1
|
He X, Huang S, Yu C, Chen Y, Zhu H, Li J, Chen S. Mst1/Hippo signaling pathway drives isoproterenol-induced inflammatory heart remodeling. Int J Med Sci 2024; 21:1718-1729. [PMID: 39006833 PMCID: PMC11241096 DOI: 10.7150/ijms.95850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 06/03/2024] [Indexed: 07/16/2024] Open
Abstract
Isoproterenol (ISO) administration is a well-established model for inducing myocardial injury, replicating key features of human myocardial infarction (MI). The ensuing inflammatory response plays a pivotal role in the progression of adverse cardiac remodeling, characterized by myocardial dysfunction, fibrosis, and hypertrophy. The Mst1/Hippo signaling pathway, a critical regulator of cellular processes, has emerged as a potential therapeutic target in cardiovascular diseases. This study investigates the role of Mst1 in ISO-induced myocardial injury and explores its underlying mechanisms. Our findings demonstrate that Mst1 ablation in cardiomyocytes attenuates ISO-induced cardiac dysfunction, preserving cardiomyocyte viability and function. Mechanistically, Mst1 deletion inhibits cardiomyocyte apoptosis, oxidative stress, and calcium overload, key contributors to myocardial injury. Furthermore, Mst1 ablation mitigates endoplasmic reticulum (ER) stress and mitochondrial fission, both of which are implicated in ISO-mediated cardiac damage. Additionally, Mst1 plays a crucial role in modulating the inflammatory response following ISO treatment, as its deletion suppresses pro-inflammatory cytokine expression and neutrophil infiltration. To further investigate the molecular mechanisms underlying ISO-induced myocardial injury, we conducted a bioinformatics analysis using the GSE207581 dataset. GO and KEGG pathway enrichment analyses revealed significant enrichment of genes associated with DNA damage response, DNA repair, protein ubiquitination, chromatin organization, autophagy, cell cycle, mTOR signaling, FoxO signaling, ubiquitin-mediated proteolysis, and nucleocytoplasmic transport. These findings underscore the significance of Mst1 in ISO-induced myocardial injury and highlight its potential as a therapeutic target for mitigating adverse cardiac remodeling. Further investigation into the intricate mechanisms of Mst1 signaling may pave the way for novel therapeutic interventions for myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Xiuling He
- Department of Cardiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, 100048, China
| | - Shuai Huang
- Department of Cardio-Thoracic Surgery, The Third Affiliated Hospital, Sun Yat-Sen University, 510630, Guangzhou, China
| | - Chijia Yu
- Department of Clinical Laboratory Medicine, The First Medical Centre, Medical School of Chinese PLA, Beijing, China
| | - Ye Chen
- Department of Cardiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Hang Zhu
- Department of Cardiology, School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Cardiology, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing, 100048, China
| | - Jianwei Li
- Department of Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan 528403, China
| | - Shanshan Chen
- Department of Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan 528403, China
| |
Collapse
|
2
|
Ushida T, Cotechini T, Protopapas N, Atallah A, Collyer C, Toews AJ, Macdonald-Goodfellow SK, Tse MY, Winn LM, Pang SC, Adams MA, Othman M, Kotani T, Kajiyama H, Graham CH. Aberrant inflammation in rat pregnancy leads to cardiometabolic alterations in the offspring and intrauterine growth restriction in the F2 generation. J Dev Orig Health Dis 2022; 13:706-718. [PMID: 35593438 DOI: 10.1017/s2040174422000265] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Children of women with pre-eclampsia have increased risk of cardiovascular (CV) and metabolic disease in adult life. Furthermore, the risk of pregnancy complications is higher in daughters born to women affected by pre-eclampsia than in daughters born after uncomplicated pregnancies. While aberrant inflammation contributes to the pathophysiology of pregnancy complications, including pre-eclampsia, the contribution of maternal inflammation to subsequent risk of CV and metabolic disease as well as pregnancy complications in the offspring remains unclear. Here, we demonstrate that 24-week-old female rats (F1) born to dams (F0) exposed to lipopolysaccharide (LPS) during pregnancy (to induce inflammation) exhibited mild systolic dysfunction, increased cardiac growth-related gene expression, altered glucose tolerance, and coagulopathy; whereas male F1 offspring exhibited altered glucose tolerance and increased visceral fat accumulation compared with F1 sex-matched offspring born to saline-treated dams. Both male and female F1 offspring born to LPS-treated dams had evidence of anemia. Fetuses (F2) from F1 females born to LPS-treated dams were growth restricted, and this reduction in fetal growth was associated with increased CD68 positivity (indicative of macrophage presence) and decreased expression of glucose transporter-1 in their utero-placental units. These results indicate that abnormal maternal inflammation can contribute to increased risk of CV and metabolic disease in the offspring, and that the effects of inflammation may cross generations. Our findings provide evidence in support of early screening for CV and metabolic disease, as well as pregnancy complications in offspring affected by pre-eclampsia or other pregnancy complications associated with aberrant inflammation.
Collapse
Affiliation(s)
- Takafumi Ushida
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nicole Protopapas
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Aline Atallah
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Charlotte Collyer
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Alexa J Toews
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | | | - M Yat Tse
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Louise M Winn
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Stephen C Pang
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Michael A Adams
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Maha Othman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
- School of Baccalaureate Nursing, St. Lawrence College, Kingston, Ontario, Canada
| | - Tomomi Kotani
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroaki Kajiyama
- Department of Gynecology and Obstetrics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Charles H Graham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
3
|
Wu M, Xing Q, Duan H, Qin G, Sang N. Suppression of NADPH oxidase 4 inhibits PM 2.5-induced cardiac fibrosis through ROS-P38 MAPK pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 837:155558. [PMID: 35504386 DOI: 10.1016/j.scitotenv.2022.155558] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 04/14/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Fine particulate matter (PM2.5) has been consistently linked to cardiovascular diseases, and cardiac fibrosis plays a crucial role in the occurrence and development of heart diseases. It is reported that NOX4-dependent redox signaling are responsible for TGFβ-mediated profibrotic responses. The current study was designed to explore the possible mechanisms of cardiac fibrosis by PM2.5 both in vitro and in vivo. Female C57BL/6 mice received PM2.5 (3 mg/kg b.w.) exposure with/without NOX4 inhibitor (apocynin, 25 mg/kg b.w.) or ROS scavenger (NALC, 50 mg/kg b.w.), every other day, for 4 weeks. H9C2 cells were incubated with PM2.5 (3 μg/mL) with/without 5 mM NALC, TGFβ inhibitor (SB431542, 10 μM), or siRNA-NOX4 for 24 h. The results demonstrated that PM2.5 induced evident collagen deposition and elevated expression of fibrosis biomarkers (Col1a1 & Col3a1). Significant systemic inflammatory response and cardiac oxidative stress were triggered by PM2.5. PM2.5 increased the protein expression of TGFβ1, NOX4, and P38 MAPK. Notably, the increased effects of PM2.5 could be suppressed by SB431542, siRNA-NOX4 in vitro or apocynin in vivo, and NALC. The reverse verification experiments further supported the involvement of the TGFβ/NOX4/ROS/P38 MAPK signaling pathway in the myocardial fibrosis induced by PM2.5. In summary, the current study provided evidence that PM2.5 challenge led to cardiac fibrosis through oxidative stress, systemic inflammation, and subsequent TGFβ/NOX4/ROS/P38 MAPK pathway and may offer new therapeutic targets in cardiac fibrosis.
