1
|
Dabral S, Noh M, Werner F, Krebes L, Völker K, Maier C, Aleksic I, Novoyatleva T, Hadzic S, Schermuly RT, Perez VADJ, Kuhn M. C-type natriuretic peptide/cGMP/FoxO3 signaling attenuates hyperproliferation of pericytes from patients with pulmonary arterial hypertension. Commun Biol 2024; 7:693. [PMID: 38844781 PMCID: PMC11156916 DOI: 10.1038/s42003-024-06375-3] [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: 07/27/2023] [Accepted: 05/23/2024] [Indexed: 06/09/2024] Open
Abstract
Pericyte dysfunction, with excessive migration, hyperproliferation, and differentiation into smooth muscle-like cells contributes to vascular remodeling in Pulmonary Arterial Hypertension (PAH). Augmented expression and action of growth factors trigger these pathological changes. Endogenous factors opposing such alterations are barely known. Here, we examine whether and how the endothelial hormone C-type natriuretic peptide (CNP), signaling through the cyclic guanosine monophosphate (cGMP) -producing guanylyl cyclase B (GC-B) receptor, attenuates the pericyte dysfunction observed in PAH. The results demonstrate that CNP/GC-B/cGMP signaling is preserved in lung pericytes from patients with PAH and prevents their growth factor-induced proliferation, migration, and transdifferentiation. The anti-proliferative effect of CNP is mediated by cGMP-dependent protein kinase I and inhibition of the Phosphoinositide 3-kinase (PI3K)/AKT pathway, ultimately leading to the nuclear stabilization and activation of the Forkhead Box O 3 (FoxO3) transcription factor. Augmentation of the CNP/GC-B/cGMP/FoxO3 signaling pathway might be a target for novel therapeutics in the field of PAH.
Collapse
Affiliation(s)
- Swati Dabral
- Institute of Physiology, University of Würzburg, Würzburg, Germany.
| | - Minhee Noh
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Franziska Werner
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Lisa Krebes
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Katharina Völker
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Christopher Maier
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Ivan Aleksic
- Department of Thoracic and Cardiovascular Surgery, University hospital Würzburg, Würzburg, Germany
| | - Tatyana Novoyatleva
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Stefan Hadzic
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Ralph Theo Schermuly
- Justus-Liebig-University Giessen (JLU), Giessen, Germany
- Universities of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Giessen, Germany
- Member of the German Center for Lung Research (DZL), Giessen, Germany
| | - Vinicio A de Jesus Perez
- Divisions of Pulmonary and Critical Care Medicine and Stanford Cardiovascular Institute, Stanford University, California, USA
| | - Michaela Kuhn
- Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
2
|
Miyazawa K, Itoh Y, Fu H, Miyazono K. Receptor-activated transcription factors and beyond: multiple modes of Smad2/3-dependent transmission of TGF-β signaling. J Biol Chem 2024; 300:107256. [PMID: 38569937 PMCID: PMC11063908 DOI: 10.1016/j.jbc.2024.107256] [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: 01/19/2024] [Revised: 02/28/2024] [Accepted: 03/05/2024] [Indexed: 04/05/2024] Open
Abstract
Transforming growth factor β (TGF-β) is a pleiotropic cytokine that is widely distributed throughout the body. Its receptor proteins, TGF-β type I and type II receptors, are also ubiquitously expressed. Therefore, the regulation of various signaling outputs in a context-dependent manner is a critical issue in this field. Smad proteins were originally identified as signal-activated transcription factors similar to signal transducer and activator of transcription proteins. Smads are activated by serine phosphorylation mediated by intrinsic receptor dual specificity kinases of the TGF-β family, indicating that Smads are receptor-restricted effector molecules downstream of ligands of the TGF-β family. Smad proteins have other functions in addition to transcriptional regulation, including post-transcriptional regulation of micro-RNA processing, pre-mRNA splicing, and m6A methylation. Recent technical advances have identified a novel landscape of Smad-dependent signal transduction, including regulation of mitochondrial function without involving regulation of gene expression. Therefore, Smad proteins are receptor-activated transcription factors and also act as intracellular signaling modulators with multiple modes of function. In this review, we discuss the role of Smad proteins as receptor-activated transcription factors and beyond. We also describe the functional differences between Smad2 and Smad3, two receptor-activated Smad proteins downstream of TGF-β, activin, myostatin, growth and differentiation factor (GDF) 11, and Nodal.
Collapse
Affiliation(s)
- Keiji Miyazawa
- 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
| | - Hao Fu
- Department of Biochemistry, Graduate School of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Kohei Miyazono
- Department of Applied Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan; Laboratory for Cancer Invasion and Metastasis, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| |
Collapse
|
3
|
Vishwakarma VK, Shah S, Kaur T, Singh AP, Arava SK, Kumar N, Yadav RK, Yadav S, Arora T, Yadav HN. Effect of vinpocetine alone and in combination with enalapril in experimental model of diabetic cardiomyopathy in rats: possible involvement of PDE-1/TGF-β/ Smad 2/3 signalling pathways. J Pharm Pharmacol 2023; 75:1198-1211. [PMID: 37229596 DOI: 10.1093/jpp/rgad043] [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: 08/25/2022] [Accepted: 04/25/2023] [Indexed: 05/27/2023]
Abstract
OBJECTIVE Diabetic cardiomyopathy (DC) is one of the severe secondary complications of diabetes mellitus in humans. Vinpocetine is an alkaloid having pleiotropic pharmacological effects. The present study is designed to investigate the effect of vinpocetine in DC in rats. METHODS Rats were fed a high-fat diet for nine weeks along with single dose of streptozotocin after the second week to induce DC. The haemodynamic evaluation was performed to assess the functional status of rats using the Biopac system. Cardiac echocardiography, biochemical, oxidative stress parameters and inflammatory cytokine level were analysed in addition to haematoxylin-eosin and Masson's trichome staining to study histological changes, cardiomyocyte diameter and fibrosis, respectively. Phosphodiesterase-1 (PDE-1), transforming growth factor-β (TGF-β) and p-Smad 2/3 expression in cardiac tissues were quantified using western blot/RT-PCR. KEY FINDING Vinpocetine treatment and its combination with enalapril decreased the glucose levels compared to diabetic rats. Vinpocetine improved the echocardiographic parameters and cardiac functional status of rats. Vinpocetine decreased the cardiac biochemical parameters, oxidative stress, inflammatory cytokine levels, cardiomyocyte diameter and fibrosis in rats. Interestingly, expressions of PDE-1, TGF-β and p-Smad 2/3 were ameliorated by vinpocetine alone and in combination with enalapril. CONCLUSIONS Vinpocetine is a well-known inhibitor of PDE-1 and the protective effect of vinpocetine in DC is exerted by inhibition of PDE-1 and subsequent inhibition of the expression of TGF-β/Smad 2/3.
Collapse
Affiliation(s)
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India
| | - Tajpreet Kaur
- Department of Pharmacology, Khalsa College of Pharmacy, Amritsar, India
| | - Amrit Pal Singh
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sudheer Kumar Arava
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Niraj Kumar
- Department of Neuroanesthesiogy and Critical Care, All India Institute of Medical Sciences, New Delhi, India
| | - Raj Kanwar Yadav
- Department Nephrology, All India Institute of Medical Sciences, New Delhi, India
| | - Sushma Yadav
- Department of Obstetrics and Gynaecology, SHKM Government Medical College, Nuh, Haryana, India
| | - Taruna Arora
- RBMCH, ICMR-Head Quarter's Ansari Nagar, New Delhi, India
| | | |
Collapse
|
4
|
Nakamura H, Zhou Y, Sakamoto Y, Yamazaki A, Kurumiya E, Yamazaki R, Hayashi K, Kasuya Y, Watanabe K, Kasahara J, Takabatake M, Tatsumi K, Yoshino I, Honda T, Murayama T. N-butyldeoxynojirimycin (miglustat) ameliorates pulmonary fibrosis through inhibition of nuclear translocation of Smad2/3. Biomed Pharmacother 2023; 160:114405. [PMID: 36804125 DOI: 10.1016/j.biopha.2023.114405] [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: 12/13/2022] [Revised: 02/10/2023] [Accepted: 02/10/2023] [Indexed: 02/17/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic progressive lung disease. The disease involves excessive accumulation of fibroblasts and myofibroblasts, and myofibroblasts differentiated by pro-fibrotic factors promote the deposition of extracellular matrix proteins such as collagen and fibronectin. Transforming growth factor-β1 is a pro-fibrotic factor that promotes fibroblast-to-myofibroblast differentiation (FMD). Therefore, inhibition of FMD may be an effective strategy for IPF treatment. In this study, we screened the anti-FMD effects of various iminosugars and showed that some compounds, including N-butyldeoxynojirimycin (NB-DNJ, miglustat, an inhibitor of glucosylceramide synthase (GCS)), a clinically approved drug for treating Niemann-Pick disease type C and Gaucher disease type 1, inhibited TGF-β1-induced FMD by inhibiting the nuclear translocation of Smad2/3. N-butyldeoxygalactonojirimycin having GCS inhibitory effect did not attenuate the TGF-β1-induced FMD, suggesting that NB-DNJ exerts the anti-FMD effects by GCS inhibitory effect independent manner. N-butyldeoxynojirimycin did not inhibit TGF-β1-induced Smad2/3 phosphorylation. In a mouse model of bleomycin (BLM)-induced pulmonary fibrosis, intratracheal or oral administration of NB-DNJ at an early fibrotic stage markedly ameliorated lung injury and deterioration of respiratory functions, such as specific airway resistance, tidal volume, and peak expiratory flow. Furthermore, the anti-fibrotic effects of NB-DNJ in the BLM-induced lung injury model were similar to those of pirfenidone and nintedanib, which are clinically approved drugs for the treatment of IPF. These results suggest that NB-DNJ may be effective for IPF treatment.
