1
|
Liu B, Wang P, Lv X. Phytol-mixed micelles alleviate dexamethasone-induced osteoporosis in zebrafish: Activation of the MMP3-OPN-MAPK pathway-mediating bone remodeling. Open Life Sci 2025; 20:20221015. [PMID: 40129470 PMCID: PMC11931661 DOI: 10.1515/biol-2022-1015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 03/26/2025] Open
Abstract
This research investigates the therapeutic efficacy of phytol-mixed micelles in mitigating dexamethasone (Dex)-induced osteoporosis in zebrafish, with a particular focus on scale regeneration. Osteoporosis was induced in zebrafish through exposure to Dex, and the effects of phytol-mixed micelles were evaluated in this model. Following phytol therapy, bone mineralization was assessed using calcium, phosphorus, and alizarin red staining tests. Additionally, commercially available kits quantified the levels of tartrate-resistant acid phosphatase (TRAP), hydroxyproline (HP), and alkaline phosphatase (ALP). The mRNA expression levels of MMP3, osteopontin (OPN), and mitogen-activated protein kinase (MAPK) were examined using reverse transcription polymerase chain reaction (RT-PCR). The findings indicated that phytol significantly increased calcium and phosphorus concentrations. Phytol-mixed micelle therapy led to increased calcium deposition and enhanced bone formation, as evidenced by alizarin red staining. Moreover, phytol administration resulted in increased HP content and upregulated ALP and TRAP activities in zebrafish. RT-PCR tests demonstrated that phytol plays a role in the restoration of the MMP3-OPN-MAPK pathway. In summary, this research highlights the potential of phytol-mixed micelles in ameliorating Dex-induced osteoporosis in zebrafish. Clarifying phytol's mechanism, particularly its stimulation of the MMP3-OPN-MAPK pathway, provides insight into its role in facilitating bone remodeling.
Collapse
Affiliation(s)
- Bo Liu
- Department of Trauma Surgery, Hebei Port Group Co LTD. Qinhuangdao Hospital of Integrated Chinese and Western Medicine, Qinhuangdao, Hebei, 066003, China
| | - Peng Wang
- Department of Spine Surgery, Shengli Oilfield Central Hospital,
Dongying, Shandong, 257000, China
| | - Xiangyang Lv
- Department of Orthopedics, Xi’an Qinhuang Hospital, Xi’an, Shaanxi, 710600, China
| |
Collapse
|
2
|
Sun P, Yang L, Yu K, Wang J, Chao J. Scaffold Proteins in Fibrotic Diseases of Visceral Organs. Biomolecules 2025; 15:420. [PMID: 40149956 PMCID: PMC11940551 DOI: 10.3390/biom15030420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 03/29/2025] Open
Abstract
Fibrosis, characterized by excessive extracellular matrix (ECM) deposition, disrupts tissue architecture and impairs organ function, ultimately leading to severe health consequences and even failure of vital organs such as the lung, heart, liver, and kidney. Despite significant advances in understanding the molecular mechanisms underlying fibrosis, effective therapeutic options remain limited. Emerging evidence highlights scaffold proteins as critical regulators in the progression of fibrosis. These multifunctional proteins serve as molecular platforms that organize and coordinate key signaling pathways-including those governing ECM remodeling, cytoskeletal organization, and cell migration-thereby integrating both profibrotic and antifibrotic signals. Their pivotal role in linking mechanotransduction, inflammatory, and developmental signals offers a unique therapeutic window, as targeted interventions (e.g., small-molecule inhibitors, peptides, biologics, and gene therapy) are emerging to modulate these pathways. This review synthesizes recent findings on scaffold protein functions across multiple organs and discusses novel therapeutic strategies to manage and potentially reverse fibrosis.
Collapse
Affiliation(s)
| | | | | | | | - Jie Chao
- Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
| |
Collapse
|
3
|
Peng Y, Xiong R, Wang B, Chen X, Ning Y, Zhao Y, Yang N, Zhang J, Li C, Zhou Y, Li P. The Essential Role of Angiogenesis in Adenosine 2A Receptor Deficiency-mediated Impairment of Wound Healing Involving c-Ski via the ERK/CREB Pathways. Int J Biol Sci 2024; 20:4532-4550. [PMID: 39247808 PMCID: PMC11380447 DOI: 10.7150/ijbs.98856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 08/07/2024] [Indexed: 09/10/2024] Open
Abstract
Adenosine receptor-mediated signaling, especially adenosine A2A receptor (A2AR) signaling, has been implicated in wound healing. However, the role of endothelial cells (ECs) in A2AR-mediated wound healing and the mechanism underlying this effect are still unclear. Here, we showed that the expression of A2AR substantially increased after wounding and was especially prominent in granulation tissue. The delaying effects of A2AR knockout (KO) on wound healing are due mainly to the effect of A2AR on endothelial cells, as shown with A2AR-KO and EC-A2AR-KO mice. Moreover, the expression of c-Ski, which is especially prominent in CD31-positive cells in granulation tissue, increased after wounding and was decreased by both EC-A2AR KO and A2AR KO. In human microvascular ECs (HMECs), A2AR activation induced EC proliferation, migration, tubule formation and c-Ski expression, whereas c-Ski depletion by RNAi abolished these effects. Mechanistically, A2AR activation promotes the expression of c-Ski through an ERK/CREB-dependent pathway. Thus, A2AR-mediated angiogenesis plays a critical role in wound healing, and c-Ski is involved mainly in the regulation of angiogenesis by A2AR via the ERK/CREB pathway. These findings identify A2AR as a therapeutic target in wound repair and other angiogenesis-dependent tissue repair processes.
Collapse
Affiliation(s)
- Yan Peng
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Renping Xiong
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Bo Wang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Xing Chen
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Yalei Ning
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Yan Zhao
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Nan Yang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Jing Zhang
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Changhong Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Yuanguo Zhou
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| | - Ping Li
- Department of Army Occupational Disease, State Key Laboratory of Trauma, Burn and Combined Injury, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang Zhilu, Chongqing 400042, People's Republic of China
| |
Collapse
|
4
|
Zhai Y, Ye SY, Wang QS, Xiong RP, Fu SY, Du H, Xu YW, Peng Y, Huang ZZ, Yang N, Zhao Y, Ning YL, Li P, Zhou YG. Overexpressed ski efficiently promotes neurorestoration, increases neuronal regeneration, and reduces astrogliosis after traumatic brain injury. Gene Ther 2023; 30:75-87. [PMID: 35132206 DOI: 10.1038/s41434-022-00320-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 12/31/2021] [Accepted: 01/20/2022] [Indexed: 11/09/2022]
Abstract
Traumatic brain injury (TBI) survivors suffer from long-term disability and neuropsychiatric sequelae due to irreparable brain tissue destruction. However, there are still few efficient therapies to promote neurorestoration in damaged brain tissue. This study aimed to investigate whether the pro-oncogenic gene ski can promote neurorestoration after TBI. We established a ski-overexpressing experimental TBI mouse model using adenovirus-mediated overexpression through immediate injection after injury. Hematoxylin-eosin staining, MRI-based 3D lesion volume reconstruction, neurobehavioral tests, and analyses of neuronal regeneration and astrogliosis were used to assess neurorestorative efficiency. The effects of ski overexpression on the proliferation of cultured immature neurons and astrocytes were evaluated using imaging flow cytometry. The Ski protein level increased in the perilesional region at 3 days post injury. ski overexpression further elevated Ski protein levels up to 14 days post injury. Lesion volume was attenuated by approximately 36-55% after ski overexpression, with better neurobehavioral recovery, more newborn immature and mature neurons, and less astrogliosis in the perilesional region. Imaging flow cytometry results showed that ski overexpression elevated the proliferation rate of immature neurons and reduced the proliferation rate of astrocytes. These results show that ski can be considered a novel neurorestoration-related gene that effectively promotes neurorestoration, facilitates neuronal regeneration, and reduces astrogliosis after TBI.
Collapse
Affiliation(s)
- Yu Zhai
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Shi-Yang Ye
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Qiu-Shi Wang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.,Department of Pathology, Research Institute of Surgery and Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ren-Ping Xiong
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Sheng-Yu Fu
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Hao Du
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ya-Wei Xu
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Yan Peng
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Zhi-Zhong Huang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Nan Yang
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Yan Zhao
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ya-Lei Ning
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China
| | - Ping Li
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.
| | - Yuan-Guo Zhou
- The Molecular Biology Centre, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital, Army Medical University (The Third Military Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
5
|
Park M, Singh S, Khan SR, Abrar MA, Grisanti F, Rahman MS, Samee MAH. Multinomial Convolutions for Joint Modeling of Regulatory Motifs and Sequence Activity Readouts. Genes (Basel) 2022; 13:genes13091614. [PMID: 36140783 PMCID: PMC9498894 DOI: 10.3390/genes13091614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
A common goal in the convolutional neural network (CNN) modeling of genomic data is to discover specific sequence motifs. Post hoc analysis methods aid in this task but are dependent on parameters whose optimal values are unclear and applying the discovered motifs to new genomic data is not straightforward. As an alternative, we propose to learn convolutions as multinomial distributions, thus streamlining interpretable motif discovery with CNN model fitting. We developed MuSeAM (Multinomial CNNs for Sequence Activity Modeling) by implementing multinomial convolutions in a CNN model. Through benchmarking, we demonstrate the efficacy of MuSeAM in accurately modeling genomic data while fitting multinomial convolutions that recapitulate known transcription factor motifs.
Collapse
Affiliation(s)
- Minjun Park
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Salvi Singh
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Samin Rahman Khan
- Computer Science and Engineering, Bangladesh University of Engineering and Technology, Dhaka 1000, Bangladesh
| | | | - Francisco Grisanti
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - M. Sohel Rahman
- Computer Science and Engineering, Bangladesh University of Engineering and Technology, Dhaka 1000, Bangladesh
| | - Md. Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
6
|
Smad-dependent pathways in the infarcted and failing heart. Curr Opin Pharmacol 2022; 64:102207. [DOI: 10.1016/j.coph.2022.102207] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/10/2022] [Accepted: 02/22/2022] [Indexed: 02/08/2023]
|
7
|
Norollahi SE, Foumani MG, Pishkhan MK, Shafaghi A, Alipour M, Jamkhaneh VB, Marghoob MN, Vahidi S. DNA Methylation Profiling of MYC, SMAD2/3 and DNMT3A in Colorectal Cancer. Oman Med J 2021; 36:e315. [PMID: 34804598 PMCID: PMC8581152 DOI: 10.5001/omj.2020.93] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/04/2020] [Indexed: 11/18/2022] Open
Abstract
Epigenetic modifications, particularly DNA methylation, is commonplace and a remarkable factor in carcinogenesis transformation. Conspicuously, previous findings have presented a cluster of irregular promoter methylation alterations related with silencing of tumor suppressor genes, little is accepted regarding their sequential DNA methylation (hypo and hyper) modifications during the cancer progression. In this way, fluctuations of DNA methylation of many genes, especially MYC, SMAD2/3, and DNMT3A, have an impressive central key role in many different cancers, including colorectal cancer (CRC). CRC is distinguished by DNA methylation, which is related to tumorigenesis and also genomic instability. Importantly, molecular heterogeneity between multiple adenomas in different patients with CRC may show diverse developmental phenotypes for these kinds of tumors. Conclusively, studying factors that are involved in CRC carcinogenesis, especially the alterations in epigenetic elements, such as DNA methylation besides RNA remodeling, and histone modification, acetylation and phosphorylation, can be influential to find new therapeutic and diagnostic biomarkers in this type of malignancy. In this account, we discuss and address the potential significant methylated modifications of these genes and their importance during the development of CRC carcinogenesis.
