1
|
Adewale AT, Sharma S, Mouawad JE, Nguyen XX, Bradshaw AD, Feghali-Bostwick C. IGF-II regulates lysyl oxidase propeptide and mediates its effects in part via basic helix-loop-helix E40. Matrix Biol 2024; 132:24-33. [PMID: 38852924 PMCID: PMC11329355 DOI: 10.1016/j.matbio.2024.06.002] [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: 02/29/2024] [Revised: 05/31/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Pulmonary fibrosis (PF) is a clinically severe and commonly fatal complication of Systemic Sclerosis (SSc). Our group has previously reported profibrotic roles for Insulin-like Growth Factor II (IGF-II) and Lysyl Oxidase (LOX) in SSc-PF. We sought to identify downstream regulatory mediators of IGF-II. In the present work, we show that SSc lung tissues have higher baseline levels of the total (N-glycosylated/unglycosylated) LOX-Propeptide (LOX-PP) than control lung tissues. LOX-PP-mediated changes were consistent with the extracellular matrix (ECM) deregulation implicated in SSc-PF progression. Furthermore, Tolloid-like 1 (TLL1) and Bone Morphogenetic Protein 1 (BMP1), enzymes that can cleave ProLOX to release LOX-PP, were increased in SSc lung fibrosis and the bleomycin (BLM)-induced murine lung fibrosis model, respectively. In addition, IGF-II regulated the levels of ProLOX, active LOX, LOX-PP, BMP1, and isoforms of TLL1. The Class E Basic Helix-Loop-Helix protein 40 (BHLHE40) transcription factor localized to the nucleus in response to IGF-II. BHLHE40 silencing downregulated TLL1 isoforms and LOX-PP, and restored features of ECM deregulation triggered by IGF-II. Our findings indicate that IGF-II, BHLHE40, and LOX-PP may serve as targets of therapeutic intervention to halt SSc-PF progression.
Collapse
Affiliation(s)
- Adegboyega Timothy Adewale
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA
| | - Shailza Sharma
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA.
| | - Joe E Mouawad
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA
| | - Xinh-Xinh Nguyen
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA
| | - Amy D Bradshaw
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA
| | - Carol Feghali-Bostwick
- Department of Medicine, Medical University of South Carolina, Charleston 29425, 96 Jonathan Lucas Street, MSC637, SC, USA.
| |
Collapse
|
2
|
Sato F, Bhawal UK, Oikawa K, Muragaki Y. Loss of Dec1 inhibits alcohol-induced hepatic lipid accumulation and circadian rhythm disorder. BMC Mol Cell Biol 2024; 25:1. [PMID: 38166556 PMCID: PMC10763066 DOI: 10.1186/s12860-023-00497-y] [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: 06/28/2023] [Accepted: 12/18/2023] [Indexed: 01/04/2024] Open
Abstract
Chronic alcohol exposure increases liver damage such as lipid accumulation and hepatitis, resulting in hepatic cirrhosis. Chronic alcohol intake is known to disturb circadian rhythms in humans and animals. DEC1, a basic helix-loop-helix transcription factor, plays an important role in the circadian rhythm, inflammation, immune responses, and tumor progression. We have previously shown that Dec1 deficiency inhibits stresses such as periodontal inflammation and perivascular fibrosis of the heart. However, the significance of Dec1 deficiency in chronic alcohol consumption remains unclear. In the present study, we investigated whether the biological stress caused by chronic alcohol intake is inhibited in Dec1 knockout mice. We treated control and Dec1 knockout mice for three months by providing free access to 10% alcohol. The Dec1 knockout mice consumed more alcohol than control mice, however, we observed severe hepatic lipid accumulation and circadian rhythm disturbance in control mice. In contrast, Dec1 knockout mice exhibited little effect on these outcomes. We also investigated the expression of peroxisome proliferator-activated receptors (PPARs) and AMP-activated protein kinase (AMPK), which are involved in the regulation of fatty acid metabolism. Immunohistochemical analysis revealed increases of phosphorylation AMPK and PPARa but decreases PPARg in Dec1 knockout mice compared to that in control mice. This indicates a molecular basis for the inhibition of hepatic lipid accumulation in alcohol-treated Dec1 knockout mice. These results suggest a novel function of Dec1 in alcohol-induced hepatic lipid accumulation and circadian rhythm disorders.