Collapse
Affiliation(s)
- Meiqiong Wu
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China; Department of Children and Adolescences Health, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi 030001, PR China.
| | - Qisong Xing
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Huiling Duan
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China
| | - Guohua Qin
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, Shanxi 030006, PR China.
| |
Collapse
|
4
|
Ding N, Zheng C. Secreted frizzled-related protein 5 promotes angiogenesis of human umbilical vein endothelial cells and alleviates myocardial injury in diabetic mice with myocardial infarction by inhibiting Wnt5a/JNK signaling. Bioengineered 2022; 13:11656-11667. [PMID: 35506262 PMCID: PMC9275896 DOI: 10.1080/21655979.2022.2070964] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The purpose of this study is to investigate whether secreted frizzled-related protein 5 (SFRP5) affects the proliferation, migration and angiogenesis of human umbilical vein endothelial cells (HUVECs) induced by high glucose (HG). HUVECs were treated with different concentrations of glucose. MTT, wound healing, angiogenesis, and ELISA assays were used to detect cell cytotoxicity, migration, tube formation, and VEGF165 and VEGF165b levels, respectively. The mice model of type 2 diabetes mellitus (T2DM) complicated with myocardial infarction (MI) was established. SFRP5 was injected intrabitoneally for 2 weeks. cardiac output (CO), left ventricular ejection fraction (LVEF) and left ventricular shortening fraction (LVSF) were detected by echocardiography. Western blot was used to detect the protein levels of SFRP5, Wnt5a, JNK1/2/3, p-JNK1/2/3, TGF-β1, Caspase3, Bax, and Bcl-2. The expression of SFRP5 was declined in HG-induced HUVECs and T2DM-MI. Intervention of SFRP5 promoted the migration of HUVECs and angiogenesis, as evidenced by a lower expression of Bax and caspase3, but a higher expression of Bcl-2. Meanwhile, SFRP5 inhibition repress Wnt5a and p-JNK expression. Howerver, The JNK inhibitor (SP600125) enhanced the down-regulation of Wnt5a/JNK pathway proteins by SFRP5. SFRP5 intervention increased the levels of CO, LVSF, and LVEF in T2DM-MI mice. SFRP5 inhibited myocardial pathological injury and fibrosis in T2DM-MI mice and SFRP5 could down-regulate Wnt5a and p-JNK1/2/3 activation. SFRP5 promotes the proliferation, migration and angiogenesis of HUVECs induced by HG, and inhibits cardiac dysfunction, pathological damage, fibrosis, and myocardial angiogenesis in diabetic myocardial ischemia mice, which is achieved by inhibiting Wnt5a/JNK signaling.
Collapse
Affiliation(s)
- Nian Ding
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Chenghong Zheng
- Clinical College of Traditional Chinese medicine, Hubei University of Chinese Medicine, Wuhan, China.,Medical Ward, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China
| |
Collapse
|
5
|
Wu S, Wang S, Wang L, Peng H, Zhang S, Yang Q, Huang M, Li Y, Guan S, Jiang W, Zhang Z, Bi Q, Li L, Gao Y, Xiong P, Zhong Z, Xu B, Deng Y, Deng Y. Docosahexaenoic acid supplementation represses the early immune response against murine cytomegalovirus but enhances NK cell effector function. BMC Immunol 2022; 23:17. [PMID: 35439922 PMCID: PMC9017742 DOI: 10.1186/s12865-022-00492-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 04/12/2022] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Docosahexaenoic acid (DHA) supplementation is beneficial for several chronic diseases; however, its effect on immune regulation is still debated. Given the prevalence of cytomegalovirus (CMV) infection and because natural killer (NK) cells are a component of innate immunity critical for controlling CMV infection, the current study explored the effect of a DHA-enriched diet on susceptibility to murine (M) CMV infection and the NK cell effector response to MCMV infection. RESULTS Male C57BL/6 mice fed a control or DHA-enriched diet for 3 weeks were infected with MCMV and sacrificed at the indicated time points postinfection. Compared with control mice, DHA-fed mice had higher liver and spleen viral loads at day 7 postinfection, but final MCMV clearance was not affected. The total numbers of NK cells and their terminal mature cell subset (KLRG1+ and Ly49H+ NK cells) were reduced compared with those in control mice at day 7 postinfection but not day 21. DHA feeding resulted in higher IFN-γ and granzyme B expression in splenic NK cells at day 7 postinfection. A mechanistic analysis showed that the splenic NK cells of DHA-fed mice had enhanced glucose uptake, increased CD71 and CD98 expression, and higher mitochondrial mass than control mice. In addition, DHA-fed mice showed reductions in the total numbers and activation levels of CD4+ and CD8+ T cells. CONCLUSIONS These results suggest that DHA supplementation represses the early response to CMV infection but preserves NK cell effector functions by improving mitochondrial activity, which may play critical roles in subsequent MCMV clearance.
Collapse
Affiliation(s)
- Shuting Wu
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Shanshan Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Lili Wang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Hongyan Peng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Shuju Zhang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Qinglan Yang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Minghui Huang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Yana Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Shuzhen Guan
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Wenjuan Jiang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Zhaohui Zhang
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Qinghua Bi
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Liping Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Yuan Gao
- Southwest Hospital/Southwest Eye Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Peiwen Xiong
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China
| | - Zhaoyang Zhong
- Cancer Center, Daping Hospital and Research Institute of Surgery, Army Medical University (Third Military Medical University), Chongqing, 400042, People's Republic of China
| | - Bo Xu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, 221002, People's Republic of China.
| | - Yafei Deng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, Changsha, Hunan, People's Republic of China.
- Pediatric Intensive Care Unit, Hunan Children's Hospital, University of South China, Changsha, Hunan, People's Republic of China.
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
6
|
Guo Y, Zhang BY, Peng YF, Chang LC, Li ZQ, Zhang XX, Zhang DJ. Mechanism of Action of Flavonoids of Oxytropis falcata on the Alleviation of Myocardial Ischemia–Reperfusion Injury. Molecules 2022; 27:molecules27051706. [PMID: 35268807 PMCID: PMC8911915 DOI: 10.3390/molecules27051706] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 11/16/2022] Open
Abstract
Oxytropis falcata Bunge is a plant used in traditional Tibetan medicine, with reported anti-inflammatory and antioxidants effects and alleviation of myocardial ischemia reperfusion injury (MIRI). However, the underlying mechanism against MIRI and the phytochemical composition of O. falcata are vague. One fraction named OFF1 with anti-MIRI activity was obtained from O. falcata, and the chemical constituents were identified by ultra-high-performance liquid chromatography coupled with tandem mass spectrometry (UHPLC–MS). The potential targets and signaling pathways involved in the action of O. falcata against MIRI were predicted by network pharmacology analysis, and its molecular mechanism on MIRI was determined by in vitro assays. The results revealed that flavonoids are the dominant constituents of OFF1. A total of 92 flavonoids reported in O. falcata targeted 213 potential MIRI-associated factors, including tumor necrosis factor (TNF), prostaglandin-endoperoxide synthase 2 (PTGS2), and the NF-κB signaling pathway. The in vitro assay on H9c2 cardiomyocytes subjected to hypoxia/reoxygenation injury confirmed that the flavonoids in OFF1 reduced myocardial marker levels, apoptotic rate, and the inflammatory response triggered by oxidative stress. Moreover, OFF1 attenuated MIRI by downregulating the ROS-mediated JNK/p38MAPK/NF-κB pathway. Collectively, these findings provide novel insights into the molecular mechanism of O. falcata in alleviating MIRI, being a potential therapeutic candidate.
Collapse
Affiliation(s)
- Yang Guo
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China; (Y.G.); (Z.-Q.L.)
| | - Ben-Yin Zhang
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China; (B.-Y.Z.); (Y.-F.P.)
| | - Yan-Feng Peng
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China; (B.-Y.Z.); (Y.-F.P.)
| | - Leng Chee Chang
- Department of Pharmaceutical Sciences, Daniel K. Inouye College of Pharmacy, University of Hawai’i, Hilo, HI 96720, USA;
| | - Zhan-Qiang Li
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China; (Y.G.); (Z.-Q.L.)
| | - Xin-Xin Zhang
- School of Pharmacy, Xi’an Jiaotong Univeristy, Xining 710061, China;
| | - De-Jun Zhang
- Research Center for High Altitude Medicine, Key Laboratory of High-Altitude Medicine (Ministry of Education), Key Laboratory of Application and Foundation for High Altitude Medicine Research in Qinghai Province (Qinghai-Utah Joint Research Key Lab for High Altitude Medicine), Qinghai University, Xining 810001, China; (Y.G.); (Z.-Q.L.)
- College of Eco-Environmental Engineering, Qinghai University, Xining 810016, China; (B.-Y.Z.); (Y.-F.P.)
