1
|
Ouyang Y, Shen R, Chu L, Fu C, Hu W, Huang H, Zhang Z, Jiang M, Chen X. Combining single-cell and bulk RNA sequencing, NK cell marker genes reveal a prognostic and immune status in pancreatic ductal adenocarcinoma. Sci Rep 2024; 14:15037. [PMID: 38951569 PMCID: PMC11217423 DOI: 10.1038/s41598-024-65917-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
The NK cell is an important component of the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC), also plays a significant role in PDAC development. This study aimed to explore the relationship between NK cell marker genes and prognosis, immune response of PDAC patients. By scRNA-seq data, we found the proportion of NK cells were significantly downregulated in PDAC and 373 NK cell marker genes were screened out. By TCGA database, we enrolled 7 NK cell marker genes to construct the signature for predicting prognosis in PDAC patients. Cox analysis identified the signature as an independent factor for pancreatic cancer. Subsequently, the predictive power of signature was validated by 6 GEO datasets and had an excellent evaluation. Our analysis of relationship between the signature and patients' immune status revealed that the signature has a strong correlation with immunocyte infiltration, inflammatory reaction, immune checkpoint inhibitors (ICIs) response. The NK cell marker genes are closely related to the prognosis and immune capacity of PDAC patients, and they have potential value as a therapeutic target.
Collapse
Affiliation(s)
- Yonghao Ouyang
- Research Institute of General Surgery, Jinling Hospital, Nanjing University Medical School, 305 Zhong Shan East Road, Nanjing, 210002, China.
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China.
| | - Rongxi Shen
- Research Institute of General Surgery, Jinling Hospital, Nanjing University Medical School, 305 Zhong Shan East Road, Nanjing, 210002, China.
| | - Lihua Chu
- Jinggangshan University, Ji'an, 334000, China
| | - Chengchao Fu
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China
| | - Wang Hu
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China
| | - Haoxuan Huang
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China
| | - Zhicheng Zhang
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China
| | - Ming Jiang
- Nanchang University, 461 Bayi Avenue, Nanchang, 330006, Jiangxi, China
| | - Xin Chen
- Jiangxi University of Chinese Medicine, Nanchang, 330000, China
| |
Collapse
|
2
|
Guo Y, Sun Y, Ma M, Huang Y, Zhang S, Tian Q. Djsnon, a downstream gene of Djfoxk1, is required for the regeneration of the planarian central nervous system. Biochem Biophys Res Commun 2023; 643:8-15. [PMID: 36584589 DOI: 10.1016/j.bbrc.2022.12.074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022]
Abstract
Regulators of adult neurogenesis are crucial targets for neuronal repair. Freshwater planarians are ideal model systems for studying neuronal regeneration as they can regenerate their entire central nervous system (CNS) using pluripotent adult stem cells. Here, we identified Djfoxk1 in planarian Dugesia japonica to be required for planarian CNS regeneration. Knockdown of Djfoxk1 inhibits the regeneration of the cephalic ganglia, resulting in the failure of eye regeneration. By RNAi screening of Djfoxk1 downstream genes, we identified Djsnon as another regulator of planarian neuronal regeneration. Inhibition of Djsnon with RNA interference (RNAi) results in similar phenotypes caused by Djfoxk1 RNAi without affecting cell proliferation and wound healing. Our findings show that Djsnon as a downstream gene of Djfoxk1 regulates the regeneration of the planarian CNS.
Collapse
Affiliation(s)
- Yajun Guo
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yujia Sun
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengwen Ma
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Yongding Huang
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Shoutao Zhang
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China; Longhu Laboratory of Advanced Immunology, Zhengzhou, Henan, China.
| | - Qingnan Tian
- School of LifeSciences, Zhengzhou University, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Maheshwari A, Traub TM, Garg PM, Ethawi Y, Buonocore G. Necrotizing Enterocolitis: Clinical Features, Histopathological Characteristics, and Genetic Associations. Curr Pediatr Rev 2022; 18:210-225. [PMID: 35125082 DOI: 10.2174/1573396318666220204113858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 11/22/2022]
Abstract
Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis seen in premature infants. Although the etiopathogenesis of NEC is unclear, genetic factors may alter a patient's susceptibility, clinical course, and outcomes. This review draws from existing studies focused on individual genes and others based on microarray-based high-throughput discovery techniques. We have included evidence from our own studies and from an extensive literature search in the databases PubMed, EMBASE, and Scopus. To avoid bias in the identification of studies, keywords were short-listed a priori from anecdotal experience and PubMed's Medical Subject Heading (MeSH) thesaurus.
Collapse
Affiliation(s)
| | - Terri M Traub
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Parvesh M Garg
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, University of Mississippi, Jackson, Mississippi, USA
| | - Yahya Ethawi
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics, Saudi American Hospital, Ajman, United Arab Emirates
| | - Giuseppe Buonocore
- Global Newborn Society, Clarksville, Maryland, USA.,Department of Pediatrics/ Neonatology, University of Siena, Siena, Italy
| |
Collapse
|
4
|
Liang L, Li S, Liu H, Mao Y, Liu L, Zhang X, Peng W, Xiao Y, Zhang F, Shi M, Wang Y, Guo B. Blood glucose control contributes to protein stability of Ski-related novel protein N in a rat model of diabetes. Exp Ther Med 2021; 22:1341. [PMID: 34630695 DOI: 10.3892/etm.2021.10776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 06/30/2021] [Indexed: 11/05/2022] Open
Abstract
Ski-related novel protein N (SnoN) negatively regulates the transforming growth factor-β1 (TGF-β1)/Smads signaling pathway and is present at a low level during diabetic nephropathy (DN), but its underlying regulatory mechanism is currently unknown. The present study aimed to assess the effects of insulin-controlled blood glucose on renal SnoN expression and fibrosis in rats with diabetes mellitus (DM). Streptozotocin-induced DM rats were treated with insulin glargine (INS group) following successful model establishment. Blood samples were collected and centrifuged for biochemical indexes and the kidneys were collected for morphological analysis. In vitro, rat renal proximal tubular epithelial cells were treated with high-glucose medium for 24 h and transferred to normal glucose medium for 24 h. The expression levels of TGF-β1, SnoN, Smad ubiquitin regulatory factor 2 (Smurf2), Arkadia, Smads, E-cadherin, α-smooth muscle actin and collagen III were assessed by western blotting and immunohistochemistry. The ubiquitylation of SnoN was detected by immunoprecipitation, and the expression levels of SnoN mRNA were evaluated by reverse transcription-quantitative PCR. The biochemical parameters and morphology indicated that renal fibrosis was notable in the DM group and mitigated in the INS group. Compared with the control group, TGF-β1, phosphor (p)-Smad2, p-Smad3, Smurf2 and Arkadia levels were enhanced in the DM group, and the levels of SnoN protein were decreased, whereas the levels of SnoN mRNA and ubiquitylation were increased in renal tissues. Notably, treatment with insulin reversed this trend. Furthermore, changing the glucose levels in the medium from high to normal glucose suppressed the epithelial-mesenchymal transition of NRK-52E cells by restoring the SnoN protein levels, and this phenomenon was impaired by the knockout of SnoN. SnoN protein levels were likely reduced through a mechanism enhanced by the ubiquitin proteasome system, which reversed the transcriptional activation of SnoN during DN progression. In addition, controlling blood glucose may delay DN fibrosis by rescuing the protein stability of SnoN.
