and HNF1α functional inactivation have been found to correlate with a change of its posttranslational modification profile. Collectively, the results obtained in this work unveil a new level of HNF1α functional inactivation by TGFβ and contribute to shed light on the early events triggering EMT in hepatocytes. Moreover, these data suggest that the use of HNF1α as anti-EMT tool in a TGFβ-containing microenvironment may require the design of new therapeutic strategies overcoming the TGFβ-induced HNF1α inactivation.
Collapse
Affiliation(s)
- Francesca Bisceglia
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Cecilia Battistelli
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Valeria Noce
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Claudia Montaldo
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | - Agatino Zammataro
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Raffaele Strippoli
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | - Marco Tripodi
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
- National Institute for Infectious Diseases L. Spallanzani, IRCCS, Rome, Italy
| | - Laura Amicone
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Alessandra Marchetti
- Istituto Pasteur Italia–Fondazione Cenci Bolognetti, Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
217
|
Brindisi M, Saraswati AP, Brogi S, Gemma S, Butini S, Campiani G. Old but Gold: Tracking the New Guise of Histone Deacetylase 6 (HDAC6) Enzyme as a Biomarker and Therapeutic Target in Rare Diseases. J Med Chem 2019; 63:23-39. [PMID: 31415174 DOI: 10.1021/acs.jmedchem.9b00924] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Epigenetic regulation orchestrates many cellular processes and greatly influences key disease mechanisms. Histone deacetylase (HDAC) enzymes play a crucial role either as biomarkers or therapeutic targets owing to their involvement in specific pathophysiological pathways. Beyond their well-characterized role as histone modifiers, HDACs also interact with several nonhistone substrates and their increased expression has been highlighted in specific diseases. The HDAC6 isoform, due to its unique cytoplasmic localization, modulates the acetylation status of tubulin, HSP90, TGF-β, and peroxiredoxins. HDAC6 also exerts noncatalytic activities through its interaction with ubiquitin. Both catalytic and noncatalytic functions of HDACs are being actively studied in the field of specific rare disorders beyond the well-established role in carcinogenesis. This Perspective outlines the application of HDAC(6) inhibitors in rare diseases, such as Rett syndrome, inherited retinal disorders, idiopathic pulmonary fibrosis, and Charcot-Marie-Tooth disease, highlighting their therapeutic potential as innovative and targeted disease-modifying agents.
Collapse
Affiliation(s)
- Margherita Brindisi
- Department of Pharmacy, Department of Excellence 2018-2022 , University of Naples Federico II , Via D. Montesano 49 , I-80131 Naples , Italy
| | - A Prasanth Saraswati
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Simone Brogi
- Department of Pharmacy , University of Pisa , via Bonanno 6 , 56126 , Pisa , Italy
| | - Sandra Gemma
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Stefania Butini
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| | - Giuseppe Campiani
- Department of Biotechnology, Chemistry and Pharmacy, Department of Excellence 2018-2022 , University of Siena , via Aldo Moro 2 , 53100 , Siena , Italy
| |
Collapse
|
218
|
Pelullo M, Zema S, Nardozza F, Checquolo S, Screpanti I, Bellavia D. Wnt, Notch, and TGF-β Pathways Impinge on Hedgehog Signaling Complexity: An Open Window on Cancer. Front Genet 2019; 10:711. [PMID: 31552081 PMCID: PMC6736567 DOI: 10.3389/fgene.2019.00711] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
Constitutive activation of the Hedgehog (Hh) signaling pathway is associated with increased risk of developing several malignancies. The biological and pathogenic importance of Hh signaling emphasizes the need to control its action tightly, both physiologically and therapeutically. Evidence of crosstalk between Hh and other signaling pathways is reported in many tumor types. Here, we provide an overview of the current knowledge about the communication between Hh and major signaling pathways, such as Notch, Wnt, and transforming growth factor β (TGF-β), which play critical roles in both embryonic and adult life. When these pathways are unbalanced, impaired crosstalk contributes to disease development. It is reported that more than one of these pathways are active in different type of tumors, at the same time. Therefore, starting from a plethora of stimuli that activate multiple signaling pathways, we describe the signals that preferentially converge on the Hh signaling cascade that influence its activity. Moreover, we highlight several connection points between Hh and Notch, Wnt, or TGF-β pathways, showing a reciprocal synergism that contributes to tumorigenesis, supporting a more malignant behavior by tumor cells, such as in leukemia and brain tumors. Understanding the importance of these molecular interlinking networks will provide a rational basis for combined anticancer drug development.
Collapse
Affiliation(s)
- Maria Pelullo
- Center of Life Nano Science Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Sabrina Zema
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| | | | - Saula Checquolo
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, Italy
| | | | - Diana Bellavia
- Department of Molecular Medicine, Sapienza University, Rome, Italy
| |
Collapse
|
219
|
Sun J, Tao R, Mao T, Feng Z, Guo Q, Zhang X. The involvement of lipid raft pathway in suppression of TGFβ-mediated metastasis by tolfenamic acid in hepatocellular carcinoma cells. Toxicol Appl Pharmacol 2019; 380:114696. [PMID: 31381904 DOI: 10.1016/j.taap.2019.114696] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 07/30/2019] [Accepted: 07/31/2019] [Indexed: 01/18/2023]
Abstract
TGFβ signaling plays an important role in orchestrating a favorable microenvironment for tumor cell growth and promoting epithelial-mesenchymal transition. As a conventional nonsteroidal anti-inflammation drugs, tolfenamic acid (TA) has been previously reported to exhibit anti-cancer activity. Herein, we investigated the effect of TA on TGFβ-mediated pro-metastatic activity and the underlying mechanisms in hepatocellular carcinoma (HCC). As a result, TA suppresses TGFβ-induced migration and glycolysis in HCC cells, which is accompanied with reduced Smad phosphorylation and subsequent nuclear transcription activity. Mechanistically, TA promotes lipid raft-caveolar internalization pathway of TGFβ receptor, therefore leading to its rapid turnover. Consistently, TA inhibits constitutively active TGFβ type I receptor induced Smad phosphorylation and EMT markers, whereas ectopic expression of TGFβ type II receptor could partially rescue TGFβ-mediated Smad2 phosphorylation and downstream genes expression in the presence of TA. Furthermore, TA inhibited HCC cells invasion in nude mice, associated with the alteration of characteristics related with EMT and glycolysis of cancer cells. Our study suggests TA could activate lipid raft pathway and modulate TGFβ mediated metastasis, implicating the potential application of TA as a modulator of tumor microenvironment in HCC.
Collapse
Affiliation(s)
- Jingfang Sun
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Ran Tao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Tianxiao Mao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Zhi Feng
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | - Xiaobo Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China.
| |
Collapse
|
220
|
Lee HR, Pyo MC, Chae SA, Hong CO, Lee KW. Inhibitory Effect of Chebulic Acid on Alveolar Epithelial to Mesenchymal Transition in Response to Urban Particulate Matter Using Co-treatment and Post-treatment Exposure. Biol Pharm Bull 2019; 42:1322-1331. [DOI: 10.1248/bpb.b19-00061] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Hee-Ra Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Min Cheol Pyo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Seung A Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Chung-Oui Hong
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University
| |
Collapse
|
221
|
Tang C, Liu T, Wang K, Wang X, Xu S, He D, Zeng J. Transcriptional regulation of FoxM1 by HIF‑1α mediates hypoxia‑induced EMT in prostate cancer. Oncol Rep 2019; 42:1307-1318. [PMID: 31364741 PMCID: PMC6718104 DOI: 10.3892/or.2019.7248] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/26/2019] [Indexed: 01/06/2023] Open
Abstract
Hypoxia is a tumorigenesis-related microenvironment change which usually occurs in the earliest stage of prostate cancer (PCa) development. Accumulating evidence has demonstrated that hypoxia/hypoxia-inducing factor (HIF) is involved in the induction of epithelial-mesenchymal transition (EMT) and increased metastatic potential in PCa. However, the mechanism by which hypoxia/HIF regulates EMT remains unclear. In the present study, we demonstrated the molecular mechanisms of hypoxia-induced EMT in PCa, focusing on HIF-1α/Forkhead box M1 (FoxM1) signaling pathway. PCa PC3 and DU145 cell lines were used as the model system in vitro. Our data revealed that hypoxia induced EMT in PCa cells. Bioinformatics analysis identified the possible association between HIF-1α and FoxM1. Additionally, FoxM1 was significantly associated with PCa development and Gleason scores of PCa. Exposure to hypoxia resulted in the increased expression of HIF-1α and FoxM1. Genetic knockdown FoxM1 abolished hypoxia-induced EMT in PCa, while exogenous overexpression of FoxM1 facilitated hypoxia-induced EMT. Furthermore, the increase of FoxM1 during hypoxia was due to the transcriptional regulation on the FoxM1 promoter by HIF-1α. We also confirmed the binding site of HIF-1α on the FoxM1 promoter by different lengths promoter sequences. These findings provide new insights into how EMT is regulated in PCa under hypoxic stress. It is worthwhile to investigate in future that inhibition of FoxM1 as a potential target may be an effective therapeutic strategy against PCa.