- Correspondence: ; Tel.: +86-0971-5310586
| |
Collapse
|
7
|
Cardiac Immunology: A New Era for Immune Cells in the Heart. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021. [PMID: 32910424 DOI: 10.1007/5584_2020_576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2023]
Abstract
The immune system is essential for the development and homeostasis of the human body. Our current understanding of the immune system on disease pathogenesis has drastically expanded over the last decade with the definition of additional non-canonical roles in various tissues. Recently, tissue-resident immune cells have become an important research topic for understanding their roles in the prevention, pathogenesis, and recovery from the diseases. Heart resident immune cells, particularly macrophage subtypes, and their characteristic morphology, distribution in the cardiac tissue, and transcriptional profile have been recently reported in the experimental animal models, unrevealing novel and unexpected roles in electrophysiological regulation of the heart both at the steady-state and diseased state. Immunological processes have been widely studied in both sterile cardiac disorders, such as myocardial infarction, autoimmune cardiac diseases, or infectious cardiac diseases, such as myocarditis, endocarditis, and acute rheumatic carditis. Following cardiac injury, innate and adaptive immunity have critical roles in pro- and anti-inflammatory processes. Heart resident immune cells not only provide defense against infectious diseases but also contribute to the homeostasis. In recent years, physiological changes and pathological processes were demonstrated to alter the abundance, distribution, polarization, and diversity of immune cells in the heart. Accumulating evidence indicates that cardiac remodeling is controlled by the complex crosstalk between cardiomyocytes and cardiac immune cells through the gap junctions, providing the ion flow to achieve synchronization and modulation of contractility. This review article aims to review the well-documented roles of both resident and recruited immune cell in the heart, as well as their recently uncovered unconventional roles in both cardiac homeostasis and cardiovascular diseases. We have mostly focused on studies on animal models used in preclinical research, underlying the need for further investigations in humans or in vitro human models. It may be foreseen that the further comprehensive investigations of cardiac immunology might harbor new therapeutic options for cardiac disorders that have tremendous medical potential.
Collapse
|
8
|
Deng Y, Yang Q, Yang Y, Li Y, Peng H, Wu S, Zhang S, Yao B, Li S, Gao Y, Li X, Li L, Deng Y. Conditional knockout of Tsc1 in RORγt-expressing cells induces brain damage and early death in mice. J Neuroinflammation 2021; 18:107. [PMID: 33957945 PMCID: PMC8101034 DOI: 10.1186/s12974-021-02153-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/15/2021] [Indexed: 11/23/2022] Open
Abstract
Background Tuberous sclerosis complex 1 (Tsc1) is known to regulate the development and function of various cell types, and RORγt is a critical transcription factor in the immune system. However, whether Tsc1 participates in regulating RORγt-expressing cells remains unknown. Methods We generated a mouse model in which Tsc1 was conditionally deleted from RORγt-expressing cells (Tsc1RORγt) to study the role of RORγt-expressing cells with Tsc1 deficiency in brain homeostasis. Results Type 3 innate lymphoid cells (ILC3s) in Tsc1RORγt mice displayed normal development and function, and the mice showed normal Th17 cell differentiation. However, Tsc1RORγt mice exhibited spontaneous tonic-clonic seizures and died between 4 and 6 weeks after birth. At the age of 4 weeks, mice in which Tsc1 was specifically knocked out in RORγt-expressing cells had cortical neuron defects and hippocampal structural abnormalities. Notably, over-activation of neurons and astrogliosis were observed in the cortex and hippocampus of Tsc1RORγt mice. Moreover, expression of the γ-amino butyric acid (GABA) receptor in the brains of Tsc1RORγt mice was decreased, and GABA supplementation prolonged the lifespan of the mice to some extent. Further experiments revealed the presence of a group of rare RORγt-expressing cells with high metabolic activity in the mouse brain. Conclusions Our study verifies the critical role of previously unnoticed RORγt-expressing cells in the brain and demonstrates that the Tsc1 signaling pathway in RORγt-expressing cells is important for maintaining brain homeostasis. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02153-8.
Collapse
Affiliation(s)
- Yafei Deng
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Qinglan Yang
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yana Li
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Hongyan Peng
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Shuting Wu
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Shuju Zhang
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China
| | - Baige Yao
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, 410000, China
| | - Shuhui Li
- Department of Clinical Biochemistry, Faculty of Pharmacy and Laboratory Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yuan Gao
- Southwest Hospital/Southwest Eye Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Liping Li
- Hunan Children's Research Institute (HCRI), Hunan Children's Hospital, Changsha, 410000, China.
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
9
|
Susceptibility to Heart Defects in Down Syndrome Is Associated with Single Nucleotide Polymorphisms in HAS 21 Interferon Receptor Cluster and VEGFA Genes. Genes (Basel) 2020; 11:genes11121428. [PMID: 33260695 PMCID: PMC7761327 DOI: 10.3390/genes11121428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/26/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Congenital heart defects (CHDs) are present in about 40-60% of newborns with Down syndrome (DS). Patients with DS can also develop acquired cardiac disorders. Mouse models suggest that a critical 3.7 Mb region located on human chromosome 21 (HSA21) could explain the association with CHDs. This region includes a cluster of genes (IFNAR1, IFNAR2, IFNGR2, IL10RB) encoding for interferon receptors (IFN-Rs). Other genes located on different chromosomes, such as the vascular endothelial growth factor A (VEGFA), have been shown to be involved in cardiac defects. So, we investigated the association between single nucleotide polymorphisms (SNPs) in IFNAR2, IFNGR2, IL10RB and VEGFA genes, and the presence of CHDs or acquired cardiac defects in patients with DS. METHODS Individuals (n = 102) with DS, and age- and gender-matched controls (n = 96), were genotyped for four SNPs (rs2229207, rs2834213, rs2834167 and rs3025039) using KASPar assays. RESULTS We found that the IFNGR2 rs2834213 G homozygous genotype and IL10RB rs2834167G-positive genotypes were more common in patients with DSand significantly associated with heart disorders, while VEGFA rs3025039T-positive genotypes (T/*) were less prevalent in patients with CHDs. CONCLUSIONS We identified some candidate risk SNPs for CHDs and acquired heart defects in DS. Our data suggest that a complex architecture of risk alleles with interplay effects may contribute to the high variability of DS phenotypes.
Collapse
|
10
|
Fan S, Zhang D, Liu F, Yang Y, Xu H. Artesunate alleviates myocardial ischemia/reperfusion-induced myocardial necrosis in rats and hypoxia/reoxygenation-induced apoptosis in H9C2 cells via regulating the FAK/PI3K/Akt pathway. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:1291. [PMID: 33209871 PMCID: PMC7661874 DOI: 10.21037/atm-20-5182] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background The various anti-inflammatory, anti-apoptotic, and antioxidant effects of Artesunate (Art) have been explored in numerous studies. This study aimed to evaluate the function of Art on myocardial necrosis in apoptotic cardiomyocytes in vivo and in vitro. Methods Sprague Dawley (SD) rats were randomly divided into groups: a control group, a myocardial ischemia reperfusion (MI/R) group, and MI/R+ Art groups. To establish a MI/R model, rats were subjected to left anterior descending artery ischemia for 45 minutes, and then reperfusion for 2 hours. Hypoxia was induced in H9C2 cells by subjecting them to hypoxic conditions at 37 °C for 4 hours, before placing them in a normoxic chamber for 2 hours. The test methods were used in this test, such as echocardiography, enzyme-linked immunosorbent assay (ELISA), HE staining, TUNEL staining, immunohistochemistry, flow cytometry, western blot, and CCK-8 assay. Results Art improved myocardial systolic function caused by MI/R injury in vivo. Simultaneously, Art reduced the levels of cardiac troponin I (cTnl), creatine kinase-MB (CK-MB) and myohemoglobin (Mb) in vivo and in vitro. Moreover, Art inhibited cardiomyocyte apoptosis in vivo and in vitro. The focal adhesion kinase (FAK)/phosphatidylinositide-3 kinases (PI3K)/AKT signaling pathway was also activated by Art in vivo and in vitro. Furthermore, after inhibitor PF573228 was added, Art inhibited apoptosis in H9C2 cells via activation of the FAK/PI3K/AKT signaling pathway in vitro. Conclusions This study confirms that Art alleviated MI/R injury and inhibited cardiomyocyte apoptosis in vivo and in vitro. Art exerted an inhibitory effect on cardiomyocyte apoptosis by activating the FAK/PI3K/AKT signaling pathway. Therefore, Art may serve as an alternative treatment for MI/R injury.