Collapse
Affiliation(s)
- Seyedeh Elham Norollahi
- Cancer Research Center and Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
| | | | | | - Afshin Shafaghi
- Faculty of Medicine, Department of Gastroenterology, Guilan University of Medical Sciences, Rasht, Iran
| | - Majid Alipour
- Department of Biology, Islamic Azad University of Babol Branch, Babol, Iran
| | - Vida Baloui Jamkhaneh
- Department of Veterinary Medicine, Islamic Azad University of Babol Branch, Babol, Iran
| | - Mohammad Namayan Marghoob
- Department of Microbiology, Faculty of Biological Sciences, Shahid Beheshti University, Tehran, Iran.,Gastroenterology and Liver Disease Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sogand Vahidi
- Clinical Research Development Unit of Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
8
|
Hara T, Sakuma M, Fujie T, Kaji T, Yamamoto C. Cadmium induces plasminogen activator inhibitor-1 via Smad2/3 signaling pathway in human endothelial EA.hy926 cells. J Toxicol Sci 2021; 46:249-253. [PMID: 33952801 DOI: 10.2131/jts.46.249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Modulation of the blood coagulation fibrinolytic system is an essential function of vascular endothelial cells. Tissue plasminogen activator (t-PA) and plasminogen activator inhibitor-1 (PAI-1) are major fibrinolytic regulatory proteins synthesized by vascular endothelial cells; fibrinolytic activity is dependent on the balance between these proteins. Previously, we have reported that cadmium, an initiator of ischemic heart disease, induces PAI-1 expression and suppresses fibrinolytic activity in cultured human vascular endothelial cells. However, the key molecules involved in cadmium-induced PAI-1 induction remain unclear. Herein, we investigated the contribution of Smad2 and Smad3, transcriptional factors involved in PAI-1 induction via transforming growth factor-β, using the human vascular endothelial cell line EA.hy926 cells in culture. Our findings indicated that cadmium induces PAI-1 expression without affecting t-PA expression up to 20 µM, a non-cytotoxic concentration, and PAI-1 induction by cadmium is partly mediated via Smad2 and Smad3. This study provides a possible mechanism underlying cadmium-induced vascular disorders.
Collapse
Affiliation(s)
- Takato Hara
- Faculty of Pharmaceutical Sciences, Toho University
| | - Miki Sakuma
- Faculty of Pharmaceutical Sciences, Toho University
| | - Tomoya Fujie
- Faculty of Pharmaceutical Sciences, Toho University
| | - Toshiyuki Kaji
- Faculty of Pharmaceutical Sciences, Tokyo University of Science
| | | |
Collapse
|
9
|
de Oliveira Camargo R, Abual'anaz B, Rattan SG, Filomeno KL, Dixon IMC. Novel factors that activate and deactivate cardiac fibroblasts: A new perspective for treatment of cardiac fibrosis. Wound Repair Regen 2021; 29:667-677. [PMID: 34076932 DOI: 10.1111/wrr.12947] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
Heart disease with attendant cardiac fibrosis kills more patients in developed countries than any other disease, including cancer. We highlight the recent literature on factors that activate and also deactivate cardiac fibroblasts. Activation of cardiac fibroblasts results in myofibroblasts phenotype which incorporates aSMA to stress fibres, express ED-A fibronectin, elevated PDGFRα and are hypersecretory ECM components. These cells facilitate both acute wound healing (infarct site) and chronic cardiac fibrosis. Quiescent fibroblasts are associated with normal myocardial tissue and provide relatively slow turnover of the ECM. Deactivation of activated myofibroblasts is a much less studied phenomenon. In this context, SKI is a known negative regulator of TGFb1 /Smad signalling, and thus may share functional similarity to PPARγ activation. The discovery of SKI's potent anti-fibrotic role, and its ability to deactivate and/or myofibroblasts is featured and contrasted with PPARγ. While myofibroblasts are typically recruited from pools of potential precursor cells in a variety of organs, the importance of activation of resident cardiac fibroblasts has been recently emphasised. Myofibroblasts deposit ECM components at an elevated rate and contribute to both systolic and diastolic dysfunction with attendant cardiac fibrosis. A major knowledge gap exists as to specific proteins that may signal for fibroblast deactivation. As SKI may be a functionally pluripotent protein, we suggest that it serves as a scaffold to proteins other than R-Smads and associated Smad signal proteins, and thus its anti-fibrotic effects may extend beyond binding R-Smads. While cardiac fibrosis is causal to heart failure, the treatment of cardiac fibrosis is hampered by the lack of availability of effective pharmacological anti-fibrotic agents. The current review will provide an overview of work highlighting novel factors which cause fibroblast activation and deactivation to underscore putative therapeutic avenues for improving disease outcomes in cardiac patients with fibrosed hearts.
Collapse
Affiliation(s)
- Rebeca de Oliveira Camargo
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Besher Abual'anaz
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Sunil G Rattan
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Krista L Filomeno
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada
| | - Ian M C Dixon
- Institute of Cardiovascular Sciences, Albrechtsen Research Centre, Winnipeg, Canada.,Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Canada.,Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
10
|
Yap JKY, Pickard BS, Gan SY, Chan EWL. Genes associated with amyloid-beta-induced inflammasome-mediated neuronal death identified using functional gene trap mutagenesis approach. Int J Biochem Cell Biol 2021; 136:106014. [PMID: 34022435 DOI: 10.1016/j.biocel.2021.106014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 02/18/2021] [Accepted: 05/11/2021] [Indexed: 01/15/2023]
Abstract
Alzheimer's disease is an irreversible neurodegenerative disease, which accounts for most dementia cases. Neuroinflammation is increasingly recognised for its roles in Alzheimer's disease pathogenesis which, in part, links amyloid-beta to neuronal death. Neuroinflammatory signalling can be exhibited by neurons themselves, potentially leading to widespread neuronal cell death, although neuroinflammation is commonly associated with glial cells. The presence of the inflammasomes such as nucleotide-binding leucine-rich repeat receptors protein 1 in neurons accelerates amyloid-beta -induced neuroinflammation and has been shown to trigger neuronal pyroptosis in murine Alzheimer's disease models. However, the pathways involved in amyloid-beta activation of inflammasomes have yet to be elucidated. In this study, a gene trap mutagenesis approach was utilised to resolve the genes functionally involved in inflammasome signalling within neurons, and the mechanism behind amyloid-beta-induced neuronal death. The results indicate that amyloid-beta significantly accelerated neuroinflammatory cell death in the presence of a primed inflammasome (the NLR family pyrin domain-containing 1). The mutagenesis screen discovered the atypical mitochondrial Ras homolog family member T1 as a significant contributor to amyloid-beta-induced inflammasome -mediated neuronal death. The mutagenesis screen also identified two genes involved in transforming growth factor beta signalling, namely Transforming Growth Factor Beta Receptor 1 and SNW domain containing 1. Additionally, a gene associated with cytoskeletal reorganisation, SLIT-ROBO Rho GTPase Activating Protein 3 was found to be neuroprotective. In conclusion, these genes could play important roles in inflammasome signalling in neurons, which makes them promising therapeutic targets for future drug development against neuroinflammation in Alzheimer's disease.
Collapse
Affiliation(s)
- Jeremy Kean Yi Yap
- School of Postgraduate Studies, International Medical University, Kuala Lumpur, Malaysia
| | - Benjamin Simon Pickard
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, United Kingdom
| | - Sook Yee Gan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Elaine Wan Ling Chan
- Institute for Research, Development and Innovation, International Medical University, Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
SKI activates the Hippo pathway via LIMD1 to inhibit cardiac fibroblast activation. Basic Res Cardiol 2021; 116:25. [PMID: 33847835 PMCID: PMC8043893 DOI: 10.1007/s00395-021-00865-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 03/24/2021] [Indexed: 01/14/2023]
Abstract
We have previously shown that overexpression of SKI, an endogenous TGF-β1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-β signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI’s effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI’s capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.
Collapse
|
12
|
Abdel Mouti M, Pauklin S. TGFB1/INHBA Homodimer/Nodal-SMAD2/3 Signaling Network: A Pivotal Molecular Target in PDAC Treatment. Mol Ther 2021; 29:920-936. [PMID: 33429081 PMCID: PMC7934636 DOI: 10.1016/j.ymthe.2021.01.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/17/2020] [Accepted: 01/02/2021] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer remains a grueling disease that is projected to become the second-deadliest cancer in the next decade. Standard treatment of pancreatic cancer is chemotherapy, which mainly targets the differentiated population of tumor cells; however, it paradoxically sets the roots of tumor relapse by the selective enrichment of intrinsically chemoresistant pancreatic cancer stem cells that are equipped with an indefinite capacity for self-renewal and differentiation, resulting in tumor regeneration and an overall anemic response to chemotherapy. Crosstalk between pancreatic tumor cells and the surrounding stromal microenvironment is also involved in the development of chemoresistance by creating a supportive niche, which enhances the stemness features and tumorigenicity of pancreatic cancer cells. In addition, the desmoplastic nature of the tumor-associated stroma acts as a physical barrier, which limits the intratumoral delivery of chemotherapeutics. In this review, we mainly focus on the transforming growth factor beta 1 (TGFB1)/inhibin subunit beta A (INHBA) homodimer/Nodal-SMAD2/3 signaling network in pancreatic cancer as a pivotal central node that regulates multiple key mechanisms involved in the development of chemoresistance, including enhancement of the stem cell-like properties and tumorigenicity of pancreatic cancer cells, mediating cooperative interactions between pancreatic cancer cells and the surrounding stroma, as well as regulating the deposition of extracellular matrix proteins within the tumor microenvironment.
Collapse
Affiliation(s)
- Mai Abdel Mouti
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Headington, University of Oxford, Oxford OX3 7LD, UK
| | - Siim Pauklin
- Botnar Research Centre, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Headington, University of Oxford, Oxford OX3 7LD, UK.
| |
Collapse
|
13
|
Wang F, Fan K, Zhao Y, Xie ML. Apigenin attenuates TGF-β1-stimulated cardiac fibroblast differentiation and extracellular matrix production by targeting miR-155-5p/c-Ski/Smad pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 265:113195. [PMID: 32800930 DOI: 10.1016/j.jep.2020.113195] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/06/2020] [Accepted: 07/15/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Apigenin is a natural flavonoid compound present in chamomile (Matricaia chamomilla L.) from the Asteraceae family, which is used in the treatment of cardiovascular diseases by traditional healers, but its effects on differentiation and extracellular matrix (ECM) production of cardiac fibroblasts (CFs) induced by transforming growth factor beta 1 (TGF-β1) are poorly understood. AIM OF THE STUDY This study aimed to examine these effects and potential molecular mechanisms and to provide a new application of apigenin in the prevention and treatment of cardiac fibrosis. MATERIALS AND METHODS The TGF-β1-stimulated CFs or the combination of TGF-β1-stimulated and microRNA-155-5p (miR-155-5p) inhibitor- or mimic-transfected CFs were treated with or without apigenin. The expression levels of intracellular related mRNA and proteins were detected by real-time polymerase chain reaction and Western blot methods, respectively. The luciferase reporter gene containing cellular Sloan-Kettering Institute (c-Ski) wild or mutant type 3'-UTR was used and the luciferase activity was examined to verify the direct link of miR-155-5p and c-Ski. RESULTS After treatment of TGF-β1-stimulated CFs with 6-24 μM apigenin, the expression of c-Ski was increased, while levels of miR-155-5p, α-smooth muscle actin, collagen Ⅰ/Ⅲ, Smad2/3, and p-Smad2/3 were decreased. After transfection of CFs with the miR-155-5p inhibitor or mimic, the similar or inverse results were respectively observed as well. The combination of TGF-β1 and miR-155-5p inhibitor or mimic might cause an antagonistical or synergistic effect, respectively, and apigenin addition could enhance the effects of the inhibitor and antagonize the effects of the mimic. Luciferase reporter gene assay demonstrated that c-Ski was a direct target of miR-155-5p. CONCLUSION These findings suggested that apigenin could inhibit the differentiation and ECM production in TGF-β1-stimulated CFs, and its mechanisms might partly be attributable to the reduction of miR-155-5p expression and subsequent increment of c-Ski expression, which might result in the inhibition of Smad2/3 and p-Smad2/3 expressions.
