1
|
Molina MA, Biswas S, Jiménez-Vázquez O, Bodily JM. Regulation of epithelial growth factor receptors by the oncoprotein E5 during the HPV16 differentiation-dependent life cycle. Tumour Virus Res 2025; 19:200315. [PMID: 40057277 PMCID: PMC11928765 DOI: 10.1016/j.tvr.2025.200315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 03/06/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025] Open
Abstract
Human papillomavirus (HPV) 16 infection initiates upon viral entry into the basal cells of the epithelium. The virus manipulates signaling pathways to complete its life cycle, which depends on cellular differentiation. The virus expresses the oncoproteins E5, E6, and E7 to promote immune evasion, cell cycle progression, apoptosis inhibition, and viral replication. The least studied viral oncoprotein is E5 (16E5), which can regulate epithelial growth factor receptor (GFR) signaling pathways. GFRs such as transforming growth factor-beta receptor (TGFBR), epidermal growth factor receptor (EGFR), and keratinocyte growth factor receptor (KGFR) have essential roles in cell growth, differentiation, and proliferation. These receptors obtain their ligands from the microenvironment, and once activated, regulate cellular behavior in the epithelium. GFRs therefore represent valuable targets for the virus to establish and maintain a cellular environment supportive of infection. The ability of 16E5 to regulate proliferation and differentiation varies through the differentiating epithelium, making it necessary to adequately describe the association between 16E5 and GFRs. Here we summarize the regulation of GFR signaling pathways by 16E5, discuss the roles of stromal growth factors, and outline unresolved questions over cellular differentiation and proliferation during the HPV life cycle.
Collapse
Affiliation(s)
- Mariano A Molina
- Department of Pathology, Vrije Universiteit Amsterdam, Amsterdam UMC Location VUmc, Amsterdam, the Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, the Netherlands; Instituto de Ciencias Médicas, Las Tablas, Panama.
| | | | | | - Jason M Bodily
- Department of Microbiology and Immunology and Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
2
|
Machahua C, Marti TM, Dorn P, Funke-Chambour M. Fibrosis in PCLS: comparing TGF-β and fibrotic cocktail. Respir Res 2025; 26:44. [PMID: 39875887 PMCID: PMC11776118 DOI: 10.1186/s12931-025-03110-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025] Open
Abstract
INTRODUCTION Fibrotic cocktail (FC) is a combination of pro-fibrotic and pro-inflammatory mediators that induces early fibrotic changes in organotypic lung models. We hypothesised that transforming growth factor beta 1 (TGF-β1) alone induces a pro-fibrotic effect similar to FC. Our aim was to compare the pro-fibrotic effects of TGF-β1 with FC in human precision-cut lung slices (PCLS). METHODS PCLS from "healthy" lung tissue of cancer patients undergoing surgery (n = 7) were incubated with TGF-β1, FC or control for 72 h. Gene expression markers for myofibroblasts differentiation, extracellular matrix (ECM), as well as TGF-β receptors were assessed (RT-qPCR). ECM proteins expression in lysates and supernatant was assessed by ELISA and immunofluorescence. RESULTS We found that TGF-β1 significantly increased gene expression of ACTA2, COL1A1, CCN2, and VIM compared to control but also compared to FC. FC showed a significant increase of matrix metalloproteinase (MMP) 7 and 1 compared to control, while TGF-β receptor 2 was lower after FC compared to TGF-β1 or control. FC or TGF-β1 showed similar fibronectin protein expression in lysates and supernatants, while type I collagen protein expression in lysates was significantly greater with TGF-β1 compared to control. CONCLUSIONS Our findings show that TGF-β1 induces consistent pro-fibrotic changes in PCLS after 72 h. Compared to TGF-β1, FC treatment resulted in reduced gene expression of TGF-β receptor 2 and increased MMPs expression, potentially mitigating the early pro-fibrotic effects. Selecting specific pro-fibrotic stimuli may be preferable depending on the research question and time point of interest in lung fibrosis studies using PCLS.
Collapse
Affiliation(s)
- Carlos Machahua
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Thomas M Marti
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBRM), University of Bern, Bern, Switzerland
| | - Patrick Dorn
- Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Bern, Switzerland
- Department for BioMedical Research (DBRM), University of Bern, Bern, Switzerland
| | - Manuela Funke-Chambour
- Department for Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| |
Collapse
|
3
|
Dogru S, Alba GM, Pierce KC, Wang T, Kia DS, Albro MB. Cell mediated reactions create TGF-β delivery limitations in engineered cartilage. Acta Biomater 2024; 190:178-190. [PMID: 39447669 PMCID: PMC11614674 DOI: 10.1016/j.actbio.2024.10.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/26/2024]
Abstract
During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. These regulatory paradigms appear to be at odds with TGF-β delivery needs in tissue engineering (TE) where administered TGF-β is required to transport long distances or reside in tissues for extended durations. In this study, we perform a novel examination of the influence of cell-mediated reactions on the spatiotemporal distribution of administered TGF-β in cartilage TE applications. Reaction rates of TGF-β binding to cell-deposited ECM and TGF-β internalization by cell receptors are experimentally characterized in bovine chondrocyte-seeded tissue constructs. TGF-β binding to the construct ECM exhibits non-linear Brunauer-Emmett-Teller (BET) adsorption behavior, indicating that as many as seven TGF-β molecules can aggregate at a binding site. Cell-mediated TGF-β internalization rates exhibit a biphasic trend, following a Michaelis-Menten relation (Vmax = 2.4 molecules cell-1 s-1, Km = 1.7 ng mL-1) at low ligand doses (≤130 ng/mL), but exhibit an unanticipated non-saturating power trend at higher doses (≥130 ng/mL). Computational models are developed to illustrate the influence of these reactions on TGF-β spatiotemporal delivery profiles for conventional TGF-β administration platforms. For TGF-β delivery via supplementation in culture medium, these reactions give rise to pronounced steady state TGF-β spatial gradients; TGF-β concentration decays by ∼90 % at a depth of only 500 μm from the media-exposed surface. For TGF-β delivery via heparin-conjugated affinity scaffolds, cell mediated internalization reactions significantly reduce the TGF-β scaffold retention time (160-360-fold reduction) relative to acellular heparin scaffolds. This work establishes the significant limitations that cell-mediated chemical reactions engender for TGF-β delivery and highlights the need for novel delivery platforms that account for these reactions to achieve optimal TGF-β exposure profiles. STATEMENT OF SIGNIFICANCE: During native cartilage development, endogenous TGF-β activity is tightly regulated by cell-mediated chemical reactions in the extracellular milieu (e.g., matrix and receptor binding), providing spatiotemporal control in a manner that is localized and short acting. However, the effect of these reactions on the delivery of exogenous TGF-β to engineered cartilage tissues remains not well understood. In this study, we demonstrate that cell-mediated reactions significantly restrict the delivery of TGF-β to cells in engineered cartilage tissue constructs. For delivery via media supplementation, reactions significantly limit TGF-β penetration into constructs. For delivery via scaffold loading, reactions significantly limit TGF-β residence time in constructs. Overall, these results illustrate the impact of cell-mediated chemical reactions on TGF-β delivery profiles and support the importance of accounting for these reactions when designing TGF-β delivery platforms for promoting cartilage regeneration.
Collapse
Affiliation(s)
- Sedat Dogru
- College of Engineering, Boston University, Boston, MA, United States
| | - Gabriela M Alba
- College of Engineering, Boston University, Boston, MA, United States
| | - Kirk C Pierce
- College of Engineering, Boston University, Boston, MA, United States
| | - Tianbai Wang
- College of Engineering, Boston University, Boston, MA, United States
| | | | - Michael B Albro
- College of Engineering, Boston University, Boston, MA, United States.
| |
Collapse
|
4
|
Tang L, Qiu H, Xu B, Su Y, Nyarige V, Li P, Chen H, Killham B, Liao J, Adam H, Yang A, Yu A, Jang M, Rubart M, Xie J, Zhu W. Microparticle Mediated Delivery of Apelin Improves Heart Function in Post Myocardial Infarction Mice. Circ Res 2024; 135:777-798. [PMID: 39145385 PMCID: PMC11392624 DOI: 10.1161/circresaha.124.324608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 08/16/2024]
Abstract
BACKGROUND Apelin is an endogenous prepropeptide that regulates cardiac homeostasis and various physiological processes. Intravenous injection has been shown to improve cardiac contractility in patients with heart failure. However, its short half-life prevents studying its impact on left ventricular remodeling in the long term. Here, we aim to study whether microparticle-mediated slow release of apelin improves heart function and left ventricular remodeling in mice with myocardial infarction (MI). METHODS A cardiac patch was fabricated by embedding apelin-containing microparticles in a fibrin gel scaffold. MI was induced via permanent ligation of the left anterior descending coronary artery in adult C57BL/6J mice followed by epicardial patch placement immediately after (acute MI) or 28 days (chronic MI) post-MI. Four groups were included in this study, namely sham, MI, MI plus empty microparticle-embedded patch treatment, and MI plus apelin-containing microparticle-embedded patch treatment. Cardiac function was assessed by transthoracic echocardiography. Cardiomyocyte morphology, apoptosis, and cardiac fibrosis were evaluated by histology. Cardioprotective pathways were determined by RNA sequencing, quantitative polymerase chain reaction, and Western blot. RESULTS The level of endogenous apelin was largely reduced in the first 7 days after MI induction and it was normalized by day 28. Apelin-13 encapsulated in poly(lactic-co-glycolic acid) microparticles displayed a sustained release pattern for up to 28 days. Treatment with apelin-containing microparticle-embedded patch inhibited cardiac hypertrophy and reduced scar size in both acute and chronic MI models, which is associated with improved cardiac function. Data from cellular and molecular analyses showed that apelin inhibits the activation and proliferation of cardiac fibroblasts by preventing transforming growth factor-β-mediated activation of Smad2/3 (supporessor of mothers against decapentaplegic 2/3) and downstream profibrotic gene expression. CONCLUSIONS Poly(lactic-co-glycolic acid) microparticles prolonged the apelin release time in the mouse hearts. Epicardial delivery of the apelin-containing microparticle-embedded patch protects mice from both acute and chronic MI-induced cardiac dysfunction, inhibits cardiac fibrosis, and improves left ventricular remodeling.
Collapse
Affiliation(s)
- Ling Tang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Huiliang Qiu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Bing Xu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Yajuan Su
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Verah Nyarige
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Pengsheng Li
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Houjia Chen
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Brady Killham
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington (H.C., B.K., J.L.)
| | - Henderson Adam
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Aaron Yang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Alexander Yu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michelle Jang
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| | - Michael Rubart
- Department of Pediatrics, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis (M.R.)
| | - Jingwei Xie
- Department of Surgery-Transplant and Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha (Y.S., J.X.)
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic Arizona, Scottsdale (L.T., H.Q., B.X., V.N., P.L., H.A., A. Yang, A. Yu, M.J., W.Z.)
| |
Collapse
|
5
|
Hapeman JD, Galwa R, Carneiro CS, Nedelcu AM. In vitro evidence for the potential of EGFR inhibitors to decrease the TGF-β1-induced dispersal of circulating tumour cell clusters mediated by EGFR overexpression. Sci Rep 2024; 14:19980. [PMID: 39198539 PMCID: PMC11358385 DOI: 10.1038/s41598-024-70358-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
Most cancer-related deaths are due to the spread of tumour cells throughout the body-a process known as metastasis. While in the vasculature, these cells are referred to as circulating tumour cells (CTCs) and can be found as either single cells or clusters of cells (often including platelets), with the latter having the highest metastatic potential. However, the biology of CTC clusters is poorly understood, and there are no therapies that specifically target them. We previously developed an in vitro model system for CTC clusters and proposed a new extravasation model that involves cluster dissociation, adherence, and single-cell invasion in response to TGF-β1 released by platelets. Here, we investigated TGF-β1-induced gene expression changes in this model, focusing on genes for which targeted drugs are available. In addition to the upregulation of the TGF-β1 signalling pathway, we found that (i) genes in the EGF/EGFR pathway, including those coding for EGFR and several EGFR ligands, were also induced, and (ii) Erlotinib and Osimertinib, two therapeutic EGFR/tyrosine kinase inhibitors, decreased the TGF-β1-induced adherence and invasion of the CTC cluster-like line despite the line expressing wild-type EGFR. Overall, we suggest that EGFR inhibitors have the potential to decrease the dispersal of CTC clusters that respond to TGF-β1 and overexpress EGFR (irrespective of its status) and thus could improve patient survival.
Collapse
Affiliation(s)
- Jorian D Hapeman
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Rakshit Galwa
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Caroline S Carneiro
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada
| | - Aurora M Nedelcu
- Department of Biology, University of New Brunswick, Fredericton, NB, E3B 5A3, Canada.
| |
Collapse
|
6
|
Lewko B, Wodzińska M, Daca A, Płoska A, Obremska K, Kalinowski L. Urolithin A Ameliorates the TGF Beta-Dependent Impairment of Podocytes Exposed to High Glucose. J Pers Med 2024; 14:914. [PMID: 39338168 PMCID: PMC11433157 DOI: 10.3390/jpm14090914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/10/2024] [Accepted: 08/14/2024] [Indexed: 09/30/2024] Open
Abstract
Increased activity of transforming growth factor-beta (TGF-β) is a key factor mediating kidney impairment in diabetes. Glomerular podocytes, the crucial component of the renal filter, are a direct target of TGF-β action, resulting in irreversible cell loss and progression of chronic kidney disease (CKD). Urolithin A (UA) is a member of the family of polyphenol metabolites produced by gut microbiota from ellagitannins and ellagic acid-rich foods. The broad spectrum of biological activities of UA makes it a promising candidate for the treatment of podocyte disorders. In this in vitro study, we investigated whether UA influences the changes exerted in podocytes by TGF-β and high glucose. Following a 7-day incubation in normal (NG, 5.5 mM) or high (HG, 25 mM) glucose, the cells were treated with UA and/or TGF-β1 for 24 h. HG and TGF-β1, each independent and in concert reduced expression of nephrin, increased podocyte motility, and up-regulated expression of b3 integrin and fibronectin. These typical-for-epithelial-to-mesenchymal transition (EMT) effects were inhibited by UA in both HG and NG conditions. UA also reduced the typically elevated HG expression of TGF-β receptors and activation of the TGF-β signal transducer Smad2. Our results indicate that in podocytes cultured in conditions mimicking the diabetic milieu, UA inhibits and reverses changes underlying podocytopenia in diabetic kidneys. Hence, UA should be considered as a potential therapeutic agent in podocytopathies.