Collapse
Affiliation(s)
- Cong Tang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Tianjie Liu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Ke Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Xinyang Wang
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shan Xu
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Dalin He
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Jin Zeng
- Department of Urology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
222
|
Marzagalli M, Raimondi M, Fontana F, Montagnani Marelli M, Moretti RM, Limonta P. Cellular and molecular biology of cancer stem cells in melanoma: Possible therapeutic implications. Semin Cancer Biol 2019; 59:221-235. [PMID: 31265892 DOI: 10.1016/j.semcancer.2019.06.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 06/27/2019] [Indexed: 01/17/2023]
Abstract
Malignant melanoma is a tumor characterized by a very high level of heterogeneity, responsible for its malignant behavior and ability to escape from standard therapies. In this review we highlight the molecular and biological features of the subpopulation of cancer stem cells (CSCs), well known to be characterized by self-renewal properties, deeply involved in triggering the processes of tumor generation, metastasis, progression and drug resistance. From the molecular point of view, melanoma CSCs are identified and characterized by the expression of stemness markers, such as surface markers, ATP-binding cassette (ABC) transporters, embryonic stem cells and intracellular markers. These cells are endowed with different functional features. In particular, they play pivotal roles in the processes of tumor dissemination, epithelial-to-mesenchymal transition (EMT) and angiogenesis, mediated by specific intracellular signaling pathways; moreover, they are characterized by a unique metabolic reprogramming. As reported for other types of tumors, the CSCs subpopulation in melanoma is also characterized by a low immunogenic profile as well as by the ability to escape the immune system, through the expression of a negative modulation of T cell functions and the secretion of immunosuppressive factors. These biological features allow melanoma CSCs to escape standard treatments, thus being deeply involved in tumor relapse. Targeting the CSCs subpopulation is now considered an attractive treatment strategy; in particular, combination treatments, based on both CSCs-targeting and standard drugs, will likely increase the therapeutic options for melanoma patients. The characterization of CSCs in liquid biopsies from single patients will pave the way towards precision medicine.
Collapse
Affiliation(s)
- Monica Marzagalli
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy
| | - Michela Raimondi
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy
| | - Fabrizio Fontana
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy
| | | | - Roberta M Moretti
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy
| | - Patrizia Limonta
- Department of Pharmacological and Biomolecular Sciences, University of Milano, Milano, Italy.
| |
Collapse
|
223
|
Watanabe K, Panchy N, Noguchi S, Suzuki H, Hong T. Combinatorial perturbation analysis reveals divergent regulations of mesenchymal genes during epithelial-to-mesenchymal transition. NPJ Syst Biol Appl 2019; 5:21. [PMID: 31275609 PMCID: PMC6570767 DOI: 10.1038/s41540-019-0097-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/28/2019] [Indexed: 12/14/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT), a fundamental transdifferentiation process in development, produces diverse phenotypes in different physiological or pathological conditions. Many genes involved in EMT have been identified to date, but mechanisms contributing to the phenotypic diversity and those governing the coupling between the dynamics of epithelial (E) genes and that of the mesenchymal (M) genes are unclear. In this study, we employed combinatorial perturbations to mammary epithelial cells to induce a series of EMT phenotypes by manipulating two essential EMT-inducing elements, namely TGF-β and ZEB1. By measuring transcriptional changes in more than 700 E-genes and M-genes, we discovered that the M-genes exhibit a significant diversity in their dependency to these regulatory elements and identified three groups of M-genes that are controlled by different regulatory circuits. Notably, functional differences were detected among the M-gene clusters in motility regulation and in survival of breast cancer patients. We computationally predicted and experimentally confirmed that the reciprocity and reversibility of EMT are jointly regulated by ZEB1. Our integrative analysis reveals the key roles of ZEB1 in coordinating the dynamics of a large number of genes during EMT, and it provides new insights into the mechanisms for the diversity of EMT phenotypes.
Collapse
Affiliation(s)
- Kazuhide Watanabe
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Nicholas Panchy
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, Knoxville, TN 37996 USA
- National Institute for Mathematical and Biological Synthesis, Knoxville, TN 37996 USA
| | - Shuhei Noguchi
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Harukazu Suzuki
- RIKEN Center for Integrative Medical Sciences, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, The University of Tennessee, Knoxville, Knoxville, TN 37996 USA
- National Institute for Mathematical and Biological Synthesis, Knoxville, TN 37996 USA
| |
Collapse
|
224
|
Mota STS, Vecchi L, Zóia MAP, Oliveira FM, Alves DA, Dornelas BC, Bezerra SM, Andrade VP, Maia YCP, Neves AF, Goulart LR, Araújo TG. New Insights into the Role of Polybromo-1 in Prostate Cancer. Int J Mol Sci 2019; 20:ijms20122852. [PMID: 31212728 PMCID: PMC6627401 DOI: 10.3390/ijms20122852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/29/2019] [Accepted: 05/30/2019] [Indexed: 01/22/2023] Open
Abstract
The human protein Polybromo-1 (PBMR1/BAF180) is a component of the SWI/SNF chromatin-remodeling complex that has been reported to be deregulated in tumors. However, its role in prostate cancer (PCa) is largely unknown. In this study, we described the PBRM1 transcriptional levels and the protein expression/localization in tissues of PCa patients and in prostatic cell lines. Increased PBRM1 mRNA levels were found in PCa samples, when compared to benign disease, and were correlated with higher Gleason score. We also verified that only the nuclear localization of PBRM1 protein is correlated with a more aggressive disease and high Prostate-Specific Antigen (PSA) levels in tissue microarrays. Intriguing expression patterns of mRNA and protein were identified in the cell lines. Although PBRM1 protein was restricted to the nuclei, in tumor cell lines in non-neoplastic cells, it was also present in vesicular-like structures that were dispersed within the cytoplasm. We knocked-down PBRM1 in the castration-resistant PCa (CRPC) cell line PC-3 and we verified that PBRM1 promotes the expression of several markers of aggressiveness, including EpCAM, TGF-β, and N-Cadherin. Therefore, our data supported the hypothesis that PBRM1 displays a pivotal role in the promotion and maintenance of the malignant behavior of PCa, especially in CRPC.
Collapse
Affiliation(s)
- Sara T S Mota
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Lara Vecchi
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Mariana A P Zóia
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Fabrícia M Oliveira
- Faculty of Mathematics, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
| | - Douglas A Alves
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Bruno C Dornelas
- Pathology Division, Internal Medicine, University Hospital, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | | | | | - Yara C P Maia
- Medical Faculty, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| | - Adriana F Neves
- Laboratory of Molecular Biology, Federal University of Goias-GO, Goiânia-GO 75704-020, Brazil.
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
- University of California Davis, Department of Medical Microbiology and Immunology, Davis, CA 95616, USA.
| | - Thaise G Araújo
- Laboratory of Genetics and Biotechnology, Institute of Biotechnology, Federal University of Uberlandia, Patos de Minas-MG 387400-128, Brazil.
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlandia, Uberlandia-MG 38400-902, Brazil.
| |
Collapse
|
225
|
Liu R, Liu Q, Pan Z, Liu X, Ding J. Cell Type and Nuclear Size Dependence of the Nuclear Deformation of Cells on a Micropillar Array. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2019; 35:7469-7477. [PMID: 30226387 DOI: 10.1021/acs.langmuir.8b02510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
While various cellular responses to materials have been published, little concerns the deformation of cell nuclei. Herein we fabricated a polymeric micropillar array of appropriate dimensions to trigger the significant self-deformation of cell nuclei and examined six cell types, which could be classified into cancerous cells (Hela and HepG2) versus healthy cells (HCvEpC, MC3T3-E1, NIH3T3, and hMSC) or epithelial-like cells (Hela, HepG2, and HCvEpC) versus fibroblast-like cells (MC3T3-E1, NIH3T3, and hMSC). While all of the cell types exhibited severe nuclear deformation on the poly(lactide- co-glycolide) (PLGA) micropillar array, the difference between the epithelial-like and fibroblast-like cells was much more significant than that between the cancerous and healthy cells. We also examined the statistics of nuclear shape indexes of cells with an inevitable dispersity of nuclear sizes. It was found that larger nuclei favored more significant deformation on the micropillar array for each cell type. In the same region of nuclear size, the parts of the epithelial-like cells exhibited more significant nuclear deformation than those of the fibroblast-like cells. Hence, this article reports the nuclear size dependence of the self-deformation of cell nuclei on micropillar arrays for the first time and meanwhile strengthens the cell-type dependence.
