51
|
Shi Y, Chen X, Huang C, Pollock C. RIPK3: A New Player in Renal Fibrosis. Front Cell Dev Biol 2020; 8:502. [PMID: 32613000 PMCID: PMC7308494 DOI: 10.3389/fcell.2020.00502] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 05/26/2020] [Indexed: 12/28/2022] Open
Abstract
Chronic kidney disease (CKD) is the end result of a plethora of renal insults, including repeated episodes of acute or toxic kidney injury, glomerular, or diabetic kidney disease. It affects a large number of the population worldwide, resulting in significant personal morbidity and mortality and economic cost to the community. Hence it is appropriate to focus on treatment strategies that interrupt the development of kidney fibrosis, the end result of all forms of CKD, in addition to upstream factors that may be specific to certain diseases. However, the current clinical approach to prevent or manage renal fibrosis remains unsatisfactory. The rising importance of receptor-interacting serine/threonine-protein kinase (RIPK) 3 in the inflammatory response and TGF-β1 signaling is increasingly recognized. We discuss here the biological functions of RIPK3 and its role in the development of renal fibrosis.
Collapse
Affiliation(s)
- Ying Shi
- Nephrology, School of Medicine, Stanford University, Palo Alto, CA, United States
| | - Xinming Chen
- Kolling Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Chunling Huang
- Kolling Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| | - Carol Pollock
- Kolling Institute of Medical Research, Sydney Medical School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
52
|
Larson C, Oronsky B, Carter CA, Oronsky A, Knox SJ, Sher D, Reid TR. TGF-beta: a master immune regulator. Expert Opin Ther Targets 2020; 24:427-438. [PMID: 32228232 DOI: 10.1080/14728222.2020.1744568] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Transforming Growth Factor-Beta (TGF-β) is a master regulator of numerous cellular functions including cellular immunity. In cancer, TGF-β can function as a tumor promoter via several mechanisms including immunosuppression. Since the immune checkpoint pathways are co-opted in cancer to induce T cell tolerance, this review posits that TGF-β is a master checkpoint in cancer, whose negative regulatory influence overrides and controls that of other immune checkpoints.Areas Covered: This review examines therapeutic agents that target TGF-β and its signaling pathways for the treatment of cancer which may be classifiable as checkpoint inhibitors in the broadest sense. This concept is supported by the observations that 1) only a subset of patients benefit from current checkpoint inhibitor therapies, 2) the presence of TGF-β in the tumor microenvironment is associated with excluded or cold tumors, and resistance to checkpoint inhibitors, and 3) existing biomarkers such as PD-1, PD-L1, microsatellite instability and tumor mutational burden are inadequate to reliably and adequately identify immuno-responsive patients. By contrast, TGF-β overexpression is a widespread and profoundly negative molecular hallmark in multiple tumor types.Expert Opinion: TGF-β status may serve as a biomarker to predict responsiveness and as a therapeutic target to increase the activity of immunotherapies.
Collapse
Affiliation(s)
| | | | | | - Arnold Oronsky
- EpicentRx, San Diego, CA, USA.,InterWest Partners, Menlo Park, CA, USA
| | - Susan J Knox
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - David Sher
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Tony R Reid
- Department of Medical Oncology, UC San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
53
|
Tzavlaki K, Moustakas A. TGF-β Signaling. Biomolecules 2020; 10:biom10030487. [PMID: 32210029 PMCID: PMC7175140 DOI: 10.3390/biom10030487] [Citation(s) in RCA: 411] [Impact Index Per Article: 102.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 03/19/2020] [Accepted: 03/20/2020] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) represents an evolutionarily conserved family of secreted polypeptide factors that regulate many aspects of physiological embryogenesis and adult tissue homeostasis. The TGF-β family members are also involved in pathophysiological mechanisms that underlie many diseases. Although the family comprises many factors, which exhibit cell type-specific and developmental stage-dependent biological actions, they all signal via conserved signaling pathways. The signaling mechanisms of the TGF-β family are controlled at the extracellular level, where ligand secretion, deposition to the extracellular matrix and activation prior to signaling play important roles. At the plasma membrane level, TGF-βs associate with receptor kinases that mediate phosphorylation-dependent signaling to downstream mediators, mainly the SMAD proteins, and mediate oligomerization-dependent signaling to ubiquitin ligases and intracellular protein kinases. The interplay between SMADs and other signaling proteins mediate regulatory signals that control expression of target genes, RNA processing at multiple levels, mRNA translation and nuclear or cytoplasmic protein regulation. This article emphasizes signaling mechanisms and the importance of biochemical control in executing biological functions by the prototype member of the family, TGF-β.
Collapse
|
54
|
Zhang Y, Que J. BMP Signaling in Development, Stem Cells, and Diseases of the Gastrointestinal Tract. Annu Rev Physiol 2020; 82:251-273. [PMID: 31618602 DOI: 10.1146/annurev-physiol-021119-034500] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The bone morphogenetic protein (BMP) pathway is essential for the morphogenesis of multiple organs in the digestive system. Abnormal BMP signaling has also been associated with disease initiation and progression in the gastrointestinal (GI) tract and associated organs. Recent studies using animal models, tissue organoids, and human pluripotent stem cells have significantly expanded our understanding of the roles played by BMPs in the development and homeostasis of GI organs. It is clear that BMP signaling regulates GI function and disease progression that involve stem/progenitor cells and inflammation in a tissue-specific manner. In this review we discuss these new findings with a focus on the esophagus, stomach, and intestine.
Collapse
Affiliation(s)
- Yongchun Zhang
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA; .,Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| | - Jianwen Que
- Division of Digestive and Liver Diseases, Department of Medicine, Columbia University, New York, NY 10032, USA; .,Columbia Center for Human Development, Columbia University Medical Center, New York, NY 10032, USA
| |
Collapse
|
55
|
The Role of Ubiquitination in Regulating Embryonic Stem Cell Maintenance and Cancer Development. Int J Mol Sci 2019; 20:ijms20112667. [PMID: 31151253 PMCID: PMC6600158 DOI: 10.3390/ijms20112667] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/19/2019] [Accepted: 05/28/2019] [Indexed: 12/18/2022] Open
Abstract
Ubiquitination regulates nearly every aspect of cellular events in eukaryotes. It modifies intracellular proteins with 76-amino acid polypeptide ubiquitin (Ub) and destines them for proteolysis or activity alteration. Ubiquitination is generally achieved by a tri-enzyme machinery involving ubiquitin activating enzymes (E1), ubiquitin conjugating enzymes (E2) and ubiquitin ligases (E3). E1 activates Ub and transfers it to the active cysteine site of E2 via a transesterification reaction. E3 coordinates with E2 to mediate isopeptide bond formation between Ub and substrate protein. The E1-E2-E3 cascade can create diverse types of Ub modifications, hence effecting distinct outcomes on the substrate proteins. Dysregulation of ubiquitination results in severe consequences and human diseases. There include cancers, developmental defects and immune disorders. In this review, we provide an overview of the ubiquitination machinery and discuss the recent progresses in the ubiquitination-mediated regulation of embryonic stem cell maintenance and cancer biology.
Collapse
|
56
|
Kloth K, Bierhals T, Johannsen J, Harms FL, Juusola J, Johnson MC, Grange DK, Kutsche K. Biallelic variants in SMAD6 are associated with a complex cardiovascular phenotype. Hum Genet 2019; 138:625-634. [DOI: 10.1007/s00439-019-02011-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/03/2019] [Indexed: 01/10/2023]
|
57
|
Balagangadharan K, Trivedi R, Vairamani M, Selvamurugan N. Sinapic acid-loaded chitosan nanoparticles in polycaprolactone electrospun fibers for bone regeneration in vitro and in vivo. Carbohydr Polym 2019; 216:1-16. [PMID: 31047045 DOI: 10.1016/j.carbpol.2019.04.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/23/2022]
Abstract
Sinapic acid (SA) is a plant-derived phenolic compound known for its multiple biological properties, but its role in the promotion of bone formation is not yet well-studied. Moreover, the delivery of SA is hindered by its complex hydrophobic nature, limiting its bioavailability. In this study, we fabricated a drug delivery system using chitosan nanoparticles (nCS) loaded with SA at different concentrations. These were incorporated into polycaprolactone (PCL) fibers via an electrospinning method. nCS loaded with 50 μM SA in PCL fibers promoted osteoblast differentiation. Furthermore, SA treatment activated the osteogenesis signaling pathways in mouse mesenchymal stem cells. A critical-sized rat calvarial bone defect model system identified that the inclusion of SA into PCL/nCS fibers accelerated bone formation. Collectively, these data suggest that SA promoted osteoblast differentiation in vitro and bone formation in vivo, possibly by activating the TGF-β1/BMP/Smads/Runx2 signaling pathways, suggesting SA might have therapeutic benefits in bone regeneration.
Collapse
Affiliation(s)
- Kalimuthu Balagangadharan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Ritu Trivedi
- Division of Endocrinology, Central Drug Research Institute (Council of Scientific and Industrial Research), Lucknow 226031, Uttar Pradesh, India
| | - Mariappanadar Vairamani
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India
| | - Nagarajan Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603203, Tamil Nadu, India.
| |
Collapse
|
58
|
Hu B, Mao Z, Du Q, Jiang X, Wang Z, Xiao Z, Zhu D, Wang X, Zhu Y, Wang H. miR-93-5p targets Smad7 to regulate the transforming growth factor-β1/Smad3 pathway and mediate fibrosis in drug-resistant prolactinoma. Brain Res Bull 2019; 149:21-31. [PMID: 30946881 DOI: 10.1016/j.brainresbull.2019.03.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/23/2019] [Accepted: 03/28/2019] [Indexed: 12/29/2022]
Abstract
Prolactinoma is a common subtype of pituitary tumors. Dopamine receptor agonists are the preferred treatment for prolactinoma; however, with this therapy, drug resistance often occurs. In our previous work, we found that partial resistant prolactinomas showed increased fibrosis and that the transforming growth factor (TGF)-β1/Smad3 signaling pathway mediated fibrosis and was involved in drug resistance. Additionally, the success of surgery is known to be heavily influenced by the consistency of the pituitary adenoma. Therefore, in this study, we aimed to clarify the mechanisms of fibrosis in prolactinoma. Using high-throughput sequencing for analysis of microRNAs, we found that miR-93-5p was significantly upregulated in prolactinoma samples with a high degree of fibrosis compared with that in samples without fibrosis. Furthermore, we found that miR-93-5p was negatively correlated with the relative expression of Smad7 and positively correlated with the relative expression of TGF-β1 in clinical prolactinoma samples. In addition, luciferase reporter assays showed that miR-93-5p could downregulate the Smad7 gene, an important inhibitor of the TGF-β1/Smad3 signaling pathway, and activate TGF-β1/Smad3 signaling-mediated fibrosis in a feed-forward loop. Moreover, miR-93-5p could enhance the drug resistance of prolactinoma cells by regulation of TGF-β1/Smad3-dependent fibrosis. Taken together, our findings demonstrated that miR-93-5p may be a potential therapeutic target for inhibiting fibrosis and reducing drug resistance in prolactinoma cells.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhigang Mao
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiu Du
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaobing Jiang
- Department of Neurosurgery, Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zheng Xiao
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dimin Zhu
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xin Wang
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Yonghong Zhu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Haijun Wang
- Department of Neurosurgery and Pituitary Tumor Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
59
|
Derynck R, Budi EH. Specificity, versatility, and control of TGF-β family signaling. Sci Signal 2019; 12:12/570/eaav5183. [PMID: 30808818 DOI: 10.1126/scisignal.aav5183] [Citation(s) in RCA: 492] [Impact Index Per Article: 98.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Encoded in mammalian cells by 33 genes, the transforming growth factor-β (TGF-β) family of secreted, homodimeric and heterodimeric proteins controls the differentiation of most, if not all, cell lineages and many aspects of cell and tissue physiology in multicellular eukaryotes. Deregulation of TGF-β family signaling leads to developmental anomalies and disease, whereas enhanced TGF-β signaling contributes to cancer and fibrosis. Here, we review the fundamentals of the signaling mechanisms that are initiated upon TGF-β ligand binding to its cell surface receptors and the dependence of the signaling responses on input from and cooperation with other signaling pathways. We discuss how cells exquisitely control the functional presentation and activation of heteromeric receptor complexes of transmembrane, dual-specificity kinases and, thus, define their context-dependent responsiveness to ligands. We also introduce the mechanisms through which proteins called Smads act as intracellular effectors of ligand-induced gene expression responses and show that the specificity and impressive versatility of Smad signaling depend on cross-talk from other pathways. Last, we discuss how non-Smad signaling mechanisms, initiated by distinct ligand-activated receptor complexes, complement Smad signaling and thus contribute to cellular responses.