Collapse
Affiliation(s)
- Barbara Lewko
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Agnieszka Daca
- Department of Physiopathology, Faculty of Medicine, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
| | | | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics-Fahrenheit Biobank BBMRI, Faculty of Pharmacy, Medical University of Gdansk, 80-210 Gdansk, Poland
- BioTechMed Center, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 80-223 Gdansk, Poland
| |
Collapse
|
7
|
Snyder Y, Mann FAT, Middleton J, Murashita T, Carney J, Bianco RW, Jana S. Non-immune factors cause prolonged myofibroblast phenotype in implanted synthetic heart valve scaffolds. APPLIED MATERIALS TODAY 2024; 39:102323. [PMID: 39131741 PMCID: PMC11308761 DOI: 10.1016/j.apmt.2024.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/13/2024]
Abstract
The clinical application of heart valve scaffolds is hindered by complications associated with the activation of valvular interstitial cell-like (VIC-like) cells and their transdifferentiation into myofibroblasts. This study aimed to examine several molecular pathway(s) that may trigger the overactive myofibroblast phenotypes in the implanted scaffolds. So, we investigated the influence of three molecular pathways - macrophage-induced inflammation, the TGF-β1-SMAD2, and WNT/β-catenin β on VIC-like cells during tissue engineering of heart valve scaffolds. We implanted electrospun heart valve scaffolds in adult sheep for up to 6 months in the right ventricular outflow tract (RVOT) and analyzed biomolecular (gene and protein) expression associated with the above three pathways by the scaffold infiltrating cells. The results showed a gradual increase in gene and protein expression of markers related to the activation of VIC-like cells and the myofibroblast phenotypes over 6 months of scaffold implantation. Conversely, there was a gradual increase in macrophage activity for the first three months after scaffold implantation. However, a decrease in macrophage activity from three to six months of scaffold tissue engineering suggested that immunological signal factors were not the primary cause of myofibroblast phenotype. Similarly, the gene and protein expression of factors associated with the TGF-β1-SMAD2 pathway in the cells increased in the first three months but declined in the next three months. Contrastingly, the gene and protein expression of factors associated with the WNT/β-catenin pathway increased significantly over the six-month study. Thus, the WNT/β-catenin pathway could be the predominant mechanism in activating VIC-like cells and subsequent myofibroblast phenotype.
Collapse
Affiliation(s)
- Yuriy Snyder
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO 65211, USA
| | - FA Tony Mann
- Veterinary Health Center, University of Missouri, 900 East Campus Drive, Columbia, MO 65211-0001
| | - John Middleton
- Veterinary Health Center, University of Missouri, 900 East Campus Drive, Columbia, MO 65211-0001
| | - Takashi Murashita
- Department of Surgery, School of Medicine, University of Missouri, One Hospital Drive, Columbia, MO 65212
| | - John Carney
- Experimental Surgical Services, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455
| | - Richard W. Bianco
- Experimental Surgical Services, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN 55455
| | - Soumen Jana
- Department of Chemical and Biomedical Engineering, University of Missouri, 1406 Rollins Street, Columbia, MO 65211, USA
| |
Collapse
|
8
|
Chia ZJ, Cao YN, Little PJ, Kamato D. Transforming growth factor-β receptors: versatile mechanisms of ligand activation. Acta Pharmacol Sin 2024; 45:1337-1348. [PMID: 38351317 PMCID: PMC11192764 DOI: 10.1038/s41401-024-01235-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/28/2024] [Indexed: 02/19/2024]
Abstract
Transforming growth factor-β (TGF-β) signaling is initiated by activation of transmembrane TGF-β receptors (TGFBR), which deploys Smad2/3 transcription factors to control cellular responses. Failure or dysregulation in the TGF-β signaling pathways leads to pathological conditions. TGF-β signaling is regulated at different levels along the pathways and begins with the liberation of TGF-β ligand from its latent form. The mechanisms of TGFBR activation display selectivity to cell types, agonists, and TGF-β isoforms, enabling precise control of TGF-β signals. In addition, the cell surface compartments used to release active TGF-β are surprisingly vibrant, using thrombospondins, integrins, matrix metalloproteinases and reactive oxygen species. The scope of TGFBR activation is further unfolded with the discovery of TGFBR activation initiated by other signaling pathways. The unique combination of mechanisms works in series to trigger TGFBR activation, which can be explored as therapeutic targets. This comprehensive review provides valuable insights into the diverse mechanisms underpinning TGFBR activation, shedding light on potential avenues for therapeutic exploration.
Collapse
Affiliation(s)
- Zheng-Jie Chia
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia
| | - Ying-Nan Cao
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Peter J Little
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia
- Department of Pharmacy, Guangzhou Xinhua University, Guangzhou, 510520, China
| | - Danielle Kamato
- School of Pharmacy, The University of Queensland, Brisbane, QLD, 4102, Australia.
- Discovery Biology, School of Environment and Science, Griffith University, Brisbane, QLD, 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, 4111, Australia.
| |
Collapse
|
9
|
Flores-Hermenegildo JM, Hernández-Cázares FDJ, Pérez-Pérez D, Romero-Ramírez H, Rodríguez-Alba JC, Licona-Limon P, Kilimann MW, Santos-Argumedo L, López-Herrera G. Lrba participates in the differentiation of IgA+ B lymphocytes through TGFβR signaling. Front Immunol 2024; 15:1386260. [PMID: 38975349 PMCID: PMC11224471 DOI: 10.3389/fimmu.2024.1386260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Introduction Lrba is a cytoplasmic protein involved in vesicular trafficking. Lrba-deficient (Lrba-/-) mice exhibit substantially higher levels of IgA in both serum and feces than wild-type (WT) mice. Transforming growth factor β1 (TGFβ1) and its receptors (TGFβR I and II) is essential for differentiating IgA+ B cells. Furthermore, increased IgA production suggests a potential connection between Lrba and the TGFβR signaling pathway in IgA production. However, the specific function of Lrba in B cell biology remains unknown. Aim Given the increased IgA levels in Lrba-/- mice, the goal in this work was to explore the lymph organs where the switch to IgA occurs, and if TGFβR function is affected. Methods Non-immunized Lrba-/- mice were compared with Lrba+/+ mice. IgA levels in the serum and feces, as well as during peripheral B cell development, were determined. IgA+ B cells and plasma cells were assessed in the small intestine and secondary lymphoid organs, such as the spleen, mesenteric lymph nodes, and Peyer's patches. The TGFβR signaling pathway was evaluated by determining the expression of TGFβR on B cells. Additionally, SMAD2 phosphorylation was measured under basal conditions and in response to recombinant TGFβ. Finally, confocal microscopy was performed to investigate a possible interaction between Lrba and TGFβR in B cells. Results Lrba-/- mice exhibited significantly higher levels of circulating IgA, IgA+ B, and plasma cells than in peripheral lymphoid organs those in WT mice. TGFβR expression on the membrane of B cells was similar in both Lrba-/- and Lrba+/+ mice. However, intracellular TGFβR expression was reduced in Lrba-/- mice. SMAD2 phosphorylation showed increased levels under basal conditions; stimulation with recombinant TGFβ elicited a poorer response than in that in Lrba+/+ B cells. Finally, we found that Lrba colocalizes with TGFβR in B cells. Conclusion Lrba is essential in controlling TGFβR signaling, subsequently regulating SMAD2 phosphorylation on B cells. This mechanism may explain the increased differentiation of IgA+ B cells and production of IgA-producing plasma cells.
Collapse
Affiliation(s)
- José Mizael Flores-Hermenegildo
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
| | - Felipe de Jesús Hernández-Cázares
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Daniela Pérez-Pérez
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
- Programa de Doctorado en Ciencias Biológicas, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Héctor Romero-Ramírez
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Juan Carlos Rodríguez-Alba
- Unidad de Neuroinmunología y Neurooncología, Instituto Nacional de Neurología y Neurocirugia (NINN), Ciudad de México, Mexico
- Facultad de Medicina y Cirugía, Universidad Autónoma Benito Juárez de Oaxaca (UABJO), Ciudad de Oaxaca, Mexico
| | - Paula Licona-Limon
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Manfred W. Kilimann
- Department of Molecular Neurobiology, Max-Planck-Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Leopoldo Santos-Argumedo
- Departamento de Biomedicina, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (CINVESTAV), Ciudad de México, Mexico
| | - Gabriela López-Herrera
- Laboratorio de Inmunodeficiencias, Instituto Nacional de Pediatría (INP), Ciudad de México, Mexico
| |
Collapse
|
10
|
Brunner P, Kiwitz L, Li L, Thurley K. Diffusion-limited cytokine signaling in T cell populations. iScience 2024; 27:110134. [PMID: 39678490 PMCID: PMC11639737 DOI: 10.1016/j.isci.2024.110134] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/30/2024] [Accepted: 05/25/2024] [Indexed: 12/17/2024] Open
Abstract
Effective immune-cell responses depend on collective decision-making mediated by diffusible intercellular signaling proteins called cytokines. Here, we designed a three-dimensional spatiotemporal modeling framework and a precise finite-element simulation setup to systematically investigate the origin and consequences of spatially inhomogeneous cytokine distributions in lymph nodes. We found that such inhomogeneities are critical for effective paracrine signaling, and they do not arise by diffusion and uptake alone, but rather depend on properties of the cell population such as an all-or-none behavior of cytokine secreting cells. Furthermore, we assessed the regulatory properties of negative and positive feedback in combination with diffusion-limited signaling dynamics, and we derived statistical quantities to characterize the spatiotemporal signaling landscape in the context of specific tissue architectures. Overall, our simulations highlight the complex spatiotemporal dynamics imposed by cell-cell signaling with diffusible ligands, which entails a large potential for fine-tuned biological control especially if combined with feedback mechanisms.
Collapse
Affiliation(s)
- Patrick Brunner
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz-Institute, Berlin, Germany
- Institute of Biology, Humboldt University, Berlin, Germany
| | - Lukas Kiwitz
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz-Institute, Berlin, Germany
- Institute of Biology, Humboldt University, Berlin, Germany
| | - Lisa Li
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| | - Kevin Thurley
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz-Institute, Berlin, Germany
| |
Collapse
|
11
|
Nitta Y, Kurioka T, Mogi S, Sano H, Yamashita T. Suppression of the TGF-β signaling exacerbates degeneration of auditory neurons in kanamycin-induced ototoxicity in mice. Sci Rep 2024; 14:10910. [PMID: 38740884 PMCID: PMC11091189 DOI: 10.1038/s41598-024-61630-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
Transforming growth factor-β (TGF-β) signaling plays a significant role in multiple biological processes, including inflammation, immunity, and cell death. However, its specific impact on the cochlea remains unclear. In this study, we aimed to investigate the effects of TGF-β signaling suppression on auditory function and cochlear pathology in mice with kanamycin-induced ototoxicity. Kanamycin and furosemide (KM-FS) were systemically administered to 8-week-old C57/BL6 mice, followed by immediate topical application of a TGF-β receptor inhibitor (TGF-βRI) onto the round window membrane. Results showed significant TGF-β receptor upregulation in spiral ganglion neurons (SGNs) after KM-FA ototoxicity, whereas expression levels in the TGF-βRI treated group remained unchanged. Interestingly, despite no significant change in cochlear TGF-β expression after KM-FS ototoxicity, TGF-βRI treatment resulted in a significant decrease in TGF-β signaling. Regarding auditory function, TGF-βRI treatment offered no therapeutic effects on hearing thresholds and hair cell survival following KM-FS ototoxicity. However, SGN loss and macrophage infiltration were significantly increased with TGF-βRI treatment. These results imply that inhibition of TGF-β signaling after KM-FS ototoxicity promotes cochlear inflammation and SGN degeneration.
Collapse
Affiliation(s)
- Yoshihiro Nitta
- Department of Otorhinolaryngology and Head and Neck Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan.
| | - Takaomi Kurioka
- Department of Otorhinolaryngology, National Defense Medical College, 3-2 Namiki, Tokorozawa, Saitama, 359-8513, Japan.
| | - Sachiyo Mogi
- Department of Otorhinolaryngology and Head and Neck Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| | - Hajime Sano
- School of Allied Health Sciences, Kitasato University, Kanagawa, Japan
| | - Taku Yamashita
- Department of Otorhinolaryngology and Head and Neck Surgery, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa, 252-0374, Japan
| |
Collapse
|
12
|
Thipboonchoo N, Fongsupa S, Sureram S, Sa-nguansak S, Kesornpun C, Kittakoop P, Soodvilai S. Altenusin, a fungal metabolite, alleviates TGF-β1-induced EMT in renal proximal tubular cells and renal fibrosis in unilateral ureteral obstruction. Heliyon 2024; 10:e24983. [PMID: 38318047 PMCID: PMC10839986 DOI: 10.1016/j.heliyon.2024.e24983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/07/2024] Open
Abstract
Renal fibrosis is a pathological feature of chronic kidney disease (CKD), progressing toward end-stage kidney disease (ESKD). The aim of this study is to investigate the therapeutic potential of altenusin, a farnesoid X receptor (FXR) agonist derived from fungi, on renal fibrosis. The effect of altenusin was determined (i) in vitro using the transforming growth factor β1 (TGF-β1)-induced epithelial to mesenchymal transition (EMT) of human renal proximal tubular cells and (ii) in vivo using mouse unilateral ureteral obstruction (UUO). The findings revealed that incubation of 10 ng/ml TGF-β1 promotes morphological change in RPTEC/TERT1 cells, a human renal proximal tubular cell line, from epithelial to fibroblast-like cells. TGF-β1 markedly increased EMT markers namely α-smooth muscle actin (α-SMA), fibronectin, and matrix metalloproteinase 9 (MMP-9), while decreased the epithelial marker E-cadherin. Co-incubation TGF-β1 with altenusin preserved the epithelial characteristics of the renal epithelial cells by antagonizing TGF-β/Smad signaling pathway, specifically a decreased phosphorylation of Smad2/3 with an increased level of Smad7. Interestingly, the antagonizing effect of altenusin does not require FXR activation. Moreover, altenusin could reverse TGF-β1-induced fibroblast-like cells to epithelial-like cells. Treatment on UUO mice with 30 mg/kg altenusin significantly reduced the expression of α-SMA, fibronectin, and collagen type 1A1 (COL1A1). The reduction in the renal fibrosis markers is correlated with the decreased phosphorylation of Smad2/3 levels but does not improve E-cadherin protein expression. Collectively, altenusin reduces EMT in human renal proximal tubular cells and renal fibrosis by antagonizing the TGF-β/Smad signaling pathway.