Collapse
Affiliation(s)
- Feng Wang
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Ke Fan
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Ying Zhao
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu Province, China
| | - Mei-Lin Xie
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, Jiangsu Province, China.
| |
Collapse
|
14
|
Landry NM, Dixon IMC. Fibroblast mechanosensing, SKI and Hippo signaling and the cardiac fibroblast phenotype: Looking beyond TGF-β. Cell Signal 2020; 76:109802. [PMID: 33017619 DOI: 10.1016/j.cellsig.2020.109802] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022]
Abstract
Cardiac fibroblast activation to hyper-synthetic myofibroblasts following a pathological stimulus or in response to a substrate with increased stiffness may be a key tipping point for the evolution of cardiac fibrosis. Cardiac fibrosis per se is associated with progressive loss of heart pump function and is a primary contributor to heart failure. While TGF-β is a common cytokine stimulus associated with fibroblast activation, a druggable target to quell this driver of fibrosis has remained an elusive therapeutic goal due to its ubiquitous use by different cell types and also in the signaling complexity associated with SMADs and other effector pathways. More recently, mechanical stimulus of fibroblastic cells has been revealed as a major point of activation; this includes cardiac fibroblasts. Further, the complexity of TGF-β signaling has been offset by the discovery of members of the SKI family of proteins and their inherent anti-fibrotic properties. In this respect, SKI is a protein that may bind a number of TGF-β associated proteins including SMADs, as well as signaling proteins from other pathways, including Hippo. As SKI is also known to directly deactivate cardiac myofibroblasts to fibroblasts, this mode of action is a putative candidate for further study into the amelioration of cardiac fibrosis. Herein we provide a synthesis of this topic and highlight novel candidate pathways to explore in the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Natalie M Landry
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada.
| |
Collapse
|
15
|
Cappelli C, Sepulveda H, Rivas S, Pola V, Urzúa U, Donoso G, Sagredo E, Carrero D, Casanova-Ortiz E, Sagredo A, González M, Manterola M, Nardocci G, Armisén R, Montecino M, Marcelain K. Ski Is Required for Tri-Methylation of H3K9 in Major Satellite and for Repression of Pericentromeric Genes: Mmp3, Mmp10 and Mmp13, in Mouse Fibroblasts. J Mol Biol 2020; 432:3222-3238. [PMID: 32198114 DOI: 10.1016/j.jmb.2020.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/23/2020] [Accepted: 03/11/2020] [Indexed: 11/27/2022]
Abstract
Several mechanisms directing a rapid transcriptional reactivation of genes immediately after mitosis have been described. However, little is known about the maintenance of repressive signals during mitosis. In this work, we address the role of Ski in the repression of gene expression during M/G1 transition in mouse embryonic fibroblasts (MEFs). We found that Ski localises as a distinct pair of dots at the pericentromeric region of mitotic chromosomes, and the absence of the protein is related to high acetylation and low tri-methylation of H3K9 in pericentromeric major satellite. Moreover, differential expression assays in early G1 cells showed that the presence of Ski is significantly associated with repression of genes localised nearby to pericentromeric DNA. In mitotic cells, chromatin immunoprecipitation assays confirmed the association of Ski to major satellite and the promoters of the most repressed genes: Mmp3, Mmp10 and Mmp13. These genes are at pericentromeric region of chromosome 9. In these promoters, the presence of Ski resulted in increased H3K9 tri-methylation levels. This Ski-dependent regulation is also observed during interphase. Consequently, Mmp activity is augmented in Ski-/- MEFs. Altogether, these data indicate that association of Ski with the pericentromeric region of chromosomes during mitosis is required to maintain the silencing bookmarks of underlying chromatin.
Collapse
Affiliation(s)
- Claudio Cappelli
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile; Instituto de Bioquimica y Microbiologia, Facultad de Ciencias, Universidad Austral de Chile, Valdivia, Chile
| | - Hugo Sepulveda
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile
| | - Solange Rivas
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Víctor Pola
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ulises Urzúa
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gerardo Donoso
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Eduardo Sagredo
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - David Carrero
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Emmanuel Casanova-Ortiz
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Alfredo Sagredo
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marisel González
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Marcia Manterola
- Instituto de Ciencias Biomédicas. Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gino Nardocci
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile; FONDAP Center for Genome Regulation, Santiago, Chile
| | - Ricardo Armisén
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile; Centro de Genética y Genómica, Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina Clínica Alemana Universidad del Desarrollo, Santiago, Chile
| | - Martin Montecino
- Instituto de Ciencias Biomédicas, Facultad de Medicina y Facultad de Ciencias de la Vida, Universidad Andres Bello, Santiago, Chile; FONDAP Center for Genome Regulation, Santiago, Chile
| | - Katherine Marcelain
- Departamento de Oncología Básico Clínica. Facultad de Medicina, Universidad de Chile, Santiago, Chile.
| |
Collapse
|
16
|
A new mutational hotspot in the SKI gene in the context of MFS/TAA molecular diagnosis. Hum Genet 2020; 139:461-472. [PMID: 31980905 DOI: 10.1007/s00439-019-02102-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/10/2019] [Indexed: 12/14/2022]
Abstract
SKI pathogenic variations are associated with Shprintzen-Goldberg Syndrome (SGS), a rare systemic connective tissue disorder characterized by craniofacial, skeletal and cardiovascular features. So far, the clinical description, including intellectual disability, has been relatively homogeneous, and the known pathogenic variations were located in two different hotspots of the SKI gene. In the course of diagnosing Marfan syndrome and related disorders, we identified nine sporadic probands (aged 2-47 years) carrying three different likely pathogenic or pathogenic variants in the SKI gene affecting the same amino acid (Thr180). Seven of these molecular events were confirmed de novo. All probands displayed a milder morphological phenotype with a marfanoid habitus that did not initially lead to a clinical diagnosis of SGS. Only three of them had learning disorders, and none had intellectual disability. Six out of nine presented thoracic aortic aneurysm, which led to preventive surgery in the oldest case. This report extends the phenotypic spectrum of variants identified in the SKI gene. We describe a new mutational hotspot associated with a marfanoid syndrome with no intellectual disability. Cardiovascular involvement was confirmed in a significant number of cases, highlighting the importance of accurately diagnosing SGS and ensuring appropriate medical treatment and follow-up.
Collapse
|
17
|
Skip is essential for Notch signaling to induce Sox2 in cerebral arteriovenous malformations. Cell Signal 2020; 68:109537. [PMID: 31927035 DOI: 10.1016/j.cellsig.2020.109537] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/08/2020] [Accepted: 01/09/2020] [Indexed: 11/24/2022]
Abstract
Notch signaling and Sry-box (Sox) family transcriptional factors both play critical roles in endothelial cell (EC) differentiation in vascularization. Recent studies have shown that excessive Notch signaling induces Sox2 to cause cerebral arteriovenous malformations (AVMs). Here, we examine human pulmonary AVMs and find no induction of Sox2. Results of epigenetic studies also show less alteration of Sox2-DNA binding in pulmonary AVMs than in cerebral AVMs. We identify high expression of ski-interacting protein (Skip) in brain ECs, a Notch-associated chromatin-modifying protein that is lacking in lung ECs. Knockdown of Skip abolished Notch-induction of Sox2 in brain ECs, while restoration of Skip in lung ECs enabled Notch-mediated Sox2 induction. The results suggest that Skip is a key factor for induction of Sox2 in cerebral AVMs.
Collapse
|
18
|
Feld C, Sahu P, Frech M, Finkernagel F, Nist A, Stiewe T, Bauer UM, Neubauer A. Combined cistrome and transcriptome analysis of SKI in AML cells identifies SKI as a co-repressor for RUNX1. Nucleic Acids Res 2019; 46:3412-3428. [PMID: 29471413 PMCID: PMC5909421 DOI: 10.1093/nar/gky119] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/09/2018] [Indexed: 11/16/2022] Open
Abstract
SKI is a transcriptional co-regulator and overexpressed in various human tumors, for example in acute myeloid leukemia (AML). SKI contributes to the origin and maintenance of the leukemic phenotype. Here, we use ChIP-seq and RNA-seq analysis to identify the epigenetic alterations induced by SKI overexpression in AML cells. We show that approximately two thirds of differentially expressed genes are up-regulated upon SKI deletion, of which >40% harbor SKI binding sites in their proximity, primarily in enhancer regions. Gene ontology analysis reveals that many of the differentially expressed genes are annotated to hematopoietic cell differentiation and inflammatory response, corroborating our finding that SKI contributes to a myeloid differentiation block in HL60 cells. We find that SKI peaks are enriched for RUNX1 consensus motifs, particularly in up-regulated SKI targets upon SKI deletion. RUNX1 ChIP-seq displays that nearly 70% of RUNX1 binding sites overlap with SKI peaks, mainly at enhancer regions. SKI and RUNX1 occupy the same genomic sites and cooperate in gene silencing. Our work demonstrates for the first time the predominant co-repressive function of SKI in AML cells on a genome-wide scale and uncovers the transcription factor RUNX1 as an important mediator of SKI-dependent transcriptional repression.