Collapse
Affiliation(s)
- Fuyuki Sato
- Department of Diagnostic Pathology, Shizuoka Cancer Center, Sunto-gun, 411-8777, Japan.
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan.
| | - Ujjal K Bhawal
- Research Institute of Oral Science, Nihon University School of Dentistry at Matsudo, Chiba, 271-8587, Japan
- Center for Global Health Research , Saveetha Medical College and Hospitals , Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, 600077, India
| | - Kosuke Oikawa
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan
| | - Yasuteru Muragaki
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, 641- 8509, Japan
| |
Collapse
|
3
|
Sato F, Kohsaka A, Tanimoto T, Bhawal UK, Muragaki Y. Histological analysis of a Becker muscular dystrophy case, diurnal expression of dystrophin in control mice and decreased expression of dystrophin in Bmal1 knockout mice. Histol Histopathol 2023; 38:165-170. [PMID: 35876434 DOI: 10.14670/hh-18-499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/24/2023]
Abstract
Becker muscular dystrophy (BMD) is a hereditary disease characterized by dystrophin deletion that consequently induces muscle weakness, cardiac hypertrophy and cardiac failure; These conditions are similar to those in Duchenne muscular dystrophy. The circadian rhythm is a physiological phenomenon that is predominantly regulated by the transcription and translation of clock genes. Bmal1 (Brain and muscle Arnt-like protein 1) is one of the core clock genes, and its deficiency disturbs the circadian rhythm, results in cardiac hypertrophy and cardiac failure. Dystrophin expression under diurnal conditions and in Bmal1 deficiency is yet to be elucidated. In this study, we analyzed the heart and lungs sampled during a BMD autopsy. Macroscopical examination revealed a large heart and dilated cardiomyopathy. Microscopical examination revealed an undulated structure, as well as the degeneration, and necrosis of myocardial cells. We also analyzed dystrophin expression in tissues obtained from human autopsies and mice. In human autopsy cases, dystrophin expression was lower in the heart with BMD compared that in the heart with non-BMD hypertrophy. In the heart and muscle of control mice, dystrophin expression was higher at ZT0 than at ZT12. The dystrophin expression was found to be lower in heart-specific Bmal1 knockout mice compared to that in the control mice. Hence, our study indicated that BMD was closely associated with cardiac hypertrophy and cardiac failure, while dystrophin had a diurnal expression pattern in control mice that was regulated by Bmal1.
Collapse
Affiliation(s)
- Fuyuki Sato
- Department of Diagnostic Pathology, Shizuoka Cancer Center, Sunto-gun, Japan. .,Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Akira Kohsaka
- Department of Physiology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Takashi Tanimoto
- Cardiovascular Medicine, Shingu Municipal Medical Center, Wakayama, Japan.,Cardiovascular Medicine, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Ujjal K Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan.,Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Yasuteru Muragaki
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan
| |
Collapse
|
4
|
Sui Y, Jiang H, Kellogg CM, Oh S, Janknecht R. Promotion of colorectal cancer by transcription factor BHLHE40 involves upregulation of ADAM19 and KLF7. Front Oncol 2023; 13:1122238. [PMID: 36890812 PMCID: PMC9986587 DOI: 10.3389/fonc.2023.1122238] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/30/2023] [Indexed: 02/22/2023] Open
Abstract
BHLHE40 is a transcription factor, whose role in colorectal cancer has remained elusive. We demonstrate that the BHLHE40 gene is upregulated in colorectal tumors. Transcription of BHLHE40 was jointly stimulated by the DNA-binding ETV1 protein and two associated histone demethylases, JMJD1A/KDM3A and JMJD2A/KDM4A, which were shown to also form complexes on their own and whose enzymatic activity was required for BHLHE40 upregulation. Chromatin immunoprecipitation assays revealed that ETV1, JMJD1A and JMJD2A interacted with several regions within the BHLHE40 gene promoter, suggesting that these three factors directly control BHLHE40 transcription. BHLHE40 downregulation suppressed both growth and clonogenic activity of human HCT116 colorectal cancer cells, strongly hinting at a pro-tumorigenic role of BHLHE40. Through RNA sequencing, the transcription factor KLF7 and the metalloproteinase ADAM19 were identified as putative BHLHE40 downstream effectors. Bioinformatic analyses showed that both KLF7 and ADAM19 are upregulated in colorectal tumors as well as associated with worse survival and their downregulation impaired HCT116 clonogenic activity. In addition, ADAM19, but not KLF7, downregulation reduced HCT116 cell growth. Overall, these data have revealed a ETV1/JMJD1A/JMJD2A→BHLHE40 axis that may stimulate colorectal tumorigenesis through upregulation of genes such as KLF7 and ADAM19, suggesting that targeting this axis represents a potential novel therapeutic avenue.