Collapse
Affiliation(s)
- Shunyang Fan
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Deyin Zhang
- Department of Galactphore, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fuyun Liu
- Department of Pediatric Orthopaedic, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuqi Yang
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Hongliang Xu
- Department of Heart Center, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Chen HI, Hu WS, Hung MY, Ou HC, Huang SH, Hsu PT, Day CH, Lin KH, Viswanadha VP, Kuo WW, Huang CY. Protective effects of luteolin against oxidative stress and mitochondrial dysfunction in endothelial cells. Nutr Metab Cardiovasc Dis 2020; 30:1032-1043. [PMID: 32402583 DOI: 10.1016/j.numecd.2020.02.014] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 02/13/2020] [Accepted: 02/18/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND AND AIMS Luteolin is a common flavonoid that is abundantly present in various edible plants, it is known to exhibit beneficial effects on cardiovascular system. However, the mechanisms which underlie the protective effects of luteolin on endothelial cell damage caused by oxidative stress remains unclear. The purpose of this study is to test the hypothesis which states that luteolin protects against H2O2-induced oxidative stress via modulating ROS-mediated P38 MAPK/NF-κB and calcium-evoked mitochondrial apoptotic signalling pathways. METHODS AND RESULTS Human umbilical vein endothelial cells (HUVECs) were pretreated with luteolin prior to being stimulated by 600 μM H2O2 for another 24 h. The expression of native and phosphorylated-P38, IκB, NF-κB, native eNOS, phosphorylated-eNOS, iNOS and several apoptosis-related proteins were analyzed by Western blot. In addition, intracellular calcium was determined by fura-2 AM and mitochondrial membrane potential was examined by using JC1. Using the data gathered, we found indications that H2O2 induced P38 MAPK/NF-κB activation. H2O2 downregulated the expression of eNOS and upregulated iNOS, which in turn contribute to an elevated NO generation and protein nitrosylation. However, pretreatment with luteolin markedly reversed all of these alterations dose-dependently. Additionally, an intracellular calcium rise and subsequent mitochondrial membrane potential collapse, P53 phosphorylation, reduced BcL-2/Bax ratio in the mitochondrial membrane, release cytochrome c from mitochondria, leading to the subsequent activation of caspase 3 activation by H2O2 were all markedly suppressed in the presence of luteolin. CONCLUSION Results from this study may provide the possible molecular mechanisms underlying cardiovascular protective effects of luteolin.
Collapse
Affiliation(s)
- Hsiu-I Chen
- Department of Physical Therapy, Hungkuang University, Taichung, Taiwan
| | - Wei-Syun Hu
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan
| | - Meng-Yu Hung
- Graduate Institute of Biomedicine, China Medical University and Hospital, Taichung, Taiwan
| | - Hsiu-Chung Ou
- Department of Physical Therapy, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Su-Hua Huang
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Pei-Tzu Hsu
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | | | - Kuan-Ho Lin
- School of Medicine, College of Medicine, China Medical University, Taichung, 40402, Taiwan; Division of Emergency, Department of Medicine, China Medical University Hospital, Taichung, 40447, Taiwan
| | - Vijaya P Viswanadha
- Department of Biotechnology, Bharathiar University, Coimbatore, 641 046, India
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedicine, China Medical University and Hospital, Taichung, Taiwan; Department of Biotechnology, Asia University, Taichung, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan; Cardiovascular and Mitochondrial Related Diseases Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, 970, Taiwan.
| |
Collapse
|
12
|
Li W, Yang J, Lyu Q, Wu G, Lin S, Yang Q, Hu J. Taurine attenuates isoproterenol-induced H9c2 cardiomyocytes hypertrophy by improving antioxidative ability and inhibiting calpain-1-mediated apoptosis. Mol Cell Biochem 2020; 469:119-132. [PMID: 32304004 DOI: 10.1007/s11010-020-03733-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/08/2020] [Indexed: 12/18/2022]
Abstract
Pathological cardiac hypertrophy is ultimately accompanied by cardiomyocyte apoptosis. Apoptosis mainly related to calpain-1-mediated apoptotic pathways. Studies had proved that taurine can maintain heart health through antioxidation and antiapoptotic functions, but the effect of taurine on cardiac hypertrophy is still unclear. This study aimed to determine whether taurine could inhibit calpain-1-mediated mitochondria-dependent apoptotic pathways in isoproterenol (ISO)-induced hypertrophic cardiomyocytes. We found that taurine could inhibit the increase in cell surface area and reduce the protein expression levels of the hypertrophic markers atrial natriuretic peptide, brain natriuretic polypeptide, and β-myosin heavy chain. Taurine also reduced ROS, intracellular Ca2+ overload and mitochondrial membrane potential. Moreover, taurine inhibited cardiomyocyte apoptosis by decreasing the protein expression of calpain-1, Bax, t-Bid, cytosolic cytochrome c, Apaf-1, cleaved caspase-9 and cleaved caspase-3 and by enhancing calpastatin and Bcl-2 protein expression. Calpain-1 small interfering RNA transfection results showed similar antiapoptotic effects as the taurine prevention group. However, compared with the two treatments, taurine inhibited the expression of cleaved caspase-9 more significantly. Therefore, we believe that taurine prevents ISO-induced H9c2 cardiomyocyte hypertrophy by inhibiting oxidative stress, intracellular Ca2+ overload, the calpain-1-mediated mitochondria-dependent apoptotic pathway and cleaved caspase-9 levels.
Collapse
Affiliation(s)
- Weiwei Li
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Jiancheng Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qiufeng Lyu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Gaofeng Wu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Shumei Lin
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China
| | - Qunhui Yang
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| | - Jianmin Hu
- Liaoning Provincial Key Laboratory of Zoonosis, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, Liaoning, People's Republic of China.
| |
Collapse
|
13
|
Zhou Q, Song J, Wang Y, Lin T. Remifentanil attenuates cardiac dysfunction, lipid peroxidation and immune disorder in rats with isoproterenol-induced myocardial injury via JNK/NF-KB p65 inhibition. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:551. [PMID: 32411774 PMCID: PMC7214888 DOI: 10.21037/atm-20-3134] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Myocardial injury caused by myocardial ischemia (MI) is still a severe condition that can result in apoptosis, oxidative stress, and inflammation. Remifentanil is a selective, ultra-short-acting, µ-opioid receptor agonist opioid. It can improve sinusoidal heart rate patterns in the fetus, for bupivacaine-induced cardiotoxicity, and with lipopolysaccharide (LPS)-induced cardiomyocytes injuries. This study aimed to explore the cardioprotective effects of remifentanil in MI model rats. Methods Sprague Dawley (SD) rats were split into five groups at random, including a control group, Isop group, low-dose remifentanil treatment group (10 µg/kg), medium-dose remifentanil treatment group (20 µg/kg), and a high-dose remifentanil treatment group (40 µg/kg). The MI model was achieved by subcutaneously injecting rats with isoproterenol (85 mg/kg) for two consecutive days. With the expression of apoptotic molecules, myocardial systolic function index, inflammation, antioxidant enzymes, and the myocardial enzyme taken into account, the data was analyzed. Results After treatment with remifentanil, the left ventricular wall thickness (LVWT), left ventricular end-systolic volume (LVESV), left ventricular ejection fraction (LVEF), fraction shortening (FS), and heart rate (HR) were significantly increased in comparison with the Isop group. Creatine kinase-MB (CK-MB), Mb, and cTnl expressions were decreased. Meanwhile, the levels of cleaved caspase-3 and caspase-9 were decreased. Remarkably, the levels of reactive oxidative species (ROS), malondialdehyde (MDA), and lactate dehydrogenase (LDH) were observed to be repressed, while the levels of superoxide dismutase (SOD) was significantly increased. More importantly, the levels of tumor necrosis factor-α (TNF-α), interleukin (IL)-1β, IL-6, and interferon (IFN)-γ were decreased. Conclusions Remifentanil has significant potential as a therapeutic intervention strategy for ameliorating myocardial injury after MI and these findings provide the rationale for further clinical studies.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Junmei Song
- Department of Cardiac Function, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Yu Wang
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| | - Tao Lin
- Department of Anesthesiology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610072, China
| |
Collapse
|
14
|
Grinnemo KH, Löfling M, Nathanson L, Baumgartner R, Ketelhuth DFJ, Beljanski V, Davies LC, Österholm C. Immunomodulatory effects of interferon-γ on human fetal cardiac mesenchymal stromal cells. Stem Cell Res Ther 2019; 10:371. [PMID: 31801632 PMCID: PMC6894330 DOI: 10.1186/s13287-019-1489-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 10/09/2019] [Accepted: 11/05/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Mesenchymal stromal cells (MSCs), due to their regenerative and immunomodulatory properties, are therapeutically used for diseases, including heart failure. As early gestational-phase embryonic tissues exhibit extraordinary regenerative potential, fetal MSCs exposed to inflammation offer a unique opportunity to evaluate molecular mechanisms underlying preferential healing, and investigate their inherent abilities to communicate with the immune system during development. The principal aim of this study was to evaluate the effects of interferon-γ (IFNγ) on the immunomodulatory effects of first-trimester human fetal cardiac (hfc)-MSCs. METHODS hfcMSCs (gestational week 8) were exposed to IFNγ, with subsequent analysis of the whole transcriptome, based on RNA sequencing. Exploration of surface-expressed immunoregulatory mediators and modulation of T cell responses were performed by flow cytometry. Presence and activity of soluble mediators were assessed by ELISA or high-performance liquid chromatography. RESULTS Stimulation of hfcMSCs with IFNγ revealed significant transcriptional changes, particularly in respect to the expression of genes belonging to antigen presentation pathways, cell cycle control, and interferon signaling. Expression of immunomodulatory genes and associated functional changes, including indoleamine 2,3-dioxygenase activity, and regulation of T cell activation and proliferation via programmed cell death protein (PD)-1 and its ligands PD-L1 and PD-L2, were significantly upregulated. These immunoregulatory molecules diminished rapidly upon withdrawal of inflammatory stimulus, indicating a high degree of plasticity by hfcMSCs. CONCLUSIONS To our knowledge, this is the first study performing a systematic evaluation of inflammatory responses and immunoregulatory properties of first-trimester cardiac tissue. In summary, our study demonstrates the dynamic responsiveness of hfcMSCs to inflammatory stimuli. Further understanding as to the immunoregulatory properties of hfcMSCs may be of benefit in the development of novel stromal cell therapeutics for cardiovascular disease.
