151
|
Liu J, Wang C, Ma X, Tian Y, Wang C, Fu Y, Luo Y. High expression of CCR5 in melanoma enhances epithelial-mesenchymal transition and metastasis via TGFβ1. J Pathol 2019; 247:481-493. [PMID: 30474221 DOI: 10.1002/path.5207] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Revised: 10/16/2018] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
Chemokine receptors are highly expressed in various cancers and play crucial roles in tumor progression. However, their expression patterns and functions in melanoma are unclear. The present study aimed to identify the chemokine receptors that play critical roles in melanoma progression and unravel the underlying molecular mechanisms. We found that CCR5 was more abundant in melanoma cells than normal cells and was positively associated with tumor malignancy in clinical patients. Animal experiments suggested that CCR5 deficiency in B16/F10 or A375 cells suppressed primary tumor growth and lung metastasis, whereas CCR5 overexpression in B16/F0 cells enhanced primary tumor growth and lung metastasis. CCR5 played a critical role in proliferation and migration of melanoma cells in vitro. Importantly, CCR5 was required for maintenance of the mesenchymal phenotype of metastatic melanoma cells. Mechanistically, CCR5 positively regulated expression of TGFβ1, which in turn induced epithelial-mesenchymal transition and migration via PI3K/AKT/GSK3β signaling. Collectively, our results establish a critical role of CCR5 expressed by melanoma cells in cancer progression and reveal the novel mechanisms controlling this process, which suggests the prognostic value of CCR5 in melanoma patients and provides novel insights into CCR5-targeted strategies for melanoma treatment. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Jie Liu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Caihong Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Xuhui Ma
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yang Tian
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Chunying Wang
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yan Fu
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| | - Yongzhang Luo
- The National Engineering Laboratory for Anti-Tumor Protein Therapeutics, Tsinghua University, Beijing, PR China.,Beijing Key Laboratory for Protein Therapeutics, Tsinghua University, Beijing, PR China.,Cancer Biology Laboratory, School of Life Sciences, Tsinghua University, Beijing, PR China
| |
Collapse
|
152
|
Lin HY, Tey SL, Ho Y, Chin YT, Wang K, Whang-Peng J, Shih YJ, Chen YR, Yang YN, Chen YC, Liu YC, Tang HY, Yang YCS. Heteronemin Induces Anti-Proliferation in Cholangiocarcinoma Cells via Inhibiting TGF-β Pathway. Mar Drugs 2018; 16:md16120489. [PMID: 30563284 PMCID: PMC6316595 DOI: 10.3390/md16120489] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 12/13/2022] Open
Abstract
A marine sesterterpenoid-type natural product, heteronemin, retains anticancer effects. In the current study, we investigate the antitumor mechanism of heteronemin in cholangiocarcinoma cells and further explore its molecular targets. Initially, heteronemin exhibited potent cytotoxic effects against cholangiocarcinoma HuccT1 and SSP-25 cells. In vitro, heteronemin altered the abilities of cell adhesion and cell migration in HuccT1 and SSP-25 cell lines. It repressed messenger ribonucleic acid (mRNA) expression levels of transforming growth factor (TGF)-β, mothers against decapentaplegic homolog (SMAD) and Myc, whose protein products play important roles in regulating cell growth, angiogenesis, and metastasis. In addition, heteronemin altered several signaling pathways. The results indicate that heteronemin was able to modulate cell adhesion, the expression of extracellular matrix (ECM) receptors, the TGF-β pathway, cell motility, the membrane integration, metastasis response, matrix metalloproteinase (MMP) remodeling, the regulation of metabolism, sprouting angiogenesis, transcription factors, and vasculogenesis in cholangiocarcinoma cell lines. The results also suggest that it activated multiple signal transduction pathways to induce an anti-proliferation effect and anti-metastasis in cholangiocarcinoma. In conclusion, heteronemin may be used as a potential medicine for anticancer therapy.
Collapse
Affiliation(s)
- Hung-Yun Lin
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Traditional Herbal Medicine Research Center of Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan.
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| | - Shu-Leei Tey
- Department of Pediatrics, E-DA Hospital, Kaohsiung 824, Taiwan.
- School of Medicine, I-Shou University, Kaohsiung 824, Taiwan.
| | - Yih Ho
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yung-Tang Chin
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.
| | - Kuan Wang
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Jacqueline Whang-Peng
- Graduate Institute of Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- Cancer Center, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ya-Jung Shih
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yi-Ru Chen
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Nanomedicine and Medical Engineering, College of Medical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| | - Yung-Ning Yang
- Department of Pediatrics, E-DA Hospital, Kaohsiung 824, Taiwan.
- School of Medicine, I-Shou University, Kaohsiung 824, Taiwan.
| | - Yu-Cheng Chen
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung 404, Taiwan.
| | - Yi-Chang Liu
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
- Department of Internal Medicine, Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| | - Heng-Yuan Tang
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, NY 12208, USA.
| | - Yu-Chen Sh Yang
- Joint Biobank, Office of Human Research, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
153
|
TGF-β downregulation-induced cancer cell death is finely regulated by the SAPK signaling cascade. Exp Mol Med 2018; 50:1-19. [PMID: 30523245 PMCID: PMC6283885 DOI: 10.1038/s12276-018-0189-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 09/06/2018] [Accepted: 09/11/2018] [Indexed: 02/08/2023] Open
Abstract
Transforming growth factor (TGF)-β signaling is increasingly recognized as a key driver in cancer. In progressive cancer tissues, TGF-β promotes tumor formation, and its increased expression often correlates with cancer malignancy. In this study, we utilized adenoviruses expressing short hairpin RNAs against TGF-β1 and TGF-β2 to investigate the role of TGF-β downregulation in cancer cell death. We found that the downregulation of TGF-β increased the phosphorylation of several SAPKs, such as p38 and JNK. Moreover, reactive oxygen species (ROS) production was also increased by TGF-β downregulation, which triggered Akt inactivation and NOX4 increase-derived ROS in a cancer cell-type-specific manner. We also revealed the possibility of substantial gene fluctuation in response to TGF-β downregulation related to SAPKs. The expression levels of Trx and GSTM1, which encode inhibitory proteins that bind to ASK1, were reduced, likely a result of the altered translocation of Smad complex proteins rather than from ROS production. Instead, both ROS and ROS-mediated ER stress were responsible for the decrease in interactions between ASK1 and Trx or GSTM1. Through these pathways, ASK1 was activated and induced cytotoxic tumor cell death via p38/JNK activation and (or) induction of ER stress. Reducing the levels of the multifunctional protein transforming growth factor (TGF)-β in cancer cells prevents tumor growth in mice. Previous studies have shown that high levels of TGF-β in cancerous tissue are associated with accelerated disease progression. Hye Jin Choi and Jae J Song at Yonsei University in Seoul, South Korea, and colleagues infected cancer cells with genetically modified viruses that reduced the expression of the gene encoding TGF-β. The resulting decrease in TGF-β protein led to cell death by stimulating the production of reactive oxygen species and signaling through the apoptosis signal-regulating kinase 1 (ASK1) pathway. When tumor-bearing mice were infected with these modified viruses, their overall survival was improved. Further understanding the mechanisms through which TGF-β regulates cancer cell survival will contribute to the development of new approaches in cancer treatment.
Collapse
|
154
|
Rossato VV, Silveira DA, Gupta S, Mombach JCM. Towards the contribution of the p38MAPK pathway to the dual role of TGFβ in cancer: A boolean model approach. Comput Biol Med 2018; 104:235-240. [PMID: 30530226 DOI: 10.1016/j.compbiomed.2018.11.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 11/28/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022]
Abstract
The transforming growth factor-beta (TGF-β) pathway is involved in the regulation of cell growth and differentiation. In normal cells or in the early stages of cancer, this pathway can control proliferation stimuli by inducing cell cycle arrest or apoptosis (through the MAP-kinase protein p38MAPK), while in late stages it seems to act as a tumor promoter. This feature is known as the TGF-β dual role in cancer and it is not completely explained. This seems to arise through the accumulation of mutations in cancer development that affect the normal function of these pathways. In this work we propose a Boolean model of the crosstalk between the TGF-β, p38 MAPK and cell cycle checkpoint pathways which qualitatively describes this dual behavior. The model shows that for the wild type case, TGF-β acts as tumor supressor by inducing cell cycle arrest or apoptosis, as expected. However, the loss of function (LoF) of its two signaling proteins: SMAD2 and SMAD3 has immortalization effects due to the activation of the PI3K/AKT pathway that contributes to inhibit apoptosis. In silico mutations of the model elements were compared with cell phenotypes in experiments presenting agreement. In addition, we performed a series of double gene perturbations (that simulate random deleterious mutations) to determine the main regulators of the network. The results suggest that SMAD2/3 and p38MAPK are key players in processing the network input. In addition, when the LoF of SMAD2/3 is combined with the LoF of p38MAPK and p53, cell cycle arrest is completely abrogated. In conclusion, the model allows to visualize, through in silico mutations, the dual role of TGF-β: for the wild-type case TGF-β is able to block proliferation, however deleterious mutations can impair cell cycle arrest promoting cellular proliferation.
Collapse
Affiliation(s)
| | - Daner A Silveira
- Departamento de Física, Universidade Federal de Santa Maria, Brazil
| | - Shantanu Gupta
- Departamento de Física, Universidade Federal de Santa Maria, Brazil
| | | |
Collapse
|
155
|
Kurtzeborn K, Cebrian C, Kuure S. Regulation of Renal Differentiation by Trophic Factors. Front Physiol 2018; 9:1588. [PMID: 30483151 PMCID: PMC6240607 DOI: 10.3389/fphys.2018.01588] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Classically, trophic factors are considered as proteins which support neurons in their growth, survival, and differentiation. However, most neurotrophic factors also have important functions outside of the nervous system. Especially essential renal growth and differentiation regulators are glial cell line-derived neurotrophic factor (GDNF), bone morphogenetic proteins (BMPs), and fibroblast growth factors (FGFs). Here we discuss how trophic factor-induced signaling contributes to the control of ureteric bud (UB) branching morphogenesis and to maintenance and differentiation of nephrogenic mesenchyme in embryonic kidney. The review includes recent advances in trophic factor functions during the guidance of branching morphogenesis and self-renewal versus differentiation decisions, both of which dictate the control of kidney size and nephron number. Creative utilization of current information may help better recapitulate renal differentiation in vitro, but it is obvious that significantly more basic knowledge is needed for development of regeneration-based renal therapies.
Collapse
Affiliation(s)
- Kristen Kurtzeborn
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
| | - Cristina Cebrian
- Developmental Biology Division, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Satu Kuure
- Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Medicum, University of Helsinki, Helsinki, Finland
- GM-Unit, Laboratory Animal Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| |
Collapse
|
156
|
Kim S, Ham S, Yang K, Kim K. Protein kinase CK2 activation is required for transforming growth factor β-induced epithelial-mesenchymal transition. Mol Oncol 2018; 12:1811-1826. [PMID: 30171795 PMCID: PMC6165993 DOI: 10.1002/1878-0261.12378] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 07/29/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
Transforming growth factor β (TGFβ) is overexpressed in advanced cancers and promotes tumorigenesis by inducing epithelial–mesenchymal transition (EMT), which enhances invasiveness and metastasis. Although we previously reported that EMT could be induced by increasing CK2 activity alone, it is not known whether CK2 also plays an essential role in TGFβ‐induced EMT. Therefore, in the present study, we investigated whether TGFβ signaling could activate CK2 and, if so, whether such activation is required for TGFβ‐induced EMT. We found that CK2 is activated by TGFβ treatment, and that activity peaks at 48 h after treatment. CK2 activation is dependent on TGFβ receptor (TGFBR) I kinase activity, but independent of SMAD4. Inhibition of CK2 activation through the use of either a CK2 inhibitor or shRNA against CSNK2A1 inhibited TGFβ‐induced EMT. TGFβ signaling decreased CK2β but did not affect CK2α protein levels, resulting in a quantitative imbalance between the catalytic α and regulatory β subunits, thereby increasing CK2 activity. The decrease in CK2β expression was dependent on TGFBRI kinase activity and the ubiquitin–proteasome pathway. The E3 ubiquitin ligases responsible for TGFβ‐induced CK2β degradation were found to be CHIP and WWP1. Okadaic acid (OA) pretreatment protected CK2β from TGFβ‐induced degradation, suggesting that dephosphorylation of CK2β by an OA‐sensitive phosphatase might be required for CK2 activation in TGFβ‐induced EMT. Collectively, our results suggest CK2 as a therapeutic target for the prevention of EMT and metastasis of cancers.