Collapse
Affiliation(s)
- Yuan Sui
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Hanlin Jiang
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Collyn M Kellogg
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sangphil Oh
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Stephenson Cancer Center, Oklahoma City, OK, United States
| | - Ralf Janknecht
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States.,Stephenson Cancer Center, Oklahoma City, OK, United States
| |
Collapse
|
5
|
Li X, Zhang X, Hou X, Bing X, Zhu F, Wu X, Guo N, Zhao H, Xu F, Xia M. Obstructive sleep apnea-increased DEC1 regulates systemic inflammation and oxidative stress that promotes development of pulmonary arterial hypertension. Apoptosis 2022; 28:432-446. [PMID: 36484960 DOI: 10.1007/s10495-022-01797-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2022] [Indexed: 12/14/2022]
Abstract
Obstructive sleep apnea (OSA), characterized by chronic intermittent hypoxia (CIH), is a common risk factor for pulmonary arterial hypertension (PAH). As a hypoxia-induced transcription factor, differentially expressed in chondrocytes (DEC1) negatively regulates the transcription of peroxisome proliferative activated receptor-γ (PPARγ), a recognized protective factor of PAH. However, whether and how DEC1 is associated with PAH pathogenesis remains unclear. In the present study, we found that DEC1 was increased in lungs and pulmonary arterial smooth muscle cells (PASMCs) of rat models of OSA-associated PAH. Oxidative indicators and inflammatory cytokines were also elevated in the blood of the rats. Similarly, hypoxia-treated PASMCs displayed enhanced DEC1 expression and reduced PPARγ expression in vitro. Functionally, DEC1 overexpression exacerbated reactive oxygen species (ROS) production and the expression of pro-inflammatory cytokines (such as TNFα, IL-1β, IL-6, and MCP-1) in PASMCs. Conversely, shRNA knockdown of Dec1 increased PPARγ expression but attenuated hypoxia-induced oxidative stress and inflammatory responses in PASMCs. Additionally, DEC1 overexpression promoted PASMC proliferation, which was drastically attenuated by a PPARγ agonist rosiglitazone. Collectively, these results suggest that hypoxia-induced DEC1 inhibits PPARγ, and that this is a predominant mechanism underpinning oxidative stress and inflammatory responses in PASMCs during PAH. DEC1 could be used as a potential target to treat PAH.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiang Zhang
- Department of Pharmacy, Central Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaozhi Hou
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Xin Bing
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Fangyuan Zhu
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Xinhao Wu
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Na Guo
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Hui Zhao
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China
| | - Fenglei Xu
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China.
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital affiliated to Shandong First Medical University, 250021, Jinan, Shandong Province, China.
