1
|
Truchan K, Osyczka AM. Noggin promotes osteogenesis in human adipose-derived mesenchymal stem cells via FGFR2/Src/Akt and ERK signaling pathway. Sci Rep 2024; 14:6724. [PMID: 38509118 PMCID: PMC10954655 DOI: 10.1038/s41598-024-56858-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/12/2024] [Indexed: 03/22/2024] Open
Abstract
The balance between Noggin and bone morphogenetic proteins (BMPs) is important during early development and skeletal regenerative therapies. Noggin binds BMPs in the extracellular space, thereby preventing BMP signaling. However, Noggin may affect cell response not necessarily through the modulation of BMP signaling, raising the possibility of direct Noggin signaling through yet unspecified receptors. Here we show that in osteogenic cultures of adipose-derived stem cells (ASCs), Noggin activates fibroblast growth factor receptors (FGFRs), Src/Akt and ERK kinases, and it stabilizes TAZ proteins in the presence of dexamethasone. Overall, this leads ASCs to increased expression of osteogenic markers and robust mineral deposition. Our results also indicate that Noggin can induce osteogenic genes expression in normal human bone marrow stem cells and alkaline phosphatase activity in normal human dental pulp stem cells. Besides, Noggin can specifically activate FGFR2 in osteosarcoma cells. We believe our findings open new research avenues to further explore the involvement of Noggin in cell fate modulation by FGFR2/Src/Akt/ERK signaling and potential applications of Noggin in bone regenerative therapies.
Collapse
Affiliation(s)
- Karolina Truchan
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| | - Anna Maria Osyczka
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Jagiellonian University, Gronostajowa St. 9, 30-387, Kraków, Poland.
| |
Collapse
|
2
|
He W, Chen Z, Li H, Wu W, He P, Zhong D, Jiang Y, Cheng W, Xu Z, Li J. Decreased phosphorylation facilitates the degradation of the endogenous protective molecule c-Ski in vascular smooth muscle cells. Cell Signal 2021; 87:110116. [PMID: 34390788 DOI: 10.1016/j.cellsig.2021.110116] [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: 05/13/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 01/04/2023]
Abstract
The dysfunction of vascular smooth muscle cells (VSMCs) is critical for atherosclerosis (AS) progression. Autophagy is indispensable during phenotypic switching and proliferation of VSMCs, contribute to AS development. Cellular Sloan-Kettering Institute (c-Ski), the repressor of TGF-β signaling, is involved in diverse physiological and pathological processes. We previously defined c-Ski also as an endogenous protective molecule against AS via inhibiting abnormal proliferation and autophagy of VSMCs. However, the endogenous level of c-Ski in VSMCs is markedly decreased during the progression of AS, so that the protective effect is drastically weakened. Elucidating the molecular mechanisms is key to the understanding of AS development and treatment. We determined that oxidized low-density lipoprotein (ox-LDL) and platelet-derived growth factor (PDGF) directly induced the degradation of c-Ski protein, closely associated with reducing its phosphorylation. Serine383 (S383) was identified as the crucial phosphorylation site for stabilizing protein expression and nuclear location of c-Ski, which was responsible for its transcriptional suppression of autophagy-related genes. Decreased S383 phosphorylation facilitated nuclear export and degradation of c-Ski, thereby lessened its inhibitory effect on induction of autophagy genes. These findings provide a novel view of c-Ski modification and function modulation under some vascular injury factors, which point to a new potential therapeutic strategy by targeting c-Ski.
Collapse
Affiliation(s)
- Wenhui He
- Department of Biochemistry and Molecular Biology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zongtao Chen
- Health Management Centre, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Haoyang Li
- Brigade 5 of Medical Undergraduate, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | | | - Ping He
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Dan Zhong
- Department of Biochemistry and Molecular Biology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yu Jiang
- Department of Biochemistry and Molecular Biology, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
3
|
Zheng Y, Pei Y, Tang R, Zhou X, Feng Z, Li D, Chen H, Zeng Z, Jiang L, Cai J, Mao P, Wang L. ACBD3 is up-regulated in gastric cancer and promotes cell cycle G1-to-S transition in an AKT-dependent manner. Exp Cell Res 2021; 406:112752. [PMID: 34332983 DOI: 10.1016/j.yexcr.2021.112752] [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: 03/11/2021] [Revised: 06/06/2021] [Accepted: 07/26/2021] [Indexed: 12/01/2022]
Abstract
It has been reported that ACBD3 is closely related to the malignant process of cells, but its role in gastric cancer has not been elucidated. This study aims to investigate the expression and function of ACBD3 in human gastric cancer. The Cancer Genome Atlas (TCGA) database were selected to analyze mRNA levels of ACBD3 in gastric cancer tissues and normal gastric epithelial tissues. qPCR and Western blot were conducted to detect the expression of ACBD3 in two normal gastric epithelial cell lines and five gastric cancer cell lines which were cultured in our laboratory. To exclude differences in individual background between different patients, we further detected the expression of ACBD3 in 8 pairs of malignant/non-malignant clinical gastric tissues. Through the establishment of stable cells, in vitro cell experiments and in vivo xenotransplantation models in mice, the role of ACBD3 in the proliferation of gastric cancer cells has been further explored. AKT inhibitors were used to deeply explore the molecular regulation mechanism of ACBD3. The results showed that the elevated ACBD3 in gastric cancer tissue were positively correlated with the clinical grade and prognosis of gastric cancer. In terms of molecular function, we found that ACBD3 can enhance the production and growth of gastric cancer cells. At the same time, the activation of AKT kinase played an important role in ACBD3's promotion of G1-to-S transition. The experiments generally indicate that ACBD3 is expected to become a potential diagnostic molecule or therapeutic target for gastric cancer.