Collapse
Affiliation(s)
- Seongrak Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea.,Integrated Genomic Research Center for Metabolic Regulation, Seoul, Korea
| | - Sunyoung Ham
- Quality Evaluation Team, Samsung Bioepis, Incheon, Korea
| | - Kyungmi Yang
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea
| | - Kunhong Kim
- Department of Biochemistry and Molecular Biology, Yonsei University College of Medicine, Seoul, Korea.,Integrated Genomic Research Center for Metabolic Regulation, Seoul, Korea
| |
Collapse
|
157
|
Sun Y, Jiang X, Lu Y, Zhu J, Yu L, Ma B, Zhang Q. Oridonin prevents epithelial-mesenchymal transition and TGF-β1-induced epithelial-mesenchymal transition by inhibiting TGF-β1/Smad2/3 in osteosarcoma. Chem Biol Interact 2018; 296:57-64. [PMID: 30243739 DOI: 10.1016/j.cbi.2018.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/02/2018] [Accepted: 09/18/2018] [Indexed: 01/08/2023]
Abstract
Osteosarcoma is the most common primary bone tumor with highly invasive characteristic and low long-term survival. Recently, epithelial-mesenchymal transition (EMT) is reported as a key event in cancer invasion and metastasis. Oridonin, a bioactive diterpenoid, has been proved to possess anti-cancer effects. However, the effect of oridonin on EMT and metastasis of osteosarcoma is unclear. In this study, we investigated the underlying mechanism of oridonin on EMT and metastasis of osteosarcoma. We found that oridonin inhibited migration and invasion of MG-63 and 143B cells. Moreover, oridonin increased the protein expression of E-cadherin and decreased that of N-cadherin and Vimentin. Oridonin upregulated the transcription of E-cadherin and downregulated N-cadherin and Vimentin. Oridonin inhibited the protein and mRNA levels of Snail and Slug. Furthermore, oridonin inhibited TGF-β-induced phosphorylation of Smad 2/3, prevented Smad dimer translocation into the nucleus. Finally, we established metastatic models of osteosarcoma 143B cells, and found that oridonin inhibited lung metastasis in vivo. Oridonin increased the protein expression of E-cadherin and reduced N-cadherin and Vimentin. Oridonin inhibited the protein expression of Snail and Slug as well as Smad 2/3 activation. In conclusion, our study demonstrated that oridonin inhibited EMT and TGF-β1-induced EMT by inhibiting TGF-β1/Smad2/3 signaling pathway in osteosarcoma.
Collapse
Affiliation(s)
- Yang Sun
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China.
| | - Xiubo Jiang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Ying Lu
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Jianwei Zhu
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Lisha Yu
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Bo Ma
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China
| | - Qi Zhang
- School of Pharmaceutical Sciences, Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing, 211816, PR China.
| |
Collapse
|
158
|
MiR‐499 regulates myoblast proliferation and differentiation by targeting transforming growth factor β receptor 1. J Cell Physiol 2018; 234:2523-2536. [DOI: 10.1002/jcp.26903] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 06/12/2018] [Indexed: 12/21/2022]
|
159
|
TGF-β receptors: In and beyond TGF-β signaling. Cell Signal 2018; 52:112-120. [PMID: 30184463 DOI: 10.1016/j.cellsig.2018.09.002] [Citation(s) in RCA: 288] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 08/07/2018] [Accepted: 09/01/2018] [Indexed: 02/07/2023]
Abstract
Transforming growth factor β (TGF-β) plays an important role in normal development and homeostasis. Dysregulation of TGF-β responsiveness and its downstream signaling pathways contribute to many diseases, including cancer initiation, progression, and metastasis. TGF-β ligands bind to three isoforms of the TGF-β receptor (TGFBR) with different affinities. TGFBR1 and 2 are both serine/threonine and tyrosine kinases, but TGFBR3 does not have any kinase activity. They are necessary for activating canonical or noncanonical signaling pathways, as well as for regulating the activation of other signaling pathways. Another prominent feature of TGF-β signaling is its context-dependent effects, temporally and spatially. The diverse effects and context dependency are either achieved by fine-tuning the downstream components or by regulating the expressions and activities of the ligands or receptors. Focusing on the receptors in events in and beyond TGF-β signaling, we review the membrane trafficking of TGFBRs, the kinase activity of TGFBR1 and 2, the direct interactions between TGFBR2 and other receptors, and the novel roles of TGFBR3.
Collapse
|
160
|
Ohta M, Chosa N, Kyakumoto S, Yokota S, Okubo N, Nemoto A, Kamo M, Joh S, Satoh K, Ishisaki A. IL‑1β and TNF‑α suppress TGF‑β‑promoted NGF expression in periodontal ligament‑derived fibroblasts through inactivation of TGF‑β‑induced Smad2/3‑ and p38 MAPK‑mediated signals. Int J Mol Med 2018; 42. [PMID: 29901090 PMCID: PMC6089780 DOI: 10.3892/ijmm_2018.3714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Mechanosensitive (MS) neurons in the periodontal ligament (PDL) pass information to the trigeminal ganglion when excited by mechanical stimulation of the tooth. During occlusal tooth trauma of PDL tissues, MS neurons are injured, resulting in atrophic neurites and eventual degeneration of MS neurons. Nerve growth factor (NGF), a neurotrophic factor, serves important roles in the regeneration of injured sensory neurons. In the present study, the effect of pro‑inflammatory cytokines, including interleukin 1β (IL‑1β) and tumor necrosis factor α (TNF‑α), on transforming growth factor β1 (TGF‑β1)‑induced NGF expression was evaluated in rat PDL‑derived SCDC2 cells. It was observed that TGF‑β1 promoted NGF expression via Smad2/3 and p38 mitogen‑activated protein kinase (MAPK) activation. IL‑1β and TNF‑α suppressed the TGF‑β1‑induced activation of Smad2/3 and p38 MAPK, resulting in the abrogation of NGF expression. NGF secreted by TGF‑β1‑treated SCDC2 cells promoted neurite extension and the expression of tyrosine hydroxylase, a rate‑limiting enzyme in dopamine synthesis in rat pheochromocytoma PC12 cells. These results suggested that pro‑inflammatory cytokines suppressed the TGF‑β‑mediated expression of NGF in PDL‑derived fibroblasts through the inactivation of TGF‑β‑induced Smad2/3 and p38 MAPK signaling, possibly resulting in the disturbance of the regeneration of injured PDL neurons.
Collapse
Affiliation(s)
- Maiko Ohta
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694,Division of Dental Anesthesia, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University, Morioka, Iwate 020-8505
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694
| | - Seiko Kyakumoto
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694
| | - Seiji Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694
| | - Naoto Okubo
- Laboratory of Pathophysiology and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-ku, Sapporo 060-0812
| | - Akira Nemoto
- Division of Operative Dentistry and Endodontics, Department of Conservative Dentistry
| | - Masaharu Kamo
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694
| | - Shigeharu Joh
- Division of Oral and Dysphasia Rehabilitation, Department of Prosthodontics, Iwate Medical University, Morioka, Iwate 020-8505, Japan
| | - Kenichi Satoh
- Division of Dental Anesthesia, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University, Morioka, Iwate 020-8505
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa-gun, Iwate 028-3694,Correspondence to: Dr Akira Ishisaki, Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan, E-mail:
| |
Collapse
|
161
|
Ma JY, You D, Li WY, Lu XL, Sun S, Li HW. Bone morphogenetic proteins and inner ear development. J Zhejiang Univ Sci B 2018; 20:131-145. [PMID: 30112880 DOI: 10.1631/jzus.b1800084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone morphogenetic proteins (BMPs) are the largest subfamily of the transforming growth factor-β superfamily, and they play important roles in the development of numerous organs, including the inner ear. The inner ear is a relatively small organ but has a highly complex structure and is involved in both hearing and balance. Here, we discuss BMPs and BMP signaling pathways and then focus on the role of BMP signal pathway regulation in the development of the inner ear and the implications this has for the treatment of human hearing loss and balance dysfunction.
Collapse
Affiliation(s)
- Jiao-Yao Ma
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Dan You
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Wen-Yan Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Xiao-Ling Lu
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Shan Sun
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China
| | - Hua-Wei Li
- Ear, Nose & Throat Institute and Otorhinolaryngology Department of Affiliated Eye and ENT Hospital, NHC Key Laboratory of Hearing Medicine, Shanghai Engineering Research Centre of Cochlear Implant, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200031, China.,Institutes of Biomedical Sciences and the Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
162
|
Ricard-Blum S, Baffet G, Théret N. Molecular and tissue alterations of collagens in fibrosis. Matrix Biol 2018; 68-69:122-149. [DOI: 10.1016/j.matbio.2018.02.004] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 02/02/2018] [Accepted: 02/02/2018] [Indexed: 02/07/2023]
|
163
|
Li P, Liu P, Peng Y, Zhang ZH, Li XM, Xiong RP, Chen X, Zhao Y, Ning YL, Yang N, Zhang B, Zhou YG. The ERK/CREB pathway is involved in the c-Ski expression induced by low TGF-β1 concentrations during primary fibroblast proliferation. Cell Cycle 2018; 17:1319-1328. [PMID: 29950153 DOI: 10.1080/15384101.2018.1480221] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
Abstract
Increasing evidence has suggested that bidirectional regulation of cell proliferation is one important effect of TGF-β1 in wound healing. Increased c-Ski expression plays a role in promoting fibroblast proliferation at low TGF-β1 concentrations, but the mechanism by which low TGF-β1 concentrations regulate c-Ski levels remains unclear. In this study, the proliferation of rat primary fibroblasts was assessed with an ELISA BrdU kit. The mRNA and protein expression and phosphorylation levels of corresponding factors were measured by RT-qPCR, immunohistochemistry or Western blotting. We first found that low TGF-β1 concentrations not only promoted c-ski mRNA and protein expression in rat primary fibroblasts but also increased the phosphorylation levels of Extracellular Signal-Regulated Kinases (ERK) and cAMP response element binding (CREB) protein. An ERK kinase (mitogen-activated protein kinase kinase, MEK) inhibitor significantly inhibited ERK1/2 phosphorylation levels, markedly reducing c-Ski expression and CREB phosphorylation levels and abrogating the growth-promoting effect of low TGF-β1 concentrations. At the same time, Smad2/3 phosphorylation levels were not significantly changed. Taken together, these results suggest that the increased cell proliferation induced by low TGF-β1 concentrations mediates c-Ski expression potentially through the ERK/CREB pathway rather than through the classic TGF-β1/Smad pathway.