Collapse
Affiliation(s)
- Karl-Henrik Grinnemo
- Department of Molecular Medicine and Surgery, Karolinska Institutet, BioClinicum J10:20, SE-171 64, Solna, Sweden
- Division of Cardiothoracic Surgery and Anesthesiology, Department of Surgical Sciences, Uppsala University, Akademiska University Hospital, 751 85, Uppsala, Sweden
| | - Marie Löfling
- Department of Molecular Medicine and Surgery, Karolinska Institutet, BioClinicum J10:20, SE-171 64, Solna, Sweden
| | - Lubov Nathanson
- Institute for Neuroimmune Medicine, Dr Kiran C. Patel College of Osteopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Roland Baumgartner
- Department of Medicine Solna, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institutet, SE-171 64, Solna, Sweden
| | - Daniel F J Ketelhuth
- Department of Medicine Solna, Cardiovascular Medicine Unit, Center for Molecular Medicine, Karolinska Institutet, SE-171 64, Solna, Sweden
| | - Vladimir Beljanski
- Cell Therapy Institute, Dr Kiran C. Patel College of Allopathic Medicine, Nova Southeastern University, Davie, FL, USA
| | - Lindsay C Davies
- Department of Laboratory Medicine, Karolinska Institutet, SE-141 52, Huddinge, Sweden
| | - Cecilia Österholm
- Department of Molecular Medicine and Surgery, Karolinska Institutet, BioClinicum J10:20, SE-171 64, Solna, Sweden.
| |
Collapse
|
15
|
Spermidine Prevents Heart Injury in Neonatal Rats Exposed to Intrauterine Hypoxia by Inhibiting Oxidative Stress and Mitochondrial Fragmentation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:5406468. [PMID: 31217839 PMCID: PMC6537013 DOI: 10.1155/2019/5406468] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/14/2019] [Accepted: 04/23/2019] [Indexed: 12/11/2022]
Abstract
Intrauterine hypoxia (IUH) is a common intrauterine dysplasia that can cause programming of the offspring cardiovascular system. In this study, we hypothesized that placental treatment with spermidine (SPD) can prevent heart injury in neonatal offspring exposed to IUH. Pregnant rats were exposed to 21% O2 or 10% O2 (hypoxia) for 7 days prior to term or were exposed to hypoxia and intraperitoneally administered SPD or SPD+difluromethylornithine (DFMO) on gestational days 15-21. Seven-day-old offspring were then sacrificed to assess several parameters. Our results demonstrated that IUH led to decreased myocardial ornithine decarboxylase (ODC) and increased spermidine/spermine N1-acetyltransferase (SSAT) expression in the offspring. IUH also resulted in decreased offspring body weight, heart weight, cardiomyocyte proliferation, and antioxidant capacity and increased cardiomyocyte apoptosis and fibrosis. Furthermore, IUH caused mitochondrial structure abnormality, dysfunction, and decreased biogenesis and led to a fission/fusion imbalance in offspring hearts. In vitro, hypoxia induced mitochondrial ROS accumulation, decreased membrane potential, and increased fragmentation. Notably, all hypoxia-induced changes analyzed in this study were prevented by SPD. Thus, in utero SPD treatment is a potential strategy for preventing IUH-induced neonatal cardiac injury.
Collapse
|
16
|
Dysregulation of miR-135a-5p promotes the development of rat pulmonary arterial hypertension in vivo and in vitro. Acta Pharmacol Sin 2019; 40:477-485. [PMID: 30038339 DOI: 10.1038/s41401-018-0076-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 05/20/2018] [Indexed: 11/08/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is the most common form of pulmonary hypertension. Pulmonary arterial remodeling is closely related to the abnormal proliferation of pulmonary artery smooth muscle cells (PASMCs), which leads to the thickening of the medial layer of muscular arteries and then results in the narrowing or occlusion of the precapillary arterioles and PAH. However, the mechanisms underlying the abnormal proliferation of PASMCs remain unclear. In this study, we established rat primary PAH models using monocrotaline (MCT) injection or hypoxic exposure, then investigated the expression patterns of seven miRNAs associated with multiple pathogenic pathways central to pulmonary hypertension, and further explored the roles and the possible mechanisms of miR-135a during the development of PAH. In the rat primary PAH models, we observed that the expression of miR-135a-5p in lungs was drastically decreased at the initial stage of PAH development after MCT administration or hypoxic exposure, but it increased by 12-fold or 10-fold at the later stage. In vitro study in PASMCs showed a similar pattern of miR-135a-5p expression, with downregulation at 6 h but upregulation at 18, 24, and 48 h after hypoxic exposure. Early, but not late, administration of a miR-135a-5p mimic inhibited hypoxia-induced proliferation of PASMCs. The protective role of early miR-135a-5p agomir in the PAH rat model further supported the hypothesis that the early decrease in the expression of miR-135a-5p contributes to the proliferation of PASMCs and development of PAH, as early administration of miR-135a-5p agomir (10 nM, i.v.) reversed the elevated mean pulmonary arterial pressure and pulmonary vascular remodeling in MCT-treated rats. We revealed that miR-135a-5p directly bound to the 3'-UTR sequence of rat transient receptor potential channel 1 (TRPC1) mRNA and decreased TRPC1 protein expression, thus inhibiting PASMC proliferation. Collectively, our data suggest that dysregulation of miR-135a-5p in PASMCs contributes to the abnormal proliferation of PASMCs and the pathogenesis of PAH. Increasing miR-135a-5p expression at the early stage of PAH is a potential new avenue to prevent PAH development.
Collapse
|
17
|
Hu S, Cheng M, Guo X, Wang S, Liu B, Jiang H, Huang C, Wu G. Down-regulation of miR-200c attenuates AngII-induced cardiac hypertrophy via targeting the MLCK-mediated pathway. J Cell Mol Med 2019; 23:2505-2516. [PMID: 30680929 PMCID: PMC6433679 DOI: 10.1111/jcmm.14135] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 11/21/2018] [Accepted: 12/09/2018] [Indexed: 01/07/2023] Open
Abstract
Background MicroRNAs (miRNAs) have been shown to commonly contribute to cardiac hypertrophy (CH). The aim of this study was to test the hypothesis that miR‐200c plays an important role in the progression of CH by targeting myosin light chain kinase (MLCK/MYLK). Methods and results Cardiac hypertrophy was induced by aortic banding (AB) in rats. Cellular hypertrophy in neonatal rat cardiomyocytes (NCMs) was induced by AngII treatment. Echocardiography, histology and molecular measurements were used to assess the results of the experiments. The levels of apoptosis and reactive oxygen species (ROS) were also measured. Quantitative real‐time PCR (qRT‐PCR) and Western blotting were used to measure mRNA and protein levels respectively. The present results showed that miR‐200c expression was increased in response to CH both in vivo and in vitro. The down‐regulation of miRNA‐200c by a specific inhibitor markedly ameliorated CH resulting from AngII treatment, and the mRNA levels of atrial natriuretic peptide, brain natriuretic peptide and β‐myosin heavy chain were simultaneously decreased. Notably, minimal apoptosis and ROS accumulation were identified in AngII‐induced hypertrophic cardiomyocytes. Conversely, the up‐regulation of miR‐200c using specific mimics reversed these effects. Mechanistic investigations demonstrated that the MLCK gene is a direct target of miR‐200c; an increase in miR‐200c levels led to a decrease in the expression of MLCK and its downstream effector, p‐MLC2, while miR‐200c inhibition increased the expression of these proteins. Furthermore, inhibiting MLCK impaired the anti‐hypertrophic effects contributions produced by the knockdown of miR‐200c. Conclusion Our studies suggest that miR‐200c may serve as a potential therapeutic target that could delay hypertrophy. We have also uncovered a relationship between miR‐200c and MLCK, identifying MLCK as a direct mediator of miR‐200c.