Collapse
Affiliation(s)
- Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.
| | - Yuan Zhou
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Yuka Sakamoto
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Ayako Yamazaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Eon Kurumiya
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Risa Yamazaki
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Kyota Hayashi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Yoshitoshi Kasuya
- Deprtment of Biomedical Science, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan; Department of Biochemistry and Molecular Pharmacology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Kazuaki Watanabe
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Junya Kasahara
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Mamoru Takabatake
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Koichiro Tatsumi
- Department of Respirology, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Ichiro Yoshino
- Department of General Thoracic Surgery, Graduate School of Medicine, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8670, Japan
| | - Takuya Honda
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
5
|
Bueno M, Calyeca J, Khaliullin T, Miller MP, Alvarez D, Rosas L, Brands J, Baker C, Nasser A, Shulkowski S, Mathien A, Uzoukwu N, Sembrat J, Mays BG, Fiedler K, Hahn SA, Salvatore SR, Schopfer FJ, Rojas M, Sandner P, Straub AC, Mora AL. CYB5R3 in type II alveolar epithelial cells protects against lung fibrosis by suppressing TGF-β1 signaling. JCI Insight 2023; 8:e161487. [PMID: 36749633 PMCID: PMC10077481 DOI: 10.1172/jci.insight.161487] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 01/27/2023] [Indexed: 02/08/2023] Open
Abstract
Type II alveolar epithelial cell (AECII) redox imbalance contributes to the pathogenesis of idiopathic pulmonary fibrosis (IPF), a deadly disease with limited treatment options. Here, we show that expression of membrane-bound cytochrome B5 reductase 3 (CYB5R3), an enzyme critical for maintaining cellular redox homeostasis and soluble guanylate cyclase (sGC) heme iron redox state, is diminished in IPF AECIIs. Deficiency of CYB5R3 in AECIIs led to sustained activation of the pro-fibrotic factor TGF-β1 and increased susceptibility to lung fibrosis. We further show that CYB5R3 is a critical regulator of ERK1/2 phosphorylation and the sGC/cGMP/protein kinase G axis that modulates activation of the TGF-β1 signaling pathway. We demonstrate that sGC agonists (BAY 41-8543 and BAY 54-6544) are effective in reducing the pulmonary fibrotic outcomes of in vivo deficiency of CYB5R3 in AECIIs. Taken together, these results show that CYB5R3 in AECIIs is required to maintain resilience after lung injury and fibrosis and that therapeutic manipulation of the sGC redox state could provide a basis for treating fibrotic conditions in the lung and beyond.
Collapse
Affiliation(s)
- Marta Bueno
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jazmin Calyeca
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Timur Khaliullin
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Megan P. Miller
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Diana Alvarez
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Lorena Rosas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Judith Brands
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Christian Baker
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amro Nasser
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Stephanie Shulkowski
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - August Mathien
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Nneoma Uzoukwu
- Aging Institute, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - John Sembrat
- Division of Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Brenton G. Mays
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kaitlin Fiedler
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Scott A. Hahn
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Francisco J. Schopfer
- Heart, Lung, Blood, and Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Pharmacology and Chemical Biology
- Pittsburgh Liver Research Center (PLRC), and
- Center for Metabolism and Mitochondrial Medicine (C3M), University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mauricio Rojas
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| | - Peter Sandner
- Bayer Pharmaceuticals Wuppertal, Germany
- Hannover Medical School, Hannover, Germany
| | | | - Ana L. Mora
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care & Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
7
|
Xiong Q, Lu F, Xie X, Zhou W. Hypoxia-induced endothelial cell-derived exosome stimulates vascular smooth muscle cell proliferation and migration. Biomed Res 2023; 44:245-255. [PMID: 38008423 DOI: 10.2220/biomedres.44.245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2023]
Abstract
This study mainly used human VSMCs and ECs cultured in vitro to investigate whether exosomes (Exos) are involved in the communication between ECs and VSMCs under hypoxia, and to explore the role and mechanism of ECs-derived exosomes in the abnormal proliferation of VSMCs. VSMCs proliferation and migration were assessed by a series of cell function assays after culturing VSMCs alone or co-culturing ECs under hypoxia or normoxia. Next, exosomes were extracted from ECs under hypoxia or normoxia and characterized. We then introduced ECs-Exos to observe their effects on VSMCs proliferation and migration, and further evaluated the expression of transforming growth factor-beta receptor 1 (TGFBR1) pathway-related proteins. Finally, the effect of ECs-Exos on VSMCs function was evaluated after knocking down TGFBR1 in ECs. VSMCs treated with ECs-Exos exhibited increased proliferation and migration ability in hypoxic environment, and the expression of TGFBR1 pathway-related proteins was upregulated. Administration of ECs-Exos with TGFβ1 knockdown conspicuously reversed the promoting effects of ECs-Exos on cell proliferation and migration under hypoxia. In summary, hypoxia affected the secretion of extracellular vesicles by endothelial cells, which can be internalized by VSMCs and accelerate the abnormal proliferation and migration of VSMCs by delivering TGFBR1.
Collapse
Affiliation(s)
- Qinggen Xiong
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Fei Lu
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Xiaoming Xie
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| | - Wei Zhou
- Intervention Department (Vascular Surgery Department) of The Second Affiliated Hospital of Nanchang University, Nanchang, 330008, China
| |
Collapse
|
8
|
Genetic Disruption of Guanylyl Cyclase/Natriuretic Peptide Receptor-A Triggers Differential Cardiac Fibrosis and Disorders in Male and Female Mutant Mice: Role of TGF-β1/SMAD Signaling Pathway. Int J Mol Sci 2022; 23:ijms231911487. [PMID: 36232788 PMCID: PMC9569686 DOI: 10.3390/ijms231911487] [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: 07/20/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 01/01/2023] Open
Abstract
The global targeted disruption of the natriuretic peptide receptor-A (NPRA) gene (Npr1) in mice provokes hypertension and cardiovascular dysfunction. The objective of this study was to determine the mechanisms regulating the development of cardiac fibrosis and dysfunction in Npr1 mutant mice. Npr1 knockout (Npr1-/-, 0-copy), heterozygous (Npr1+/-, 1-copy), and wild-type (Npr1+/+, 2-copy) mice were treated with the transforming growth factor (TGF)-β1 receptor (TGF-β1R) antagonist GW788388 (2 µg/g body weight/day; ip) for 28 days. Hearts were isolated and used for real-time quantitative reverse transcription polymerase chain reaction (qRT-PCR), Western blot, and immunohistochemical analyses. The Npr1-/- (0-copy) mice showed a 6-fold induction of cardiac fibrosis and dysfunction with markedly induced expressions of collagen-1α (3.8-fold), monocyte chemoattractant protein (3.7-fold), connective tissue growth factor (CTGF, 5.3-fold), α-smooth muscle actin (α-SMA, 6.1-fold), TGF-βRI (4.3-fold), TGF-βRII (4.7-fold), and phosphorylated small mothers against decapentaplegic (pSMAD) proteins, including pSMAD-2 (3.2-fold) and pSMAD-3 (3.7-fold), compared with wild-type mice. The expressions of phosphorylated extracellular-regulated kinase ERK1/2 (pERK1/2), matrix metalloproteinases-2, -9, (MMP-2, -9), and proliferating cell nuclear antigen (PCNA) were also significantly upregulated in Npr1 0-copy mice. The treatment of mutant mice with GW788388 significantly blocked the expression of fibrotic markers, SMAD proteins, MMPs, and PCNA compared with the vehicle-treated control mice. The treatment with GW788388 significantly prevented cardiac dysfunctions in a sex-dependent manner in Npr1 0-copy and 1-copy mutant mice. The results suggest that the development of cardiac fibrosis and dysfunction in mutant mice is predominantly regulated through the TGF-β1-mediated SMAD-dependent pathway.
Collapse
|
9
|
Zheng Y, Zhang H, Sun H. Metformin inhibits the proliferation and invasion of ovarian cancer cells by suppressing TRIM37-induced TRAF2 ubiquitination. Cancer Sci 2022; 113:3776-3786. [PMID: 35950370 PMCID: PMC9633302 DOI: 10.1111/cas.15524] [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: 05/26/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian cancer is the leading cause of death in gynecological malignancies worldwide. Our previous studies have proved that metformin inhibited the proliferation and invasion of ovarian cancer in vitro and in vivo. However, the underlying mechanisms have not been fully elucidated. Immunohistochemistry was carried out to detect the expression of tripartite motif‐containing 37 (TRIM37), Ki‐67, and MMP‐9 in ovarian cancer and normal tissues. The influence of TRIM37 on the proliferation and invasion of ovarian cancer cells was verified by the real‐time cellular analysis proliferation test, colony formation test, and Transwell assay. Western blot analysis and immunoprecipitation were used to detect the expression of the nuclear factor‐κB (NF‐κB) pathway and the interaction between TRIM37 and tumor necrosis factor receptor‐associated factor 2 (TRAF2). Ubiquitination detection was carried out to detect the ubiquitination level of TRAF2. The present study revealed that TRIM37 expression was significantly increased in ovarian cancer tissues compared with normal control tissues, and its overexpression was closely associated with proliferation and metastasis. Metformin inhibited the NF‐κB signaling pathway by downregulating TRIM37. Metformin also inhibited the ubiquitination of TRAF2 induced by TRIM37 overexpression. Metformin inhibits the proliferation and invasion of ovarian cancer cells by suppressing TRIM37‐induced TRAF2 ubiquitination.