Collapse
Affiliation(s)
- Natechanok Thipboonchoo
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Somsak Fongsupa
- Department of Medical Technology, Faculty of Allied Health Science, Thammasat University Rangsit Campus, Thailand
| | - Sanya Sureram
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok 10210, Thailand
| | - Suliporn Sa-nguansak
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok 10210, Thailand
| | - Chatchai Kesornpun
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok 10210, Thailand
| | - Prasat Kittakoop
- Chulabhorn Research Institute, Kamphaeng Phet 6 Road, Laksi, Bangkok 10210, Thailand
- Chulabhorn Graduate Institute, Program in Chemical Sciences, Chulabhorn Royal Academy, Laksi, Bangkok 10210, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, Ministry of Higher Education, Science, Research and Innovation, Bangkok 10400, Thailand
| | - Sunhapas Soodvilai
- Research Center of Transport Protein for Medical Innovation, Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, Ministry of Higher Education, Science, Research and Innovation, Bangkok 10400, Thailand
- Excellent Center for Drug Discovery, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
13
|
Bačenková D, Trebuňová M, Demeterová J, Živčák J. Human Chondrocytes, Metabolism of Articular Cartilage, and Strategies for Application to Tissue Engineering. Int J Mol Sci 2023; 24:17096. [PMID: 38069417 PMCID: PMC10707713 DOI: 10.3390/ijms242317096] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/02/2023] [Indexed: 12/18/2023] Open
Abstract
Hyaline cartilage, which is characterized by the absence of vascularization and innervation, has minimal self-repair potential in case of damage and defect formation in the chondral layer. Chondrocytes are specialized cells that ensure the synthesis of extracellular matrix components, namely type II collagen and aggregen. On their surface, they express integrins CD44, α1β1, α3β1, α5β1, α10β1, αVβ1, αVβ3, and αVβ5, which are also collagen-binding components of the extracellular matrix. This article aims to contribute to solving the problem of the possible repair of chondral defects through unique methods of tissue engineering, as well as the process of pathological events in articular cartilage. In vitro cell culture models used for hyaline cartilage repair could bring about advanced possibilities. Currently, there are several variants of the combination of natural and synthetic polymers and chondrocytes. In a three-dimensional environment, chondrocytes retain their production capacity. In the case of mesenchymal stromal cells, their favorable ability is to differentiate into a chondrogenic lineage in a three-dimensional culture.
Collapse
Affiliation(s)
- Darina Bačenková
- Department of Biomedical Engineering and Measurement, Faculty of Mechanical Engineering, Technical University of Košice, Letná 9, 042 00 Košice, Slovakia; (M.T.); (J.D.); (J.Ž.)
| | | | | | | |
Collapse
|
14
|
Atef MM, Mostafa YM, Ahmed AAM, El-Sayed NM. Simvastatin attenuates aluminium chloride-induced neurobehavioral impairments through activation of TGF-β1/ SMAD2 and GSK3β/β-catenin signalling pathways. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2023; 102:104220. [PMID: 37454825 DOI: 10.1016/j.etap.2023.104220] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterised by the presence of β-amyloid plaques and acetylcholine depletion leading to neurobehavioral defects. AD was contributed also with downregulation of TGF-β1/SMAD2 and GSK3β/β-catenin pathways. Simvastatin (SMV) improved memory function experimentally and clinically. Hence, this study aimed to investigate the mechanistic role of SMV against aluminium chloride (AlCl3) induced neurobehavioral impairments. AD was induced by AlCl3 (50 mg/kg) for 6 weeks. Mice received Simvastatin (10 or 20 mg/kg) or Donepezil (3 mg/kg) for 6 weeks after that the histopathological, immunohistochemical and biochemical test were examined. Treatment with SMV improved the memory deterioration induced by AlCl3 with significant recovery of the histopathological changes. This was concomitant with the decrease of AChE and Aβ (1-42). SMV provides its neuroprotective effect through upregulating the protein expression of β-catenin, TGF-β1 and downregulating the expression of GSK3β, TLR4 and p-SMAD2.
Collapse
Affiliation(s)
| | - Yasser M Mostafa
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt; Department of Pharmacology & Toxicology, Faculty of Pharmacy, Badr University in Cairo, Egypt
| | - Amal A M Ahmed
- Department of Cytology & Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Norhan M El-Sayed
- Department of Pharmacology & Toxicology, Faculty of Pharmacy, Suez Canal University, Ismailia 41522, Egypt.
| |
Collapse
|
15
|
Shiying S, Weihong W, Xiuqiong T, Yemei Q. TGFB3 gene mutation associated with mandibular coronoid process hyperplasia: a family investigation. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 136:e109-e115. [PMID: 37246056 DOI: 10.1016/j.oooo.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/30/2023]
Abstract
OBJECTIVE Coronoid process hyperplasia (CPH) of the mandible can lead to restricted mouth opening and maxillofacial deformities, which have been hypothesized to be closely associated with genetics. This study investigated the relationship between congenital CPH and TGFB3 mutation in a family of patients with CPH. STUDY DESIGN A limited mouth opening proband with CPH underwent whole-exome gene sequencing in November 2019, and the results confirmed compound heterozygous mutations in the TGFB3 gene. Subsequently, clinical imaging and genetic testing were performed on 10 other individuals in his family. RESULTS A total of 9 people in this family have CPH. Among them, 6 have the same exon compound heterozygous mutation sites of the TGFB3 gene (chr14-76446905 and chr14-76429713), accompanied by homozygous or heterozygous mutations in the 3'untranslated region (3'UTR) of the TGFB3 gene (chr14:76429555). The other 3 individuals have a homozygous mutation in the 3'untranslated region of the TGFB3 gene. CONCLUSION The heterogeneous compound mutation of the TGFB3 gene or the homozygous mutation of 3'UTR of the TGFB3 gene may be correlated with CPH. In addition, the specifically related mechanism needs to be confirmed by further genetic animal experiments.
Collapse
Affiliation(s)
- Shen Shiying
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital, Kunming Medical University, Kunming, Yunnan 650106, China
| | - Wang Weihong
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital, Kunming Medical University, Kunming, Yunnan 650106, China.
| | - Tang Xiuqiong
- Department of Stomatology, Luoping County People's Hospital, Qujing, Yunnan 655800, China
| | - Qian Yemei
- Department of Oral and Maxillofacial Surgery, Affiliated Stomatology Hospital, Kunming Medical University, Kunming, Yunnan 650106, China
| |
Collapse
|
16
|
Zujur D, Al-Akashi Z, Nakamura A, Zhao C, Takahashi K, Aritomi S, Theoputra W, Kamiya D, Nakayama K, Ikeya M. Enhanced chondrogenic differentiation of iPS cell-derived mesenchymal stem/stromal cells via neural crest cell induction for hyaline cartilage repair. Front Cell Dev Biol 2023; 11:1140717. [PMID: 37234772 PMCID: PMC10206169 DOI: 10.3389/fcell.2023.1140717] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
Background: To date, there is no effective long-lasting treatment for cartilage tissue repair. Primary chondrocytes and mesenchymal stem/stromal cells are the most commonly used cell sources in regenerative medicine. However, both cell types have limitations, such as dedifferentiation, donor morbidity, and limited expansion. Here, we report a stepwise differentiation method to generate matrix-rich cartilage spheroids from induced pluripotent stem cell-derived mesenchymal stem/stromal cells (iMSCs) via the induction of neural crest cells under xeno-free conditions. Methods: The genes and signaling pathways regulating the chondrogenic susceptibility of iMSCs generated under different conditions were studied. Enhanced chondrogenic differentiation was achieved using a combination of growth factors and small-molecule inducers. Results: We demonstrated that the use of a thienoindazole derivative, TD-198946, synergistically improves chondrogenesis in iMSCs. The proposed strategy produced controlled-size spheroids and increased cartilage extracellular matrix production with no signs of dedifferentiation, fibrotic cartilage formation, or hypertrophy in vivo. Conclusion: These findings provide a novel cell source for stem cell-based cartilage repair. Furthermore, since chondrogenic spheroids have the potential to fuse within a few days, they can be used as building blocks for biofabrication of larger cartilage tissues using technologies such as the Kenzan Bioprinting method.
Collapse
Affiliation(s)
- Denise Zujur
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ziadoon Al-Akashi
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Anna Nakamura
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Chengzhu Zhao
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Laboratory of Skeletal Development and Regeneration, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Kazuma Takahashi
- Research Institute for Bioscience Product and Fine Chemicals, Ajinomoto Co., Inc, Kawasaki, Japan
| | - Shizuka Aritomi
- Research Institute for Bioscience Product and Fine Chemicals, Ajinomoto Co., Inc, Kawasaki, Japan
| | - William Theoputra
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Daisuke Kamiya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Takeda-CiRA Joint Program (T-CiRA), Kanagawa, Japan
| | - Koichi Nakayama
- Center for Regenerative Medicine Research, Faculty of Medicine, Saga University, Saga, Japan
| | - Makoto Ikeya
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
- Takeda-CiRA Joint Program (T-CiRA), Kanagawa, Japan
| |
Collapse
|
17
|
Soomro A, Khajehei M, Li R, O’Neil K, Zhang D, Gao B, MacDonald M, Kakoki M, Krepinsky JC. A therapeutic target for CKD: activin A facilitates TGFβ1 profibrotic signaling. Cell Mol Biol Lett 2023; 28:10. [PMID: 36717814 PMCID: PMC9885651 DOI: 10.1186/s11658-023-00424-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND TGFβ1 is a major profibrotic mediator in chronic kidney disease (CKD). Its direct inhibition, however, is limited by adverse effects. Inhibition of activins, also members of the TGFβ superfamily, blocks TGFβ1 profibrotic effects, but the mechanism underlying this and the specific activin(s) involved are unknown. METHODS Cells were treated with TGFβ1 or activins A/B. Activins were inhibited generally with follistatin, or specifically with neutralizing antibodies or type I receptor downregulation. Cytokine levels, signaling and profibrotic responses were assessed with ELISA, immunofluorescence, immunoblotting and promoter luciferase reporters. Wild-type or TGFβ1-overexpressing mice with unilateral ureteral obstruction (UUO) were treated with an activin A neutralizing antibody. RESULTS In primary mesangial cells, TGFβ1 induces secretion primarily of activin A, which enables longer-term profibrotic effects by enhancing Smad3 phosphorylation and transcriptional activity. This results from lack of cell refractoriness to activin A, unlike that for TGFβ1, and promotion of TGFβ type II receptor expression. Activin A also supports transcription through regulating non-canonical MRTF-A activation. TGFβ1 additionally induces secretion of activin A, but not B, from tubular cells, and activin A neutralization prevents the TGFβ1 profibrotic response in renal fibroblasts. Fibrosis induced by UUO is inhibited by activin A neutralization in wild-type mice. Worsened fibrosis in TGFβ1-overexpressing mice is associated with increased renal activin A expression and is inhibited to wild-type levels with activin A neutralization. CONCLUSIONS Activin A facilitates TGFβ1 profibrotic effects through regulation of both canonical (Smad3) and non-canonical (MRTF-A) signaling, suggesting it may be a novel therapeutic target for preventing fibrosis in CKD.