Collapse
Affiliation(s)
- Ruili Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Qiong Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Zhen Pan
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Xiangnan Liu
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| | - Jiandong Ding
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science , Fudan University , Shanghai 200438 , China
| |
Collapse
|
226
|
MEK Inhibition Targets Cancer Stem Cells and Impedes Migration of Pancreatic Cancer Cells In Vitro and In Vivo. Stem Cells Int 2019; 2019:8475389. [PMID: 31281387 PMCID: PMC6589314 DOI: 10.1155/2019/8475389] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 03/10/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains a devastating disease with a very poor prognosis. At the same time, its incidence is on the rise, and PDAC is expected to become the second leading cause of cancer-related death by 2030. Despite extensive work on new therapeutic approaches, the median overall survival is only 6-12 months after diagnosis and the 5-year survival is less than 7%. While pancreatic cancer is particularly difficult to treat, patients usually succumb not to the growth of the primary tumor, but to extensive metastasis; therefore, strategies to reduce the migratory and metastatic capacity of pancreatic cancer cells merit close attention. The vast majority of pancreatic cancers harbor RAS mutations. The outstanding relevance of the RAS/MEK/ERK pathway in pancreatic cancer biology has been extensively shown previously. Due to their high dependency on Ras mutations, pancreatic cancers might be particularly sensitive to inhibitors acting downstream of Ras. Herein, we use a genetically engineered mouse model of pancreatic cancer and primary pancreatic cancer cells were derived from this model to demonstrate that small-molecule MEK inhibitors functionally abrogate cancer stem cell populations as demonstrated by reduced sphere and organoid formation capacity. Furthermore, we demonstrate that MEK inhibition suppresses TGFβ-induced epithelial-to-mesenchymal transition and migration in vitro and ultimately results in a highly significant reduction in circulating tumor cells in mice.
Collapse
|
227
|
Saad ESA, Lam JSY, Al-Khan AA, Tayebi M, Day MJ, Richardson SJ, Danks JA. A Comparative Review of Mixed Mammary Tumors in Mammals. J Mammary Gland Biol Neoplasia 2019; 24:125-137. [PMID: 30488318 DOI: 10.1007/s10911-018-9422-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 11/21/2018] [Indexed: 11/30/2022] Open
Abstract
Mixed tumors are characterized by the histological identification of two or more cell types. Commonly, a mixture of epithelial and myoepithelial cells is included in abundant stroma, which can consist of myxoid, chondroid or bony matrices. Spontaneously arising mixed tumors are rare lesions in the human breast but are common in human salivary glands and canine mammary glands. Subtle histopathological characteristics and overlapping attributes of malignant lesions with other benign lesions can lead to a diagnostic challenge. Mixed tumors can present as benign or malignant. While malignant mixed tumors are quite rare in the human breast they have a poor prognosis. Benign mixed mammary tumors occur more frequently in female dogs than in humans and are usually associated with a good prognosis. This review will provide a comprehensive overview of mixed mammary tumors, across various mammalian species.
Collapse
Affiliation(s)
- Eman S A Saad
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Jacqueline S Y Lam
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
- Laverty Pathology, Macquarie Park, Sydney, Australia
| | - Awf A Al-Khan
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, New South Wales, Australia
| | - Michael J Day
- School of Veterinary and Life Sciences, Murdoch University, Western Australia, Australia
| | - Samantha J Richardson
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Janine A Danks
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia.
- Department of Medicine, Austin Health, The University of Melbourne, Heidelberg, Victoria, Australia.
| |
Collapse
|
228
|
Li X, Chen S, Yan L, Wang J, Pei M. Prospective application of stem cells to prevent post-operative skeletal fibrosis. J Orthop Res 2019; 37:1236-1245. [PMID: 30835890 PMCID: PMC9202416 DOI: 10.1002/jor.24266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Accepted: 02/17/2019] [Indexed: 02/04/2023]
Abstract
Post-operative skeletal fibrosis is considered one of the major complications causing dysfunction of the skeletal system and compromising the outcomes of clinical treatment. Limited success has been achieved using current therapies; more effective therapies to reduce post-operative skeletal fibrosis are needed. Stem cells possess the ability to repair and regenerate damaged tissue. Numerous studies show that stem cells serve as a promising therapeutic approach for fibrotic diseases in tissues other than the skeletal system by inhibiting the inflammatory response and secreting favorable cytokines through activating specific signaling pathways, acting as so-called medicinal signaling cells. In this review, current therapies are summarized for post-operative skeletal fibrosis. Given that stem cells are used as a promising therapeutic approach for fibrotic diseases, little effort has been undertaken to use stem cells to prevent post-operative skeletal fibrosis. This review aims at providing useful information for the potential application of stem cells in preventing post-operative skeletal fibrosis in the near future. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1236-1245, 2019.
Collapse
Affiliation(s)
- Xiaolei Li
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China,Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Song Chen
- Department of Orthopaedics, Chengdu Military General Hospital, Chengdu, Sichuan, 610083, China
| | - Lianqi Yan
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China
| | - Jingcheng Wang
- Department of Orthopaedics, Orthopaedics Institute, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou, Jiangsu, 225001, China,Co-Corresponding author: Jingcheng Wang, MD, Department of Orthopaedics, Subei People’s Hospital, 98 West Nantong Road, Yangzhou 225001, China;
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA,Exercise Physiology, West Virginia University, Morgantown, WV, 26506, USA,WVU Cancer Institute, Robert C. Byrd Health Sciences Center, West Virginia University, Morgantown, WV, 26506, USA,Corresponding author: Ming Pei MD, PhD, Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, PO Box 9196, 64 Medical Center Drive, Morgantown, WV 26506-9196, USA, Telephone: 304-293-1072; Fax: 304-293-7070;
| |
Collapse
|
229
|
Dai B, Zhou G, Hu Z, Zhu G, Mao B, Su H, Jia Q. MiR-205 suppresses epithelial-mesenchymal transition and inhibits tumor growth of human glioma through down-regulation of HOXD9. Biosci Rep 2019; 39:BSR20181989. [PMID: 30992394 PMCID: PMC6522733 DOI: 10.1042/bsr20181989] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 03/19/2019] [Accepted: 04/01/2019] [Indexed: 12/02/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) plays a pivotal role in cancer progression. Hsa-miR-205 is considered one of the fundamental regulators of EMT. In the present study, we found that miR-205 was down-regulated in glioma tissues and human glioma cells U87 and U251. Meanwhile, miR-205 overexpression enhanced E-cadherin, reduced mesenchymal markers, and decreased cell proliferation, migration, and invasion in vitro. In vivo, miR-205 suppressed tumor growth. Additionally, HOXD9 was confirmed as a direct target of miR-205. Suppression of HOXD9 by miR-205 was demonstrated by luciferase reporter assay, quantitative real time-PCR analysis, and western blot. Moreover, we observed a negative correlation between miR-205 and HOXD9 in human glioma tissues. In summary, our findings demonstrated that miR-205 suppresses glioma tumor growth, invasion, and reverses EMT through down-regulating its target HOXD9.