Collapse
Affiliation(s)
- Shan Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Mian Cheng
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Guo
- Department of Cardiology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Beilei Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Congxin Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Cardiovascular Research Institute, Wuhan University, Hubei Key Laboratory of Cardiology, Wuhan, China
| |
Collapse
|
18
|
Qi D, Wang Q, Wu M, Zhang X. Comprehensive bioinformatics analysis of lncRNAs in gastric cancer. Oncol Lett 2019; 17:1279-1291. [PMID: 30655896 PMCID: PMC6313033 DOI: 10.3892/ol.2018.9707] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 07/03/2018] [Indexed: 12/17/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have been generally considered to serve important roles in various types of cancer, including gastric cancer. However, a comprehensive understanding of lncRNAs in gastric cancer requires further study. The present study performed an in-depth study revealed 50 differently expressed lncRNAs. The changed cellular pathways and biological process in gastric cancer were determined. To further confirm the functions of the differently expressed lncRNAs, co-expression networks were constructed between the lncRNAs and mRNA; this lead to the identification of 6 modules, which participated in various cellular pathways. In addition, 2 lncRNAs were identified which were associated with clinical outcome. The biological analysis and experimental evidence suggested that LINC00982 inhibited, while LL22NC03-N14H11.1 promoted the proliferation of gastric cancer cells. These lncRNAs may be considered as potential prognostic factor in gastric cancer.
Collapse
Affiliation(s)
- Dongdong Qi
- Department of Clinical Laboratory, Hulunbuir Mental Health Center, Hulunbuir, Inner Mongolia 022150, P.R. China
| | - Qiang Wang
- Department of General Surgery, Inner Mongolia Forestry General Hospital, Hulunbuir, Inner Mongolia 022150, P.R. China
| | - Meiqing Wu
- Dermatological Department, Inner Mongolia Forestry General Hospital, Hulunbuir, Inner Mongolia 022150, P.R. China
| | - Xiong Zhang
- Hulunbuir Mental Health Center, Hulunbuir, Inner Mongolia 022150, P.R. China
| |
Collapse
|
19
|
Gao Y, Bai D, Zhao Y, Zhu Q, Zhou Y, Li Z, Lu N. LL202 ameliorates colitis against oxidative stress of macrophage by activation of the Nrf2/HO‐1 pathway. J Cell Physiol 2018; 234:10625-10639. [DOI: 10.1002/jcp.27739] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 10/18/2018] [Indexed: 12/14/2022]
Affiliation(s)
- Yuan Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| | - Dongsheng Bai
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| | - Yue Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| | - Qin Zhu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| | - Yihui Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| | - Zhiyu Li
- Department of Medicinal Chemistry China Pharmaceutical University Nanjing China
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention Department of Basic Medicine School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University Nanjing China
| |
Collapse
|
20
|
Kim DH, Shin EA, Kim B, Shim BS, Kim SH. Reactive oxygen species-mediated phosphorylation of p38 signaling is critically involved in apoptotic effect of Tanshinone I in colon cancer cells. Phytother Res 2018; 32:1975-1982. [DOI: 10.1002/ptr.6126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/11/2018] [Accepted: 05/15/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Dong Hee Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Eun Ah Shin
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Bonglee Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Bum Sang Shim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| | - Sung-Hoon Kim
- College of Korean Medicine; Kyung Hee University; Seoul South Korea
| |
Collapse
|
21
|
Deng Y, Song L, Nie X, Shou W, Li X. Prenatal inflammation exposure-programmed cardiovascular diseases and potential prevention. Pharmacol Ther 2018; 190:159-172. [PMID: 29803628 DOI: 10.1016/j.pharmthera.2018.05.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In recent years, the rapid development of medical and pharmacological interventions has led to a steady decline in certain noncommunicable chronic diseases (NCDs), such as cancer. However, the overall incidence of cardiovascular diseases (CVDs) has not seemed to decline. CVDs have become even more prevalent in many countries and represent a global health threat and financial burden. An increasing number of epidemiological and experimental studies have demonstrated that maternal insults not only can result in birth defects but also can cause developmental functional defects that contribute to adult NCDs. In the current review, we provide an overview of evidence from both epidemiological investigations and experimental animal studies supporting the concept of developmental reprogramming of adult CVDs in offspring that have experienced prenatal inflammation exposure (PIE) during fetal development (PIE-programmed CVDs), a disease-causing event that has not been effectively controlled. This review describes the epidemiological observations, data from animal models, and related mechanisms for the pathogenesis of PIE-programmed CVDs. In addition, the potential therapeutic interventions of PIE-programmed CVDs are discussed. Finally, we also deliberate the need for future mechanistic studies and biomarker screenings in this important field, which creates a great opportunity to combat the global increase in CVDs by managing the adverse effects of inflammation for prepregnant and pregnant individuals who are at risk for PIE-programmed CVDs.
Collapse
Affiliation(s)
- Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Center of Translational Medicine, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China.
| | - Liang Song
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Center of Translational Medicine, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China
| | - Xuqiang Nie
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Center of Translational Medicine, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China
| | - Weinian Shou
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Center of Translational Medicine, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4 W302D, Indianapolis, IN 46202, USA
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China; Center of Translational Medicine, College of Pharmacy, Army Medical University (Third Military Medical University), 30# Gaotanyan Rd., Shapingba District, Chongqing 400038, China.
| |
Collapse
|
22
|
Yu S, Wen Y, Li J, Zhang H, Liu Y. Prenatal Lipopolysaccharide Exposure Promotes Dyslipidemia in the Male Offspring Rats. Front Physiol 2018; 9:542. [PMID: 29867579 PMCID: PMC5964359 DOI: 10.3389/fphys.2018.00542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 04/26/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammation is critical to the pathogenesis of cardiovascular diseases (CVDs). We have uncovered intrauterine inflammation induced by lipopolysaccharide (LPS) increases CVDs in adult offspring rats. The present study aimed to explore the role of prenatal exposure to LPS on the lipid profiles in male offspring rats and to further assess their susceptibility to high fat diet (HFD). Maternal LPS (0.79 mg/kg) exposure produced a significant increase in serum and hepatic levels of total cholesterol, triglycerides, low-density lipoprotein cholesterol, aspartate amino transferase as well as liver morphological abnormalities in 8-week-old offspring rats. Meanwhile, disturbed gene expressions involved in hepatic lipid metabolism and related signaling pathways were found, especially the up-regulated very-low density lipoprotein receptor (VLDLR) and down-regulated transmembrane 7 superfamily member 2 (TM7SF2). Following HFD treatment, however, the lipid profile shifts and liver dysfunction were exacerbated compared to the offsprings treated with prenatal LPS exposure alone. Compared with that in control offsprings, the hepatic mitochondria (Mt) in offspring rats solely treated with HFD exhibited remarkably higher ATP level, enforced Complex IV expression and a sharp reduction of its activity, whereas the offsprings from LPS-treated dams showed the loss of ATP content, diminished membrane potential, decline in protein expression and activity of mitochondrial respiratory complex IV, increased level of MtDNA deletion as well. Furthermore, treatment with HFD deteriorated these mitochondrial disorders in the prenatally LPS-exposed offspring rats. Taken together, maternal LPS exposure reinforces dyslipidemia in response to a HFD in adult offsprings, which should be associated with mitochondrial abnormalities and disturbed gene expressions of cholesterol metabolism.