Collapse
Affiliation(s)
- Ya Zheng
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P. R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, P. R. China
| | - Haiyan Zhang
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P. R. China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Shanghai, P. R. China
| | - Hong Sun
- Department of Gynecology, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, P. R. China
| |
Collapse
|
10
|
Xie C, Chen C, Wu L, Xiong Y, Xing C, Mao H. BRCC36 prevents vascular calcification in chronic kidney disease through the β-catenin signalling pathway. Exp Cell Res 2022; 413:113051. [PMID: 35149088 DOI: 10.1016/j.yexcr.2022.113051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 11/04/2022]
Abstract
Vascular calcification (VC) is a strong predictor of cardiovascular mortality and overall mortality in patients with chronic kidney disease (CKD); however, the molecular mechanisms underlying VC have yet to be elucidated. Here, we report the role of the deubiquitinating enzyme BRCC36 in the process of VC in CKD. We established an in vitro VC model of vascular smooth muscle cells (VSMCs) and an adenine-induced CKD mouse model. The expression of BRCC36 was significantly decreased in both the in vivo and in vitro VC models. Alizarin red staining and calcium content assays showed that BRCC36 overexpression reduced calcium deposition in the presence of calcifying medium, while the contractile protein α-smooth muscle actin (α-SMA) was upregulated and phosphorylated β-catenin was downregulated. Cell immunofluorescence showed that BRCC36 overexpression also reduced the expression of phosphorylated β-catenin in the nucleus in the presence of calcifying medium. In addition, coimmunoprecipitation showed that BRCC36 can bind to β-catenin. These results suggest that BRCC36 can interact with β-catenin, the main effector protein of the Wnt/β-catenin pathway, inhibiting the phosphorylation of β-catenin and negatively regulating the cell signalling pathway, thereby inhibiting VC. This may provide new insights into the molecular mechanisms of VC in the context of CKD.
Collapse
Affiliation(s)
- Caidie Xie
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Nephrology, Nanjing Public Health Medical Center, Nanjing Second Hospital, Nanjing Hospital Affiliated to Nanjing University of Traditional Chinese Medicine, Nanjing, China.
| | - Cheng Chen
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China; Department of Medical Science, Yangzhou Polytechnic College, Yangzhou, China.
| | - Lin Wu
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yiqing Xiong
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Changying Xing
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Huijuan Mao
- Department of Nephrology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Hofmann F. The cGMP system: components and function. Biol Chem 2021; 401:447-469. [PMID: 31747372 DOI: 10.1515/hsz-2019-0386] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
The cyclic guanosine monophosphate (cGMP) signaling system is one of the most prominent regulators of a variety of physiological and pathophysiological processes in many mammalian and non-mammalian tissues. Targeting this pathway by increasing cGMP levels has been a very successful approach in pharmacology as shown for nitrates, phosphodiesterase (PDE) inhibitors and stimulators of nitric oxide-guanylyl cyclase (NO-GC) and particulate GC (pGC). This is an introductory review to the cGMP signaling system intended to introduce those readers to this system, who do not work in this area. This article does not intend an in-depth review of this system. Signal transduction by cGMP is controlled by the generating enzymes GCs, the degrading enzymes PDEs and the cGMP-regulated enzymes cyclic nucleotide-gated ion channels, cGMP-dependent protein kinases and cGMP-regulated PDEs. Part A gives a very concise introduction to the components. Part B gives a very concise introduction to the functions modulated by cGMP. The article cites many recent reviews for those who want a deeper insight.
Collapse
Affiliation(s)
- Franz Hofmann
- Pharmakologisches Institut, Technische Universität München, Biedersteiner Str. 29, D-80802 München, Germany
| |
Collapse
|
12
|
DiNicolantonio JJ, McCarty MF, Barroso-Aranda J, Assanga S, Lujan LML, O'Keefe JH. A nutraceutical strategy for downregulating TGFβ signalling: prospects for prevention of fibrotic disorders, including post-COVID-19 pulmonary fibrosis. Open Heart 2021; 8:openhrt-2021-001663. [PMID: 33879509 PMCID: PMC8061562 DOI: 10.1136/openhrt-2021-001663] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/30/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- James J DiNicolantonio
- Preventive Cardiology, Saint Luke's Mid America Heart Institute, Kansas City, Missouri, USA
| | | | | | - Simon Assanga
- Department of Research and Postgraduate Studies in Food, University of Sonora, Sonora, Mexico
| | | | - James H O'Keefe
- University of Missouri-Kansas City, Saint Lukes Mid America Heart Institute, Kansas City, Missouri, USA
| |
Collapse
|
13
|
Friebe A, Englert N. NO-sensitive guanylyl cyclase in the lung. Br J Pharmacol 2020; 179:2328-2343. [PMID: 33332689 DOI: 10.1111/bph.15345] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/21/2022] Open
Abstract
In the late 1960s, several labatories identified guanylyl cyclase (GC) as the cGMP-producing enzyme. Subsequently, two different types of GC were described that differed in their cellular localization. Primarily found in the cytosol, nitric oxide (NO)-sensitive guanylyl cyclase (NO-GC) acts as receptor for the signalling molecule NO, in contrast the membrane-bound isoenzyme is activated by natriuretic peptides. The lung compared with other tissues exhibits the highest expression of NO-GC. The enzyme has been purified from lung for biochemical analysis. Although expressed in smooth muscle cells (SMCs) and in pericytes, the function of NO-GC in lung, especially in pericytes, is still not fully elucidated. However, pharmacological compounds that target NO-GC are available and have been implemented for the therapy of pulmonary arterial hypertension. In addition, NO-GC has been suggested as drug target for the therapy of asthma, acute respiratory distress syndrome and pulmonary fibrosis.
Collapse
Affiliation(s)
- Andreas Friebe
- Physiological Institute, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Nils Englert
- Physiological Institute, Julius Maximilian University of Würzburg, Würzburg, Germany
| |
Collapse
|
14
|
Nio Y, Ookawara M, Yamasaki M, Hanauer G, Tohyama K, Shibata S, Sano T, Shimizu F, Anayama H, Hazama M, Matsuo T. Ameliorative effect of phosphodiesterase 4 and 5 inhibitors in deoxycorticosterone acetate-salt hypertensive uni-nephrectomized KKA y mice. FASEB J 2020; 34:14997-15014. [PMID: 32939821 DOI: 10.1096/fj.202001084r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 08/04/2020] [Accepted: 08/31/2020] [Indexed: 11/11/2022]
Abstract
Diabetic nephropathy (DN) is a leading cause of end-stage renal disease (ESRD). Hypertension increases kidney stress, which deteriorates function, and leads to peripheral renal vascular resistance. Long-term hypoperfusion promotes interstitial fibrosis and glomerular sclerosis, resulting in nephrosclerosis. Although hypertension and DN are frequent ESRD complications, relevant animal models remain unavailable. We generated a deoxycorticosterone acetate (DOCA)-salt hypertensive uni-nephrectomized (UNx) KKAy mouse model demonstrating hypertension, hyperglycemia, cardiac hypertrophy, kidney failure, increased urinary albumin creatinine ratio (UACR), and increased renal PDE4D and cardiac PDE5A mRNA levels. We hypothesized that the novel PDE4 selective inhibitor, compound A, and PDE5 inhibitor, sildenafil, exhibit nephroprotective, and cardioprotective effects in this new model. Compound A, sildenafil, and the angiotensin II receptor blocker, irbesartan, significantly reduced ventricular hypertrophy and pleural effusion volume. Meanwhile, compound A and sildenafil significantly suppressed the UACR, urinary kidney injury molecule-1, and monocyte chemoattractant protein-1 levels, as well as that of renal pro-fibrotic marker mRNAs, including collagen 1A1, fibronectin, and transforming growth factor-beta (TGF-β). Moreover, compound A significantly suppressed TGF-β-induced pro-fibrotic mRNA expression in vitro in all major kidney lesions, including within the glomerular mesangial region, podocytes, and epithelial region. Hence, PDE4 and PDE5 inhibitors may be promising treatments, in combination with irbesartan, for DN with hypertension as they demonstrate complementary mechanisms.
Collapse
Affiliation(s)
- Yasunori Nio
- Extra-Value Generation and General Medicine DDU, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Mitsugi Ookawara
- Extra-Value Generation and General Medicine DDU, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Midori Yamasaki
- Extra-Value Generation and General Medicine DDU, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Guido Hanauer
- Takeda Pharmaceuticals International AG, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Kimio Tohyama
- Drug Metabolism & Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Sachio Shibata
- Drug Metabolism & Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoya Sano
- Drug Safety Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Fumi Shimizu
- Drug Safety Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hisashi Anayama
- Drug Safety Research Laboratories, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Masatoshi Hazama
- Extra-Value Generation and General Medicine DDU, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Takanori Matsuo
- Extra-Value Generation and General Medicine DDU, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
15
|
ANP/NPRA Inhibits Epithelial-Mesenchymal Transition of Airway by Targeting Smad3 in Asthma. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-018-09804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Chu S, Zhang X, Sun Y, Liang Y, Sun J, Lu M, Huang J, Jiang M, Ma L. Atrial natriuretic peptide inhibits epithelial-mesenchymal transition (EMT) of bronchial epithelial cells through cGMP/PKG signaling by targeting Smad3 in a murine model of allergic asthma. Exp Lung Res 2019; 45:245-254. [PMID: 31496319 DOI: 10.1080/01902148.2019.1660734] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background: Atrial natriuretic peptide (ANP) inhibits TGF-β1-induced epithelial-mesenchymal transition (EMT) in human airway cells. We aim to explore the role and mechanism of ANP on EMT of bronchial epithelial cells from murine model of allergic asthma in vitro. Methods: Murine model of allergic asthma was established with BALB/c mice using ovalbumin (OVA). Bronchial epithelial cells were isolated from OVA-exposed mice, and then were cocultured with TGF-β1, ANP, natriuretic peptide receptor A antagonist, cGMP analog, cGMP inhibitor or/and protein kinase G (PKG) inhibitor, respectively. We assessed expressions of E-Cadherin, α-SMA, cGMP, Smad3 and p-Smad3 in the murine cells before and after Smad3 silence. Results: Compared with bronchial epithelial cells from controls and OVA-exposed mice without additional stimulation, the mRNA and protein expressions of E-Cadherin were decreased but α-SMA expressions were increased in cells with TGF-β1 stimulation from OVA-exposed mice in vitro. That could be reversed by ANP. The effect of ANP could be mimicked by the cGMP analog, which could be reversed by cGMP or PKG inhibitor. Moreover, the phosphorylated Smad3 expression was consistent with that of α-SMA. When Smad3 was silenced, Smad3 was mostly expressed in cytoplasm. In contrast, it is mostly expressed in nucleus of non-silenced cells during EMT. Conclusions: In a murine model of allergic asthma, ANP could inhibit TGF-β1-induced EMT of bronchial epithelial cells through cGMP/PKG signaling, targeting TGF-β1/Smad3 via attenuating phosphorylation of Smad3 in vitro, which may provide potential of ANP in treating allergic asthma with airway remodeling.
