1
|
Dorry S, Perla S, Bennett AM. Mitogen-Activated Protein Kinase Phosphatase-5 is Required for TGF-β Signaling Through a JNK-Dependent Pathway. Mol Cell Biol 2025; 45:17-31. [PMID: 39607740 PMCID: PMC11693473 DOI: 10.1080/10985549.2024.2426665] [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: 06/25/2024] [Revised: 10/31/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in tissue fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
2
|
Gould ML, Downes NJ, Woolley AG, Hussaini HM, Ratnayake JT, Ali MA, Friedlander LT, Cooper PR. Harnessing the Regenerative Potential of Purified Bovine Dental Pulp and Dentin Extracellular Matrices in a Chitosan/Alginate Hydrogel. Macromol Biosci 2024; 24:e2400254. [PMID: 38938070 DOI: 10.1002/mabi.202400254] [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: 05/28/2024] [Revised: 06/24/2024] [Indexed: 06/29/2024]
Abstract
When a tooth is diseased or damaged through caries, bioactive molecules are liberated from the pulp and dentin as part of the natural response to injury and these are key molecules for stimulating stem cell responses for tissue repair. Incorporation of these extracellular-matrix (ECM)-derived molecules into a hydrogel model can mimic in vivo conditions to enable dentin-pulp complex regeneration. Here, a chitosan/alginate (C/A) hydrogel is developed to sequester bovine ECM extracts. Human dental pulp cells (hDPCs) are cultured with these constructs and proliferation and cytotoxicity assays confirm that these C/A hydrogels are bioactive. Sequential z-axis fluorescent imaging visualizes hDPCs protruding into the hydrogel as it degraded. Alizarin red S staining shows that hDPCs cultured with the hydrogels display increased calcium-ion deposition, with dentin ECM stimulating the highest levels. Alkaline phosphatase activity is increased, as is expression of transforming growth factor-beta as demonstrated using immunocytochemistry. Directional analysis following phase contrast kinetic image capture demonstrates that both dentin and pulp ECM molecules act as chemoattractants for hDPCs. Data from this study demonstrate that purified ECM from dental pulp and dentin when delivered in a C/A hydrogel stimulates dental tissue repair processes in vitro.
Collapse
Affiliation(s)
- Maree L Gould
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Nerida J Downes
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Adele G Woolley
- Maurice Wilkins Centre for Biodiscovery, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Haizal M Hussaini
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
- Faculty of Dental Medicine, University of Airlangga, Surabaya, 60132, Indonesia
| | - Jithendra T Ratnayake
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Mohammad Azam Ali
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Lara T Friedlander
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| | - Paul R Cooper
- Faculty of Dentistry, Sir John Walsh Research Institute, University of Otago, P.O. Box 56, Dunedin, 9054, New Zealand
| |
Collapse
|
3
|
Zhang L, Wu M, Zhang J, Liu T, Fu S, Wang Y, Xu Z. The pivotal role of glucose transporter 1 in diabetic kidney disease. Life Sci 2024; 353:122932. [PMID: 39067659 DOI: 10.1016/j.lfs.2024.122932] [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: 04/05/2024] [Revised: 07/12/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024]
Abstract
Diabetes mellitus (DM) is a significant public health problem. Diabetic kidney disease (DKD) is the most common complication of DM, and its incidence has been increasing with the increasing prevalence of DM. Given the association between DKD and mortality in patients with DM, DKD is a significant burden on public health resources. Despite its significance in DM progression, the pathogenesis of DKD remains unclear. Aberrant glucose uptake by cells is an important pathophysiological mechanism underlying DKD renal injury. Glucose is transported across the bilayer cell membrane by a glucose transporter (GLUT) located on the cell membrane. Multiple GLUT proteins have been identified in the kidney, and GLUT1 is one of the most abundantly expressed isoforms. GLUT1 is a crucial regulator of intracellular glucose metabolism and plays a key pathological role in the phenotypic changes in DKD mesangial cells. In an attempt to understand the pathogenesis of DKD better, we here present a review of studies on the role of GLUT1 in the development and progression of DKD.
Collapse
Affiliation(s)
- Li Zhang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meiyan Wu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jizhou Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun 130021, China
| | - Tingting Liu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Shaojie Fu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yue Wang
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China
| | - Zhonggao Xu
- Department of Nephrology, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
4
|
Dorry S, Perla S, Bennett AM. MAPK Phosphatase-5 is required for TGF-β signaling through a JNK-dependent pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.600976. [PMID: 38979264 PMCID: PMC11230413 DOI: 10.1101/2024.06.27.600976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Mitogen-activated protein kinase (MAPK) phosphatases (MKPs) constitute members of the dual-specificity family of protein phosphatases that dephosphorylate the MAPKs. MKP-5 dephosphorylates the stress-responsive MAPKs, p38 MAPK and JNK, and has been shown to promote tissue fibrosis. Here, we provide insight into how MKP-5 regulates the transforming growth factor-β (TGF-β) pathway, a well-established driver of fibrosis. We show that MKP-5-deficient fibroblasts in response to TGF-β are impaired in SMAD2 phosphorylation at canonical and non-canonical sites, nuclear translocation, and transcriptional activation of fibrogenic genes. Consistent with this, pharmacological inhibition of MKP-5 is sufficient to block TGF-β signaling, and that this regulation occurs through a JNK-dependent pathway. By utilizing RNA sequencing and transcriptomic analysis, we identify TGF-β signaling activators regulated by MKP-5 in a JNK-dependent manner, providing mechanistic insight into how MKP-5 promotes TGF-β signaling. This study elucidates a novel mechanism whereby MKP-5-mediated JNK inactivation is required for TGF-β signaling and provides insight into the role of MKP-5 in fibrosis.
Collapse
Affiliation(s)
- Sam Dorry
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sravan Perla
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, Connecticut, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
5
|
Liu Y, Luo Z. Repurposing Anticancer Drugs Targeting the MAPK/ERK Signaling Pathway for the Treatment of Respiratory Virus Infections. Int J Mol Sci 2024; 25:6946. [PMID: 39000055 PMCID: PMC11240997 DOI: 10.3390/ijms25136946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Revised: 06/16/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Respiratory virus infections remain a significant challenge to human health and the social economy. The symptoms range from mild rhinitis and nasal congestion to severe lower respiratory tract dysfunction and even mortality. The efficacy of therapeutic drugs targeting respiratory viruses varies, depending upon infection time and the drug resistance engendered by a high frequency of viral genome mutations, necessitating the development of new strategies. The MAPK/ERK pathway that was well delineated in the 1980s represents a classical signaling cascade, essential for cell proliferation, survival, and differentiation. Since this pathway is constitutively activated in many cancers by oncogenes, several drugs inhibiting Raf/MEK/ERK have been developed and currently used in anticancer treatment. Two decades ago, it was reported that viruses such as HIV and influenza viruses could exploit the host cellular MAPK/ERK pathway for their replication. Thus, it would be feasible to repurpose this category of the pathway inhibitors for the treatment of respiratory viral infections. The advantage is that the host genes are not easy to mutate such that the drug resistance rarely occurs during short-period treatment of viruses. Therefore, in this review we will summarize the research progress on the role of the MAPK/ERK pathway in respiratory virus amplification and discuss the potential of the pathway inhibitors (MEK inhibitors) in the treatment of respiratory viral infections.