Collapse
Affiliation(s)
- Luqun Liang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Shuang Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Huiming Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Yanwen Mao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Lingling Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Xiaohuan Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Wei Peng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Ying Xiao
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Yuanyuan Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China.,Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
5
|
Role of macrophages in fetal development and perinatal disorders. Pediatr Res 2021; 90:513-523. [PMID: 33070164 DOI: 10.1038/s41390-020-01209-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/17/2020] [Accepted: 09/25/2020] [Indexed: 02/06/2023]
Abstract
In the fetus and the neonate, altered macrophage function has been implicated not only in inflammatory disorders but also in developmental abnormalities marked by altered onset, interruption, or imbalance of key structural changes. The developmental role of macrophages were first noted nearly a century ago, at about the same time when these cells were being identified as central effectors in phagocytosis and elimination of microbes. Since that time, we have made considerable progress in understanding the diverse roles that these cells play in both physiology and disease. Here, we review the role of fetal and neonatal macrophages in immune surveillance, innate immunity, homeostasis, tissue remodeling, angiogenesis, and repair of damaged tissues. We also discuss the possibility of therapeutic manipulation of the relative abundance and activation status of macrophage subsets in various diseases. This article combines peer-reviewed evidence from our own studies with results of an extensive literature search in the databases PubMed, EMBASE, and Scopus. IMPACT: We have reviewed the structure, differentiation, and classification of macrophages in the neonatal period. Neonatal macrophages are derived from embryonic, hepatic, and bone marrow precursors. Macrophages play major roles in tissue homeostasis, innate immunity, inflammation, tissue repair, angiogenesis, and apoptosis of various cellular lineages in various infectious and inflammatory disorders. Macrophages and related inflammatory mediators could be important therapeutic targets in several neonatal diseases.
Collapse
|
6
|
Sader F, Roy S. Tgf-β superfamily and limb regeneration: Tgf-β to start and Bmp to end. Dev Dyn 2021; 251:973-987. [PMID: 34096672 DOI: 10.1002/dvdy.379] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/19/2022] Open
Abstract
Axolotls represent a popular model to study how nature solved the problem of regenerating lost appendages in tetrapods. Our work over many years focused on trying to understand how these animals can achieve such a feat and not end up with a scarred up stump. The Tgf-β superfamily represents an interesting family to target since they are involved in wound healing in adults and pattern formation during development. This family is large and comprises Tgf-β, Bmps, activins and GDFs. In this review, we present work from us and others on Tgf-β & Bmps and highlight interesting observations between these two sub-families. Tgf-β is important for the preparation phase of regeneration and Bmps for the redevelopment phase and they do not overlap with one another. We present novel data showing that the Tgf-β non-canonical pathway is also not active during redevelopment. Finally, we propose a molecular model to explain how Tgf-β and Bmps maintain distinct windows of expression during regeneration in axolotls.
Collapse
Affiliation(s)
- Fadi Sader
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada
| | - Stéphane Roy
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, Québec, Canada.,Department of Stomatology, Faculty of Dentistry, Université de Montréal, Montréal, Québec, Canada
| |
Collapse
|
7
|
Zhang KJ, Hu Y, Luo N, Li X, Chen FY, Yuan JQ, Guo L. miR‑574‑5p attenuates proliferation, migration and EMT in triple‑negative breast cancer cells by targeting BCL11A and SOX2 to inhibit the SKIL/TAZ/CTGF axis. Int J Oncol 2020; 56:1240-1251. [PMID: 32319565 DOI: 10.3892/ijo.2020.4995] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/21/2019] [Indexed: 11/05/2022] Open
Abstract
Triple‑negative breast cancer (TNBC) is a subtype of breast cancer with a high degree of malignancy. TNBC is prone to distant metastasis and has a poor prognosis. A number of TNBC‑related microRNAs (miRNAs) have been studied and identified. However, the detailed roles of miR‑574‑5p in TNBC remain poorly understood. miR‑574‑5p, SRY (sex determining region Y)‑box 2 (SOX2), B‑cell lymphoma/leukaemia 11A (BCL11A), SKI like proto‑oncogene (SKIL) and epithelial‑mesenchymal transition (EMT)‑related miRNAs and proteins were measured by reverse transcription‑quantitative PCR and western blotting analysis, respectively. A luciferase reporter assay was employed to validate the direct targeting of SOX2 and BCL11A by miR‑574‑5p. MTT, colony formation and Transwell assays were performed to analyse the biological functions of miR‑574‑5p in TNBC cells. A nude mouse xenograft model was used to verify the effects of miR‑574‑5p on the tumorigenesis of TNBC in vivo. The results demonstrated that miR‑574‑5p levels were decreased in breast cancer tissues and cells. miR‑574‑5p repressed proliferation, migration and EMT in TNBC cells. Further experiments confirmed that miR‑574‑5p reduced tumour size and metastasis in vivo. miR‑574‑5p targeted BCL11A and SOX2 to inhibit the SKIL/transcriptional co‑activator with PDZ‑binding motif/connective tissue growth factor axis, and the inhibitory effect of miR‑574‑5p in TNBC cells was at least partly dependent on SOX2 and BCL11A. In addition, the regulation of downstream oncogenes by SOX2 was dependent on BCL11A. To the best of our knowledge, this is the first study to report the association between the miR‑574‑5p/BCL11A/SOX2 axis and the tumorigenesis of TNBC, which provides a new mechanism for understanding the progression of TNBC.
Collapse
Affiliation(s)
- Ke-Jing Zhang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Yu Hu
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Na Luo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Xin Li
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fei-Yu Chen
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Jia-Qi Yuan
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Lei Guo
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
8
|
Zhu Q, Chang A, Xu A, Luo K. The regulatory protein SnoN antagonizes activin/Smad2 protein signaling and thereby promotes adipocyte differentiation and obesity in mice. J Biol Chem 2018; 293:14100-14111. [PMID: 30030373 DOI: 10.1074/jbc.ra118.003678] [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: 04/25/2018] [Revised: 07/17/2018] [Indexed: 12/25/2022] Open
Abstract
Ski-related oncogene SnoN (SnoN or SKIL) regulates multiple signaling pathways in a tissue- and developmental stage-dependent manner and has broad functions in embryonic angiogenesis, mammary gland alveologenesis, cancer, and aging. Here, we report that SnoN also plays a critical role in white adipose tissue (WAT) development by regulating mesenchymal stem cell (MSC) self-renewal and differentiation. We found that SnoN promotes MSC differentiation in the adipocyte lineage by antagonizing activin A/Smad2, but not TGFβ/Smad3 signaling. Mice lacking SnoN or expressing a mutant SnoN defective in binding to the Smads were protected from high-fat diet-induced obesity and insulin resistance, and MSCs lacking a functional SnoN exhibited defective differentiation. We further demonstrated that activin, via Smad2, appears to be the major regulator of WAT development in vivo We also noted that activin A is abundantly expressed in WAT and adipocytes through an autocrine mechanism and promotes MSC self-renewal and inhibits adipogenic differentiation by inducing expression of the gene encoding the homeobox transcription factor Nanog. Of note, SnoN repressed activin/Smad2 signaling and activin A expression, enabling expression of adipocyte-specific transcription factors and promoting adipogenic differentiation. In conclusion, our study has revealed that SnoN plays an important in vivo role in adipocyte differentiation and WAT development in vivo by decreasing activity in the activin/Smad2 signaling pathway.