Collapse
Affiliation(s)
- Shuyuan Chu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Xiufeng Zhang
- Department of Respiratory Medicine, The Second Affiliated Hospital of Hainan Medical University , Haikou , Hainan , China
| | - Yabing Sun
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Yaxi Liang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Jingyi Sun
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Minyan Lu
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Jianwei Huang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Ming Jiang
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| | - Libing Ma
- Department of Respiratory and Critical Care Medicine, The Affiliated Hospital of Guilin Medical University , Guilin , Guangxi , China.,Institute of Respiratory Diseases, Guilin Medical University , Guilin , Guangxi , China.,Key Laboratory of Respiratory Diseases of Colleges and Universities Affiliated Education Department of Guangxi Zhuang Autonomous Region , Guilin , Guangxi , China
| |
Collapse
|
17
|
Luo Y, Xie L, Liu HM, Liu B. [Effect of low-concentration paclitaxel on collagen deposition outside rat pulmonary artery smooth muscle cells and related mechanism]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:924-929. [PMID: 31506155 PMCID: PMC7390237 DOI: 10.7499/j.issn.1008-8830.2019.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/05/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To study the effect of low-concentration paclitaxel (PTX) on transforming growth factor-β1 (TGF-β1)-induced collagen deposition outside rat pulmonary artery smooth muscle cells (PASMCs) and related mechanism. METHODS Primary rat PASMCs were divided into a blank control group (n=3), a model group (n=3), and a drug intervention group (n=3). No treatment was given for the blank control group. The model group was treated with TGF-β1 with a final concentration of 10 ng/mL. The drug intervention group was treated with PTX with a final concentration of 100 nmol/L in addition to the treatment in the model group. MTT colorimetry was used to measure cell proliferation. Quantitative real-time PCR was used to measure the relative mRNA expression of collagen type I (COL-I) and collagen type III (COL-III). ELISA was used to measure the OD value of COL-I and COL-III proteins. Western blot was used to measure the relative protein expression of COL-I, COL-III, and the key proteins of the TGF-β1/Smad3 signaling pathway (Smad3 and p-Smad3). RESULTS Compared with the blank control group, the model group had significant increases in proliferation ability, relative mRNA and protein expression of COL-I and COL-III, and relative protein expression of p-Smad3 (P<0.05). Compared with the model group, the drug intervention group had significant reductions in the above indicators, but which were still higher than those in the blank control group (P<0.05). There was no significant difference in the relative protein expression of Smad3 among the three groups (P>0.05). CONCLUSIONS Low-concentration PTX exerts a marked inhibitory effect on TGF-β1-induced collagen deposition outside PASMCs, possibly by regulating the phosphorylation of Smad3 protein.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pediatrics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | | | | | | |
Collapse
|
18
|
Luo Y, Xie L, Liu HM, Liu B. [Effect of low-concentration paclitaxel on collagen deposition outside rat pulmonary artery smooth muscle cells and related mechanism]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2019; 21:924-929. [PMID: 31506155 PMCID: PMC7390237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 08/05/2019] [Indexed: 08/01/2024]
Abstract
OBJECTIVE To study the effect of low-concentration paclitaxel (PTX) on transforming growth factor-β1 (TGF-β1)-induced collagen deposition outside rat pulmonary artery smooth muscle cells (PASMCs) and related mechanism. METHODS Primary rat PASMCs were divided into a blank control group (n=3), a model group (n=3), and a drug intervention group (n=3). No treatment was given for the blank control group. The model group was treated with TGF-β1 with a final concentration of 10 ng/mL. The drug intervention group was treated with PTX with a final concentration of 100 nmol/L in addition to the treatment in the model group. MTT colorimetry was used to measure cell proliferation. Quantitative real-time PCR was used to measure the relative mRNA expression of collagen type I (COL-I) and collagen type III (COL-III). ELISA was used to measure the OD value of COL-I and COL-III proteins. Western blot was used to measure the relative protein expression of COL-I, COL-III, and the key proteins of the TGF-β1/Smad3 signaling pathway (Smad3 and p-Smad3). RESULTS Compared with the blank control group, the model group had significant increases in proliferation ability, relative mRNA and protein expression of COL-I and COL-III, and relative protein expression of p-Smad3 (P<0.05). Compared with the model group, the drug intervention group had significant reductions in the above indicators, but which were still higher than those in the blank control group (P<0.05). There was no significant difference in the relative protein expression of Smad3 among the three groups (P>0.05). CONCLUSIONS Low-concentration PTX exerts a marked inhibitory effect on TGF-β1-induced collagen deposition outside PASMCs, possibly by regulating the phosphorylation of Smad3 protein.
Collapse
Affiliation(s)
- Yan Luo
- Department of Pediatrics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, China.
| | | | | | | |
Collapse
|
19
|
Muraki Y, Naito T, Tohyama K, Shibata S, Kuniyeda K, Nio Y, Hazama M, Matsuo T. Improvement of pulmonary arterial hypertension, inflammatory response, and epithelium injury by dual activation of cAMP/cGMP pathway in a rat model of monocrotaline-induced pulmonary hypertension. Biosci Biotechnol Biochem 2019; 83:1000-1010. [PMID: 30835622 DOI: 10.1080/09168451.2019.1584520] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pulmonary hypertension (PH) is a life-threatening lung disease. PH with concomitant lung diseases, e.g., idiopathic pulmonary fibrosis, is associated with poor prognosis. Development of novel therapeutic vasodilators for treatment of these patients is a key imperative. We evaluated the efficacy of dual activation of cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP) using an active, small-molecule phosphodiesterase (PDE4)/PDE5 dual inhibitor (Compound A). Compound A increased both cAMP and cGMP levels in WI-38 lung fibroblasts and suppressed the expressions of type-1 collagen α1 chain and fibronectin. Additionally, compound A reduced right ventricular weight/left ventricular weight+septal weight ratio, brain natriuretic peptide expression levels in right ventricle, C─C motif chemokine ligand 2 expression levels in lung, and plasma surfactant protein D. Our data indicate that dual activation of cAMP/cGMP pathways may be a novel treatment strategy for PH.
Collapse
Affiliation(s)
- Yo Muraki
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Takako Naito
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Kimio Tohyama
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Sachio Shibata
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Kanako Kuniyeda
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Yasunori Nio
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Masatoshi Hazama
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| | - Takanori Matsuo
- a Pharmaceutical Research Division , Takeda Pharmaceutical Company Limited , Fujisawa , Kanagawa , Japan
| |
Collapse
|
20
|
Cai L, Huo Z, Yang H, He F, Cao Z, Wu F, Liu L, Sun B. Gene co-expression network analysis identifies BRCC3 as a key regulator in osteogenic differentiation of osteoblasts through a β-catenin signaling dependent pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2019; 22:173-178. [PMID: 30834083 PMCID: PMC6396986 DOI: 10.22038/ijbms.2018.29498.7123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 09/25/2018] [Indexed: 11/17/2022]
Abstract
OBJECTIVES The prognosis of osteoporosis is very poor, and it is very important to identify a biomarker for prevention of osteoporosis. In this study, we aimed to identify candidate markers in osteoporosis and to investigate the role of candidate markers in osteogenic differentiation. MATERIALS AND METHODS Using Weighted Gene Co-Expression Network analysis, we identified three hub genes might associate with osteoporosis. The mRNA expression of hub genes in osteoblasts from osteoporosis patients or healthy donor was detected by qRT-PCR. Using siRNA and overexpression, we investigated the role of hub gene BRCC3 in osteogenic differentiation by alkaline phosphatase staining and Alizarin red staining. Moreover, the role of β-catenin signaling in the osteogenic differentiation was detected by using β-catenin signaling inhibitor XAV939. RESULTS We identified three hub genes that might associate with osteoporosis including BRCC3, UBE2N, and UBE2K. UBE2N mRNA and UBE2K mRNA were not changed in osteoblasts isolated from osteoporosis patients, compared with healthy donors, whereas BRCC3 mRNA was significantly increased. Depletion of BRCC3 promoted the activation of alkaline phosphatase and formation of calcified nodules in osteoblasts isolated from osteoporosis patients and up-regulated β-catenin expression. XAV939 reversed the BRCC3 siRNA-induced osteogenic differentiation. Additionally, inhibited osteogenic differentiation was also observed after BACC3 overexpression, and this was accompanied by decreased β-catenin expression. CONCLUSION BRCC3 is an important regulator for osteogenic differentiation of osteoblasts through β-catenin signaling, and it might be a promising target for osteoporosis treatment.