Collapse
Affiliation(s)
- Christine Feld
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany.,Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen and Marburg, Baldingerstr., 35043 Marburg, Germany
| | - Peeyush Sahu
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| | - Miriam Frech
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen and Marburg, Baldingerstr., 35043 Marburg, Germany
| | - Florian Finkernagel
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| | - Andrea Nist
- Genomics Core Facility, Philipps University Marburg, Hans-Meerwein-Str. 3, 35043 Marburg, Germany
| | - Thorsten Stiewe
- Genomics Core Facility, Philipps University Marburg, Hans-Meerwein-Str. 3, 35043 Marburg, Germany.,Institute of Molecular Oncology, Philipps University Marburg, Hans-Meerwein-Str. 3, 35043 Marburg, Germany
| | - Uta-Maria Bauer
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Hans-Meerwein-Str. 2, 35043 Marburg, Germany
| | - Andreas Neubauer
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, University Hospital Giessen and Marburg, Baldingerstr., 35043 Marburg, Germany
| |
Collapse
|
19
|
Höflmayer D, Willich C, Hube-Magg C, Simon R, Lang D, Neubauer E, Jacobsen F, Hinsch A, Luebke AM, Tsourlakis MC, Huland H, Graefen M, Haese A, Heinzer H, Minner S, Büscheck F, Sauter G, Schlomm T, Steurer S, Clauditz TS, Burandt E, Wilczak W, Bernreuther C. SNW1 is a prognostic biomarker in prostate cancer. Diagn Pathol 2019; 14:33. [PMID: 31043167 PMCID: PMC6495565 DOI: 10.1186/s13000-019-0810-8] [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: 12/07/2018] [Accepted: 04/11/2019] [Indexed: 12/16/2022] Open
Abstract
Background SNW1 is a nuclear receptor co-activator involved in splicing and transcription control, including androgen receptor signaling. Overexpression of SNW1 has been linked to adverse prognosis in different cancer types, but studies on the role of SNW1 in prostate cancer are lacking. Methods Using immunohistochemistry, we analyzed SNW1 expression in 10,310 prostate cancers in a tissue microarray (TMA) with attached clinical and molecular data. Results The comparison with normal prostate tissue revealed an up regulation of SNW1 in a subset of cancer samples. SNW1 staining was considered weak in 31.5%, moderate in 37.7% and strong in 14% of cancers. Strong SNW1 expression was markedly more frequent in prostate cancers harboring the TMPRSS2:ERG fusion (24%) than in ERG negative cancers (7%, p < 0.0001). Significant associations with Gleason grade, stage, nodal status and early biochemical recurrence were observed in the ERG negative and positive subset. Multivariable modeling revealed that the prognostic value of SNW1 up regulation was independent from the established preoperative histopathological and clinical parameters. Conclusion These results demonstrate that SNW1 overexpression is an independent prognostic marker in prostate cancer with potential clinical utility. Electronic supplementary material The online version of this article (10.1186/s13000-019-0810-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Doris Höflmayer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Carla Willich
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Claudia Hube-Magg
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany.
| | - Dagmar Lang
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Emily Neubauer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Frank Jacobsen
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Andrea Hinsch
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Andreas M Luebke
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Marie Christina Tsourlakis
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Hartwig Huland
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Markus Graefen
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Alexander Haese
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Hans Heinzer
- Martini-Clinic, Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Sarah Minner
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Franziska Büscheck
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Thorsten Schlomm
- Department of Urology, Charité - Universitätsmedizin Berlin, Charitéplatz 1, D-10117, Berlin, Germany
| | - Stefan Steurer
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Till S Clauditz
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Eike Burandt
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Waldemar Wilczak
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| | - Christian Bernreuther
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, D-20246, Hamburg, Germany
| |
Collapse
|
20
|
Preventive Effects of Evodiamine on Dexamethasone-Induced Osteoporosis in Zebrafish. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5859641. [PMID: 30805367 PMCID: PMC6362467 DOI: 10.1155/2019/5859641] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 12/30/2018] [Indexed: 12/02/2022]
Abstract
The aim of this study was to investigate the effect of evodiamine (EV) on dexamethasone-induced osteoporosis in zebrafish. Zebrafish larvae were exposed to different concentrations of dexamethasone to obtain the osteoporosis in zebrafish. Calcium, phosphorus, and alizarin red staining determination were performed to evaluate the effects of EV on bone mineralization. Alkaline phosphatase (ALP), hydroxyproline (HP), and tartrate resistant acid phosphatase (TRAP) were also measured by commercial kits. The expression of MMP3-OPN-MAPK pathway in zebrafish was measured by Western blot. RT-PCR was used to determine mRNA levels of MMP3, OPN, and MAPK. EV could significantly increase the content of calcium and phosphorus. The results of alizarin red staining showed that EV could significantly increase the calcium sink of horse fish, increasing the area of bone formation. EV could increase the content of hydroxyproline in zebrafish. EV also increased ALP and TRAP in zebrafish. Western blot and RT-PCR results showed that EV restored the MMP3-OPN-MAPK pathway in zebrafish. In conclusion, we found that EV can alleviate dexamethasone-induced osteoporosis in zebrafish. The mechanism is related to activating MMP3-OPN-MAPK pathway and then activating bone remodeling.
Collapse
|
21
|
Zeglinski MR, Moghadam AR, Ande SR, Sheikholeslami K, Mokarram P, Sepehri Z, Rokni H, Mohtaram NK, Poorebrahim M, Masoom A, Toback M, Sareen N, Saravanan S, Jassal DS, Hashemi M, Marzban H, Schaafsma D, Singal P, Wigle JT, Czubryt MP, Akbari M, Dixon IM, Ghavami S, Gordon JW, Dhingra S. Myocardial Cell Signaling During the Transition to Heart Failure. Compr Physiol 2018; 9:75-125. [DOI: 10.1002/cphy.c170053] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Muench DE, Ferchen K, Velu CS, Pradhan K, Chetal K, Chen X, Weirauch MT, Colmenares C, Verma A, Salomonis N, Grimes HL. SKI controls MDS-associated chronic TGF-β signaling, aberrant splicing, and stem cell fitness. Blood 2018; 132:e24-e34. [PMID: 30249787 PMCID: PMC6251005 DOI: 10.1182/blood-2018-06-860890] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/18/2018] [Indexed: 01/19/2023] Open
Abstract
The transforming growth factor beta (TGF-β) signaling pathway controls hematopoietic stem cell (HSC) behavior in the marrow niche; however, TGF-β signaling becomes chronic in early-stage myelodysplastic syndrome (MDS). Although TGF-β signaling normally induces negative feedback, in early-stage MDS, high levels of microRNA-21 (miR-21) contribute to chronic TGF-β signaling. We found that a TGF-β signal-correlated gene signature is sufficient to identify an MDS patient population with abnormal RNA splicing (eg, CSF3R) independent of splicing factor mutations and coincident with low HNRNPK activity. Levels of SKI messenger RNA (mRNA) encoding a TGF-β antagonist are sufficient to identify these patients. However, MDS patients with high SKI mRNA and chronic TGF-β signaling lack SKI protein because of miR-21 activity. To determine the impact of SKI loss, we examined murine Ski -/- HSC function. First, competitive HSC transplants revealed a profound defect in stem cell fitness (competitive disadvantage) but not specification, homing, or multilineage production. Aged recipients of Ski -/- HSCs exhibited mild phenotypes similar to phenotypes in those with macrocytic anemia. Second, blastocyst complementation revealed a dramatic block in Ski -/- hematopoiesis in the absence of transplantation. Similar to SKI-high MDS patient samples, Ski -/- HSCs strikingly upregulated TGF-β signaling and deregulated expression of spliceosome genes (including Hnrnpk). Moreover, novel single-cell splicing analyses demonstrated that Ski -/- HSCs and high levels of SKI expression in MDS patient samples share abnormal alternative splicing of common genes (including those that encode splicing factors). We conclude that miR-21-mediated loss of SKI activates TGF-β signaling and alternative splicing to impair the competitive advantage of normal HSCs (fitness), which could contribute to selection of early-stage MDS-genic clones.
Collapse
Affiliation(s)
- David E Muench
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kyle Ferchen
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Chinavenmeni S Velu
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Kith Pradhan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | | | - Matthew T Weirauch
- Division of Biomedical Informatics
- Center for Autoimmune Genomics and Etiology, and
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
| | - Clemencia Colmenares
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Amit Verma
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY; and
| | | | - H Leighton Grimes
- Division of Immunobiology and Center for Systems Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| |
Collapse
|
23
|
Turner AW, Wong D, Dreisbach CN, Miller CL. GWAS Reveal Targets in Vessel Wall Pathways to Treat Coronary Artery Disease. Front Cardiovasc Med 2018; 5:72. [PMID: 29988570 PMCID: PMC6026658 DOI: 10.3389/fcvm.2018.00072] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 05/29/2018] [Indexed: 12/22/2022] Open
Abstract
Coronary artery disease (CAD) is the leading cause of mortality worldwide and poses a considerable public health burden. Recent genome-wide association studies (GWAS) have revealed >100 genetic loci associated with CAD susceptibility in humans. While a number of these loci harbor gene targets of currently approved therapies, such as statins and PCSK9 inhibitors, the majority of the annotated genes at these loci encode for proteins involved in vessel wall function with no known drugs available. Importantly many of the associated genes linked to vascular (smooth muscle, endothelial, and macrophage) cell processes are now organized into distinct functional pathways, e.g., vasodilation, growth factor responses, extracellular matrix and plaque remodeling, and inflammation. In this mini-review, we highlight the most recently identified loci that have predicted roles in the vessel wall and provide genetic context for pre-existing therapies as well as new drug targets informed from GWAS. With the development of new modalities to target these pathways, (e.g., antisense oligonucleotides, CRISPR/Cas9, and RNA interference) as well as the computational frameworks to prioritize or reposition therapeutics, there is great opportunity to close the gap from initial genetic discovery to clinical translation for many patients affected by this common disease.
Collapse
Affiliation(s)
- Adam W Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States
| | - Doris Wong
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States
| | - Caitlin N Dreisbach
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States.,Data Science Institute, University of Virginia, Charlottesville, VA, United States
| | - Clint L Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, United States.,Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, United States.,Data Science Institute, University of Virginia, Charlottesville, VA, United States.,Department of Public Health Sciences, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
24
|
Tecalco-Cruz AC, Ríos-López DG, Vázquez-Victorio G, Rosales-Alvarez RE, Macías-Silva M. Transcriptional cofactors Ski and SnoN are major regulators of the TGF-β/Smad signaling pathway in health and disease. Signal Transduct Target Ther 2018; 3:15. [PMID: 29892481 PMCID: PMC5992185 DOI: 10.1038/s41392-018-0015-8] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 02/16/2018] [Accepted: 03/15/2018] [Indexed: 12/19/2022] Open
Abstract
The transforming growth factor-β (TGF-β) family plays major pleiotropic roles by regulating many physiological processes in development and tissue homeostasis. The TGF-β signaling pathway outcome relies on the control of the spatial and temporal expression of >500 genes, which depend on the functions of the Smad protein along with those of diverse modulators of this signaling pathway, such as transcriptional factors and cofactors. Ski (Sloan-Kettering Institute) and SnoN (Ski novel) are Smad-interacting proteins that negatively regulate the TGF-β signaling pathway by disrupting the formation of R-Smad/Smad4 complexes, as well as by inhibiting Smad association with the p300/CBP coactivators. The Ski and SnoN transcriptional cofactors recruit diverse corepressors and histone deacetylases to repress gene transcription. The TGF-β/Smad pathway and coregulators Ski and SnoN clearly regulate each other through several positive and negative feedback mechanisms. Thus, these cross-regulatory processes finely modify the TGF-β signaling outcome as they control the magnitude and duration of the TGF-β signals. As a result, any alteration in these regulatory mechanisms may lead to disease development. Therefore, the design of targeted therapies to exert tight control of the levels of negative modulators of the TGF-β pathway, such as Ski and SnoN, is critical to restore cell homeostasis under the specific pathological conditions in which these cofactors are deregulated, such as fibrosis and cancer. Proteins that repress molecular signaling through the transforming growth factor-beta (TGF-β) pathway offer promising targets for treating cancer and fibrosis. Marina Macías-Silva and colleagues from the National Autonomous University of Mexico in Mexico City review the ways in which a pair of proteins, called Ski and SnoN, interact with downstream mediators of TGF-β to inhibit the effects of this master growth factor. Aberrant levels of Ski and SnoN have been linked to diverse range of diseases involving cell proliferation run amok, and therapies that regulate the expression of these proteins could help normalize TGF-β signaling to healthier physiological levels. For decades, drug companies have tried to target the TGF-β pathway, with limited success. Altering the activity of these repressors instead could provide a roundabout way of remedying pathogenic TGF-β activity in fibrosis and oncology.