Collapse
Affiliation(s)
- Qingwei Zhu
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Amanda Chang
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720
| | - Albert Xu
- the Department of Cellular and Molecular Pharmacology, Howard Hughes Medical Institute, University of California, San Francisco, California 94158-2140
| | - Kunxin Luo
- From the Department of Molecular and Cell Biology, University of California, Berkeley, California 94720, .,the Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720 and
| |
Collapse
|
9
|
Wang Y, Xiao Y, Li S, Shi L, Liu L, Zhang Y, Shi M, Guo B. BMP-7 enhances SnoN mRNA expression in renal tubular epithelial cells under high-glucose conditions. Mol Med Rep 2017; 16:3308-3314. [PMID: 28765970 PMCID: PMC5548011 DOI: 10.3892/mmr.2017.7025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 07/04/2017] [Indexed: 12/22/2022] Open
Abstract
The present study aimed to identify any association between bone morphogenetic protein-7 (BMP-7) and the expression of the transcriptional co-repressor Ski-related novel protein N (SnoN), in addition to alterations in tubulointerstitial fibrosis, during the development and progression of diabetic nephropathy (DN). Streptozotocin was injected into the tail veins of 20 healthy and specific pathogen-free male Sprague-Dawley rats. The rats were sacrificed to detect the appropriate biochemical indicators of renal pathological alterations following 24 weeks. Then, various doses of human recombinant BMP-7 were added to high glucose-cultured NRK-52E cells. Immunohistochemistry, immunofluorescence staining and western blotting were used to determine the expression of SnoN, BMP-7, Smad ubiquitin regulatory factor (Smurf)2, Arkadia, E-cadherin, α-smooth muscle actin and Collagen III. Reverse transcription-quantitative polymerase chain reaction was used to detect SnoN mRNA expression. With the progression of DN, the expression of BMP-7 in rat renal tissue was downregulated, whereas the expression of Smurf2 and Arkadia increased. Furthermore, the expression of SnoN mRNA increased however the expression of SnoN protein decreased, accompanied by renal tubular epithelial cell mesenchymal transition, extracellular matrix (ECM) deposition and severe renal function disorder. The exogenous recombinant human BMP-7 alleviated high glucose-induced phenotypic transformation and ECM synthesis of NRK-52E in vitro and upregulated SnoN transcription and protein expression, however no effect was observed on the expression of Smurf2 and Arkadia. BMP-7 may ameliorate DN and renal fibrosis via increasing the expression of SnoN mRNA and protein in renal tubular epithelial cells, rather than directly inhibiting the degradation of SnoN by E3 ubiquitin ligase.
Collapse
Affiliation(s)
- Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Shuang Li
- Department of Pathophysiology, Guizhou Medical Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Lei Shi
- Department of Pathology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, Guizhou 556000, P.R. China
| | - Lirong Liu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Yingying Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, Guizhou 550025, P.R. China
| |
Collapse
|
10
|
MohanKumar K, Namachivayam K, Chapalamadugu K, Garzon SA, Premkumar MH, Tipparaju S, Maheshwari A. Smad7 interrupts TGF-β signaling in intestinal macrophages and promotes inflammatory activation of these cells during necrotizing enterocolitis. Pediatr Res 2016; 79:951-61. [PMID: 26859364 PMCID: PMC4899224 DOI: 10.1038/pr.2016.18] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 11/18/2015] [Indexed: 12/31/2022]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. Based on our recent findings of increased Smad7 expression in surgically resected bowel affected by NEC, we hypothesized that NEC macrophages undergo inflammatory activation because increased Smad7 expression renders these cells resistant to normal, gut-specific, transforming growth factor (TGF)-β-mediated suppression of inflammatory pathways. METHODS We used surgically resected human NEC tissue, murine models of NEC-like injury, bone marrow-derived and intestinal macrophages, and RAW264.7 cells. Smad7 and IκB kinase-beta (IKK-β) were measured by quantitative PCR, western blots, and immunohistochemistry. Promoter activation was confirmed in luciferase reporter and chromatin immunoprecipitation assays. RESULTS NEC macrophages showed increased Smad7 expression, particularly in areas with severe tissue damage and high bacterial load. Lipopolysaccharide-induced Smad7 expression suppressed TGF-β signaling and augmented nuclear factor-kappa B (NF-κB) activation and cytokine production in macrophages. Smad7-mediated NF-κB activation was likely mediated via increased expression of IKK-β, which, further increased Smad7 expression in a feed-forward loop. We show that Smad7 induced IKK-β expression through direct binding to the IKK-β promoter and its transcriptional activation. CONCLUSION Smad7 expression in NEC macrophages interrupts TGF-β signaling and promotes NF-κB-mediated inflammatory signaling in these cells through increased expression of IKK-β.
Collapse
Affiliation(s)
- Krishnan MohanKumar
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kopperuncholan Namachivayam
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - Kalyan Chapalamadugu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, USA
| | - Steven A. Garzon
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | - Srinivas Tipparaju
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, Florida, USA
| | - Akhil Maheshwari
- Department of Pediatrics, University of Illinois at Chicago, Chicago, Illinois, USA, Department of Pediatrics, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA, Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA, Department of Community and Family Health, College of Public Health, University of South Florida, Tampa, Florida, USA,Address for correspondence: Akhil Maheshwari, 1 Tampa General Circle, Suite F170, Tampa, FL 33606, USA; Phone: 813-844-3437; Fax: 813-844-1671;
| |
Collapse
|
11
|
Zhu Q, Le Scolan E, Jahchan N, Ji X, Xu A, Luo K. SnoN Antagonizes the Hippo Kinase Complex to Promote TAZ Signaling during Breast Carcinogenesis. Dev Cell 2016; 37:399-412. [PMID: 27237790 DOI: 10.1016/j.devcel.2016.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 04/01/2016] [Accepted: 05/01/2016] [Indexed: 12/12/2022]
Abstract
SnoN regulates multiple signaling pathways, including TGF-β/Smad and p53, and displays both pro-oncogenic and anti-oncogenic activities in human cancer. We have observed previously that both its intracellular localization and expression levels are sensitive to cell density, suggesting that it may crosstalk with Hippo signaling. Here we report that, indeed, SnoN interacts with multiple components of the Hippo pathway to inhibit the binding of Lats2 to TAZ and the subsequent phosphorylation of TAZ, leading to TAZ stabilization. Consistently, SnoN enhances the transcriptional and oncogenic activities of TAZ, and reducing SnoN decreases TAZ expression as well as malignant progression of breast cancer cells. Interestingly, SnoN itself is downregulated by Lats2 that is activated by the Scribble basolateral polarity protein. Thus, SnoN is a critical component of the Hippo regulatory network that receives signals from the tissue architecture and polarity to coordinate the activity of intracellular signaling pathways.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA
| | | | - Nadine Jahchan
- ORIC Pharmaceuticals, South San Francisco, CA 94080, USA
| | - Xiaodan Ji
- Departments of Cancer Biology, Cell & Developmental Biology, and Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 190104-6160, USA
| | - Albert Xu
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, 16 Barker Hall, MC3204, Berkeley, CA 94720, USA; Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA.
| |
Collapse
|
12
|
Abstract
The abundance and cross-linking of intramuscular connective tissue contributes to the background toughness of meat, and is thus undesirable. Connective tissue is mainly synthesized by intramuscular fibroblasts. Myocytes, adipocytes and fibroblasts are derived from a common pool of progenitor cells during the early embryonic development. It appears that multipotent mesenchymal stem cells first diverge into either myogenic or non-myogenic lineages; non-myogenic mesenchymal progenitors then develop into the stromal-vascular fraction of skeletal muscle wherein adipocytes, fibroblasts and derived mesenchymal progenitors reside. Because non-myogenic mesenchymal progenitors mainly undergo adipogenic or fibrogenic differentiation during muscle development, strengthening progenitor proliferation enhances the potential for both intramuscular adipogenesis and fibrogenesis, leading to the elevation of both marbling and connective tissue content in the resulting meat product. Furthermore, given the bipotent developmental potential of progenitor cells, enhancing their conversion to adipogenesis reduces fibrogenesis, which likely results in the overall improvement of marbling (more intramuscular adipocytes) and tenderness (less connective tissue) of meat. Fibrogenesis is mainly regulated by the transforming growth factor (TGF) β signaling pathway and its regulatory cascade. In addition, extracellular matrix, a part of the intramuscular connective tissue, provides a niche environment for regulating myogenic differentiation of satellite cells and muscle growth. Despite rapid progress, many questions remain in the role of extracellular matrix on muscle development, and factors determining the early differentiation of myogenic, adipogenic and fibrogenic cells, which warrant further studies.