Collapse
Affiliation(s)
- Ping Li
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Ping Liu
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China.,b Department 4, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Yan Peng
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Zhuo-Hang Zhang
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Xiao-Ming Li
- b Department 4, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Ren-Ping Xiong
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Xing Chen
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Yan Zhao
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Ya-Lei Ning
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Nan Yang
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Bo Zhang
- b Department 4, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| | - Yuan-Guo Zhou
- a The Molecular Biology Center, State Key Laboratory of Trauma, Burn and Combined Injury, Research Institute of Surgery and Daping Hospital , Third Military Medical University , Chongqing , People's Republic of China
| |
Collapse
|
164
|
Takeda N, Hara H, Fujiwara T, Kanaya T, Maemura S, Komuro I. TGF-β Signaling-Related Genes and Thoracic Aortic Aneurysms and Dissections. Int J Mol Sci 2018; 19:ijms19072125. [PMID: 30037098 PMCID: PMC6073540 DOI: 10.3390/ijms19072125] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
Transforming growth factor-β (TGF)-β signaling plays a crucial role in the development and maintenance of various organs, including the vasculature. Accordingly, the mutations in TGF-β signaling pathway-related genes cause heritable disorders of the connective tissue, such as Marfan syndrome (MFS), Loeys-Dietz syndrome (LDS), and Shprintzen-Goldberg syndrome (SGS), and these syndromes may affect skeletal, ocular, pulmonary, and cardiovascular systems. Aortic root aneurysms are common problems that can result in aortic dissection or rupture, which is the leading cause of sudden death in the natural history of MFS and LDS, and recent improvements in surgical treatment have improved life expectancy. However, there is currently no genotype-specific medical treatment. Accumulating evidence suggest that not only structural weakness of connective tissue but also increased TGF-β signaling contributes to the complicated pathogenesis of aortic aneurysm formation, but a comprehensive understanding of governing molecular mechanisms remains lacking. Inhibition of angiotensin II receptor signaling and endothelial dysfunction have gained attention as a possible MFS treatment strategy, but interactions with TGF-β signaling remain elusive. Heterozygous loss-of-function mutations in TGF-β receptors 1 and 2 (TGFBR1 and TGFBR2) cause LDS, but TGF-β signaling is activated in the aorta (referred to as the TGF-β paradox) by mechanisms yet to be elucidated. In this review, we present and discuss the current understanding of molecular mechanisms responsible for aortopathies of MFS and related disorders.
Collapse
Affiliation(s)
- Norifumi Takeda
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Hironori Hara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Takayuki Fujiwara
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Tsubasa Kanaya
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Sonoko Maemura
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo Hospital, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan.
| |
Collapse
|
165
|
Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND, Zhao YY. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem Biol Interact 2018; 292:76-83. [PMID: 30017632 DOI: 10.1016/j.cbi.2018.07.008] [Citation(s) in RCA: 679] [Impact Index Per Article: 113.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Revised: 07/01/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β1 (TGF-β1) is considered as a crucial mediator in tissue fibrosis and causes tissue scarring largely by activating its downstream small mother against decapentaplegic (Smad) signaling. Different TGF-β signalings play different roles in fibrogenesis. TGF-β1 directly activates Smad signaling which triggers pro-fibrotic gene overexpression. Excessive studies have demonstrated that dysregulation of TGF-β1/Smad pathway was an important pathogenic mechanism in tissue fibrosis. Smad2 and Smad3 are the two major downstream regulator that promote TGF-β1-mediated tissue fibrosis, while Smad7 serves as a negative feedback regulator of TGF-β1/Smad pathway thereby protects against TGF-β1-mediated fibrosis. This review presents an overview of the molecular mechanisms of TGF-β/Smad signaling pathway in renal, hepatic, pulmonary and cardiac fibrosis, followed by an in-depth discussion of their molecular mechanisms of intervention effects both in vitro and in vivo. The role of TGF-β/Smad signaling pathway in tumor or cancer is also discussed. Additionally, the current advances also highlight targeting TGF-β/Smad signaling pathway for the prevention of tissue fibrosis. The review reveals comprehensive pathophysiological mechanisms of tissue fibrosis. Particular challenges are presented and placed within the context of future applications against tissue fibrosis.
Collapse
Affiliation(s)
- He-He Hu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Dan-Qian Chen
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Yan-Ni Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Ya-Long Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, No. 548 Binwen Road, Hangzhou, Zhejiang, 310053, China
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, School of Medicine, University of California Irvine, Irvine, CA, 92897, USA
| | - Ying-Yong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Science, Northwest University, No. 229 Taibai North Road, Xi'an, Shaanxi, 710069, China.
| |
Collapse
|
166
|
Li C, Rezov V, Joensuu E, Vartiainen V, Rönty M, Yin M, Myllärniemi M, Koli K. Pirfenidone decreases mesothelioma cell proliferation and migration via inhibition of ERK and AKT and regulates mesothelioma tumor microenvironment in vivo. Sci Rep 2018; 8:10070. [PMID: 29968778 PMCID: PMC6030186 DOI: 10.1038/s41598-018-28297-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
Malignant mesothelioma is an aggressive cancer with poor prognosis. It is characterized by prominent extracellular matrix, mesenchymal tumor cell phenotypes and chemoresistance. In this study, the ability of pirfenidone to alter mesothelioma cell proliferation and migration as well as mesothelioma tumor microenvironment was evaluated. Pirfenidone is an anti-fibrotic drug used in the treatment of idiopathic pulmonary fibrosis and has also anti-proliferative activities. Mesothelioma cell proliferation was decreased by pirfenidone alone or in combination with cisplatin. Pirfenidone also decreased significantly Transwell migration/invasion and 3D collagen invasion. This was associated with increased BMP pathway activity, decreased GREM1 expression and downregulation of MAPK/ERK and AKT/mTOR signaling. The canonical Smad-mediated TGF-β signaling was not affected by pirfenidone. However, pirfenidone blocked TGF-β induced upregulation of ERK and AKT pathways. Treatment of mice harboring mesothelioma xenografts with pirfenidone alone did not reduce tumor proliferation in vivo. However, pirfenidone modified the tumor microenvironment by reducing the expression of extracellular matrix associated genes. In addition, GREM1 expression was downregulated by pirfenidone in vivo. By reducing two major upregulated pathways in mesothelioma and by targeting tumor cells and the microenvironment pirfenidone may present a novel anti-fibrotic and anti-cancer adjuvant therapy for mesothelioma.
Collapse
Affiliation(s)
- Chang Li
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Soochow, China
| | - Veronika Rezov
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Emmi Joensuu
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Ville Vartiainen
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.,University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Mikko Rönty
- Department of Pathology, University of Helsinki and Fimlab laboratories, Pathology, Tampere, Finland
| | - Miao Yin
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland
| | - Marjukka Myllärniemi
- University of Helsinki and Helsinki University Hospital, Heart and Lung Center and HUH diagnostics, Pulmonary Medicine, Helsinki, Finland
| | - Katri Koli
- Research Programs Unit, Translational Cancer Biology, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
167
|
Zhu X, Gao Q, Zhao G, Wang H, Liu L, Chen Z, Chen Y, Wu L, Xu Z, Li W. Comparison Study of Bone Defect Healing Effect of Raw and Processed Pyritum in Rats. Biol Trace Elem Res 2018; 184:136-147. [PMID: 28980123 DOI: 10.1007/s12011-017-1166-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 09/25/2017] [Indexed: 01/06/2023]
Abstract
To evaluate and compare the effect of raw and processed pyritum on tibial defect healing, 32 male Sprague Dawley rats were randomly divided into four groups. After tibial defect, animals were produced and grouped: sham and control group were orally administrated with distilled water (1 mL/100 g), while treatment groups were given aqueous extracts of raw and processed pyritum (1.5 g/kg) for successive 42 days. Radiographic examination showed that bone defect healing effect of the treatment groups was obviously superior compared to that of the control group. Bone mineral density of whole tibia was increased significantly after treating with pyritum. Inductively coupled plasma-optical emission spectrometry showed that the contents of Ca, P, and Mg in callus significantly increased in the treatment groups comparing with the control. Moreover, serological analysis showed that the concentration of serum phosphorus of the treatment groups significantly increased compared with that of the control group. By in vitro study, we have evaluated the effects of drug-containing serum of raw and processed pyritum on osteoblasts. It was manifested that both the drug-containing sera of raw and processed pyritum significantly increased the mRNA levels of alkaline phosphatase and collagen type I. Protein levels of phosphorylated Smad2/3 also increased. The mRNA levels of osteocalcin and transforming growth factor β (TGF-β) type I and II receptors, as well as the protein levels of TGF-β1 in the processed groups, were higher than those in the control. In summary, both raw and processed pyritum-containing sera exhibited positive effects on osteoblasts, which maybe via the TGF-β1/Smad signaling pathway. Notably, the tibia defect healing effect of pyritum was significantly enhanced after processing.
Collapse
Affiliation(s)
- Xingyu Zhu
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Qianqian Gao
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Genhua Zhao
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Heng Wang
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Ling Liu
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Zhipeng Chen
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yijun Chen
- Modern Analysis Center of Nanjing University, Nanjing, China
| | - Li Wu
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China
| | - Zisheng Xu
- Wuhu Pure Sunshine Natural Medicine Company Limited, Wuhu, Anhui, People's Republic of China.
| | - Weidong Li
- College of Pharmacy, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing, 210023, Jiangsu, People's Republic of China.
- Engineering Center of State Ministry of Education for Standardization of Chinese Medicine Processing, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
168
|
Abstract
Transforming Growth Factor beta (TGF-β) is a pleiotropic cytokine produced in large amounts within cancer microenvironments that will ultimately promote neoplastic progression, notably by suppressing the host’s T-cell immunosurveillance. This effect is mostly due to the well-known inhibitory effect of TGF-β on T cell proliferation, activation, and effector functions. Moreover, TGF-β subverts T cell immunity by favoring regulatory T-cell differentiation, further reinforcing immunosuppression within tumor microenvironments. These findings stimulated the development of many strategies to block TGF-β or its signaling pathways, either as monotherapy or in combination with other therapies, to restore anti-cancer immunity. Paradoxically, recent studies provided evidence that TGF-β can also promote differentiation of certain inflammatory populations of T cells, such as Th17, Th9, and resident-memory T cells (Trm), which have been associated with improved tumor control in several models. Here, we review current advances in our understanding of the many roles of TGF-β in T cell biology in the context of tumor immunity and discuss the possibility to manipulate TGF-β signaling to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Amina Dahmani
- Centre de Recherche de L'hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, QC H1T 2M4, Canada.
| | - Jean-Sébastien Delisle
- Centre de Recherche de L'hôpital Maisonneuve-Rosemont, 5415 Boul. de L'Assomption, Montréal, QC H1T 2M4, Canada.
- Hematology-Oncology service, Hôpital Maisonneuve-Rosemont, Department of Medicine, Université de Montréal, Montréal, QC H1T 2M4, Canada.
| |
Collapse
|
169
|
TGF-β in T Cell Biology: Implications for Cancer Immunotherapy. Cancers (Basel) 2018; 10:cancers10060194. [PMID: 29891791 PMCID: PMC6025055 DOI: 10.3390/cancers10060194] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 06/07/2018] [Accepted: 06/07/2018] [Indexed: 12/25/2022] Open
Abstract
Transforming Growth Factor beta (TGF-β) is a pleiotropic cytokine produced in large amounts within cancer microenvironments that will ultimately promote neoplastic progression, notably by suppressing the host’s T-cell immunosurveillance. This effect is mostly due to the well-known inhibitory effect of TGF-β on T cell proliferation, activation, and effector functions. Moreover, TGF-β subverts T cell immunity by favoring regulatory T-cell differentiation, further reinforcing immunosuppression within tumor microenvironments. These findings stimulated the development of many strategies to block TGF-β or its signaling pathways, either as monotherapy or in combination with other therapies, to restore anti-cancer immunity. Paradoxically, recent studies provided evidence that TGF-β can also promote differentiation of certain inflammatory populations of T cells, such as Th17, Th9, and resident-memory T cells (Trm), which have been associated with improved tumor control in several models. Here, we review current advances in our understanding of the many roles of TGF-β in T cell biology in the context of tumor immunity and discuss the possibility to manipulate TGF-β signaling to improve cancer immunotherapy.