Collapse
Affiliation(s)
- Rik Derynck
- Department of Cell and Tissue Biology and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA.
| | - Erine H Budi
- Department of Cell and Tissue Biology and Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
60
|
Tang Y, Reissig S, Glasmacher E, Regen T, Wanke F, Nikolaev A, Gerlach K, Popp V, Karram K, Fantini MC, Schattenberg JM, Galle PR, Neurath MF, Weigmann B, Kurschus FC, Hövelmeyer N, Waisman A. Alternative Splice Forms of CYLD Mediate Ubiquitination of SMAD7 to Prevent TGFB Signaling and Promote Colitis. Gastroenterology 2019; 156:692-707.e7. [PMID: 30315770 DOI: 10.1053/j.gastro.2018.10.023] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 10/03/2018] [Accepted: 10/04/2018] [Indexed: 12/30/2022]
Abstract
BACKGROUND & AIMS The CYLD lysine 63 deubiquitinase gene (CYLD) encodes tumor suppressor protein that is mutated in familial cylindromatosus, and variants have been associated with Crohn disease (CD). Splice forms of CYLD that lack exons 7 and 8 regulate transcription factors and functions of immune cells. We examined the expression of splice forms of CYLD in colon tissues from patients with CD and their effects in mice. METHODS We performed immunohistochemical analyses of colon tissues from patients with untreated CD and patients without inflammatory bowel diseases (controls). We obtained mice that expressed splice forms of CYLD (sCYLD mice) without or with SMAD7 (sCYLD/SMAD7 mice) from transgenes and CYLD-knockout mice (with or without transgenic expression of SMAD7) and performed endoscopic analyses. Colitis was induced in Rag1-/- mice by transfer of CD4+ CD62L+ T cells from C57/Bl6 or transgenic mice. T cells were isolated from mice and analyzed by flow cytometry and quantitative real-time polymerase chain reaction and intestinal tissues were analyzed by histology and immunohistochemistry. CYLD forms were expressed in mouse embryonic fibroblasts, primary T cells, and HEK293T cells, which were analyzed by immunoblot, mobility shift, and immunoprecipitation assays. RESULTS The colonic lamina propria from patients with CD was infiltrated by T cells and had higher levels of sCYLD (but not full-length CYLD) and SMAD7 than tissues from controls. Incubation of mouse embryonic fibroblasts and T cells with transforming growth factor β increased their production of sCYLD and decreased full-length CYLD. Transgenic expression of sCYLD and SMAD7 in T cells prevented the differentiation of regulatory T cells and T-helper type 17 cells and increased the differentiation of T-helper type 1 cells. The same effects were observed in colon tissues from sCYLD/SMAD7 mice but not in those from CYLD-knockout SMAD7 mice. The sCYLD mice had significant increases in the numbers of T-helper type 1 cells and CD44high CD62Llow memory-effector CD4+ T cells in the spleen and mesenteric lymph nodes compared with wild-type mice; sCYLD/SMAD7 mice had even larger increases. The sCYLD/SMAD7 mice spontaneously developed severe colitis, with infiltration of the colon by dendritic cells, neutrophils, macrophages, and CD4+ T cells and increased levels of Ifng, Il6, Il12a, Il23a, and Tnf mRNAs. Co-transfer of regulatory T cells from wild-type, but not from sCYLD/SMAD7, mice prevented the induction of colitis in Rag1-/- mice by CD4+ T cells. We found increased levels of poly-ubiquitinated SMAD7 in sCYLD CD4+ T cells. CYLD formed a nuclear complex with SMAD3, whereas sCYLD recruited SMAD7 to the nucleus, which inhibited the expression of genes regulated by SMAD3 and SMAD4. We found that sCYLD mediated lysine 63-linked ubiquitination of SMAD7. The sCYLD-SMAD7 complex inhibited transforming growth factor β signaling in CD4+ T cells. CONCLUSIONS Levels of the spliced form of CYLD are increased in colon tissues from patients with CD. sCYLD mediates ubiquitination and nuclear translocation of SMAD7 and thereby decreases transforming growth factor β signaling in T cells. This prevents immune regulatory mechanisms and leads to colitis in mice.
Collapse
Affiliation(s)
- Yilang Tang
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Sonja Reissig
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Elke Glasmacher
- Institute for Diabetes and Obesity, Helmholtz Zentrum München, Garching, Germany
| | - Tommy Regen
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Florian Wanke
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Alexei Nikolaev
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Katharina Gerlach
- Department of Internal Medicine I, University Hospital Erlangen, University Erlangen-Nürnberg, Erlangen, Germany
| | - Vanessa Popp
- Department of Internal Medicine I, University Hospital Erlangen, University Erlangen-Nürnberg, Erlangen, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Massimo C Fantini
- Department of Systems Medicine, University of Tor Vergata, Rome, Italy
| | - Jörn M Schattenberg
- Department of Internal Medicine I, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Peter R Galle
- Department of Internal Medicine I, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Markus F Neurath
- Department of Internal Medicine I, University Hospital Erlangen, University Erlangen-Nürnberg, Erlangen, Germany
| | - Benno Weigmann
- Department of Internal Medicine I, University Hospital Erlangen, University Erlangen-Nürnberg, Erlangen, Germany
| | - Florian C Kurschus
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Nadine Hövelmeyer
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Centre, Johannes Gutenberg University of Mainz, Mainz, Germany.
| |
Collapse
|
61
|
Samba-Mondonga M, Calvé A, Mallette FA, Santos MM. MyD88 Regulates the Expression of SMAD4 and the Iron Regulatory Hormone Hepcidin. Front Cell Dev Biol 2018; 6:105. [PMID: 30234111 PMCID: PMC6127602 DOI: 10.3389/fcell.2018.00105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 08/14/2018] [Indexed: 12/26/2022] Open
Abstract
The myeloid differentiation primary response gene 88 (MyD88) is an adaptive protein that is essential for the induction of inflammatory cytokines through almost all the Toll-like receptors (TLRs). TLRs recognize molecular patterns present in microorganisms called pathogen-associated molecular patterns. Therefore, MyD88 plays an important role in innate immunity since its activation triggers the first line of defense against microorganisms. Herein, we describe the first reported role of MyD88 in an interconnection between innate immunity and the iron-sensing pathway (BMP/SMAD4). We found that direct interaction of MyD88 with SMAD4 protein activated hepcidin expression. The iron regulatory hormone hepcidin is indispensable for the intestinal regulation of iron absorption and iron recycling by macrophages. We show that MyD88 induces hepcidin expression in a manner dependent on the proximal BMP responsive element on the hepcidin gene (HAMP) promoter. We identified the Toll/interleukin-1 receptor (TIR) domain of MyD88 as the domain of interaction with SMAD4. Furthermore, we show that BMP6 stimulation, which activates SMAD6 expression, also induces MyD88 proteosomal degradation as a negative feedback mechanism to limit hepcidin induction. Finally, we report that the MyD88 gain-of-function L265P mutation, frequently encountered in B-cell lymphomas such as Waldenström’s macroglobulinemia, enhances hepcidin expression and iron accumulation in B cell lines. Our results reveal a new potential role for MyD88 in the SMAD signaling pathway and iron homeostasis regulation.
Collapse
Affiliation(s)
- Macha Samba-Mondonga
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| | - Annie Calvé
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada
| | - Frédérick A Mallette
- Département de Médecine, Université de Montréal, Montréal, QC, Canada.,Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC, Canada
| | - Manuela M Santos
- Nutrition and Microbiome Laboratory, Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montréal, QC, Canada.,Département de Médecine, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
62
|
The Dynamic Roles of TGF-β Signalling in EBV-Associated Cancers. Cancers (Basel) 2018; 10:cancers10080247. [PMID: 30060514 PMCID: PMC6115974 DOI: 10.3390/cancers10080247] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/23/2018] [Accepted: 07/25/2018] [Indexed: 02/07/2023] Open
Abstract
The transforming growth factor-β (TGF-β) signalling pathway plays a critical role in carcinogenesis. It has a biphasic action by initially suppressing tumorigenesis but promoting tumour progression in the later stages of disease. Consequently, the functional outcome of TGF-β signalling is strongly context-dependent and is influenced by various factors including cell, tissue and cancer type. Disruption of this pathway can be caused by various means, including genetic and environmental factors. A number of human viruses have been shown to modulate TGF-β signalling during tumorigenesis. In this review, we describe how this pathway is perturbed in Epstein-Barr virus (EBV)-associated cancers and how EBV interferes with TGF-β signal transduction. The role of TGF-β in regulating the EBV life cycle in tumour cells is also discussed.
Collapse
|
63
|
Biological processes and signal transduction pathways regulated by the protein methyltransferase SETD7 and their significance in cancer. Signal Transduct Target Ther 2018; 3:19. [PMID: 30013796 PMCID: PMC6043541 DOI: 10.1038/s41392-018-0017-6] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/05/2018] [Accepted: 03/21/2018] [Indexed: 02/07/2023] Open
Abstract
Protein methyltransferases have been shown to methylate histone and non-histone proteins, leading to regulation of several biological processes that control cell homeostasis. Over the past few years, the histone-lysine N-methyltransferase SETD7 (SETD7; also known as SET7/9, KIAA1717, KMT7, SET7, SET9) has emerged as an important regulator of at least 30 non-histone proteins and a potential target for the treatment of several human diseases. This review discusses current knowledge of the structure and subcellular localization of SETD7, as well as its function as a histone and non-histone methyltransferase. This work also underlines the putative contribution of SETD7 to the regulation of gene expression, control of cell proliferation, differentiation and endoplasmic reticulum stress, which indicate that SETD7 is a candidate for novel targeted therapies with the aim of either stimulating or inhibiting its activity, depending on the cell signaling context.