| |
Collapse
|
6
|
Ren KW, Yu XH, Gu YH, Xie X, Wang Y, Wang SH, Li HH, Bi HL. Cardiac-specific knockdown of Bhlhe40 attenuates angiotensin II (Ang II)-Induced atrial fibrillation in mice. Front Cardiovasc Med 2022; 9:957903. [PMID: 36304536 PMCID: PMC9592817 DOI: 10.3389/fcvm.2022.957903] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 09/20/2022] [Indexed: 11/13/2022] Open
Abstract
Atrial fibrosis and atrial inflammation are associated with the pathogenesis of atrial fibrillation (AF). Basic helix–loop–helix family member E40 (Bhlhe40) is an important transcription factor, which is involved in tumors, inflammation, apoptosis, viral infection, and hypoxia. However, its role and molecular mechanism in AF remain unclear. In this study, a mouse model of AF was induced by Ang II infusion. The atrial diameter was evaluated using echocardiography. Induction and duration of AF were measured by programmed electrical stimulation. Atrial structural remodeling was detected using routine histologic examinations. Our results showed that Bhlhe40 was significantly upregulated in angiotensin II (Ang II)-stimulated atrial cardiomyocytes and atrial tissues and in tissues from patients with AF. Cardiac-specific knockdown of Bhlhe40 in mice by a type 9 recombinant adeno-associated virus (rAAV9)-shBhlhe40 significantly ameliorated Ang II-induced atrial dilatation, atrial fibrosis, and atrial inflammation, as well as the inducibility and duration of AF. Mechanistically, cardiac-specific knockdown of Bhlhe40 attenuated Ang II-induced activation of NF-κB/NLRP3, TGF-1β/Smad2 signals, the increased expression of CX43, and the decreased expression of Kv4.3 in the atria. This is the first study to suggest that Bhlhe40 is a novel regulator of AF progression, and identifying Bhlhe40 may be a new therapeutic target for hypertrophic remodeling and heart failure.
Collapse
Affiliation(s)
- Kai-Wen Ren
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiao-Hong Yu
- Department of Cardiology, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu-Hui Gu
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xin Xie
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yu Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Shi-hao Wang
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Hui-Hua Li
- Department of Emergency Medicine, Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China,*Correspondence: Hui-Hua Li,
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, China,Hai-Lian Bi,
| |
Collapse
|
7
|
DEC1 represses cardiomyocyte hypertrophy by recruiting PRP19 as an E3 ligase to promote ubiquitination-proteasome-mediated degradation of GATA4. J Mol Cell Cardiol 2022; 169:96-110. [PMID: 35659652 DOI: 10.1016/j.yjmcc.2022.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 04/18/2022] [Accepted: 05/12/2022] [Indexed: 12/14/2022]
Abstract
Although the pro-hypertrophic role of GATA binding protein 4 (GATA4) during cardiac hypertrophy has been well established, the negative regulatory mechanism to counteract its hyperactivation remains elusive. We hypothesized that the hyperactivation of GATA4 could be a result of loss of interaction between GATA4 with specific suppressors. Using high throughput mass spectrometry technology, we carried out a proteomic screen for endogenous suppressor of GATA4, which disassociated with GATA4 during the hypertrophic response in a cultured cardiac myoblast cell line (H9C2 cells). We identified differentiated embryo chondrocyte 1 (DEC1) negatively regulated the function of GATA4 through physical interaction and negatively regulated cardiac hypertrophy both in vivo and in vitro. Particularly, DEC1 promoted the ubiquitination and proteasome-mediated degradation of GATA4, but did not function as an E3 ligase. Again, using mass spectrometry technology, we systematically identified pre-mRNA processing factor 19 (PRP19) as a newfound E3 ligase, which promoted the K6-linked ubiquitination of GATA4 at its lysine 256. Functional experiments performed in cultured neonatal rat ventricular myocytes and H9C2 cells demonstrated that both DEC1 and PRP19 negatively regulated agonist-induced cardiomyocyte hypertrophic responses. Furthermore, rescue experiments performed in these cells revealed that DEC1 and PRP19 suppressed cardiomyocyte hypertrophy by inhibiting the function of GATA4. Our study thus defined the novel DEC1-PRP19-GATA4 axis to be a previously unknown mechanism in regulating cardiomyocyte hypertrophy. Although GATA4 is indispensable for normal cardiac function, harnessing DEC1- or PRP19-mediated negative regulation to counteract the hyperactivation of GATA4 might serve as a novel therapeutic strategy for pathological cardiac hypertrophy.