Collapse
|
170
|
Ohta M, Chosa N, Kyakumoto S, Yokota S, Okubo N, Nemoto A, Kamo M, Joh S, Satoh K, Ishisaki A. IL‑1β and TNF‑α suppress TGF‑β‑promoted NGF expression in periodontal ligament‑derived fibroblasts through inactivation of TGF‑β‑induced Smad2/3‑ and p38 MAPK‑mediated signals. Int J Mol Med 2018; 42:1484-1494. [PMID: 29901090 DOI: 10.3892/ijmm.2018.3714] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/11/2018] [Indexed: 01/11/2023] Open
Abstract
Mechanosensitive (MS) neurons in the periodontal ligament (PDL) pass information to the trigeminal ganglion when excited by mechanical stimulation of the tooth. During occlusal tooth trauma of PDL tissues, MS neurons are injured, resulting in atrophic neurites and eventual degeneration of MS neurons. Nerve growth factor (NGF), a neurotrophic factor, serves important roles in the regeneration of injured sensory neurons. In the present study, the effect of pro‑inflammatory cytokines, including interleukin 1β (IL‑1β) and tumor necrosis factor α (TNF‑α), on transforming growth factor β1 (TGF‑β1)‑induced NGF expression was evaluated in rat PDL‑derived SCDC2 cells. It was observed that TGF‑β1 promoted NGF expression via Smad2/3 and p38 mitogen‑activated protein kinase (MAPK) activation. IL‑1β and TNF‑α suppressed the TGF‑β1‑induced activation of Smad2/3 and p38 MAPK, resulting in the abrogation of NGF expression. NGF secreted by TGF‑β1‑treated SCDC2 cells promoted neurite extension and the expression of tyrosine hydroxylase, a rate‑limiting enzyme in dopamine synthesis in rat pheochromocytoma PC12 cells. These results suggested that pro‑inflammatory cytokines suppressed the TGF‑β‑mediated expression of NGF in PDL‑derived fibroblasts through the inactivation of TGF‑β‑induced Smad2/3 and p38 MAPK signaling, possibly resulting in the disturbance of the regeneration of injured PDL neurons.
Collapse
Affiliation(s)
- Maiko Ohta
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| | - Naoyuki Chosa
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| | - Seiko Kyakumoto
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| | - Seiji Yokota
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| | - Naoto Okubo
- Laboratory of Pathophysiology and Therapeutics, Division of Pharmasciences, Faculty of Pharmaceutical Sciences, Hokkaido University, Kita‑ku, Sapporo 060‑0812, Japan
| | - Akira Nemoto
- Division of Operative Dentistry and Endodontics, Department of Conservative Dentistry, Iwate Medical University, Morioka, Iwate 020‑8505, Japan
| | - Masaharu Kamo
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| | - Shigeharu Joh
- Division of Oral and Dysphasia Rehabilitation, Department of Prosthodontics, Iwate Medical University, Morioka, Iwate 020‑8505, Japan
| | - Kenichi Satoh
- Division of Dental Anesthesia, Department of Reconstructive Oral and Maxillofacial Surgery, Iwate Medical University, Morioka, Iwate 020‑8505, Japan
| | - Akira Ishisaki
- Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, Shiwa‑gun, Iwate 028‑3694, Japan
| |
Collapse
|
171
|
Kurosaki F, Uchibori R, Sehara Y, Saga Y, Urabe M, Mizukami H, Hagiwara K, Kume A. AAV6-Mediated IL-10 Expression in the Lung Ameliorates Bleomycin-Induced Pulmonary Fibrosis in Mice. Hum Gene Ther 2018; 29:1242-1251. [PMID: 29598007 DOI: 10.1089/hum.2018.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fibroproliferative disorder with limited therapeutic options. An aberrant wound healing process in response to repetitive lung injury has been suggested for its pathogenesis, and a number of cytokines including transforming growth factor β1 play pivotal roles in the induction and progression of fibrosis. Thus, the regulation of these pro-inflammatory conditions may reduce the progression of IPF and ameliorate its symptoms in patients. Interleukin-10 (IL-10), a pleiotropic cytokine, exerts anti-inflammatory and anti-fibrotic effects in numerous biological settings. In the present study, we investigated the preventive effects of IL-10 on bleomycin-induced pulmonary fibrosis in mice with the continuous expression of this cytokine via an adeno-associated virus serotype 6 vector. Mice were administered the adeno-associated virus serotype 6 vector encoding mouse IL-10 by intratracheal injection, and osmotic minipumps containing bleomycin were subcutaneously implanted seven days later. Lung histology and the expression levels of pro-inflammatory cytokines and fibrogenic cytokines were then analyzed. In mice exhibiting persistent IL-10 expression on day 35, the number of infiltrated inflammatory cells and the development of fibrosis in lung tissues were significantly reduced. Increases in transforming growth factor β1 and decreases in IFN-γ were also suppressed in treated animals, with changes in these cytokines playing important roles in the pathogenesis of pulmonary fibrosis. Furthermore, IL-10 significantly improved survival in bleomycin-induced mice. Our results provide insights into the potential benefit of the anti-fibrotic effects of IL-10 as a novel therapeutic approach for IPF.
Collapse
Affiliation(s)
- Fumio Kurosaki
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan .,2 Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Ryosuke Uchibori
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan .,3 Division of Immuno-Gene and Cell Therapy (Takara Bio), Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Yoshihide Sehara
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Yasushi Saga
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan .,4 Department of Obstetrics and Gynecology, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Masashi Urabe
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Hiroaki Mizukami
- 1 Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Koichi Hagiwara
- 2 Division of Pulmonary Medicine, Department of Medicine, Jichi Medical University , Shimotsuke, Tochigi, Japan
| | - Akihiro Kume
- 5 Support Center for Clinical Investigation, Jichi Medical University , Shimotsuke, Tochigi, Japan
| |
Collapse
|
172
|
Li XY, Ban GF, Al-Shameri B, He X, Liang DZ, Chen WX. High-temperature Requirement Protein A1 Regulates Odontoblastic Differentiation of Dental Pulp Cells via the Transforming Growth Factor Beta 1/Smad Signaling Pathway. J Endod 2018; 44:765-772. [PMID: 29580722 DOI: 10.1016/j.joen.2018.02.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 01/22/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Dentinogenesis includes odontoblast differentiation and extracellular matrix maturation as well as dentin mineralization. It is regulated by numerous molecules. High-temperature requirement protein A1 (HtrA1) plays crucial roles in bone mineralization and development and is closely associated with the transforming growth factor beta (TGF-β) signal in osteogenesis differentiation. Simultaneously, the TGF-β1/small mother against decapentaplegic (Smad) signaling pathway is an important signaling pathway in various physiological processes and as a downstream regulation factor of HtrA1. However, the role of HtrA1 and its relationship with the TGF-β1/Smad signaling pathway in dentin mineralization is unknown. METHODS We detected the role of HtrA1 and its relationship with the TGF-β1/Smad signaling pathway in odontoblastic differentiation of human dental pulp cells (hDPCs) in this study. First, hDPCs were cultured in mineralized medium, and odontoblastic differentiation was confirmed by investigating mineralized nodule formation, alkaline phosphatase (ALP) activity, and the expression of mineral-associated genes, including ALP, collagen I, and dentin sialophosphoprotein. Then, the expression of HtrA1 and TGF-β1/Smad in hDPCs was investigated in hDPCs during mineralized induction. After HtrA1 knockdown by lentivirus, the mineralized nodule formation, ALP activity, and expression of mineral-associated genes and TGF-β1/Smad genes were investigated to confirm the effect of HtrA1 on odontoblastic differentiation and its relationship with the TGF-β1/Smad signaling pathway. RESULTS The expression of HtrA1 and TGF-β1 was increased during odontoblastic differentiation of hDPCs along with the messenger RNA expression of downstream factors of the TGF-β1/Smad signaling pathway. In addition, lentivirus-mediated HtrA1 knockdown inhibited the process of mineralization and the expression of HtrA1 and TGF-β1/Smad genes. CONCLUSIONS These findings suggest that HtrA1 might positively regulate odontoblastic differentiation of hDPCs through activation of the TGF-β1/Smad signaling pathway.
Collapse
Affiliation(s)
- Xian-Yu Li
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China
| | - Gui-Fei Ban
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China
| | - Basheer Al-Shameri
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China
| | - Xuan He
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China
| | - Deng-Zhong Liang
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China
| | - Wen-Xia Chen
- Department of Operative Dentistry and Endodontology, College and Hospital of Stomatology, Guangxi Medical University, Nanning, Guagnxi, China.
| |
Collapse
|
173
|
Karlsson T, Sundar R, Widmark A, Landström M, Persson E. Osteoblast-derived factors promote metastatic potential in human prostate cancer cells, in part via non-canonical transforming growth factor β (TGFβ) signaling. Prostate 2018; 78:446-456. [PMID: 29383751 DOI: 10.1002/pros.23489] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 01/10/2018] [Indexed: 01/01/2023]
Abstract
BACKGROUND Transforming growth factor β (TGFβ) functions as a double-edged sword in prostate cancer tumorigenesis. In initial stages of the disease, TGFβ acts as a growth inhibitor upon tumor cells, whereas it in later stages of disease rather promotes invasion and metastatic potential. One well-known cellular source of TGFβ in the bone metastatic site is the bone-forming osteoblasts. Here we have studied the effects by osteoblast-derived factors on metastatic potential in several human prostate cancer cell lines. METHODS Effects on metastatic potential in prostate cancer cells by osteoblast-derived factors were studied in vitro using several methods, including Transwell migration and evaluation of formation of pro-migratory protrusions. Confocal microscopy was used to evaluate possible changes in differentiation state in tumor cells by analysis of markers for epithelial-to-mesenchymal transition (EMT). The Matrigel-on-top 3D culture method was used for further assessment of metastatic characteristics in tumor cells by analysis of formation of filopodium-like protrusions (FLPs). RESULTS Osteoblast-derived factors increased migration of PC-3U cells, an effect less prominent in cells overexpressing a mutated type I TGFβ receptor (TβRI) preventing non-canonical TRAF6-dependent TGFβ signaling. Osteoblast-derived factors also increased the formation of long protrusions and loss of cell-cell contacts in PC-3U cells, suggesting induction of a more aggressive phenotype. In addition, treatment with TGFβ or osteoblast-derived factors of PC-3U cells in Matrigel-on-top 3D cultures promoted formation of FLPs, previously shown to be essential for metastatic establishment. CONCLUSIONS These findings suggests that factors secreted from osteoblasts, including TGFβ, can induce several cellular traits involved in metastatic potential of PC-3U cells, further strengthening the role for bone cells to promote metastatic tumor cell behavior.