Collapse
|
13
|
Caligaris C, Vázquez-Victorio G, Sosa-Garrocho M, Ríos-López DG, Marín-Hernández A, Macías-Silva M. Actin-cytoskeleton polymerization differentially controls the stability of Ski and SnoN co-repressors in normal but not in transformed hepatocytes. Biochim Biophys Acta Gen Subj 2015; 1850:1832-41. [PMID: 26002202 DOI: 10.1016/j.bbagen.2015.05.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 04/21/2015] [Accepted: 05/12/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Ski and SnoN proteins function as transcriptional co-repressors in the TGF-β pathway. They regulate cell proliferation and differentiation, and their aberrant expression results in altered TGF-β signalling, malignant transformation, and alterations in cell proliferation. METHODS We carried out a comparative characterization of the endogenous Ski and SnoN protein regulation by TGF-β, cell adhesion disruption and actin-cytoskeleton rearrangements between normal and transformed hepatocytes; we also analyzed Ski and SnoN protein stability, subcellular localization, and how their protein levels impact the TGF-β/Smad-driven gene transcription. RESULTS Ski and SnoN protein levels are lower in normal hepatocytes than in hepatoma cells. They exhibit a very short half-life and a nuclear/cytoplasmic distribution in normal hepatocytes opposed to a high stability and restricted nuclear localization in hepatoma cells. Interestingly, while normal cells exhibit a transient TGF-β-induced gene expression, the hepatoma cells are characterized by a strong and sustained TGF-β-induced gene expression. A novel finding is that Ski and SnoN stability is differentially regulated by cell adhesion and cytoskeleton rearrangements in the normal hepatocytes. The inhibition of protein turnover down-regulated both Ski and SnoN co-repressors impacting the kinetic of expression of TGF-β-target genes. CONCLUSION Normal regulatory mechanisms controlling Ski and SnoN stability, subcellular localization and expression are altered in hepatocarcinoma cells. GENERAL SIGNIFICANCE This work provides evidence that Ski and SnoN protein regulation is far more complex in normal than in transformed cells, since many of the normal regulatory mechanisms are lost in transformed cells.
Collapse
Affiliation(s)
- Cassandre Caligaris
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Genaro Vázquez-Victorio
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Marcela Sosa-Garrocho
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Diana G Ríos-López
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México
| | - Alvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología, México D.F., 14080, México
| | - Marina Macías-Silva
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México. México D.F., 04510, México.
| |
Collapse
|
14
|
Liu C, Zhang H, Zang X, Wang C, Kong Y, Zhang H. The influence of SnoN gene silencing by siRNA on the cell proliferation and apoptosis of human pancreatic cancer cells. Diagn Pathol 2015; 10:30. [PMID: 25907906 PMCID: PMC4407884 DOI: 10.1186/s13000-015-0267-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2014] [Accepted: 04/07/2015] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND The prognosis for pancreatic cancer (PC) is very poor. The SnoN gene may have a role in cell proliferation and apoptosis in human cancer. However, the influence of SnoN on cell proliferation and apoptosis in human PC cells remains unknown. METHODS SnoN expression was assessed in SW1990 PC cell lines using real-time polymerase chain reaction (PCR). A luciferase reporter assay was used to confirm the target associations. The effect of SnoN on cell proliferation in vitro was confirmed using Cell Counting Kit-8. Apoptosis was confirmed using flow cytometry. Gene and protein expression were examined using real time PCR and Western blotting, respectively. RESULTS SnoN siRNA significantly inhibited the growth of SW1990 cells by decreasing cell proliferation (P < 0.05) and increasing cell apoptosis (P < 0.05), compared with the blank group and the negative control group. The highest inhibition of cell proliferation appeared at 3 days post-transfection. Cell apoptosis more obvious at 48 h after transfection. CONCLUSIONS In summary, our results reveal that the RNAi-mediated downregulation of SnoN effectively inhibited the proliferation of PC cells. SnoN-siRNA also enhanced SW1990 PC cell apoptosis. These findings indicate that SnoN gene plays an important role in pancreatic cancer development, and might serve as a potential therapeutic target for pancreatic cancer. However, further in vivo studies are needed to clarify the influence of SnoN gene silencing by siRNA on pancreatic cancer therapy. VIRTUAL SLIDES The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/7609324661510147.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Hui Zhang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Xiaoxia Zang
- Department of Stomatology, Air Force General Hospital of PLA, Beijing, China.
| | - Cheng Wang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Yalin Kong
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| | - Hongyi Zhang
- Department of Hepatobiliary Surgery, Air Force General Hospital of PLA, 30 Fucheng Road, Beijing, 100142, China.
| |
Collapse
|
15
|
Zeglinski MR, Hnatowich M, Jassal DS, Dixon IMC. SnoN as a novel negative regulator of TGF-β/Smad signaling: a target for tailoring organ fibrosis. Am J Physiol Heart Circ Physiol 2015; 308:H75-82. [DOI: 10.1152/ajpheart.00453.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Remodeling of the extracellular matrix is beneficial during the acute wound healing stage following tissue injury. In the short term, resident fibroblasts and myofibroblasts regulate the matrix remodeling process through production of matricellular protein components that provide structural support to the damaged tissue. This process is largely governed by the transforming growth factor-β1 (TGF-β1) pathway, a critical mediator of the remodeling process. In the long term, chronic activation of the TGF-β1 pathway promotes excessive synthesis and deposition of matrix proteins, including fibrillar collagens, which ultimately leads to organ failure. SnoN (and its alternatively-spliced isoforms SnoN2, SnoA, and SnoI) is one of four members of a family of negative regulators of TGF-β1 signaling that includes Ski and functional Smad-suppressing elements on chromosomes 15 and 18. SnoN has been shown to be structurally and functionally similar to Ski and has been demonstrated to directly interact with Ski to abrogate gene expression. Despite this, little progress has been made in delineating a specific role for SnoN in the regulation of myofibroblast phenotype and function. This review outlines the current body of knowledge of what we refer to as the “Ski-Sno superfamily,” with a focus on the structural and functional importance of SnoN in mediating the fibrotic response by myofibroblasts following tissue injury.