Collapse
|
8
|
Hu X, Zou M, Ni L, Zhang M, Zheng W, Liu B, Cheng Z. Dec1 Deficiency Ameliorates Pulmonary Fibrosis Through the PI3K/AKT/GSK-3β/β-Catenin Integrated Signaling Pathway. Front Pharmacol 2022; 13:829673. [PMID: 35355710 PMCID: PMC8959854 DOI: 10.3389/fphar.2022.829673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/23/2022] [Indexed: 12/14/2022] Open
Abstract
Tissue remodeling/fibrosis is a main feature of idiopathic pulmonary fibrosis (IPF), which results in the replacement of normal lung parenchyma with a collagen-rich extracellular matrix produced by fibroblasts and myofibroblasts. Epithelial-mesenchymal transition (EMT) in type 2 lung epithelial cells is a key process in IPF, which leads to fibroblasts and myofibroblasts accumulation and excessive collagen deposition. DEC1, a structurally distinct class of basic helix-loop-helix proteins, is associated with EMT in cancer. However, the functional role of DEC1 in pulmonary fibrosis (PF) remains elusive. Herein, we aimed to explore DEC1 expression in IPF and bleomycin (BLM)-induced PF in mice and the mechanisms underlying the fibrogenic effect of DEC1 in PF in vivo and in vitro by Dec1-knockout (Dec1 -/-) mice, knockdown and overexpression of DEC1 in alveolar epithelial cells (A549 cells). We found that the expression of DEC1 was increased in IPF and BLM-injured mice. More importantly, Dec1 -/- mice had reduced PF after BLM challenge. Additionally, DEC1 deficiency relieved EMT development and repressed the PI3K/AKT/GSK-3β/β-catenin integrated signaling pathway in mice and in A549 cells, whereas DEC1 overexpression in vitro had converse effects. Moreover, the PI3K/AKT and Wnt/β-catenin signaling inhibitors, LY294002 and XAV-939, ameliorated BLM-meditated PF in vivo and relieved EMT in vivo and in vitro. These pathways are interconnected by the GSK-3β phosphorylation status. Our findings indicated that during PF progression, DEC1 played a key role in EMT via the PI3K/AKT/GSK-3β/β-catenin integrated signaling pathway. Consequently, targeting DEC1 may be a potential novel therapeutic approach for IPF.
Collapse
Affiliation(s)
- Xingxing Hu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Menglin Zou
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Lan Ni
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Mingyang Zhang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Weishuai Zheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bing Liu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhenshun Cheng
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, China.,Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China
| |
Collapse
|
9
|
Li X, Liu C, Qi W, Meng Q, Zhao H, Teng Z, Xu R, Wu X, Zhu F, Qin Y, Zhao M, Xu F, Xia M. Endothelial Dec1-PPARγ Axis Impairs Proliferation and Apoptosis Homeostasis Under Hypoxia in Pulmonary Arterial Hypertension. Front Cell Dev Biol 2021; 9:757168. [PMID: 34765605 PMCID: PMC8576361 DOI: 10.3389/fcell.2021.757168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/27/2021] [Indexed: 11/15/2022] Open
Abstract
Background: The hypoxia-induced pro-proliferative and anti-apoptotic characteristics of pulmonary arterial endothelial cells (PAECs) play critical roles in pulmonary vascular remodeling and contribute to hypoxic pulmonary arterial hypertension (PAH) pathogenesis. However, the mechanism underlying this hypoxic disease has not been fully elucidated. Methods: Bioinformatics was adopted to screen out the key hypoxia-related genes in PAH. Gain- and loss-function assays were then performed to test the identified hypoxic pathways in vitro. Human PAECs were cultured under hypoxic (3% O2) or normoxic (21% O2) conditions. Hypoxia-induced changes in apoptosis and proliferation were determined by flow cytometry and Ki-67 immunofluorescence staining, respectively. Survival of the hypoxic cells was estimated by cell counting kit-8 assay. Expression alterations of the target hypoxia-related genes, cell cycle regulators, and apoptosis factors were investigated by Western blot. Results: According to the Gene Expression Omnibus dataset (GSE84538), differentiated embryo chondrocyte expressed gene 1-peroxisome proliferative-activated receptor-γ (Dec1-PPARγ) axis was defined as a key hypoxia-related signaling in PAH. A negative correlation was observed between Dec1 and PPARγ expression in patients with hypoxic PAH. In vitro observations revealed an increased proliferation and a decreased apoptosis in PAECs under hypoxia. Furthermore, hypoxic PAECs exhibited remarkable upregulation of Dec1 and downregulation of PPARγ. Dec1 was confirmed to be crucial for the imbalance of proliferation and apoptosis in hypoxic PAECs. Furthermore, the pro-surviving effect of hypoxic Dec1 was mediated through PPARγ inhibition. Conclusion: For the first time, Dec1-PPARγ axis was identified as a key determinant hypoxia-modifying signaling that is necessary for the imbalance between proliferation and apoptosis of PAECs. These novel endothelial signal transduction events may offer new diagnostic and therapeutic options for patients with hypoxic PAH.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Chengcheng Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China.,Department of Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Wenwen Qi