Collapse
Affiliation(s)
- Bin Dai
- Department of Neurosurgery, Beijing Shijitan Hospital, Capital Medical University, No.10 Tieyi Rd, Yangfangdian, Haidian District, Beijing, 100038, P.R. China
| | - Guanghua Zhou
- Department of Neurosurgery, Liaocheng People's Hospital of Shandong, No.67 Dongchang West Road, Liaocheng, Shandong Province, 252000, P.R. China
| | - Zhiqiang Hu
- Department of Neurosurgery, Beijing Shijitan Hospital, Capital Medical University, No.10 Tieyi Rd, Yangfangdian, Haidian District, Beijing, 100038, P.R. China
| | - Guangtong Zhu
- Department of Neurosurgery, Beijing Shijitan Hospital, Capital Medical University, No.10 Tieyi Rd, Yangfangdian, Haidian District, Beijing, 100038, P.R. China
| | - Beibei Mao
- Department of Neurosurgery, Beijing Shijitan Hospital, Capital Medical University, No.10 Tieyi Rd, Yangfangdian, Haidian District, Beijing, 100038, P.R. China
| | - Haiyang Su
- Department of Neurosurgery, Beijing Shijitan Hospital, Capital Medical University, No.10 Tieyi Rd, Yangfangdian, Haidian District, Beijing, 100038, P.R. China
| | - Qingbin Jia
- Department of Neurosurgery, Liaocheng People's Hospital of Shandong, No.67 Dongchang West Road, Liaocheng, Shandong Province, 252000, P.R. China
| |
Collapse
|
230
|
Kajimura T, Sato S, Murakami A, Hayashi-Okada M, Nakashima K, Sueoka K, Sugino N. Overexpression of carbonyl reductase 1 inhibits malignant behaviors and epithelial mesenchymal transition by suppressing TGF-β signaling in uterine leiomyosarcoma cells. Oncol Lett 2019; 18:1503-1512. [PMID: 31423217 PMCID: PMC6607169 DOI: 10.3892/ol.2019.10429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/22/2019] [Indexed: 12/31/2022] Open
Abstract
Carbonyl reductase 1 (CBR1) has been reported to be involved in cancer progression. Recently, we found that CBR1 overexpression inhibited malignant behaviors and the epithelial mesenchymal transition (EMT) in uterine cervical cancer. It remained unclear whether this was also the case in uterine leiomyosarcoma (uLMS), which is derived from mesenchymal cells and is a much more malignant gynecological tumor. A number of previous studies suggested that malignant behaviors are associated with EMT, even in mesenchymal malignant tumors. In the present study, we investigated whether CBR1 inhibits malignant behaviors and EMT in uLMS. We established clones of uLMS cells (SKN cells) and uterine sarcoma cells (MES-SA cells) that overexpressed CBR1. Cell proliferative, migratory and invasive activities were suppressed by CBR1 overexpression, accompanied by increases in the expressions of epithelial markers (E-cadherin and cytokeratin) and decreases in the expressions of mesenchymal markers (N-cadherin and fibronectin), suggesting that CBR1 overexpression inhibits malignant behaviors and EMT in uLMS cells. In addition, transforming growth factor-β (TGF-β) production and the subsequent signaling and phosphorylation of Smad were suppressed in the clones. To investigate the association between TGF-β and EMT, SKN cells were treated with TGF-β or a TGF-β receptor blocker (SB431542). EMT was promoted by TGF-β and inhibited by SB431542. In conclusion, this is the first study, to the best of the authors' knowledge, showing that CBR1 overexpression inhibits malignant behaviors and EMT in uLMS cells. The present study provided novel insight demonstrating that the suppressive effect of CBR1 is mediated through TGF-β signaling.
Collapse
Affiliation(s)
- Takuya Kajimura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Akihiro Murakami
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Maki Hayashi-Okada
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kengo Nakashima
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kotaro Sueoka
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
231
|
Jiang Y, Wells A, Sylakowski K, Clark AM, Ma B. Adult Stem Cell Functioning in the Tumor Micro-Environment. Int J Mol Sci 2019; 20:ijms20102566. [PMID: 31130595 PMCID: PMC6566759 DOI: 10.3390/ijms20102566] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/18/2019] [Accepted: 05/23/2019] [Indexed: 12/14/2022] Open
Abstract
Tumor progression from an expanded cell population in a primary location to disseminated lethal growths subverts attempts at cures. It has become evident that these steps are driven in a large part by cancer cell-extrinsic signaling from the tumor microenvironment (TME), one cellular component of which is becoming more appreciated for potential modulation of the cancer cells directly and the TME globally. That cell is a heterogenous population referred to as adult mesenchymal stem cells/multipotent stromal cells (MSCs). Herein, we review emerging evidence as to how these cells, both from distant sources, mainly the bone marrow, or local resident cells, can impact the progression of solid tumors. These nascent investigations raise more questions than they answer but paint a picture of an orchestrated web of signals and interactions that can be modulated to impact tumor progression.
Collapse
Affiliation(s)
- Yuhan Jiang
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- Department of Computational & Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA 15213, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Kyle Sylakowski
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Amanda M Clark
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| | - Bo Ma
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
- VA Pittsburgh Healthcare System, Pittsburgh, PA 15213, USA.
| |
Collapse
|
232
|
Cabezas F, Farfán P, Marzolo MP. Participation of the SMAD2/3 signalling pathway in the down regulation of megalin/LRP2 by transforming growth factor beta (TGF-ß1). PLoS One 2019; 14:e0213127. [PMID: 31120873 PMCID: PMC6532859 DOI: 10.1371/journal.pone.0213127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 05/08/2019] [Indexed: 12/19/2022] Open
Abstract
Megalin/LRP2 is a receptor that plays important roles in the physiology of several organs, such as kidney, lung, intestine, and gallbladder and also in the physiology of the nervous system. Megalin expression is reduced in diseases associated with fibrosis, including diabetic nephropathy, hepatic fibrosis and cholelithiasis, as well as in some breast and prostate cancers. One of the hallmarks of these conditions is the presence of the cytokine transforming growth factor beta (TGF-ß). Although TGF-ß has been implicated in the reduction of megalin levels, the molecular mechanism underlying this regulation is not well understood. Here, we show that treatment of two epithelial cell lines (from kidney and gallbladder) with TGF-ß1 is associated with decreased megalin mRNA and protein levels, and that these effects are reversed by inhibiting the TGF-ß1 type I receptor (TGF-ßRI). Based on in silico analyses, the two SMAD-binding elements (SBEs) in the megalin promoter are located at positions -57 and -605. Site-directed mutagenesis of the SBEs and chromatin immunoprecipitation (ChIP) experiments revealed that SMAD2/3 transcription factors interact with SBEs. Both the presence of SMAD2/3 and intact SBEs were associated with repression of the megalin promoter, in the absence as well in the presence of TGF-ß1. Also, reduced megalin expression and promoter activation triggered by high concentration of albumin are dependent on the expression of SMAD2/3. Interestingly, the histone deacetylase inhibitor Trichostatin A (TSA), which induces megalin expression, reduced the effects of TGF-ß1 on megalin mRNA levels. These data show the significance of TGF-ß and the SMAD2/3 signalling pathway in the regulation of megalin and explain the decreased megalin levels observed under conditions in which TGF-ß is upregulated, including fibrosis-associated diseases and cancer.
Collapse
Affiliation(s)
- Felipe Cabezas
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pamela Farfán
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María-Paz Marzolo
- Laboratorio de Tráfico Intracelular y Señalización, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
233
|
Amerizadeh F, Bahrami A, Khazaei M, Hesari A, Rezayi M, Talebian S, Maftouh M, Moetamani-Ahmadi M, Seifi S, Shahidsales S, Joudi-Mashhad M, Ferns GA, Ghasemi F, Avan A. Current status and future prospects of transforming growth factor-β as a potential prognostic and therapeutic target in the treatment of breast cancer. J Cell Biochem 2019; 120:6962-6971. [PMID: 30672016 DOI: 10.1002/jcb.27831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/14/2018] [Indexed: 01/24/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway is one of the important pathways involved in the cancer cell proliferation, invasion, migration, angiogenesis, apoptosis, as well as in metastasis by agitation or invasion of metastasis-related factors, including matrix metalloproteinase (MMP), epithelial-to-mesenchymal transition (EMT), tumor microenvironment (TME), cancer stem cells (CSCs), and cell adhesion molecules (CAMs). These data suggest its potential value as a therapeutic object in the treatment of malignancies including breast cancer. Several pharmacological approaches have been established to suppress TGF-β pathway; such as vaccines, small molecular inhibitors, antisense oligonucleotides, and monoclonal antibodies. Some of these are now approved by the US Food and Drug Administration for targeting the TGF-β signaling pathway. This study attempts to summarize the current data about the functions of TGF-β in cancer cells, and their probable application in the cancer therapy with a specific emphasis on recent preclinical and clinical research in the treatment of breast cancer and its prognostic value.