Collapse
|
64
|
Liang ZG, Yao H, Xie RS, Gong CL, Tian Y. MicroRNA‑20b‑5p promotes ventricular remodeling by targeting the TGF‑β/Smad signaling pathway in a rat model of ischemia‑reperfusion injury. Int J Mol Med 2018; 42:975-987. [PMID: 29786750 PMCID: PMC6034914 DOI: 10.3892/ijmm.2018.3695] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 04/26/2018] [Indexed: 01/04/2023] Open
Abstract
Myocardial ischemic injury results from severe impairment of the coronary blood supply and may lead to metabolic and ultrastructural changes, thereby causing irreversible damage. MicroRNA (miR)-20b-5p has been demonstrated to be involved in malignancies of the breast, colorectum, stomach, blood and oropharynx. The present study aimed to investigate the effects of miR-20b-5p on ventricular remodeling following myocardial ischemia-reperfusion (IR) injury in rats by targeting small mothers against decapentaplegic homolog 7 (Smad7) via the transforming growth factor-β (TGF-β)/Smad signaling pathway. A total of 70 adult male Sprague-Dawley rats were divided into seven groups: Sham group, IR group, negative control group, miR-20b-5p mimics group, miR-20b-5p inhibitors group, small interfering RNA (siRNA)-Smad7 group, and miR-20b-5p inhibitors + siRNA-Smad7 group. Dual luciferase reporter gene assays were used to verify the association between miR-20b-5p and Smad7. Myocardial infarction size, myocardial collagen volume fraction and perivascular collagen area were detected separately using triphenyltetrazolium chloride and Masson's staining. The rate of positive expression of Smad7 was detected using immunohistochemistry, and the expression levels of miR-20b-5p, TGF-β1, Smad3 and Smad7 were detected using reverse transcription-quantitative polymerase chain reaction and western blot analyses. The findings revealed that miR-20b-5p inhibited Smad7. Compared with the sham group, the other six groups had increased myocardial infarction size, myocardial collagen, and expression of miR-20b-5p, TGF-β1 and Smad3, and decreased expression of Smad7. Compared with the IR group, the miR-20b-5p mimics group and the siRNA-Smad7 group had increased myocardial infarction size and myocardial collagen, increased expression of TGF-β1 and Smad3, and decreased expression of Smad7. The expression of miR-20b-5p was markedly increased in the miR-20b-5p mimics group, but did not differ significantly from that in the siRNA-Smad7 group. The results demonstrated that miR-20b-5p promoted ventricular remodeling following myocardial IR injury in rats by inhibiting the expression of Smad7 through activating the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Zhao-Guang Liang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Hong Yao
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Rong-Sheng Xie
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Chun-Lin Gong
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| | - Ye Tian
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150001, P.R. China
| |
Collapse
|
65
|
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
|
66
|
DiVito KA, Daniele MA, Roberts SA, Ligler FS, Adams AA. Microfabricated blood vessels undergo neoangiogenesis. Biomaterials 2017; 138:142-152. [DOI: 10.1016/j.biomaterials.2017.05.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/25/2017] [Accepted: 05/07/2017] [Indexed: 01/06/2023]
|
67
|
Ayerst BI, Merry CLR, Day AJ. The Good the Bad and the Ugly of Glycosaminoglycans in Tissue Engineering Applications. Pharmaceuticals (Basel) 2017; 10:E54. [PMID: 28608822 PMCID: PMC5490411 DOI: 10.3390/ph10020054] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/05/2017] [Accepted: 06/05/2017] [Indexed: 12/14/2022] Open
Abstract
High sulfation, low cost, and the status of heparin as an already FDA- and EMA- approved product, mean that its inclusion in tissue engineering (TE) strategies is becoming increasingly popular. However, the use of heparin may represent a naïve approach. This is because tissue formation is a highly orchestrated process, involving the temporal expression of numerous growth factors and complex signaling networks. While heparin may enhance the retention and activity of certain growth factors under particular conditions, its binding 'promiscuity' means that it may also inhibit other factors that, for example, play an important role in tissue maintenance and repair. Within this review we focus on articular cartilage, highlighting the complexities and highly regulated processes that are involved in its formation, and the challenges that exist in trying to effectively engineer this tissue. Here we discuss the opportunities that glycosaminoglycans (GAGs) may provide in advancing this important area of regenerative medicine, placing emphasis on the need to move away from the common use of heparin, and instead focus research towards the utility of specific GAG preparations that are able to modulate the activity of growth factors in a more controlled and defined manner, with less off-target effects.
Collapse
Affiliation(s)
- Bethanie I Ayerst
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| | - Catherine L R Merry
- Stem Cell Glycobiology Group, Wolfson Centre for Stem Cells, Tissue Engineering & Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester M13 9PL, UK.
| |
Collapse
|
68
|
Gillis E, Kumar AA, Luyckx I, Preuss C, Cannaerts E, van de Beek G, Wieschendorf B, Alaerts M, Bolar N, Vandeweyer G, Meester J, Wünnemann F, Gould RA, Zhurayev R, Zerbino D, Mohamed SA, Mital S, Mertens L, Björck HM, Franco-Cereceda A, McCallion AS, Van Laer L, Verhagen JMA, van de Laar IMBH, Wessels MW, Messas E, Goudot G, Nemcikova M, Krebsova A, Kempers M, Salemink S, Duijnhouwer T, Jeunemaitre X, Albuisson J, Eriksson P, Andelfinger G, Dietz HC, Verstraeten A, Loeys BL. Candidate Gene Resequencing in a Large Bicuspid Aortic Valve-Associated Thoracic Aortic Aneurysm Cohort: SMAD6 as an Important Contributor. Front Physiol 2017; 8:400. [PMID: 28659821 PMCID: PMC5469151 DOI: 10.3389/fphys.2017.00400] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 05/26/2017] [Indexed: 12/30/2022] Open
Abstract
Bicuspid aortic valve (BAV) is the most common congenital heart defect. Although many BAV patients remain asymptomatic, at least 20% develop thoracic aortic aneurysm (TAA). Historically, BAV-related TAA was considered as a hemodynamic consequence of the valve defect. Multiple lines of evidence currently suggest that genetic determinants contribute to the pathogenesis of both BAV and TAA in affected individuals. Despite high heritability, only very few genes have been linked to BAV or BAV/TAA, such as NOTCH1, SMAD6, and MAT2A. Moreover, they only explain a minority of patients. Other candidate genes have been suggested based on the presence of BAV in knockout mouse models (e.g., GATA5, NOS3) or in syndromic (e.g., TGFBR1/2, TGFB2/3) or non-syndromic (e.g., ACTA2) TAA forms. We hypothesized that rare genetic variants in these genes may be enriched in patients presenting with both BAV and TAA. We performed targeted resequencing of 22 candidate genes using Haloplex target enrichment in a strictly defined BAV/TAA cohort (n = 441; BAV in addition to an aortic root or ascendens diameter ≥ 4.0 cm in adults, or a Z-score ≥ 3 in children) and in a collection of healthy controls with normal echocardiographic evaluation (n = 183). After additional burden analysis against the Exome Aggregation Consortium database, the strongest candidate susceptibility gene was SMAD6 (p = 0.002), with 2.5% (n = 11) of BAV/TAA patients harboring causal variants, including two nonsense, one in-frame deletion and two frameshift mutations. All six missense mutations were located in the functionally important MH1 and MH2 domains. In conclusion, we report a significant contribution of SMAD6 mutations to the etiology of the BAV/TAA phenotype.
Collapse
Affiliation(s)
- Elisabeth Gillis
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Ajay A Kumar
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Ilse Luyckx
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Christoph Preuss
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Université de MontrealMontreal, QC, Canada
| | - Elyssa Cannaerts
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Gerarda van de Beek
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Björn Wieschendorf
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium.,Department of Cardiac and Thoracic Vascular Surgery, University Hospital Schleswig-HolsteinLübeck, Germany
| | - Maaike Alaerts
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Nikhita Bolar
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Geert Vandeweyer
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Josephina Meester
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Florian Wünnemann
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Université de MontrealMontreal, QC, Canada
| | - Russell A Gould
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of MedicineBaltimore, MD, United States
| | - Rustam Zhurayev
- Department of Clinical pathology, Lviv National Medical University after Danylo HalytskyLviv, Ukraine
| | - Dmytro Zerbino
- Department of Clinical pathology, Lviv National Medical University after Danylo HalytskyLviv, Ukraine
| | - Salah A Mohamed
- Department of Cardiac and Thoracic Vascular Surgery, University Hospital Schleswig-HolsteinLübeck, Germany
| | - Seema Mital
- Cardiovascular Research, SickKids University HospitalToronto, ON, Canada
| | - Luc Mertens
- Cardiovascular Research, SickKids University HospitalToronto, ON, Canada
| | - Hanna M Björck
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska InstituteStockholm, Sweden
| | - Anders Franco-Cereceda
- Cardiothoracic Surgery Unit, Department of Molecular Medicine and Surgery, Karolinska InstituteStockholm, Sweden
| | - Andrew S McCallion
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of MedicineBaltimore, MD, United States
| | - Lut Van Laer
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Judith M A Verhagen
- Department of Clinical Genetics, Erasmus University Medical CenterRotterdam, Netherlands
| | | | - Marja W Wessels
- Department of Clinical Genetics, Erasmus University Medical CenterRotterdam, Netherlands
| | - Emmanuel Messas
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Université Paris Descartes, Paris Sorbonne Cité; Institut National de la Santé et de la Recherche Médicale, UMRSParis, France
| | - Guillaume Goudot
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Université Paris Descartes, Paris Sorbonne Cité; Institut National de la Santé et de la Recherche Médicale, UMRSParis, France
| | - Michaela Nemcikova
- Department of Biology and Medical Genetics, 2nd Faculty of Medicine-Charles University and Motol University HospitalPrague, Czechia
| | - Alice Krebsova
- Institute of Clinical and Experimental MedicinePrague, Czechia
| | - Marlies Kempers
- Department of Human Genetics, Radboud University Medical CentreNijmegen, Netherlands
| | - Simone Salemink
- Department of Human Genetics, Radboud University Medical CentreNijmegen, Netherlands
| | - Toon Duijnhouwer
- Department of Human Genetics, Radboud University Medical CentreNijmegen, Netherlands
| | - Xavier Jeunemaitre
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Université Paris Descartes, Paris Sorbonne Cité; Institut National de la Santé et de la Recherche Médicale, UMRSParis, France
| | - Juliette Albuisson
- Assistance Publique-Hôpitaux de Paris, Hôpital Européen Georges Pompidou; Université Paris Descartes, Paris Sorbonne Cité; Institut National de la Santé et de la Recherche Médicale, UMRSParis, France
| | - Per Eriksson
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska InstituteStockholm, Sweden
| | - Gregor Andelfinger
- Cardiovascular Genetics, Department of Pediatrics, CHU Sainte-Justine, Université de MontrealMontreal, QC, Canada
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of MedicineBaltimore, MD, United States.,Howard Hughes Medical InstituteBaltimore, MD, United States
| | - Aline Verstraeten
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium
| | - Bart L Loeys
- Faculty of Medicine and Health Sciences, Center of Medical Genetics, University of Antwerp and Antwerp University HospitalAntwerp, Belgium.,Department of Human Genetics, Radboud University Medical CentreNijmegen, Netherlands
| | | |
Collapse
|
69
|
Gen Y, Yasui K, Kitaichi T, Iwai N, Terasaki K, Dohi O, Hashimoto H, Fukui H, Inada Y, Fukui A, Jo M, Moriguchi M, Nishikawa T, Umemura A, Yamaguchi K, Konishi H, Naito Y, Itoh Y. ASPP2 suppresses invasion and TGF-β1-induced epithelial-mesenchymal transition by inhibiting Smad7 degradation mediated by E3 ubiquitin ligase ITCH in gastric cancer. Cancer Lett 2017; 398:52-61. [PMID: 28400336 DOI: 10.1016/j.canlet.2017.04.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 03/21/2017] [Accepted: 04/02/2017] [Indexed: 12/12/2022]
Abstract
ASPP2 regulates cell polarity and cell-cell adhesion by binding to, and co-localizing with PAR3 at tight junctions. Here we show a novel role of ASPP2 in suppressing gastric cancer (GC) invasiveness. Immunoprecipitation and immunofluorescence analyses showed that ASPP2 promoted the recruitment of PAR3 to cell-cell junctions in GC cells. Diminished expression of ASPP2 and loss of junctional PAR3 localization were significantly associated with diffuse-type histology, deeper invasion depth, positive peritoneal dissemination and worse prognosis in primary GC. ASPP2 suppressed migration and invasion of GC cells in vitro and peritoneal dissemination of GC cells in vivo in a mouse model. ASPP2 suppressed epithelial-mesenchymal transition (EMT) induced by TGF-β1-Smad2/3 signaling in GC cells through suppression of the degradation of Smad7, a negative regulator of TGF-β1-Smad2/3 signaling, by interacting with the E3 ubiquitin ligase ITCH. In conclusion, ASPP2 suppresses invasion, peritoneal dissemination and TGF-β1-induced EMT by inhibiting Smad7 degradation mediated by ITCH.