Collapse
Affiliation(s)
- Yingchun Zheng
- School of Life Sciences and Biopharmaceutics, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yuanyuan Pei
- Shenzhen Long-gang Maternal and Child Health Hospital Centralab, Shenzhen, 518172, China
| | - Ruiming Tang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou, 511518, China
| | - Xiulan Zhou
- Shenzhen Long-gang Maternal and Child Health Hospital Centralab, Shenzhen, 518172, China
| | - Zhengfu Feng
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou, 511518, China
| | - Difeng Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou, 511518, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Han Chen
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou, 511518, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhi Zeng
- School of Life Sciences and Biopharmaceutics, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lili Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Guangzhou, 511518, China; Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Key Laboratory of Protein Modification and Degradation, State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Junchao Cai
- Department of Immunology, Sun Yat-sen University, Zhongshan School of Medicine, Guangzhou, 510080, China.
| | - Pu Mao
- The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
| | - Lan Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
4
|
Zhao X, Fang Y, Wang X, Yang Z, Li D, Tian M, Kang P. Knockdown of Ski decreases osteosarcoma cell proliferation and migration by suppressing the PI3K/Akt signaling pathway. Int J Oncol 2019; 56:206-218. [PMID: 31746363 PMCID: PMC6910224 DOI: 10.3892/ijo.2019.4914] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/23/2019] [Indexed: 02/05/2023] Open
Abstract
Ski, an evolutionary conserved protein, is involved in the development of a number of tumors, such as Barrett's esophagus, leukemia, colorectal cancer, gastric cancer, pancreatic cancer, hemangiomas and melanoma. However, studies on the functions of Ski in osteosarcoma (OS) are limited. In this study, firstly the differential expression of Ski in OS tissues and osteochondroma tissues was detected, and the expression of Ski in both human OS cell lines (MG63 and U2OS) and normal osteoblasts (hFoB1.19) was then detected. The results demonstrated that Ski expression was significantly upregulated in both human OS tissues and cell lines. The results led us to hypothesize that Ski may play an essential role in the pathological process of OS. Thus, Ski specific small interfere RNA (Ski-siRNA) was used. The results revealed that OS cell proliferation was markedly inhibited following the knockdown of Ski, which was identified by CCK8 assay, EdU staining and cell cycle analysis. In addition, OS cell migration was significantly suppressed following Ski knockdown, which was identified by wound healing assay. Moreover, the protein levels of p-PI3K and p-Akt in OS cells declined prominently following Ski knockdown. On the whole, the findings of this study revealed that Ski expression was significantly upregulated in OS tissue and OS cells. The knockdown of Ski decreased OS cell proliferation and migration, which was mediated by blocking the PI3K/Akt signaling pathway. Thus, Ski may act as a tumor promoter gene in tumorigenesis, and Ski may prove to be a potential therapeutic target for the treatment of OS.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Yuying Fang
- Weifang Maternal and Child Health Hospital, Weifang, Shandong 261000, P.R. China
| | - Xingwen Wang
- The Second Clinical Medical College of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Zhouyuan Yang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Donghai Li
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Meng Tian
- Neurosurgery Research Laboratory, West China Hospital, Sichuan Univerisity, Chengdu, Sichuan 610041, P.R. China
| | - Pengde Kang
- Department of Orthopedic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| |
Collapse
|
5
|
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: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
6
|
Jiang B, Guan Y, Shen HJ, Zhang LH, Jiang JX, Dong XW, Shen HH, Xie QM. Akt/PKB signaling regulates cigarette smoke-induced pulmonary epithelial-mesenchymal transition. Lung Cancer 2018; 122:44-53. [DOI: 10.1016/j.lungcan.2018.05.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/19/2018] [Accepted: 05/21/2018] [Indexed: 01/31/2023]
|
7
|
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: 9.8] [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
|
8
|
Ji X, Hu X, Zou C, Ruan H, Fan X, Tang C, Shi W, Mei L, Zhu H, Hussain M, Zeng L, Zhang X, Wu X. Vitamin C deficiency exacerbates diabetic glomerular injury through activation of transforming growth factor-β signaling. Biochim Biophys Acta Gen Subj 2017; 1861:2186-2195. [PMID: 28652077 DOI: 10.1016/j.bbagen.2017.06.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Revised: 06/20/2017] [Accepted: 06/22/2017] [Indexed: 01/04/2023]