Collapse
Affiliation(s)
- Angeles C Tecalco-Cruz
- 1Instituto de Investigaciones Biomédicas at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Diana G Ríos-López
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | | | - Reyna E Rosales-Alvarez
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| | - Marina Macías-Silva
- 2Instituto de Fisiología Celular at Universidad Nacional Autónoma de México, Mexico city, 04510 Mexico
| |
Collapse
|
25
|
Frech M, Teichler S, Feld C, Bouchard C, Berberich H, Sorg K, Mernberger M, Bullinger L, Bauer UM, Neubauer A. MYB induces the expression of the oncogenic corepressor SKI in acute myeloid leukemia. Oncotarget 2018; 9:22423-22435. [PMID: 29854289 PMCID: PMC5976475 DOI: 10.18632/oncotarget.25051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/21/2018] [Indexed: 11/25/2022] Open
Abstract
Acute myeloid leukemia (AML) arises through clonal expansion of transformed myeloid progenitor cells. The SKI proto-oncogene is highly upregulated in different solid tumors and leukemic cells, but little is known about its transcriptional regulation during leukemogenesis. MYB is an important hematopoietic transcription factor involved in proliferation as well as differentiation and upregulated in most human acute leukemias. Here, we find that MYB protein binds within the regulatory region of the SKI gene in AML cells. Reporter gene assays using MYB binding sites present in the SKI gene locus show MYB-dependent transcriptional activation. SiRNA-mediated depletion of MYB in leukemic cell lines reveals that MYB is crucial for SKI gene expression. Consistently, we observed a positive correlation of MYB and SKI expression in leukemic cell lines and in samples of AML patients. Moreover, MYB and SKI both were downregulated by treatment with histone deacetylase inhibitors. Strikingly, differentiation of AML cells induced by depletion of MYB is attenuated by overexpression of SKI. Our findings identify SKI as a novel MYB target gene, relevant for the MYB-induced differentiation block in leukemic cells.
Collapse
Affiliation(s)
- Miriam Frech
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Sabine Teichler
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Christine Feld
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany.,Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Caroline Bouchard
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Hannah Berberich
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Katharina Sorg
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| | - Marco Mernberger
- Institute of Molecular Oncology, Philipps University Marburg, Marburg 35043, Germany
| | - Lars Bullinger
- Department of Internal Medicine III, University Hospital of Ulm, Ulm 89081, Germany
| | - Uta-Maria Bauer
- Institute of Molecular Biology and Tumor Research (IMT), School of Medicine, Philipps University Marburg, Marburg 35043, Germany
| | - Andreas Neubauer
- Department of Internal Medicine and Hematology, Oncology and Immunology, Philipps University Marburg, Marburg 35033, Germany.,University Hospital Giessen and Marburg, Marburg 35033, Germany
| |
Collapse
|
26
|
Tomasi ML, Ramani K. SUMOylation and phosphorylation cross-talk in hepatocellular carcinoma. Transl Gastroenterol Hepatol 2018; 3:20. [PMID: 29780898 DOI: 10.21037/tgh.2018.04.04] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 04/11/2018] [Indexed: 01/22/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a primary malignancy of the liver and occurs predominantly in patients with underlying chronic liver disease and cirrhosis. The large spectrum of protein post-translational modification (PTM) includes numerous critical signaling events that occur during neoplastic transformation. PTMs occur to nearly all proteins and increase the functional diversity of proteins. We have reviewed the role of two major PTMs, SUMOylation and phosphorylation, in the altered signaling of key players in HCC. SUMOylation is a PTM that involves addition of a small ubiquitin-like modifiers (SUMO) group to proteins. It is known to regulate protein stability, protein-protein interactions, trafficking and transcriptional activity. The major pathways that are regulated by SUMOylation and may influence HCC are regulation of transcription, cell growth pathways associated with B-cell lymphoma 2 (Bcl-2) and methionine adenosyltransferases (MAT), oxidative stress pathways [nuclear erythroid 2-related factor 2 (Nrf2)], tumor suppressor pathways (p53), hypoxia-inducible signaling [hypoxia-inducible factor-1 (HIF-1)], glucose and lipid metabolism, nuclear factor kappa B (NF-κB) and β-Catenin signaling. Phosphorylation is an extensively studied PTM in HCC. The mitogen-activated protein kinase (MAPK), phosphatidyl inositol/AK-strain transforming (PI3K/AKT), and C-SRC pathways have been extensively studied for deregulation of kinases and alteration in signaling of targets through phosphorylation of their substrates. Cross-talk between phosphorylation and SUMOylation is known to influence transcriptional activity of proteins and protein-protein interactions. In HCC, several SUMOylation-dependent phosphorylation events have been studied such as MAPK activation and c-SRC activity that have been reviewed in this work. The drastic effects of site-specific phosphorylation or SUMOylation on enzyme activity of signaling players and its effect on growth and tumorigenesis suggests that these PTMs are novel targets for therapeutic intervention in HCC.
Collapse
Affiliation(s)
- Maria Lauda Tomasi
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Komal Ramani
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
27
|
Silencing of c-Ski augments TGF-b1-induced epithelial-mesenchymal transition in cardiomyocyte H9C2 cells. Cardiol J 2018; 26:66-76. [PMID: 29570207 DOI: 10.5603/cj.a2018.0009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/09/2017] [Accepted: 01/15/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The shRNA lentiviral vector was constructed to silence c-Ski expression in cardiac mus- cle cells, with the aim of exploring the role of c-Ski in transforming growth factor b1 (TGF-b1)-induced epithelial-mesenchymal transitions (EMT) in H9C2 cells. METHODS Real-time polymerase chain reaction (RT-PCR) and western blot were used to detect c-Ski ex- pression at protein and messenger ribonucleic acid (mRNA) levels in 5 different cell lines. Then, lentiviral vector was constructed to silence or overexpress c-Ski in H9C2 cells. MTT and/or soft agar assay and tran- swell assay were used to detect cell proliferation and migration, respectively. The expression levels of c-Ski under different concentrations of TGF-b1 stimulation were detected by RT-qPCR and immunocytochemi- cal analysis. In the presence or absence of TGF-b1 stimulation, the proteins' expression levels of a-SMA, FN and E-cadherin, which are closely correlated with the process of EMT, were measured by western blot after c-Ski silencing or overexpression. Meanwhile, the effect of c-Ski on Samd3 phosphorylation with TGF-b1 stimulation was investigated. RESULTS There is a high expression of c-Ski at protein and mRNA levels in H9C2 cell line, which first demonstrated the presence of c-Ski expression in H9C2 cells. Overexpression of c-Ski significantly increased H9C2 cell proliferation. The ability of c-Ski gene silencing to suppress cell proliferation was gradually enhanced, and inhibition efficiency was the highest after 6 to 7 d of transfection. Moreover, H9C2 cells with c-Ski knockdown gained significantly aggressive invasive potential when compared with the control group. TGF-b1 stimulation could dose-independently reduce c-Ski expression in H9C2 cells and lead to obvious down-regulated expression of E-cadherin. Interestingly, c-Ski could restore E-cadherin expression while suppressing a-SMA and/or FN expression stimulated by TGF-b1. How- ever, shRNA-induced c-Ski knockdown aggravated only the TGF-b1-induced EMT. Moreover, c-Ski- -shRNA also promoted the phosphorylation of Samd3 induced by TGF-b1. CONCLUSIONS c-Ski expression in cardiac muscle cells could be down-regulated by TGF-b1. Silencing of c-Ski gene was accompanied by down-regulation of E-cadherin, up-regulation of a-SMA and/or FN and Smad3 phosphorylation induced by TGF-b1, promoting EMT process. Therefore, c-Ski may be closely associated with TGF-b1-induced EMT and play an important role in cardiac fibrosis develop- ment and progression.
Collapse
|
28
|
Yin H, Wang S, Zhang Y, Wu M, Wang J, Ma Y. Zuogui Pill improves the dexamethasone-induced osteoporosis progression in zebrafish larvae. Biomed Pharmacother 2017; 97:995-999. [PMID: 29136778 DOI: 10.1016/j.biopha.2017.11.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 10/28/2017] [Accepted: 11/03/2017] [Indexed: 12/12/2022] Open
Abstract
The current study was to evaluate the protective effects of Chinese medicine prescription Zuogui Pill (ZGP) on osteoporosis (OP) in zebrafish larvae induced by dexamethasone. Alizarin Red staining, calcium and phosphorus determination were performed to evaluate the effect of ZGP on bone mineralization. Hydroxyproline (HP), Alkaline phosphatase (ALP), and tartrate resistant acid phosphatase (TRAP) were also measured by commercial kits. We found that ZGP had positive effects in increasing bone mineral content (BMC), strengthening bone biomechanical, promoting bone formation, inhibiting bone resorption, and mediating protein levels of TGF-β1/Smads signaling pathway. The findings demonstrated that ZGP treatments inhibited the phosphorylation of TGF-β and p-Smad 3 as well as the expressions of collagen I and collagen II by western blot. Taken together, we demonstrated that ZGP may prevent osteoporosis via reversing the imbalance of bone fomation/bone resorption and activating the TGF-β-Smad signal.
Collapse
Affiliation(s)
- Heng Yin
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Shanfu Wang
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Yafeng Zhang
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Mao Wu
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - JianWei Wang
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China.
| | - Yong Ma
- Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
29
|
Xie M, Wu X, Zhang J, Zhang J, Li X. Ski regulates Smads and TAZ signaling to suppress lung cancer progression. Mol Carcinog 2017; 56:2178-2189. [PMID: 28398634 DOI: 10.1002/mc.22661] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 03/28/2017] [Accepted: 04/08/2017] [Indexed: 12/11/2022]
Abstract
Ski, the transforming protein of the avian Sloan-Kettering retrovirus, displays both pro- and anti-oncogenic activities in human cancer. The mechanisms underlying these conflicting observations have not been fully understood. Herein, we investigated the mechanism underlying the tumor suppressor activity of Ski. To investigate the effect of Ski re-activation on TGF-β and Hippo/TAZ pathway, we measured its effect on the endogenous Smad target genes (PAI-1 and P15INK4B ) and TAZ target gene CTGF. The results revealed that Ski exerted its inhibitory activity in TGF-β1/Smad signaling pathway. Ski inhibited TAZ by increasing their phosphorylation by Lats2 and did not alter the localization of TAZ. Ski inhibited lung cancer growth and invasion. Ski methylation correlated with decreased mRNA expression in human lung cancer cell lines. Thus, Ski inhibited the function of TGF-β and TAZ through multiple mechanisms in human lung cancer.
Collapse
Affiliation(s)
- Mian Xie
- China State Key Laboratory of Respiratory Disease and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaojun Wu
- State Key Laboratory of Oncology in Southern China, Department of Colorectal Surgery, Sun Yat-Sen University Cancer Center, Collaborative Innovation Center of Cancer Medicine, Guangzhou, China
| | - Jinjun Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiexia Zhang
- China State Key Laboratory of Respiratory Disease and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangxiang Li
- China State Key Laboratory of Respiratory Disease and Guangzhou Institute of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Walldén K, Nyman T, Hällberg BM. SnoN Stabilizes the SMAD3/SMAD4 Protein Complex. Sci Rep 2017; 7:46370. [PMID: 28397834 PMCID: PMC5387736 DOI: 10.1038/srep46370] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 03/15/2017] [Indexed: 12/30/2022] Open
Abstract
TGF-β signaling regulates cellular processes such as proliferation, differentiation and apoptosis through activation of SMAD transcription factors that are in turn modulated by members of the Ski-SnoN family. In this process, Ski has been shown to negatively modulate TGF-β signaling by disrupting active R-SMAD/Co-SMAD heteromers. Here, we show that the related regulator SnoN forms a stable complex with the R-SMAD (SMAD3) and the Co-SMAD (SMAD4). To rationalize this stabilization at the molecular level, we determined the crystal structure of a complex between the SAND domain of SnoN and the MH2-domain of SMAD4. This structure shows a binding mode that is compatible with simultaneous coordination of R-SMADs. Our results show that SnoN, and SMAD heteromers can form a joint structural core for the binding of other transcription modulators. The results are of fundamental importance for our understanding of the molecular mechanisms behind the modulation of TGF-β signaling.