Collapse
Affiliation(s)
- Lixiong Cai
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Zhiqian Huo
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Haiyun Yang
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Fengchun He
- Department of Spine Osteopathy, Nanhai Hospital of Southen Medical University, Foshan 528000, China
| | - Zhenglin Cao
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Feng Wu
- Department of Traumatology and Orthopedics, Foshan Hospital of TCM, Foshan 528000, China
| | - Lianjun Liu
- Department of Ophthalmology Myopia Treatment, Foshan Aier Eye Hospital, Foshan 528000, China
| | - Bingyin Sun
- Department of Orthopaedics, Foshan Jiangxiang Hospital, Foshan 528200, China
| |
Collapse
|
21
|
Childers RC, Sunyecz I, West TA, Cismowski MJ, Lucchesi PA, Gooch KJ. Role of the cytoskeleton in the development of a hypofibrotic cardiac fibroblast phenotype in volume overload heart failure. Am J Physiol Heart Circ Physiol 2018; 316:H596-H608. [PMID: 30575422 DOI: 10.1152/ajpheart.00095.2018] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hemodynamic load regulates cardiac remodeling. In contrast to pressure overload (increased afterload), hearts subjected to volume overload (VO; preload) undergo a distinct pattern of eccentric remodeling, chamber dilation, and decreased extracellular matrix content. Critical profibrotic roles of cardiac fibroblasts (CFs) in postinfarct remodeling and in response to pressure overload have been well established. Little is known about the CF phenotype in response to VO. The present study characterized the phenotype of primary cultures of CFs isolated from hearts subjected to 4 wk of VO induced by an aortocaval fistula. Compared with CFs isolated from sham hearts, VO CFs displayed a "hypofibrotic" phenotype, characterized by a ~50% decrease in the profibrotic phenotypic markers α-smooth muscle actin, connective tissue growth factor, and collagen type I, despite increased levels of profibrotic transforming growth factor-β1 and an intact canonical transforming growth factor-β signaling pathway. Actin filament dynamics were characterized, which regulate the CF phenotype in response to biomechanical signals. Actin polymerization was determined by the relative amounts of G-actin monomers versus F-actin. Compared with sham CFs, VO CFs displayed ~78% less F-actin and an increased G-actin-to-F-actin ratio (G/F ratio). In sham CFs, treatment with the Rho kinase inhibitor Y-27632 to increase the G/F ratio resulted in recapitulation of the hypofibrotic CF phenotype observed in VO CFs. Conversely, treatment of VO CFs with jasplakinolide to decrease the G/F ratio restored a more profibrotic response (>2.5-fold increase in α-smooth muscle actin, connective tissue growth factor, and collagen type I). NEW & NOTEWORTHY The present study is the first to describe a "hypofibrotic" phenotype of cardiac fibroblasts isolated from a volume overload model. Our results suggest that biomechanical regulation of actin microfilament stability and assembly is a critical mediator of cardiac fibroblast phenotypic modulation.
Collapse
Affiliation(s)
- Rachel C Childers
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University , Columbus, Ohio
| | - Ian Sunyecz
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - T Aaron West
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Mary J Cismowski
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio
| | - Pamela A Lucchesi
- The Center for Cardiovascular Research, The Research Institute at Nationwide Children's Hospital , Columbus, Ohio.,Department of Pediatrics, The Ohio State University , Columbus, Ohio
| | - Keith J Gooch
- Department of Biomedical Engineering, The Ohio State University , Columbus, Ohio.,The Dorothy M. Davis Heart & Lung Research Institute, The Ohio State University , Columbus, Ohio
| |
Collapse
|
22
|
Münzel T, Daiber A, Steven S, Tran LP, Ullmann E, Kossmann S, Schmidt FP, Oelze M, Xia N, Li H, Pinto A, Wild P, Pies K, Schmidt ER, Rapp S, Kröller-Schön S. Effects of noise on vascular function, oxidative stress, and inflammation: mechanistic insight from studies in mice. Eur Heart J 2018; 38:2838-2849. [PMID: 28329261 DOI: 10.1093/eurheartj/ehx081] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
Aims Epidemiological studies indicate that traffic noise increases the incidence of coronary artery disease, hypertension and stroke. The underlying mechanisms remain largely unknown. Field studies with nighttime noise exposure demonstrate that aircraft noise leads to vascular dysfunction, which is markedly improved by vitamin C, suggesting a key role of oxidative stress in causing this phenomenon. Methods and results We developed a novel animal model to study the vascular consequences of aircraft noise exposure. Peak sound levels of 85 and mean sound level of 72 dBA applied by loudspeakers for 4 days caused an increase in systolic blood pressure, plasma noradrenaline and angiotensin II levels and induced endothelial dysfunction. Noise increased eNOS expression but reduced vascular NO levels because of eNOS uncoupling. Noise increased circulating levels of nitrotyrosine, interleukine-6 and vascular expression of the NADPH oxidase subunit Nox2, nitrotyrosine-positive proteins and of endothelin-1. FACS analysis demonstrated an increase in infiltrated natural killer-cells and neutrophils into the vasculature. Equal mean sound pressure levels of white noise for 4 days did not induce these changes. Comparative Illumina sequencing of transcriptomes of aortic tissues from aircraft noise-treated animals displayed significant changes of genes in part responsible for the regulation of vascular function, vascular remodelling, and cell death. Conclusion We established a novel and unique aircraft noise stress model with increased blood pressure and vascular dysfunction associated with oxidative stress. This animal model enables future studies of molecular mechanisms, mitigation strategies, and pharmacological interventions to protect from noise-induced vascular damage.
Collapse
Affiliation(s)
- Thomas Münzel
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Andreas Daiber
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main
| | - Sebastian Steven
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Lan P Tran
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Elisabeth Ullmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Frank P Schmidt
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Obere Zahlbacher , Mainz, Germany
| | - Antonio Pinto
- Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Philipp Wild
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main.,Preventive Cardiology and Preventive Medicine, Center for Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Kai Pies
- Engineering Office for Noise Protection, Mainz, Germany
| | - Erwin R Schmidt
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Steffen Rapp
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I - Laboratory of Molecular Cardiology, University Medical Center at the Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131 Mainz, Germany
| |
Collapse
|
23
|
Münzel T, Sørensen M, Schmidt F, Schmidt E, Steven S, Kröller-Schön S, Daiber A. The Adverse Effects of Environmental Noise Exposure on Oxidative Stress and Cardiovascular Risk. Antioxid Redox Signal 2018; 28:873-908. [PMID: 29350061 PMCID: PMC5898791 DOI: 10.1089/ars.2017.7118] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 11/11/2017] [Accepted: 11/14/2017] [Indexed: 12/29/2022]
Abstract
Epidemiological studies have provided evidence that traffic noise exposure is linked to cardiovascular diseases such as arterial hypertension, myocardial infarction, and stroke. Noise is a nonspecific stressor that activates the autonomous nervous system and endocrine signaling. According to the noise reaction model introduced by Babisch and colleagues, chronic low levels of noise can cause so-called nonauditory effects, such as disturbances of activity, sleep, and communication, which can trigger a number of emotional responses, including annoyance and subsequent stress. Chronic stress in turn is associated with cardiovascular risk factors, comprising increased blood pressure and dyslipidemia, increased blood viscosity and blood glucose, and activation of blood clotting factors, in animal models and humans. Persistent chronic noise exposure increases the risk of cardiometabolic diseases, including arterial hypertension, coronary artery disease, diabetes mellitus type 2, and stroke. Recently, we demonstrated that aircraft noise exposure during nighttime can induce endothelial dysfunction in healthy subjects and is even more pronounced in coronary artery disease patients. Importantly, impaired endothelial function was ameliorated by acute oral treatment with the antioxidant vitamin C, suggesting that excessive production of reactive oxygen species contributes to this phenomenon. More recently, we introduced a novel animal model of aircraft noise exposure characterizing the underlying molecular mechanisms leading to noise-dependent adverse oxidative stress-related effects on the vasculature. With the present review, we want to provide an overview of epidemiological, translational clinical, and preclinical noise research addressing the nonauditory, adverse effects of noise exposure with focus on oxidative stress. Antioxid. Redox Signal. 28, 873-908.
Collapse
Affiliation(s)
- Thomas Münzel
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Mette Sørensen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Frank Schmidt
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Erwin Schmidt
- Institute for Molecular Genetics, Johannes Gutenberg University, Mainz, Germany
| | - Sebastian Steven
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Swenja Kröller-Schön
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Andreas Daiber
- The Center for Cardiology, Cardiology 1, Johannes Gutenberg University Medical Center, Mainz, Germany
| |
Collapse
|
24
|
Holland NA, Francisco JT, Johnson SC, Morgan JS, Dennis TJ, Gadireddy NR, Tulis DA. Cyclic Nucleotide-Directed Protein Kinases in Cardiovascular Inflammation and Growth. J Cardiovasc Dev Dis 2018; 5:E6. [PMID: 29367584 PMCID: PMC5872354 DOI: 10.3390/jcdd5010006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 01/17/2018] [Accepted: 01/19/2018] [Indexed: 02/08/2023] Open
Abstract
Cardiovascular disease (CVD), including myocardial infarction (MI) and peripheral or coronary artery disease (PAD, CAD), remains the number one killer of individuals in the United States and worldwide, accounting for nearly 18 million (>30%) global deaths annually. Despite considerable basic science and clinical investigation aimed at identifying key etiologic components of and potential therapeutic targets for CVD, the number of individuals afflicted with these dreaded diseases continues to rise. Of the many biochemical, molecular, and cellular elements and processes characterized to date that have potential to control foundational facets of CVD, the multifaceted cyclic nucleotide pathways continue to be of primary basic science and clinical interest. Cyclic adenosine monophosphate (cyclic AMP) and cyclic guanosine monophosphate (cyclic GMP) and their plethora of downstream protein kinase effectors serve ubiquitous roles not only in cardiovascular homeostasis but also in the pathogenesis of CVD. Already a major target for clinical pharmacotherapy for CVD as well as other pathologies, novel and potentially clinically appealing actions of cyclic nucleotides and their downstream targets are still being discovered. With this in mind, this review article focuses on our current state of knowledge of the cyclic nucleotide-driven serine (Ser)/threonine (Thr) protein kinases in CVD with particular emphasis on cyclic AMP-dependent protein kinase (PKA) and cyclic GMP-dependent protein kinase (PKG). Attention is given to the regulatory interactions of these kinases with inflammatory components including interleukin 6 signals, with G protein-coupled receptor and growth factor signals, and with growth and synthetic transcriptional platforms underlying CVD pathogenesis. This article concludes with a brief discussion of potential future directions and highlights the importance for continued basic science and clinical study of cyclic nucleotide-directed protein kinases as emerging and crucial controllers of cardiac and vascular disease pathologies.