- Department of Otolaryngology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiu Meng
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hui Zhao
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhenxiao Teng
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Runtong Xu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xinhao Wu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fangyuan Zhu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yiming Qin
- College of Chemical Engineering and Materials Science, Shandong Normal University, Jinan, China
| | - Miaoqing Zhao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Fenglei Xu
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ming Xia
- Department of Otolaryngology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.,Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
10
|
Methatham T, Tomida S, Kimura N, Imai Y, Aizawa K. Inhibition of the canonical Wnt signaling pathway by a β-catenin/CBP inhibitor prevents heart failure by ameliorating cardiac hypertrophy and fibrosis. Sci Rep 2021; 11:14886. [PMID: 34290289 PMCID: PMC8295328 DOI: 10.1038/s41598-021-94169-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
In heart failure (HF) caused by hypertension, the myocyte size increases, and the cardiac wall thickens. A low-molecular-weight compound called ICG001 impedes β-catenin-mediated gene transcription, thereby protecting both the heart and kidney. However, the HF-preventive mechanisms of ICG001 remain unclear. Hence, we investigated how ICG001 can prevent cardiac hypertrophy and fibrosis induced by transverse aortic constriction (TAC). Four weeks after TAC, ICG001 attenuated cardiac hypertrophy and fibrosis in the left ventricular wall. The TAC mice treated with ICG001 showed a decrease in the following: mRNA expression of brain natriuretic peptide (Bnp), Klf5, fibronectin, β-MHC, and β-catenin, number of cells expressing the macrophage marker CD68 shown in immunohistochemistry, and macrophage accumulation shown in flow cytometry. Moreover, ICG001 may mediate the substrates in the glycolysis pathway and the distinct alteration of oxidative stress during cardiac hypertrophy and HF. In conclusion, ICG001 is a potential drug that may prevent cardiac hypertrophy and fibrosis by regulating KLF5, immune activation, and the Wnt/β-catenin signaling pathway and inhibiting the inflammatory response involving macrophages.
Collapse
Affiliation(s)
- Thanachai Methatham
- grid.410804.90000000123090000Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498 Japan
| | - Shota Tomida
- grid.410804.90000000123090000Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498 Japan
| | - Natsuka Kimura
- grid.410804.90000000123090000Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498 Japan
| | - Yasushi Imai
- grid.410804.90000000123090000Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498 Japan
| | - Kenichi Aizawa
- grid.410804.90000000123090000Division of Clinical Pharmacology, Department of Pharmacology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi, 329-0498 Japan
| |
Collapse
|
11
|
Patel JC, Singh A, Tulswani R, Sharma YK, Khurana P, Ragumani S. Identification of VEGFA-centric temporal hypoxia-responsive dynamic cardiopulmonary network biomarkers. Life Sci 2021; 281:119718. [PMID: 34147483 DOI: 10.1016/j.lfs.2021.119718] [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: 02/23/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 10/21/2022]
Abstract
AIMS Hypoxia, a pathophysiological condition, is profound in several cardiopulmonary diseases (CPD). Every individual's lethality to a hypoxia state differs in terms of hypoxia exposure time, dosage units and dependent on the individual's genetic makeup. Most of the proposed markers for CPD were generally aim to distinguish disease samples from normal samples. Although, as per the 2018 GOLD guidelines, clinically useful biomarkers for several cardio pulmonary disease patients in stable condition have yet to be identified. We attempt to address these key issues through the identification of Dynamic Network Biomarkers (DNB) to detect hypoxia induced early warning signals of CPD before the catastrophic deterioration. MATERIALS AND METHODS The human microvascular endothelial tissues microarray datasets (GSE11341) of lung and cardiac expose to hypoxia (1% O2) for 3, 24 and 48 h were