Collapse
Affiliation(s)
- Terese Karlsson
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| | - Reshma Sundar
- Department of Medical Biosciences, Section for Pathology, Umeå University, Umeå, Sweden
| | - Anders Widmark
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| | - Maréne Landström
- Department of Medical Biosciences, Section for Pathology, Umeå University, Umeå, Sweden
| | - Emma Persson
- Department of Radiation Sciences, Section for Oncology, Umeå University, Umeå, Sweden
| |
Collapse
|
174
|
Mallikarjuna P, Sitaram RT, Landström M, Ljungberg B. VHL status regulates transforming growth factor-β signaling pathways in renal cell carcinoma. Oncotarget 2018; 9:16297-16310. [PMID: 29662646 PMCID: PMC5893241 DOI: 10.18632/oncotarget.24631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 02/27/2018] [Indexed: 12/26/2022] Open
Abstract
To evaluate the role of pVHL in the regulation of TGF-β signaling pathways in clear cell renal cell carcinoma (ccRCC) as well as in non-ccRCC; the expression of pVHL, and the TGF-β pathway components and their association with clinicopathological parameters and patient’s survival were explored. Tissue samples from 143 ccRCC and 58 non-ccRCC patients were examined by immunoblot. ccRCC cell lines were utilized for mechanistic in-vitro studies. Expression levels of pVHL were significantly lower in ccRCC compared with non-ccRCC. Non-ccRCC and ccRCC pVHL-High expressed similar levels of pVHL. Expression of the TGF-β type I receptor (ALK5) and intra-cellular domain were significantly higher in ccRCC compared with non-ccRCC. In non-ccRCC, expressions of ALK5-FL, ALK5-ICD, pSMAD2/3, and PAI-1 had no association with clinicopathological parameters and survival. In ccRCC pVHL-Low, ALK5-FL, ALK5-ICD, pSMAD2/3, and PAI-1 were significantly related with tumor stage, size, and survival. In ccRCC pVHL-High, the expression of PAI-1 was associated with stage and survival. In-vitro studies revealed that pVHL interacted with ALK5 to downregulate its expression through K48-linked poly-ubiquitination and proteasomal degradation, thus negatively controlling TGF-β induced cancer cell invasiveness. The pVHL status controls the ALK5 and can thereby regulate the TGF-β pathway, aggressiveness of tumors, and survival of the ccRCC and non-ccRCC patients.
Collapse
Affiliation(s)
- Pramod Mallikarjuna
- Department of Medical Biosciences, Pathology, Umeå University, Umeå SE-90187, Sweden
| | - Raviprakash T Sitaram
- Department of Medical Biosciences, Pathology, Umeå University, Umeå SE-90187, Sweden.,Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå SE-90187, Sweden
| | - Maréne Landström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå SE-90187, Sweden
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå SE-90187, Sweden
| |
Collapse
|
175
|
Abstract
The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR)-dependent pathway is one of the most integral pathways linked to cell metabolism, proliferation, differentiation, and survival. This pathway is dysregulated in a variety of diseases, including neoplasia, immune-mediated diseases, and fibroproliferative diseases such as pulmonary fibrosis. The mTOR kinase is frequently referred to as the master regulator of this pathway. Alterations in mTOR signaling are closely associated with dysregulation of autophagy, inflammation, and cell growth and survival, leading to the development of lung fibrosis. Inhibitors of mTOR have been widely studied in cancer therapy, as they may sensitize cancer cells to radiation therapy. Studies also suggest that mTOR inhibitors are promising modulators of fibroproliferative diseases such as idiopathic pulmonary fibrosis (IPF) and radiation-induced pulmonary fibrosis (RIPF). Therefore, mTOR represents an attractive and unique therapeutic target in pulmonary fibrosis. In this review, we discuss the pathological role of mTOR kinase in pulmonary fibrosis and examine how mTOR inhibitors may mitigate fibrotic progression.
Collapse
|
176
|
Mirzaei H, Faghihloo E. Viruses as key modulators of the TGF-β pathway; a double-edged sword involved in cancer. Rev Med Virol 2018; 28:e1967. [PMID: 29345394 PMCID: PMC7169117 DOI: 10.1002/rmv.1967] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 12/11/2017] [Accepted: 12/12/2017] [Indexed: 12/12/2022]
Abstract
Transforming growth factor-β (TGF-β) signaling pathway is a key network in cell signaling that controls vital processes such as proliferation, differentiation, apoptosis, epithelial-mesenchymal transition, and migration, thus acting as a double-edged sword in normal development and diseases, in particular organ fibrosis, vascular disorders, and cancer. Early in tumorigenesis, the pathway exerts anti-tumor effects through suppressing cell cycle and inducing apoptosis, while during late stages, it functions as a tumor promoter by enhancing tumor invasiveness and metastasis. This signaling pathway can be perturbed by environmental and genetic factors such as microbial interference and mutation, respectively. In this way, the present review describes the modulation of the TGF-β pathway by oncogenic human viral pathogens and other viruses. The main mechanisms by which viruses interferes with TGF-β signaling seems to be through (1) the alteration of either TGF-β protein expression or activation, (2) the modulation of the TGF-β receptors or SMADs factors (by interfering with their levels and functions), (3) the alteration of none-SMAD pathways, and (4) indirect interaction with the pathway by the modulation of transcriptional co-activator/repressor and regulators of the pathway. Given the axial role of this pathway in tumorigenesis, it can be regarded as an attractive target for cancer therapy. Hence, further investigations on this subject may represent molecular targets among either TGF-β signaling molecules or viral factors for the treatment and management of viral infection consequences such as cancer.
Collapse
Affiliation(s)
- Habibollah Mirzaei
- Department of Virology, School of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
- Hepatitis Research CenterLorestan University of Medical SciencesKhorramabadIRIran
| | - Ebrahim Faghihloo
- Department of Microbiology, School of MedicineShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
177
|
Abstract
Fanconi anemia is an inherited disease characterized by genomic instability, hypersensitivity to DNA cross-linking agents, bone marrow failure, short stature, skeletal abnormalities, and a high relative risk of myeloid leukemia and epithelial malignancies. The 21 Fanconi anemia genes encode proteins involved in multiple nuclear biochemical pathways that effect DNA interstrand crosslink repair. In the past, bone marrow failure was attributed solely to the failure of stem cells to repair DNA. Recently, non-canonical functions of many of the Fanconi anemia proteins have been described, including modulating responses to oxidative stress, viral infection, and inflammation as well as facilitating mitophagic responses and enhancing signals that promote stem cell function and survival. Some of these functions take place in non-nuclear sites and do not depend on the DNA damage response functions of the proteins. Dysfunctions of the canonical and non-canonical pathways that drive stem cell exhaustion and neoplastic clonal selection are reviewed, and the potential therapeutic importance of fully investigating the scope and interdependences of the canonical and non-canonical pathways is emphasized.
Collapse
Affiliation(s)
- Grover Bagby
- Departments of Medicine and Molecular and Medical Genetics, Division of Hematology and Medical Oncology, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
178
|
Ethanol sensitizes hepatocytes for TGF-β-triggered apoptosis. Cell Death Dis 2018; 9:51. [PMID: 29352207 PMCID: PMC5833779 DOI: 10.1038/s41419-017-0071-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 09/19/2017] [Accepted: 10/09/2017] [Indexed: 12/14/2022]
Abstract
Alcohol abuse is a global health problem causing a substantial fraction of chronic liver diseases. Abundant TGF-β—a potent pro-fibrogenic cytokine—leads to disease progression. Our aim was to elucidate the crosstalk of TGF-β and alcohol on hepatocytes. Primary murine hepatocytes were challenged with ethanol and TGF-β and cell fate was determined. Fluidigm RNA analyses revealed transcriptional effects that regulate survival and apoptosis. Mechanistic insights were derived from enzyme/pathway inhibition experiments and modulation of oxidative stress levels. To substantiate findings, animal model specimens and human liver tissue cultures were investigated. Results: On its own, ethanol had no effect on hepatocyte apoptosis, whereas TGF-β increased cell death. Combined treatment led to massive hepatocyte apoptosis, which could also be recapitulated in human HCC liver tissue treated ex vivo. Alcohol boosted the TGF-β pro-apoptotic gene signature. The underlying mechanism of pathway crosstalk involves SMAD and non-SMAD/AKT signaling. Blunting CYP2E1 and ADH activities did not prevent this effect, implying that it was not a consequence of alcohol metabolism. In line with this, the ethanol metabolite acetaldehyde did not mimic the effect and glutathione supplementation did not prevent the super-induction of cell death. In contrast, blocking GSK-3β activity, a downstream mediator of AKT signaling, rescued the strong apoptotic response triggered by ethanol and TGF-β. This study provides novel information on the crosstalk between ethanol and TGF-β. We give evidence that ethanol directly leads to a boost of TGF-β’s pro-apoptotic function in hepatocytes, which may have implications for patients with chronic alcoholic liver disease.
Collapse
|
179
|
Ashry M, Rajput SK, Folger JK, Knott JG, Hemeida NA, Kandil OM, Ragab RS, Smith GW. Functional role of AKT signaling in bovine early embryonic development: potential link to embryotrophic actions of follistatin. Reprod Biol Endocrinol 2018; 16:1. [PMID: 29310676 PMCID: PMC5759257 DOI: 10.1186/s12958-017-0318-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 12/25/2017] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND TGF-β signaling pathways regulate several crucial processes in female reproduction. AKT is a non-SMAD signaling pathway regulated by TGF-β ligands essential for oocyte maturation and early embryonic development in the mouse, but its regulatory role in bovine early embryonic development is not well established. Previously, we demonstrated a stimulatory role for follistatin (a binding protein for specific members of TGF-β superfamily) in early bovine embryonic development. The objectives of the present studies were to determine the functional role of AKT signaling in bovine early embryonic development and embryotrophic actions of follistatin. METHODS We used AKT inhibitors III and IV as pharmacological inhibitors of AKT signaling pathway during the first 72 h of in vitro embryo culture. Effects of AKT inhibition on early embryonic development and AKT phosphorylation were investigated in the presence or absence of exogenous follistatin. RESULTS Pharmacological inhibition of AKT signaling resulted in a significant reduction in early embryo cleavage, and development to the 8- to 16-cell and blastocyst stages (d7). Treatment with exogenous follistatin increased AKT phosphorylation and rescued the inhibitory effect of AKT inhibitors III and IV on AKT phosphorylation and early embryonic development. CONCLUSIONS Collectively, results suggest a potential requirement of AKT for bovine early embryonic development, and suggest a potential role for follistatin in regulation of AKT signaling in early bovine embryos.
Collapse
Affiliation(s)
- Mohamed Ashry
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Sandeep K. Rajput
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Joseph K. Folger
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Jason G. Knott
- Developmental Epigenetics Laboratory, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| | - Nabil A. Hemeida
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Omaima M. Kandil
- Department of Animal Reproduction and Artificial Insemination, Veterinary Research Division, National Research Center, Giza, Egypt
| | - Refaat S. Ragab
- Department of Theriogenology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - George W. Smith
- Laboratory of Mammalian Reproductive Biology and Genomics, Michigan State University, East Lansing, MI 48824 USA
- Department of Animal Science, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
180
|
Yan X, Xiong X, Chen YG. Feedback regulation of TGF-β signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:37-50. [PMID: 29228156 DOI: 10.1093/abbs/gmx129] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a multi-functional polypeptide that plays a critical role in regulating a broad range of cellular functions and physiological processes. Signaling is initiated when TGF-β ligands bind to two types of cell membrane receptors with intrinsic Ser/Thr kinase activity and transmitted by the intracellular Smad proteins, which act as transcription factors to regulate gene expression in the nucleus. Although it is relatively simple and straight-forward, this TGF-β/Smad pathway is regulated by various feedback loops at different levels, including the ligand, the receptor, Smads and transcription, and is thus fine-tuned in terms of signaling robustness, duration, specificity, and plasticity. The precise control gives rise to versatile and context-dependent pathophysiological functions. In this review, we firstly give an overview of TGF-β signaling, and then discuss how each step of TGF-β signaling is finely controlled by distinct modes of feedback mechanisms, involving both protein regulators and miRNAs.