Collapse
Affiliation(s)
- Shiyun Yu
- Department of Pharmaceutics, College of Pharmacy, Institute of Materia Medica, Third Military Medical University, Chongqing, China
| | - Yan Wen
- Department of Pharmaceutics, College of Pharmacy, Institute of Materia Medica, Third Military Medical University, Chongqing, China.,Department of General Surgery, Southwest Hospital of Third Military Medical University, Chongqing, China
| | - Jingmei Li
- Department of Pharmaceutics, College of Pharmacy, Institute of Materia Medica, Third Military Medical University, Chongqing, China
| | - Haigang Zhang
- Department of Pharmacology, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ya Liu
- Department of Pharmaceutics, College of Pharmacy, Institute of Materia Medica, Third Military Medical University, Chongqing, China
| |
Collapse
|
23
|
Priya LB, Baskaran R, Huang CY, Padma VV. Neferine ameliorates cardiomyoblast apoptosis induced by doxorubicin: possible role in modulating NADPH oxidase/ROS-mediated NFκB redox signaling cascade. Sci Rep 2017; 7:12283. [PMID: 28947826 PMCID: PMC5612945 DOI: 10.1038/s41598-017-12060-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/29/2017] [Indexed: 12/04/2022] Open
Abstract
Doxorubicin (DOX) mediated cardiomyopathy is a major challenge in cancer chemotherapy. Redox-cycling of doxorubicin by flavoenzymes makes the heart more vulnerable to oxidative stress leading to cardiac dysfunction. The present study evaluates the role of neferine, a bisbenzylisoquinoline alkaloid, in curbing the molecular consequences of DOX-exposure in H9c2 cardiomyoblasts. Neferine pre-treatment increased cell viability upon DOX-exposure. DOX activates NADPH oxidase subunits, (p22phox, p47phox, gp91phox) as the primary event followed by peak in [Ca2+]i accumulation by 2 h, ROS by 3 h and activated ERK1/2 and p38 MAPKinases, time dependently along with the activation and translocation of NFκB and up-regulated COX2 and TNF-α expressions. Neferine pre-treatment modulated NADPH oxidase/ROS system, inhibited MAPKinases and NFκB activation, reduced sub G1 cell population and concomitantly increased cyclin D1 expression reducing DOX-mediated apoptosis. The study demonstrates for the first time, the molecular sequential events behind DOX toxicity and the mechanism of protection offered by neferine with specific relevance to NADPH oxidase system, MAPKinases, inflammation and apoptosis in H9c2 cells. Our data suggests the use of neferine as a new approach in pharmacological interventions against cardiovascular disorders as secondary complications.
Collapse
Affiliation(s)
- Lohanathan Bharathi Priya
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India
| | - Rathinasamy Baskaran
- National Institute of Cancer Research, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Graduate Institute of Chinese Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| | - Viswanadha Vijaya Padma
- Translational Research Laboratory, Department of Biotechnology, School of Biotechnology and Genetic Engineering, Bharathiar University, Coimbatore, 641046, Tamil Nadu, India.
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan.
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan.
| |
Collapse
|
24
|
Liu N, Cui C, Sun Y, Zhang F, Wang S, Su G, Cai X. Hydrogen peroxide promotes the expression of angiopoietin like 4 in RAW264.7 macrophages via MAPK pathways. Mol Med Rep 2017; 16:6128-6133. [PMID: 28849063 DOI: 10.3892/mmr.2017.7365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 03/23/2017] [Indexed: 11/06/2022] Open
Abstract
Previous studies including some vivo experiments and large scale clinical trials have indicated that angiopoietin like 4 (ANGPTL4) is involved in atherosclerosis. However, the specific mechanism underlying the process remains unresolved. Similarly, cumulative evidence indicated that hydrogen peroxide (H2O2) is closely related to the occurrence and development of atherosclerosis. The current study investigated whether H2O2 treatment can affect ANGPTL4 release in macrophage cells cell viability assay, western blot analysis, ELISA and immunofluorescence. It was determined that treatment with 0.25 and 0.5 mM H2O2 resulted in a significant increase in ANGPTL4 protein expression in macrophage cells. Mitogen‑activated protein kinase (MAPK) pathways were implicated in the secretion of ANGPTL4 regulated by H2O2, and specific inhibitors of MAPK1 (also known as ERK) and p38 MAPK significantly decreased H2O2 induced ANGPTL4 protein expression. Accordingly, it was demonstrated that ANGPTL4 expression was regulated by H2O2 via ERK and p38 MAPK, but not the MAPK8 (also known as JNK) pathway. In view of the effects of H2O2 and ANGPTL4 on atherosclerosis, the influence of H2O2 on ANGPTL4 provided new insight into the mechanism of atherosclerosis.
Collapse
Affiliation(s)
- Nan Liu
- Department of Cardiovascular Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Changxia Cui
- Department of Cardiovascular Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Yue Sun
- Department of Cardiology, Shandong University, Cheeloo College of Medicine, Jinan, Shandong 250013, P.R. China
| | - Feng Zhang
- Department of Cardiology, Tengzhou Central People's Hospital, Tengzhou, Shandong 277500, P.R. China
| | - Shuya Wang
- Department of Cardiovascular Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Guohai Su
- Department of Cardiovascular Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| | - Xiaojun Cai
- Department of Cardiovascular Medicine, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013, P.R. China
| |
Collapse
|
25
|
Zhang D, Cui Y, Li B, Luo X, Li B, Tang Y. miR-155 regulates high glucose-induced cardiac fibrosis via the TGF-β signaling pathway. MOLECULAR BIOSYSTEMS 2017; 13:215-224. [PMID: 27924974 DOI: 10.1039/c6mb00649c] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiac fibrosis, as a pathological process, plays an important role in various cardiac diseases. microRNA-155 (miR-155) is one of the most important miRNAs, and previous studies have shown that it is a regulatory factor in various fibrotic diseases. However, the mechanism by which miR-155 affects myocardial fibrosis remains unclear. In this study, we aim to establish the biological function of miR-155 in myocardial fibrosis induced by diabetes in mice. We used normal C57BL/6 wild type (WT) and miR-155 knockout (KO) mice to establish the diabetic model by intraperitoneal injection of streptozotocin, and we utilized echocardiography to evaluate the cardiac function at 30 and 60 days post-modeling. Hematoxylin-eosin (HE) and sirius-red (SR) staining were used to evaluate the degree of myocardial lesions. Furthermore, we extracted cardiac fibroblasts (CFs) from the WT mice and transfected them with miR-155 inhibitors, mimics and negative control siRNAs to analyze the specific mechanism involved in the development of myocardial fibrosis. The results showed that miR-155 deficiency could prevent cardiac fibrosis induced by diabetes in mice and also that attenuated collagen synthesis is induced by high glucose (HG) in CFs. We found that miR-155 regulated cardiac fibrosis via the TGF-β1-Smad 2 signaling pathway. These findings suggest that miR-155 may be a therapeutic target for preventing cardiac fibrosis induced by diabetes.
Collapse
Affiliation(s)
- Dong Zhang
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| | - Yongchun Cui
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| | - Bin Li
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| | - Xiaokang Luo
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| | - Bo Li
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| | - Yue Tang
- Chinese Academy of Medical Sciences, Peking Union Medical College, National Centre for Cardiovascular Disease, Fuwai Hospital, State Key Laboratory of Cardiovascular Disease, Beijing Key Laboratory of Pre-Clinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, Beijing 100037, China.
| |
Collapse
|
26
|
Choi JY, Hwang CJ, Lee HP, Kim HS, Han SB, Hong JT. Inhibitory effect of ethanol extract of Nannochloropsis oceanica on lipopolysaccharide-induced neuroinflammation, oxidative stress, amyloidogenesis and memory impairment. Oncotarget 2017; 8:45517-45530. [PMID: 28489589 PMCID: PMC5542205 DOI: 10.18632/oncotarget.17268] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Accepted: 03/26/2017] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress and neuroinflammation is implicated in the pathogenesis and development of Alzheimer's disease (AD). Here, we investigated the suppressive possibility of ethanol extract of Nannochloropsis oceanica (N. oceanica) on memory deficiency along with the fundamental mechanisms in lipopolysaccharide (LPS)-treated mice model. Among several extracts of 32 marine microalgae, ethanol extract of N. oceanica showed the most significant inhibitory effect on nitric oxide (NO) generation, NF-κB activity and β-secretase activity in cultured BV-2 cells, neuronal cells and Raw 264.7 cells. Ethanol extract of N. oceanica (50, 100 mg/kg) also ameliorated LPS (250 μg/kg)-induced memory impairment. We also found that ethanol extract of N. oceanica inhibited the LPS-induced expression of iNOS and COX-2. Furthermore, the production of reactive oxygen species (ROS), malondialdehyde (MDA) level as well as glutathione (GSH) level was also decreased by treatment of ethanol extract of N.oceanica. The ethanol extract of N. oceanica also suppresses IκB degradation as well as p50 and p65 translocation into the nucleus in LPS-treated mice brain. Associated with the inhibitory effect on neuroinflammation and oxidative stress, ethanol extract of N. oceanica suppressed Aβ1-42 generation through down-regulation of APP and BACE1 expression in in vivo. These results suggest that ethanol extract of N. oceanica ameliorated memory impairment via anti-inflammatory, anti-oxidant and anti-amyloidogenic mechanisms.