Collapse
Affiliation(s)
- Nathan A Holland
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Jake T Francisco
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Sean C Johnson
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Joshua S Morgan
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Troy J Dennis
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - Nishitha R Gadireddy
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| | - David A Tulis
- Department of Physiology, Brody School of Medicine, East Carolina University, 600 Moye Boulevard, Greenville, NC 27834, USA.
| |
Collapse
|
25
|
Wang Y, Del Borgo M, Lee HW, Baraldi D, Hirmiz B, Gaspari TA, Denton KM, Aguilar MI, Samuel CS, Widdop RE. Anti-fibrotic Potential of AT 2 Receptor Agonists. Front Pharmacol 2017; 8:564. [PMID: 28912715 PMCID: PMC5583590 DOI: 10.3389/fphar.2017.00564] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 08/09/2017] [Indexed: 12/23/2022] Open
Abstract
There are a number of therapeutic targets to treat organ fibrosis that are under investigation in preclinical models. There is increasing evidence that stimulation of the angiotensin II type 2 receptor (AT2R) is a novel anti-fibrotic strategy and we have reviewed the published in vivo preclinical data relating to the effects of compound 21 (C21), which is the only nonpeptide AT2R agonist that is currently available for use in chronic preclinical studies. In particular, the differential influence of AT2R on extracellular matrix status in various preclinical fibrotic models is discussed. Collectively, these studies demonstrate that pharmacological AT2R stimulation using C21 decreases organ fibrosis, which has been most studied in the setting of cardiovascular and renal disease. In addition, AT2R-mediated anti-inflammatory effects may contribute to the beneficial AT2R-mediated anti-fibrotic effects seen in preclinical models.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Mark Del Borgo
- Department of Biochemistry and Molecular Biology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Huey W Lee
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Dhaniel Baraldi
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Baydaa Hirmiz
- Department of Biochemistry and Molecular Biology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Tracey A Gaspari
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Kate M Denton
- Department of Physiology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Marie-Isabel Aguilar
- Department of Biochemistry and Molecular Biology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Chrishan S Samuel
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| | - Robert E Widdop
- Department of Pharmacology, Cardiovascular Disease Program, Biomedicine Discovery Institute, Monash University, ClaytonVIC, Australia
| |
Collapse
|
26
|
Nio Y, Tanaka M, Hirozane Y, Muraki Y, Okawara M, Hazama M, Matsuo T. Phosphodiesterase 4 inhibitor and phosphodiesterase 5 inhibitor combination therapy has antifibrotic and anti‐inflammatory effects in mdx mice with Duchenne muscular dystrophy. FASEB J 2017; 31:5307-5320. [DOI: 10.1096/fj.201700249r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 07/25/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Yasunori Nio
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masayuki Tanaka
- Inflammation Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yoshihiko Hirozane
- Biomolecular Research LaboratoriesPharmaceutical Research DivisionTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Yo Muraki
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Mitsugi Okawara
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Masatoshi Hazama
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| | - Takanori Matsuo
- Extra Value Generation and General Medicine Drug Discovery UnitTakeda Pharmaceutical Company Limited Fujisawa Japan
| |
Collapse
|
27
|
Schinner E, Wetzl V, Schramm A, Kees F, Sandner P, Stasch JP, Hofmann F, Schlossmann J. Inhibition of the TGFβ signalling pathway by cGMP and cGMP-dependent kinase I in renal fibrosis. FEBS Open Bio 2017; 7:550-561. [PMID: 28396839 PMCID: PMC5377407 DOI: 10.1002/2211-5463.12202] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/16/2017] [Accepted: 01/23/2017] [Indexed: 11/29/2022] Open
Abstract
Agents that enhance production of nitric oxide (NO) and cyclic guanosine monophosphate (cGMP) ameliorate the progression of renal fibrosis. However, the molecular mechanism of this process is not fully understood. We hypothesize that the antifibrotic effects of cGMP and cGMP‐dependent kinase I (cGKI) are mediated via regulation of the TGFβ signalling pathway, both via ERK and the Smad‐dependent route. Kidney fibrosis was induced by unilateral ureter obstruction (UUO) in wild‐type and cGKI‐deficient (cGKI‐KO) mice. The cGMP/cGKI signalling pathway was activated by application of the soluble guanylate cyclase (sGC) stimulator BAY 41‐8543 (BAY), beginning 1 day after UUO. After 7 days, the antifibrotic effects of BAY were analysed by measuring mRNA and protein expression of characteristic fibrotic biomarkers. The effects of cGMP/TGFβ on cultured fibroblasts were also analysed in vitro. BAY application influenced the activity of the extracellular matrix (ECM)‐degrading matrix metalloproteases (MMP2 and MMP9) and their inhibitor tissue inhibitors of metalloproteinase‐1, the secretion of cytokines (e.g. IL‐6) and the expression pattern of ECM proteins (e.g. collagen, fibronectin) and profibrotic mediators (e.g. connective tissue growth factors and plasminogen‐activator inhibitor‐1). Activation of the cGMP/cGKI signalling pathway showed protective effects against fibrosis which were mediated by inhibition of P‐Erk1/2 and translocation of P‐smad3. The elucidation of these signalling mechanisms might support the development of new therapeutic options regarding cGMP/cGKI‐mediated antifibrotic actions.
Collapse
Affiliation(s)
- Elisabeth Schinner
- Department of Pharmacology and Toxicology University of Regensburg Germany
| | - Veronika Wetzl
- Department of Pharmacology and Toxicology University of Regensburg Germany; Novartis Pharma GmbH Nuremberg Germany
| | - Andrea Schramm
- Department of Pharmacology and Toxicology University of Regensburg Germany
| | - Frieder Kees
- Department of Pharmacology and Toxicology University of Regensburg Germany
| | | | | | - Franz Hofmann
- Institute of Pharmacology and Toxicology Technical University of Munich Germany
| | - Jens Schlossmann
- Department of Pharmacology and Toxicology University of Regensburg Germany
| |
Collapse
|
28
|
Atrial natriuretic peptide protects against bleomycin-induced pulmonary fibrosis via vascular endothelial cells in mice : ANP for pulmonary fibrosis. Respir Res 2017; 18:1. [PMID: 28049526 PMCID: PMC5210263 DOI: 10.1186/s12931-016-0492-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2016] [Accepted: 12/14/2016] [Indexed: 12/20/2022] Open
Abstract
Background Pulmonary fibrosis is a life-threatening disease characterized by progressive dyspnea and worsening pulmonary function. Atrial natriuretic peptide (ANP), a heart-derived secretory peptide used clinically in Japan for the treatment of acute heart failure, exerts a wide range of protective effects on various organs, including the heart, blood vessels, kidneys, and lungs. Its therapeutic properties are characterized by anti-inflammatory and anti-fibrotic activities mediated by the guanylyl cyclase-A (GC-A) receptor. We hypothesized that ANP would have anti-fibrotic and anti-inflammatory effects on bleomycin (BLM)-induced pulmonary fibrosis in mice. Methods Mice were divided into three groups: normal control, BLM with vehicle, and BLM with ANP. ANP (0.5 μg/kg/min via osmotic-pump, subcutaneously) or vehicle administration was started before BLM administration (1 mg/kg) and continued until the mice were sacrificed. At 7 or 21 days after BLM administration, fibrotic changes and infiltration of inflammatory cells in the lungs were assessed based on histological findings and analysis of bronchoalveolar lavage fluid. In addition, fibrosis and inflammation induced by BLM were evaluated in vascular endothelium-specific GC-A overexpressed mice. Finally, attenuation of transforming growth factor-β (TGF-β) signaling by ANP was studied using immortalized mouse endothelial cells stably expressing GC-A receptor. Results ANP significantly decreased lung fibrotic area and infiltration of inflammatory cells in lungs after BLM administration. Furthermore, similar effects of ANP were observed in vascular endothelium–specific GC-A overexpressed mice. In cultured mouse endothelial cells, ANP reduced phosphorylation of Smad2 after TGF-β stimulation. Conclusions ANP exerts protective effects on BLM-induced pulmonary fibrosis via vascular endothelial cells.
Collapse
|
29
|
Xiao PJ, Mitchell AM, Huang L, Li C, Samulski RJ. Disruption of Microtubules Post-Virus Entry Enhances Adeno-Associated Virus Vector Transduction. Hum Gene Ther 2016; 27:309-24. [PMID: 26942476 DOI: 10.1089/hum.2016.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perinuclear retention of viral particles is a poorly understood phenomenon observed during many virus infections. In this study, we investigated whether perinuclear accumulation acts as a barrier to limit recombinant adeno-associated virus (rAAV) transduction. After nocodazole treatment to disrupt microtubules at microtubule-organization center (MT-MTOC) after virus entry, we observed higher rAAV transduction. To elucidate the role of MT-MTOC in rAAV infection and study its underlying mechanisms, we demonstrated that rAAV's perinuclear localization was retained by MT-MTOC with fluorescent analysis, and enhanced rAAV transduction from MT-MTOC disruption was dependent on the rAAV capsid's nuclear import signals. Interestingly, after knocking down RhoA or inhibiting its downstream effectors (ROCK and Actin), MT-MTOC disruption failed to increase rAAV transduction or nuclear entry. These data suggest that enhancement of rAAV transduction is the result of increased trafficking to the nucleus via the RhoA-ROCK-Actin pathway. Ten-fold higher rAAV transduction was also observed by disrupting MT-MTOC in brain, liver, and tumor in vivo. In summary, this study indicates that virus perinuclear accumulation at MT-MTOC is a barrier-limiting parameter for effective rAAV transduction and defines a novel defense mechanism by which host cells restrain viral invasion.