Collapse
Affiliation(s)
| | - Zhijun Luo
- Medical Department, Queen Mary School, Nanchang University, Nanchang 330031, China;
| |
Collapse
|
6
|
De Masi R, Orlando S, Carata E, Panzarini E. Ultrastructural Characterization of PBMCs and Extracellular Vesicles in Multiple Sclerosis: A Pilot Study. Int J Mol Sci 2024; 25:6867. [PMID: 38999977 PMCID: PMC11241448 DOI: 10.3390/ijms25136867] [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: 05/03/2024] [Revised: 06/10/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Growing evidence identifies extracellular vesicles (EVs) as important cell-to-cell signal transducers in autoimmune disorders, including multiple sclerosis (MS). If the etiology of MS still remains unknown, its molecular physiology has been well studied, indicating peripheral blood mononuclear cells (PBMCs) as the main pathologically relevant contributors to the disease and to neuroinflammation. Recently, several studies have suggested the involvement of EVs as key mediators of neuroimmune crosstalk in central nervous system (CNS) autoimmunity. To assess the role of EVs in MS, we applied electron microscopy (EM) techniques and Western blot analysis to study the morphology and content of plasma-derived EVs as well as the ultrastructure of PBMCs, considering four MS patients and four healthy controls. Through its exploratory nature, our study was able to detect significant differences between groups. Pseudopods and large vesicles were more numerous at the plasmalemma interface of cases, as were endoplasmic vesicles, resulting in an activated aspect of the PBMCs. Moreover, PBMCs from MS patients also showed an increased number of multivesicular bodies within the cytoplasm and amorphous material around the vesicles. In addition, we observed a high number of plasma-membrane-covered extensions, with multiple associated large vesicles and numerous autophagosomal vacuoles containing undigested cytoplasmic material. Finally, the study of EV cargo evidenced a number of dysregulated molecules in MS patients, including GANAB, IFI35, Cortactin, Septin 2, Cofilin 1, and ARHGDIA, that serve as inflammatory signals in a context of altered vesicular dynamics. We concluded that EM coupled with Western blot analysis applied to PBMCs and vesiculation can enhance our knowledge in the physiopathology of MS.
Collapse
Affiliation(s)
- Roberto De Masi
- Complex Operative Unit of Neurology, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Stefania Orlando
- Laboratory of Neuroproteomics, Multiple Sclerosis Centre, “F. Ferrari” Hospital, Casarano, 73042 Lecce, Italy
| | - Elisabetta Carata
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of the Salento, 73100 Lecce, Italy;
| | - Elisa Panzarini
- Department of Biological and Environmental Sciences and Technologies (Di.S.Te.B.A.), University of the Salento, 73100 Lecce, Italy;
| |
Collapse
|
7
|
Lin Y, Gahn J, Banerjee K, Dobreva G, Singhal M, Dubrac A, Ola R. Role of endothelial PDGFB in arterio-venous malformations pathogenesis. Angiogenesis 2024; 27:193-209. [PMID: 38070064 PMCID: PMC11021264 DOI: 10.1007/s10456-023-09900-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 11/05/2023] [Indexed: 04/17/2024]
Abstract
Arterial-venous malformations (AVMs) are direct connections between arteries and veins without an intervening capillary bed. Either familial inherited or sporadically occurring, localized pericytes (PCs) drop is among the AVMs' hallmarks. Whether impaired PC coverage triggers AVMs or it is a secondary event is unclear. Here we evaluated the role of the master regulator of PC recruitment, Platelet derived growth factor B (PDGFB) in AVM pathogenesis. Using tamoxifen-inducible deletion of Pdgfb in endothelial cells (ECs), we show that disruption of EC Pdgfb-mediated PC recruitment and maintenance leads to capillary enlargement and organotypic AVM-like structures. These vascular lesions contain non-proliferative hyperplastic, hypertrophic and miss-oriented capillary ECs with an altered capillary EC fate identity. Mechanistically, we propose that PDGFB maintains capillary EC size and caliber to limit hemodynamic changes, thus restricting expression of Krüppel like factor 4 and activation of Bone morphogenic protein, Transforming growth factor β and NOTCH signaling in ECs. Furthermore, our study emphasizes that inducing or activating PDGFB signaling may be a viable therapeutic approach for treating vascular malformations.
Collapse
Affiliation(s)
- Yanzhu Lin
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johannes Gahn
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Kuheli Banerjee
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- German Centre for Cardiovascular Research (DZHK), Heidelberg, Germany
| | - Mahak Singhal
- Laboratory of AngioRhythms, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Alexandre Dubrac
- Centre de Recherche, CHU St. Justine, Montreal, QC, H3T 1C5, Canada
- Département de Pathologie et Biologie Cellulaire, Université de Montréal, Montreal, QC, H3T 1J4, Canada
| | - Roxana Ola
- Experimental Pharmacology Mannheim (EPM), European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
8
|
Cao M, Fan B, Zhen T, Das A, Wang J. Ruthenium biochanin-A complex ameliorates lung carcinoma through the downregulation of the TGF-β/PPARγ/PI3K/TNF-α pathway in association with caspase-3-mediated apoptosis. Toxicol Res 2023; 39:455-475. [PMID: 37398567 PMCID: PMC10313601 DOI: 10.1007/s43188-023-00177-1] [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: 12/10/2022] [Revised: 03/08/2023] [Accepted: 03/23/2023] [Indexed: 07/04/2023] Open
Abstract
Lung cancer is the most often reported cancer with a terrible prognosis worldwide. Flavonoid metal complexes have exhibited potential chemotherapeutic effects with substantially low adverse effects. This study investigated the chemotherapeutic effect of the ruthenium biochanin-A complex on lung carcinoma in both in vitro and in vivo model systems. The synthesized organometallic complex was characterized via UV‒visible spectroscopy, FTIR, mass spectrometry, and scanning electron microscopy. Moreover, the DNA binding activity of the complex was determined. The in vitro chemotherapeutic assessment was performed on the A549 cell line through MTT assay, flow cytometry, and western blot analysis. An in vivo toxicity study was performed to determine the chemotherapeutic dose of the complex, and subsequently, chemotherapeutic activity was assessed in benzo-α-pyrene-induced lung cancer mouse model by evaluating the histopathology, immunohistochemistry, and TUNEL assays. The IC50 value of the complex in A549 cells was found to be 20 µM. The complex demonstrated significant apoptosis induction, enhanced caspase-3 expression and cell cycle arrest with downregulated PI3K, PPARγ, TGF-β, and TNF-α expression in A549 cells. The in vivo study suggested that ruthenium biochanin-A therapy restored the morphological architecture of lung tissue in a benzo-α-pyrene-induced lung cancer model and inhibited the expression of Bcl2. Additionally, increased apoptotic events were identified with upregulation of caspase-3 and p53 expression. In conclusion, the ruthenium biochanin-A complex successfully amelioratedlung cancer incidence in both in vitro and in vivo models through the alteration of the TGF-β/PPARγ/PI3K/TNF-α axis with the induction of the p53/caspase-3-mediated apoptotic pathway.
Collapse
Affiliation(s)
- Ming Cao
- Department of Thoracic Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan, 250014 Shandong Province China
| | - Bo Fan
- Department of Thoracic Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan, 250014 Shandong Province China
| | - Tianchang Zhen
- Department of Thoracic Surgery, The First Hospital Affiliated with Shandong First Medical University, Jinan, 250014 Shandong Province China
| | - Abhijit Das
- Department of Pharmacology, NSHM Knowledge Campus, Kolkata- Group of Institutions, 124 B.L. Saha Road, Kolkata, West Bengal 700053 India
| | - Junling Wang
- Department of Respiratory and Critical Care, The First Hospital Affiliated with Shandong First Medical University, No.16766, Lixia District, Jingshi Road, Jinan, 250014 Shandong Province China
| |
Collapse
|
9
|
Therapeutic or lifelong training effects on pancreatic morphological and functional parameters in an animal model of aging and obesity. Exp Gerontol 2023; 175:112144. [PMID: 36907475 DOI: 10.1016/j.exger.2023.112144] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 03/02/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023]
Abstract
AIMS Obesity, aging, and physical training are factors influencing pancreatic functional and morphological parameters. Aiming to clarify the impact of the interaction of these factors, we analyzed the effect of therapeutic or lifelong physical training on body adiposity and pancreatic functional and morphological parameters of aged and obese rats. METHODS 24 male Wistar rats were (initial age = 4 months and final age = 14 months) randomly divided into three aged and obese experimental groups (n = 8/group): untrained, therapeutic trained, and lifelong trained. Body adiposity, plasmatic concentration and pancreatic immunostaining of insulin, markers of tissue inflammation, lipid peroxidation, activity and immunostaining of antioxidant enzymes, and parameters of pancreatic morphology were evaluated. RESULTS Lifelong physical training improved the body adiposity, plasmatic insulin concentration, and macrophage immunostaining in the pancreas. The animals submitted to therapeutic and lifelong training showed an increase in the density of the pancreatic islets; lower insulin, Nuclear Factor Kappa B (NF-κB), and Transforming Growth Factor beta (TGF-β) immunostaining in the pancreatic parenchyma, as well as lower pancreatic tissue lipid peroxidation, lower fibrosis area, increased catalase and glutathione peroxidase (GPx) activity and increased heme oxygenase-1 (HO-1) immunostaining, with the greatest effect in the lifelong training group. CONCLUSION Lifelong training promoted greater beneficial effects on the pancreatic functional and morphological parameters of aged and obese animals compared to therapeutic exercise.