Collapse
Affiliation(s)
- Forouzan Amerizadeh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsane Bahrami
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - AmirReza Hesari
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Rezayi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sahar Talebian
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mona Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Sima Seifi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mona Joudi-Mashhad
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Brighton, UK
| | - Faezeh Ghasemi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
234
|
Long Noncoding RNA MALAT1 Acts as a Competing Endogenous RNA to Regulate TGF- β2 Induced Epithelial-Mesenchymal Transition of Lens Epithelial Cells by a MicroRNA-26a-Dependent Mechanism. BIOMED RESEARCH INTERNATIONAL 2019; 2019:1569638. [PMID: 31143769 PMCID: PMC6501259 DOI: 10.1155/2019/1569638] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/31/2019] [Accepted: 04/08/2019] [Indexed: 02/07/2023]
Abstract
The aim of the present study was to characterize whether the long noncoding RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1)/miR-26a/Smad4 axis is involved in epithelial–mesenchymal transition (EMT) of lens epithelial cells (LECs). Primary human LECs were separated and cultured. Microarray analysis showed that a total of 568 lncRNAs are differentially expressed in primary HLECs in the presence of TGF-β2 and MALAT1 is mostly significantly dysregulated lncRNAs, which is increased by nearly 17-fold. In addition, upregulation of MALAT1 and downregulation of miR-26a were detected in human posterior capsule opacification (PCO) attached LECs and the LECs obtained from patients with anterior polar cataracts by quantitative RT-PCR (qRT-PCR). Next, our results showed that TGF-β2 induces overexpression of EMT markers in primary HLECs via a MALAT1-dependent mechanism. The mechanism is that MALAT1 negatively regulates miR-26a and miR-26a directly targets Smad4 by luciferase reporter assays and RNA-binding protein immunoprecipitation assay. In summary, TGF-β2 induces MALAT1 overexpression, which in turn MALAT1 acts as a ceRNA targeting Smad4 by binding miR-26a and promotes the progression of EMT of LECs.
Collapse
|
235
|
Zhang L, Song X, Chen X, Wang Q, Zheng X, Wu C, Jiang J. Circular RNA CircCACTIN Promotes Gastric Cancer Progression by Sponging MiR-331-3p and Regulating TGFBR1 Expression. Int J Biol Sci 2019; 15:1091-1103. [PMID: 31182928 PMCID: PMC6535790 DOI: 10.7150/ijbs.31533] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/27/2019] [Indexed: 01/05/2023] Open
Abstract
Background: Circular RNAs (circRNAs) are a new class of RNAs that play a significant role in regulating gene expression and biological function. However, the expression profile and function of circRNAs in gastric cancer (GC) remain mostly uncertain. In the present study, we researched the expression profile of circRNAs in human GC tissues and explored the role of circCACTIN (hsa_circ_0092303). Methods: Circular RNA microarray assays were performed to detect circular RNA expression profiles of GC and circCACTIN was identified for further investigation. Quantitative real-time PCR was used to detect the expression of circCACTIN, miR-331-3p and TGFBR1 in GC specimens and cell lines. CircCACTIN was stably silenced and overexpressed in GC cells, and cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), as well as tumorigenesis in nude mice were performed to assess the effect of circCACTIN on GC. Results: CircCACTIN expression was obviously up-regulated in GC tissues and cell lines. Knockdown of circCACTIN inhibited GC cells proliferation, migration, invasion and EMT. Enforced-expression of circCACTIN promoted GC cells migration, invasion and EMT, but had no effect on GC cells proliferation. Moreover, in vivo experiments, circCACTIN up-regulation promoted GC tumor growth and EMT, and circCACTIN down-regulation inhibited GC tumor growth and EMT. Binding interactions were detected between circCACTIN and miR-331-3p, and between miR-331-3p and TGFBR1 by Dual-luciferase reporter assays. Mechanistically, we demonstrated that circCACTIN promoted gastric cancer progression by sponging miRNA-331-3p and regulating TGFBR1 mRNA expression. Conclusion: The circCACTIN/miR-331-3p/TGFBR1 axis affected the proliferation, migration, invasion and EMT of GC through the mechanism of competing endogenous RNAs (ceRNA). Furthermore, our results identified circCACTIN as a novel oncogenic circRNA in GC.
Collapse
Affiliation(s)
- Luo Zhang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xing Song
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xin Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Qi Wang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, China
| |
Collapse
|
236
|
Lien HC, Lee YH, Juang YL, Lu YT. Fibrillin-1, a novel TGF-beta-induced factor, is preferentially expressed in metaplastic carcinoma with spindle sarcomatous metaplasia. Pathology 2019; 51:375-383. [PMID: 31010590 DOI: 10.1016/j.pathol.2019.02.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/18/2019] [Accepted: 02/03/2019] [Indexed: 11/28/2022]
Abstract
TGF-β induces epithelial-mesenchymal transition (EMT), which is involved in tumour progression. This study aims to identify and characterise novel factors potentially related to TGF-β-mediated tumour aggression in breast cancer. We treated the human mammary epithelial cell line MCF10A with TGF-β and observed TGF-β-dependent upregulation of FBN1, involving demethylation of CpG sites, in MCF10A cells undergoing EMT. The biological importance of fibrillin-1, encoded by FBN1, was evaluated through immunohistochemistry on 225 breast cancer specimens of various subtypes. Fibrillin-1 expression was observed only in metaplastic carcinoma of the breast (MCB) (51.7%), and the expression was observed in spindle sarcomatous metaplasia (SSM), but not in other metaplasia, including matrix-producing, pleomorphic, and squamous metaplasia, and carcinomatous components of both MCB and non-MCB. Fibrillin-1 expression was also restricted to the SSM of non-mammary carcinosarcomas of various organs. Overall, fibrillin-1 expression was enriched in MCB and non-mammary carcinosarcoma with SSM (93.7% and 93.3%, respectively), but not in MCBs and non-mammary carcinosarcoma without SSM. FBN1 knockdown in MDA-MB-231 cells with high FBN1 expression did not compromise migration, invasion, and tumourigenesis, and did not alter the expression of other EMT-related markers. In conclusion, fibrillin-1 is a novel TGF-β-induced marker. Fibrillin-1 expression in SSM, but not in other metaplasia and carcinomatous components, in both MCBs and non-mammary carcinosarcomas, together with the inability of FBN1-knockdown to compromise migration and invasion, indicates that fibrillin-1 is a marker induced solely in spindle metaplasia during EMT and does not induce EMT nor lead to tumour aggressiveness.
Collapse
Affiliation(s)
- Huang-Chun Lien
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan; Department of Pathology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan.
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital, National Taiwan University, Taipei, Taiwan
| | - Yu-Lin Juang
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| | - Yueh-Tong Lu
- Graduate Institute of Pathology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
237
|
The Role of Fibrosis and Liver-Associated Fibroblasts in the Pathogenesis of Hepatocellular Carcinoma. Int J Mol Sci 2019. [PMID: 30959975 DOI: 10.3390/ijms20071723.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive types of cancer and lacks effective therapeutic approaches. Most HCC develops in the setting of chronic liver injury, hepatic inflammation, and fibrosis. Hepatic stellate cells (HSCs) and cancer-associated fibroblasts (CAFs) are key players in liver fibrogenesis and hepatocarcinogenesis, respectively. CAFs, which probably derive from HSCs, activate into extracellular matrix (ECM)-producing myofibroblasts and crosstalk with cancer cells to affect tumor growth and invasion. In this review, we describe the different components which form the HCC premalignant microenvironment (PME) and the tumor microenvironment (TME), focusing on the liver fibrosis process and the biology of CAFs. We will describe the CAF-dependent mechanisms which have been suggested to promote hepatocarcinogenesis, such as the alteration of ECM, CAF-dependent production of cytokines and angiogenic factors, CAF-dependent reduction of immuno-surveillance, and CAF-dependent promotion of epithelial-mesenchymal transition (EMT). New knowledge of the fibrosis process and the role of CAFs in HCC may pave the way for new therapeutic strategies for liver cancer.
Collapse
|
238
|
Baglieri J, Brenner DA, Kisseleva T. The Role of Fibrosis and Liver-Associated Fibroblasts in the Pathogenesis of Hepatocellular Carcinoma. Int J Mol Sci 2019; 20:ijms20071723. [PMID: 30959975 PMCID: PMC6479943 DOI: 10.3390/ijms20071723] [Citation(s) in RCA: 218] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 03/29/2019] [Accepted: 04/05/2019] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most aggressive types of cancer and lacks effective therapeutic approaches. Most HCC develops in the setting of chronic liver injury, hepatic inflammation, and fibrosis. Hepatic stellate cells (HSCs) and cancer-associated fibroblasts (CAFs) are key players in liver fibrogenesis and hepatocarcinogenesis, respectively. CAFs, which probably derive from HSCs, activate into extracellular matrix (ECM)-producing myofibroblasts and crosstalk with cancer cells to affect tumor growth and invasion. In this review, we describe the different components which form the HCC premalignant microenvironment (PME) and the tumor microenvironment (TME), focusing on the liver fibrosis process and the biology of CAFs. We will describe the CAF-dependent mechanisms which have been suggested to promote hepatocarcinogenesis, such as the alteration of ECM, CAF-dependent production of cytokines and angiogenic factors, CAF-dependent reduction of immuno-surveillance, and CAF-dependent promotion of epithelial-mesenchymal transition (EMT). New knowledge of the fibrosis process and the role of CAFs in HCC may pave the way for new therapeutic strategies for liver cancer.