Collapse
Affiliation(s)
- Xiaohua Yan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Xiangyang Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang 330006, China
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
181
|
Molecular Mechanisms Responsible for Anti-inflammatory and Immunosuppressive Effects of Mesenchymal Stem Cell-Derived Factors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1084:187-206. [PMID: 31175638 DOI: 10.1007/5584_2018_306] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mesenchymal stem cells (MSCs) are self-renewable cells capable for multilineage differentiation and immunomodulation. MSCs are able to differentiate into all cell types of mesodermal origin and, due to their plasticity, may generate cells of neuroectodermal or endodermal origin in vitro. In addition to the enormous differentiation potential, MSCs efficiently modulate innate and adaptive immune response and, accordingly, were used in large number of experimental and clinical trials as new therapeutic agents in regenerative medicine. Although MSC-based therapy was efficient in the treatment of many inflammatory and degenerative diseases, unwanted differentiation of engrafted MSCs represents important safety concern. MSC-based beneficial effects are mostly relied on the effects of MSC-derived immunomodulatory, pro-angiogenic, and trophic factors which attenuate detrimental immune response and inflammation, reduce ischemic injuries, and promote tissue repair and regeneration. Accordingly, MSC-conditioned medium (MSC-CM), which contains MSC-derived factors, has the potential to serve as a cell-free, safe therapeutic agent for the treatment of inflammatory diseases. Herein, we summarized current knowledge regarding identification, isolation, ontogeny, and functional characteristics of MSCs and described molecular mechanisms responsible for MSC-CM-mediated anti-inflammatory and immunosuppressive effects in the therapy of inflammatory lung, liver, and kidney diseases and ischemic brain injury.
Collapse
|
182
|
Xie F, Ling L, van Dam H, Zhou F, Zhang L. TGF-β signaling in cancer metastasis. Acta Biochim Biophys Sin (Shanghai) 2018; 50:121-132. [PMID: 29190313 DOI: 10.1093/abbs/gmx123] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Indexed: 02/06/2023] Open
Abstract
The transforming growth factor (TGF)-β signaling events are well known to control diverse processes and numerous responses, such as cell proliferation, differentiation, apoptosis, and migration. TGF-β signaling plays context-dependent roles in cancer: in pre-malignant cells TGF-β primarily functions as a tumor suppressor, while in the later stages of cancer TGF-β signaling promotes invasion and metastasis. Recent studies have also suggested that the cross-talk between TGF-β signaling and other signaling pathways, such as Hippo, Wnt, EGFR/RAS, and PI3K/AKT pathways, may substantially contribute to our current understanding of TGF-β signaling and cancer. As a result of the wide-ranging effects of TGF-β, blockade of TGF-β and its downstream signaling components provides multiple therapeutic opportunities. Therefore, the outlook for anti-TGF-β signaling therapy for numerous diseases appears bright and will provide valuable information and thinking on the drug molecular design. In this review, we focus on recent insights into the regulation of TGF-β signaling in cancer metastasis which may contribute to the development of novel cancer-targeting therapies.
Collapse
Affiliation(s)
- Feng Xie
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Li Ling
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Hans van Dam
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, The Netherlands
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Long Zhang
- Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| |
Collapse
|
183
|
Sitaram RT, Mallikarjuna P, Landström M, Ljungberg B. Transforming growth factor-β promotes aggressiveness and invasion of clear cell renal cell carcinoma. Oncotarget 2017; 7:35917-35931. [PMID: 27166254 PMCID: PMC5094972 DOI: 10.18632/oncotarget.9177] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 04/19/2016] [Indexed: 12/22/2022] Open
Abstract
The molecular mechanisms whereby transforming growth factor-β (TGF-β) promotes clear cell renal cell carcinoma (ccRCC) progression is elusive. The cell membrane bound TGF-β type I receptor (ALK5), was recently found to undergo proteolytic cleavage in aggressive prostate cancer cells, resulting in liberation and subsequent nuclear translocation of its intracellular domain (ICD), suggesting that ALK5-ICD might be a useful cancer biomarker. Herein, the possible correlation between ALK5 full length (ALK5-FL) and ALK5-ICD protein, phosphorylated Smad2/3 (pSmad2/3), and expression of TGF-β target gene PAI-1, was investigated in a clinical ccRCC material, in relation to tumor grade, stage, size and cancer specific survival. Expression of ALK5-FL, ALK5-ICD, pSmad2/3 and PAI-1 protein levels were significantly higher in higher stage and associated with adverse survival. ALK5-ICD, pSmad2/3 and PAI-1 correlated with higher grade, and ALK5-FL, pSmad2/3 and PAI-1 protein levels were significantly correlated with larger tumor size. Moreover, the functional role of the TGF-β - ALK5-ICD pathway were investigated in two ccRCC cell lines by treatment with ADAM/MMP2 inhibitor TAPI-2, which prevented TGF-β-induced ALK5-ICD generation, nuclear translocation, as well as cell invasion. The present study demonstrated that canonical TGF-β Smad2/3 pathway and generation of ALK5-ICD correlates with poor survival and invasion of ccRCC in vitro.
Collapse
Affiliation(s)
- Raviprakash T Sitaram
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| | | | - Maréne Landström
- Department of Medical Biosciences, Pathology, Umeå University, Umeå, Sweden
| | - Börje Ljungberg
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umeå University, Umeå, Sweden
| |
Collapse
|
184
|
Witte D, Otterbein H, Förster M, Giehl K, Zeiser R, Lehnert H, Ungefroren H. Negative regulation of TGF-β1-induced MKK6-p38 and MEK-ERK signalling and epithelial-mesenchymal transition by Rac1b. Sci Rep 2017; 7:17313. [PMID: 29229918 PMCID: PMC5725500 DOI: 10.1038/s41598-017-15170-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 10/23/2017] [Indexed: 01/26/2023] Open
Abstract
Prompted by earlier findings that the Rac1-related isoform Rac1b inhibits transforming growth factor (TGF)-β1-induced canonical Smad signalling, we studied here whether Rac1b also impacts TGF-β1-dependent non-Smad signalling such as the MKK6-p38 and MEK-ERK mitogen-activated protein kinase (MAPK) pathways and epithelial-mesenchymal transition (EMT). Transient depletion of Rac1b protein in pancreatic cancer cells by RNA interference increased the extent and duration of TGF-β1-induced phosphorylation of p38 MAPK in a Smad4-independent manner. Rac1b depletion also strongly increased basal ERK activation - independent of the kinase function of the TGF-β type I receptor ALK5 - and sensitised cells towards further upregulation of phospho-ERK levels by TGF-β1, while ectopic overexpression of Rac1b had the reverse effect. Rac1b depletion increased an EMT phenotype as evidenced by cell morphology, gene expression of EMT markers, cell migration and growth inhibition. Inhibition of MKK6-p38 or MEK-ERK signalling partially relieved the Rac1b depletion-dependent increase in TGF-β1-induced gene expression and cell migration. Rac1b depletion also enhanced TGF-β1 autoinduction of crucial TGF-β pathway components and decreased that of TGF-β pathway inhibitors. Our results show that Rac1b antagonises TGF-β1-dependent EMT by inhibiting MKK6-p38 and MEK-ERK signalling and by controlling gene expression in a way that favors attenuation of TGF-β signalling.
Collapse
Affiliation(s)
- David Witte
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck, 23538, Lübeck, Germany
| | - Hannah Otterbein
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck, 23538, Lübeck, Germany
| | - Maria Förster
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck, 23538, Lübeck, Germany
| | - Klaudia Giehl
- Signal Transduction of Cellular Motility, Internal Medicine V, Justus-Liebig-University Giessen, 35392, Giessen, Germany
| | - Robert Zeiser
- Department of Hematology and Oncology, Freiburg University Medical Center, Albert-Ludwigs-University, 79106, Freiburg i.Br., Germany
| | - Hendrik Lehnert
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck, 23538, Lübeck, Germany
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, and University of Lübeck, 23538, Lübeck, Germany. .,Department of General and Thoracic Surgery, UKSH, Campus Kiel, 24105, Kiel, Germany.
| |
Collapse
|
185
|
Seoane J, Gomis RR. TGF-β Family Signaling in Tumor Suppression and Cancer Progression. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022277. [PMID: 28246180 DOI: 10.1101/cshperspect.a022277] [Citation(s) in RCA: 342] [Impact Index Per Article: 48.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transforming growth factor-β (TGF-β) induces a pleiotropic pathway that is modulated by the cellular context and its integration with other signaling pathways. In cancer, the pleiotropic reaction to TGF-β leads to a diverse and varied set of gene responses that range from cytostatic and apoptotic tumor-suppressive ones in early stage tumors, to proliferative, invasive, angiogenic, and oncogenic ones in advanced cancer. Here, we review the knowledge accumulated about the molecular mechanisms involved in the dual response to TGF-β in cancer, and how tumor cells evolve to evade the tumor-suppressive responses of this signaling pathway and then hijack the signal, converting it into an oncogenic factor. Only through the detailed study of this complexity can the suitability of the TGF-β pathway as a therapeutic target against cancer be evaluated.
Collapse
Affiliation(s)
- Joan Seoane
- Translational Research Program, Vall d'Hebron Institute of Oncology, 08035 Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
| | - Roger R Gomis
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain.,Oncology Program, Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, 08028 Barcelona, Spain
| |
Collapse
|
186
|
Song J, Landström M. TGFβ activates PI3K-AKT signaling via TRAF6. Oncotarget 2017; 8:99205-99206. [PMID: 29245887 PMCID: PMC5725078 DOI: 10.18632/oncotarget.22275] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/30/2017] [Indexed: 01/16/2023] Open
Affiliation(s)
- Jie Song
- Maréne Landström: Medical Biosciences Department, Umeå University, Umeå, Sweden
| | - Maréne Landström
- Maréne Landström: Medical Biosciences Department, Umeå University, Umeå, Sweden
| |
Collapse
|
187
|
Targeting Interleukin-1β Protects from Aortic Aneurysms Induced by Disrupted Transforming Growth Factor β Signaling. Immunity 2017; 47:959-973.e9. [DOI: 10.1016/j.immuni.2017.10.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 07/18/2017] [Accepted: 10/26/2017] [Indexed: 01/11/2023]
|
188
|
Hou X, Zhang J, Wang Y, Xiong W, Mi J. TGFBR-IDH1-Cav1 axis promotes TGF-β signalling in cancer-associated fibroblast. Oncotarget 2017; 8:83962-83974. [PMID: 29137396 PMCID: PMC5663568 DOI: 10.18632/oncotarget.20861] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 08/23/2017] [Indexed: 11/25/2022] Open
Abstract
TGF-β signalling plays an important role in fibroblasts activation and tumour progression. Here, we report that the TGFBR-IDH1-Cav1 axis promotes TGF- β signalling in fibroblasts. Our data demonstrated that IDH1 was downregulated by TGF-β signalling in fibroblasts, and downregulation of IDH1 increased cellular concentration of α-ketoglutarate (α-KG) by accelerating glutamine metabolization. Interestingly, α-KG suppressed Cav1 expression through reducing the trimethylation of histone H3K4. Furthermore, Cav1 downregulation inhibited TGFBR protein degradation. In turn, the activated TGFBR promoted TGF-β signalling. These findings demonstrated that metabolic enzyme IDH1 regulates TGF-β signalling by feedback mechanism through α-KG and TGFBR-IDH1-Cav1 axis is important for TGF-β signalling.