Collapse
|
10
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
11
|
Wang Y, Liu P, Chen X, Yang W. Circ_CHMP5 aggravates oxidized low-density lipoprotein-induced damage to human umbilical vein endothelial cells through miR-516b-5p/TGFβR2 axis. Clin Hemorheol Microcirc 2023; 85:325-339. [PMID: 37212088 DOI: 10.3233/ch-231722] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
BACKGROUND Atherosclerosis (AS) was one of the main causes of death in the elderly, and lesions in human umbilical vein endothelial cells (HUVECs) could lead to AS. CircRNA-charged multivesicular body protein 5 (circ_CHMP5) was reported to participate in the progression of AS. METHODS Quantitative real-time polymerase chain reaction (qRT-PCR) was used to analyze the levels of circ_CHMP5, miR-516b-5p, and transforming growth factor beta receptor 2 (TGFβR2) in AS patients or ox-LDL-induced HUVECs. 5-Ethynyl-2'-deoxyuridine and cell counting kit-8 assays were performed to detect cell proliferation. Proteins expression was assessed by western blot assay. Cell apoptosis was examined by flow cytometry. Tube formation assay was utilized to measure the tube formation ability of HUVCEs. The targeting relationships between miR-516b-5p and circ_CHMP5 or TGFβR2 were confirmed by dual-luciferase reporter assay and RNA-pull down assay. RESULTS Circ_CHMP5 was enhanced in the serum of AS patients and ox-LDL-exposure HUVECs. Ox-LDL blocked proliferation and tube formation of HUVECs and induced cell apoptosis, and circ_CHMP5 knockdown reversed these effects. In addition, circ_CHMP5 regulated the growth of ox-LDL-induced HUVECs through miR-516b-5p and TGFβR2. Moreover, the effects of circ_CHMP5 knockdown on ox-LDL-induced HUVECs were obviously recovered by downregulation of miR-516b-5p, and overexpression of TGFβR2 restored the effects of miR-516b-5p upregulation on ox-LDL-stimulated HUVECs. CONCLUSION Silence of circ_CHMP5 overturned ox-LDL-treated inhibition of HUVECs proliferation and angiogenesis by miR-516b-5p and TGFβR2. These results provided new solutions for the treatment of AS.
Collapse
Affiliation(s)
- Yueru Wang
- Department of Internal Medicine-Cardiovascular, Shanxi Provincial People's Hospital, Taiyuan City, Shanxi, China
| | - Ping Liu
- Shanxi Provincial Medical Service Evaluation Center, Taiyuan City, Shanxi, China
| | - Xiaoyan Chen
- Department of Ultrasound, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
| | - Wuxiao Yang
- Department of Cardiology, Shanxi Provincial People's Hospital, Taiyuan City, Shanxi, China
| |
Collapse
|
12
|
Maruszewska-Cheruiyot M, Stear MJ, Machcińska M, Donskow-Łysoniewska K. Importance of TGFβ in Cancer and Nematode Infection and Their Interaction-Opinion. Biomolecules 2022; 12:1572. [PMID: 36358922 PMCID: PMC9687433 DOI: 10.3390/biom12111572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/20/2022] [Accepted: 10/24/2022] [Indexed: 09/29/2023] Open
Abstract
Historically, there has been little interaction between parasitologists and oncologists, although some helminth infections predispose to the development of tumours. In addition, both parasites and tumours need to survive immune attack. Recent research suggests that both tumours and parasites suppress the immune response to increase their chances of survival. They both co-opt the transforming growth factor beta (TGFβ) signalling pathway to modulate the immune response to their benefit. In particular, there is concern that suppression of the immune response by nematodes and their products could enhance susceptibility to tumours in both natural and artificial infections.
Collapse
Affiliation(s)
| | - Michael James Stear
- Department of Animal, Plant and Soil Science, Agribio, La Trobe University, Bundoora 3086, Australia
| | - Maja Machcińska
- Department of Experimental Immunotherapy, Faculty of Medicine, Lazarski University, 02-662 Warsaw, Poland
| | | |
Collapse
|
13
|
Hachana S, Larrivée B. TGF-β Superfamily Signaling in the Eye: Implications for Ocular Pathologies. Cells 2022; 11:2336. [PMID: 35954181 PMCID: PMC9367584 DOI: 10.3390/cells11152336] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/06/2023] Open
Abstract
The TGF-β signaling pathway plays a crucial role in several key aspects of development and tissue homeostasis. TGF-β ligands and their mediators have been shown to be important regulators of ocular physiology and their dysregulation has been described in several eye pathologies. TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis. Inadequate TGF-β signaling has been associated with defective angiogenesis, vascular barrier function, unfavorable inflammatory responses, and tissue fibrosis. In addition, experimental models of corneal neovascularization, diabetic retinopathy, proliferative vitreoretinopathy, glaucoma, or corneal injury suggest that aberrant TGF-β signaling may contribute to the pathological features of these conditions, showing the potential of modulating TGF-β signaling to treat eye diseases. This review highlights the key roles of TGF-β family members in ocular physiology and in eye diseases, and reviews approaches targeting the TGF-β signaling as potential treatment options.
Collapse
Affiliation(s)
- Soumaya Hachana
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| | - Bruno Larrivée
- Maisonneuve-Rosemont Hospital Research Center, Montreal, QC H1T 2M4, Canada
- Department of Ophthalmology, Université de Montréal, Montreal, QC H3C 3J7, Canada
| |
Collapse
|
14
|
Baba AB, Rah B, Bhat GR, Mushtaq I, Parveen S, Hassan R, Hameed Zargar M, Afroze D. Transforming Growth Factor-Beta (TGF-β) Signaling in Cancer-A Betrayal Within. Front Pharmacol 2022; 13:791272. [PMID: 35295334 PMCID: PMC8918694 DOI: 10.3389/fphar.2022.791272] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 02/09/2022] [Indexed: 12/11/2022] Open
Abstract
A ubiquitously expressed cytokine, transforming growth factor-beta (TGF-β) plays a significant role in various ongoing cellular mechanisms. The gain or loss-of-function of TGF-β and its downstream mediators could lead to a plethora of diseases includes tumorigenesis. Specifically, at the early onset of malignancy TGF-β act as tumour suppressor and plays a key role in clearing malignant cells by reducing the cellular proliferation and differentiation thus triggers the process of apoptosis. Subsequently, TGF-β at an advanced stage of malignancy promotes tumorigenesis by augmenting cellular transformation, epithelial-mesenchymal-transition invasion, and metastasis. Besides playing the dual roles, depending upon the stage of malignancy, TGF-β also regulates cell fate through immune and stroma components. This oscillatory role of TGF-β to fight against cancer or act as a traitor to collaborate and crosstalk with other tumorigenic signaling pathways and its betrayal within the cell depends upon the cellular context. Therefore, the current review highlights and understands the dual role of TGF-β under different cellular conditions and its crosstalk with other signaling pathways in modulating cell fate.