Collapse
Affiliation(s)
- Jacopo Baglieri
- Department of Medicine, UC San Diego, La Jolla, CA 92093, USA.
| | - David A Brenner
- Department of Medicine, UC San Diego, La Jolla, CA 92093, USA.
| | | |
Collapse
|
239
|
Zhang Y, Jiang X, Ren L. Optimization of the adipose-derived mesenchymal stem cell delivery time for radiation-induced lung fibrosis treatment in rats. Sci Rep 2019; 9:5589. [PMID: 30944348 PMCID: PMC6447528 DOI: 10.1038/s41598-019-41576-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/11/2019] [Indexed: 12/14/2022] Open
Abstract
The present study attempts to identify the optimal time duration for the administration of Ad-MSCs, in order to maximize its therapeutic benefits, and compare the degree of fibrosis among three different administration time points using the RILF rat model system. Ad-MSCs were delivered to Sprague-Dawley rats through the tail vein at the following different time points after thorax irradiation: two hours, seven days, and two hours + seven days. Post Ad-MSCs transplantation and the histopathological analysis of the lungs were performed along with analysis of inflammatory cytokine levels, including interleukin (IL)-1, IL-2, IL-6, IL-10 and tumor necrosis factor-α (TNF-α). In particular, pro-fibrotic factors (TGF-β1 and α-SMA) were also evaluated in serum and lung tissues. In addition, it was also determined whether Ad-MSCs had any role in inhibiting the transition of type II alveolar epithelial cells into fibroblasts in the lungs of injured rats. The present results demonstrated that the intravenous delivery of Ad-MSCs twice at the 2-hour and 7-day (R + MSC2h+7d group) was effective in reducing lung fibrosis for long term durations, when compared with single delivery either at the two-hour or 7-day time points. In addition, a marked anti-inflammatory effect was also observed in RILF rats in the R + MSC2h+7d group, as indicated by the reduced serum levels of pro-inflammatory cytokines (TNF-α, IL-1 and IL-6) and increased levels of anti-inflammatory cytokines IL-10 and IL-2. Rats that were delivered twice with Ad-MSCs (R + MSC2h+7d group) exhibited significantly reduced TGF-β1 and α-SMA levels, in contrast to rats in the R + MSC7d or R + MSC2h groups, after four weeks. Furthermore, it was also noted that after four weeks, Ad-MSCs increased the number of lung epithelial cells (SP-C) and inhibited the lung fibroblastic cells (α-SMA) of rats in the R + MSC2h and R + MSC2h+7d groups. The present study concluded that two injections of Ad-MSCs (R + MSC2h+7d group) appear to be optimal for therapeutic efficacy and safety during RILF.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Experimental Pharmacology and Toxicology, Pharmaceutical Science of Jilin University, Changchun, Jilin, 130021, China.,Department of Vascular Surgery, The first Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xinping Jiang
- Department of Oncological Radiotherapy, The First Hospital of Jilin University, Changchun, 130021, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, Pharmaceutical Science of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
240
|
Zhao J, Feng M, Liu D, Liu H, Shi M, Zhang J, Qu J. Antagonism between HTRA3 and TGFβ1 Contributes to Metastasis in Non-Small Cell Lung Cancer. Cancer Res 2019; 79:2853-2864. [PMID: 30940659 DOI: 10.1158/0008-5472.can-18-2507] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/02/2019] [Accepted: 03/29/2019] [Indexed: 11/16/2022]
Abstract
High temperature requirement A3 (HTRA3, long and short isoforms) is a member of the HtrA family and has been implicated as a tumor suppressor in cancer progression in multiple cancer types, yet its molecular functions in non-small cell lung cancer (NSCLC) are not well understood. Here, we report that decreased levels of HTRA3 negatively correlate with elevated TGFβ1 in lung tumor tissue with metastasis. Furthermore, high expression of HTRA3 indicated better prognosis independent of TGFβ1 expression. In NSCLC cell lines, exogenous TGFβ1 significantly downregulated the level of HTRA3, especially the long isoform, during induction of epithelial-mesenchymal transition (EMT). Mechanistically, c-Jun, which is elevated by TGFβ1, directly bound the promoter of HTRA3-L and inhibited its transcription. As a negative feedback loop, overexpression of HTRA3-L attenuated TGFβ1-mediated invasion-metastasis cascades via activation of SMAD2/3 and sensitized cells to anti-PD-L1 treatment. Taken together, our findings suggest that in the early stages of cancer, overexpressed HTRA3 acts as a brake on the oncogenic effects of TGFβ1 and inhibits tumor metastasis. In later stages, the role of HTRA3 is weakened and TGFβ1 efficiently promotes EMT in the absence of the HTRA3 brake. SIGNIFICANCE: This study provides new mechanistic insight of the interaction between HTRA3 and TGFβ in lung cancer by illustrating that HTRA3 is a novel mediator acting as a suppressor of TGFβ1-related oncogenic effects.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/metabolism
- Adenocarcinoma/secondary
- Adult
- Aged
- Aged, 80 and over
- Animals
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/metabolism
- Carcinoma, Non-Small-Cell Lung/secondary
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/secondary
- Cell Movement
- Cell Proliferation
- Epithelial-Mesenchymal Transition
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Lung Neoplasms/genetics
- Lung Neoplasms/metabolism
- Lung Neoplasms/pathology
- Male
- Mice
- Mice, Inbred BALB C
- Middle Aged
- Prognosis
- Serine Endopeptidases/genetics
- Serine Endopeptidases/metabolism
- Smad2 Protein/genetics
- Smad2 Protein/metabolism
- Survival Rate
- Transforming Growth Factor beta1/genetics
- Transforming Growth Factor beta1/metabolism
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Jingya Zhao
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Mingxiang Feng
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dong Liu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Haixia Liu
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Mengmeng Shi
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jing Zhang
- Department of Pulmonary Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jieming Qu
- Department of Pulmonary and Critical Care Medicine, Institute of Respiratory Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
- Department of Pulmonary Medicine, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
241
|
Zhao Y, Liu J, Chen F, Feng XH. C-terminal domain small phosphatase-like 2 promotes epithelial-to-mesenchymal transition via Snail dephosphorylation and stabilization. Open Biol 2019; 8:rsob.170274. [PMID: 29618518 PMCID: PMC5936716 DOI: 10.1098/rsob.170274] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 03/12/2018] [Indexed: 12/16/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a cellular reprogramming process converting epithelial cells into mesenchymal cell morphology. Snail is a critical regulator of EMT by both suppressing epithelial gene expression and promoting mesenchymal gene expression. Expression and activity of Snail are tightly controlled at transcriptional and post-translational levels. It has previously been reported that Snail undergoes phosphorylation and ubiquitin-dependent proteasome degradation. Here, we report nuclear phosphatase SCP4/CTDSPL2 acts as a novel Snail phosphatase. SCP4 physically interacts with and directly dephosphorylates Snail. SCP4-mediated dephosphorylation of Snail suppresses the ubiquitin-dependent proteasome degradation of Snail and consequently enhances TGFβ-induced EMT. The knockdown of SCP4 in MCF10A mammary epithelial cells leads to attenuated cell migration. Collectively, our finding demonstrates that SCP4 plays a critical role in EMT through Snail dephosphorylation and stabilization.
Collapse
Affiliation(s)
- Yulan Zhao
- Life Sciences Institute, and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Jinquan Liu
- Life Sciences Institute, and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Fenfang Chen
- Life Sciences Institute, and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China
| | - Xin-Hua Feng
- Life Sciences Institute, and Innovation Center for Cell Signaling Network, Zhejiang University, Hangzhou, Zhejiang 310058, People's Republic of China .,Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA.,Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
242
|
Zhang L, Wang X, Lai C, Zhang H, Lai M. PMEPA1 induces EMT via a non-canonical TGF-β signalling in colorectal cancer. J Cell Mol Med 2019; 23:3603-3615. [PMID: 30887697 PMCID: PMC6484414 DOI: 10.1111/jcmm.14261] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 12/27/2018] [Accepted: 02/05/2019] [Indexed: 12/21/2022] Open
Abstract
Prostate transmembrane protein androgen induced 1 (PMEPA1) has been reported to promote cancer progression. Metastasis is the main factor leading to cancer progression and poor prognosis, and at the beginning of metastasis, epithelial‐to‐mesenchymal transition (EMT) is a crucial activation. However, the relationship between PMEPA1 and EMT in colorectal cancer metastasis is still poorly understood. In this study, we first testified that PMEPA1 expresses higher in tumour than normal tissue in Gene Expression Omnibus database, in the Cancer Genome Atlas (TCGA) as well as in the clinical data we collected. Moreover, the higher expression was associated with poor prognosis. We furthermore demonstrated PMEPA1 promotes colorectal cancer metastasis and EMT in vivo and in vitro. We found that PMEPA1 activates the bone morphogenetic proteins (BMP) signalling of TGF‐β signalling resulting in promoting EMT and accelerating the proliferation and metastasis of colorectal cancer.