Abstract
BACKGROUND The hyperglycemia and hyperoxidation that characterize diabetes lead to reduced vitamin C (VC) in diabetic humans and experimentally diabetic animals. Herein, we access the effects of VC deficiency on the diabetic kidney injury and explore the underlying mechanism. METHODS l-gulonolactone oxidase conventional knockout (Gulo-/-) mice genetically unable to synthesize VC were subjected to streptozotocin-induced diabetic kidney injury and the role of VC deficiency was evaluated by biochemical and histological approaches. Rat mesangial cells were cultured to investigate the underlying mechanism. RESULTS Functionally, VC deficiency aggravates the streptozotocin-induced renal insufficiency, exhibiting the increased urine albumin, water intake, and urine volume in Gulo-/- mice. Morphologically, VC deficiency exacerbates the streptozotocin-induced kidney injury, exhibiting the increased glomerular expansion, deposition of Periodic Acid-Schiff- and Masson-positive materials, and expression of α-smooth muscle actin, fibronectin and type 4 collagen in glomeruli of Gulo-/- mice. Mechanistically, VC activates protein kinase B (Akt) to destabilize Ski and thereby induce the expression of Smad7, resulting in suppression of TGF-β/Smad signaling and extracellular matrix deposition in mesangial cells. CONCLUSIONS VC is essential for the renal function maintenance in diabetes. GENERAL SIGNIFICANCE Compensation for the loss of VC could be an effective remedy for diabetic kidney injury.
Collapse
Affiliation(s)
- Xing Ji
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Xinhua Hu
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Chaochun Zou
- Department of Endocrinology, the Affiliated Children Hospital, Zhejiang University Medical School, Hangzhou 310006, China
| | - Hongfeng Ruan
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Xueying Fan
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Chao Tang
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Wei Shi
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Liu Mei
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Haibin Zhu
- Department of Gynecology and Obstetrics, the First Affiliated Hospital, Zhejiang University Medical School, Hangzhou 310009, China
| | - Musaddique Hussain
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China
| | - Linghui Zeng
- Department of Pharmacology, Zhejiang University City College, Hangzhou 310023, China
| | - Xiaodong Zhang
- Department of Cell Biology, Wuhan University College of Life Science, Wuhan 430072, China
| | - Ximei Wu
- Department of Pharmacology, Zhejiang University Medical School, Hangzhou 310058, China.
| |
Collapse
|
9
|
Li J, Zou L, Zhou Y, Li L, Zhu Y, Yang Y, Gong Y, Lou J, Ke J, Zhang Y, Tian J, Zou D, Peng X, Chang J, Gong J, Zhong R, Zhou X, Miao X. A low-frequency variant in SMAD7 modulates TGF-β signaling and confers risk for colorectal cancer in Chinese population. Mol Carcinog 2017; 56:1798-1807. [PMID: 28218435 DOI: 10.1002/mc.22637] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 02/08/2017] [Accepted: 02/16/2017] [Indexed: 12/22/2022]
Abstract
The TGF-β pathway plays an essential role in regulating cell proliferation and differentiation. GWASs and candidate approaches have identified a battery of genetic variants in the TGF-β pathway contributing to colorectal cancer (CRC). However, most of the significant variants are common variants and their functions remain ambiguous. To identify causal variants with low-frequency in the TGF-β pathway contributing to CRC susceptibility in Chinese population, we performed targeted sequencing of 12 key genes in TGF-β signaling in CRC patients followed by a two-stage case-control study with a total of 5109 cases and 5169 controls. Bioinformatic annotations and biochemical experiments were applied to reveal the potential functions of significant variants. Seven low-frequency genetic variants were captured through targeted sequencing. The two stage association studies showed that missense variant rs3764482 (c. 83C>T; p. S28F) in the gene SMAD7 was consistently and significantly associated with CRC risk. Compared with the wild type, the ORs for variant allele were 1.37 (95%CI: 1.10-1.70, P = 0.005), 1.55 (95%CI: 1.30-1.86, P = 1.15 × 106 ), and 1.48 (1.29-1.70, P = 2.44 × 10;8 ) in stage 1, stage 2, and the combined analyses, respectively. Functional annotations revealed that the minor allele T of rs3764482 was more effective than the major allele C in blocking the TGF-β signaling and inhibiting the phosphorylation of receptor-regulated SMADs (R-SMADs). In conclusion, low-frequency coding variant rs3764482 in SMAD7 is associated with CRC risk in Chinese population. The rs3764482 variant may block the TGF-β signaling via impeding the activation of downstream genes, leading to cancer cell proliferation, thus contributing to CRC pathogenesis.