Collapse
Affiliation(s)
- Matthew R. Zeglinski
- Department of Physiology, Institute of Cardiovascular Sciences, Saint Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Mark Hnatowich
- Department of Physiology, Institute of Cardiovascular Sciences, Saint Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Davinder S. Jassal
- Department of Physiology, Institute of Cardiovascular Sciences, Saint Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ian M. C. Dixon
- Department of Physiology, Institute of Cardiovascular Sciences, Saint Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
The proteasome inhibitor, MG132, attenuates diabetic nephropathy by inhibiting SnoN degradation in vivo and in vitro. BIOMED RESEARCH INTERNATIONAL 2014; 2014:684765. [PMID: 25003128 PMCID: PMC4070544 DOI: 10.1155/2014/684765] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 05/09/2014] [Accepted: 05/14/2014] [Indexed: 01/13/2023]
Abstract
Transforming growth factor-β (TGF-β) has been shown to be involved in diabetic nephropathy (DN). The SnoN protein can regulate TGF-β signaling through interaction with Smad proteins. Recent studies have shown that SnoN is mainly degraded by the ubiquitin-proteasome pathway. However, the role of SnoN in the regulation of TGF-β/Smad signaling in DN is still unclear. In this study, diabetic rats were randomly divided into a diabetic control group (DC group) and a proteasome inhibitor (MG132) diabetes therapy group (DT group). Kidney damage parameters and the expression of SnoN, Smurf2, and TGF-β were observed. Simultaneously, we cultured rat glomerular mesangial cells (GMCs) stimulated with high glucose, and SnoN and Arkadia expression were measured. Results demonstrated that 24-hour urine protein, ACR, BUN, and the expression of Smurf2 and TGF-β were significantly increased (P < 0.05), whereas SnoN was significantly decreased in the DC group (P < 0.05). However, these changes diminished after treatment with MG132. SnoN expression in GMCs decreased significantly (P < 0.05), but Arkadia expression gradually increased due to high glucose stimulation (P < 0.05), which could be almost completely reversed by MG132 (P < 0.05). The present results support the hypothesis that MG132 may alleviate kidney damage by inhibiting SnoN degradation and TGF-β activation, suggesting that the ubiquitin-proteasome pathway may become a new therapeutic target for DN.
Collapse
|
17
|
Li C, Peart N, Xuan Z, Lewis DE, Xia Y, Jin J. PMA induces SnoN proteolysis and CD61 expression through an autocrine mechanism. Cell Signal 2014; 26:1369-78. [PMID: 24637302 DOI: 10.1016/j.cellsig.2014.03.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2013] [Revised: 02/19/2014] [Accepted: 03/06/2014] [Indexed: 10/25/2022]
Abstract
Phorbol-12-myristate-13-acetate, also called PMA, is a small molecule that activates protein kinase C and functions to differentiate hematologic lineage cells. However, the mechanism of PMA-induced cellular differentiation is not fully understood. We found that PMA triggers global enhancement of protein ubiquitination in K562, a myelogenous leukemia cell line and one of the enhanced-ubiquitination targets is SnoN, an inhibitor of the Smad signaling pathway. Our data indicated that PMA stimulated the production of Activin A, a cytokine of the TGF-β family. Activin A then activated the phosphorylation of both Smad2 and Smad3. In consequence, SnoN is ubiquitinated by the APC(Cdh1) ubiquitin ligase with the help of phosphorylated Smad2. Furthermore, we found that SnoN proteolysis is important for the expression of CD61, a marker of megakaryocyte. These results indicate that protein ubiquitination promotes megakaryopoiesis via degrading SnoN, an inhibitor of CD61 expression, strengths the roles of ubiquitination in cellular differentiation.
Collapse
Affiliation(s)
- Chonghua Li
- Department of Biochemistry and Molecular Biology, Medical School, United States
| | - Natoya Peart
- Department of Biochemistry and Molecular Biology, Medical School, United States; Program of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Zhenyu Xuan
- Department of Molecular and Cellular Biology, The University of Texas at Dallas, Dallas, TX, United States
| | - Dorothy E Lewis
- Department of Internal Medicine, Medical School, United States
| | - Yang Xia
- Department of Biochemistry and Molecular Biology, Medical School, United States; Program of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Jianping Jin
- Department of Biochemistry and Molecular Biology, Medical School, United States; Program of Biochemistry and Molecular Biology, Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States.
| |
Collapse
|
18
|
Marques R, Maia CJ, Vaz C, Correia S, Socorro S. The diverse roles of calcium-binding protein regucalcin in cell biology: from tissue expression and signalling to disease. Cell Mol Life Sci 2014; 71:93-111. [PMID: 23519827 PMCID: PMC11113322 DOI: 10.1007/s00018-013-1323-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 02/21/2013] [Accepted: 03/07/2013] [Indexed: 02/06/2023]
Abstract
Regucalcin (RGN) is a calcium (Ca(2+))-binding protein widely expressed in vertebrate and invertebrate species, which is also known as senescence marker protein 30, due to its molecular weight (33 kDa) and a characteristically diminished expression with the aging process. RGN regulates intracellular Ca(2+) homeostasis and the activity of several proteins involved in intracellular signalling pathways, namely, kinases, phosphatases, phosphodiesterase, nitric oxide synthase and proteases, which highlights its importance in cell biology. In addition, RGN has cytoprotective effects reducing intracellular levels of oxidative stress, also playing a role in the control of cell survival and apoptosis. Multiple factors have been identified regulating the cell levels of RGN transcripts and protein, and an altered expression pattern of this interesting protein has been found in cases of reproductive disorders, neurodegenerative diseases and cancer. Moreover, RGN is a serum-secreted protein, and its levels have been correlated with the stage of disease, which strongly suggests the usefulness of this protein as a potential biomarker for monitoring disease onset and progression. The present review aims to discuss the available information concerning RGN expression and function in distinct cell types and tissues, integrating cellular and molecular mechanisms in the context of normal and pathological conditions. Insight into the cellular actions of RGN will be a key step towards deepening the knowledge of the biology of several human diseases.
Collapse
Affiliation(s)
- Ricardo Marques
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cláudio J. Maia
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cátia Vaz
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sara Correia
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Sílvia Socorro
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| |
Collapse
|
19
|
Zhu Q, Kim YH, Wang D, Oh SP, Luo K. SnoN facilitates ALK1-Smad1/5 signaling during embryonic angiogenesis. ACTA ACUST UNITED AC 2013; 202:937-50. [PMID: 24019535 PMCID: PMC3776356 DOI: 10.1083/jcb.201208113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In endothelial cells, two type I receptors of the transforming growth factor β (TGF-β) family, ALK1 and ALK5, coordinate to regulate embryonic angiogenesis in response to BMP9/10 and TGF-β. Whereas TGF-β binds to and activates ALK5, leading to Smad2/3 phosphorylation and inhibition of endothelial cell proliferation and migration, BMP9/10 and TGF-β also bind to ALK1, resulting in the activation of Smad1/5. SnoN is a negative regulator of ALK5 signaling through the binding and repression of Smad2/3. Here we uncover a positive role of SnoN in enhancing Smad1/5 activation in endothelial cells to promote angiogenesis. Upon ligand binding, SnoN directly bound to ALK1 on the plasma membrane and facilitated the interaction between ALK1 and Smad1/5, enhancing Smad1/5 phosphorylation. Disruption of this SnoN-Smad interaction impaired Smad1/5 activation and up-regulated Smad2/3 activity. This resulted in defective angiogenesis and arteriovenous malformations, leading to embryonic lethality at E12.5. Thus, SnoN is essential for TGF-β/BMP9-dependent biological processes by its ability to both positively and negatively modulate the activities of Smad-dependent pathways.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | | | | | | | | |
Collapse
|
20
|
Sonoda A, Mukaisho KI, Nakayama T, Diem VTN, Hattori T, Andoh A, Fujiyama Y, Sugihara H. Genetic lineages of undifferentiated-type gastric carcinomas analysed by unsupervised clustering of genomic DNA microarray data. BMC Med Genomics 2013; 6:25. [PMID: 23866769 PMCID: PMC3728264 DOI: 10.1186/1755-8794-6-25] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 07/11/2013] [Indexed: 12/13/2022] Open
Abstract
Background It is suspected that early gastric carcinoma (GC) is a dormant variant that rarely progresses to advanced GC. We demonstrated that the dormant and aggressive variants of tubular adenocarcinomas (TUBs) of the stomach are characterized by loss of MYC and gain of TP53 and gain of MYC and/or loss of TP53, respectively. The aim of this study is to determine whether this is also the case in undifferentiated-type GCs (UGCs) of different genetic lineages: one with a layered structure (LS+), derived from early signet ring cell carcinomas (SIGs), and the other, mostly poorly differentiated adenocarcinomas, without LS but with a minor tubular component (TC), dedifferentiated from TUBs (LS−/TC+). Methods Using 29 surgically resected stomachs with 9 intramucosal and 20 invasive UGCs (11 LS+ and 9 LS−/TC+), 63 genomic DNA samples of mucosal and invasive parts and corresponding reference DNAs were prepared from formalin-fixed, paraffin-embedded tissues with laser microdissection, and were subjected to array-based comparative genomic hybridization (aCGH), using 60K microarrays, and subsequent unsupervised, hierarchical clustering. Of 979 cancer-related genes assessed, we selected genes with mean copy numbers significantly different between the two major clusters. Results Based on similarity in genomic copy-number profile, the 63 samples were classified into two major clusters. Clusters A and B, which were rich in LS+ UGC and LS−/TC+ UGC, respectively, were discriminated on the basis of 40 genes. The aggressive pattern was more frequently detected in LS−/TC+ UGCs, (20/26; 77%), than in LS+UGCs (17/37; 46%; P = 0.0195), whereas no dormant pattern was detected in any of the UGC samples. Conclusions In contrast to TUBs, copy number alterations of MYC and TP53 exhibited an aggressive pattern in LS+ SIG at early and advanced stages, indicating that early LS+ UGCs inevitably progress to an advanced GC. Cluster B (enriched in LS−/TC+) exhibited more frequent gain of driver genes and a more frequent aggressive pattern than cluster A, suggesting potentially worse prognosis in UGCs of cluster B.