Collapse
Affiliation(s)
- Karin Walldén
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Tomas Nyman
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - B Martin Hällberg
- Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden.,Röntgen-Ångström-Cluster, Karolinska Institutet Outstation, Centre for Structural Systems Biology, DESY-Campus, 22603 Hamburg, Germany.,European Molecular Biology Laboratory, Hamburg Unit, 22603 Hamburg, Germany
| |
Collapse
|
31
|
Hara T, Yoshida E, Fujiwara Y, Yamamoto C, Kaji T. Transforming Growth Factor-β 1 Modulates the Expression of Syndecan-4 in Cultured Vascular Endothelial Cells in a Biphasic Manner. J Cell Biochem 2017; 118:2009-2017. [PMID: 28019669 PMCID: PMC5485002 DOI: 10.1002/jcb.25861] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/22/2016] [Indexed: 01/06/2023]
Abstract
Proteoglycans are macromolecules that consist of a core protein and one or more glycosaminoglycan side chains. Previously, we reported that transforming growth factor‐β1 (TGF‐β1) regulates the synthesis of a large heparan sulfate proteoglycan, perlecan, and a small leucine‐rich dermatan sulfate proteoglycan, biglycan, in vascular endothelial cells depending on cell density. Recently, we found that TGF‐β1 first upregulates and then downregulates the expression of syndecan‐4, a transmembrane heparan sulfate proteoglycan, via the TGF‐β receptor ALK5 in the cells. In order to identify the intracellular signal transduction pathway that mediates this modulation, bovine aortic endothelial cells were cultured and treated with TGF‐β1. Involvement of the downstream signaling pathways of ALK5—the Smad and MAPK pathways—in syndecan‐4 expression was examined using specific siRNAs and inhibitors. The data indicate that the Smad3–p38 MAPK pathway mediates the early upregulation of syndecan‐4 by TGF‐β1, whereas the late downregulation is mediated by the Smad2/3 pathway. Multiple modulations of proteoglycan synthesis may be involved in the regulation of vascular endothelial cell functions by TGF‐β1. J. Cell. Biochem. 118: 2009–2017,2017. © 2016 The Authors. Journal of Cellular Biochemistry Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Takato Hara
- Faculty of Pharmaceutical SciencesDepartment of Environmental HealthTokyo University of ScienceNoda 278‐8510Japan
| | - Eiko Yoshida
- Faculty of Pharmaceutical SciencesDepartment of Environmental HealthTokyo University of ScienceNoda 278‐8510Japan
| | - Yasuyuki Fujiwara
- Department of Environmental HealthSchool of PharmacyTokyo University of Pharmacy and Life SciencesHachioji 192‐0392Japan
| | - Chika Yamamoto
- Faculty of Pharmaceutical SciencesDepartment of Environmental HealthToho UniversityFunabashi 274‐8510Japan
| | - Toshiyuki Kaji
- Faculty of Pharmaceutical SciencesDepartment of Environmental HealthTokyo University of ScienceNoda 278‐8510Japan
| |
Collapse
|
32
|
Abstract
Thoracic aortic aneurysm is a potentially life-threatening condition in that it places patients at risk for aortic dissection or rupture. However, our modern understanding of the pathogenesis of thoracic aortic aneurysm is quite limited. A genetic predisposition to thoracic aortic aneurysm has been established, and gene discovery in affected families has identified several major categories of gene alterations. The first involves mutations in genes encoding various components of the transforming growth factor beta (TGF-β) signaling cascade (FBN1, TGFBR1, TGFBR2, TGFB2, TGFB3, SMAD2, SMAD3 and SKI), and these conditions are known collectively as the TGF-β vasculopathies. The second set of genes encode components of the smooth muscle contractile apparatus (ACTA2, MYH11, MYLK, and PRKG1), a group called the smooth muscle contraction vasculopathies. Mechanistic hypotheses based on these discoveries have shaped rational therapies, some of which are under clinical evaluation. This review discusses published data on genes involved in thoracic aortic aneurysm and attempts to explain divergent hypotheses of aneurysm origin.
Collapse
Affiliation(s)
- Eric M Isselbacher
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Christian Lacks Lino Cardenas
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston
| | - Mark E Lindsay
- From Thoracic Aortic Center (E.M.I., C.L.L.C., M.E.L.), Cardiovascular Genetics Program (M.E.L.), Cardiovascular Research Center (C.L.L.C., M.E.L.), and Cardiology Division (E.M.I., C.L.L.C., M.E.L.), Department of Medicine, and Pediatric Cardiology Division, Department of Pediatrics (M.E.L.), Massachusetts General Hospital, Harvard Medical School, Boston.
| |
Collapse
|
33
|
Smith MP, Brunton H, Rowling EJ, Ferguson J, Arozarena I, Miskolczi Z, Lee JL, Girotti MR, Marais R, Levesque MP, Dummer R, Frederick DT, Flaherty KT, Cooper ZA, Wargo JA, Wellbrock C. Inhibiting Drivers of Non-mutational Drug Tolerance Is a Salvage Strategy for Targeted Melanoma Therapy. Cancer Cell 2016; 29:270-284. [PMID: 26977879 PMCID: PMC4796027 DOI: 10.1016/j.ccell.2016.02.003] [Citation(s) in RCA: 183] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 12/18/2015] [Accepted: 02/08/2016] [Indexed: 01/19/2023]
Abstract
Once melanomas have progressed with acquired resistance to mitogen-activated protein kinase (MAPK)-targeted therapy, mutational heterogeneity presents a major challenge. We therefore examined the therapy phase before acquired resistance had developed and discovered the melanoma survival oncogene MITF as a driver of an early non-mutational and reversible drug-tolerance state, which is induced by PAX3-mediated upregulation of MITF. A drug-repositioning screen identified the HIV1-protease inhibitor nelfinavir as potent suppressor of PAX3 and MITF expression. Nelfinavir profoundly sensitizes BRAF and NRAS mutant melanoma cells to MAPK-pathway inhibitors. Moreover, nelfinavir is effective in BRAF and NRAS mutant melanoma cells isolated from patients progressed on MAPK inhibitor (MAPKi) therapy and in BRAF/NRAS/PTEN mutant tumors. We demonstrate that inhibiting a driver of MAPKi-induced drug tolerance could improve current approaches of targeted melanoma therapy.
Collapse
Affiliation(s)
- Michael P Smith
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Holly Brunton
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Emily J Rowling
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jennifer Ferguson
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Imanol Arozarena
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Zsofia Miskolczi
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Jessica L Lee
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK
| | - Maria R Girotti
- Molecular Oncology Group, CRUK Manchester Institute for Cancer Research, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4BX, UK
| | - Richard Marais
- Molecular Oncology Group, CRUK Manchester Institute for Cancer Research, Manchester Cancer Research Centre, Wilmslow Road, Manchester, M20 4BX, UK
| | - Mitchell P Levesque
- Department of Dermatology, UniversitätsSpital Zürich, University of Zürich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, UniversitätsSpital Zürich, University of Zürich, Gloriastrasse 31, 8091 Zurich, Switzerland
| | - Dennie T Frederick
- Department of Medicine, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA 02114-2696, USA
| | - Keith T Flaherty
- Department of Medicine, Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, MA 02114-2696, USA
| | - Zachary A Cooper
- Divison of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA
| | - Jennifer A Wargo
- Divison of Surgical Oncology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Houston, TX 77030, USA
| | - Claudia Wellbrock
- Manchester Cancer Research Centre, Wellcome Trust Centre for Cell-Matrix Research, The University of Manchester, Michael Smith Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
34
|
Ye L, Jiang WG. Bone morphogenetic proteins in tumour associated angiogenesis and implication in cancer therapies. Cancer Lett 2015; 380:586-597. [PMID: 26639195 DOI: 10.1016/j.canlet.2015.10.036] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/17/2015] [Accepted: 10/12/2015] [Indexed: 02/09/2023]
Abstract
Bone morphogenetic protein (BMP) belongs to transforming growth factor-β superfamily. To date, more than 20 BMPs have been identified in humans. BMPs play a critical role in embryonic and postnatal development, and also in maintaining homeostasis in different organs and tissues by regulating cell differentiation, proliferation, survival and motility. They play important roles in the development and progression of certain malignancies, including prostate cancer, breast cancer, lung cancer, etc. Recently, more evidence shows that BMPs are also involved in tumour associated angiogenesis. For example BMP can either directly regulate the functions of vascular endothelial cells or indirectly influence the angiogenesis via regulation of angiogenic factors, such as vascular endothelial growth factor (VEGF). Such crosstalk can also be reflected in the interaction with other angiogenic factors, like hepatocyte growth factor (HGF) and basic fibroblast growth factor (bFGF). All these factors are involved in the orchestration of the angiogenic process during tumour development and progression. Review of the relevant studies will provide a comprehensive prospective on current understanding and shed light on the corresponding therapeutic opportunity.
Collapse
Affiliation(s)
- Lin Ye
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK.
| | - Wen G Jiang
- Metastasis & Angiogenesis Research Group, Cardiff University-Peking University Cancer Institute, Institute of Cancer and Genetics, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| |
Collapse
|
35
|
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: 0.9] [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
|
36
|
YANG HAIPING, ZHAN LEI, YANG TIANJIE, WANG LONGQIANG, LI CHANG, ZHAO JUN, LEI ZHE, LI XIANGDONG, ZHANG HONGTAO. Ski prevents TGF-β-induced EMT and cell invasion by repressing SMAD-dependent signaling in non-small cell lung cancer. Oncol Rep 2015; 34:87-94. [DOI: 10.3892/or.2015.3961] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/13/2015] [Indexed: 11/06/2022] Open
|
37
|
Vázquez-Victorio G, Caligaris C, Del Valle-Espinosa E, Sosa-Garrocho M, González-Arenas NR, Reyes-Cruz G, Briones-Orta MA, Macías-Silva M. Novel regulation of Ski protein stability and endosomal sorting by actin cytoskeleton dynamics in hepatocytes. J Biol Chem 2015; 290:4487-99. [PMID: 25561741 DOI: 10.1074/jbc.m114.579532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
TGF-β-induced antimitotic signals are highly regulated during cell proliferation under normal and pathological conditions, such as liver regeneration and cancer. Up-regulation of the transcriptional cofactors Ski and SnoN during liver regeneration may favor hepatocyte proliferation by inhibiting TGF-β signals. In this study, we found a novel mechanism that regulates Ski protein stability through TGF-β and G protein-coupled receptor (GPCR) signaling. Ski protein is distributed between the nucleus and cytoplasm of normal hepatocytes, and the molecular mechanisms controlling Ski protein stability involve the participation of actin cytoskeleton dynamics. Cytoplasmic Ski is partially associated with actin and localized in cholesterol-rich vesicles. Ski protein stability is decreased by TGF-β/Smads, GPCR/Rho signals, and actin polymerization, whereas GPCR/cAMP signals and actin depolymerization promote Ski protein stability. In conclusion, TGF-β and GPCR signals differentially regulate Ski protein stability and sorting in hepatocytes, and this cross-talk may occur during liver regeneration.