Collapse
|
15
|
Patnaik S, Sahoo L, Mohanty M, Bit A, Meher PK, Das S, Jayasankar P, Saha JN, Das P. Activin receptor type IIB in rohu (Labeo rohita): molecular characterization, tissue distribution and immunohistochemical localization during different stages of gonadal maturation. FISH PHYSIOLOGY AND BIOCHEMISTRY 2021; 47:1353-1367. [PMID: 34273063 DOI: 10.1007/s10695-021-00973-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/26/2021] [Indexed: 06/13/2023]
Abstract
Activin receptor type IIB (ActRIIB) is a transmembrane serine/threonine kinase receptor which plays a pivotal role in regulating the reproduction in vertebrates including teleost. Earlier studies have documented its importance in governing gonadal maturation in higher vertebrates. However, reports on the regulation of fish reproductive system by ActRIIB gene are still limited. Here, we report the identification and characterization of ActRIIB cDNA of Labeo rohita, a commercially important fish species of the Indian subcontinent. The full-length gene encoding rohu ActRIIB was cloned and found to be of 1674 bp in length. Functional similarities were evident from evolutionary analysis across vertebrates. Real-time PCR to measure the expression of ActRIIB transcript in rohu revealed significant mRNA levels in gonads followed by non-reproductive tissues, including the brain, pituitary and muscle. With respect to different gonadal maturation stages, predominant expression of ActRIIB mRNA was observed during the pre-spawning phase of both sexes. To further delineate its role in rohu reproduction, a recombinant protein of the extracellular domain of ActRIIB (rECD-ActRIIB) was produced, and polyclonal antibody is raised against the protein for its immuno-localization studies during different gonadal maturation stages. Strong immunoreactivity was noticed in the pre-vitellogenic oocytes which decreased dramatically in the fully mature oocytes. Similarly, the strong and intense immunoreactivity was found in the spermatids and spermatocytes of the immature testis, and eventually the intensity reduced with the progression of the maturation stage. These results provide the first evidence of the presence of ActRIIB in rohu gonadal tissues. Taken together, our observations lay the groundwork for further understanding and investigating on the potential role of ActRIIB in fish reproduction system in the event of gonadal maturation.
Collapse
Affiliation(s)
- Siddhi Patnaik
- Department of Cancer and Stem Cell Biology, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Lakshman Sahoo
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | - Mausumee Mohanty
- Barcode Biosciences, Dr. Shivaram Karanth Nagar, Bengaluru, 560077, India
| | - Amrita Bit
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | - Prem Kumar Meher
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | - Sachidananda Das
- PG Department of Zoology, Utkal University, Bhubaneswar, 751004, Odisha, India
| | - Pallipuram Jayasankar
- Marine Biotechnology Division, ICAR-Central Marine Fisheries Research Institute, Kochi, 682018, India
| | - Jatindra Nath Saha
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India
| | - Paramananda Das
- Fish Genetics and Biotechnology Division, ICAR-Central Institute of Freshwater Aquaculture, Kausalyaganga, Bhubaneswar, 751002, Odisha, India.
| |
Collapse
|
16
|
Kumari A, Shonibare Z, Monavarian M, Arend RC, Lee NY, Inman GJ, Mythreye K. TGFβ signaling networks in ovarian cancer progression and plasticity. Clin Exp Metastasis 2021; 38:139-161. [PMID: 33590419 PMCID: PMC7987693 DOI: 10.1007/s10585-021-10077-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 02/03/2021] [Indexed: 02/06/2023]
Abstract
Epithelial ovarian cancer (EOC) is a leading cause of cancer-related death in women. Late-stage diagnosis with significant tumor burden, accompanied by recurrence and chemotherapy resistance, contributes to this poor prognosis. These morbidities are known to be tied to events associated with epithelial-mesenchymal transition (EMT) in cancer. During EMT, localized tumor cells alter their polarity, cell-cell junctions, cell-matrix interactions, acquire motility and invasiveness and an exaggerated potential for metastatic spread. Key triggers for EMT include the Transforming Growth Factor-β (TGFβ) family of growth factors which are actively produced by a wide array of cell types within a specific tumor and metastatic environment. Although TGFβ can act as either a tumor suppressor or promoter in cancer, TGFβ exhibits its pro-tumorigenic functions at least in part via EMT. TGFβ regulates EMT both at the transcriptional and post-transcriptional levels as outlined here. Despite recent advances in TGFβ based therapeutics, limited progress has been seen for ovarian cancers that are in much need of new therapeutic strategies. Here, we summarize and discuss several recent insights into the underlying signaling mechanisms of the TGFβ isoforms in EMT in the unique metastatic environment of EOCs and the current therapeutic interventions that may be relevant.
Collapse
Affiliation(s)
- Asha Kumari
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, WTI 320B, 1824 Sixth Avenue South, Birmingham, AL, 35294, USA
| | - Zainab Shonibare
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, WTI 320B, 1824 Sixth Avenue South, Birmingham, AL, 35294, USA
| | - Mehri Monavarian
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, WTI 320B, 1824 Sixth Avenue South, Birmingham, AL, 35294, USA
| | - Rebecca C Arend
- Department of Obstetrics and Gynecology-Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Nam Y Lee
- Division of Pharmacology, Chemistry and Biochemistry, College of Medicine, University of Arizona, Tucson, AZ, 85721, USA
| | - Gareth J Inman
- Cancer Research UK Beatson Institute and Institute of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Karthikeyan Mythreye
- Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at Birmingham, WTI 320B, 1824 Sixth Avenue South, Birmingham, AL, 35294, USA.
| |
Collapse
|
17
|
The Overexpression of Keratin 23 Promotes Migration of Ovarian Cancer via Epithelial-Mesenchymal Transition. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8218735. [PMID: 33204716 PMCID: PMC7652601 DOI: 10.1155/2020/8218735] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 09/28/2020] [Accepted: 10/14/2020] [Indexed: 01/13/2023]
Abstract
Background Keratin 23 (KRT23) is a new member of the KRT gene family and known to be involved in the development and migration of various types of tumors. However, the role of KRT23 in ovarian cancer (OC) remains unclear. This study is aimed at investigating the function of KRT23 in OC. Methods The expression of KRT23 in normal ovarian and OC tissues was determined using the Oncomine database and immunohistochemical staining. Reverse transcription quantitative polymerase chain reaction assay was used to analyze the expression of KRT23 in normal ovarian epithelial cell lines and OC cell lines. Small interfering RNA (siRNA), wound healing assay, and transwell assay were conducted to detect the effects of KRT23 on OC cell migration and invasion. Further mechanistic studies were verified by the Gene Expression Profiling Interactive Analysis platform, Western blotting, and immunofluorescence staining. Results KRT23 was highly expressed in OC tissues and cell lines. High KRT23 expression could regulate OC cell migration and invasion, and the reduction of KRT23 by siRNA inhibited the migration and invasion of OC cells in vitro. Furthermore, KRT23 mediated epithelial-mesenchymal transition (EMT) by regulating p-Smad2/3 levels in the TGF-β/Smad signaling pathway. Conclusions These results demonstrate that KRT23 plays an important role in OC migration via EMT by regulating the TGF-β/Smad signaling pathway.