Collapse
Affiliation(s)
- Yasuyuki Gen
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Molecular Cytogenetics, Medical Research Institute and Graduate School of Medical and Dental Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohichiroh Yasui
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Tomoko Kitaichi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Naoto Iwai
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kei Terasaki
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osamu Dohi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hikaru Hashimoto
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hayato Fukui
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yutaka Inada
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akifumi Fukui
- Department of Gastroenterology and Hepatology, North Medical Center Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masayasu Jo
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Michihisa Moriguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Taichiro Nishikawa
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Atushi Umemura
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kanji Yamaguchi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyuki Konishi
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yuji Naito
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshito Itoh
- Department of Molecular Gastroenterology and Hepatology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
70
|
Abstract
Inhibitory Smads (I-Smads) have conserved carboxy-terminal MH2 domains but highly divergent amino-terminal regions when compared with receptor-regulated Smads (R-Smads) and common-partner Smads (co-Smads). Smad6 preferentially inhibits Smad signaling initiated by the bone morphogenetic protein (BMP) type I receptors ALK-3 and ALK-6, whereas Smad7 inhibits both transforming growth factor β (TGF-β)- and BMP-induced Smad signaling. I-Smads also regulate some non-Smad signaling pathways. Here, we discuss the vertebrate I-Smads, their roles as inhibitors of Smad activation and regulators of receptor stability, as scaffolds for non-Smad signaling, and their possible roles in the nucleus. We also discuss the posttranslational modification of I-Smads, including phosphorylation, ubiquitylation, acetylation, and methylation.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
71
|
Abstract
Inhibitory Smads (I-Smads) have conserved carboxy-terminal MH2 domains but highly divergent amino-terminal regions when compared with receptor-regulated Smads (R-Smads) and common-partner Smads (co-Smads). Smad6 preferentially inhibits Smad signaling initiated by the bone morphogenetic protein (BMP) type I receptors ALK-3 and ALK-6, whereas Smad7 inhibits both transforming growth factor β (TGF-β)- and BMP-induced Smad signaling. I-Smads also regulate some non-Smad signaling pathways. Here, we discuss the vertebrate I-Smads, their roles as inhibitors of Smad activation and regulators of receptor stability, as scaffolds for non-Smad signaling, and their possible roles in the nucleus. We also discuss the posttranslational modification of I-Smads, including phosphorylation, ubiquitylation, acetylation, and methylation.
Collapse
Affiliation(s)
- Keiji Miyazawa
- Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi 409-3898, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
72
|
Yang F, Wang N, Wang Y, Yu T, Wang H. Activin-SMAD signaling is required for maintenance of porcine iPS cell self-renewal through upregulation of NANOG and OCT4 expression. J Cell Physiol 2017; 232:2253-2262. [PMID: 27996082 DOI: 10.1002/jcp.25747] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/17/2016] [Accepted: 12/19/2016] [Indexed: 12/25/2022]
Abstract
Porcine induced pluripotent stem cells (piPSCs) retain the enormous potential for farm animal reproduction and translational medicine, and have been reported by many laboratories worldwide. Some piPSC lines were bFGF-dependence and showed mouse EpiSC-like morphology; other lines were LIF-dependence and showed mouse ESC-like morphology. Metastable state of piPSC line that required both LIF and bFGF was also reported. Because bona fide pig embryonic stem cells were not available, uncovering piPSC state-specific regulatory circuitries was the most important task. In this study, we explored the function of Activin-SMAD signaling pathway and its downstream activated target genes in piPSCs. Transcriptome analysis showed that genes involved in Activin-SMAD signaling pathway were evidently activated during porcine somatic cell reprogramming, regardless piPSCs were LIF- or bFGF-dependent. Addition of Activin A and overexpression of SMAD2/3 significantly promoted expressions of porcine NANOG and OCT4, whereas inhibition of Activin-SMAD signaling by SB431542 and SMAD7 reduced NANOG and OCT4 expressions, and induced piPSCs differentiation exiting from pluripotent state. Our data demonstrate that activation of Activin-SMAD signaling pathway by addition of Activin A in culture medium is necessary for maintenance of self-renewal in porcine pluripotent stem cells.
Collapse
Affiliation(s)
- Fan Yang
- Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ning Wang
- Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yaxian Wang
- Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Tong Yu
- Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Huayan Wang
- Department of Animal Biotechnology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
73
|
Sharum IB, Granados-Aparici S, Warrander FC, Tournant FP, Fenwick MA. Serine threonine kinase receptor associated protein regulates early follicle development in the mouse ovary. Reproduction 2017; 153:221-231. [DOI: 10.1530/rep-16-0612] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 11/14/2016] [Accepted: 11/22/2016] [Indexed: 12/13/2022]
Abstract
The molecular mechanisms involved in regulating the development of small, gonadotrophin-independent follicles are poorly understood; however, many studies have highlighted an essential role for TGFB ligands. Canonical TGFB signalling is dependent upon intracellular SMAD proteins that regulate transcription. STRAP has been identified in other tissues as an inhibitor of the TGFB–SMAD signalling pathway. Therefore, in this study we aimed to determine the expression and role of STRAP in the context of early follicle development. Using qPCR, Strap, Smad3 and Smad7 revealed similar expression profiles in immature ovaries from mice aged 4–16 days containing different populations of early growing follicles. STRAP and SMAD2/3 proteins co-localised in granulosa cells of small follicles using immunofluorescence. Using an established culture model, neonatal mouse ovary fragments with a high density of small non-growing follicles were used to examine the effects of Strap knockdown using siRNA and STRAP protein inhibition by immuno-neutralisation. Both interventions caused a reduction in the proportion of small, non-growing follicles and an increase in the proportion and size of growing follicles in comparison to untreated controls, suggesting inhibition of STRAP facilitates follicle activation. Recombinant STRAP protein had no effect on small, non-growing follicles, but increased the mean oocyte size of growing follicles in the neonatal ovary model and also promoted the growth of isolated preantral follicles in vitro. Overall findings indicate STRAP is expressed in the mouse ovary and is capable of regulating development of small follicles in a stage-dependent manner.
Collapse
|
74
|
Green YS, Kwon S, Mimoto MS, Xie Y, Christian JL. Tril targets Smad7 for degradation to allow hematopoietic specification in Xenopus embryos. Development 2016; 143:4016-4026. [PMID: 27633996 DOI: 10.1242/dev.141812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/01/2016] [Indexed: 12/21/2022]
Abstract
In Xenopus laevis, bone morphogenetic proteins (Bmps) induce expression of the transcription factor Gata2 during gastrulation, and Gata2 is required in both ectodermal and mesodermal cells to enable mesoderm to commit to a hematopoietic fate. Here, we identify tril as a Gata2 target gene that is required in both ectoderm and mesoderm for primitive hematopoiesis to occur. Tril is a transmembrane protein that functions as a co-receptor for Toll-like receptors to mediate innate immune responses in the adult brain, but developmental roles for this molecule have not been identified. We show that Tril function is required both upstream and downstream of Bmp receptor-mediated Smad1 phosphorylation for induction of Bmp target genes. Mechanistically, Tril triggers degradation of the Bmp inhibitor Smad7. Tril-dependent downregulation of Smad7 relieves repression of endogenous Bmp signaling during gastrulation and this enables mesodermal progenitors to commit to a blood fate. Thus, Tril is a novel component of a Bmp-Gata2 positive-feedback loop that plays an essential role in hematopoietic specification.
Collapse
Affiliation(s)
- Yangsook Song Green
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah, School of Medicine, Salt Lake City, UT 84132, USA
| | - Sunjong Kwon
- Department of Cell and Developmental Biology, Oregon Health and Sciences University, School of Medicine, Portland, OR 97239-3098, USA
| | - Mizuho S Mimoto
- Department of Cell and Developmental Biology, Oregon Health and Sciences University, School of Medicine, Portland, OR 97239-3098, USA
| | - Yuanyuan Xie
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah, School of Medicine, Salt Lake City, UT 84132, USA
| | - Jan L Christian
- Department of Neurobiology and Anatomy and Internal Medicine, Division of Hematology and Hematologic Malignancies, University of Utah, School of Medicine, Salt Lake City, UT 84132, USA
| |
Collapse
|
75
|
Elevating CLIC4 in Multiple Cell Types Reveals a TGF- Dependent Induction of a Dominant Negative Smad7 Splice Variant. PLoS One 2016; 11:e0161410. [PMID: 27536941 PMCID: PMC4990216 DOI: 10.1371/journal.pone.0161410] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Accepted: 08/04/2016] [Indexed: 12/22/2022] Open
Abstract
CLIC4 (Chloride intracellular channel 4) belongs to a family of putative intracellular chloride channel proteins expressed ubiquitously in multiple tissues. CLIC4 is predominantly soluble and traffics between the cytoplasm and nucleus and participates in cell cycle control and differentiation. Transforming growth factor beta (TGF-β) elevates CLIC4, which enhances TGF-β signaling through CLIC4 mediated stabilization of phospho-Smad2/3. CLIC4 is essential for TGF-β induced conversion of fibroblasts to myofibroblasts and expression of matrix proteins, signaling via the p38MAPK pathway. Therefore, regulation of TGF-β signaling is a major mechanism by which CLIC4 modifies normal growth and differentiation. We now report that elevated CLIC4 alters Smad7 function, a feedback inhibitor of the TGF-β pathway. Overexpression of CLIC4 in keratinocytes, mouse embryonic fibroblasts and other mouse and human cell types increases the expression of Smad7Δ, a novel truncated form of Smad7. The alternatively spliced Smad7Δ variant is missing 94bp in exon 4 of Smad 7 and is conserved between mouse and human cells. The deletion is predicted to lack the TGF-β signaling inhibitory MH2 domain of Smad7. Treatment with exogenous TGF-β1 also enhances expression of Smad7Δ that is amplified in the presence of CLIC4. While Smad7 expression inhibits TGF-β signaling, exogenously expressed Smad7Δ does not inhibit TGF-β signaling as determined by TGF-β dependent proliferation, reporter assays and phosphorylation of Smad proteins. Instead, exogenous Smad7Δ acts as a dominant negative inhibitor of Smad7, thus increasing TGF-β signaling. This discovery adds another dimension to the myriad ways by which CLIC4 modifies TGF-β signaling.