Collapse
Affiliation(s)
- Xiaodan Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Center of Systems Medicine, Chinese Academy of Medical Sciences, Suzhou Institute of Systems Medicine, Suzhou, China
| | - Jieying Zhang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongbin Wang
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wujun Xiong
- Shanghai East Hospital, Affiliated to Tongji University, Shanghai, China
| | - Jun Mi
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
189
|
Santibanez JF, Bjelica S. Transforming Growth Factor-Beta1 and Myeloid-Derived Suppressor Cells Interplay in Cancer. ACTA ACUST UNITED AC 2017. [DOI: 10.2174/1876401001706010001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background:
Transforming growth factor-beta1 (TGF-β1) is a pleiotropic cytokine with a double role in cancer through its capacity to inhibit early stages of tumors while enhancing tumor progression at late stages of tumor progression. Moreover, TGF-β1 is a potent immunosuppressive cytokine within the tumor microenvironment that allows cancer cells to escape from immune surveillance, which largely contributes to the tumor progression.
Method:
It has been established that the cancer progression is commonly associated with increased number of Myeloid-derived suppressor cells (MDSC) that are a hallmark of cancer and a key mechanism of immune evasion.
Result:
MDSC represent a population of heterogeneous myeloid cells comprised of macrophages, granulocytes and dendritic cells at immature stages of development. MDSC promote tumor progression by regulating immune responses as well as tumor angiogenesis and cancer metastasis.
Conclusion:
In this review, we present an overview of the main key functions of both TGF-β1 and MDSC in cancer and in the immune system. Furthermore, the mutual contribution between TGF-β1 and MDSC in the regulation of immune system and cancer development will be analyzed.
Collapse
|
190
|
OM-101 Decreases the Fibrotic Response Associated with Proliferative Vitreoretinopathy. J Ophthalmol 2017; 2017:1606854. [PMID: 29109865 PMCID: PMC5646338 DOI: 10.1155/2017/1606854] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 09/06/2017] [Indexed: 11/17/2022] Open
Abstract
Purpose This study aimed to investigate the effect of OM-101 on the fibrotic response occurring in proliferative vitreoretinopathy (PVR) in an animal model. Methods Antifibrotic effect of OM-101 was investigated in vivo. As control, eight weeks old c57black mice underwent intravitreal injection with Hepes (group A) or dispase (0.3 units), to induce retinal detachment (RD) and PVR. The dispase-injected mice were randomly divided into two groups B and C (N = 25 mice); in group C, the eyes were treated with intravitreal injection of OM-101 (3 μl), and group B with PBS, as a control. After additional five days, mice were injected with the same initial treatment. Three days later, mice were euthanized, and the eyes were enucleated and processed for histological analysis. Results Intravitreal injection of dispase caused RD in 64% of the mice in group B, and 93% of those mice had PVR. Only 32% of mice treated with OM-101 and dispase (group C) developed RD, and only 25% of those developed PVR. Conclusions OM-101 was found effective in reducing the incidence of RD and PVR maintaining the normal architecture of the retina. This study suggests that OM-101 is a potentially effective and safe drug for the treatment of PVR patients.
Collapse
|
191
|
Gentile D, Lazzerini PE, Gamberucci A, Natale M, Selvi E, Vanni F, Alì A, Taddeucci P, Del-Ry S, Cabiati M, Della-Latta V, Abraham DJ, Morales MA, Fulceri R, Laghi-Pasini F, Capecchi PL. Searching Novel Therapeutic Targets for Scleroderma: P2X7-Receptor Is Up-regulated and Promotes a Fibrogenic Phenotype in Systemic Sclerosis Fibroblasts. Front Pharmacol 2017; 8:638. [PMID: 28955239 PMCID: PMC5602350 DOI: 10.3389/fphar.2017.00638] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/29/2017] [Indexed: 11/13/2022] Open
Abstract
Objectives: Systemic sclerosis (SSc) is a connective tissue disorder presenting fibrosis of the skin and internal organs, for which no effective treatments are currently available. Increasing evidence indicates that the P2X7 receptor (P2X7R), a nucleotide-gated ionotropic channel primarily involved in the inflammatory response, may also have a key role in the development of tissue fibrosis in different body districts. This study was aimed at investigating P2X7R expression and function in promoting a fibrogenic phenotype in dermal fibroblasts from SSc patients, also analyzing putative underlying mechanistic pathways. Methods: Fibroblasts were isolated by skin biopsy from 9 SSc patients and 8 healthy controls. P2X7R expression, and function (cytosolic free Ca2+ fluxes, α-smooth muscle actin [α-SMA] expression, cell migration, and collagen release) were studied. Moreover, the role of cytokine (interleukin-1β, interleukin-6) and connective tissue growth factor (CTGF) production, and extracellular signal-regulated kinases (ERK) activation in mediating P2X7R-dependent pro-fibrotic effects in SSc fibroblasts was evaluated. Results: P2X7R expression and Ca2+ permeability induced by the selective P2X7R agonist 2'-3'-O-(4-benzoylbenzoyl)ATP (BzATP) were markedly higher in SSc than control fibroblasts. Moreover, increased αSMA expression, cell migration, CTGF, and collagen release were observed in lipopolysaccharides-primed SSc fibroblasts after BzATP stimulation. While P2X7-induced cytokine changes did not affect collagen production, it was completely abrogated by inhibition of the ERK pathway. Conclusion: In SSc fibroblasts, P2X7R is overexpressed and its stimulation induces Ca2+-signaling activation and a fibrogenic phenotype characterized by increased migration and collagen production. These data point to the P2X7R as a potential, novel therapeutic target for controlling exaggerated collagen deposition and tissue fibrosis in patients with SSc.
Collapse
Affiliation(s)
- Daniela Gentile
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pietro E Lazzerini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Alessandra Gamberucci
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Mariarita Natale
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Enrico Selvi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Francesca Vanni
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Alessandra Alì
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Paolo Taddeucci
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | | | | | | | - David J Abraham
- Division of Medicine, Department of Inflammation, Centre for Rheumatology and Connective Tissue Diseases, University College London, London, United Kingdom
| | | | - Rosella Fulceri
- Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Franco Laghi-Pasini
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| | - Pier L Capecchi
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
192
|
Wang T, Liu Y, Zou JF, Cheng ZS. Interleukin-17 induces human alveolar epithelial to mesenchymal cell transition via the TGF-β1 mediated Smad2/3 and ERK1/2 activation. PLoS One 2017; 12:e0183972. [PMID: 28873461 PMCID: PMC5584923 DOI: 10.1371/journal.pone.0183972] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 08/15/2017] [Indexed: 12/31/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and usually progressive lung disease and the epithelial-mesenchymal transition (EMT) may play an important role in the pathogenesis of pulmonary fibrosis. IL-17 is a proinflammatory cytokine which promotes EMT profiles in lung inflammatory diseases. In this study, we investigated the effect of IL-17 on EMT in alveolar epithelial cell line A549 and the role of TGFβ1-Smad and ERK signaling pathways in the process. Morphological observation on the cells was performed under inverted microscope. The mRNA and protein expressions of E-cad and α-SMA were detected by quantitative RT-PCR and western blotting. The mRNA and protein expressions of TGF-β1 were analyzed via quantitative RT-PCR and ELISA. Expressions of Smad2/3, p-Smad2/3, ERK1/2, p-ERK1/2 and p-JNK were examined by western blotting. The results indicated that IL-17 can induce A549 cells to undergo morphological changes and phenotypic markers changes, such as down-regulated E-cad expression and up-regulated α-SMA expression. Additionally, IL-17 enhanced TGF-β1 expression and stimulated Smad2/3 and ERK1/2 phosphorylation in A549 cells. However, there were no significant differences in the expression of phosphorylated JNK in A549 cells with or without IL-17 treatment. SB431542 or U0126 treated cells showed inhibited morphological changes and phenotypic markers expression, such as up-regulated E-cad expression and down-regulated α-SMA expression. In summary, our results suggest that IL-17 can induce A549 alveolar epithelial cells to undergo EMT via the TGF-β1 mediated Smad2/3 and ERK1/2 activation.
Collapse
Affiliation(s)
- Ting Wang
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Liu
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jing-Feng Zou
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shun Cheng
- Department of Respiratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
- * E-mail:
| |
Collapse
|
193
|
Kanhaiya K, Czeizler E, Gratie C, Petre I. Controlling Directed Protein Interaction Networks in Cancer. Sci Rep 2017; 7:10327. [PMID: 28871116 PMCID: PMC5583175 DOI: 10.1038/s41598-017-10491-y] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023] Open
Abstract
Control theory is a well-established approach in network science, with applications in bio-medicine and cancer research. We build on recent results for structural controllability of directed networks, which identifies a set of driver nodes able to control an a-priori defined part of the network. We develop a novel and efficient approach for the (targeted) structural controllability of cancer networks and demonstrate it for the analysis of breast, pancreatic, and ovarian cancer. We build in each case a protein-protein interaction network and focus on the survivability-essential proteins specific to each cancer type. We show that these essential proteins are efficiently controllable from a relatively small computable set of driver nodes. Moreover, we adjust the method to find the driver nodes among FDA-approved drug-target nodes. We find that, while many of the drugs acting on the driver nodes are part of known cancer therapies, some of them are not used for the cancer types analyzed here; some drug-target driver nodes identified by our algorithms are not known to be used in any cancer therapy. Overall we show that a better understanding of the control dynamics of cancer through computational modelling can pave the way for new efficient therapeutic approaches and personalized medicine.
Collapse
Affiliation(s)
- Krishna Kanhaiya
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
| | - Eugen Czeizler
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
- National Institute for Research and Development for Biological Sciences, Bucharest, Romania
| | - Cristian Gratie
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland
| | - Ion Petre
- Computational Biomodeling Laboratory, Turku Centre for Computer Science, and Department of Computer Science, Åbo Akademi University, Turku, 20500, Finland.
| |
Collapse
|
194
|
Mechanism of chimeric vaccine stimulation of indoleamine 2,3-dioxygenase biosynthesis in human dendritic cells is independent of TGF-β signaling. Cell Immunol 2017; 319:43-52. [PMID: 28864263 DOI: 10.1016/j.cellimm.2017.08.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 07/27/2017] [Accepted: 08/18/2017] [Indexed: 01/17/2023]
Abstract
Cholera toxin B subunit fusion to autoantigens such as proinsulin (CTB-INS) down regulate dendritic cell (DC) activation and stimulate synthesis of DC immunosuppressive cytokines. Recent studies of CTB-INS induction of immune tolerance in human DCs indicate that increased biosynthesis of indoleamine 2,3-dioxygenase (IDO1) may play an important role in CTB-INS vaccine suppression of DC activation. Studies in murine models suggest a role for transforming growth factor beta (TGF-β) in the stimulation of IDO1 biosynthesis, for the induction of tolerance in DCs. Here, we investigated the contribution of TGF-β superfamily proteins to CTB-INS induction of IDO1 biosynthesis in human monocyte-derived DCs (moDCs). We show that CTB-INS upregulates the level of TGF-β1, activin-A and the TGF-β activator, integrin αvβ8 in human DCs. However, inhibition of endogenous TGF-β, activin-A or addition of biologically active TGF-β1, and activin-A, did not inhibit or stimulate IDO1 biosynthesis in human DCs treated with CTB-INS. While inhibition with the kinase inhibitor, RepSox, blocked SMAD2/3 phosphorylation and diminished IDO1 biosynthesis in a concentration dependent manner. Specific blocking of the TGF-β type 1 kinase receptor with SB-431542 did not arrest IDO1 biosynthesis, suggesting the involvement of a different kinase pathway other than TGF-β type 1 receptor kinase in CTB-INS induction of IDO1 in human moDCs. Together, our experimental findings identify additional immunoregulatory proteins induced by the CTB-INS fusion protein, suggesting CTB-INS may utilize multiple mechanisms in the induction of tolerance in human moDCs.