Collapse
Affiliation(s)
- Jiaoyuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Li Zou
- Department of Health Care, Bao'an Maternal and Child Health Hospital, Shenzhen, China
| | - Ying Zhou
- Institute of Orthopaedics, the First Affiliated Hospital of Chinese PLA General Hospial, Beijing, China
| | - Lu Li
- Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Ying Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Yang Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Yajie Gong
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Jiao Lou
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Juntao Ke
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Yi Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Jianbo Tian
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Danyi Zou
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Xiating Peng
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Jiang Chang
- Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Jing Gong
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Rong Zhong
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| | - Xiaobo Zhou
- Brigham and Women's Hospital, Channing Division of Network Medicine, Boston, Massachusetts
| | - Xiaoping Miao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Key Laboratory of Environment and Health, Tongji Medical College, Huazhong University of Science and Technology, Ministry of Education & Ministry of Environmental Protection, Wuhan, China
| |
Collapse
|
10
|
Rivas S, Armisén R, Rojas DA, Maldonado E, Huerta H, Tapia JC, Espinoza J, Colombo A, Michea L, Hayman MJ, Marcelain K. The Ski Protein is Involved in the Transformation Pathway of Aurora Kinase A. J Cell Biochem 2016; 117:334-43. [PMID: 26138431 DOI: 10.1002/jcb.25275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 06/29/2015] [Indexed: 12/19/2022]
Abstract
Oncogenic kinase Aurora A (AURKA) has been found to be overexpresed in several tumors including colorectal, breast, and hematological cancers. Overexpression of AURKA induces centrosome amplification and aneuploidy and it is related with cancer progression and poor prognosis. Here we show that AURKA phosphorylates in vitro the transcripcional co-repressor Ski on aminoacids Ser326 and Ser383. Phosphorylations on these aminoacids decreased Ski protein half-life. Reduced levels of Ski resulted in centrosomes amplification and multipolar spindles formation, same as AURKA overexpressing cells. Importantly, overexpression of Ski wild type, but not S326D and S383D mutants inhibited centrosome amplification and cellular transformation induced by AURKA. Altogether, these results suggest that the Ski protein is a target in the transformation pathway mediated by the AURKA oncogene.
Collapse
Affiliation(s)
- Solange Rivas
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Ricardo Armisén
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Diego A Rojas
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Edio Maldonado
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Hernán Huerta
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Julio C Tapia
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Jaime Espinoza
- Department of Pathology, UC-Center for Investigational Oncology (CITO), School of Medicine, Pontificia Universidad Católica de Chile 8330034, Santiago, Chile
| | - Alicia Colombo
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Luis Michea
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Millenium Institute of Immunology and Immunotherapy, Santiago, Chile
| | - Michael J Hayman
- Department of Microbiology and Molecular Genetics, Stony Brook University, Stony Brook, New York 11794
| | - Katherine Marcelain
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile.,Centro de Investigación y Tratamiento del Cáncer, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| |
Collapse
|
11
|
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: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/13/2015] [Indexed: 11/06/2022] Open
|
12
|
Xiao L, Peng X, Liu F, Tang C, Hu C, Xu X, Wang M, Luo Y, Yang S, Song P, Xiao P, Kanwar YS, Sun L. AKT regulation of mesothelial-to-mesenchymal transition in peritoneal dialysis is modulated by Smurf2 and deubiquitinating enzyme USP4. BMC Cell Biol 2015; 16:7. [PMID: 25885904 PMCID: PMC4369877 DOI: 10.1186/s12860-015-0055-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 02/19/2015] [Indexed: 02/24/2023] Open
Abstract
Background Transforming growth factor-β1 (TGF-β1) plays a key role in mesothelial-to-mesenchymal transition (MMT) during peritoneal dialysis (PD). However, the role of Akt in MMT transformation in PD is not clear. Results In this study, we observed that the phosphorylated form of protein kinase B (Akt), termed as pAkt, was up-regulated in the peritoneum of mice undergoing PD. It was associated with thickening of the peritoneum and up-regulation of TGF-β1. Upregulation of pAkt paralleled with the increased expression of Smad ubiquitination regulatory factor 2 (Smurf2), Vimentin and fibronectin (FN), and decreased expression of mothers against decapentaplegic homolog 7 (Smad7) and Zonula Occludens protein 1(ZO-1) in mice undergoing PD treatment and in TGF-β1 induced human peritoneal mesothelial cells (HPMCs). These changes were reversed with the treatment of a PI3K/Akt inhibitor LY294002 in vivo or in cells transfected with Akt dominant-negative (Akt-DN) plasmids in vitro. Increased Smurf2 expression in HPMCs, induced by TGF-β1 was accompanied with altered expression of Transforming growth factor receptor I (TβR-I), Smad7, ZO-1, Vimentin and FN via Akt modulation. In addition, inhibition of Ubiquitin carboxyl-terminal hydrolase 4 (USP4) decreased TGF- β1-induced expression of TβR-I and reversed the altered expression of Smad7, Smurf2, ZO-1 and Vimentin. Moreover, TGF-β1 accentuated the interactions between Smurf2 and Smad7, while reduced the association between TβR-I and Smurf2. These interactions were reversed by the treatment of Akt-DN and USP4 siRNA, respectively. Conclusions These data implied that Akt mediated MMT in PD via Smurf2 modulation/and or Smad7 degradation while conceivably maintaining the TβRI stability, most likely by the USP4.