Collapse
|
76
|
Jin L, Zhu C, Wang X, Li C, Cao C, Yuan J, Li S. Urocortin attenuates TGFβ1-induced Snail1 and slug expressions: inhibitory role of Smad7 in Smad2/3 signaling in breast cancer cells. J Cell Biochem 2016; 116:2494-503. [PMID: 26138318 DOI: 10.1002/jcb.25194] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 04/14/2015] [Indexed: 01/08/2023]
Abstract
Corticortropin-releasing hormone (CRH) family are multifunctional endocrine-factors that regulate proliferation, apoptosis, and migration of various types of cancer cells. Deregulation of the transforming growth factor β1(TGFβ1) signal transduction promotes aggressive metastatic properties in late-stage breast cancers. We previously have demonstrated in breast cancer cell line that CRH suppressed TGFβ1-induced Epithelial-Mesenchymal Transition (EMT) via induction of E-cadherin. Our present data in MCF-7 and MDA-MB-231 cells showed that Urocortin (Ucn, a member of CRH family) inhibited TGFβ1 signaling by reducing Smad2/3 activation and subsequent nuclear translocation through increasing Smad7 expression, leading to downregulation of Snail1 and Slug, the two EMT promoters. We further found that Antalarmin (CRH receptor type 1, CRHR1 antagonist) and Antisauvagine-30 (CRH receptor type2, CRHR2 antagonist) abrogated the effects of Ucn on TGFβ1 signaling, implying that both active CRHR1 and CRHR2 participate in Ucn-repressed TGFβ1 signaling. Our findings, for the fist time, identify Ucn as a potential mediator that inhibits oncogenic signaling by TGFβ1 and suggest that activating CRHR1 and R2 may prove effective in diminishing breast cancer progression stimulated by TGFβ1.
Collapse
Affiliation(s)
- Lai Jin
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Chao Zhu
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Xiaofei Wang
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Chuanhua Li
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Chunxuan Cao
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Jie Yuan
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| | - Shengnan Li
- Department of Pharmacology, Jiangsu Provincial key Lab of Cardiovascular Diseases and Molecular Intervention, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
77
|
Davey JR, Watt KI, Parker BL, Chaudhuri R, Ryall JG, Cunningham L, Qian H, Sartorelli V, Sandri M, Chamberlain J, James DE, Gregorevic P. Integrated expression analysis of muscle hypertrophy identifies Asb2 as a negative regulator of muscle mass. JCI Insight 2016; 1. [PMID: 27182554 DOI: 10.1172/jci.insight.85477] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The transforming growth factor-β (TGF-β) signaling network is a critical regulator of skeletal muscle mass and function and, thus, is an attractive therapeutic target for combating muscle disease, but the underlying mechanisms of action remain undetermined. We report that follistatin-based interventions (which modulate TGF-β network activity) can promote muscle hypertrophy that ameliorates aging-associated muscle wasting. However, the muscles of old sarcopenic mice demonstrate reduced response to follistatin compared with healthy young-adult musculature. Quantitative proteomic and transcriptomic analyses of young-adult muscles identified a transcription/translation signature elicited by follistatin exposure, which included repression of ankyrin repeat and SOCS box protein 2 (Asb2). Increasing expression of ASB2 reduced muscle mass, thereby demonstrating that Asb2 is a TGF-β network-responsive negative regulator of muscle mass. In contrast to young-adult muscles, sarcopenic muscles do not exhibit reduced ASB2 abundance with follistatin exposure. Moreover, preventing repression of ASB2 in young-adult muscles diminished follistatin-induced muscle hypertrophy. These findings provide insight into the program of transcription and translation events governing follistatin-mediated adaptation of skeletal muscle attributes and identify Asb2 as a regulator of muscle mass implicated in the potential mechanistic dysfunction between follistatin-mediated muscle growth in young and old muscles.
Collapse
Affiliation(s)
| | - Kevin I Watt
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Benjamin L Parker
- Charles Perkins Centre, School of Molecular Bioscience, The University of Sydney, Sydney, Australia
| | - Rima Chaudhuri
- Charles Perkins Centre, School of Molecular Bioscience, The University of Sydney, Sydney, Australia
| | - James G Ryall
- National Institute of Arthritis Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA; Department of Physiology, The University of Melbourne, Melbourne, Australia
| | | | - Hongwei Qian
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Vittorio Sartorelli
- National Institute of Arthritis Musculoskeletal and Skin Diseases (NIAMS), NIH, Bethesda, Maryland, USA
| | - Marco Sandri
- Venetian Institute of Molecular Medicine, The University of Padova, Padova, Italy
| | - Jeffrey Chamberlain
- Department of Neurology, The University of Washington, Seattle, Washington, USA
| | - David E James
- Charles Perkins Centre, School of Molecular Bioscience, The University of Sydney, Sydney, Australia; Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Paul Gregorevic
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Physiology, The University of Melbourne, Melbourne, Australia; Department of Neurology, The University of Washington, Seattle, Washington, USA; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Australia
| |
Collapse
|
78
|
TGF-β signaling is activated in patients with chronic HBV infection and repressed by SMAD7 overexpression after successful antiviral treatment. Inflamm Res 2016; 65:355-65. [PMID: 26856334 DOI: 10.1007/s00011-016-0921-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 11/07/2015] [Accepted: 01/27/2016] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVES Although animal studies demonstrated that Smad7 induction ameliorates TGF-β/SMAD-mediated fibrogenesis, its role in human hepatic diseases is rather obscure. Our study explored the activation status of TGF-β/activin pathway in patients with chronic liver diseases, and how it is affected by successful antiviral treatment in chronic HBV hepatitis (CHB). METHODS Thirty-seven CHB patients (19 with active disease, 14 completely remitted on long-term antiviral treatment and 4 with relapse after treatment withdrawal), 18 patients with chronic HCV hepatitis, 12 with non-alcoholic fatty liver disease (NAFLD), and 3 controls were enrolled in the study. Liver mRNA levels of CTGF, all TGF-β/activin isoforms, their receptors and intracellular mediators (SMADs) were evaluated using qRT-PCR and were correlated with the grade of liver inflammation and fibrosis staging. The expression and localization of pSMAD2 and pSMAD3 were assessed by immunohistochemistry. RESULTS TGF-β signalling is activated in CHB patients with active disease, while SMAD7 is up-regulated during the resolution of inflammation after successful treatment. SMAD7 overexpression was also observed in NAFLD patients exhibiting no or minimal fibrosis, despite the activation of TGF-β/activin signaling. CONCLUSIONS SMAD7 overexpression might represent a mechanism limiting TGF-β-mediated fibrogenesis in human hepatic diseases; therefore, SMAD7 induction likely represents a candidate for novel therapeutic approaches.
Collapse
|
79
|
Abstract
TGF-β signaling plays a key role in the temporal and spatial regulation of bone remodeling. During osteoclast bone resorption, TGF-β is released from the bone matrix and activated. Active TGF-β recruits mesenchymal stem cells to the bone resorption pit through the SMAD signaling pathway. Mesenchymal stem cells undergo osteoblast differentiation and deposit new bone filling in the resorption pit and maintaining the structural integrity of the skeleton. Thus, TGF-β signaling plays a key role in the coupling process and disruptions to the TGF-β signaling pathway lead to loss of skeletal integrity. This chapter describes methods on how to quantitate bone matrix TGF-β and assess its role in mesenchymal stem cell migration both in vitro and in vivo.
Collapse
Affiliation(s)
- Janet L Crane
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lingling Xian
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Ross Building, Room 231, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
| |
Collapse
|
80
|
Seet LF, Toh LZ, Finger SN, Chu SWL, Stefanovic B, Wong TT. Valproic acid suppresses collagen by selective regulation of Smads in conjunctival fibrosis. J Mol Med (Berl) 2015; 94:321-34. [PMID: 26507880 PMCID: PMC4803820 DOI: 10.1007/s00109-015-1358-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 09/28/2015] [Accepted: 10/20/2015] [Indexed: 04/17/2023]
Abstract
Overproduction of type I collagen is associated with a wide range of fibrotic diseases as well as surgical failure such as in glaucoma filtration surgery (GFS). Its modulation is therefore of clinical importance. Valproic acid (VPA) is known to reduce collagen in a variety of tissues with unclear mechanism of action. In this report, we demonstrate that VPA inhibited collagen production in both conjunctival fibroblasts and the mouse model of GFS. In fibroblasts, VPA decreased type I collagen expression which intensified with longer drug exposure and suppressed steady-state type I collagen promoter activity. Moreover, VPA decreased Smad2, Smad3 and Smad4 but increased Smad6 expression with a similar intensity-exposure profile. Reduction of Smad3 using small hairpin RNA and/or overexpression of Smad6 resulted in decreased collagen expression which was exacerbated when VPA was simultaneously present. Furthermore, fibrogenic TGF-β2 failed to induce collagen when VPA was present, as opposed to the myofibroblast markers, beta-actin, alpha-smooth muscle actin and tenascin-C, which were elevated by TGF-β2. VPA suppressed p3TP-Lux luciferase activity and selectively rescued Smad6 expression from suppression by TGF-β2. Notably, SMAD6 overexpression reduced the effectiveness of TGF-β2 in inducing collagen expression. In corroboration, VPA inhibited type I collagen but increased Smad6 expression in the late phase of wound healing in the mouse model of GFS. Taken together, our data indicate that VPA has the capacity to effectively suppress both steady-state and fibrogenic activation of type I collagen expression by modulating Smad expression. Hence, VPA is potentially applicable as an anti-fibrotic therapeutic by targeting collagen. Key message: • VPA modulates type I collagen expression via members of the Smad family. • VPA suppresses Smad2, Smad3 and Smad4 but upregulates Smad6. • Smad3 and Smad6 are involved in VPA regulation of steady-state collagen expression. • Smad6 is involved in VPA modulation of TGF-β-stimulated collagen expression. • VPA reduces collagen and upregulates Smad6 in the mouse model of glaucoma filtration surgery.