retrieved from the public repository. The time dependent differentially expressed genes were subjected to tissue specificity and promoter analysis to filtrate the noise levels in the networks and to dissect the tissue specific hypoxia induced genes. These filtered out genes were used to construct the dynamic segmentation networks. The hypoxia induced dynamic differentially expressed genes were validated in the lung and heart tissues of male rats. These rats were exposed to hypobaric hypoxia (simulated altitude of 25,000 or PO2 - 282 mm of Hg) progressively for 3, 24 and 48 h. KEY FINDINGS To identify the temporal key genes regulated in hypoxia, we ranked the dominant genes based on their consolidated topological features from tissue specific networks, time dependent networks and dynamic networks. Overall topological ranking described VEGFA as a single node dynamic hub and strongly communicated with tissue specific genes to carry forward their tissue specific information. We named this type of VEGFAcentric dynamic networks as "V-DNBs". As a proof of principle, our methodology helped us to identify the V-DNBs specific for lung and cardiac tissues namely V-DNBL and V-DNBC respectively. SIGNIFICANCE Our experimental studies identified VEGFA, SLC2A3, ADM and ENO2 as the minimum and sufficient candidates of V-DNBL. The dynamic expression patterns could be readily exploited to capture the pre disease state of hypoxia induced pulmonary vascular remodelling. Whereas in V-DNBC the minimum and sufficient candidates are VEGFA, SCL2A3, ADM, NDRG1, ENO2 and BHLHE40. The time dependent single node expansion indicates V-DNBC could also be the pre disease state pathological hallmark for hypoxia-associated cardiovascular remodelling. The network cross-talk and expression pattern between V-DNBL and V-DNBC are completely distinct. On the other hand, the great clinical advantage of V-DNBs for pre disease predictions, a set of samples during the healthy condition should suffice. Future clinical studies might further shed light on the predictive power of V-DNBs as prognostic and diagnostic biomarkers for CPD.
Collapse
Affiliation(s)
- Jai Chand Patel
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Ajeet Singh
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Rajkumar Tulswani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Yogendra Kumar Sharma
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Pankaj Khurana
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India
| | - Sugadev Ragumani
- Defence Institute of Physiology and Allied Sciences, Defence Research and Development Organization, Lucknow Road, Timarpur, Delhi, India.
| |
Collapse
|
12
|
Li X, Le HT, Sato F, Kang TH, Makishima M, Zhong L, Liu Y, Guo L, Bhawal UK. Dec1 deficiency protects the heart from fibrosis, inflammation, and myocardial cell apoptosis in a mouse model of cardiac hypertrophy. Biochem Biophys Res Commun 2020; 532:513-519. [PMID: 32896382 DOI: 10.1016/j.bbrc.2020.08.058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 08/19/2020] [Indexed: 10/23/2022]
Abstract
Cardiac inflammation and fibrosis triggered by left ventricular pressure overload are the major causes of heart dysfunction. Differentiated embryonic chondrocyte gene 1 (Dec1) is a basic helix-loop-helix transcription factor that is comprehensively involved in inflammation and tissue fibrosis, but its role in cardiac hypertrophy remains unclear. This study explored the effects of Dec1 on cardiac fibrosis, inflammation, and apoptosis in hypertrophic conditions. Transverse aortic constriction (TAC) was performed to induce cardiac hypertrophy in wild-type (WT) mice and in Dec1 knock out (KO) mice for 4 weeks. Using the TAC mouse model, prominent differences in cardiac hypertrophy at the morphological, functional, and molecular levels were delineated by Masson's Trichrome and TUNEL staining, immunohistochemistry, RT-PCR and Western Blot. DNA microarray and microRNA (miRNA) array analyses were carried out to identify gene and miRNA expression patterns. Dec1KO mice exhibited a more severe hypertrophic heart, whereas WT mice showed a more pronounced perivascular fibrosis after TAC at 4 weeks. The Dec1 deficiency promoted M2 phenotype macrophages. Dec1KO TAC mice showed fewer apoptotic cells than WT TAC mice. APEX1, WNT16, FGF10 and MMP-10 were differentially expressed according to DNA microarray analysis and expression levels of those genes and the corresponding miRNAs (miR-295, miR-200 b, miR-130a, miR-92a) showed the same trends. Furthermore, luciferase reporter assay confirmed that FGF10 is the direct target gene of miR-130. In conclusion, a Dec1 deficiency protects the heart from perivascular fibrosis, regulates M1/M2 macrophage polarization and reduces cell apoptosis, which may provide a novel insight for the treatment of cardiac hypertrophy.