Collapse
Affiliation(s)
- Li Xiao
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Xiang Peng
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Fuyou Liu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Chengyuan Tang
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Chun Hu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Xiaoxuan Xu
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Ming Wang
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Ying Luo
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Shikun Yang
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Panai Song
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Ping Xiao
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| | - Yashpal S Kanwar
- Departments of Pathology & Medicine, Northwestern University, Chicago, USA.
| | - Lin Sun
- Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
13
|
Amiri M, Jafari M, Azimzadeh Jamalkandi S, Davoodi SM. Atopic dermatitis-associated protein interaction network lead to new insights in chronic sulfur mustard skin lesion mechanisms. Expert Rev Proteomics 2014; 10:449-60. [PMID: 24117202 DOI: 10.1586/14789450.2013.841548] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic sulfur mustard skin lesions (CSMSLs) are the most common complications of sulfur mustard exposure; however, its mechanism is not completely understood.According to clinical signs, there are similarities between CSMSL and atopic dermatitis (AD). In this study, proteomic results of AD were reviewed and the AD-associated protein-protein interaction network (PIN) was analyzed. According to centrality measurements, 16 proteins were designated as pivotal elements in AD mechanisms. Interestingly, most of these proteins had been reported in some sulfur mustard-related studies in late and acute phases separately. Based on the gene enrichment analysis, aging, cell response to stress, cancer, Toll- and NOD-like receptor and apoptosis signaling pathways have the greatest impact on the disease. By the analysis of directed protein interaction networks, it is concluded that TNF, IL-6, AKT1, NOS3 and CDKN1A are the most important proteins. It is possible that these proteins play role in the shared complications of AD and CSMSL including xerosis and itching.
Collapse
Affiliation(s)
- Mojtaba Amiri
- Chemical Injuries Research Center, Baqiyatallah University of Medical Sciences, Tehran,P.O. 1949613711, Iran
| | | | | | | |
Collapse
|
14
|
Wang J, Zheng X, Zeng G, Zhou Y, Yuan H. Purified vitexin compound 1 inhibits growth and angiogenesis through activation of FOXO3a by inactivation of Akt in hepatocellular carcinoma. Int J Mol Med 2013; 33:441-8. [PMID: 24337611 DOI: 10.3892/ijmm.2013.1587] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2013] [Accepted: 12/02/2013] [Indexed: 11/06/2022] Open
Abstract
Vitexins, isolated from the seeds of the Chinese herb Vitex negundo, is known to exert antitumor activity in cancer xenograft models and cell lines. The aim of the current study was to examine whether the Akt/forkhead box protein O3a (FOXO3a) pathway mediates the biological effects of purified vitexin compound 1 (VB-1) in hepatocellular carcinoma (HCC) cells. The effect of VB-1 on the viability of the HCC cell lines HepG2, Hep3B, Huh-7 and the human embryonic liver cells L-02 was investigated using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. Growth inhibition was assessed by clonogenic assay, and cell cycle arrest was investigated using flow cytometry. Inhibition of angiogenesis was evaluated using a matrigel in vitro HUVEC tube formation assay. The effects on the Akt/FOXO3a pathway were detected by western blotting. VB-1 suppressed the proliferation of HepG2, Hep3B, Huh-7 cells, but had little effect on L-02 cells. VB-1 inhibited anchorage-dependent and -independent HepG2 cell growth in a concentration-dependent manner by induction of cell cycle arrest at G1/G0. VB-1 also reduced the secretion of vascular endothelial growth factor (VEGF), resulting in the inhibition of endothelial tube formation. Phosphorylated Akt and its downstream effector FOXO3a were downregulated in VB-1-treated HepG2 cells. Knockdown of Akt1 by small interfering RNA (siRNA) enhanced growth inhibition, and silencing FOXO3a by siRNA attenuated this action. VB-1 inhibited growth and induced cell cycle arrest at G1/G0 by regulating the Akt/FOXO3a pathway. The findings suggested that VB-1 is a potentially promising candidate for the treatment of HCC.