Collapse
Affiliation(s)
- Asfia Soomro
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Mohammad Khajehei
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Renzhong Li
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Kian O’Neil
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Dan Zhang
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Bo Gao
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Melissa MacDonald
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada
| | - Masao Kakoki
- grid.410711.20000 0001 1034 1720Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC USA
| | - Joan C. Krepinsky
- grid.25073.330000 0004 1936 8227Division of Nephrology, Department of Medicine, McMaster University, Hamilton, Canada ,grid.416721.70000 0001 0742 7355St. Joseph’s Hospital, 50 Charlton Ave East, Rm T3311, Hamilton, ON L8N 4A6 Canada
| |
Collapse
|
18
|
Dynamics of hepatocyte-cholangiocyte cell-fate decisions during liver development and regeneration. iScience 2022; 25:104955. [PMID: 36060070 PMCID: PMC9437857 DOI: 10.1016/j.isci.2022.104955] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/17/2022] [Accepted: 08/12/2022] [Indexed: 11/25/2022] Open
Abstract
The immense regenerative potential of the liver is attributed to the ability of its two key cell types – hepatocytes and cholangiocytes – to trans-differentiate to one another either directly or through intermediate progenitor states. However, the dynamic features of decision-making between these cell-fates during liver development and regeneration remains elusive. Here, we identify a core gene regulatory network comprising c/EBPα, TGFBR2, and SOX9 which is multistable in nature, enabling three distinct cell states – hepatocytes, cholangiocytes, and liver progenitor cells (hepatoblasts/oval cells) – and stochastic switching among them. Predicted expression signature for these three states are validated through multiple bulk and single-cell transcriptomic datasets collected across developmental stages and injury-induced liver repair. This network can also explain the experimentally observed spatial organization of phenotypes in liver parenchyma and predict strategies for efficient cellular reprogramming. Our analysis elucidates how the emergent dynamics of underlying regulatory networks drive diverse cell-fate decisions in liver development and regeneration. Identified minimal regulatory network to model liver development and regeneration Changes in phenotypic landscapes by in-silico perturbations of regulatory networks Ability to explain physiological spatial patterning of liver cell types Decoded strategies for efficient reprogramming among liver cell phenotypes
Collapse
|
19
|
Duan Q, Shen X, He D, Xu Y, Zheng Z, Zheng Z, Jiang X, Ren M, Chen L, Zhang T, Lu Y, Ye L, Xie X. Role and Mechanism of Epithelial-Mesenchymal Transition Mediated by Inflammatory Stress-Induced TGF- β1 in Promoting Arteriovenous Fistula Stenosis. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:9454843. [PMID: 37671238 PMCID: PMC10477026 DOI: 10.1155/2022/9454843] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 05/27/2022] [Indexed: 09/07/2023]
Abstract
Objective To explore the role and mechanism of epithelial-mesenchymal transition (EMT) mediated by inflammatory stress-induced TGF-β1 in promoting arteriovenous fistula stenosis. Methods The inflammatory cells HK-2 were cultured by adding TGF-β1. The optimal stimulation time was determined after TGF-β1 was added. HK-2 cells were divided into two groups, DMEM/F12 medium was added to one group (the control group), and the other group was treated with TGF-β1 (10 ng/ml) in serum-free DMEM/F12 medium to stimulate cell differentiation to mesenchymal. Results TGF-β1 was stably expressed after being transfected into EMT. The expression of TGF-β1 in the experimental group was higher than that in the control group (P < 0.05) 7 days after transfection. Western blot showed that TGF-β1 protein expression was higher in the experimental group 7 days after transfection, and no TGF-β1 protein expression was detected in the control group. The smooth muscle cells showed α-SMA expression in the control group, but no cells with expression of SMA and CD31/vWF were found at the same time; α-SMA expression was shown in smooth muscle cells and proliferative myofibroblasts, but no cells with expressions of SMA and CD31/vWF were found at the same time. The observation group showed that the expression of α-SMA was detected in smooth muscle cells and proliferative myofibroblasts, CD31/vWF was also expressed in endothelial cells, and α-SMA and vWF were also observed in endothelial cells, but no CD31 expression was found. Conclusion The inflammatory stress-induced TGF-β1 could act on epithelial-mesenchymal transition and promote the degree of arteriovenous fistula stenosis.
Collapse
Affiliation(s)
- Qingqing Duan
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Xiaogang Shen
- Nephrology Department, Zhejiang Provincial Peoples' Hospital, Hangzhou 310014, China
| | - Dongyuan He
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Yuankai Xu
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Zhigui Zheng
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Zhibo Zheng
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Xinxin Jiang
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Min Ren
- The Department of Obstetrics and Gynecology, Zhejiang Hospital, Hangzhou 310012, China
| | - Lili Chen
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Ting Zhang
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Yunan Lu
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Luxi Ye
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| | - Xiaohui Xie
- Nephrology Department, Zhejiang Hospital, Hangzhou 310012, China
| |
Collapse
|
20
|
Smith SS, Chu D, Qu T, Aggleton JA, Schneider RA. Species-specific sensitivity to TGFβ signaling and changes to the Mmp13 promoter underlie avian jaw development and evolution. eLife 2022; 11:e66005. [PMID: 35666955 PMCID: PMC9246370 DOI: 10.7554/elife.66005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 06/03/2022] [Indexed: 12/02/2022] Open
Abstract
Precise developmental control of jaw length is critical for survival, but underlying molecular mechanisms remain poorly understood. The jaw skeleton arises from neural crest mesenchyme (NCM), and we previously demonstrated that these progenitor cells express more bone-resorbing enzymes including Matrix metalloproteinase 13 (Mmp13) when they generate shorter jaws in quail embryos versus longer jaws in duck. Moreover, if we inhibit bone resorption or Mmp13, we can increase jaw length. In the current study, we uncover mechanisms establishing species-specific levels of Mmp13 and bone resorption. Quail show greater activation of and sensitivity to transforming growth factor beta (TGFβ) signaling than duck; where intracellular mediators like SMADs and targets like Runt-related transcription factor 2 (Runx2), which bind Mmp13, become elevated. Inhibiting TGFβ signaling decreases bone resorption, and overexpressing Mmp13 in NCM shortens the duck lower jaw. To elucidate the basis for this differential regulation, we examine the Mmp13 promoter. We discover a SMAD-binding element and single nucleotide polymorphisms (SNPs) near a RUNX2-binding element that distinguish quail from duck. Altering the SMAD site and switching the SNPs abolish TGFβ sensitivity in the quail Mmp13 promoter but make the duck promoter responsive. Thus, differential regulation of TGFβ signaling and Mmp13 promoter structure underlie avian jaw development and evolution.
Collapse
Affiliation(s)
- Spenser S Smith
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Daniel Chu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Tiange Qu
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Jessye A Aggleton
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
21
|
Wong DCP, Lee EHC, Er J, Yow I, Koean RAG, Ang O, Xiao J, Low BC, Ding JL. Lung Cancer Induces NK Cell Contractility and Cytotoxicity Through Transcription Factor Nuclear Localization. Front Cell Dev Biol 2022; 10:871326. [PMID: 35652099 PMCID: PMC9149376 DOI: 10.3389/fcell.2022.871326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Actomyosin-mediated cellular contractility is highly conserved for mechanotransduction and signalling. While this phenomenon has been observed in adherent cell models, whether/how contractile forces regulate the function of suspension cells like natural killer (NK) cells during cancer surveillance, is unknown. Here, we demonstrated in coculture settings that the evolutionarily conserved NK cell transcription factor, Eomes, undergoes nuclear shuttling during lung cancer cell surveillance. Biophysical and biochemical analyses revealed mechanistic enhancement of NK cell actomyosin-mediated contractility, which is associated with nuclear flattening, thus enabling nuclear entry of Eomes associated with enhanced NK cytotoxicity. We found that NK cells responded to the presumed immunosuppressive TGFβ in the NK-lung cancer coculture medium to sustain its intracellular contractility through myosin light chain phosphorylation, thereby promoting Eomes nuclear localization. Therefore, our results demonstrate that lung cancer cells provoke NK cell contractility as an early phase activation mechanism and that Eomes is a plausible mechano-responsive protein for increased NK cytotoxicity. There is scope for strategic application of actomyosin-mediated contractility modulating drugs ex vivo, to reinvigorate NK cells prior to adoptive cancer immunotherapy in vivo (177 words).
Collapse
Affiliation(s)
- Darren Chen Pei Wong
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute Singapore, National University of Singapore, Singapore, Singapore
| | - E Hui Clarissa Lee
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Junzhi Er
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ivan Yow
- Mechanobiology Institute Singapore, National University of Singapore, Singapore, Singapore
| | | | - Owen Ang
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Jingwei Xiao
- Mechanobiology Institute Singapore, National University of Singapore, Singapore, Singapore
| | - Boon Chuan Low
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Mechanobiology Institute Singapore, National University of Singapore, Singapore, Singapore
- University Scholars Programme, National University of Singapore, Singapore, Singapore
| | - Jeak Ling Ding
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
- Integrative Sciences and Engineering Programme, National University of Singapore, Singapore, Singapore
| |
Collapse
|
22
|
Yadav P, Kundu P, Pandey VK, Amin PJ, Nair J, Shankar BS. Effects of prolonged treatment of TGF-βR inhibitor SB431542 on radiation-induced signaling in breast cancer cells. Int J Radiat Biol 2022; 98:1630-1644. [PMID: 35446183 DOI: 10.1080/09553002.2022.2069299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 01/04/2022] [Accepted: 03/31/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE We have earlier characterized increased TGF-β signaling in radioresistant breast cancer cells. In this study, we wanted to determine the effect of prolonged treatment of TGF-βR inhibitor SB431542 on radiation-induced signaling, viz., genes regulating apoptosis, EMT, anti and pro-inflammatory cytokines. MATERIALS AND METHODS Breast cancer cells were pretreated with TGF-βR inhibitor (SB 431542) followed by exposure to 6 Gy and recovery period of 7 days (D7-6G). We assessed cell survival by MTT assay, cytokines by ELISA and expression analysis by RT-PCR, flow cytometry, and western blot. We carried out migration assays using trans well inserts. We performed bioinformatics analyses of human cancer database through cBioportal. RESULTS There was an upregulation of TGF-β1 and 3 and downregulation of TGF-β2, TGF-βR1, and TGF-βR2 in invasive breast carcinoma samples compared to normal tissue. TGF-β1 and TNF-α was higher in radioresistant D7-6G cells with upregulation of pSMAD3, pNF-kB, and ERK signaling. Pretreatment of D7-6G cells with TGF-βR inhibitor SB431542 abrogated pSMAD3, increased proliferation, and migration along with an increase in apoptosis and pro-apoptotic genes. This was associated with hybrid E/M phenotype and downregulation of TGF-β downstream genes, HMGA2 and Snail. There was complete agreement in the expression of mRNA and protein data in genes like vimentin, Snail and HMGA2 in different treatment groups. However, there was disagreement in expression of mRNA and protein in genes like Bax, Bcl-2, E-cadherin, Zeb-1 among the different treatment groups indicating post-transcriptional and post-translational processing of these proteins. Treatment of cells with only SB431542 also increased expression of some E/M genes indicating TGF-β independent effects. Increased IL-6 and IL-10 secretion by SB431542 along with increase in pSTAT3 and pCREB1 could probably explain these TGF-β/Smad3 independent effects. CONCLUSION These results highlight that TGF-β-pSMAD3 and TNF-α-pNF-kB are the predominant signaling pathways in radioresistant cells and possibility of some TGF-β/Smad3 independent effects on prolonged treatment with the drug SB431542.
Collapse
Affiliation(s)
- Poonam Yadav
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Priya Kundu
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| | - Vipul K Pandey
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Prayag J Amin
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Jisha Nair
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
| | - Bhavani S Shankar
- Radiation Biology and Health Sciences Division, Bio-Science Group, Bhabha Atomic Research Centre, Mumbai, India
- Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
23
|
Mukti AI, Ilyas S, Warli SM, Putra A, Rasyid N, Munir D, Siregar KB, Ichwan M. Umbilical Cord-Derived Mesenchymal Stem Cells Improve TGF-β, α-SMA and Collagen on Erectile Dysfunction in Streptozotocin-Induced Diabetic Rats. Med Arch 2022; 76:4-11. [PMID: 35422561 PMCID: PMC8976889 DOI: 10.5455/medarh.2022.76.4-11] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 02/25/2022] [Indexed: 11/04/2022] Open
Abstract
Background A Erectile dysfunction (ED) is one of the well-known comorbidities in males with diabetes mellitus (DM), whose pathogenesis might be induced by dysregulation of corpus cavernosum smooth muscle cells. UC-MSCs are multipotent cells that attract considerable interest due to immunoregulatory properties and might be a potential strategy to regulate and recover the functional cells and tissues, including tissue improvement in DMED. Objective This study aims to determine the efficacy of UC-MSCs in improving the erectile function of DMED rats through analyzing the expression of TGF-β, α-SMA, and collagen. Methods Total number of 30 male Sprague-Dawley rats (6 to 8 weeks old) were randomly divided into four groups (negative control group, positive control group, T1 group, and T2 group). After 16 h fast, 24 rats were randomly selected and intraperitoneally injected with streptozotocin to induce DM. At 8 weeks after STZ injection, rats with DMED were identified by unresponsive erectile stimulation within 30 min. PC group received 500 μL; T1 rats treated with 500 μL PBS containing 1x106 UC-MSCs; T2 rats treated with 500 μL PBS containing 3x106 UC-MSCs. After MSCs treatment, the rats were sacrificed and the corpus cavernosum tissues were prepared for histological observations. Results This study resulted in the administration of UC-MSCs could downregulate the expression of TGF-β, α-SMA, and collagen leading to the improvement of DMED. Conclusion UC-MSCs improve the expression of TGF-β, α-SMA, and collagen on erectile dysfunction in streptozotocin-induced diabetic rats.
Collapse
Affiliation(s)
- Ade Indra Mukti
- Departement of Doctoral Degree Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Syafruddin Ilyas
- Department of Biology, Faculty of Mathematics and Natural Sciences, Universitas Sumatera Utara, Medan, Indonesia
| | - Syah Mirsya Warli
- Department of Urology, Faculty of Medicine / Universitas Sumatera Utara Hospital, Medan Indonesia
| | - Agung Putra
- Stem Cell and Cancer Research (SCCR), Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia
- Department of Pathology, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia
- Department of Postgraduate Biomedical Science, Medical Faculty, Sultan Agung Islamic University (UNISSULA), Semarang, Indonesia
| | - Nur Rasyid
- Department of Urology, Faculty of Medicine Universitas Indonesia, Cipto Mangunkusumo General Hospital, Jakarta, Indonesia
| | - Delfitri Munir
- Departement of Doctoral Degree Program, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
- Departement of Otorhinolaryngology-Head and Neck Surgery, Faculty of Medicine, Universitas Sumatera Utara, Medan, Medan, Indonesia
- Pusat Unggulan Inovasi (PUI) Stem Cell, Universitas Sumatera Utara (USU), Medan, Indonesia
| | - Kamal Basri Siregar
- Oncology Surgery Department, Faculty of Medicine, Universitas Sumatera Utara, Universitas Sumatera Utara Hospital, Medan, Indonesia
| | - Muhammad Ichwan
- Pusat Unggulan Inovasi (PUI) Stem Cell, Universitas Sumatera Utara (USU), Medan, Indonesia
- Departement of Pharmacology and Therapeutics, Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
24
|
Suliman HB, Healy Z, Zobi F, Kraft BD, Welty-Wolf K, Smith J, Barkauskas C, Piantadosi CA. Nuclear respiratory factor-1 negatively regulates TGF-β1 and attenuates pulmonary fibrosis. iScience 2022; 25:103535. [PMID: 34977500 PMCID: PMC8683592 DOI: 10.1016/j.isci.2021.103535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 09/02/2021] [Accepted: 11/25/2021] [Indexed: 12/27/2022] Open
Abstract
The preclinical model of bleomycin-induced lung fibrosis is useful to study mechanisms related to human pulmonary fibrosis. Using BLM in mice, we find low HO-1 expression. Although a unique Rhenium-CO-releasing molecule (ReCORM) up-regulates HO-1, NRF-1, CCN5, and SMAD7, it reduces TGFβ1, TGFβr1, collagen, α-SMA, and phosphorylated Smad2/3 levels in mouse lung and in human lung fibroblasts. ChIP assay studies confirm NRF-1 binding to the promoters of TGFβ1 repressors CCN5 and Smad7. ReCORM did not blunt lung fibrosis in Hmox1-deficient alveolar type 2 cell knockout mice, suggesting this gene participates in lung protection. In human lung fibroblasts, TGFβ1-dependent production of α-SMA is abolished by ReCORM or by NRF-1 gene transfection. We demonstrate effective HO-1/NRF-1 signaling in lung AT2 cells protects against BLM induced lung injury and fibrosis by maintaining mitochondrial health, function, and suppressing the TGFβ1 pathway. Thus, protection of AT2 cell mitochondrial integrity via HO-1/NRF-1 presents an innovative therapeutic target.