Collapse
Affiliation(s)
- Lei Zhang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xue Wang
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Chong Lai
- Department of Urology, School of Medicine, The First Affiliated Hospital, Zhejiang University, Hangzhou, China
| | - Honghe Zhang
- Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
| | - Maode Lai
- School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Pathology, Key Laboratory of Disease Proteomics of Zhejiang Province, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
243
|
Li J, He Y, Cao Y, Yu Y, Chen X, Gao X, Hu Q. Upregulation of Twist is involved in Gli1 induced migration and invasion of hepatocarcinoma cells. Biol Chem 2019; 399:911-919. [PMID: 29908118 DOI: 10.1515/hsz-2018-0131] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/11/2018] [Indexed: 12/25/2022]
Abstract
Hedgehog (Hh) signaling is involved in the progression of hepatocellular carcinoma (HCC), while its detailed mechanisms are not well illustrated. Our present study revealed that the expression of Gli1, while not Gli2 or Gli3, is significantly increased in HCC cell lines and 20/28 (71.4%) HCC tissues as compared with their corresponding controls. Over expression of Gli1 can promote the migration, invasion and epithelial-mesenchymal transition (EMT) of HCC cells. Gli1 can increase the expression of Twist, while not other EMT transcription factors such as Snail, ZEB1 or Slug. Gli1 increases the transcription of Twist while it has no significant effect on the protein or mRNA stability. Chromatin immunoprecipitation-polymerase chain reaction confirms that Gli1 can directly bind to the promoter of Twist, in which the third binding site is essential for Gli1 induced transcription. Collectively, our data suggest that upregulation of Twist is involved in Gli1 induced migration and invasion of HCC cells.
Collapse
Affiliation(s)
- Juan Li
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuting He
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yuan Cao
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Yan Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaolong Chen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Xiaojuan Gao
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| | - Qiuyue Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China
| |
Collapse
|
244
|
Guo Y, Bao C, Ma D, Cao Y, Li Y, Xie Z, Li S. Network-Based Combinatorial CRISPR-Cas9 Screens Identify Synergistic Modules in Human Cells. ACS Synth Biol 2019; 8:482-490. [PMID: 30762338 DOI: 10.1021/acssynbio.8b00237] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Tumorigenesis is a complex process that is driven by a combination of networks of genes and environmental factors; however, efficient approaches to identifying functional networks that are perturbed by the process of tumorigenesis are lacking. In this study, we provide a comprehensive network-based strategy for the systematic discovery of functional synergistic modules that are causal determinants of inflammation-induced tumorigenesis. Our approach prioritizes candidate genes selected by integrating clinical-based and network-based genome-wide gene prediction methods and identifies functional synergistic modules based on combinatorial CRISPR-Cas9 screening. On the basis of candidate genes inferred de novo from experimental and computational methods to be involved in inflammation and cancer, we used an existing TGFβ1-induced cellular transformation model in colonic epithelial cells and a new combinatorial CRISPR-Cas9 screening strategy to construct an inflammation-induced differential genetic interaction network. The inflammation-induced differential genetic interaction network that we generated yielded functional insights into the genes and functional module combinations, and showed varied responses to the inflammation agents as well as active traditional Chinese medicine compounds. We identified opposing differential genetic interactions of inflammation-induced tumorigenesis: synergistic promotion and suppression. The synergistic promotion state was primarily caused by deletions in the immune and metabolism modules; the synergistic suppression state was primarily induced by deletions in the proliferation and immune modules or in the proliferation and metabolism modules. These results provide insight into possible early combinational targets and biomarkers for inflammation-induced tumorigenesis and highlight the synergistic effects that occur among immune, proliferation, and metabolism modules. In conclusion, this approach deepens the understanding of the underlying mechanisms that cause inflammation to potentially increase the cancer risk of colonic epithelial cells and accelerate the translation into novel functional modules or synergistic module combinations that modulate complex disease phenotypes.
Collapse
Affiliation(s)
- Yucheng Guo
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Chen Bao
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Dacheng Ma
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yubing Cao
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanda Li
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Zhen Xie
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Shao Li
- MOE Key Laboratory of Bioinformatics and TCM-X Center/Bioinformatics Division/TFIDT, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| |
Collapse
|
245
|
Zha L, Garrett S, Sun J. Salmonella Infection in Chronic Inflammation and Gastrointestinal Cancer. Diseases 2019; 7:E28. [PMID: 30857369 PMCID: PMC6473780 DOI: 10.3390/diseases7010028] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Revised: 02/16/2019] [Accepted: 03/06/2019] [Indexed: 12/19/2022] Open
Abstract
Salmonella not only causes acute infections, but can also cause patients to become chronic "asymptomatic" carriers. Salmonella has been verified as a pathogenic factor that contributes to chronic inflammation and carcinogenesis. This review summarizes the acute and chronic Salmonella infection and describes the current research progress of Salmonella infection contributing to inflammatory bowel disease and cancer. Furthermore, this review explores the underlying biological mechanism of the host signaling pathways manipulated by Salmonella effector molecules. Using experimental animal models, researchers have shown that Salmonella infection is related to host biological processes, such as host cell transformation, stem cell maintenance, and changes of the gut microbiota (dysbiosis). Finally, this review discusses the current challenges and future directions in studying Salmonella infection and its association with human diseases.
Collapse
Affiliation(s)
- Lang Zha
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Shari Garrett
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
246
|
Wu W, Hutcheon AEK, Sriram S, Tran JA, Zieske JD. Initiation of fibrosis in the integrin Αvβ6 knockout mice. Exp Eye Res 2019; 180:23-28. [PMID: 30500364 PMCID: PMC6540115 DOI: 10.1016/j.exer.2018.11.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 11/24/2018] [Accepted: 11/27/2018] [Indexed: 01/22/2023]
Abstract
We previously demonstrated that β6 knockout mice showed impaired wound repair in corneal debridement and keratectomy wounds. In the current investigation, we continued our examination of integrin αvβ6 in order to determine if it was required for the initiation of wound healing in a corneal wound model that normally heals in a fibrotic manner. A full-thickness corneal incision was made in C57BL/6 J wild type (WT) and C57BL/6-Itgb6 KO (β6-/-) mice. The mice were observed at 3, 7, 14, and 28 days post-incision. The morphology of corneal restoration was observed in tissue sections stained with hemotoxilin and eosin (H&E). In addition, indirect-immunofluorescence (IF) was performed on sections and/or whole mounts to evaluate the immunolocalization of α-smooth muscle actin (SMA) and thrombospondin-1 (TSP-1). H&E staining revealed that the corneas in β6-/- mice healed slower than those in WT mice, with an obvious delay in the restoration of the stromal matrix and epithelium. In sections at 3 and 7 days, SMA and TSP-1 were greatly reduced in the β6-/- mice as compared to WT, but peaked at 28 days after incision. Whole mount SMA IF results were consistent with those from sections. Therefore, the initiation of fibrosis was inhibited by the lack of αvβ6; however, there appeared to be an alternate mechanism that initiated fibrosis 7-14 days later. Localization of TSP-1 correlated with expression of SMA whether wound healing was delayed or initiated immediately after wounding.