Collapse
Affiliation(s)
- Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.
| | - Hue Thi Le
- Department of Physiology, Hanoi Medical University, Hanoi, Viet Nam.
| | - Fuyuki Sato
- Pathology Division, Shizuoka Cancer Center, Shizuoka, Japan.
| | - Tong Ho Kang
- Graduate School of Biotechnology, Kyung Hee University, Republic of Korea.
| | - Makoto Makishima
- Division of Biochemistry, Department of Biomedical Sciences, Nihon University, School of Medicine, Tokyo, Japan.
| | - Liangjun Zhong
- Department of Stomatology, Hangzhou Normal University, Hangzhou, China.
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, Beijing, China.
| | - Lijia Guo
- Department of Orthodontics, School of Stomatology, Capital Medical University, Beijing, China.
| | - Ujjal K Bhawal
- Department of Biochemistry and Molecular Biology, Nihon University School of Dentistry at Matsudo, Chiba, Japan.
| |
Collapse
|
13
|
Guo NL, Bello D, Ye Q, Tagett R, Chanetsa L, Singh D, Poh TY, Setyawati MI, Chotirmall SH, Ng KW, Demokritou P. Pilot deep RNA sequencing of worker blood samples from Singapore printing industry for occupational risk assessment. NANOIMPACT 2020; 19:100248. [PMID: 33511305 PMCID: PMC7840153 DOI: 10.1016/j.impact.2020.100248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/09/2023]
Abstract
Several engineered nanomaterials (ENMs) are used in toner-based printing equipment (TPE) including laser printers and photocopiers to improve toner performance. High concentration of airborne nanoparticles due to TPE emissions has been documented in copy centers and chamber studies. Recent animal inhalation studies by our group suggested exposure to laser printer-emitted nanoparticles (PEPs) increased cardiovascular risk by impairing ventricular performance and inducing hypertension and arrhythmia, consistent with global transcriptomic and metabolomic profiling results. There has been no genome-wide transcriptomic analysis of workers exposed to TPE emissions to systematically assess the occupational exposure health risks. In this pilot study, deep RNA sequencing of blood samples of workers in two printing companies in Singapore was performed. The genome-scale analysis of the blood samples from TPE exposed workers revealed perturbed transcriptional activities related to inflammatory and immune responses, metabolism, cardiovascular impairment, neurological diseases, oxidative stress, physical morphogenesis/deformation, and cancer, when compared with the control peers (office workers). Many of these disease risks associated with particle inhalation exposures in such work environments were consistent with the observation from the PEPs rat inhalation studies. In particular, the cell adhesion molecules (CAMs) was a top significantly perturbed pathway in blood samples from exposed workers compared with the office workers in both companies. The protein expression of sICAM was verified in plasma of exposed workers, showing a positive correlation with daily average nanoparticle concentration in indoor air measured in these two companies. Larger scale genomic and molecular epidemiology studies in copier operators are warranted in order to assess potential risks from such particulate matter exposures.
Collapse
Affiliation(s)
- Nancy Lan Guo
- West Virginia University Cancer Institute/School of Public Health, West Virginia University, Morgantown, WV 26506
- Correspondence: ; Tel: 1-304-293-6455
| | - Dhimiter Bello
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts, Lowell, MA 01854
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Qing Ye
- Lane Department of Computer Science and Electrical Engineering, Statler College of Engineering and Mineral Resources, West Virginia University, Morgantown, WV 26506
| | - Rebecca Tagett
- BRCF Bioinformatics Core, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Lucia Chanetsa
- Department of Biomedical and Nutritional Sciences, Zuckerberg College of Health Sciences, University of Massachusetts, Lowell, MA 01854
| | - Dilpreet Singh
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Tuang Yeow Poh
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Magdiel Inggrid Setyawati
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Sanjay H. Chotirmall
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Kee Woei Ng
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Environmental Chemistry and Materials Center, Nanyang Environment & Water Research Institute, Singapore 637141, Singapore
| | - Philip Demokritou
- Center for Nanotechnology and Nanotoxicology, Department of Environmental Health, T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|