Collapse
|
18
|
Moein S, Moradzadeh K, Javanmard SH, Nasiri SM, Gheisari Y. In vitro versus in vivo models of kidney fibrosis: Time-course experimental design is crucial to avoid misinterpretations of gene expression data. JOURNAL OF RESEARCH IN MEDICAL SCIENCES 2020; 25:84. [PMID: 33273929 PMCID: PMC7698384 DOI: 10.4103/jrms.jrms_906_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 04/11/2020] [Accepted: 05/14/2020] [Indexed: 11/22/2022]
Abstract
Background: In vitro models are common tools in nephrology research. However, their validity has rarely been scrutinized. Materials and Methods: Considering the critical role of transforming growth factor (TGF)-β and hypoxia pathways in kidney fibrosis, kidney-derived cells were exposed to TGF-β and/or hypoxic conditions and the expression levels of some genes related to these two signaling pathways were quantified in a time-course manner. Furthermore, a unilateral ureteral obstruction mouse model was generated, and the expressions of the same genes were assessed. Results: In all in vitro experimental groups, the expression of the genes was noisy with no consistent pattern. However, in the animal model, TGF-β pathway-related genes demonstrated considerable overexpression in the ureteral obstruction group compared with the sham controls. Interestingly, hypoxia pathway genes had prominent fluctuations with very similar patterns in both animal groups, suggesting a periodical pattern not affected by the intervention. Conclusion: The findings of this study suggest that in vitro findings should be interpreted cautiously and if possible are substituted or supported by animal models that are more consistent and reliable. Furthermore, we underscore the importance of time-course evaluation of both case and control groups in gene expression studies to avoid misconceptions caused by gene expression noise or intrinsic rhythms.
Collapse
Affiliation(s)
- Shiva Moein
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Kobra Moradzadeh
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Seyed Mahdi Nasiri
- Department of Clinical Pathology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Yousof Gheisari
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran.,Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
19
|
Silva TA, Ferreira LFDC, Pereira MCDS, Calvet CM. Differential Role of TGF-β in Extracellular Matrix Regulation During Trypanosoma cruzi-Host Cell Interaction. Int J Mol Sci 2019; 20:E4836. [PMID: 31569452 PMCID: PMC6801917 DOI: 10.3390/ijms20194836] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 08/03/2019] [Accepted: 08/07/2019] [Indexed: 12/19/2022] Open
Abstract
Transforming growth factor beta (TGF-β) is a determinant for inflammation and fibrosis in cardiac and skeletal muscle in Chagas disease. To determine its regulatory mechanisms, we investigated the response of Trypanosoma cruzi-infected cardiomyocytes (CM), cardiac fibroblasts (CF), and L6E9 skeletal myoblasts to TGF-β. Cultures of CM, CF, and L6E9 were infected with T. cruzi (Y strain) and treated with TGF-β (1-10 ng/mL, 1 h or 48 h). Fibronectin (FN) distribution was analyzed by immunofluorescence and Western blot (WB). Phosphorylated SMAD2 (PS2), phospho-p38 (p-p38), and phospho-c-Jun (p-c-Jun) signaling were evaluated by WB. CF and L6E9 showed an increase in FN from 1 ng/mL of TGF-β, while CM displayed FN modulation only after 10 ng/mL treatment. CF and L6E9 showed higher PS2 levels than CM, while p38 was less stimulated in CF than CM and L6E9. T. cruzi infection resulted in localized FN disorganization in CF and L6E9. T. cruzi induced an increase in FN in CF cultures, mainly in uninfected cells. Infected CF cultures treated with TGF-β showed a reduction in PS2 and an increase in p-p38 and p-c-Jun levels. Our data suggest that p38 and c-Jun pathways may be participating in the fibrosis regulatory process mediated by TGF-β after T. cruzi infection.
Collapse
Affiliation(s)
- Tatiana Araújo Silva
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| | | | | | - Claudia Magalhães Calvet
- Cellular Ultrastructure Laboratory, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
20
|
Gu L, Saha ST, Thomas J, Kaur M. Targeting cellular cholesterol for anticancer therapy. FEBS J 2019; 286:4192-4208. [DOI: 10.1111/febs.15018] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/30/2019] [Accepted: 07/24/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Liang Gu
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Sourav Taru Saha
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Jodie Thomas
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| | - Mandeep Kaur
- School of Molecular and Cell Biology University of the Witwatersrand Johannesburg South Africa
| |
Collapse
|
21
|
Xue K, Zhang J, Li C, Li J, Wang C, Zhang Q, Chen X, Yu X, Sun L, Yu X. The role and mechanism of transforming growth factor beta 3 in human myocardial infarction-induced myocardial fibrosis. J Cell Mol Med 2019; 23:4229-4243. [PMID: 30983140 PMCID: PMC6533491 DOI: 10.1111/jcmm.14313] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 02/11/2019] [Accepted: 03/04/2019] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor beta (TGFβ) plays a crucial role in tissue fibrosis. A number of studies have shown that TGFβ3 significantly attenuated tissue fibrosis. However, the mechanism involved in this effect is poorly understood. In this study we found that the expression level of TGFβ3 was higher in human myocardial infarction (MI) tissues than in normal tissues, and interestingly, it increased with the development of fibrosis post‐myocardial infarction (post‐MI). In vitro, human cardiac fibroblasts (CFs) were incubated with angiotensin II (Ang II) to mimic the ischaemic myocardium microenvironment and used to investigate the anti‐fibrotic mechanism of TGFβ3. Then, fibrosis‐related proteins were detected by Western blot. It was revealed that TGFβ3 up‐regulation attenuated the proliferation, migration of human CFs and the expression of collagens, which are the main contributors to fibrosis, promoted the phenotype shift and the cross‐linking of collagens. Importantly, the expression of collagens was higher in the si‐smad7 groups than in the control groups, while silencing smad7 increased the phosphorylation level of the TGFβ/smad signalling pathway. Collectively, these results indicated that TGFβ3 inhibited fibrosis via the TGFβ/smad signalling pathway, possibly attributable to the regulation of smad7, and that TGFβ3 might serve as a potential therapeutic target for myocardial fibrosis post‐MI.
Collapse
Affiliation(s)
- Ke Xue
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jun Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Cong Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Jing Li
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Cong Wang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Qingqing Zhang
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xianlu Chen
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiaotang Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Lei Sun
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| | - Xiao Yu
- Department of Pathology and Forensic Medicine, College of Basic Medical Sciences, Dalian Medical University, Dalian, China
| |
Collapse
|
22
|
Sun L, Xiu M, Wang S, Brigstock DR, Li H, Qu L, Gao R. Lipopolysaccharide enhances TGF-β1 signalling pathway and rat pancreatic fibrosis. J Cell Mol Med 2018; 22:2346-2356. [PMID: 29424488 PMCID: PMC5867168 DOI: 10.1111/jcmm.13526] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 12/08/2017] [Indexed: 12/20/2022] Open
Abstract
Pancreatic stellate cells (PSCs) play a critical role in fibrogenesis during alcoholic chronic pancreatitis (ACP). Transforming growth factor-beta1 (TGF-β1) is a key regulator of extracellular matrix production and PSC activation. Endotoxin lipopolysaccharide (LPS) has been recognized as a trigger factor in the pathogenesis of ACP. This study aimed to investigate the mechanisms by which LPS modulates TGF-β1 signalling and pancreatic fibrosis. Sprague-Dawley rats fed with a Lieber-DeCarli alcohol (ALC) liquid diet for 10 weeks with or without LPS challenge during the last 3 weeks. In vitro studies were performed using rat macrophages (Mφs) and PSCs (RP-2 cell line). The results showed that repeated LPS challenge resulted in significantly more collagen production and PSC activation compared to rats fed with ALC alone. LPS administration caused overexpression of pancreatic TLR4 or TGF-β1 which was paralleled by an increased number of TLR4-positive or TGF-β1-positive Mφs or PSCs in ALC-fed rats. In vitro, TLR4 or TGF-β1 production in Mφs or RP-2 cells was up-regulated by LPS. LPS alone or in combination with TGF-β1 significantly increased type I collagen and α-SMA production and Smad2 and 3 phosphorylation in serum-starved RP-2 cells. TGF-β pseudoreceptor BAMBI production was repressed by LPS, which was antagonized by Si-TLR4 RNA or by inhibitors of MyD88/NF-kB. Additionally, knockdown of Bambi with Si-Bambi RNA significantly increased TGF-β1 signalling in RP-2 cells. These findings indicate that LPS increases TGF-β1 production through paracrine and autocrine mechanisms and that LPS enhances TGF-β1 signalling in PSCs by repressing BAMBI via TLR4/MyD88/NF-kB activation.