Collapse
Affiliation(s)
- Genaro Vázquez-Victorio
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Cassandre Caligaris
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Eugenio Del Valle-Espinosa
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Marcela Sosa-Garrocho
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Nelly R González-Arenas
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Guadalupe Reyes-Cruz
- the Departamento de Biología Celular, CINVESTAV-IPN, México, D. F. 07000, México
| | - Marco A Briones-Orta
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| | - Marina Macías-Silva
- From the Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, México, D. F. 04510, México and
| |
Collapse
|
38
|
Chen Z, Li W, Ning Y, Liu T, Shao J, Wang Y. Ski diminishes TGF-β1-induced myofibroblast phenotype via up-regulating Meox2 expression. Exp Mol Pathol 2014; 97:542-9. [PMID: 25445500 DOI: 10.1016/j.yexmp.2014.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Revised: 09/26/2014] [Accepted: 11/04/2014] [Indexed: 10/24/2022]
Abstract
OBJECTIVE The aim of the present work was to investigate the mechanism of transforming growth factor (TGF)-β1 and Sloan-Kettering Institute (Ski) in the pathogenesis of hypertrophic scars (HS). BACKGROUND Wound healing is an inherent process, but the aberrant wound healing of skin injury may lead to HS. There has been growing evidence suggesting a role for TGF-β1 and Ski in the pathogenesis of fibrosis. MATERIAL AND METHODS The MTT assay was used to detect the cell proliferation induced by TGF-β1. The Ski gene was transduced into cells with an adenovirus, and then the function of Ski in cell proliferation and differentiation was observed. Ski mRNA levels were measured by RT-PCR. Western blotting was used to detect the protein expression of α-SMA, E-cadherin, Meox1, Meox2, Zeb1 and Zeb2. RESULTS TGF-β1 can promote human skin fibroblast (HSF) cell proliferation in a time-dependent manner, but the promoting effect could be suppressed by Ski. TGF-β1 also induces the formation of the myofibroblast phenotype and the effect of TGF-β1 could be diminished by Ski. Also, Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by up-regulating the expression of Meox2. CONCLUSIONS Ski diminishes the myofibroblast phenotype induced by TGF-β1 through the suppression of Zeb2 by up-regulating the expression of Meox2.
Collapse
Affiliation(s)
- Zhaowei Chen
- Department of Burns and Plastic Surgery, Liao Cheng Hospital, Liao Cheng 252000, China.
| | - Wenjing Li
- Department of Burns and Plastic Surgery, Liao Cheng Hospital, Liao Cheng 252000, China
| | - Yan Ning
- Department of Burns and Plastic Surgery, Liao Cheng Hospital, Liao Cheng 252000, China
| | - Tong Liu
- Department of Burns and Plastic Surgery, Liao Cheng Hospital, Liao Cheng 252000, China
| | - Jingxiang Shao
- Department of Burns and Plastic Surgery, Liao Cheng Hospital, Liao Cheng 252000, China
| | - Yaojun Wang
- Department of Burns and Skin Surgery, Xi Jing Hospital, Xian 710032, China
| |
Collapse
|
39
|
Schepers D, Doyle AJ, Oswald G, Sparks E, Myers L, Willems PJ, Mansour S, Simpson MA, Frysira H, Maat-Kievit A, Van Minkelen R, Hoogeboom JM, Mortier GR, Titheradge H, Brueton L, Starr L, Stark Z, Ockeloen C, Lourenco CM, Blair E, Hobson E, Hurst J, Maystadt I, Destrée A, Girisha KM, Miller M, Dietz HC, Loeys B, Van Laer L. The SMAD-binding domain of SKI: a hotspot for de novo mutations causing Shprintzen-Goldberg syndrome. Eur J Hum Genet 2014; 23:224-8. [PMID: 24736733 DOI: 10.1038/ejhg.2014.61] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Revised: 02/24/2014] [Accepted: 03/05/2014] [Indexed: 01/10/2023] Open
Abstract
Shprintzen-Goldberg syndrome (SGS) is a rare, systemic connective tissue disorder characterized by craniofacial, skeletal, and cardiovascular manifestations that show a significant overlap with the features observed in the Marfan (MFS) and Loeys-Dietz syndrome (LDS). A distinguishing observation in SGS patients is the presence of intellectual disability, although not all patients in this series present this finding. Recently, SGS was shown to be due to mutations in the SKI gene, encoding the oncoprotein SKI, a repressor of TGFβ activity. Here, we report eight recurrent and three novel SKI mutations in eleven SGS patients. All were heterozygous missense mutations located in the R-SMAD binding domain, except for one novel in-frame deletion affecting the DHD domain. Adding our new findings to the existing data clearly reveals a mutational hotspot, with 73% (24 out of 33) of the hitherto described unrelated patients having mutations in a stretch of five SKI residues (from p.(Ser31) to p.(Pro35)). This implicates that the initial molecular testing could be focused on mutation analysis of the first half of exon 1 of SKI. As the majority of the known mutations are located in the R-SMAD binding domain of SKI, our study further emphasizes the importance of TGFβ signaling in the pathogenesis of SGS.
Collapse
Affiliation(s)
- Dorien Schepers
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Alexander J Doyle
- 1] McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Howard Hughes Medical Institute, Baltimore, MD, USA
| | - Gretchen Oswald
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Elizabeth Sparks
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Loretha Myers
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Sahar Mansour
- SW Thames Regional Genetics Service, St George's, University of London, London, UK
| | - Michael A Simpson
- Division of Genetics and Molecular Medicine, Department of Medical and Molecular Genetics, King's College London School of Medicine, London, UK
| | - Helena Frysira
- Department of Medical Genetics, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Anneke Maat-Kievit
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Rick Van Minkelen
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Jeanette M Hoogeboom
- Department of Clinical Genetics, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Geert R Mortier
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Hannah Titheradge
- Department of Clinical Genetics, Birmingham Women's Hospital, Birmingham, UK
| | - Louise Brueton
- Department of Clinical Genetics, Birmingham Women's Hospital, Birmingham, UK
| | - Lois Starr
- Clinical Genetics, Munroe-Meyer Institute for Genetics and Rehabilitation, Nebraska Medical Center, Omaha, NE, USA
| | - Zornitza Stark
- Victorian Clinical Genetics Services, Murdoch Childrens Research Institute, Parkville, Victoria, Australia
| | - Charlotte Ockeloen
- Department of Human Genetics, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - Charles Marques Lourenco
- Department of Medical Genetics, School of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Ed Blair
- Department of Clinical Genetics, Churchill Hospital, Oxford, UK
| | - Emma Hobson
- Department of Clinical Genetics, Chapel Allerton Hospital, Leeds, UK
| | - Jane Hurst
- Department of Clinical Genetics, Great Ormond Street Hospital, London, UK
| | - Isabelle Maystadt
- Center for Human Genetics, Institute for Pathology and Genetics (IPG), Gosselies, Belgium
| | - Anne Destrée
- Center for Human Genetics, Institute for Pathology and Genetics (IPG), Gosselies, Belgium
| | - Katta M Girisha
- Division of Medical Genetics, Department of Pediatrics, Kasturba Medical College, Manipal University, Manipal, India
| | - Michelle Miller
- Department of Cardiology, All Childrens Hospital, St. Petersburg, FL, USA
| | - Harry C Dietz
- 1] McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA [2] Howard Hughes Medical Institute, Baltimore, MD, USA
| | - Bart Loeys
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| | - Lut Van Laer
- Center for Medical Genetics, Faculty of Medicine and Health Sciences, University of Antwerp and Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
40
|
Wang L, Zhang M, Wu Y, Cheng C, Huang Y, Shi Z, Huang H. SKIP expression is correlated with clinical prognosis in patients with bladder cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:1695-1701. [PMID: 24817966 PMCID: PMC4014250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 02/26/2014] [Indexed: 06/03/2023]
Abstract
The Ski-interacting protein (SKIP) is a transcriptional cofactor distinct from other cofactors and is involved in regulation of many cancer-related proteins. However, its distribution and clinical significances in bladder cancer remains poorly understood. In this study, Quantitative real-time PCR and immunohistochemistry were performed to detect the expression of SKIP in clinical bladder cancer samples. In addition, the correlation of SKIP expression and clinicopathological features and clinical outcomes were analyzed. The expression levels of SKIP in clinical bladder cancer were much higher than that in paired adjacent noncancerous tissues. High expression of SKIP was closely related with histological grades and the poor prognosis of bladder cancer. Based on our data, we speculated that SKIP may be a potential prognostic marker in bladder cancer.
Collapse
Affiliation(s)
- Longwang Wang
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Mei Zhang
- Department of Neurology, Wuhan Central HospitalHubei Province, 430014, China
| | - Yong Wu
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Cheng Cheng
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Yawei Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Zimin Shi
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| | - Hongwei Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang UniversityNanchang 330006, China
| |
Collapse
|
41
|
Cunnington RH, Northcott JM, Ghavami S, Filomeno KL, Jahan F, Kavosh MS, Davies JJL, Wigle JT, Dixon IMC. The Ski-Zeb2-Meox2 pathway provides a novel mechanism for regulation of the cardiac myofibroblast phenotype. J Cell Sci 2013; 127:40-9. [PMID: 24155330 DOI: 10.1242/jcs.126722] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cardiac fibrosis is linked to fibroblast-to-myofibroblast phenoconversion and proliferation but the mechanisms underlying this are poorly understood. Ski is a negative regulator of TGF-β-Smad signaling in myofibroblasts, and might redirect the myofibroblast phenotype back to fibroblasts. Meox2 could alter TGF-β-mediated cellular processes and is repressed by Zeb2. Here, we investigated whether Ski diminishes the myofibroblast phenotype by de-repressing Meox2 expression and function through repression of Zeb2 expression. We show that expression of Meox1 and Meox2 mRNA and Meox2 protein is reduced during phenoconversion of fibroblasts to myofibroblasts. Overexpression of Meox2 shifts the myofibroblasts into fibroblasts, whereas the Meox2 DNA-binding mutant has no effect on myofibroblast phenotype. Overexpression of Ski partially restores Meox2 mRNA expression levels to those in cardiac fibroblasts. Expression of Zeb2 increased during phenoconversion and Ski overexpression reduces Zeb2 expression in first-passage myofibroblasts. Furthermore, expression of Meox2 is decreased in scar following myocardial infarction, whereas Zeb2 protein expression increases in the infarct scar. Thus Ski modulates the cardiac myofibroblast phenotype and function through suppression of Zeb2 by upregulating the expression of Meox2. This cascade might regulate cardiac myofibroblast phenotype and presents therapeutic options for treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Ryan H Cunnington
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Transforming Growth Factor-β (TGF-β) superfamily ligands regulate many aspects of cell identity, function, and survival in multicellular animals. Genes encoding five TGF-β family members are present in the genome of C. elegans. Two of the ligands, DBL-1 and DAF-7, signal through a canonical receptor-Smad signaling pathway; while a third ligand, UNC-129, interacts with a noncanonical signaling pathway. No function has yet been associated with the remaining two ligands. Here we summarize these signaling pathways and their biological functions.