Collapse
|
195
|
Ritelli M, Morlino S, Giacopuzzi E, Bernardini L, Torres B, Santoro G, Ravasio V, Chiarelli N, D'Angelantonio D, Novelli A, Grammatico P, Colombi M, Castori M. A recognizable systemic connective tissue disorder with polyvalvular heart dystrophy and dysmorphism associated with TAB2
mutations. Clin Genet 2017; 93:126-133. [DOI: 10.1111/cge.13032] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/29/2017] [Accepted: 04/01/2017] [Indexed: 12/14/2022]
Affiliation(s)
- M. Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - S. Morlino
- Division/Laboratory of Medical Genetics, Department of Molecular Medicine; University “La Sapienza”, San Camillo-Forlanini Hospital; Rome Italy
| | - E. Giacopuzzi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - L. Bernardini
- Unità di Citogenetica; IRCCS Casa Sollievo della Sofferenza; San Giovanni Rotondo (Foggia) Italy
| | - B. Torres
- Unità di Citogenetica; IRCCS Casa Sollievo della Sofferenza; San Giovanni Rotondo (Foggia) Italy
| | - G. Santoro
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - V. Ravasio
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - N. Chiarelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - D. D'Angelantonio
- Division/Laboratory of Medical Genetics, Department of Molecular Medicine; University “La Sapienza”, San Camillo-Forlanini Hospital; Rome Italy
| | - A. Novelli
- Laboratory of Medical Genetics; Bambino Gesù Children's Hospital, IRCCS; Rome Italy
| | - P. Grammatico
- Division/Laboratory of Medical Genetics, Department of Molecular Medicine; University “La Sapienza”, San Camillo-Forlanini Hospital; Rome Italy
| | - M. Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine; University of Brescia; Brescia Italy
| | - M. Castori
- Unità di Genetica Medica; IRCCS Casa Sollievo della Sofferenza; San Giovanni Rotondo (Foggia) Italy
| |
Collapse
|
196
|
Differential expression of transforming growth factor-beta1, connective tissue growth factor, phosphorylated-SMAD2/3 and phosphorylated-ERK1/2 during mouse tooth development. J Mol Histol 2017; 48:347-355. [PMID: 28825193 DOI: 10.1007/s10735-017-9733-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 08/16/2017] [Indexed: 10/19/2022]
Abstract
Connective tissue growth factor (CTGF) is a downstream mediator of transforming growth factor-beta 1 (TGF-β1) and TGF-β1-induced CTGF expression is regulated through SMAD and mitogen-activated protein kinase (MAPK) signaling pathways. The fine modulation of TGF-β1 signaling is very important to the process of tooth development. However, little is known about the localization of CTGF, MAPK and SMAD in the context of TGF-β1 signaling during odontogenesis. Hence, we aimed to investigate the expression of TGF-β1, CTGF, phosphorylated-SMAD2/3 (p-SMAD2/3) and phosphorylated-ERK1/2 (p-ERK1/2). ICR mice heads of embryonic (E) day 13.5, E14.5, E16.5, postnatal (PN) day 0.5 and PN3.5 were processed for immunohistochemistry. Results revealed that at E13.5, TGF-β1 and CTGF were strongly expressed in dental epithelium (DE) and dental mesenchyme (DM), while p-SMAD2/3 was intensely expressed in the internal side of DE. p-ERK1/2 was not present in DE or DM. At E14.5 and E16.5, strong staining for TGF-β1 and CTGF was detected in enamel knot (EK) and dental papilla (DPL). DPL was intensely stained for p-ERK1/2 but negatively stained for p-SMAD2/3. There was no staining for p-SMAD2/3 and p-ERK1/2 in EK. At PN0.5 and PN3.5, moderate to intense staining for TGF-β1 and CTGF was evident in preameloblasts (PA), secretary ameloblasts (SA) and dental pulp (DP). p-SMAD2/3 was strongly expressed in SA and DP but sparsely localized in PA. p-ERK1/2 was intensely expressed in DP, although negative staining was observed in PA and SA. These data demonstrate that TGF-β1 and CTGF show an identical expression pattern, while p-SMAD2/3 and p-ERK1/2 exhibit differential expression, and indicate that p-SMAD2/3 and p-ERK1/2 might play a regulatory role in TGF-β1 induced CTGF expression during tooth development.
Collapse
|
197
|
Gudey SK, Sundar R, Heldin CH, Bergh A, Landström M. Pro-invasive properties of Snail1 are regulated by sumoylation in response to TGFβ stimulation in cancer. Oncotarget 2017; 8:97703-97726. [PMID: 29228645 PMCID: PMC5716685 DOI: 10.18632/oncotarget.20097] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 05/19/2017] [Indexed: 01/11/2023] Open
Abstract
Transforming growth factor β (TGFβ) is a key regulator of epithelial-to-mesenchymal transition (EMT) during embryogenesis and in tumors. The effect of TGFβ, on ΕΜΤ, is conveyed by induction of the pro-invasive transcription factor Snail1. In this study, we report that TGFβ stimulates Snail1 sumoylation in aggressive prostate, breast and lung cancer cells. Sumoylation of Snail1 lysine residue 234 confers its transcriptional activity, inducing the expression of classical EMT genes, as well as TGFβ receptor I (TβRI) and the transcriptional repressor Hes1. Mutation of Snail1 lysine residue 234 to arginine (K234R) abolished sumoylation of Snail1, as well as its migratory and invasive properties in human prostate cancer cells. An increased immunohistochemical expression of Snail1, Sumo1, TβRI, Hes1, and c-Jun was observed in aggressive prostate cancer tissues, consistent with their functional roles in tumorigenesis.
Collapse
Affiliation(s)
| | - Reshma Sundar
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Carl-Henrik Heldin
- Ludwig Institute for Cancer Research, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anders Bergh
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Marene Landström
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| |
Collapse
|
198
|
Zhu Y, Qiu Y, Yu H, Yi S, Su W, Kijlstra A, Yang P. Aberrant DNA methylation of GATA binding protein 3 (GATA3), interleukin-4 (IL-4), and transforming growth factor-β (TGF-β) promoters in Behcet's disease. Oncotarget 2017; 8:64263-64272. [PMID: 28969068 PMCID: PMC5610000 DOI: 10.18632/oncotarget.19500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 06/12/2017] [Indexed: 12/18/2022] Open
Abstract
The pathogenesis of Behcet's disease (BD) remains poorly understood. The purpose of this study was to investigate whether an aberrant DNA methylation of transcriptional and inflammatory factors, including TBX21, GATA3, RORγt, FOXP3, IFN-γ, IL-4, IL-17A and TGF-β, in CD4+T confers risk to BD. We found that the promoter methylation level of GATA3, IL-4 and TGF-β was significantly up-regulated in active BD patients and negatively correlated with the corresponding mRNA expression. The mRNA expression of GATA3 and TGF-β was markedly down-regulated in active BD patients compared to healthy individuals. Treatment with corticosteroids and cyclosporine (CsA) resulted in a decrease of the methylation level of GATA3 and TGF-β in inactive BD patients. Our results suggest that an aberrant DNA methylation of GATA3 and TGF-β is associated with their mRNA expression and participates in the pathogenesis of BD.
Collapse
Affiliation(s)
- Yunyun Zhu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Yiguo Qiu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Hongsong Yu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Shenglan Yi
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Wencheng Su
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| | - Aize Kijlstra
- University Eye Clinic Maastricht, Maastricht, The Netherlands
| | - Peizeng Yang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Ophthalmology and Chongqing Eye Institute, Chongqing, China
| |
Collapse
|
199
|
Benn A, Hiepen C, Osterland M, Schütte C, Zwijsen A, Knaus P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence. FASEB J 2017; 31:4720-4733. [PMID: 28733457 PMCID: PMC5636702 DOI: 10.1096/fj.201700193rr] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 06/27/2017] [Indexed: 01/04/2023]
Abstract
Before the onset of sprouting angiogenesis, the endothelium is prepatterned for the positioning of tip and stalk cells. Both cell identities are not static, as endothelial cells (ECs) constantly compete for the tip cell position in a dynamic fashion. Here, we show that both bone morphogenetic protein 2 (BMP2) and BMP6 are proangiogenic in vitro and ex vivo and that the BMP type I receptors, activin receptor-like kinase 3 (ALK3) and ALK2, play crucial and distinct roles in this process. BMP2 activates the expression of tip cell-associated genes, such as delta-like ligand 4 (DLL4) and kinase insert domain receptor (KDR), and p38-heat shock protein 27 (HSP27)-dependent cell migration, thereby generating tip cell competence. Whereas BMP6 also triggers collective cell migration via the p38-HSP27 signaling axis, BMP6 induces in addition SMAD1/5 signaling, thereby promoting the expression of stalk cell-associated genes, such as hairy and enhancer of split 1 (HES1) and fms-like tyrosine kinase 1 (FLT1). Specifically, ALK3 is required for sprouting from HUVEC spheroids, whereas ALK2 represses sprout formation. We demonstrate that expression levels and respective complex formation of BMP type I receptors in ECs determine stalk vs. tip cell identity, thus contributing to endothelial plasticity during sprouting angiogenesis. As antiangiogenic monotherapies that target the VEGF or ALK1 pathways have not fulfilled efficacy objectives in clinical trials, the selective targeting of the ALK2/3 pathways may be an attractive new approach.-Benn, A., Hiepen, C., Osterland, M., Schütte, C., Zwijsen, A., Knaus, P. Role of bone morphogenetic proteins in sprouting angiogenesis: differential BMP receptor-dependent signaling pathways balance stalk vs. tip cell competence.
Collapse
Affiliation(s)
- Andreas Benn
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany.,Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Christian Hiepen
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| | - Marc Osterland
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - Christof Schütte
- Zuse Institute Berlin, Berlin, Germany.,Institute for Mathematics, Freie Universität Berlin, Berlin, Germany
| | - An Zwijsen
- Vlaams Instituut voor Biotechnologie (VIB) Center for Brain and Disease Research, KU Leuven, Leuven, Belgium.,Department of Human Genetics, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany; .,Deutsche Forschungsgemeinschaft (DFG) Graduate School 1093, Berlin School of Integrative Oncology, Berlin, Germany.,DFG Graduate School 203, Berlin-Brandenburg School for Regenerative Therapies, Berlin, Germany
| |
Collapse
|
200
|
Dichotomous roles of TGF-β in human cancer. Biochem Soc Trans 2017; 44:1441-1454. [PMID: 27911726 DOI: 10.1042/bst20160065] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/27/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-β (TGF-β) mediates numerous biological processes, including embryonic development and the maintenance of cellular homeostasis in a context-dependent manner. Consistent with its central role in maintaining cellular homeostasis, inhibition of TGF-β signaling results in disruption of normal homeostatic processes and subsequent carcinogenesis, defining the TGF-β signaling pathway as a tumor suppressor. However, once carcinogenesis is initiated, the TGF-β signaling pathway promotes cancer progression. This dichotomous function of the TGF-β signaling pathway is mediated through altering effects on both the cancer cells, by inducing apoptosis and inhibiting proliferation, and the tumor microenvironment, by promoting angiogenesis and inhibiting immunosurveillance. Current studies support inhibition of TGF-β signaling either alone, or in conjunction with anti-angiogenic therapy or immunotherapy as a promising strategy for the treatment of human cancers.
Collapse
|