Collapse
Affiliation(s)
- Li-Fong Seet
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore, Singapore. .,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Duke-NUS Graduate Medical School Singapore, Singapore, Singapore.
| | - Li Zhen Toh
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore, Singapore
| | - Sharon N Finger
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore, Singapore
| | - Stephanie W L Chu
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore, Singapore
| | - Branko Stefanovic
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL, USA
| | - Tina T Wong
- Ocular Therapeutics and Drug Delivery, Singapore Eye Research Institute, Singapore, Singapore. .,Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,Duke-NUS Graduate Medical School Singapore, Singapore, Singapore. .,Glaucoma Service, Singapore National Eye Center, 11 Third Hospital Avenue, Singapore, 168751, Singapore. .,School of Materials Science and Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
81
|
Jurberg AD, Vasconcelos-Fontes L, Cotta-de-Almeida V. A Tale from TGF-β Superfamily for Thymus Ontogeny and Function. Front Immunol 2015; 6:442. [PMID: 26441956 PMCID: PMC4564722 DOI: 10.3389/fimmu.2015.00442] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 08/14/2015] [Indexed: 12/16/2022] Open
Abstract
Multiple signaling pathways control every aspect of cell behavior, organ formation, and tissue homeostasis throughout the lifespan of any individual. This review takes an ontogenetic view focused on the large superfamily of TGF-β/bone morphogenetic protein ligands to address thymus morphogenesis and function in T cell differentiation. Recent findings on a role of GDF11 for reversing aging-related phenotypes are also discussed.
Collapse
Affiliation(s)
- Arnon Dias Jurberg
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil ; Graduate Program in Cell and Developmental Biology, Institute of Biomedical Sciences, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Larissa Vasconcelos-Fontes
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| | - Vinícius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz) , Rio de Janeiro , Brazil
| |
Collapse
|
82
|
O'Beirne SL, Walsh SM, Fabre A, Reviriego C, Worrell JC, Counihan IP, Lumsden RV, Cramton-Barnes J, Belperio JA, Donnelly SC, Boylan D, Marchal-Sommé J, Kane R, Keane MP. CXCL9 Regulates TGF-β1-Induced Epithelial to Mesenchymal Transition in Human Alveolar Epithelial Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:2788-96. [PMID: 26268659 DOI: 10.4049/jimmunol.1402008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 07/12/2015] [Indexed: 01/22/2023]
Abstract
Epithelial to mesenchymal cell transition (EMT), whereby fully differentiated epithelial cells transition to a mesenchymal phenotype, has been implicated in the pathogenesis of idiopathic pulmonary fibrosis (IPF). CXCR3 and its ligands are recognized to play a protective role in pulmonary fibrosis. In this study, we investigated the presence and extent of EMT and CXCR3 expression in human IPF surgical lung biopsies and assessed whether CXCR3 and its ligand CXCL9 modulate EMT in alveolar epithelial cells. Coexpression of the epithelial marker thyroid transcription factor-1 and the mesenchymal marker α-smooth muscle actin and CXCR3 expression was examined by immunohistochemical staining of IPF surgical lung biopsies. Epithelial and mesenchymal marker expression was examined by quantitative real-time PCR, Western blotting, and immunofluorescence in human alveolar epithelial (A549) cells treated with TGF-β1 and CXCL9, with Smad2, Smad3, and Smad7 expression and cellular localization examined by Western blotting. We found that significantly more cells were undergoing EMT in fibrotic versus normal areas of lung in IPF surgical lung biopsy samples. CXCR3 was expressed by type II pneumocytes and fibroblasts in fibrotic areas in close proximity to cells undergoing EMT. In vitro, CXCL9 abrogated TGF-β1-induced EMT. A decrease in TGF-β1-induced phosphorylation of Smad2 and Smad3 occurred with CXCL9 treatment. This was associated with increased shuttling of Smad7 from the nucleus to the cytoplasm where it inhibits Smad phosphorylation. This suggests a role for EMT in the pathogenesis of IPF and provides a novel mechanism for the inhibitory effects of CXCL9 on TGF-β1-induced EMT.
Collapse
Affiliation(s)
- Sarah L O'Beirne
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Sinead M Walsh
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Aurélie Fabre
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland
| | - Carlota Reviriego
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Julie C Worrell
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Ian P Counihan
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Robert V Lumsden
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Jennifer Cramton-Barnes
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - John A Belperio
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095; and
| | - Seamas C Donnelly
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Denise Boylan
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Joëlle Marchal-Sommé
- INSERM Unité Mixte de Recherche 700, Physiopathologie et Epidémiologie de l'Insuffisance Respiratoire, Universite Denis Diderot, Paris 7, Unité de Formation et de Recherche de Médecine, 75018 Paris, France
| | - Rosemary Kane
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - Michael P Keane
- St. Vincent's University Hospital and School of Medicine and Medical Science, University College Dublin, Dublin 4, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin 4, Ireland;
| |
Collapse
|
83
|
Girerd B, Coulet F, Jaïs X, Eyries M, Van Der Bruggen C, De Man F, Houweling A, Dorfmüller P, Savale L, Sitbon O, Vonk-Noordegraaf A, Soubrier F, Simonneau G, Humbert M, Montani D. Characteristics of pulmonary arterial hypertension in affected carriers of a mutation located in the cytoplasmic tail of bone morphogenetic protein receptor type 2. Chest 2015; 147:1385-1394. [PMID: 25429696 DOI: 10.1378/chest.14-0880] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Mutations in BMPR2 encoding bone morphogenetic protein receptor type 2 (BMPRII) is the main genetic risk factor for heritable pulmonary arterial hypertension (PAH). The suspected mechanism is considered to be a defect of BMP signaling. The BMPRII receptor exists in a short isoform without a cytoplasmic tail, which has preserved BMP signaling. METHODS This cohort study compared age at PAH diagnosis and severity between patients carrying a BMPR2 mutation affecting the cytoplasmic tail of BMPRII and affected carriers of a mutation upstream of this domain. RESULTS We identified 171 carriers affected with PAH with a mutated BMPR2. Twenty-three were carriers of a point mutation located on the cytoplasmic tail of BMPRII. This population was characterized by having an older age at diagnosis compared with other BMPR2 mutation carriers (43.2 ± 12.1 years and 35.7 ± 14.6 years, P = .040), a lower pulmonary vascular resistance (13.3 ± 3.5 and 17.4 ± 6.7, P = .023), and a higher proportion of acute vasodilator responders with a long-term response to calcium channel blockers (8.7% and 0%, P = .02). No statistically significant differences were observed in survival. An in vitro assay showed that mutations located in the cytoplasmic tail led to normal activation of the Smad pathway, whereas activation was abolished in the presence of mutations located in the kinase domain. CONCLUSIONS Patients carrying a mutation affecting the cytoplasmic tail of BMPRII were characterized by an older age at diagnosis compared with other BMPR2 mutation carriers, less severe hemodynamic characteristics, and a greater chance of being a long-term responder to calcium channel blockers. Further investigations are needed to better understand the consequences of these BMPR2 mutations in BMPRII signaling pathways and their possible role in pulmonary arterial remodeling.
Collapse
Affiliation(s)
- Barbara Girerd
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Florence Coulet
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France
| | - Xavier Jaïs
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Mélanie Eyries
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France; ICAN Institute for Cardiometabolism and Nutrition, Paris, France; Unité Mixte de Recherche en Santé (UMR_S 1166), UPMC - Université Paris-Sorbonne, and INSERM, Paris, France
| | - Cathelijne Van Der Bruggen
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Frances De Man
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Arjan Houweling
- Department of Clinical Genetics, VU University Medical Center, Amsterdam, The Netherlands
| | - Peter Dorfmüller
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Laurent Savale
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Olivier Sitbon
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Anton Vonk-Noordegraaf
- Departments of Pulmonary Medicine, Institute for Cardiovascular Research, VU University Medical Center, Amsterdam, The Netherlands
| | - Florent Soubrier
- Genetics Department, Hôpital Pitié-Salpêtrière, AP-HP, Paris, France; ICAN Institute for Cardiometabolism and Nutrition, Paris, France; Unité Mixte de Recherche en Santé (UMR_S 1166), UPMC - Université Paris-Sorbonne, and INSERM, Paris, France
| | - Gérald Simonneau
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - Marc Humbert
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France
| | - David Montani
- University Paris-Sud, Le Kremlin-Bicêtre, France; Assistance Publique-Hôpitaux de Paris (AP-HP), Service de Pneumologie, Centre de Référence de l'Hypertension Pulmonaire Sévère, DHU Thorax Innovation, Hôpital Bicêtre, Le Kremlin-Bicêtre, France; Institut National de la Santé et de la Recherche Médicale (INSERM) U999, LabEx LERMIT, Centre Chirurgical Marie Lannelongue, Le Plessis Robinson, France.
| |
Collapse
|
84
|
Tsubakihara Y, Hikita A, Yamamoto S, Matsushita S, Matsushita N, Oshima Y, Miyazawa K, Imamura T. Arkadia enhances BMP signalling through ubiquitylation and degradation of Smad6. J Biochem 2015; 158:61-71. [PMID: 25762727 DOI: 10.1093/jb/mvv024] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 01/17/2015] [Indexed: 02/02/2023] Open
Abstract
Arkadia, a positive regulator of Smad-dependent signalling via the transforming growth factor-β (TGF-β) family, is an E3 ubiquitin ligase that induces ubiquitylation and proteasome-dependent degradation of TGF-β suppressors such as Smad7, c-Ski and SnoN. In this study, we examined the effects of Arkadia on bone morphogenetic protein (BMP)-induced osteoblast differentiation. Knockdown of Arkadia reduced mineralization and expression of osteoblast differentiation markers. Furthermore, we showed that Smad6, a BMP-specific inhibitory Smad, is a target of Arkadia: wild-type (WT) Arkadia, but not the C937A (CA) mutant lacking E3 ubiquitin-ligase activity, induced ubiquitylation and proteasome-dependent degradation of Smad6. Accordingly, protein levels of Smad6, Smad7 and c-Ski were elevated in MEFs from Arkadia KO mice. Finally, expression of Arkadia attenuated blockade of BMP signalling by Smad6 in a transcriptional reporter assay. These results demonstrate that Smad6 is a novel target of Arkadia, and that Arkadia positively regulates BMP signalling via degradation of Smad6, Smad7 and c-Ski/SnoN.