Collapse
Affiliation(s)
- Wenjing Wu
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Audrey E K Hutcheon
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Sriniwas Sriram
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - Jennifer A Tran
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| | - James D Zieske
- Schepens Eye Research Institute/Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, 20 Staniford Street, Boston, MA, 02114, USA.
| |
Collapse
|
247
|
Yang M, Chen G, Zhang X, Guo Y, Yu Y, Tian L, Chang S, Chen ZK. Inhibition of class I HDACs attenuates renal interstitial fibrosis in a murine model. Pharmacol Res 2019; 142:192-204. [PMID: 30807866 DOI: 10.1016/j.phrs.2019.02.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 01/20/2019] [Accepted: 02/12/2019] [Indexed: 02/07/2023]
Abstract
Renal interstitial fibrosis is the most common of all the forms of chronic kidney disease (CKD). Research has shown that histone deacetylases (HDACs) participate in the process leading to renal fibrosis. However, the effects of class I HDAC inhibitors on the mechanisms of onset and progression of renal interstitial fibrosis are still unclear. Here, we present the effects and mechanisms of action of FK228 (a selective inhibitor of class I HDACs) in the murine model of unilateral ureteral obstruction (UUO) and in vitro models. We investigated the antifibrotic role of FK228 in a murine model of UUO. We used two key effector cell populations, rat renal interstitial fibroblasts and renal tubular epithelial cells exposed to recombinant transforming growth factor-beta 1 (TGF-β1), to explore the mechanistic pathways among in vitro models. The results indicated that FK228 significantly suppressed the production of extracellular matrix (ECM) in both in vivo and in vitro models. FK228 inhibited renal fibroblast activation and proliferation and increased the acetylation of histone H3. We found that FK228 also inhibited the small mothers against decapentaplegic (Smad) and non-Smad signaling pathways. So FK228 could significantly suppress renal interstitial fibrosis via Smad and non-Smad pathways. FK228 may be the basis for a new and effective medicine for alleviating renal fibrosis in the future.
Collapse
Affiliation(s)
- Min Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Gen Chen
- Department of Radiology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xue Zhang
- Department of Breast Surgery, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yuliang Guo
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Yan Yu
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Li Tian
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Sheng Chang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Zhonghua Klaus Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
248
|
Zhang L, Shen ZY, Wang K, Li W, Shi JM, Osoro EK, Ullah N, Zhou Y, Ji SR. C-reactive protein exacerbates epithelial-mesenchymal transition through Wnt/β-catenin and ERK signaling in streptozocin-induced diabetic nephropathy. FASEB J 2019; 33:6551-6563. [PMID: 30794428 DOI: 10.1096/fj.201801865rr] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Previous studies have reported the pathogenic role of C-reactive protein (CRP) during diabetic kidney disease (DKD) in human CRP transgenic and Crp-/- mice. However, because humans and mice have inverse acute phase expression patterns of CRP and serum amyloid P component, this could lead to the inaccurate evaluation of CRP function with the above-mentioned CRP transgenic mouse. But different from mice, rats have the same acute phase protein expression pattern as human, which might avoid this problem and be a better choice for CRP function studies. To dispel this doubt and accurately define the role of CRP during diabetic nephropathy, we created the first Crp-/- rat model, which we treated with streptozocin to induce DKD for in vivo studies. Moreover, an established cell line (human kidney 2) was used to further investigate the pathologic mechanisms of CRP. We found that CRP promotes epithelial-mesenchymal transition (EMT) through Wnt/β-catenin and ERK1/2 signaling, which are dependent on CRP binding to FcγRII on apoptotic cells. By promoting EMT, CRP was demonstrated to accelerate the development of DKD. We thus present convincing evidence demonstrating CRP as a therapeutic target for DKD treatment.-Zhang, L., Shen, Z.-Y., Wang, K., Li, W., Shi, J.-M., Osoro, E. K., Ullah, N., Zhou, Y., Ji, S.-R. C-reactive protein exacerbates epithelial-mesenchymal transition through Wnt/β-catenin and ERK signaling in streptozocin-induced diabetic nephropathy.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Zhi-Yuan Shen
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Ke Wang
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Wei Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Jing-Ming Shi
- Department of Anatomy, School of Basic Medical Sciences, Xizang Minzu University, Xianyang, China
| | - Ezra Kombo Osoro
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Naeem Ullah
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhou
- Department of Dermatology, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shang-Rong Ji
- Ministry of Education (MOE) Key Laboratory of Cell Activities and Stress Adaptations, School of Life Sciences, Lanzhou University, Lanzhou, China
| |
Collapse
|
249
|
Du Y, Song W, Chen J, Chen H, Xuan Z, Zhao L, Chen J, Jin C, Zhou M, Tuo B, Zhao Y, Zheng S, Song P. The potassium channel KCa3.1 promotes cell proliferation by activating SKP2 and metastasis through the EMT pathway in hepatocellular carcinoma. Int J Cancer 2019; 145:503-516. [PMID: 30628729 DOI: 10.1002/ijc.32121] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Revised: 11/17/2018] [Accepted: 12/19/2018] [Indexed: 12/19/2022]
Abstract
The intermediate conductance calcium-activated potassium channel (KCa3.1) plays an important role in maintaining intracellular calcium homeostasis and is involved in the tumorigenesis of many human cancers. However, it is unknown whether KCa3.1 plays a role in the genesis of hepatocellular carcinoma (HCC), one of the most common malignant tumors worldwide with a very poor prognosis. In our study, we found that the expression of KCa3.1 was significantly elevated in poorly differentiated HCC tissues compared to adjacent noncancerous tissues. In vitro and in vivo experiments showed that KCa3.1 could promote cell proliferation, migration, and invasion of HCC. Mechanistically, KCa3.1 promoted cell cycle progression and migration and invasion of HCC cells by activating S-phase protein kinase 2 (SKP2) to trigger the degradation of p21 and p27 and targeting Reelin (RELN) to induce epithelial-mesenchymal transition (EMT), respectively. Taken together, our results demonstrate that KCa3.1 plays an important role in the genesis and progression of HCC, implying that it might be a promising therapeutic target in HCC.
Collapse
Affiliation(s)
- Yehui Du
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Wenfeng Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jian Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Hao Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Zefeng Xuan
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Long Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Jun Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Cheng Jin
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Mengqiao Zhou
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Biguang Tuo
- Department of Gastroenterology, Affiliated Hospital of Zunyi Medical College, Zunyi, China
| | - Yongchao Zhao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| | - Penghong Song
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University.,NHFPC Key Laboratory of Combined Multi-organ Transplantation, Hangzhou, China.,Key Laboratory of Organ Transplantation, Zhejiang Province, Hangzhou, China.,Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, China
| |
Collapse
|
250
|
Zou Y, Li S, Li Z, Song D, Zhang S, Yao Q. MiR-146a attenuates liver fibrosis by inhibiting transforming growth factor-β1 mediated epithelial-mesenchymal transition in hepatocytes. Cell Signal 2019; 58:1-8. [PMID: 30711634 DOI: 10.1016/j.cellsig.2019.01.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Revised: 01/05/2019] [Accepted: 01/31/2019] [Indexed: 12/21/2022]
Abstract
Epithelial-mesenchymal transition (EMT) has emerged as a vital process in embryogenesis, carcinogenesis, and tissue fibrosis. Transforming growth factor-beta 1 (TGF-β1)-mediated signaling pathways play important roles in the EMT process. MicroRNA-146a (miR-146a) has been suggested as a significant regulatory molecule in fibrogenesis. Therefore, the present study aimed to evaluate the effect of miR-146a on the EMT of hepatocytes and to investigate the role of overexpressing miR-146a on rat hepatic fibrosis. The results showed that the miR-146a level decreased during the EMT process of L02 hepatocytes induced by TGF-β1 in vitro. Moreover, miR-146a overexpression led to significant reduction of EMT-related markers expression in hepatocytes. Subsequent experiments revealed that miR-146a attenuated the EMT process in hepatocytes by targeting small mothers against decapentaplegic (SMAD) 4. Meanwhile, restoration of SMAD4 expression rescued the inhibitory effect of miRNA-146a on EMT. Further in vivo studies revealed that intravenous injection of miR-146a-expressing adenovirus (Ad-miR-146a) successfully restored the miR-146a levels and mitigated fibrogenesis in the livers of CCl4-treated rats. More importantly, after Ad-miR-146a treatment, inhibition of both EMT traits and SMAD4 expression was observed. The results of the present study showed that miR-146a/SMAD4 is a key signaling cascade that inhibits hepatocyte EMT, and the introduction of miR-146a might present a promising therapeutic option for liver fibrosis.
Collapse
Affiliation(s)
- Yanting Zou
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, PR China; Shanghai Institute of Liver disease, Shanghai, PR China
| | - Shuyu Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, PR China; Shanghai Institute of Liver disease, Shanghai, PR China
| | - Zhengliang Li
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, PR China; Shanghai Institute of Liver disease, Shanghai, PR China
| | - Dongqiang Song
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Shuncai Zhang
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, PR China; Shanghai Institute of Liver disease, Shanghai, PR China.
| | - Qunyan Yao
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Fudan University, Shanghai, PR China; Shanghai Institute of Liver disease, Shanghai, PR China.
| |
Collapse
|