Collapse
Affiliation(s)
- Ping-Jie Xiao
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,2 Cell and Developmental Biology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Angela M Mitchell
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,3 Department of Microbiology and Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Lu Huang
- 4 Department of Statistics, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Chengwen Li
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - R Jude Samulski
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,5 Department of Pharmacology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| |
Collapse
|
30
|
Jung ES, Lee J, Heo NJ, Kim S, Kim DK, Joo KW, Han JS. Low-dose paclitaxel ameliorates renal fibrosis by suppressing transforming growth factor-β1-induced plasminogen activator inhibitor-1 signaling. Nephrology (Carlton) 2016; 21:574-82. [DOI: 10.1111/nep.12747] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 01/11/2016] [Accepted: 02/09/2016] [Indexed: 12/17/2022]
Affiliation(s)
- Eun Sook Jung
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| | - Jeonghwan Lee
- Department of Internal Medicine; Hallym University Hangang Sacred Heart Hospital; Seoul Korea
| | - Nam Ju Heo
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| | - Sejoong Kim
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| | - Dong Ki Kim
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| | - Kwon Wook Joo
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| | - Jin Suk Han
- Department of Internal Medicine; College of Medicine, Seoul National University; Seoul Korea
| |
Collapse
|
31
|
Milara J, Escrivá J, Ortiz JL, Juan G, Artigues E, Morcillo E, Cortijo J. Vascular effects of sildenafil in patients with pulmonary fibrosis and pulmonary hypertension: an ex vivo/in vitro study. Eur Respir J 2016; 47:1737-49. [DOI: 10.1183/13993003.01259-2015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 01/23/2016] [Indexed: 12/15/2022]
Abstract
Sildenafil improves the 6-min walking distance in patients with idiopathic pulmonary fibrosis (IPF) and right-sided ventricular systolic dysfunction.We analysed the previously unexplored role of sildenafil on vasoconstriction and remodelling of pulmonary arteries from patients with IPF and pulmonary hypertension (PH) ex vivo. Pulmonary arteries from 18 donors without lung disease, nine IPF, eight PH+IPF and four PH patients were isolated to measure vasodilator and anti-contractile effects of sildenafil in isometric organ bath. Ventilation/perfusion was explored in an animal model of bleomycin lung fibrosis.Sildenafil relaxed serotonin (5-HT) pre-contracted pulmonary arteries in healthy donors and IPF patients and, to a lesser extent, in PH+IPF and PH. Sildenafil inhibited 5-HT dose-response contraction curve mainly in PH+IPF and PH, but not in healthy donors. Sildenafil did not impair the ventilation/perfusion mismatching induced by bleomycin. Pulmonary arteries from PH+IPF patients showed a marked expression of phosphodiesterse-5 and extracellular matrix components. Sildenafil inhibited pulmonary artery endothelial and smooth muscle cell to mesenchymal transition by inhibition of extracellular regulated kinases 1 and 2 (ERK1/2) and SMAD3 phosphorylation.These results suggest an absence of direct relaxant effect and a prominent anti-contractile and anti-remodelling role of sildenafil in PH+IPF pulmonary arteries that could explain the beneficial effects of sildenafil in IPF with PH phenotype.
Collapse
|
32
|
Sen A, Kumar P, Garg R, Lindsey SH, Katakam PVG, Bloodworth M, Pandey KN. Transforming growth factor β1 antagonizes the transcription, expression and vascular signaling of guanylyl cyclase/natriuretic peptide receptor A - role of δEF1. FEBS J 2016; 283:1767-81. [PMID: 26934489 DOI: 10.1111/febs.13701] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 01/20/2016] [Accepted: 02/29/2016] [Indexed: 12/19/2022]
Abstract
The objective of this study was to determine the role of transforming growth factor β1 (TGF-β1) in transcriptional regulation and function of the guanylyl cyclase A/natriuretic peptide receptor A gene (Npr1) and whether cross-talk exists between these two hormonal systems in target cells. After treatment of primary cultured rat thoracic aortic vascular smooth muscle cells and mouse mesangial cells with TGF-β1, the Npr1 promoter construct containing a δ-crystallin enhancer binding factor 1 (δEF1) site showed 85% reduction in luciferase activity in a time- and dose-dependent manner. TGF-β1 also significantly attenuated luciferase activity of the Npr1 promoter by 62%, and decreased atrial natriuretic peptide-mediated relaxation of mouse denuded aortic rings ex vivo. Treatment of cells with TGF-β1 increased the protein levels of δEF1 by 2.4-2.8-fold, and also significantly enhanced the phosphorylation of Smad 2/3, but markedly reduced Npr1 mRNA and receptor protein levels. Over-expression of δEF1 showed a reduction in Npr1 promoter activity by 75%, while deletion or site-directed mutagenesis of δEF1 sites in the Npr1 promoter eliminated the TGF-β1-mediated repression of Npr1 transcription. TGF-β1 significantly increased the expression of α-smooth muscle actin and collagen type I α2 in rat thoracic aortic vascular smooth muscle cells, which was markedly attenuated by atrial natriuretic peptide in cells over-expressing natriuretic peptide receptor A. Together, the present results suggest that an antagonistic cascade exists between the TGF-β1/Smad/δEF1 pathways and Npr1 expression and receptor signaling that is relevant to renal and vascular remodeling, and may be critical in the regulation of blood pressure and cardiovascular homeostasis.
Collapse
Affiliation(s)
- Anagha Sen
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Prerna Kumar
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Renu Garg
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Sarah H Lindsey
- Department of Pharmacology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Prasad V G Katakam
- Department of Pharmacology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Meaghan Bloodworth
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| | - Kailash N Pandey
- Department of Physiology, Tulane University Health Sciences Center and School of Medicine, New Orleans, LA, USA
| |
Collapse
|
33
|
Schleede J, Blair SS. The Gyc76C Receptor Guanylyl Cyclase and the Foraging cGMP-Dependent Kinase Regulate Extracellular Matrix Organization and BMP Signaling in the Developing Wing of Drosophila melanogaster. PLoS Genet 2015; 11:e1005576. [PMID: 26440503 PMCID: PMC4595086 DOI: 10.1371/journal.pgen.1005576] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/16/2015] [Indexed: 12/30/2022] Open
Abstract
The developing crossveins of the wing of Drosophila melanogaster are specified by long-range BMP signaling and are especially sensitive to loss of extracellular modulators of BMP signaling such as the Chordin homolog Short gastrulation (Sog). However, the role of the extracellular matrix in BMP signaling and Sog activity in the crossveins has been poorly explored. Using a genetic mosaic screen for mutations that disrupt BMP signaling and posterior crossvein development, we identify Gyc76C, a member of the receptor guanylyl cyclase family that includes mammalian natriuretic peptide receptors. We show that Gyc76C and the soluble cGMP-dependent kinase Foraging, likely linked by cGMP, are necessary for normal refinement and maintenance of long-range BMP signaling in the posterior crossvein. This does not occur through cell-autonomous crosstalk between cGMP and BMP signal transduction, but likely through altered extracellular activity of Sog. We identify a novel pathway leading from Gyc76C to the organization of the wing extracellular matrix by matrix metalloproteinases, and show that both the extracellular matrix and BMP signaling effects are largely mediated by changes in the activity of matrix metalloproteinases. We discuss parallels and differences between this pathway and other examples of cGMP activity in both Drosophila melanogaster and mammalian cells and tissues. Signaling between cells regulates many processes, including the choices cells make between different fates during development and regeneration, and misregulation of such signaling underlies many human pathologies. To understand how such signals control developmental decisions, it is necessary to elucidate both how cells regulate and respond to different levels of signaling, and how different types of signals combine and regulate each other. We have used genetic screening in the fruitfly Drosophila melanogaster to identify mutations that reduce or eliminate signals carried by Bone Morphogenetic Proteins (BMPs), and show that BMP signaling is sensitive Gyc76C, a peptide receptor that stimulates the production of cGMP in cells. We identify downstream intracellular effectors of this cGMP activity, but provide evidence that the effects on the BMP pathway are not mediated at the intracellular level, but rather through cGMP’s effects upon the extracellular matrix and matrix-remodeling proteinases, which in turn affects the activity of extracellular BMP-binding proteins. We discuss differences and parallels with other examples of cGMP activity in Drosophila melanogaster and mammals.