Collapse
Affiliation(s)
- Ayano Sonoda
- Department of Pathology, Division of Molecular and Diagnostic Pathology, Shiga University of Medical Science, Otsu 520-2192, Japan
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Hernández-Damián J, Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Vázquez-Macías A, Caligaris C, Sosa-Garrocho M, Flores-Pérez B, Romero-Avila M, Macías-Silva M. Downregulation of SnoN oncoprotein induced by antibiotics anisomycin and puromycin positively regulates transforming growth factor-β signals. Biochim Biophys Acta Gen Subj 2013; 1830:5049-58. [PMID: 23872350 DOI: 10.1016/j.bbagen.2013.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 06/26/2013] [Accepted: 07/09/2013] [Indexed: 12/13/2022]
Abstract
BACKGROUND SnoN and Ski proteins function as Smad transcriptional corepressors and are implicated in the regulation of diverse cellular processes such as proliferation, differentiation and transformation. Transforming growth factor-β (TGF-β) signaling causes SnoN and Ski protein degradation via proteasome with the participation of phosphorylated R-Smad proteins. Intriguingly, the antibiotics anisomycin (ANS) and puromycin (PURO) are also able to downregulate Ski and SnoN proteins via proteasome. METHODS We explored the effects of ANS and PURO on SnoN protein downregulation when the activity of TGF-β signaling was inhibited by using different pharmacological and non-pharmacological approaches, either by using specific TβRI inhibitors, overexpressing the inhibitory Smad7 protein, or knocking-down TβRI receptor or Smad2 by specific shRNAs. The outcome of SnoN and Ski downregulation induced by ANS or PURO on TGF-β signaling was also studied. RESULTS SnoN protein downregulation induced by ANS and PURO did not involve the induction of R-Smad phosphorylation but it was abrogated after TGF-β signaling inhibition; this effect occurred in a cell type-specific manner and independently of protein synthesis inhibition or any other ribotoxic effect. Intriguingly, antibiotics seem to require components of the TGF-β/Smad pathway to downregulate SnoN. In addition, SnoN protein downregulation induced by antibiotics favored gene transcription induced by TGF-β signaling. CONCLUSIONS ANS and PURO require TGF-β/Smad pathway to induce SnoN and Ski protein downregulation independently of inducing R-Smad2 phosphorylation, which facilitates TGF-β signaling. GENERAL SIGNIFICANCE Antibiotic analogs lacking ribotoxic effects are useful as pharmacological tools to study TGF-β signaling by controlling Ski and SnoN protein levels.
Collapse
Affiliation(s)
- Jacqueline Hernández-Damián
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México D.F., 04510 Mexico
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang W, Liu C, Wang Y, Cao L. Effects of the downregulation of SnoN expression on HepG2 cell proliferation and apoptosis. Mol Med Rep 2013; 7:1324-8. [PMID: 23446947 DOI: 10.3892/mmr.2013.1340] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2012] [Accepted: 01/31/2013] [Indexed: 11/06/2022] Open
Abstract
Ski‑novel protein (SnoN) is a proto‑oncogene that belongs to the Ski protein family and is involved in regulating processes such as cell proliferation and apoptosis. To investigate the role of SnoN in the proliferation and apoptosis of HepG2 cells, we downregulated its expression by the use of small interfering RNA (siRNA). Three fragments predicted to have RNAi capacity were designed and synthesized as the target siRNAs (siRNA‑A, ‑B and ‑C). Following transfection, inhibition efficiency was detected by reverse transcription PCR (RT‑PCR) and western blot analysis. The siRNA with the optimal inhibition efficiency was used for the cell proliferation and apoptosis analysis. Cell proliferation was analyzed by the Cell Counting Kit‑8 (CCK‑8) and cell apoptosis was investigated by flow cytometry. In our study, all three siRNAs efficiently inhibited SnoN expression, and siRNA‑C demonstrated the optimal inhibition efficiency. We found that following downregulation of SnoN expression, HepG2 cell proliferation was significantly inhibited (P<0.05), while HepG2 cell apoptosis was significantly increased (P<0.05). SnoN‑specific siRNA is capable of effectively inhibiting the expression of SnoN in human HepG2 cells, and the downregulation of SnoN expression induces growth inhibition and apoptosis.
Collapse
Affiliation(s)
- Wenqi Wang
- Department of Gastroenterology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, Shandong 250014, P.R. China.
| | | | | | | |
Collapse
|
23
|
Jahchan NS, Ouyang G, Luo K. Expression profiles of SnoN in normal and cancerous human tissues support its tumor suppressor role in human cancer. PLoS One 2013; 8:e55794. [PMID: 23418461 PMCID: PMC3572181 DOI: 10.1371/journal.pone.0055794] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 12/30/2012] [Indexed: 11/18/2022] Open
Abstract
SnoN is a negative regulator of TGF-β signaling and also an activator of the tumor suppressor p53 in response to cellular stress. Its role in human cancer is complex and controversial with both pro-oncogenic and anti-oncogenic activities reported. To clarify its role in human cancer and provide clinical relevance to its signaling activities, we examined SnoN expression in normal and cancerous human esophageal, ovarian, pancreatic and breast tissues. In normal tissues, SnoN is expressed in both the epithelium and the surrounding stroma at a moderate level and is predominantly cytoplasmic. SnoN levels in all tumor epithelia examined are lower than or similar to that in the matched normal samples, consistent with its anti-tumorigenic activity in epithelial cells. In contrast, SnoN expression in the stroma is highly upregulated in the infiltrating inflammatory cells in high-grade esophageal and ovarian tumor samples, suggesting that SnoN may potentially promote malignant progression through modulating the tumor microenvironment in these tumor types. The overall levels of SnoN expression in these cancer tissues do not correlate with the p53 status. However, in human cancer cell lines with amplification of the snoN gene, a strong correlation between increased SnoN copy number and inactivation of p53 was detected, suggesting that the tumor suppressor SnoN-p53 pathway must be inactivated, either through downregulation of SnoN or inactivation of p53, in order to allow cancer cell to proliferate and survive. These data strongly suggest that SnoN can function as a tumor suppressor at early stages of tumorigenesis in human cancer tissues.