Collapse
Affiliation(s)
- Jiangang Wang
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Xingxing Zheng
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Guangyao Zeng
- School of Pharmaceutical Science, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yingjun Zhou
- School of Pharmaceutical Science, Central South University, Changsha, Hunan 410013, P.R. China
| | - Hong Yuan
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
15
|
Pingel J, Wienecke J, Kongsgaard M, Behzad H, Abraham T, Langberg H, Scott A. Increased mast cell numbers in a calcaneal tendon overuse model. Scand J Med Sci Sports 2013; 23:e353-60. [PMID: 23889295 PMCID: PMC4282450 DOI: 10.1111/sms.12089] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/02/2013] [Indexed: 02/06/2023]
Abstract
Tendinopathy is often discovered late because the initial development of tendon pathology is asymptomatic. The aim of this study was to examine the potential role of mast cell involvement in early tendinopathy using a high-intensity uphill running (HIUR) exercise model. Twenty-four male Wistar rats were divided in two groups: running group (n = 12); sedentary control group (n = 12). The running-group was exposed to the HIUR exercise protocol for 7 weeks. The calcaneal tendons of both hind limbs were dissected. The right tendon was used for histologic analysis using Bonar score, immunohistochemistry, and second harmonic generation microscopy (SHGM). The left tendon was used for quantitative polymerase chain reaction (qPCR) analysis. An increased tendon cell density in the runners were observed compared to the controls (P = 0.05). Further, the intensity of immunostaining of protein kinase B, P = 0.03; 2.75 ± 0.54 vs 1.17 ± 0.53, was increased in the runners. The Bonar score (P = 0.05), and the number of mast cells (P = 0.02) were significantly higher in the runners compared to the controls. Furthermore, SHGM showed focal collagen disorganization in the runners, and reduced collagen density (P = 0.03). IL-3 mRNA levels were correlated with mast cell number in sedentary animals. The qPCR analysis showed no significant differences between the groups in the other analyzed targets. The current study demonstrates that 7-week HIUR causes structural changes in the calcaneal tendon, and further that these changes are associated with an increased mast cell density.
Collapse
Affiliation(s)
- J Pingel
- Institute of Sports Medicine, Department of Orthopaedic Surgery M. Bispebjerg Hospital and Center for Healthy Aging, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
16
|
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: 0.9] [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
|
17
|
Forthun RB, SenGupta T, Skjeldam HK, Lindvall JM, McCormack E, Gjertsen BT, Nilsen H. Cross-species functional genomic analysis identifies resistance genes of the histone deacetylase inhibitor valproic acid. PLoS One 2012; 7:e48992. [PMID: 23155442 PMCID: PMC3498369 DOI: 10.1371/journal.pone.0048992] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2012] [Accepted: 10/03/2012] [Indexed: 01/01/2023] Open
Abstract
The mechanisms of successful epigenetic reprogramming in cancer are not well characterized as they involve coordinated removal of repressive marks and deposition of activating marks by a large number of histone and DNA modification enzymes. Here, we have used a cross-species functional genomic approach to identify conserved genetic interactions to improve therapeutic effect of the histone deacetylase inhibitor (HDACi) valproic acid, which increases survival in more than 20% of patients with advanced acute myeloid leukemia (AML). Using a bidirectional synthetic lethality screen revealing genes that increased or decreased VPA sensitivity in C. elegans, we identified novel conserved sensitizers and synthetic lethal interactors of VPA. One sensitizer identified as a conserved determinant of therapeutic success of HDACi was UTX (KDM6A), which demonstrates a functional relationship between protein acetylation and lysine-specific methylation. The synthetic lethal screen identified resistance programs that compensated for the HDACi-induced global hyper-acetylation, and confirmed MAPKAPK2, HSP90AA1, HSP90AB1 and ACTB as conserved hubs in a resistance program for HDACi that are drugable in human AML cell lines. Hence, these resistance hubs represent promising novel targets for refinement of combinatorial epigenetic anti-cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Emmet McCormack
- Institute of Medicine, Hematology Section, University of Bergen, Bergen, Norway
- Hematology Section, Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Bjørn Tore Gjertsen