Collapse
Affiliation(s)
- Yutaro Tsubakihara
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Atsuhiko Hikita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shin Yamamoto
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Sachi Matsushita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Natsuki Matsushita
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yusuke Oshima
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-im
| | - Keiji Miyazawa
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-imaging, Proteo-Science Center, Ehime University, Shitsukawa, Toon, Ehime; Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Tokyo; Department of Gastroenterology and Metabiology, Ehime University, Shitsukawa, Toon, Ehime; Translational Research Center, Ehime University Hospital, Shitsukawa, Toon, Ehime; and Department of Biochemistry, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime; Division of Bio-im
| |
Collapse
|
85
|
Transforming growth factor Beta family: insight into the role of growth factors in regulation of fracture healing biology and potential clinical applications. Mediators Inflamm 2015; 2015:137823. [PMID: 25709154 PMCID: PMC4325469 DOI: 10.1155/2015/137823] [Citation(s) in RCA: 178] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Accepted: 11/09/2014] [Indexed: 01/15/2023] Open
Abstract
The transforming growth factor beta (TGF-β) family forms a group of three isoforms, TGF-β1, TGF-β2, and TGF-β3, with their structure formed by interrelated dimeric polypeptide chains. Pleiotropic and redundant functions of the TGF-β family concern control of numerous aspects and effects of cell functions, including proliferation, differentiation, and migration, in all tissues of the human body. Amongst many cytokines and growth factors, the TGF-β family is considered a group playing one of numerous key roles in control of physiological phenomena concerning maintenance of metabolic homeostasis in the bone tissue. By breaking the continuity of bone tissue, a spread-over-time and complex bone healing process is initiated, considered a recapitulation of embryonic intracartilaginous ossification. This process is a cascade of local and systemic phenomena spread over time, involving whole cell lineages and various cytokines and growth factors. Numerous in vivo and in vitro studies in various models analysing cytokines and growth factors' involvement have shown that TGF-β has a leading role in the fracture healing process. This paper sums up current knowledge on the basis of available literature concerning the role of the TGF-β family in the fracture healing process.
Collapse
|
86
|
Dzik JM. Evolutionary roots of arginase expression and regulation. Front Immunol 2014; 5:544. [PMID: 25426114 PMCID: PMC4224125 DOI: 10.3389/fimmu.2014.00544] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 10/13/2014] [Indexed: 12/11/2022] Open
Abstract
Two main types of macrophage functions are known: classical (M1), producing nitric oxide, NO, and M2, in which arginase activity is primarily expressed. Ornithine, the product of arginase, is a substrate for synthesis of polyamines and collagen, important for growth and ontogeny of animals. M2 macrophages, expressing high level of mitochondrial arginase, have been implicated in promoting cell division and deposition of collagen during ontogeny and wound repair. Arginase expression is the default mode of tissue macrophages, but can also be amplified by signals, such as IL-4/13 or transforming growth factor-β (TGF-β) that accelerates wound healing and tissue repair. In worms, the induction of collagen gene is coupled with induction of immune response genes, both depending on the same TGF-β-like pathway. This suggests that the main function of M2 “heal” type macrophages is originally connected with the TGF-β superfamily of proteins, which are involved in regulation of tissue and organ differentiation in embryogenesis. Excretory–secretory products of metazoan parasites are able to induce M2-type of macrophage responses promoting wound healing without participation of Th2 cytokines IL-4/IL-13. The expression of arginase in lower animals can be induced by the presence of parasite antigens and TGF-β signals leading to collagen synthesis. This also means that the main proteins, which, in primitive metazoans, are involved in regulation of tissue and organ differentiation in embryogenesis are produced by innate immunity. The signaling function of NO is known already from the sponge stage of animal evolution. The cytotoxic role of NO molecule appeared later, as documented in immunity of marine mollusks and some insects. This implies that the M2-wound healing promoting function predates the defensive role of NO, a characteristic of M1 macrophages. Understanding when and how the M1 and M2 activities came to be in animals is useful for understanding how macrophage immunity, and immune responses operate.
Collapse
Affiliation(s)
- Jolanta Maria Dzik
- Department of Biochemistry, Faculty of Agriculture and Biology, Warsaw University of Life Sciences - SGGW , Warszawa , Poland
| |
Collapse
|
87
|
Kong Q, Li WJ, Huang HR, Zhong YQ, Fang JP. Differential gene expression profiles of peripheral blood mononuclear cells in childhood asthma. J Asthma 2014; 52:343-52. [PMID: 25329679 DOI: 10.3109/02770903.2014.971967] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Asthma is a common childhood disease with strong genetic components. This study compared whole-genome expression differences between asthmatic young children and healthy controls to identify gene signatures of childhood asthma. METHODS Total RNA extracted from peripheral blood mononuclear cells (PBMC) was subjected to microarray analysis. QRT-PCR was performed to verify the microarray results. Classification and functional characterization of differential genes were illustrated by hierarchical clustering and gene ontology analysis. Multiple logistic regression (MLR) analysis, receiver operating characteristic (ROC) curve analysis, and discriminate power were used to scan asthma-specific diagnostic markers. RESULTS For fold-change>2 and p < 0.05, there were 758 named differential genes. The results of QRT-PCR confirmed successfully the array data. Hierarchical clustering divided 29 highly possible genes into seven categories and the genes in the same cluster were likely to possess similar expression patterns or functions. Gene ontology analysis presented that differential genes primarily enriched in immune response, response to stress or stimulus, and regulation of apoptosis in biological process. MLR and ROC curve analysis revealed that the combination of ADAM33, Smad7, and LIGHT possessed excellent discriminating power. CONCLUSIONS The combination of ADAM33, Smad7, and LIGHT would be a reliable and useful childhood asthma model for prediction and diagnosis.
Collapse
Affiliation(s)
- Qian Kong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University , Guangzhou, Guangdong , P.R. China
| | | | | | | | | |
Collapse
|
88
|
Chen X, Shi W, Wang F, Du Z, Yang Y, Gao M, Yao Y, He K, Wang C, Hao A. Zinc Finger DHHC-Type Containing 13 Regulates Fate Specification of Ectoderm and Mesoderm Cell Lineages by Modulating Smad6 Activity. Stem Cells Dev 2014; 23:1899-909. [DOI: 10.1089/scd.2014.0068] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Xueran Chen
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Wei Shi
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Fen Wang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Zhaoxia Du
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Yang Yang
- Infertility Center, Qilu Hospital, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Ming Gao
- Reproductive Medical Center of Shandong University, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Yao Yao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Kun He
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Chen Wang
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| | - Aijun Hao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Histology and Embryology, Shandong University School of Medicine, Jinan, Shandong, People's Republic of China
| |
Collapse
|
89
|
Simultaneous imaging and restoration of cell function using cell permeable peptide probe. Biomaterials 2014; 35:6287-98. [DOI: 10.1016/j.biomaterials.2014.04.045] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 04/14/2014] [Indexed: 11/20/2022]
|
90
|
Luo L, Li N, Lv N, Huang D. SMAD7: a timer of tumor progression targeting TGF-β signaling. Tumour Biol 2014; 35:8379-85. [PMID: 24935472 DOI: 10.1007/s13277-014-2203-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Accepted: 06/06/2014] [Indexed: 01/02/2023] Open
Abstract
In the context of cancer, transforming growth factor β (TGF-β) is a cell growth suppressor; however, it is also a critical inducer of invasion and metastasis. SMAD is the important mediator of TGF-β signaling pathway, which includes receptor-regulated SMADs (R-SMADs), common-mediator SMADs (co-SMADs), and inhibitory SMADs (I-SMADs). I-SMADs block the activation of R-SMADs and co-SMADs and thus play important roles especially in the SMAD-dependent signaling. SMAD7 belongs to the I-SMADs. As an inhibitor of TGF-β signaling, SMAD7 is overexpressed in numerous cancer types and its abundance is positively correlated to the malignancy. Emerging evidence has revealed the switch-in-role of SMAD7 in cancer, from a TGF-β inhibiting protein at the early stages that facilitates proliferation to an enhancer of invasion at the late stages. This role change may be accompanied or elicited by the tumor microenvironment and/or somatic mutation. Hence, current knowledge suggests a tumor-favorable timer nature of SMAD7 in cancer progression. In this review, we summarized the advances and recent findings of SMAD7 and TGF-β signaling in cancer, followed by specific discussion on the possible factors that account for the functional changes of SMAD7.
Collapse
Affiliation(s)
- Lingyu Luo
- Research Institute of Digestive Diseases, The First Affiliated Hospital of Nanchang University, 17th Yongwaizen St., Nanchang, Jiangxi, 330006, People's Republic of China
| | | | | | | |
Collapse
|
91
|
Ramamoorthi G, Sivalingam N. Molecular mechanism of TGF-β signaling pathway in colon carcinogenesis and status of curcumin as chemopreventive strategy. Tumour Biol 2014; 35:7295-305. [PMID: 24668546 DOI: 10.1007/s13277-014-1840-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/11/2014] [Indexed: 02/06/2023] Open
Abstract
Colon cancer is one of the third most common cancer in man, the second most common cancer in women worldwide, and the second leading cause of mortality in the USA. There are a number of molecular pathways that have been implicated in colon carcinogenesis, including TGF-β/Smad signaling pathway. TGF-β (transforming growth factor-beta) signaling pathway has the potential to regulate various biological processes including cell growth, differentiation, apoptosis, extracellular matrix modeling, and immune response. TGF-β signaling pathway acts as a tumor suppressor, but alterations in TGF-β signaling pathway promotes colon cancer cell growth, migration, invasion, angiogenesis, and metastasis. Here we review the role of TGF-β signaling cascade in colon carcinogenesis and multiple molecular targets of curcumin in colon carcinogenesis. Elucidation of the molecular mechanism of curcumin on TGF-β signaling pathway-induced colon carcinogenesis may ultimately lead to novel and more effective treatments for colon cancer.
Collapse
Affiliation(s)
- Ganesan Ramamoorthi
- Department of Biotechnology, School of Bioengineering, SRM University, Kattankulathur, 603203, Tamilnadu, India,
| | | |
Collapse
|
92
|
Imamura T, Oshima Y, Hikita A. Regulation of TGF-β family signalling by ubiquitination and deubiquitination. J Biochem 2013; 154:481-9. [PMID: 24165200 DOI: 10.1093/jb/mvt097] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Members of the transforming growth factor-β (TGF-β) family, including TGF-βs, activin and bone morphogenetic proteins (BMPs), are multifunctional proteins that regulate a wide variety of cellular responses, such as proliferation, differentiation, migration and apoptosis. TGF-β family signalling is mainly mediated by membranous serine/threonine kinase receptors and intracellular Smad proteins. This signalling is tightly regulated by various post-translational modifications including ubiquitination. Several E3 ubiquitin ligases play a crucial role in the recognition and ubiquitin-dependent degradation of TGF-β family receptors, Smad proteins and their interacted proteins to regulate positively and negatively TGF-β family signalling. In contrast, non-degradative ubiquitin modifications also regulate TGF-β family signalling. Recently, in addition to protein ubiquitination, deubiquitination by deubiquitinating enzymes has been reported to control TGF-β family signalling pathways. Interestingly, more recent studies suggest that TGF-β signalling is not only regulated via ubiquitination and/or deubiquitination, but also it relies on ubiquitination for its effect on other pathways. Thus, ubiquitin modifications play key roles in TGF-β family signal transduction and cross-talk between TGF-β family signalling and other signalling pathways. Here, we review the current understandings of the positive and negative regulatory mechanisms by ubiquitin modifications that control TGF-β family signalling.