Collapse
Affiliation(s)
- Li Sun
- Department of Hepatic Biliary Pancreatic MedicineFirst Hospital of Jilin UniversityChangchunChina
| | - Ming Xiu
- Department of Hepatic Biliary Pancreatic MedicineFirst Hospital of Jilin UniversityChangchunChina
| | - Shuhua Wang
- Department of Surgical GastroenterologyFirst Hospital of Jilin UniversityChangchunChina
| | | | - Hongyan Li
- Department of Hepatic Biliary Pancreatic MedicineFirst Hospital of Jilin UniversityChangchunChina
| | - Limei Qu
- Department of Hepatic Biliary Pancreatic MedicineFirst Hospital of Jilin UniversityChangchunChina
| | - Runping Gao
- Department of Hepatic Biliary Pancreatic MedicineFirst Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
23
|
Çağlar HO, Yılmaz Süslüer S, Kavaklı Ş, Gündüz C, Ertürk B, Özkınay F, Haydaroğlu A. Meme kanseri kök hücrelerinde elajik asit ile indüklenmiş miRNA’ların ifadesi ve elajik asidin apoptoz üzerine etkisi. EGE TIP DERGISI 2018. [DOI: 10.19161/etd.399234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
|
24
|
Serralheiro P, Soares A, Costa Almeida CM, Verde I. TGF-β1 in Vascular Wall Pathology: Unraveling Chronic Venous Insufficiency Pathophysiology. Int J Mol Sci 2017; 18:E2534. [PMID: 29186866 PMCID: PMC5751137 DOI: 10.3390/ijms18122534] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Revised: 11/21/2017] [Accepted: 11/22/2017] [Indexed: 12/21/2022] Open
Abstract
Chronic venous insufficiency and varicose veins occur commonly in affluent countries and are a socioeconomic burden. However, there remains a relative lack of knowledge about venous pathophysiology. Various theories have been suggested, yet the molecular sequence of events is poorly understood. Transforming growth factor-beta one (TGF-β1) is a highly complex polypeptide with multifunctional properties that has an active role during embryonic development, in adult organ physiology and in the pathophysiology of major diseases, including cancer and various autoimmune, fibrotic and cardiovascular diseases. Therefore, an emphasis on understanding its signaling pathways (and possible disruptions) will be an essential requirement for a better comprehension and management of specific diseases. This review aims at shedding more light on venous pathophysiology by describing the TGF-β1 structure, function, activation and signaling, and providing an overview of how this growth factor and disturbances in its signaling pathway may contribute to specific pathological processes concerning the vessel wall which, in turn, may have a role in chronic venous insufficiency.
Collapse
Affiliation(s)
- Pedro Serralheiro
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| | - Andreia Soares
- Norfolk and Norwich University Hospital, Colney Ln, Norwich NR47UY, UK.
| | - Carlos M Costa Almeida
- Department of General Surgery (C), Coimbra University Hospital Centre, Portugal; Faculty of Medicine, University of Coimbra, Praceta Prof. Mota Pinto, 3000-075 Coimbra, Portugal.
| | - Ignacio Verde
- Faculty of Health Sciences, CICS-UBI-Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6201-506 Covilhã, Portugal.
| |
Collapse
|
25
|
Shao JB, Gao ZM, Huang WY, Lu ZB. The mechanism of epithelial-mesenchymal transition induced by TGF-β1 in neuroblastoma cells. Int J Oncol 2017; 50:1623-1633. [PMID: 28393230 PMCID: PMC5403264 DOI: 10.3892/ijo.2017.3954] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/22/2017] [Indexed: 12/12/2022] Open
Abstract
Neuroblastoma is the second most common extracranial malignant solid tumor that occurs in childhood, and metastasis is one of the major causes of death in neuroblastoma patients. The epithelial-mesenchymal transition (EMT) is an important mechanism for both the initiation of tumor invasion and subsequent metastasis. Therefore, this study investigated the mechanism by which transforming growth factor (TGF)-β1 induces EMT in human neuroblastoma cells. Using quantitative RT-qPCR and western blot analyses, we found that the mRNA and protein expression levels of E-cadherin were significantly decreased, whereas that of α-SMA was significantly increased after neuroblastoma cells were treated with different concentrations of TGF-β1. A scratch test and Transwell migration assay revealed that cell migration significantly and directly correlated with the concentration of TGF-β1 indicating that TGF-β1 induced EMT in neuroblastoma cells and led to their migration. Inhibiting Smad2/3 expression did not affect the expression of the key molecules involved in EMT. Further investigation found that the expression of the glioblastoma transcription factor (Gli) significantly increased in TGF-β1-stimulated neuroblastoma cells undergoing EMT, accordingly, interfering with Gli1/2 expression inhibited TGF-β1-induced EMT in neuroblastoma cells. GANT61, which is a targeted inhibitor of Gli1 and Gli2, decreased cell viability and promoted cell apoptosis. Thus, TGF-β1 induced EMT in neuroblastoma cells to increase their migration. Specifically, EMT induced by TGF-β1 in neuroblastoma cells did not depend on the Smad signaling pathway, and the transcription factor Gli participated in TGF-β1-induced EMT independent of Smad signaling.
Collapse
Affiliation(s)
- Jing-Bo Shao
- Department of Hematology/Oncology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200040, P.R. China
| | - Zhi-Mei Gao
- Department of Central Laboratory, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200040, P.R. China
| | - Wen-Yan Huang
- Department of Nephrology, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200040, P.R. China
| | - Zhi-Bao Lu
- Department of General Surgery, Shanghai Children's Hospital, Shanghai Jiaotong University, Shanghai 200040, P.R. China
| |
Collapse
|
26
|
Zhang K, Wang YW, Ma R. Bioinformatics analysis of dysregulated microRNAs in the nipple discharge of patients with breast cancer. Oncol Lett 2017; 13:3100-3108. [PMID: 28521415 PMCID: PMC5431374 DOI: 10.3892/ol.2017.5801] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2015] [Accepted: 01/13/2017] [Indexed: 01/18/2023] Open
Abstract
MicroRNAs (miRNAs/miRs) have been reported to be associated with the tumorigenesis and progression of various types of human cancer; however, the underlying mechanisms of this association remain unclear. The aim of the present study was to explore the potential functions of miRNAs in the development of breast cancer using bioinformatics analysis, based on the miRNA expression profile in nipple discharge. A previous study demonstrated the upregulation of miR-3646 and miR-4484, and the downregulation of miR-4732-5p in the nipple discharge of patients with breast cancer, compared with patients with benign breast lesions. In the present study, the target genes of miR-3646, −4484 and −4732-5p were predicted using TargetScan and the MicroRNA Target Prediction and Functional Study Database. The predicted target genes were further analyzed by Gene Ontology term enrichment analysis, Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis and protein-protein interaction analysis. Numerous carcinoma-associated genes, including ADIPOQ, CPEB1, DNAJB4, EIF4E, APP and BCLAF1, were revealed to be putative targets of miR-3646, −4484 and −4732-5p. Bioinformatics analysis associated miR-3646 with the Rap1 and TGF-β signaling pathways, miR-4484 with the ErbB, estrogen and focal adhesion signaling pathways, and miR-4732-5p with the proteoglycan signaling pathway. Notably, protein-protein interaction analysis identified that numerous predicted targets of these miRNAs were associated with one other. In addition, the target genes of the miRNAs were identified to be under the regulation of a number of transcription factors (TFs). The predicted target genes of miR-3646, −4484 and −4732-5p were identified to serve a role in cancer-associated signaling pathways and TF-mRNA networks, indicating that they serve a role in breast carcinogenesis and progression. These results provide a comprehensive view of the functions and molecular mechanisms of miR-3646, −4484 and −4732-5p, and will aid in future studies.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Ya-Wen Wang
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Rong Ma
- Department of Breast Surgery, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
27
|
Molecular mechanism of Antrodia cinnamomea sulfated polysaccharide on the suppression of lung cancer cell growth and migration via induction of transforming growth factor β receptor degradation. Int J Biol Macromol 2017; 95:1144-1152. [DOI: 10.1016/j.ijbiomac.2016.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/26/2016] [Accepted: 11/02/2016] [Indexed: 12/28/2022]
|
28
|
Nakano N, Tsuchiya Y, Kako K, Umezaki K, Sano K, Ikeno S, Otsuka E, Shigeta M, Nakagawa A, Sakata N, Itoh F, Nakano Y, Iemura SI, van Dinther M, Natsume T, Ten Dijke P, Itoh S. TMED10 Protein Interferes with Transforming Growth Factor (TGF)-β Signaling by Disrupting TGF-β Receptor Complex Formation. J Biol Chem 2017; 292:4099-4112. [PMID: 28115518 DOI: 10.1074/jbc.m116.769109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/13/2017] [Indexed: 01/17/2023] Open
Abstract
The intensity and duration of TGF-β signaling determine the cellular biological response. How this is negatively regulated is not well understood. Here, we identified a novel negative regulator of TGF-β signaling, transmembrane p24-trafficking protein 10 (TMED10). TMED10 disrupts the complex formation between TGF-β type I (also termed ALK5) and type II receptors (TβRII). Misexpression studies revealed that TMED10 attenuated TGF-β-mediated signaling. A 20-amino acid-long region from Thr91 to Glu110 within the extracellular region of TMED10 was found to be crucial for TMED10 interaction with both ALK5 and TβRII. Synthetic peptides corresponding to this region inhibit both TGF-β-induced Smad2 phosphorylation and Smad-dependent transcriptional reporter activity. In a xenograft cancer model, where previously TGF-β was shown to elicit tumor-promoting effects, gain-of-function and loss-of-function studies for TMED10 revealed a decrease and increase in the tumor size, respectively. Thus, we determined herein that TMED10 expression levels are the key determinant for efficiency of TGF-β receptor complex formation and signaling.