- Institute of Medicine, Hematology Section, University of Bergen, Bergen, Norway
- Hematology Section, Department of Medicine, Haukeland University Hospital, Bergen, Norway
- * E-mail: (BTG); (HN)
| | - Hilde Nilsen
- The Biotechnology Centre, University of Oslo, Oslo, Norway
- * E-mail: (BTG); (HN)
| |
Collapse
|
18
|
Band AM, Laiho M. SnoN oncoprotein enhances estrogen receptor-α transcriptional activity. Cell Signal 2011; 24:922-30. [PMID: 22227247 DOI: 10.1016/j.cellsig.2011.12.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Accepted: 12/19/2011] [Indexed: 12/13/2022]
Abstract
Estrogen receptor-α (ERα) and transforming growth factor-beta (TGF-β) signaling pathways are essential regulators during mammary gland development and tumorigenesis. Ski-related novel gene (SnoN) is an oncoprotein and a negative feedback inhibitor of TGF-β signaling. We have previously reported that low expression of SnoN in ERα positive breast carcinomas is associated with favorable prognosis (Zhang et al. Cancer Res. (2003) 63, 5005-5010). Here we have studied the mechanism of a possible cross-talk between ERα and SnoN. We find that SnoN interacts with the estrogen-activated form of ERα in the nucleus. SnoN contains two highly conserved nuclear receptor binding LxxLL-like motifs and we show that mutations in these motifs reduce the interaction of SnoN with ERα. Over-expression of SnoN enhanced the transcriptional activity of ERα in estrogen response element (ERE)-reporter assays, augmented the expression of several ERα target genes and increased the proliferation of MCF7 breast carcinoma cells in an estrogen-dependent manner. Chromatin immunoprecipitation demonstrated that SnoN interacts with ERα at the TTF1 (pS2) gene promoter. Conversely, silencing of SnoN reduced both ERE-reporter activity and the expression of ERα target genes in MCF7 and T-47D breast cancer cells. Histone deacetylase inhibition increased the level of SnoN and SnoN-dependent enhancement of ERα-dependent transcription and SnoN supported the recruitment of p300 histone acetylase to ERα. This study reveals a novel mechanism that interconnects ERα and TGF-β signaling pathways by SnoN. Accordingly, the results indicate that high SnoN level promotes ERα signaling and possibly breast cancer progression.
Collapse
Affiliation(s)
- Arja M Band
- Molecular Cancer Biology Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
19
|
Fuentes-Calvo I, Blázquez-Medela AM, Eleno N, Santos E, López-Novoa JM, Martínez-Salgado C. H-Ras isoform modulates extracellular matrix synthesis, proliferation, and migration in fibroblasts. Am J Physiol Cell Physiol 2011; 302:C686-97. [PMID: 22094331 DOI: 10.1152/ajpcell.00103.2011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ras GTPases are ubiquitous plasma membrane transducers of extracellular stimuli. In addition to their role as oncogenes, Ras GTPases are key regulators of cell function. Each of the Ras isoforms exhibits specific modulatory activity on different cellular pathways. This has prompted researchers to determine the pathophysiological roles of each isoform. There is a proven relationship between the signaling pathways of transforming growth factor-β1 (TGF-β1) and Ras GTPases. To assess the individual role of H-Ras oncogene in basal and TGF-β1-mediated extracellular matrix (ECM) synthesis, proliferation, and migration in fibroblasts, we analyzed these processes in embryonic fibroblasts obtained from H-Ras knockout mice (H-ras(-/-)). We found that H-ras(-/-) fibroblasts exhibited a higher basal phosphatidylinositol-3-kinase (PI3K)/Akt activation than wild-type (WT) fibroblasts, whereas MEK/ERK 1/2 activation was similar in both types of cells. Fibronectin and collagen synthesis were higher in H-ras(-/-) fibroblasts and proliferation was lower in H-ras(-/-) than in WT fibroblasts. Moreover, H-Ras appeared indispensable to maintain normal fibroblast motility, which was highly restricted in H-ras(-/-) cells. These results suggest that H-Ras (through downregulation of PI3K/Akt activation) could modulate fibroblast activity by reducing ECM synthesis and upregulating both proliferation and migration. TGF-β1 strongly increased ERK and Akt activation in WT but not in H-ras(-/-) fibroblasts, suggesting that H-Ras is necessary to increase ERK 1/2 activation and to maintain PI3K downregulation in TGF-β1-stimulated fibroblasts. TGF-β1 stimulated ECM synthesis and proliferation, although ECM synthesis was higher and proliferation lower in H-ras(-/-) than in WT fibroblasts. Hence, H-Ras activation seems to play a key role in the regulation of these effects.