Collapse
Affiliation(s)
- Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine; Division of Bio-imaging, Proteo-Science Center, Ehime University; Translational Research Center, Ehime University Hospital; and Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency (JST), Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | |
Collapse
|
93
|
Estrada KD, Wang W, Retting KN, Chien CT, Elkhoury FF, Heuchel R, Lyons KM. Smad7 regulates terminal maturation of chondrocytes in the growth plate. Dev Biol 2013; 382:375-84. [PMID: 23994637 DOI: 10.1016/j.ydbio.2013.08.021] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 08/20/2013] [Accepted: 08/21/2013] [Indexed: 11/16/2022]
Abstract
Members of the bone morphogenetic protein (BMP) superfamily, including transforming growth factor-betas (TGFβ), regulate multiple aspects of chondrogenesis. Smad7 is an intracellular inhibitor of BMP and TGFβ signaling. Studies in which Smad7 was overexpressed in chondrocytes demonstrated that Smad7 can impact chondrogenesis by inhibiting BMP signaling. However, whether Smad7 is actually required for endochondral ossification in vivo is unclear. Moreover, whether Smad7 regulates TGFβ in addition to BMP signaling in developing cartilage is unknown. In this study, we found that Smad7 is required for both axial and appendicular skeletal development. Loss of Smad7 led to impairment of the cell cycle in chondrocytes and to defects in terminal maturation. This phenotype was attributed to upregulation of both BMP and TGFβ signaling in Smad7 mutant growth plates. Moreover, Smad7-/- mice develop hypocellular cores in the medial growth plates, associated with elevated HIF1α levels, cell death, and intracellular retention of types II and X collagen. Thus, Smad7 may be required to mediate cell stress responses in the growth plate during development.
Collapse
Affiliation(s)
- Kristine D Estrada
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, David Geffen School of Medicine at the University of California, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | |
Collapse
|
94
|
Xu J, Wang AH, Oses-Prieto J, Makhijani K, Katsuno Y, Pei M, Yan L, Zheng YG, Burlingame A, Brückner K, Derynck R. Arginine Methylation Initiates BMP-Induced Smad Signaling. Mol Cell 2013; 51:5-19. [PMID: 23747011 PMCID: PMC3951972 DOI: 10.1016/j.molcel.2013.05.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2012] [Revised: 10/12/2012] [Accepted: 05/02/2013] [Indexed: 11/15/2022]
Abstract
Kinase activation and substrate phosphorylation commonly form the backbone of signaling cascades. Bone morphogenetic proteins (BMPs), a subclass of TGF-β family ligands, induce activation of their signaling effectors, the Smads, through C-terminal phosphorylation by transmembrane receptor kinases. However, the slow kinetics of Smad activation in response to BMP suggests a preceding step in the initiation of BMP signaling. We now show that arginine methylation, which is known to regulate gene expression, yet also modifies some signaling mediators, initiates BMP-induced Smad signaling. BMP-induced receptor complex formation promotes interaction of the methyltransferase PRMT1 with the inhibitory Smad6, resulting in Smad6 methylation and relocalization at the receptor, leading to activation of effector Smads through phosphorylation. PRMT1 is required for BMP-induced biological responses across species, as evidenced by the role of its ortholog Dart1 in BMP signaling during Drosophila wing development. Activation of signaling by arginine methylation may also apply to other signaling pathways.
Collapse
Affiliation(s)
- Jian Xu
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - A. Hongjun Wang
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Juan Oses-Prieto
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Kalpana Makhijani
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Yoko Katsuno
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Ming Pei
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Leilei Yan
- Department of Chemistry, Georgia State University, Atlanta, GA30302-4098
| | - Y. George Zheng
- Department of Chemistry, Georgia State University, Atlanta, GA30302-4098
| | - Alma Burlingame
- Department of Pharmaceutical Chemistry, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Katja Brückner
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| | - Rik Derynck
- Departments of Cell and Tissue Biology, and Anatomy, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California at San Francisco, San Francisco, California 94143-0512. USA
| |
Collapse
|
95
|
Zimmer J, Degenkolbe E, Wildemann B, Seemann P. BMP Signaling in Regenerative Medicine. Bioinformatics 2013. [DOI: 10.4018/978-1-4666-3604-0.ch064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
More than 40 years after the discovery of Bone Morphogenetic Proteins (BMPs) as bone inducers, a whole protein family of growth factors connected to a wide variety of functions in embryonic development, homeostasis, and regeneration has been characterized. Today, BMP2 and BMP7 are already used in the clinic to promote vertebral fusions and restoration of non-union fractures. Besides describing present clinical applications, the authors review ongoing trials highlighting the future possibilities of BMPs in medicine. Apparently, the physiological roles of BMPs have expanded their range from bone growth induction and connective tissue regeneration to cancer diagnosis/treatment and cardiovascular disease prevention.
Collapse
Affiliation(s)
- Julia Zimmer
- Charité-Universitätsmedizin Berlin, Berlin-Brandenburg Center for Regenerative Therapies, Germany
| | - Elisa Degenkolbe
- Charité-Universitätsmedizin Berlin, Berlin-Brandenburg Center for Regenerative Therapies, Germany
| | - Britt Wildemann
- Charité-Universitätsmedizin Berlin, Berlin-Brandenburg Center for Regenerative Therapies, Germany
| | - Petra Seemann
- Charité-Universitätsmedizin Berlin, Berlin-Brandenburg Center for Regenerative Therapies, Germany
| |
Collapse
|
96
|
Abstract
Signalling of the transforming growth factor-β (TGF-β) family is tightly regulated by various mechanisms including negative feedback by Inhibitory Sma- and Mad-related proteins (Smads) (I-Smads: Smad6 and Smad7). Smad6 preferentially inhibits bone morphogenetic protein (BMP) signalling, whereas Smad7 suppresses both TGF-β and BMP signalling. To elucidate the roles of Smad7 in murine development and in TGF-β signalling, several Smad7-deficient mouse strains have been generated. Tojo et al. (Smad7-deficient mice show growth retardation with reduced viability. J. Biochem. 2012;151:621-631.) demonstrated that Smad7 null mutation caused perinatal lethality on a C57BL/6 background. However, the Smad7-deficient mice on an ICR background survived to adulthood, but showing growth retardation. Unexpectedly, phosphorylation levels of Smad2 and Smad3 were slightly reduced in murine embryonic fibroblast (MEF) cells isolated from Smad7-deficient embryos compared with wild-type MEF cells. Together with other Smad7-mutant mouse strains, these mutant mice provide useful tools to understand important roles of Smad7 in the development of murine embryos and diseases.
Collapse
|
97
|
Li Y, Wang M, Carra C, Cucinotta FA. Modularized Smad-regulated TGFβ signaling pathway. Math Biosci 2012; 240:187-200. [DOI: 10.1016/j.mbs.2012.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2011] [Revised: 07/19/2012] [Accepted: 07/20/2012] [Indexed: 01/05/2023]
|
98
|
Mechanical stretch down-regulates expression of the Smad6 gene in cultured rat mesangial cells. Clin Exp Nephrol 2012; 16:690-6. [DOI: 10.1007/s10157-012-0630-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2011] [Accepted: 03/15/2012] [Indexed: 02/04/2023]
|
99
|
Kim H, Kim DH, Park SB, Ko YG, Kim SW, Do YJ, Park JH, Yang BS. Induction of Ski Protein Expression upon Luteinization in Rat Granulosa Cells. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2012; 25:635-41. [PMID: 25049607 PMCID: PMC4093106 DOI: 10.5713/ajas.2011.11336] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 02/02/2012] [Accepted: 11/23/2011] [Indexed: 12/14/2022]
Abstract
Ski protein is implicated in proliferation/differentiation in a variety of cells. We had previously reported that Ski protein is present in granulosa cells of atretic follicles, but not in preovulatory follicles, suggesting that Ski has a role in apoptosis of granulosa cells. The alternative fate of granulosa cells other than apoptosis is to differentiate to luteal cells; however, it is unknown whether Ski is expressed and has a role in granulosa cells undergoing luteinization. Thus, the aim of the present study was to locate Ski protein in the rat ovary during luteinizationto predict the possible role of Ski. In order to examine the expression pattern of Ski protein along with the progress of luteinization, follicular growth was induced by administration of equine chorionic gonadtropin to immature female rats, and luteinization was induced by human chorionic gonadtropin treatment to mimic luteinizing hormone (LH) surge. While no Ski-positive granulosa cells were present in preovulatory follicle, Ski protein expression was induced in response to LH surge, and was maintained after the formation of the corpus luteum (CL). Though Ski protein is absent in granulosa cells of preovulatory follicle, its mRNA (c-Ski) was expressed and the level was unchanged even after LH surge. Taken together, these results demonstrated that Ski protein expression is induced in granulosa cells upon luteinization, and suggests that its expression is regulated post-transcriptionally.
Collapse
Affiliation(s)
- Hyun Kim
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-8657, Japan ; Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Dong Hun Kim
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Soo Bong Park
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Yeoung-Gyu Ko
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Sung-Woo Kim
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Yoon Jun Do
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Jae-Hong Park
- Animal Genetic Resources Station, National Institute of Animal Science, RDA, Namwon 590-832, Korea
| | - Boh-Suk Yang
- Korean Native Cattle Experiment Station, National Institute of Animal Science, RDA, Pyeongchang 232-952, Korea
| |
Collapse
|
100
|
Tojo M, Takebe A, Takahashi S, Tanaka K, Imamura T, Miyazono K, Chiba T. Smad7-deficient mice show growth retardation with reduced viability. J Biochem 2012; 151:621-31. [PMID: 22383537 DOI: 10.1093/jb/mvs022] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Smad7 is an inhibitory molecule induced by members of the transforming growth factor-β (TGF-β) family, including TGF-β, activin, nodal and bone morphogenetic proteins (BMPs). To elucidate the in vivo functions of Smad7, we generated conditional Smad7-knockout mice in which the Mad homology 2 (MH2) domain and the poly (A) signal sequence were flanked with loxP sites (floxed). The Smad7-floxed mice exhibited no obvious phenotype. Smad7 total-null mice on a C57BL/6 background died within a few days of birth, whereas mice with an ICR background developed to adulthood but were significantly smaller than wild-type mice. Unexpectedly, phospho-Smad2 and phospho-Smad3 were decreased in Smad7-deficient mouse embryonic fibroblast (MEF) cells, whereas phospho-Smad1/5/8 was similarly expressed in wild-type and Smad7-deficient MEF cells. Moreover, expression levels of TGF-β type I receptor (ALK5) were higher in Smad7-deficient MEF cells than in wild-type MEF cells. Plasminogen activator inhibitor-1 (PAI-1) and inhibitor of differentiation-1 (Id-1) mRNA were similarly expressed in wild-type and Smad7-deficient MEF cells. Some differences were observed in mitogen-activated protein kinase (MAPK)-signalling between wild-type and Smad7-deficient MEF cells. We demonstrated that Smad7 plays an important role in normal mouse growth and provide a useful tool for analysing Smad7 functions in vivo.
Collapse
Affiliation(s)
- Masayoshi Tojo
- Department of Molecular Pathology, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|