Collapse
Affiliation(s)
- Tina L Gumienny
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center College of Medicine, College Station, TX 77843, USA
| | | |
Collapse
|
43
|
Ski protein levels increase during in vitro progression of HPV16-immortalized human keratinocytes and in cervical cancer. Virology 2013; 444:100-8. [PMID: 23809940 DOI: 10.1016/j.virol.2013.05.039] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2013] [Revised: 05/23/2013] [Accepted: 05/30/2013] [Indexed: 01/23/2023]
Abstract
We compared the levels of the Ski oncoprotein, an inhibitor of transforming growth factor-beta (TGF-β) signaling, in normal human keratinocytes (HKc), HPV16 immortalized HKc (HKc/HPV16), and differentiation resistant HKc/HPV16 (HKc/DR) in the absence and presence of TGF-β. Steady-state Ski protein levels increased in HKc/HPV16 and even further in HKc/DR, compared to HKc. TGF-β treatment of HKc, HKc/HPV16, and HKc/DR dramatically decreased Ski. TGF-β-induced Ski degradation was delayed in HKc/DR. Ski and phospho-Ski protein levels are cell cycle dependent with maximal Ski expression and localization to centrosomes and mitotic spindles during G2/M. ShRNA knock down of Ski in HKc/DR inhibited cell proliferation. More intense nuclear and cytoplasmic Ski staining and altered Ski localization were found in cervical cancer samples compared to adjacent normal tissue in a cervical cancer tissue array. Overall, these studies demonstrate altered Ski protein levels, degradation and localization in HPV16-transformed human keratinocytes and in cervical cancer.
Collapse
|
44
|
Danielpour D. Transforming Growth Factor-Beta in Prostate Cancer. Prostate Cancer 2013. [DOI: 10.1007/978-1-4614-6828-8_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
45
|
Vo BT, Cody B, Cao Y, Khan SA. Differential role of Sloan-Kettering Institute (Ski) protein in Nodal and transforming growth factor-beta (TGF-β)-induced Smad signaling in prostate cancer cells. Carcinogenesis 2012; 33:2054-64. [PMID: 22843506 DOI: 10.1093/carcin/bgs252] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) signaling pathways contain both tumor suppressor and tumor promoting activities. We have demonstrated that Nodal, another member of the TGF-β superfamily, and its receptors are expressed in prostate cancer cells. Nodal and TGF-β exerted similar biological effects on prostate cells; both inhibited proliferation in WPE, RWPE1 and DU145 cells, whereas neither had any effect on the proliferation of LNCaP or PC3 cells. Interestingly, Nodal and TGF-β induced migration in PC3 cells, but not in DU145 cells. TGF-β induced predominantly phosphorylation of Smad3, whereas Nodal induced phosphorylation of only Smad2. We also determined the expression and differential role of Ski, a corepressor of Smad2/3, in Nodal and TGF-β signaling in prostate cancer cells. Similar levels of Ski mRNA were found in several established prostate cell lines; however, high levels of Ski protein were only detected in prostate cancer cells and prostate cancer tissue samples. Exogenous Nodal and TGF-β had no effects on Ski mRNA levels. On the other hand, TGF-β induced a rapid degradation of Ski protein mediated by the proteasomal pathway, whereas Nodal had no effect on Ski protein. Reduced Ski levels correlated with increased basal and TGF-β-induced Smad2/3 phosphorylation. Knockdown of endogenous Ski reduced proliferation in DU145 cells and enhanced migration of PC3 cells. We conclude that high levels of Ski expression in prostate cancer cells may be responsible for repression of TGF-β and Smad3 signaling, but Ski protein levels do not influence Nodal and Smad2 signaling.
Collapse
Affiliation(s)
- BaoHan T Vo
- Center for Cancer Research and Therapeutic Development, Clark Atlanta University, Atlanta, GA 30314, USA
| | | | | | | |
Collapse
|
46
|
Baranek C, Atanasoski S. Modulating epigenetic mechanisms: the diverse functions of Ski during cortical development. Epigenetics 2012; 7:676-9. [PMID: 22647289 DOI: 10.4161/epi.20590] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In the developing forebrain, neural stem and progenitor cells generate a large variety of neurons with specific functions in the mature cortex. A central issue is to understand the roles of transcriptional networks and regulatory pathways that control these complex developmental processes. The proto-oncogene Ski is a transcriptional regulator linked to the human 1p36 deletion syndrome, which involves a set of phenotypes including nervous system defects. Ski shows a dynamic expression pattern during cortical development and, accordingly, the phenotype of Ski-deficient cortices is complex, involving altered cell cycle characteristics of neural progenitors, disturbed timing of neurogenesis and mis-specification of projection neurons. Ski is likely to play a role in various pathways by virtue of its ability to interact with a range of signaling molecules, thereby modulating transcriptional activity of corresponding target genes. Ski regulates proliferation and differentiation of various cell types, and more recent data from my laboratory demonstrates that Ski is also involved in the specification of cortical projection neurons. This Point-of-View elucidates the role of Ski as an essential linker between sequence-specific transcription factors and non-DNA binding cofactors with chromatin modifying activities. In particular, it puts forward the hypothesis that the diverse functions of Ski as a co-repressor might be related to its association with distinct HDAC-complexes.
Collapse
Affiliation(s)
- Constanze Baranek
- Institute of Physiology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | | |
Collapse
|
47
|
Lee JH, Lee GT, Kwon SJ, Jeong J, Ha YS, Kim WJ, Kim IY. CREBZF, a novel Smad8-binding protein. Mol Cell Biochem 2012; 368:147-53. [PMID: 22707059 DOI: 10.1007/s11010-012-1353-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2012] [Accepted: 05/16/2012] [Indexed: 12/25/2022]
Abstract
Smads are the secondary messengers of the transforming growth factor-β (TGF-β) signaling pathway. TGF-β receptors phosphorylate the Receptor Smads (R-Smads) upon ligand binding; activated R-Smads translocate to the nucleus and function as transcription factors. Among the R-Smads, Smads 1, 5, and 8 mainly mediate signals in the bone morphogenetic proteins (BMPs) pathways, while Smads 2/3 mediate TGF-β signaling. The regulation of Smads in the TGF-β signal pathway has been well defined, but the relationship of Smads 1, 5, and 8 to the BMP pathways has been relatively understudied. To understand the specific regulation of BMP mediating Smads, we performed yeast two-hybrid screening using the Mad homology 2(MH2) domain of Smad8 as bait. In this screening, novel Smad-binding protein, CREBZF-a basic region-leucine zipper (bZIP) transcription factor-was identified. The interaction of CREBZF and Smads 1, 5, and 8 was confirmed by immunoprecipitation in a human prostate cancer cell line. Overexpression of CREBZF inhibited the promoter activity of BMP response element and abolished the cell growth inhibition induced by BMP-6. Thus, CREBZF inhibits the function of BMP-6 by interacting with Smads. The identification of this novel Smads-binding protein, among others will help us understand the modulation of BMP-signaling pathways.
Collapse
Affiliation(s)
- Jae-Ho Lee
- Section of Urologic Oncology and the Dean and Betty Gallo Prostate Cancer Center, The Cancer Institute of New Jersey and Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Zieba A, Pardali K, Söderberg O, Lindbom L, Nyström E, Moustakas A, Heldin CH, Landegren U. Intercellular variation in signaling through the TGF-β pathway and its relation to cell density and cell cycle phase. Mol Cell Proteomics 2012; 11:M111.013482. [PMID: 22442258 DOI: 10.1074/mcp.m111.013482] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fundamental open questions in signal transduction remain concerning the sequence and distribution of molecular signaling events among individual cells. In this work, we have characterized the intercellular variability of transforming growth factor β-induced Smad interactions, providing essential information about TGF-β signaling and its dependence on the density of cell populations and the cell cycle phase. By employing the recently developed in situ proximity ligation assay, we investigated the dynamics of interactions and modifications of Smad proteins and their partners under native and physiological conditions. We analyzed the kinetics of assembly of Smad complexes and the influence of cellular environment and relation to mitosis. We report rapid kinetics of formation of Smad complexes, including native Smad2-Smad3-Smad4 trimeric complexes, in a manner influenced by the rate of proteasomal degradation of these proteins, and we found a striking cell to cell variation of signaling complexes. The single-cell analysis of TGF-β signaling in genetically unmodified cells revealed previously unknown aspects of regulation of this pathway, and it provided a basis for analysis of these signaling events to diagnose pathological perturbations in patient samples and to evaluate their susceptibility to drug treatment.
Collapse
Affiliation(s)
- Agata Zieba
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Se-75185 Sweden
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ding B, Sun Y, Huang J. Overexpression of SKI oncoprotein leads to p53 degradation through regulation of MDM2 protein sumoylation. J Biol Chem 2012; 287:14621-30. [PMID: 22411991 DOI: 10.1074/jbc.m111.301523] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protooncogene Ski was identified based on its ability to transform avian fibroblasts in vitro. In support of its oncogenic activity, SKI was found to be overexpressed in a variety of human cancers, although the exact molecular mechanism(s) responsible for its oncogenic activity is not fully understood. We found that SKI can negatively regulate p53 by decreasing its level through up-regulation of MDM2 activity, which is mediated by the ability of SKI to enhance sumoylation of MDM2. This stimulation of MDM2 sumoylation is accomplished through a direct interaction of SKI with SUMO-conjugating enzyme E2, Ubc9, resulting in enhanced thioester bond formation and mono-sumoylation of Ubc9. A mutant SKI defective in transformation fails to increase p53 ubiquitination and is unable to increase MDM2 levels and to increase mono-sumoylation of Ubc9, suggesting that the ability of SKI to enhance Ubc9 activity is essential for its transforming function. These results established a detailed molecular mechanism that underlies the ability of SKI to cause cellular transformation while unraveling a novel connection between sumoylation and tumorigenesis, providing potential new therapeutic targets for cancer.
Collapse
Affiliation(s)
- Boxiao Ding
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | | | | |
Collapse
|
50
|
Toma I, McCaffrey TA. Transforming growth factor-β and atherosclerosis: interwoven atherogenic and atheroprotective aspects. Cell Tissue Res 2012; 347:155-75. [PMID: 21626289 PMCID: PMC4915479 DOI: 10.1007/s00441-011-1189-3] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 05/06/2011] [Indexed: 12/15/2022]
Abstract
Age-related progression of cardiovascular disease is by far the largest health problem in the US and involves vascular damage, progressive vascular fibrosis and the accumulation of lipid-rich atherosclerotic lesions. Advanced lesions can restrict flow to key organs and can trigger occlusive thrombosis resulting in a stroke or myocardial infarction. Transforming growth factor-beta (TGF-β) is a major orchestrator of the fibroproliferative response to tissue damage. In the early stages of repair, TGF-β is released from platelets and activated from matrix reservoirs; it then stimulates the chemotaxis of repair cells, modulates immunity and inflammation and induces matrix production. At later stages, it negatively regulates fibrosis through its strong antiproliferative and apoptotic effects on fibrotic cells. In advanced lesions, TGF-β might be important in arterial calcification, commonly referred to as "hardening of the arteries". Because TGF-β can signal through multiple pathways, namely the SMADs, a MAPK pathway and the Rho/ROCK pathways, selective defects in TGF-β signaling can disrupt otherwise coordinated pathways of tissue regeneration. TGF-β is known to control cell proliferation, cell migration, matrix synthesis, wound contraction, calcification and the immune response, all being major components of the atherosclerotic process. However, many of the effects of TGF-β are essential to normal tissue repair and thus, TGF-β is often thought to be "atheroprotective". The present review attempts to parse systematically the known effects of TGF-β on both the major risk factors for atherosclerosis and to isolate the role of TGF-β in the many component pathways involved in atherogenesis.
Collapse
Affiliation(s)
- Ian Toma
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 I Street NW. Ross Hall 443, Washington DC 20037, USA
| | - Timothy A. McCaffrey
- Department of Medicine, Division of Genomic Medicine, The George Washington University Medical Center, 2300 I Street NW. Ross Hall 443, Washington DC 20037, USA
| |
Collapse
|