Collapse
Affiliation(s)
- Justin Schleede
- Department of Zoology, University of Wisconsin, Madison, Wisconsin, United States of America
- Genetics Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Seth S. Blair
- Department of Zoology, University of Wisconsin, Madison, Wisconsin, United States of America
- * E-mail:
| |
Collapse
|
34
|
Qu X, Li X, Zheng Y, Ren Y, Puelles VG, Caruana G, Nikolic-Paterson DJ, Li J. Regulation of renal fibrosis by Smad3 Thr388 phosphorylation. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:944-952. [PMID: 24485922 DOI: 10.1016/j.ajpath.2013.12.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 12/12/2013] [Accepted: 12/19/2013] [Indexed: 01/08/2023]
Abstract
Transforming growth factor-β (TGF-β) promotes tissue fibrosis via receptor-mediated phosphorylation of the receptor-activated Smad2/3, together with Smad4. Of these, Smad3 plays a major profibrotic role in mouse models of tissue fibrosis. Transcriptional activity of the Smad3 protein is regulated by phosphorylation of residues in the C-terminal domain and the linker region. Herein, we examined the role of a novel phosphorylation site within the MH2 domain (T388) in the regulation of Smad3 activity. Confocal microscopy using an Smad3 phosphorylated T388-specific antibody identified phosphorylation of Smad3 T388 in myofibroblasts and tubular epithelial cells in human focal and segmental glomerulosclerosis and mouse models of unilateral ureteric obstruction and diabetic nephropathy, whereas phosphorylated T388 was largely absent in normal kidney. In vitro, TGF-β1 induced phosphorylation of Smad3 T388 in a biphasic pattern. A point mutation of T388/V in an Smad3 construct demonstrated that phosphorylation of T388 promotes Smad3 binding to Smad4 and CDK8, but was not necessary for nuclear translocation. Furthermore, T388 phosphorylation was required for TGF-β-induced collagen I gene promoter activity and extracellular matrix production in cultured fibroblasts. In conclusion, our study identifies phosphorylation of T388 in the Smad3 MH2 domain as an important mechanism that regulates the profibrotic TGF-β/Smad3 signaling pathway, which has direct relevance to human and experimental fibrotic kidney disease.
Collapse
Affiliation(s)
- Xinli Qu
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Xueling Li
- Key Laboratory of Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, China
| | - Yaowu Zheng
- Transgenic Research Center, School of Life Sciences, Northeast Normal University, Changchun, China
| | - Yi Ren
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, Florida
| | - Victor G Puelles
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - Georgina Caruana
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia
| | - David J Nikolic-Paterson
- Department of Nephrology, Monash Health and the Department of Medicine, Monash University, Clayton, Australia
| | - Jinhua Li
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Australia.
| |
Collapse
|
35
|
Pacurari M, Xing D, Hilgers RHP, Guo YY, Yang Z, Hage FG. Endothelial cell transfusion ameliorates endothelial dysfunction in 5/6 nephrectomized rats. Am J Physiol Heart Circ Physiol 2013; 305:H1256-64. [PMID: 23955716 DOI: 10.1152/ajpheart.00132.2013] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Endothelial dysfunction is prevalent in chronic kidney disease. This study tested the hypothesis that transfusion of rat aortic endothelial cells (ECs) ameliorates endothelial dysfunction in a rat model of chronic kidney disease. Male Sprague-Dawley rats underwent sham surgery or 5/6 nephrectomy (Nx). Five weeks after Nx, EC (1.5 × 10(6) cells/rat) or vehicle were transfused intravenously. One week later, vascular reactivity of mesenteric artery was assessed on a wire myograph. Sensitivity of endothelium-dependent relaxation to acetylcholine and maximum vasodilation were impaired by Nx and improved by EC transfusion. Using selective pharmacological nitric oxide synthase isoform inhibitors, we demonstrated that the negative effect of Nx on endothelial function and rescue by EC transfusion are, at least in part, endothelial nitric oxide synthase mediated. Plasma asymmetric dimethylarginine was increased by Nx and decreased by EC transfusion, whereas mRNA expression of dimethylarginine dimethylaminohydrolases 1 (DDAH1) was decreased by Nx and restored by EC transfusion. Immunohistochemical staining confirmed that local expression of DDAH1 is decreased by Nx and increased by EC transfusion. In conclusion, EC transfusion attenuates Nx-induced endothelium-dependent vascular dysfunction by regulating DDAH1 expression and enhancing endothelial nitric oxide synthase activity. These results suggest that EC-based therapy could provide a novel therapeutic strategy to improve vascular function in chronic kidney disease.
Collapse
Affiliation(s)
- Maricica Pacurari
- Vascular Biology and Hypertension Program, Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama; and
| | | | | | | | | | | |
Collapse
|
36
|
Du L, Qu X, Zheng H, Li R, Wang J, Chen M, Zhao P, Zhang Z, Gong K. Reverse Apolipoprotein A-I Mimetic Peptide R-D4F Inhibits Neointimal Formation following Carotid Artery Ligation in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2013; 182:1932-9. [DOI: 10.1016/j.ajpath.2013.01.040] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 12/23/2012] [Accepted: 01/23/2013] [Indexed: 11/26/2022]
|
37
|
Brice A, Moseley GW. Viral interactions with microtubules: orchestrators of host cell biology? Future Virol 2013. [DOI: 10.2217/fvl.12.137] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Viral interaction with the microtubule (MT) cytoskeleton is critical to infection by many viruses. Most data regarding virus–MT interaction indicate key roles in the subcellular transport of virions/viral genomic material to sites of replication, assembly and egress. However, the MT cytoskeleton orchestrates diverse processes in addition to subcellular cargo transport, including regulation of signaling pathways, cell survival and mitosis, suggesting that viruses, expert manipulators of the host cell, may use the virus–MT interface to control multiple aspects of cell biology. Several lines of evidence support this idea, indicating that specific viral proteins can modify MT dynamics and/or structure and regulate processes such as apoptosis and innate immune signaling through MT-dependent mechanisms. Here, the authors review general aspects of virus–MT interactions, with emphasis on viral mechanisms that modify MT dynamics and functions to affect processes beyond virion transport. The emerging importance of discrete viral protein–MT interactions in pathogenic processes indicates that these interfaces may represent new targets for future therapeutics and vaccine development.
Collapse
Affiliation(s)
- Aaron Brice
- Viral Immune Evasion & Pathogenicity Laboratory, Department of Biochemistry & Molecular Biology, Monash University, Victoria 3800, Australia
| | - Gregory W Moseley
- Viral Immune Evasion & Pathogenicity Laboratory, Department of Biochemistry & Molecular Biology, Monash University, Victoria 3800, Australia.
| |
Collapse
|
38
|
HDAC6 deacetylase activity is required for hypoxia-induced invadopodia formation and cell invasion. PLoS One 2013; 8:e55529. [PMID: 23405166 PMCID: PMC3566011 DOI: 10.1371/journal.pone.0055529] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 12/27/2012] [Indexed: 02/06/2023] Open
Abstract
Despite significant progress in the cancer field, tumor cell invasion and metastasis remain a major clinical challenge. Cell invasion across tissue boundaries depends largely on extracellular matrix degradation, which can be initiated by formation of actin-rich cell structures specialized in matrix degradation called invadopodia. Although the hypoxic microenvironment within solid tumors has been increasingly recognized as an important driver of local invasion and metastasis, little is known about how hypoxia influences invadopodia biogenesis. Here, we show that histone deacetylase 6 (HDAC6), a cytoplasmic member of the histone deacetylase family, is a novel modulator of hypoxia-induced invadopodia formation. Hypoxia was found to enhance HDAC6 tubulin deacetylase activity through activation of the EGFR pathway. Activated HDAC6, in turn, triggered Smad3 phosphorylation resulting in nuclear accumulation. Inhibition of HDAC6 activity or knockdown of the protein inhibited both hypoxia-induced Smad3 activation and invadopodia formation. Our data provide evidence that hypoxia influences invadopodia formation in a biphasic manner, which involves the activation of HDAC6 deacetylase activity by EGFR, resulting in enhanced Smad phosphorylation and nuclear accumulation. The identification of HDAC6 as a key participant of hypoxia-induced cell invasion may have important therapeutic implications for the treatment of metastasis in cancer patients.
Collapse
|
39
|
Salama SA, Diaz-Arrastia CR, Kilic GS, Kamel MW. 2-Methoxyestradiol causes functional repression of transforming growth factor β3 signaling by ameliorating Smad and non-Smad signaling pathways in immortalized uterine fibroid cells. Fertil Steril 2012; 98:178-84. [PMID: 22579131 DOI: 10.1016/j.fertnstert.2012.04.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2011] [Revised: 03/30/2012] [Accepted: 04/03/2012] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the effects and the mechanism of action of 2-methoxyestradiol (2ME(2)) on transforming growth factor (TGF) β3-induced profibrotic response in immortalized human uterine fibroid smooth muscle (huLM) cells. DESIGN Laboratory study. SETTING University research laboratory. PATIENTS(S) Not applicable. INTERVENTIONS(S) Not applicable. MAIN OUTCOME MEASURE(S) huLM cells were treated with TGF-β3 (5 ηg/mL) in the presence or absence of specific Smad3 inhibitor SIS3 (1 μmol/L), inhibitor of the PI3K/Akt (LY294002, 10 μmol/L), or 2ME(2) (0.5 μmol/L), and the expression of collagen (Col) type I(αI), Col III(αI), plasminogen activator inhibitor (PAI) 1, connective tissue growth factor (CTGF), and α-smooth muscle actin (α-SMA) were determined by real-time reverse-transcription polymerase chain reaction and immunoblotting. The effect of 2ME(2) on Smad-microtubule binding was evaluated by coimmunoprecipitation. RESULT(S) Our data revealed that TGF-β3-induced fibrogenic response in huLM is mediated by both Smad-dependent and Smad-independent PI3K/Akt/mTOR signaling pathways. 2ME(2) abrogates TGF-β3-induced expression of Col I(αI), Col III(αI), PAI-1, CTGF, and α-SMA. Molecularly, 2ME(2) ameliorates TGF-β3-induced Smad2/3 phosphorylation and nuclear translocation. In addition, 2ME(2) inhibits TGF-β3-induced activation of the PI3K/Akt/mTOR pathway. CONCLUSION(S) TGF-β3-induced profibrotic response in fibroid cells is mediated by Smad-dependent and Smad-independent PI3K/Akt/mTOR pathways. 2ME(2) inhibits TGF-β3 profibrotic effects in huLM cells by ameliorating both Smad-dependent and Smad-independent signaling pathways.
Collapse
Affiliation(s)
- Salama A Salama
- Division of Gynecologic Oncology, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, Texas 77030, USA.
| | | | | | | |
Collapse
|