Collapse
Affiliation(s)
- Hagir B. Suliman
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
- Department of Anaesthesiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Zachary Healy
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Fabio Zobi
- Department of Chemistry, University of Fribourg, Fribourg, Switzerland
| | - Bryan D. Kraft
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Karen Welty-Wolf
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Joshua Smith
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Christina Barkauskas
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
| | - Claude A. Piantadosi
- Department of Medicine, Duke University School of Medicine, 200 Trent Drive, Durham, NC 27710, USA
- Department of Anaesthesiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
25
|
Mohamed R, Shajimoon A, Afroz R, Gabr M, Thomas WG, Little PJ, Kamato D. Akt acts as a switch for GPCR transactivation of the TGF-β receptor type 1. FEBS J 2021; 289:2642-2656. [PMID: 34826189 DOI: 10.1111/febs.16297] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 11/12/2021] [Accepted: 12/25/2021] [Indexed: 12/20/2022]
Abstract
Transforming growth factor (TGF)-β signalling commences with the engagement of TGF-β ligand to cell surface TGF-β receptors (TGFBR) stimulating Smad2 carboxyl-terminal phosphorylation (phospho-Smad2C) and downstream biological responses. In several cell models, G protein-coupled receptors (GPCRs) transactivate the TGF-β receptors type-1 (TGFBR1) leading to phospho-Smad2C, however, we have recently published that in keratinocytes thrombin did not transactivate the TGFBR1. The bulk of TGFBRs reside in the cytosol and in response to protein kinase B (Akt phosphorylation) can translocate to the cell surface increasing the cell's responsiveness to TGF-β. In this study, we investigate the role of Akt in GPCR transactivation of the TGFBR1. We demonstrate that angiotensin II and thrombin do not phosphorylate Smad2C in human vascular smooth muscle cells and in keratinocytes respectively. We used Akt agonist, SC79 to sensitise the cells to Akt and observed that Ang II and thrombin phosphorylate Smad2C via Akt/AS160-dependent pathways. We show that SC79 rapidly translocates TGFBRs to the cell surface thus increasing the cell's response to the GPCR agonist. These findings highlight novel mechanistic insight for the role of Akt in GPCR transactivation of the TGFBR1.
Collapse
Affiliation(s)
- Raafat Mohamed
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia
| | - Aravindra Shajimoon
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia.,School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Australia
| | - Rizwana Afroz
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia
| | - Mai Gabr
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, St Lucia, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Australia
| |
Collapse
|
26
|
Carlisle RE, Mohammed-Ali Z, Lu C, Yousof T, Tat V, Nademi S, MacDonald ME, Austin RC, Dickhout JG. TDAG51 induces renal interstitial fibrosis through modulation of TGF-β receptor 1 in chronic kidney disease. Cell Death Dis 2021; 12:921. [PMID: 34625532 PMCID: PMC8501078 DOI: 10.1038/s41419-021-04197-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/17/2021] [Accepted: 09/08/2021] [Indexed: 12/28/2022]
Abstract
Chronic kidney disease (CKD) is characterized by the gradual loss of renal function and is a major public health concern. Risk factors for CKD include hypertension and proteinuria, both of which are associated with endoplasmic reticulum (ER) stress. ER stress-induced TDAG51 protein expression is increased at an early time point in mice with CKD. Based on these findings, wild-type and TDAG51 knock-out (TDKO) mice were used in an angiotensin II/deoxycorticosterone acetate/salt model of CKD. Both wild-type and TDKO mice developed hypertension, increased proteinuria and albuminuria, glomerular injury, and tubular damage. However, TDKO mice were protected from apoptosis and renal interstitial fibrosis. Human proximal tubular cells were used to demonstrate that TDAG51 expression induces apoptosis through a CHOP-dependent mechanism. Further, a mouse model of intrinsic acute kidney injury demonstrated that CHOP is required for ER stress-mediated apoptosis. Renal fibroblasts were used to demonstrate that TGF-β induces collagen production through an IRE1-dependent mechanism; cells treated with a TGF-β receptor 1 inhibitor prevented XBP1 splicing, a downstream consequence of IRE1 activation. Interestingly, TDKO mice express significantly less TGF-β receptor 1, thus, preventing TGF-β-mediated XBP1 splicing. In conclusion, TDAG51 induces apoptosis in the kidney through a CHOP-dependent mechanism, while contributing to renal interstitial fibrosis through a TGF-β-IRE1-XBP1 pathway.
Collapse
Affiliation(s)
- Rachel E Carlisle
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Zahraa Mohammed-Ali
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Chao Lu
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Tamana Yousof
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Victor Tat
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Samera Nademi
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Melissa E MacDonald
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Richard C Austin
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada
| | - Jeffrey G Dickhout
- McMaster University and The Research Institute of St. Joe's Hamilton, Department of Medicine, Division of Nephrology, Hamilton, Canada.
| |
Collapse
|
27
|
Salesa B, Assis M, Andrés J, Serrano-Aroca Á. Carbon Nanofibers versus Silver Nanoparticles: Time-Dependent Cytotoxicity, Proliferation, and Gene Expression. Biomedicines 2021; 9:1155. [PMID: 34572341 PMCID: PMC8467915 DOI: 10.3390/biomedicines9091155] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 12/20/2022] Open
Abstract
Carbon nanofibers (CNFs) are one-dimensional nanomaterials with excellent physical and broad-spectrum antimicrobial properties characterized by a low risk of antimicrobial resistance. Silver nanoparticles (AgNPs) are antimicrobial metallic nanomaterials already used in a broad range of industrial applications. In the present study these two nanomaterials were characterized by Raman spectroscopy, transmission electron microscopy, zeta potential, and dynamic light scattering, and their biological properties were compared in terms of cytotoxicity, proliferation, and gene expression in human keratinocyte HaCaT cells. The results showed that both AgNPs and CNFs present similar time-dependent cytotoxicity (EC50 of 608.1 µg/mL for CNFs and 581.9 µg/mL for AgNPs at 24 h) and similar proliferative HaCaT cell activity. However, both nanomaterials showed very different results in the expression of thirteen genes (superoxide dismutase 1 (SOD1), catalase (CAT), matrix metallopeptidase 1 (MMP1), transforming growth factor beta 1 (TGFB1), glutathione peroxidase 1 (GPX1), fibronectin 1 (FN1), hyaluronan synthase 2 (HAS2), laminin subunit beta 1 (LAMB1), lumican (LUM), cadherin 1 CDH1, collagen type IV alpha (COL4A1), fibrillin (FBN), and versican (VCAN)) treated with the lowest non-cytotoxic concentrations in the HaCaT cells after 24 h. The AgNPs were capable of up-regulating only two genes (SOD1 and MMP1) while the CNFs were very effective in up-regulating eight genes (FN1, MMP1, CAT, CDH1, COL4A1, FBN, GPX1, and TGFB1) involved in the defense mechanisms against oxidative stress and maintaining and repairing tissues by regulating cell adhesion, migration, proliferation, differentiation, growth, morphogenesis, and tissue development. These results demonstrate CNF nanomaterials' unique great potential in biomedical applications such as tissue engineering and wound healing.
Collapse
Affiliation(s)
- Beatriz Salesa
- Biomaterials and Bioengineering Lab., Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| | - Marcelo Assis
- Department of Physical and Analytical Chemistry, University Jaume I (UJI), 12071 Castellon, Spain; (M.A.); (J.A.)
| | - Juan Andrés
- Department of Physical and Analytical Chemistry, University Jaume I (UJI), 12071 Castellon, Spain; (M.A.); (J.A.)
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab., Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, c/Guillem de Castro 94, 46001 Valencia, Spain;
| |
Collapse
|
28
|
Saleh AC, Sabry R, Mastromonaco GF, Favetta LA. BPA and BPS affect the expression of anti-Mullerian hormone (AMH) and its receptor during bovine oocyte maturation and early embryo development. Reprod Biol Endocrinol 2021; 19:119. [PMID: 34344364 PMCID: PMC8330045 DOI: 10.1186/s12958-021-00773-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 05/28/2021] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Exposure to endocrine-disrupting chemicals, such as Bisphenol A (BPA) and Bisphenol S (BPS), is widespread and has negative implications on embryonic development. Preliminary evidence revealed that in women undergoing IVF treatment, urinary BPA levels were associated with low serum anti-Mullerian hormone, however a definitive relationship between the two has not yet been characterized. METHODS This study aimed to evaluate BPA and BPS effects on in vitro oocyte maturation and early preimplantation embryo development through i) analysis of anti-Mullerian hormone (AMH) and anti-Mullerian hormone receptor II (AMHRII), ii) investigation of developmental parameters, such as cleavage, blastocyst rates and developmental arrest, iii) detection of apoptosis and iv) assessment of possible sex ratio skew. An in vitro bovine model was used as a translational model for human early embryonic development. We first assessed AMH and AMHRII levels after bisphenol exposure during oocyte maturation. Zygotes were also analyzed during cleavage and blastocysts stages. Techniques used include in vitro fertilization, quantitative polymerase chain reaction (qPCR), western blotting, TUNEL and immunofluorescence. RESULTS Our findings show that BPA significantly decreased cleavage (p < 0.001), blastocyst (p < 0.005) and overall developmental rates as well as significantly increased embryonic arrest at the 2-4 cell stage (p < 0.05). Additionally, both BPA and BPS significantly increased DNA fragmentation in 2-4 cells, 8-16 cells and blastocyst embryos (p < 0.05). Furthermore, BPA and BPS alter AMH and AMHRII at the mRNA and protein level in both oocytes and blastocysts. BPA, but not BPS, also significantly skews sex ratios towards female blastocysts (p < 0.05). CONCLUSION This study shows that BPA affects AMH and AMHRII expression during oocyte maturation and that BPS exerts its effects to a greater extent after fertilization and therefore may not be a safer alternative to BPA. Our data lay the foundation for future functional studies, such as receptor kinetics, downstream effectors, and promoter activation/inhibition to prove a functional relationship between bisphenols and the AMH signalling system.
Collapse
Affiliation(s)
- Angela Christina Saleh
- grid.34429.380000 0004 1936 8198Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario Canada
| | - Reem Sabry
- grid.34429.380000 0004 1936 8198Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario Canada
| | - Gabriela Fabiana Mastromonaco
- grid.34429.380000 0004 1936 8198Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario Canada
- grid.507770.20000 0001 0698 6008Reproductive Physiology, Toronto Zoo, Scarborough, Ontario Canada
| | - Laura Alessandra Favetta
- grid.34429.380000 0004 1936 8198Department of Biomedical Sciences, Ontario Veterinary College, University of Guelph, Guelph, Ontario Canada
| |
Collapse
|
29
|
Monteiro DA, Dole NS, Campos JL, Kaya S, Schurman CA, Belair CD, Alliston T. Fluid shear stress generates a unique signaling response by activating multiple TGFβ family type I receptors in osteocytes. FASEB J 2021; 35:e21263. [PMID: 33570811 PMCID: PMC7888383 DOI: 10.1096/fj.202001998r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/18/2022]
Abstract
Bone is a dynamic tissue that constantly adapts to changing mechanical demands. The transforming growth factor beta (TGFβ) signaling pathway plays several important roles in maintaining skeletal homeostasis by both coupling the bone‐forming and bone‐resorbing activities of osteoblasts and osteoclasts and by playing a causal role in the anabolic response of bone to applied loads. However, the extent to which the TGFβ signaling pathway in osteocytes is directly regulated by fluid shear stress (FSS) is unknown, despite work suggesting that fluid flow along canaliculi is a dominant physical cue sensed by osteocytes following bone compression. To investigate the effects of FSS on TGFβ signaling in osteocytes, we stimulated osteocytic OCY454 cells cultured within a microfluidic platform with FSS. We find that FSS rapidly upregulates Smad2/3 phosphorylation and TGFβ target gene expression, even in the absence of added TGFβ. Indeed, relative to treatment with TGFβ, FSS induced a larger increase in levels of pSmad2/3 and Serpine1 that persisted even in the presence of a TGFβ receptor type I inhibitor. Our results show that FSS stimulation rapidly induces phosphorylation of multiple TGFβ family R‐Smads by stimulating multimerization and concurrently activating several TGFβ and BMP type I receptors, in a manner that requires the activity of the corresponding ligand. While the individual roles of the TGFβ and BMP signaling pathways in bone mechanotransduction remain unclear, these results implicate that FSS activates both pathways to generate a downstream response that differs from that achieved by either ligand alone.