Collapse
Affiliation(s)
- Nadine S. Jahchan
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Gaoliang Ouyang
- College of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
- * E-mail:
| |
Collapse
|
24
|
Namachivayam K, Blanco CL, MohanKumar K, Jagadeeswaran R, Vasquez M, McGill-Vargas L, Garzon SA, Jain SK, Gill RK, Freitag NE, Weitkamp JH, Seidner SR, Maheshwari A. Smad7 inhibits autocrine expression of TGF-β2 in intestinal epithelial cells in baboon necrotizing enterocolitis. Am J Physiol Gastrointest Liver Physiol 2013; 304:G167-80. [PMID: 23154975 PMCID: PMC3543645 DOI: 10.1152/ajpgi.00141.2012] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Preterm infants may be at risk of necrotizing enterocolitis (NEC) due to deficiency of transforming growth factor-β 2 (TGF-β(2)) in the developing intestine. We hypothesized that low epithelial TGF-β(2) expression in preterm intestine and during NEC results from diminished autocrine induction of TGF-β(2) in these cells. Premature baboons delivered at 67% gestation were treated per current norms for human preterm infants. NEC was diagnosed by clinical and radiological findings. Inflammatory cytokines, TGF-β(2), Smad7, Ski, and strawberry notch N (SnoN)/Ski-like oncoprotein (SKIL) was measured using quantitative reverse transcriptase-polymerase chain reaction, immunoblots, and immunohistochemistry. Smad7 effects were examined in transfected IEC6 intestinal epithelial cells in vitro. Findings were validated in archived human tissue samples of NEC. NEC was recorded in seven premature baboons. Consistent with existing human data, premature baboon intestine expressed less TGF-β(2) than term intestine. TGF-β(2) expression was regulated in epithelial cells in an autocrine fashion, which was interrupted in the premature intestine and during NEC due to increased expression of Smad7. LPS increased Smad7 binding to the TGF-β(2) promoter and was associated with dimethylation of the lysine H3K9, a marker of transcriptional silencing, on the nucleosome of TGF-β(2). Increased Smad7 expression in preterm intestine was correlated with the deficiency of SnoN/SKIL, a repressor of the Smad7 promoter. Smad7 inhibits autocrine expression of TGF-β(2) in intestinal epithelial cells in the normal premature intestine and during NEC. Increased Smad7 expression in the developing intestine may be due to a developmental deficiency of the SnoN/SKIL oncoprotein.
Collapse
Affiliation(s)
- Kopperuncholan Namachivayam
- Departments of Pediatrics (1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease),
| | - Cynthia L. Blanco
- 3Departments of Pediatrics (Division of Neonatology), University of Texas Health Sciences Center at San Antonio, San Antonio, Texas;
| | - Krishnan MohanKumar
- Departments of Pediatrics (1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease),
| | - Ramasamy Jagadeeswaran
- Departments of Pediatrics (1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease),
| | - Margarita Vasquez
- 3Departments of Pediatrics (Division of Neonatology), University of Texas Health Sciences Center at San Antonio, San Antonio, Texas;
| | - Lisa McGill-Vargas
- 3Departments of Pediatrics (Division of Neonatology), University of Texas Health Sciences Center at San Antonio, San Antonio, Texas;
| | - Steven A. Garzon
- 2Center for Neonatal and Pediatric Gastrointestinal Disease), ,4Pathology,
| | - Sunil K. Jain
- 5Department of Pediatrics (Division of Neonatology), University of Texas Medical Branch, Galveston, Texas; and
| | - Ravinder K. Gill
- 2Center for Neonatal and Pediatric Gastrointestinal Disease), ,6Medicine (Section of Digestive Diseases and Nutrition),
| | | | - Jörn-Hendrik Weitkamp
- 8Department of Pediatrics (Division of Neonatology), Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Steven R. Seidner
- 3Departments of Pediatrics (Division of Neonatology), University of Texas Health Sciences Center at San Antonio, San Antonio, Texas;
| | - Akhil Maheshwari
- Departments of Pediatrics (1Division of Neonatology and ,2Center for Neonatal and Pediatric Gastrointestinal Disease), ,9Pharmacology, University of Illinois at Chicago, Chicago, Illinois;
| |
Collapse
|
25
|
Pan D, Zhu Q, Conboy MJ, Conboy IM, Luo K. SnoN activates p53 directly to regulate aging and tumorigenesis. Aging Cell 2012; 11:902-911. [PMID: 22805162 DOI: 10.1111/j.1474-9726.2012.00857.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
We have identified SnoN as a direct activator of p53 to accelerate aging and inhibit tumorigenesis. SnoN has been shown previously to promote proliferation and transformation by antagonizing TGFβ signaling. We show that elimination of this TGFβ antagonistic activity of SnoN in vivo results in accelerated aging and resistance to tumorigenesis. The SnoN knockin mice display a shortened lifespan, decreased reproductivity, osteoporosis, reduced regenerative capacity, and other aging phenotypes, similar to that found in mice expressing an active p53. These activities of SnoN rely on the ability of SnoN to activate p53. SnoN can bind directly to p53 and compete with Mdm2 for binding to p53, preventing p53 ubiquitination and degradation and additionally facilitating p53 acetylation and phosphorylation. SnoN also binds to p53 on the promoter of p53 responsive genes to promote transcription activation. This activation of p53 by SnoN is necessary for its antitumorigenic and progeria activities in vivo because elimination of one copy of p53 reverses the aging phenotypes and accelerates tumorigenesis. Thus, we have revealed a novel function of SnoN in regulating aging and tumorigenesis by directly activating p53.
Collapse
Affiliation(s)
- Deng Pan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Michael J Conboy
- Department of Bioengineering and QB3 University of California, Berkeley, CA 94720
| | - Irina M Conboy
- Department of Bioengineering and QB3 University of California, Berkeley, CA 94720
| | - Kunxin Luo
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| |
Collapse
|
26
|
Takaesu NT, Stinchfield MJ, Shimizu K, Arase M, Quijano JC, Watabe T, Miyazono K, Newfeld SJ. Drosophila CORL is required for Smad2-mediated activation of Ecdysone Receptor expression in the mushroom body. Development 2012; 139:3392-401. [PMID: 22874913 DOI: 10.1242/dev.079442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
CORL proteins (FUSSEL/SKOR proteins in humans) are related to Sno/Ski oncogenes but their developmental roles are unknown. We have cloned Drosophila CORL and show that its expression is restricted to distinct subsets of cells in the central nervous system. We generated a deletion of CORL and noted that homozygous individuals rarely survive to adulthood. Df(4)dCORL adult escapers display mushroom body (MB) defects and Df(4)dCORL larvae are lacking Ecdysone Receptor (EcR-B1) expression in MB neurons. This is phenocopied in CORL-RNAi and Smad2-RNAi clones in wild-type larvae. Furthermore, constitutively active Baboon (type I receptor upstream of Smad2) cannot stimulate EcR-B1 MB expression in Df(4)dCORL larvae, which demonstrates a formal requirement for CORL in Smad2 signaling. Studies of mouse Corl1 (Skor1) revealed that it binds specifically to Smad3. Overall, the data suggest that CORL facilitates Smad2 activity upstream of EcR-B1 in the MB. The conservation of neural expression and strong sequence homology of all CORL proteins suggests that this is a new family of Smad co-factors.