Collapse
Affiliation(s)
- Isabel Fuentes-Calvo
- Unidad de Fisiopatología Renal y Cardiovascular, Instituto “Reina Sofía” de Investigación Nefrológica, Departamento de Fisiología y Farmacología, Universidad de Salamanca, Salamanca, Spain
| | | | | | | | | | | |
Collapse
|
20
|
Bonnon C, Atanasoski S. c-Ski in health and disease. Cell Tissue Res 2011; 347:51-64. [DOI: 10.1007/s00441-011-1180-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 04/15/2011] [Indexed: 01/28/2023]
|
21
|
Band AM, Laiho M. Crosstalk of TGF-β and estrogen receptor signaling in breast cancer. J Mammary Gland Biol Neoplasia 2011; 16:109-15. [PMID: 21390570 DOI: 10.1007/s10911-011-9203-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2011] [Accepted: 02/28/2011] [Indexed: 01/17/2023] Open
Abstract
Estrogen receptor-α (ERα) and transforming growth factor (TGF)-β signaling pathways are major regulators during mammary gland development, function and tumorigenesis. Predominantly, they have opposing roles in proliferation and apoptosis. While ERα signaling supports growth and differentiation and is antiapoptotic, mammary gland epithelia cells are very sensitive to TGF-β-induced cell cycle arrest and apoptosis. Their regulatory pathways intersect, and ERα blocks TGF-β pathway by multiple means, including direct interactions of its signaling components, Smads. However, relatively little is known of the dysfunction of their interactions in cancer. A better understanding would help to develop new strategies for breast cancer treatment.
Collapse
Affiliation(s)
- Arja M Band
- Molecular Cancer Biology Program, Biomedicum Helsinki and Haartman Institute, University of Helsinki, Helsinki, Finland
| | | |
Collapse
|
22
|
Mosquera J, Armisen R, Zhao H, Rojas DA, Maldonado E, Tapia JC, Colombo A, Hayman MJ, Marcelain K. Identification of Ski as a target for Aurora A kinase. Biochem Biophys Res Commun 2011; 409:539-43. [PMID: 21600873 DOI: 10.1016/j.bbrc.2011.05.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 05/06/2011] [Indexed: 12/12/2022]
Abstract
Ski is a negative regulator of the transforming growth factor-β and other signalling pathways. The absence of SKI in mouse fibroblasts leads to chromosome segregation defects and genomic instability, suggesting a role for Ski during mitosis. At this stage, Ski is phosphorylated but to date little is known about the kinases involved in this process. Here, we show that Aurora A kinase is able to phosphorylate Ski in vitro. In vivo, Aurora A and Ski co-localized at the centrosomes and co-immunoprecipitated. Conversely, a C-terminal truncation mutant of Ski (SkiΔ491-728) lacking a coiled-coil domain, displayed decreased centrosomal localization. This mutant no longer co-immunoprecipitated with Aurora-A in vivo, but was still phosphorylated in vitro, indicating that the Ski-Aurora A interaction takes place at the centrosomes. These data identify Ski as a novel target of Aurora A and contribute to an understanding of the role of these proteins in the mitotic process.
Collapse
Affiliation(s)
- Jocelyn Mosquera
- Programa de Genética Humana, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Gardner S, Alzhanov D, Knollman P, Kuninger D, Rotwein P. TGF-β inhibits muscle differentiation by blocking autocrine signaling pathways initiated by IGF-II. Mol Endocrinol 2010; 25:128-37. [PMID: 21106882 DOI: 10.1210/me.2010-0292] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Skeletal muscle differentiation and regeneration are regulated by interactions between exogenous hormone- and growth factor-activated signaling cascades and endogenous muscle-specific transcriptional programs. IGF-I and IGF-II can promote muscle differentiation in vitro and can enhance muscle maintenance and repair in vivo. In contrast, members of the TGF-β superfamily prominently inhibit muscle differentiation and regeneration. In this study, we have evaluated functional interactions between IGF- and TGF-β-regulated signaling pathways during skeletal muscle differentiation. In the mouse C2 muscle cell line and in human myoblasts in primary culture, addition of TGF-β1 blocked differentiation in a dose-dependent way, inhibited expression of muscle-specific mRNAs and proteins, and impaired myotube formation. TGF-β1 also diminished stimulation of IGF-II gene expression in myoblasts, decreased IGF-II secretion, and reduced IGF-I receptor activation. To test the hypothesis that TGF-β1 prevents muscle differentiation primarily by blocking IGF-II production, we examined effects of IGF analogues on TGF-β actions in myoblasts. Although both IGF-I and IGF-II restored muscle gene and protein expression, and stimulated myotube formation in the presence of TGF-β1, they did not reduce TGF-β1-stimulated signaling, as measured by no decline in phosphorylation of SMA and mothers against decapentaplegic homolog (Smad)3, or in induction of TGF-β-activated target genes, including a Smad-dependent promoter-reporter plasmid. Our results demonstrate that TGF-β disrupts an IGF-II-stimulated autocrine amplification cascade that is necessary for muscle differentiation in vitro. Because this inhibitory pathway can be overcome by exogenous IGFs, our observations point toward potential strategies to counteract disorders that reduce muscle mass and strength.
Collapse
Affiliation(s)
- Samantha Gardner
- Department of Biochemistry and Molecular Biology, Oregon Health and Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239-3098, USA
| | | | | | | | | |
Collapse
|