Collapse
Affiliation(s)
- David A Monteiro
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Neha S Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - J Luke Campos
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA
| | - Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA
| | - Cassandra D Belair
- The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA.,Department of Urology, University of California, San Francisco, CA, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, USA.,UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA, USA.,The Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, CA, USA
| |
Collapse
|
30
|
Yang Y, Ye WL, Zhang RN, He XS, Wang JR, Liu YX, Wang Y, Yang XM, Zhang YJ, Gan WJ. The Role of TGF- β Signaling Pathways in Cancer and Its Potential as a Therapeutic Target. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6675208. [PMID: 34335834 PMCID: PMC8321733 DOI: 10.1155/2021/6675208] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway mediates various biological functions, and its dysregulation is closely related to the occurrence of malignant tumors. However, the role of TGF-β signaling in tumorigenesis and development is complex and contradictory. On the one hand, TGF-β signaling can exert antitumor effects by inhibiting proliferation or inducing apoptosis of cancer cells. On the other hand, TGF-β signaling may mediate oncogene effects by promoting metastasis, angiogenesis, and immune escape. This review summarizes the recent findings on molecular mechanisms of TGF-β signaling. Specifically, this review evaluates TGF-β's therapeutic potential as a target by the following perspectives: ligands, receptors, and downstream signaling. We hope this review can trigger new ideas to improve the current clinical strategies to treat tumors related to the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Yun Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Jing-Ru Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Xuan Liu
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Yi Wang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Soochow University, Suzhou 215124, China
| |
Collapse
|
31
|
Cesaro E, Pastore A, Polverino A, Manna L, Divisato G, Quintavalle C, Di Sanzo M, Faniello MC, Grosso M, Costanzo P. ZNF224 is a mediator of TGF-β pro-oncogenic function in melanoma. Hum Mol Genet 2021; 30:2100-2109. [PMID: 34181020 DOI: 10.1093/hmg/ddab173] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/27/2022] Open
Abstract
The zinc finger protein ZNF224 plays a dual role in cancer, operating as both tumor suppressor and oncogenic factor depending on cellular and molecular partners. In this research we investigated the role of ZNF224 in melanoma, a highly invasive and metastatic cancer, and provided evidence for the involvement of ZNF224 in the TGF-β signaling as a mediator of the TGF-β pro-oncogenic function. Our results showed that ZNF224, whose expression increased in melanoma cell lines after TGF-β stimulation, potentiated the activation induced by TGF-β on its target genes involved in epithelial-mesenchymal transition (EMT). Accordingly, overexpression of ZNF224 enhanced the tumourigenic properties of melanoma cells, promoting cell proliferation and invasiveness, while ZNF224 knockdown had the opposite effect. Moreover, ZNF224 positively modulates the expression of TGF-β itself and its type 1 and 2 receptors (TβR1 and TβR2), thus highlighting a possible mechanism by which ZNF224 could enhance the endogenous TGFβ/Smad signalling. Our findings unveil a positive regulatory loop between TGF-β and ZNF224 to promote EMT, consequently increasing the tumour metastatic potential.
Collapse
Affiliation(s)
- Elena Cesaro
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Arianna Pastore
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Alessia Polverino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Lorenzo Manna
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Giuseppina Divisato
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Cristina Quintavalle
- Institute of Experimental Endocrinology and Oncology (IEOS) G. Salvatore, CNR, 80131 Naples, Italy
| | - Maddalena Di Sanzo
- Department of Experimental and Clinical Medicine University of Catanzaro "Magna Graecia", 88100 Catanzaro, Italy
| | - Maria Concetta Faniello
- Department of Experimental and Clinical Medicine University of Catanzaro "Magna Graecia", 88100 Catanzaro, Italy
| | - Michela Grosso
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| | - Paola Costanzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy
| |
Collapse
|
32
|
Basoli V, Della Bella E, Kubosch EJ, Alini M, Stoddart MJ. Effect of expansion media and fibronectin coating on growth and chondrogenic differentiation of human bone marrow-derived mesenchymal stromal cells. Sci Rep 2021; 11:13089. [PMID: 34158528 PMCID: PMC8219706 DOI: 10.1038/s41598-021-92270-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/08/2021] [Indexed: 01/01/2023] Open
Abstract
In the field of regenerative medicine, considerable advances have been made from the technological and biological point of view. However, there are still large gaps to be filled regarding translation and application of mesenchymal stromal cell (MSC)-based therapies into clinical practice. Indeed, variables such as cell type, unpredictable donor variation, and expansion/differentiation methods lead to inconsistencies. Most protocols use bovine serum (FBS) derivatives during MSC expansion. However, the xenogeneic risks associated with FBS limits the use of MSC-based products in clinical practice. Herein we compare a chemically defined, xenogeneic-free commercial growth medium with a conventional medium containing 10% FBS and 5 ng/ml FGF2. Furthermore, the effect of a fibronectin-coated growth surface was investigated. The effect of the different culture conditions on chondrogenic commitment was assessed by analyzing matrix deposition and gene expression of common chondrogenic markers. Chondrogenic differentiation potential was similar between the FBS-containing αMEM and the chemically defined medium with fibronectin coating. On the contrary, the use of fibronectin coating with FBS-containing medium appeared to reduce the differentiation potential of MSCs. Moreover, cells that were poorly responsive to in vitro chondrogenic stimuli were shown to improve their differentiation potential after expansion in a TGF-β1 containing medium. In conclusion, the use of a xenogeneic-free medium provides a suitable alternative for human bone marrow MSC expansion, due the capability to maintain cell characteristic and potency. To further improve chondrogenic potential of BMSCs, priming the cells with TGF-β1 during expansion is a promising strategy.
Collapse
Affiliation(s)
- Valentina Basoli
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Elena Della Bella
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Eva Johanna Kubosch
- Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Albert-Ludwigs-University of Freiburg, 79106, Freiburg, Germany
| | - Mauro Alini
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland
| | - Martin J Stoddart
- Regenerative Orthopaedics, AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, Switzerland. .,Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Albert-Ludwigs-University of Freiburg, 79106, Freiburg, Germany.
| |
Collapse
|
33
|
Enhanced Intestinal TGF-β/SMAD-Dependent Signaling in Simian Immunodeficiency Virus Infected Rhesus Macaques. Cells 2021; 10:cells10040806. [PMID: 33916615 PMCID: PMC8066988 DOI: 10.3390/cells10040806] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/18/2021] [Accepted: 04/01/2021] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β signaling (TGF-β) maintains a balanced physiological function including cell growth, differentiation, and proliferation and regulation of immune system by modulating either SMAD2/3 and SMAD7 (SMAD-dependent) or SMAD-independent signaling pathways under normal conditions. Increased production of TGF-β promotes immunosuppression in Human Immunodeficiency Virus (HIV)/Simian Immunodeficiency Virus (SIV) infection. However, the cellular source and downstream events of increased TGF-β production that attributes to its pathological manifestations remain unknown. Here, we have shown increased production of TGF-β in a majority of intestinal CD3−CD20−CD68+ cells from acute and chronically SIV infected rhesus macaques, which negatively correlated with the frequency of jejunum CD4+ T cells. No significant changes in intestinal TGF-β receptor II expression were observed but increased production of the pSMAD2/3 protein and SMAD3 gene expression in jejunum tissues that were accompanied by a downregulation of SMAD7 protein and gene expression. Enhanced TGF-β production by intestinal CD3−CD20−CD68+ cells and increased TGF-β/SMAD-dependent signaling might be due to a disruption of a negative feedback loop mediated by SMAD7. This suggests that SIV infection impacts the SMAD-dependent signaling pathway of TGF-β and provides a potential framework for further study to understand the role of viral factor(s) in modulating TGF-β production and downregulating SMAD7 expression in SIV. Regulation of mucosal TGF-β expression by therapeutic TGF-β blockers may help to create effective antiviral mucosal immune responses.
Collapse
|
34
|
Motizuki M, Koinuma D, Yokoyama T, Itoh Y, Omata C, Miyazono K, Saitoh M, Miyazawa K. TGF-β-induced cell motility requires downregulation of ARHGAPs to sustain Rac1 activity. J Biol Chem 2021; 296:100545. [PMID: 33741342 PMCID: PMC8079281 DOI: 10.1016/j.jbc.2021.100545] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) signaling promotes cancer progression. In particular, the epithelial-mesenchymal transition (EMT) induced by TGF-β is considered crucial to the malignant phenotype of cancer cells. Here, we report that the EMT-associated cellular responses induced by TGF-β are mediated by distinct signaling pathways that diverge at Smad3. By expressing chimeric Smad1/Smad3 proteins in SMAD3 knockout A549 cells, we found that the β4 region in the Smad3 MH1 domain is essential for TGF-β-induced cell motility, but is not essential for other EMT-associated responses including epithelial marker downregulation. TGF-β was previously reported to enhance cell motility by activating Rac1 via phosphoinositide 3-kinase. Intriguingly, TGF-β-dependent signaling mediated by Smad3's β4 region causes the downregulation of multiple mRNAs that encode GTPase activating proteins that target Rac1 (ARHGAPs), thereby attenuating Rac1 inactivation. Therefore, two independent pathways downstream of TGF-β type I receptor contribute cooperatively to sustained Rac1 activation, thereby leading to enhanced cell motility.
Collapse
Affiliation(s)
- Mitsuyoshi Motizuki
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Daizo Koinuma
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Takashi Yokoyama
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yuka Itoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Chiho Omata
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masao Saitoh
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan; Center for Medical Education and Science, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Keiji Miyazawa
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan.
| |
Collapse
|
35
|
Positioning of nucleosomes containing γ-H2AX precedes active DNA demethylation and transcription initiation. Nat Commun 2021; 12:1072. [PMID: 33594057 PMCID: PMC7886895 DOI: 10.1038/s41467-021-21227-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 01/12/2021] [Indexed: 01/09/2023] Open
Abstract
In addition to nucleosomes, chromatin contains non-histone chromatin-associated proteins, of which the high-mobility group proteins are the most abundant. Chromatin-mediated regulation of transcription involves DNA methylation and histone modifications. However, the order of events and the precise function of high-mobility group proteins during transcription initiation remain unclear. Here we show that high-mobility group AT-hook 2 protein (HMGA2) induces DNA nicks at the transcription start site, which are required by the histone chaperone FACT complex to incorporate nucleosomes containing the histone variant H2A.X. Further, phosphorylation of H2A.X at S139 (γ-H2AX) is required for repair-mediated DNA demethylation and transcription activation. The relevance of these findings is demonstrated within the context of TGFB1 signaling and idiopathic pulmonary fibrosis, suggesting therapies against this lethal disease. Our data support the concept that chromatin opening during transcriptional initiation involves intermediates with DNA breaks that subsequently require DNA repair mechanisms to ensure genome integrity. The order of DNA methylation and histone modifications during transcription remained unclear. Here the authors show that HMGA2 induces DNA nicks at TGFB1-responsive genes, promoting nucleosome incorporation containing γ-H2AX, which is required for repair-mediated DNA demethylation and transcription.
Collapse
|
36
|
Li M, Zha X, Wang S. The role of N6-methyladenosine mRNA in the tumor microenvironment. Biochim Biophys Acta Rev Cancer 2021; 1875:188522. [PMID: 33545295 DOI: 10.1016/j.bbcan.2021.188522] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/27/2021] [Accepted: 01/30/2021] [Indexed: 12/25/2022]
Abstract
In recent years, the most widely distributed eukaryotic messenger RNA (mRNA) modification, N6-methyladenosine (m6A), has received a large amount of interest, in part due to the development and advances of high-throughput RNA sequencing. The effects of m6A mRNA on tumor progression have been the most widely studied, and large amounts of conflicting data have been reported due to differences in tumor contexts, cell types or cell states. The majority of these studies were related to the significance of m6A mRNA on tumor cells, including on proliferation, stemness, invasion capability, etc. However, it has been noted that tumorigenesis and tumor progression cannot occur without support from the tumor microenvironment (TME), which contains multiple types of stromal cells, such as infiltrating immune cells (IICs), vascular cells, mesenchymal stem cells (MSCs), cancer-associated fibroblasts (CAFs), and various environmental factors. Here, we summarized the contributions of abnormal m6A mRNA in stromal cells within the TME and described the effects of m6A mRNA on TME remodeling.
Collapse
Affiliation(s)
- Min Li
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xuan Zha
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China; Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.
| |
Collapse
|
37
|
Zhu AK, Shan YQ, Zhang J, Liu XC, Ying RC, Kong WC. Exosomal NNMT from peritoneum lavage fluid promotes peritoneal metastasis in gastric cancer. Kaohsiung J Med Sci 2021; 37:305-313. [PMID: 33508890 DOI: 10.1002/kjm2.12334] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/22/2020] [Accepted: 11/08/2020] [Indexed: 12/18/2022] Open
Abstract
Peritoneal metastasis (PM) is the major cause of recurrence in patients with gastric cancer (GC) and is associated with poor prognosis. The oncogenic role of Nicotinamide N-methyltransferase (NNMT) in GC has been reported, but the role of secreted NNMT that is transported by exosomes remains unknown. In this study, exosomes were isolated from GC patients with or without PM and from GC cell line, including GC-114, GC-026, MKN45, and SNU-16 cells. The contents of NNMT were significantly enhanced in exosomes isolated from GC patients with PM compared with those from GC patients without PM. Furthermore, the levels of NNMT were significantly enhanced in exosomes from GC cell lines relative to those from normal human gastric epithelial cell line GES-1 cells. These data indicate that NNMT may be involved in intercellular communication for peritoneal dissemination. Moreover, colocalization of GC-derived exosomal NNMT was found in human peritoneal mesothelial cell line HMrSV5 cells. Additionally, relative to GES-1 exosomes, SNU-16 exosomes significantly activated TGF-β/smad2 signaling in HMrSV5 cells. However, when NNMT was silenced, the activation of TGF-β/smad2 by SNU-16 exosomes was abolished in HMrSV5 cells. We propose that NNMT-containing exosomes derived from GC cells could promote peritoneal metastasis via TGF-β/smad2 signaling.