Collapse
Affiliation(s)
- Norma T Takaesu
- School of Life Sciences, Arizona State University, Tempe, AZ 85287-4501, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jahchan NS, Wang D, Bissell MJ, Luo K. SnoN regulates mammary gland alveologenesis and onset of lactation by promoting prolactin/Stat5 signaling. Development 2012; 139:3147-56. [PMID: 22833129 DOI: 10.1242/dev.079616] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Mammary epithelial cells undergo structural and functional differentiation at late pregnancy and parturition to produce and secrete milk. Both TGF-β and prolactin pathways are crucial regulators of this process. However, how the activities of these two antagonistic pathways are orchestrated to initiate lactation has not been well defined. Here, we show that SnoN, a negative regulator of TGF-β signaling, coordinates TGF-β and prolactin signaling to control alveologenesis and lactogenesis. SnoN expression is induced at late pregnancy by the coordinated actions of TGF-β and prolactin. The elevated SnoN promotes Stat5 signaling by enhancing its stability, thereby sharply increasing the activity of prolactin signaling at the onset of lactation. SnoN-/- mice display severe defects in alveologenesis and lactogenesis, and mammary epithelial cells from these mice fail to undergo proper morphogenesis. These defects can be rescued by an active Stat5. Thus, our study has identified a new player in the regulation of milk production and revealed a novel function of SnoN in mammary alveologenesis and lactogenesis in vivo through promotion of Stat5 signaling.
Collapse
Affiliation(s)
- Nadine S Jahchan
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | |
Collapse
|
28
|
Liu R, Wang Y, Xiao Y, Shi M, Zhang G, Guo B. SnoN as a key regulator of the high glucose-induced epithelial-mesenchymal transition in cells of the proximal tubule. Kidney Blood Press Res 2012; 35:517-28. [PMID: 22813962 DOI: 10.1159/000339172] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 04/27/2012] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS Ski-related protein N (SnoN) suppression is essential to transforming growth factor-β1 induction and the epithelial-mesenchymal transition (EMT) in several cancer cells. The role of SnoN in diabetic nephropathy is unknown. We aimed to determine the role of SnoN in the EMT of proximal tubule cells (PTCs) maintained under high glucose conditions. METHODS Immunohistochemistry, immunocytochemistry, Western blotting, small interfering RNA gene silencing, viral transduction and RT-PCR were used to assess changes in SnoN, E-cadherin, cytokeratin-18, α-smooth muscle actin and fibronectin expression using an in vivo streptozotocin-induced rat diabetic nephropathy model, and PTCs exposed to high glucose (25 mmol/l). RESULTS High glucose induced EMT in vitro and in vivo. Exposure of PTCs to a high concentration of glucose suppressed SnoN expression in a time-dependent manner compared with normal glucose and high osmolarity-treated groups. SnoN gene silencing under high glucose conditions appears to enhance the transition of PTC phenotype. Conversely, ectopic expression of exogenous SnoN after transfection conferred tubular epithelial cell resistance to high glucose-induced EMT. CONCLUSION SnoN plays a negative role in high glucose-induced EMT in PTCs. The effect of SnoN downregulation in vivo and in vitro suggests that SnoN may be a potential therapeutic target.
Collapse
Affiliation(s)
- Ruixia Liu
- Department of Pathophysiology, Guiyang Medical University, Guiyang, China
| | | | | | | | | | | |
Collapse
|
29
|
Zhu Q, Luo K. SnoN in regulation of embryonic development and tissue morphogenesis. FEBS Lett 2012; 586:1971-6. [PMID: 22710172 DOI: 10.1016/j.febslet.2012.03.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/03/2012] [Accepted: 03/05/2012] [Indexed: 01/15/2023]
Abstract
SnoN (Ski-novel protein) plays an important role in embryonic development, tumorigenesis and aging. Past studies largely focused on its roles in tumorigenesis. Recent studies of its expression patterns and functions in mouse models and mammalian cells have revealed that SnoN interacts with multiple signaling molecules at different cellular levels to modulate the activities of several signaling pathways in a tissue context and developmental stage dependent manner. These studies suggest that SnoN may have broad functions in the embryonic development and tissue morphogenesis.
Collapse
Affiliation(s)
- Qingwei Zhu
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
30
|
Defining the earliest transcriptional steps of chondrogenic progenitor specification during the formation of the digits in the embryonic limb. PLoS One 2011; 6:e24546. [PMID: 21931747 PMCID: PMC3172225 DOI: 10.1371/journal.pone.0024546] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Accepted: 08/12/2011] [Indexed: 12/12/2022] Open
Abstract
The characterization of genes involved in the formation of cartilage is of key importance to improve cell-based cartilage regenerative therapies. Here, we have developed a suitable experimental model to identify precocious chondrogenic events in vivo by inducing an ectopic digit in the developing embryo. In this model, only 12 hr after the implantation of a Tgfβ bead, in the absence of increased cell proliferation, cartilage forms in undifferentiated interdigital mesoderm and in the course of development, becomes a structurally and morphologically normal digit. Systematic quantitative PCR expression analysis, together with other experimental approaches allowed us to establish 3 successive periods preceding the formation of cartilage. The “pre-condensation stage”, occurring within the first 3 hr of treatment, is characterized by the activation of connective tissue identity transcriptional factors (such as Sox9 and Scleraxis) and secreted factors (such as Activin A and the matricellular proteins CCN-1 and CCN-2) and the downregulation of the galectin CG-8. Next, the “condensation stage” is characterized by intense activation of Smad 1/5/8 BMP-signaling and increased expression of extracellular matrix components. During this period, the CCN matricellular proteins promote the expression of extracellular matrix and cell adhesion components. The third period, designated the “pre-cartilage period”, precedes the formation of molecularly identifiable cartilage by 2–3 hr and is characterized by the intensification of Sox 9 gene expression, along with the stimulation of other pro-chondrogenic transcription factors, such as HifIa. In summary, this work establishes a temporal hierarchy in the regulation of pro-chondrogenic genes preceding cartilage differentiation and provides new insights into the relative roles of secreted factors and cytoskeletal regulators that direct the first steps of this process in vivo.
Collapse
|
31
|
Bonnon C, Atanasoski S. c-Ski in health and disease. Cell Tissue Res 2011; 347:51-64. [DOI: 10.1007/s00441-011-1180-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/15/2011] [Indexed: 01/28/2023]
|
32
|
Band AM, Laiho M. Crosstalk of TGF-β and estrogen receptor signaling in breast cancer. J Mammary Gland Biol Neoplasia 2011; 16:109-15. [PMID: 21390570 DOI: 10.1007/s10911-011-9203-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 02/28/2011] [Indexed: 01/17/2023] Open
Abstract
Estrogen receptor-α (ERα) and transforming growth factor (TGF)-β signaling pathways are major regulators during mammary gland development, function and tumorigenesis. Predominantly, they have opposing roles in proliferation and apoptosis. While ERα signaling supports growth and differentiation and is antiapoptotic, mammary gland epithelia cells are very sensitive to TGF-β-induced cell cycle arrest and apoptosis. Their regulatory pathways intersect, and ERα blocks TGF-β pathway by multiple means, including direct interactions of its signaling components, Smads. However, relatively little is known of the dysfunction of their interactions in cancer. A better understanding would help to develop new strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Arja M Band
- Molecular Cancer Biology Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|