Collapse
Affiliation(s)
- Naoko Nakano
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Yuki Tsuchiya
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kenro Kako
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Kenryu Umezaki
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Keigo Sano
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Souichi Ikeno
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Eri Otsuka
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Masashi Shigeta
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Ai Nakagawa
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Nobuo Sakata
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Fumiko Itoh
- the Laboratory of Cardiovascular Medicine, Tokyo University of Pharmacy and Life Sciences, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | - Yota Nakano
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan
| | - Shun-Ichiro Iemura
- the Translational Research Center, Fukushima Medical University, 11-25 Sakaemachi, Fukushima City, Fukushima 960-8031, Japan
| | - Maarten van Dinther
- the Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, S-1-P, 2300 RC Leiden, The Netherlands
| | - Tohru Natsume
- the Molecular Profiling Research Center for Drug Discovery (molprof), National Institute of Advanced Industrial Science and Technology (AIST), 2-4-7 Aomi, Koto-ku, Tokyo 135-0064, Japan, and
| | - Peter Ten Dijke
- the Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, S-1-P, 2300 RC Leiden, The Netherlands.,the Science for Life Laboratory, Ludwig Institute for Cancer Research, Uppsala University, Uppsala SE-751 24, Sweden
| | - Susumu Itoh
- From the Laboratory of Biochemistry, Showa Pharmaceutical University, Machida, Tokyo 194-8543, Japan,
| |
Collapse
|
29
|
Huang M, Lou D, Li HH, Cai Q, Wang YP, Yang HF. Pyrrolidine dithiocarbamate attenuates paraquat-induced acute pulmonary poisoning in vivo via transforming growth factor β1 and nuclear factor κB pathway interaction. Hum Exp Toxicol 2016; 35:1312-1318. [PMID: 26860689 DOI: 10.1177/0960327116630351] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Paraquat (PQ) exposure could cause pulmonary fibrosis. The aim of this study was to investigate the protective effect of pyrrolidine dithiocarbamate (PDTC) in an acute PQ poison model. One hundred and forty-four Sprague Dawley rats were equally divided into three experimental groups: control group, PQ group, and PQ + PDTC group. At days 1, 3, 7, 14, 28, and 56 of treatment, the serum levels of transforming growth factor β1 (TGF-β1), the levels of hydroxyproline, the protein expression of nuclear factor κB (NF-κB) pathway, and histopathological change in lung tissue were assessed. The survival rate of rats treated with PQ + PDTC was increased compared with that of rats treated only with PQ (p < 0.05), and the occurrence of pathological changes was dramatically attenuated in the PQ + PDTC group. The serum levels of TGF-β1 and the hydroxyproline levels in the PQ group were significantly increased in a time-dependent manner compared with those in the control and PQ + PDTC groups on days 7, 14, 28, and 56 (p < 0.05). Additionally, the protein levels of NF-κB proteins p65, inhibitor of κB (IκB) kinase (IKKβ, and IκB-α were significantly downregulated in the PQ + PDTC group as determined by array analysis. The present findings suggest that overexpression of TGF-β1 may play an important role in PQ-induced lung injury and that PDTC, a strong NF-κB inhibitor, can rescue PQ-induced pulmonary fibrosis by influencing the protein expression of NF-κB pathway.
Collapse
Affiliation(s)
- M Huang
- Lab of Molecular Toxicology, Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, People's Republic of China
| | - D Lou
- Shanghai Municipal Center for Disease Control and Prevention, Shanghai, People's Republic of China
| | - H-H Li
- Lab of Molecular Toxicology, Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Q Cai
- Lab of Molecular Toxicology, Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Y-P Wang
- Lab of Molecular Toxicology, Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, People's Republic of China
| | - H-F Yang
- Lab of Molecular Toxicology, Department of Occupational and Environmental Health, School of Public Health, Ningxia Medical University, Yinchuan, People's Republic of China
| |
Collapse
|
30
|
Corinaldesi C, Di Luigi L, Lenzi A, Crescioli C. Phosphodiesterase type 5 inhibitors: back and forward from cardiac indications. J Endocrinol Invest 2016; 39:143-51. [PMID: 26122487 PMCID: PMC4712255 DOI: 10.1007/s40618-015-0340-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 06/11/2015] [Indexed: 12/24/2022]
Abstract
PDE5 inhibitors (PDE5i) are widely known as treatment for erectile dysfunction (ED). This favorable action has emerged as a "side effect" from pioneering studies when PDE5i have been originally proposed as treatment for coronary artery disease (CAD). PDE5i showed marginal benefits for CAD treatment; although disappointing for that indication, they improved systemic and pulmonary vasodilation and ameliorated general endothelial function. Therefore, PDE5i have been approved and licensed also for pulmonary artery hypertension (PAH), besides ED. Nowadays, fine-tuned biomolecular mechanisms of PDE5i are well recognized to be beneficial onto myocardial contractility and geometry, to reduce tissue fibrosis, hypertrophy and apoptosis. PDE5i consistently exert benefits on heart failure, infarct, cardiomyopathy. The concept that PDE5i likely blunt Th1-driven inflammatory processes, which shift the homeostatic balance from health to disease, has emerged; PDE5i seem to decrease the release of active biomolecules from cells to tissues interested by inflammation. In this view, following clinical and basic research progresses, PDE5i can be undoubtedly "re-allocated" for cardiac indications and, hopefully, they could be approved as therapeutic tools to treat and prevent heart disease. This review aims to summarize PDE5i different clinical applications, from past to present and future, focusing on their potential power as treatment for cardiac diseases.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics
- Anti-Inflammatory Agents, Non-Steroidal/pharmacology
- Anti-Inflammatory Agents, Non-Steroidal/therapeutic use
- Cardiovascular Agents/pharmacokinetics
- Cardiovascular Agents/pharmacology
- Cardiovascular Agents/therapeutic use
- Cardiovascular Diseases/drug therapy
- Cardiovascular Diseases/immunology
- Cardiovascular Diseases/metabolism
- Cardiovascular Diseases/physiopathology
- Coronary Artery Disease/drug therapy
- Coronary Artery Disease/immunology
- Coronary Artery Disease/metabolism
- Coronary Artery Disease/physiopathology
- Cyclic Nucleotide Phosphodiesterases, Type 5/chemistry
- Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Evidence-Based Medicine
- Heart/drug effects
- Heart/physiopathology
- Humans
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/physiopathology
- Myocardium/enzymology
- Myocardium/immunology
- Myocardium/metabolism
- Phosphodiesterase 5 Inhibitors/pharmacokinetics
- Phosphodiesterase 5 Inhibitors/pharmacology
- Phosphodiesterase 5 Inhibitors/therapeutic use
- Vasodilator Agents/pharmacokinetics
- Vasodilator Agents/pharmacology
- Vasodilator Agents/therapeutic use
Collapse
Affiliation(s)
- C Corinaldesi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - L Di Luigi
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy
| | - A Lenzi
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - C Crescioli
- Department of Movement, Human and Health Sciences, Section of Health Science, Unit of Endocrinology, University of Rome "Foro Italico", Rome, Italy.