Collapse
Affiliation(s)
- A-Kao Zhu
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Yu-Qiang Shan
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Jian Zhang
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Xin-Chun Liu
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Rong-Chao Ying
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| | - Wen-Cheng Kong
- Department of General Surgery, Affiliated Hangzhou First People Hospital, Zhejiang University School of Medicine, Zhejiang Province, China
| |
Collapse
|
38
|
Ungefroren H. Autocrine TGF-β in Cancer: Review of the Literature and Caveats in Experimental Analysis. Int J Mol Sci 2021; 22:977. [PMID: 33478130 PMCID: PMC7835898 DOI: 10.3390/ijms22020977] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 12/14/2022] Open
Abstract
Autocrine signaling is defined as the production and secretion of an extracellular mediator by a cell followed by the binding of that mediator to receptors on the same cell to initiate signaling. Autocrine stimulation often operates in autocrine loops, a type of interaction, in which a cell produces a mediator, for which it has receptors, that upon activation promotes expression of the same mediator, allowing the cell to repeatedly autostimulate itself (positive feedback) or balance its expression via regulation of a second factor that provides negative feedback. Autocrine signaling loops with positive or negative feedback are an important feature in cancer, where they enable context-dependent cell signaling in the regulation of growth, survival, and cell motility. A growth factor that is intimately involved in tumor development and progression and often produced by the cancer cells in an autocrine manner is transforming growth factor-β (TGF-β). This review surveys the many observations of autocrine TGF-β signaling in tumor biology, including data from cell culture and animal models as well as from patients. We also provide the reader with a critical discussion on the various experimental approaches employed to identify and prove the involvement of autocrine TGF-β in a given cellular response.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein, Campus Lübeck, D-23538 Lübeck, Germany;
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, Campus Kiel, D-24105 Kiel, Germany
| |
Collapse
|
39
|
Zhang X, Shao X, Zhang R, Zhu R, Feng R. Integrated analysis reveals the alterations that LMNA interacts with euchromatin in LMNA mutation-associated dilated cardiomyopathy. Clin Epigenetics 2021; 13:3. [PMID: 33407844 PMCID: PMC7788725 DOI: 10.1186/s13148-020-00996-1] [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: 10/16/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Dilated cardiomyopathy (DCM) is a serious cardiac heterogeneous pathological disease, which may be caused by mutations in the LMNA gene. Lamins interact with not only lamina-associated domains (LADs) but also euchromatin by alone or associates with the lamina-associated polypeptide 2 alpha (LAP2α). Numerous studies have documented that LMNA regulates gene expression by interacting with LADs in heterochromatin. However, the role of LMNA in regulating euchromatin in DCM is poorly understood. Here, we determine the differential binding genes on euchromatin in DCM induced by LMNA mutation by performing an integrated analysis of bioinformatics and explore the possible molecular pathogenesis mechanism. RESULTS Six hundred twenty-three and 4484 differential binding genes were identified by ChIP-seq technology. The ChIP-seq analysis results and matched RNA-Seq transcriptome data were integrated to further validate the differential binding genes of ChIP-seq. Five and 60 candidate genes involved in a series of downstream analysis were identified. Finally, 4 key genes (CREBBP, PPP2R2B, BMP4, and BMP7) were harvested, and these genes may regulate LMNA mutation-induced DCM through WNT/β-catenin or TGFβ-BMP pathways. CONCLUSIONS We identified four key genes that may serve as potential biomarkers and novel therapeutic targets. Our study also illuminates the possible molecular pathogenesis mechanism that the abnormal binding between LMNA or LAP2α-lamin A/C complexes and euchromatin DNA in LMNA mutations, which may cause DCM through the changes of CREBBP, PPP2R2B, BMP4, BMP7 expressions, and the dysregulation of WNT/β-catenin or TGFβ-BMP pathways, providing valuable insights to improve the occurrence and development of DCM.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Xiuli Shao
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Ruijia Zhang
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Rongli Zhu
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China
| | - Rui Feng
- Department of Pharmaceutical Toxicology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| |
Collapse
|
40
|
McIlvenna LC, Patten RK, McAinch AJ, Rodgers RJ, Stepto NK, Moreno-Asso A. Transforming Growth Factor Beta 1 Alters Glucose Uptake but Not Insulin Signalling in Human Primary Myotubes From Women With and Without Polycystic Ovary Syndrome. Front Endocrinol (Lausanne) 2021; 12:732338. [PMID: 34707569 PMCID: PMC8544291 DOI: 10.3389/fendo.2021.732338] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/22/2021] [Indexed: 12/24/2022] Open
Abstract
Women with polycystic ovary syndrome (PCOS), commonly have profound skeletal muscle insulin resistance which can worsen other clinical features. The heterogeneity of the condition has made it challenging to identify the precise mechanisms that cause this insulin resistance. A possible explanation for the underlying insulin resistance may be the dysregulation of Transforming Growth Factor-beta (TGFβ) signalling. TGFβ signalling contributes to the remodelling of reproductive and hepatic tissues in women with PCOS. Given the systemic nature of TGFβ signalling and its role in skeletal muscle homeostasis, it may be possible that these adverse effects extend to other peripheral tissues. We aimed to determine if TGFβ1 could negatively regulate glucose uptake and insulin signalling in skeletal muscle of women with PCOS. We show that both myotubes from women with PCOS and healthy women displayed an increase in glucose uptake, independent of changes in insulin signalling, following short term (16 hr) TGFβ1 treatment. This increase occurred despite pro-fibrotic signalling increasing via SMAD3 and connective tissue growth factor in both groups following treatment with TGFβ1. Collectively, our findings show that short-term treatment with TGFβ1 does not appear to influence insulin signalling or promote insulin resistance in myotubes. These findings suggest that aberrant TGFβ signalling is unlikely to directly contribute to skeletal muscle insulin resistance in women with PCOS in the short term but does not rule out indirect or longer-term effects.
Collapse
Affiliation(s)
- Luke C. McIlvenna
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Rhiannon K. Patten
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Andrew J. McAinch
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, Melbourne, VIC, Australia
| | - Raymond J. Rodgers
- Discipline of Obstetrics and Gynaecology, School of Medicine, Robinson Research Institute, The University of Adelaide, Adelaide, SA, Australia
| | - Nigel K. Stepto
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, Melbourne, VIC, Australia
| | - Alba Moreno-Asso
- Institute for Health and Sport, Victoria University, Melbourne, VIC, Australia
- Australian Institute for Musculoskeletal Science (AIMSS), Victoria University, Melbourne, VIC, Australia
- *Correspondence: Alba Moreno-Asso,
| |
Collapse
|
41
|
Transforming growth factor beta type 1 (TGF-β) and hypoxia-inducible factor 1 (HIF-1) transcription complex as master regulators of the immunosuppressive protein galectin-9 expression in human cancer and embryonic cells. Aging (Albany NY) 2020; 12:23478-23496. [PMID: 33295886 PMCID: PMC7762483 DOI: 10.18632/aging.202343] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/15/2020] [Indexed: 12/11/2022]
Abstract
Galectin-9 is one of the key proteins employed by a variety of human malignancies to suppress anti-cancer activities of cytotoxic lymphoid cells and thus escape immune surveillance. Human cancer cells in most cases express higher levels of galectin-9 compared to non-transformed cells. However, the biochemical mechanisms underlying this phenomenon remain unclear. Here we report for the first time that in human cancer as well as embryonic cells, the transcription factors hypoxia-inducible factor 1 (HIF-1) and activator protein 1 (AP-1) are involved in upregulation of transforming growth factor beta 1 (TGF-β1) expression, leading to activation of the transcription factor Smad3 through autocrine action. This process triggers upregulation of galectin-9 expression in both malignant (mainly in breast and colorectal cancer as well as acute myeloid leukaemia (AML)) and embryonic cells. The effect, however, was not observed in mature non-transformed human cells. TGF-β1-activated Smad3 therefore displays differential behaviour in human cancer and embryonic vs non-malignant cells. This study uncovered a self-supporting biochemical mechanism underlying high levels of galectin-9 expression operated by the human cancer and embryonic cells employed in our investigations. Our results suggest the possibility of using the TGF-β1 signalling pathway as a potential highly efficient target for cancer immunotherapy.
Collapse
|
42
|
Zhou Y, Little PJ, Cao Y, Ta HT, Kamato D. Lysophosphatidic acid receptor 5 transactivation of TGFBR1 stimulates the mRNA expression of proteoglycan synthesizing genes XYLT1 and CHST3. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2020; 1867:118848. [PMID: 32920014 DOI: 10.1016/j.bbamcr.2020.118848] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/30/2020] [Accepted: 09/03/2020] [Indexed: 12/27/2022]
Abstract
Lysophosphatidic acid (LPA) via transactivation dependent signalling pathways contributes to a plethora of physiological and pathophysiological responses. In the vasculature, hyperelongation of glycosaminoglycan (GAG) chains on proteoglycans leads to lipid retention in the intima resulting in the early pathogenesis of atherosclerosis. Therefore, we investigated and defined the contribution of transactivation dependent signalling in LPA mediated GAG chain hyperelongation in human vascular smooth muscle cells (VSMCs). LPA acting via the LPA receptor 5 (LPAR5) transactivates the TGFBR1 to stimulate the mRNA expression of GAG initiation and elongation genes xylosyltransferase-1 (XYLT1) and chondroitin 6-sulfotransferase-1 (CHST3), respectively. We found that LPA stimulates ROS and Akt signalling in VSMCs, however they are not associated in LPAR5 transactivation of the TGFBR1. We observed that LPA via ROCK dependent pathways transactivates the TGFBR1 to stimulate genes associated with GAG chain elongation. We demonstrate that GPCR transactivation of the TGFBR1 occurs via a universal biochemical mechanism and the identified effectors represent potential therapeutic targets to inhibit pathophysiological effects of GPCR transactivation of the TGFBR1.
Collapse
Affiliation(s)
- Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia.
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China.
| | - Yingnan Cao
- Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Hang T Ta
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; School of Environment and Science, Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia.
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, the University of Queensland, Woolloongabba, Queensland 4102, Australia; Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China.
| |
Collapse
|
43
|
Ungefroren H, Wellner UF, Keck T, Lehnert H, Marquardt JU. The Small GTPase RAC1B: A Potent Negative Regulator of-and Useful Tool to Study-TGFβ Signaling. Cancers (Basel) 2020; 12:E3475. [PMID: 33266416 PMCID: PMC7700615 DOI: 10.3390/cancers12113475] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 02/06/2023] Open
Abstract
RAC1 and its alternatively spliced isoform, RAC1B, are members of the Rho family of GTPases. Both isoforms are involved in the regulation of actin cytoskeleton remodeling, cell motility, cell proliferation, and epithelial-mesenchymal transition (EMT). Compared to RAC1, RAC1B exhibits a number of distinctive features with respect to tissue distribution, downstream signaling and a role in disease conditions like inflammation and cancer. The subcellular locations and interaction partners of RAC1 and RAC1B vary depending on their activation state, which makes RAC1 and RAC1B ideal candidates to establish cross-talk with cancer-associated signaling pathways-for instance, interactions with signaling by transforming growth factor β (TGFβ), a known tumor promoter. Although RAC1 has been found to promote TGFβ-driven tumor progression, recent observations in pancreatic carcinoma cells surprisingly revealed that RAC1B confers anti-oncogenic properties, i.e., through inhibiting TGFβ-induced EMT. Since then, an unexpected array of mechanisms through which RAC1B cross-talks with TGFβ signaling has been demonstrated. However, rather than being uniformly inhibitory, RAC1B interacts with TGFβ signaling in a way that results in the selective blockade of tumor-promoting pathways, while concomitantly allowing tumor-suppressive pathways to proceed. In this review article, we are going to discuss the specific interactions between RAC1B and TGFβ signaling, which occur at multiple levels and include various components such as ligands, receptors, cytosolic mediators, transcription factors, and extracellular inhibitors of TGFβ ligands.
Collapse
Affiliation(s)
- Hendrik Ungefroren
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, Campus Kiel, University Hospital Schleswig-Holstein, D-24105 Kiel, Germany
| | - Ulrich F. Wellner
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | - Tobias Keck
- Clinic for Surgery, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany; (U.F.W.); (T.K.)
| | | | - Jens-Uwe Marquardt
- First Department of Medicine, Campus Lübeck, University Hospital Schleswig-Holstein, D-23538 Lübeck, Germany;
| |
Collapse
|
44
|
Posa F, Baha-Schwab EH, Wei Q, Di Benedetto A, Neubauer S, Reichart F, Kessler H, Spatz JP, Albiges-Rizo C, Mori G, Cavalcanti-Adam EA. Surface Co-presentation of BMP-2 and integrin selective ligands at the nanoscale favors α 5β 1 integrin-mediated adhesion. Biomaterials 2020; 267:120484. [PMID: 33142116 DOI: 10.1016/j.biomaterials.2020.120484] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 10/14/2020] [Accepted: 10/21/2020] [Indexed: 12/17/2022]
Abstract
Here we present the use of surface nanopatterning of covalently immobilized BMP-2 and integrin selective ligands to determine the specificity of their interactions in regulating cell adhesion and focal adhesion assembly. Gold nanoparticle arrays carrying single BMP-2 dimers are prepared by block-copolymer micellar nanolithography and azide-functionalized integrin ligands (cyclic-RGD peptides or α5β1 integrin peptidomimetics) are immobilized on the surrounding polyethylene glycol alkyne by click chemistry. Compared to BMP-2 added to the media, surface immobilized BMP-2 (iBMP-2) favors the spatial segregation of adhesion clusters and enhances focal adhesion (FA) size in cells adhering to α5β1 integrin selective ligands. Moreover, iBMP-2 copresented with α5β1 integrin ligands induces the recruitment of αvβ3 integrins in FAs. When copresented with RGD, iBMP-2 induces the assembly of a higher number of FAs, which are not affected by α5β1 integrin blocking. Our dual-functionalized platforms offer the possibility to study the crosstalk between integrins and BMP receptors, and more in general they could be used to address the spatial regulation of growth factors and adhesion receptors crosstalk on biomimetic surfaces.
Collapse
Affiliation(s)
- Francesca Posa
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Elisabeth H Baha-Schwab
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Qiang Wei
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Adriana Di Benedetto
- University of Foggia, Department of Clinical and Experimental Medicine, viale Pinto 1, Foggia, 71122, Italy
| | - Stefanie Neubauer
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Florian Reichart
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Horst Kessler
- Institute for Advanced Study and Center of Integrated Protein Science (CIPSM), Department of Chemistry, Technische Universität München, Lichtenbergstraße 4, Garching, 85748, Germany
| | - Joachim P Spatz
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany
| | - Corinne Albiges-Rizo
- Institut Albert Bonniot, Université Joseph Fourier, INSERM U823, CNRS ERL 5284, Grenoble Alpessite Santé, Grenoble Cedex, 09, F38042, France
| | - Giorgio Mori
- University of Foggia, Department of Clinical and Experimental Medicine, viale Pinto 1, Foggia, 71122, Italy
| | - Elisabetta Ada Cavalcanti-Adam
- Max Planck Institute for Medical Research, Department of Cellular Biophysics, Jahnstraße 29, Heidelberg, 69120, Germany.
| |
Collapse
|
45
|
Marudamuthu AS, Bhandary YP, Fan L, Radhakrishnan V, MacKenzie B, Maier E, Shetty SK, Nagaraja MR, Gopu V, Tiwari N, Zhang Y, Watts AB, Williams RO, Criner GJ, Bolla S, Marchetti N, Idell S, Shetty S. Caveolin-1-derived peptide limits development of pulmonary fibrosis. Sci Transl Med 2020; 11:11/522/eaat2848. [PMID: 31826982 DOI: 10.1126/scitranslmed.aat2848] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/05/2019] [Accepted: 11/08/2019] [Indexed: 12/15/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal fibrotic lung disease with a median 5-year survival of ~20%. Current U.S. Food and Drug Administration-approved pharmacotherapies slow progression of IPF, providing hope that even more effective treatments can be developed. Alveolar epithelial progenitor type II cell (AEC) apoptosis and proliferation, and accumulation of activated myofibroblasts or fibrotic lung fibroblasts (fLfs) contribute to the progression of IPF. Full-length caveolin-1 scaffolding domain peptide (CSP; amino acids 82 to 101 of Cav1: DGIWKASFTTFTVTKYWFYR) inhibits AEC apoptosis and fLf activation and expansion and attenuates PF in bleomycin (BLM)-induced lung injury in mice. Like full-length CSP, a seven-amino acid deletion fragment of CSP, CSP7 (FTTFTVT), demonstrated antifibrotic effects in murine models of lung fibrosis. When CSP7 was administered during the fibrotic phase in three preclinical models [single-dose BLM, repeated-dose BLM, and adenovirus expressing constitutively active transforming growth factor-β1 (Ad-TGF-β1)-induced established PF], CSP7 reduced extracellular matrix (ECM) markers characteristic of PF, increased AEC survival, and improved lung function. CSP7 is amenable to both systemic (intraperitoneal) or direct lung delivery in a nebulized or dry powder form. Furthermore, CSP7 treatment of end-stage human IPF lung tissue explants attenuated ECM production and promoted AEC survival. Ames testing for mutagenicity and in vitro human peripheral blood lymphocyte and in vivo mouse micronucleus transformation assays indicated that CSP7 is not carcinogenic. Together, these findings support the further development of CSP7 as an antifibrotic treatment for patients with IPF or other interstitial lung diseases.
Collapse
Affiliation(s)
- Amarnath Satheesh Marudamuthu
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Yashodhar Prabhakar Bhandary
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Liang Fan
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Vijay Radhakrishnan
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - BreAnne MacKenzie
- Lung Therapeutics Inc., 2801 Via Fortuna Suite 425 Austin, TX 78746, USA
| | - Esther Maier
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA
| | - Shwetha Kumari Shetty
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - M R Nagaraja
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Venkadesaperumal Gopu
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Nivedita Tiwari
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Yajie Zhang
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA
| | - Alan B Watts
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA
| | - Robert O Williams
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, University of Texas at Austin, 2409 University Avenue, Austin, TX 78712, USA
| | - Gerald J Criner
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, 3401 North Broad Street, Philadelphia, PA 19140, USA
| | - Sudhir Bolla
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, 3401 North Broad Street, Philadelphia, PA 19140, USA
| | - Nathaniel Marchetti
- Department of Thoracic Medicine and Surgery, Lewis Katz School of Medicine, Temple University, 3401 North Broad Street, Philadelphia, PA 19140, USA
| | - Steven Idell
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA
| | - Sreerama Shetty
- Department of Medicine, Texas Lung Injury Institute, University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX 75708, USA.
| |
Collapse
|
46
|
Liao Z, Chen L, Zhang X, Zhang H, Tan X, Dong K, Lu X, Zhu H, Liu Q, Zhang Z, Ding Z, Dong W, Zhu P, Chu L, Liang H, Datta PK, Zhang B, Chen X. PTPRε Acts as a Metastatic Promoter in Hepatocellular Carcinoma by Facilitating Recruitment of SMAD3 to TGF-β Receptor 1. Hepatology 2020; 72:997-1012. [PMID: 31903610 DOI: 10.1002/hep.31104] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/17/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND AIMS Transforming growth factor beta (TGF-β) suppresses early stages of tumorigenesis, but contributes to the migration and metastasis of cancer cells. However, the role of TGF-β signaling in invasive prometastatic hepatocellular carcinoma (HCC) is poorly understood. In this study, we investigated the roles of canonical TGF-β/mothers against decapentaplegic homolog 3 (SMAD3) signaling and identified downstream effectors on HCC migration and metastasis. APPROACH AND RESULTS By using in vitro trans-well migration and invasion assays and in vivo metastasis models, we demonstrated that SMAD3 and protein tyrosine phosphatase receptor epsilon (PTPRε) promote migration, invasion, and metastasis of HCC cells in vitro and in vivo. Further mechanistic studies revealed that, following TGF-β stimulation, SMAD3 binds directly to PTPRε promoters to activate its expression. PTPRε interacts with TGFBR1/SMAD3 and facilitates recruitment of SMAD3 to TGFBR1, resulting in a sustained SMAD3 activation status. The tyrosine phosphatase activity of PTPRε is important for binding with TGFBR1, recruitment and activation of SMAD3, and its prometastatic role in vitro. A positive correlation between pSMAD3/SMAD3 and PTPRε expression was determined in HCC samples, and high expression of SMAD3 or PTPRε was associated with poor prognosis of patients with HCC. CONCLUSIONS PTPRε positive feedback regulates TGF-β/SMAD3 signaling to promote HCC metastasis.
Collapse
Affiliation(s)
- Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Lin Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xuewu Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Hongwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xiaolong Tan
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Keshuai Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Xun Lu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - He Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Qiumeng Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Zeyang Ding
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Wei Dong
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Peng Zhu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Liang Chu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Huifang Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
| | - Pran K Datta
- Division of Hematology and Oncology, Department of Medicine, UAB Comprehensive Cancer Center, Birmingham, AL
- Birmingham Veterans Affairs Medical Center, Birmingham, AL
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
47
|
Moon HR, Jung JM, Kim SY, Song Y, Chang SE. TGF-β3 suppresses melanogenesis in human melanocytes cocultured with UV-irradiated neighboring cells and human skin. J Dermatol Sci 2020; 99:100-108. [PMID: 32620316 DOI: 10.1016/j.jdermsci.2020.06.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 06/20/2020] [Accepted: 06/22/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Ultraviolet radiation (UVR) is the most well-known cause of skin pigmentation accompanied with photoaging. Transforming growth factor (TGF)-β1 was previously shown to have anti-melanogenic property; however, it can induce scarring in skin. OBJECTIVE We investigated the effect of TGF-β3 on melanogenesis in human melanocytes cocultured with UV-irradiated skin constituent cells, and UV-irradiated human skin. METHODS UVB irradiation or treatment with stem cell factor (SCF) and endothelin-1 (ET-1) was applied to human melanocytes cocultured with keratinocytes and/or fibroblasts and ex vivo human skin. Mechanistic pathways were further explored after treatment with TGF-β3. RESULTS While UVB irradiation or SCF/ET-1 enhanced melanogenesis, TGF-β3 effectively inhibited melanin accumulation and tyrosinase activity via downregulation of the extracellular signal-regulated kinase (ERK)/microphthalmia-associated transcription factor (MITF) pathway. TGF-β3 increased the expression of differentiation markers of keratinocytes. CONCLUSION TGF-β3 effectively suppressed UVR-stimulated melanogenesis indicating that topical TGF-β3 may be a suitable candidate for the treatment of UV-associated hyperpigmentation disorders.
Collapse
Affiliation(s)
- Hye-Rim Moon
- Beautiful skin clinic, 16-26, Sanbon-ro 323beon-gil, Gunpo-si, Gyeonggi-do, Republic of Korea
| | - Joon Min Jung
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Su Yeon Kim
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Youngsup Song
- Department of Biomedical Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Sung Eun Chang
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
48
|
AGAP2: Modulating TGFβ1-Signaling in the Regulation of Liver Fibrosis. Int J Mol Sci 2020; 21:ijms21041400. [PMID: 32092977 PMCID: PMC7073092 DOI: 10.3390/ijms21041400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
AGAP2 (Arf GAP with GTP-binding protein-like domain, Ankyrin repeat and PH domain 2) isoform 2 is a protein that belongs to the Arf GAP (GTPase activating protein) protein family. These proteins act as GTPase switches for Arfs, which are Ras superfamily members, being therefore involved in signaling regulation. Arf GAP proteins have been shown to participate in several cellular functions including membrane trafficking and actin cytoskeleton remodeling. AGAP2 is a multi-tasking Arf GAP that also presents GTPase activity and is involved in several signaling pathways related with apoptosis, cell survival, migration, and receptor trafficking. The increase of AGAP2 levels is associated with pathologies as cancer and fibrosis. Transforming growth factor beta-1 (TGF-β1) is the most potent pro-fibrotic cytokine identified to date, currently accepted as the principal mediator of the fibrotic response in liver, lung, and kidney. Recent literature has described that the expression of AGAP2 modulates some of the pro-fibrotic effects described for TGF-β1 in the liver. The present review is focused on the interrelated molecular effects between AGAP2 and TGFβ1 expression, presenting AGAP2 as a new player in the signaling of this pro-fibrotic cytokine, thereby contributing to the progression of hepatic fibrosis.
Collapse
|
49
|
Role of TGF-β in Skin Chronic Wounds: A Keratinocyte Perspective. Cells 2020; 9:cells9020306. [PMID: 32012802 PMCID: PMC7072438 DOI: 10.3390/cells9020306] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/17/2020] [Accepted: 01/27/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic wounds are characterized for their incapacity to heal within an expected time frame. Potential mechanisms driving this impairment are poorly understood and current hypotheses point to the development of an unbalanced milieu of growth factor and cytokines. Among them, TGF-β is considered to promote the broadest spectrum of effects. Although it is known to contribute to healthy skin homeostasis, the highly context-dependent nature of TGF-β signaling restricts the understanding of its roles in healing and wound chronification. Historically, low TGF-β levels have been suggested as a pattern in chronic wounds. However, a revision of the available evidence in humans indicates that this could constitute a questionable argument. Thus, in chronic wounds, divergences regarding skin tissue compartments seem to be characterized by elevated TGF-β levels only in the epidermis. Understanding how this aspect affects keratinocyte activities and their capacity to re-epithelialize might offer an opportunity to gain comprehensive knowledge of the involvement of TGF-β in chronic wounds. In this review, we compile existing evidence on the roles played by TGF-β during skin wound healing, with special emphasis on keratinocyte responses. Current limitations and future perspectives of TGF-β research in chronic wounds are discussed.
Collapse
|
50
|
Li J, Chen F, Peng Y, Lv Z, Lin X, Chen Z, Wang H. N6-Methyladenosine Regulates the Expression and Secretion of TGFβ1 to Affect the Epithelial-Mesenchymal Transition of Cancer Cells. Cells 2020; 9:cells9020296. [PMID: 31991845 PMCID: PMC7072279 DOI: 10.3390/cells9020296] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/14/2020] [Accepted: 01/19/2020] [Indexed: 02/07/2023] Open
Abstract
N6-methyladenosine (m6A) is the most abundant modification on eukaryotic mRNA, which regulates all steps of the mRNA life cycle. An increasing number of studies have shown that m6A methylation plays essential roles in tumor development. However, the relationship between m6A and the progression of cancers remains to be explored. Here, we reported that transforming growth factor-β (TGFβ1)-induced epithelial–mesenchymal transition (EMT) was inhibited in methyltransferase-like 3 (METTL3) knockdown (Mettl3Mut/−) cells. The expression of TGFβ1 was up-regulated, while self-stimulated expression of TGFβ1 was suppressed in Mettl3Mut/− cells. We further revealed that m6A promoted TGFB1 mRNA decay, but impaired TGFB1 translation progress. Besides this, the autocrine of TGFβ1 was disrupted in Mettl3Mut/− cells via interrupting TGFβ1 dimer formation. Lastly, we found that Snail, which was down-regulated in Mettl3Mut/− cells, was a key factor responding to TGFβ1-induced EMT. Together, our research demonstrated that m6A performed multi-functional roles in TGFβ1 expression and EMT modulation, suggesting the critical roles of m6A in cancer progression regulation.
Collapse
Affiliation(s)
- Jiexin Li
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
| | - Feng Chen
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
| | - Yanxi Peng
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
| | - Ziyan Lv
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
| | - Xinyao Lin
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
- Correspondence: (Z.C.); (H.W.); Tel.: +86-020-87343759 (Z.C.); +86-020-39943024 (H.W.)
| | - Hongsheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; (J.L.); (F.C.); (Y.P.); (Z.L.); (X.L.)
- Correspondence: (Z.C.); (H.W.); Tel.: +86-020-87343759 (Z.C.); +86-020-39943024 (H.W.)
| |
Collapse
|