| |
Collapse
|
31
|
Choi YJ, Baek GY, Park HR, Jo SK, Jung U. Smad2/3-Regulated Expression of DLX2 Is Associated with Radiation-Induced Epithelial-Mesenchymal Transition and Radioresistance of A549 and MDA-MB-231 Human Cancer Cell Lines. PLoS One 2016; 11:e0147343. [PMID: 26799321 PMCID: PMC4723265 DOI: 10.1371/journal.pone.0147343] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 12/31/2015] [Indexed: 01/27/2023] Open
Abstract
The control of radioresistance and metastatic potential of surviving cancer cells is important for improving cancer eradication by radiotheraphy. The distal-less homeobox2 (DLX2) gene encodes for a homeobox transcription factor involved in morphogenesis and its deregulation was found in human solid tumors and hematologic malignancies. Here we investigated the role of DLX2 in association with radiation-induced epithelial to mesenchymal transition (EMT) and stem cell-like properties and its regulation by Smad2/3 signaling in irradiated A549 and MDA-MB-231 human cancer cell lines. In irradiated A549 and MDA-MB-231 cells, EMT was induced as demonstrated by EMT marker expression, phosphorylation of Smad2/3, and migratory and invasive ability. Also, irradiated A549 and MDA-MB-231 cells showed increased cancer stem cells (CSCs) marker. Interestingly, DLX2 was overexpressed upon irradiation. Therefore, we examined the role of DLX2 in radiation-induced EMT and radioresistance. The overexpression of DLX2 alone induced EMT, migration and invasion, and CSC marker expression. The reduced colony-forming ability in irradiated cells was partially restored by DLX2 overexpression. On the other hand, the depletion of DLX2 using si-RNA abolished radiation-induced EMT, CSC marker expression, and phosphorylation of Smad2/3 in irradiated A549 and MDA-MB-231 cells. Also, depletion of DLX2 increased the radiation sensitivity in both cell lines. Moreover, knockdown of Smad2/3, a key activator of TGF-β1 pathway, abrogated the radiation-induced DLX2 expression, indicating that radiation-induced DLX2 expression is dependent on Smad2/3 signaling. These results demonstrated that DLX2 plays a crucial role in radioresistance, radiation-induced EMT and CSC marker expression, and the expression of DLX2 is regulated by Smad2/3 signaling in A549 and MDA-MB-231 cell lines.
Collapse
Affiliation(s)
- Yeo-Jin Choi
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Ga-Young Baek
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
| | - Hae-Ran Park
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sung-Kee Jo
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Uhee Jung
- Radiation Biotechnology Research Division, Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute (KAERI), Jeongeup, Republic of Korea
- Department of Radiation Biotechnology and Applied Radioisotope, University of Science and Technology (UST), Daejeon, Republic of Korea
- * E-mail:
| |
Collapse
|
32
|
Tenogenic induction of equine mesenchymal stem cells by means of growth factors and low-level laser technology. Vet Res Commun 2016; 40:39-48. [PMID: 26757735 DOI: 10.1007/s11259-016-9652-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/05/2016] [Indexed: 12/20/2022]
Abstract
Tendons regenerate poorly due to a dense extracellular matrix and low cellularity. Cellular therapies aim to improve tendon repair using mesenchymal stem cells and tenocytes; however, a current limitation is the low proliferative potential of tenocytes in cases of severe trauma. The purpose of this study was to develop a method useful in veterinary medicine to improve the differentiation of Peripheral Blood equine mesenchymal stem cells (PB-MSCs) into tenocytes. PB-MSCs were used to study the effects of the addition of some growth factors (GFs) as TGFβ3 (transforming growth factor), EGF2 (Epidermal growth factor), bFGF2 (Fibroblast growth factor) and IGF-1 (insulin-like growth factor) in presence or without Low Level Laser Technology (LLLT) on the mRNA expression levels of genes important in the tenogenic induction as Early Growth Response Protein-1 (EGR1), Tenascin (TNC) and Decorin (DCN). The singular addition of GFs did not show any influence on the mRNA expression of tenogenic genes whereas the specific combinations that arrested cell proliferation in favour of differentiation were the following: bFGF2 + TGFβ3 and bFGF2 + TGFβ3 + LLLT. Indeed, the supplement of bFGF2 and TGFβ3 significantly upregulated the expression of Early Growth Response Protein-1 and Decorin, while the use of LLLT induced a significant increase of Tenascin C levels. In conclusion, the present study might furnish significant suggestions for developing an efficient approach for tenocyte induction since the external administration of bFGF2 and TGFβ3, along with LLLT, influences the differentiation of PB-MSCs towards the tenogenic fate.
Collapse
|
33
|
A Novel Matrine Derivative WM130 Inhibits Activation of Hepatic Stellate Cells and Attenuates Dimethylnitrosamine-Induced Liver Fibrosis in Rats. BIOMED RESEARCH INTERNATIONAL 2015; 2015:203978. [PMID: 26167476 PMCID: PMC4488526 DOI: 10.1155/2015/203978] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/21/2015] [Accepted: 06/02/2015] [Indexed: 02/08/2023]
Abstract
Activation of hepatic stellate cells (HSCs) is a critical event in process of hepatic fibrogenesis and cirrhosis. Matrine, the active ingredient of Sophora, had been used for clinical treatment of acute/chronic liver disease. However, its potency was low. We prepared a high potency and low toxicity matrine derivate, WM130 (C30N4H40SO5F), which exhibited better pharmacological activities on antihepatic fibrosis. This study demonstrated that WM130 results in a decreased proliferative activity of HSC-T6 cells, with the half inhibitory concentration (IC50) of 68 μM. WM130 can inhibit the migration and induce apoptosis in HSC-T6 cells at both concentrations of 68 μM (IC50) and 34 μM (half IC50). The expression of α-SMA, Collagen I, Collagen III, and TGF-β1 could be downregulated, and the protein phosphorylation levels of EGFR, AKT, ERK, Smad, and Raf (p-EGFR, p-AKT, p-ERK, p-Smad, and p-Raf) were also decreased by WM130. On the DMN-induced rat liver fibrosis model, WM130 can effectively reduce the TGF-β1, AKT, α-SMA, and p-ERK levels, decrease the extracellular matrix (ECM) formation, and inhibit rat liver fibrosis progression. In conclusion, this study demonstrated that WM130 can significantly inhibit the activation of HSC-T6 cells and block the rat liver fibrosis progression by inducing apoptosis, suppressing the deposition of ECM, and inhibiting TGF-β/Smad and Ras/ERK pathways.
Collapse
|