Collapse
Affiliation(s)
- Ji Yeon Choi
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Chul Ju Hwang
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hee Pom Lee
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Hee Sik Kim
- Sustainable Bioresource Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Yuseoung, Daejeon 305-806, Republic of Korea
| | - Sang-Bae Han
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Osong-eup, Heungdeok-gu, Cheongju, Chungbuk 28160, Republic of Korea
| |
Collapse
|
27
|
Sun F, Li X, Duan WQ, Tian W, Gao M, Yang J, Wu XY, Huang D, Xia W, Han YN, Wang JX, Liu YX, Dong CJ, Zhao D, Ban T, Chu WF. Transforming Growth Factor-β Receptor III is a Potential Regulator of Ischemia-Induced Cardiomyocyte Apoptosis. J Am Heart Assoc 2017; 6:JAHA.116.005357. [PMID: 28559372 PMCID: PMC5669164 DOI: 10.1161/jaha.116.005357] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background Myocardial infarction (MI) is often accompanied by cardiomyocyte apoptosis, which decreases heart function and leads to an increased risk of heart failure. The aim of this study was to examine the effects of transforming growth factor‐β receptor III (TGFβR3) on cardiomyocyte apoptosis during MI. Methods and Results An MI mouse model was established by left anterior descending coronary artery ligation. Cell viability, apoptosis, TGFβR3, and mitogen‐activated protein kinase signaling were assessed by methylthiazolyldiphenyl‐tetrazolium bromide assay, terminal deoxynucleotidyl transferase‐mediated dUTP nick end labeling assay, immunofluorescence, electron microscopy, and Western blotting. Our results demonstrated that TGFβR3 expression in the border region of the heart was dynamically changed during MI. After stimulation with H2O2, TGFβR3 overexpression in cardiomyocytes led to increased cell apoptosis and activation of p38 signaling, whereas TGFβR3 knockdown had the opposite effect. ERK1/2 and JNK1/2 signaling was not altered by TGFβR3 modulation, and p38 inhibitor (SB203580) reduced the effect of TGFβR3 on apoptosis, suggesting that p38 has a nonredundant function in activating apoptosis. Consistent with the in vitro observations, cardiac TGFβR3 transgenic mice showed augmented cardiomyocyte apoptosis, enlarged infarct size, increased injury, and enhanced p38 signaling upon MI. Conversely, cardiac loss of function of TGFβR3 by adeno‐associated viral vector serotype 9–TGFβR3 short hairpin RNA attenuated the effects of MI in mice. Conclusions TGFβR3 promotes apoptosis of cardiomyocytes via a p38 pathway–associated mechanism, and loss of TGFβR3 reduces MI injury, which suggests that TGFβR3 may serve as a novel therapeutic target for MI.
Collapse
Affiliation(s)
- Fei Sun
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xin Li
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wen-Qi Duan
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wei Tian
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Ming Gao
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jia Yang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Xia-Yang Wu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Di Huang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wei Xia
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan-Na Han
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Jia-Xin Wang
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Yan-Xin Liu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Chang-Jiang Dong
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Dan Zhao
- Department of Clinical Pharmacy (Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment), The 2nd Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Tao Ban
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| | - Wen-Feng Chu
- Department of Pharmacology (the State-Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin, China
| |
Collapse
|
28
|
Sun X, Han Q, Luo H, Pan X, Ji Y, Yang Y, Chen H, Wang F, Lai W, Guan X, Zhang Q, Tang Y, Chu J, Yu J, Shou W, Deng Y, Li X. Profiling analysis of long non-coding RNAs in early postnatal mouse hearts. Sci Rep 2017; 7:43485. [PMID: 28266538 PMCID: PMC5339910 DOI: 10.1038/srep43485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 01/02/2023] Open
Abstract
Mammalian cardiomyocytes undergo a critical hyperplastic-to-hypertrophic growth transition at early postnatal age, which is important in establishing normal physiological function of postnatal hearts. In the current study, we intended to explore the role of long non-coding (lnc) RNAs in this transitional stage. We analyzed lncRNA expression profiles in mouse hearts at postnatal day (P) 1, P7 and P28 via microarray. We identified 1,146 differentially expressed lncRNAs with more than 2.0-fold change when compared the expression profiles of P1 to P7, P1 to P28, and P7 to P28. The neighboring genes of these differentially expressed lncRNAs were mainly involved in DNA replication-associated biological processes. We were particularly interested in one novel cardiac-enriched lncRNA, ENSMUST00000117266, whose expression was dramatically down-regulated from P1 to P28 and was also sensitive to hypoxia, paraquat, and myocardial infarction. Knockdown ENSMUST00000117266 led to a significant increase of neonatal mouse cardiomyocytes in G0/G1 phase and reduction in G2/M phase, suggesting that ENSMUST00000117266 is involved in regulating cardiomyocyte proliferative activity and is likely associated with hyperplastic-to-hypertrophic growth transition. In conclusion, our data have identified a large group of lncRNAs presented in the early postnatal mouse heart. Some of these lncRNAs may have important functions in cardiac hyperplastic-to-hypertrophic growth transition.
Collapse
Affiliation(s)
- Xiongshan Sun
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Han
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hongqin Luo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaodong Pan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yan Ji
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hanying Chen
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wenjing Lai
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao Guan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Zhang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Tang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jianhong Chu
- Suzhou Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Weinian Shou
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China.,Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
29
|
Lu XL, Zhao CH, Zhang H, Yao XL. iRhom2 is involved in lipopolysaccharide-induced cardiac injury in vivo and in vitro through regulating inflammation response. Biomed Pharmacother 2017; 86:645-653. [DOI: 10.1016/j.biopha.2016.11.075] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/16/2016] [Accepted: 11/16/2016] [Indexed: 01/17/2023] Open
|
30
|
Deng Y, Zhang Q, Luo H, Chen X, Han Q, Wang F, Huang P, Lai W, Guan X, Pan X, Ji Y, Guo W, Che L, Tang Y, Gu L, Yu J, Namaka M, Deng Y, Li X. Sustained elevation of NF-κB activity sensitizes offspring of maternal inflammation to hypertension via impairing PGC-1α recovery. Sci Rep 2016; 6:32642. [PMID: 27616627 PMCID: PMC5018852 DOI: 10.1038/srep32642] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 08/11/2016] [Indexed: 02/07/2023] Open
Abstract
Growing evidence has demonstrated that maternal detrimental factors, including inflammation, contribute to the development of hypertension in the offspring. The current study found that offspring subjected to prenatal exposure of inflammation by lipopolysaccharide (LPS) challenge during the second semester showed significantly increased systolic blood pressure. In addition, these offspring also displayed augmented vascular damage and reactive oxygen species (ROS) levels in thoracic aortas when challenged with deoxycorticosterone acetate and high-salt diet (DOCA-salt). Interestingly, the antioxidant N-acetyl-L-cysteine markedly reversed these changes. Mechanistically, prenatal LPS exposure led to pre-existing elevated peroxisome proliferators-activated receptor-γ co-activator (PGC)-1α, a critical master of ROS metabolism, which up-regulated the ROS defense capacity and maintained the balance of ROS generation and elimination under resting state. However, continued elevation of NF-κB activity significantly suppressed the rapid recovery of PGC-1α expression response to DOCA-salt challenge in offspring that underwent prenatal inflammatory stimulation. This was further confirmed by using a NF-κB inhibitor (N-p-Tosyl-L-phenylalanine chloromethyl ketone) that restored PGC-1α recovery and prevented blood pressure elevation induced by DOCA-salt. Our results suggest that maternal inflammation programmed proneness to NF-κB over-activation which impaired PGC-1α-mediated anti-oxidant capacity resulting in the increased sensitivity of offspring to hypertensive damage.
Collapse
Affiliation(s)
- Yafei Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Zhang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hongqin Luo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xianhua Chen
- Diagosis and Treatment Center for Servicemen, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Qi Han
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Pei Huang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wenjing Lai
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao Guan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaodong Pan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yan Ji
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wei Guo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ling Che
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Tang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Liangqi Gu
- The Center for Disease Control and Prevention of Chengdu Military Command, Chengdu, China
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Michael Namaka
- Colleges of Pharmacy and Medicine, University of Manitoba, Winnipeg, MB, Canada
- Joint Laboratory of Biological Psychiatry Between Shantou University Medical College and the College of Medicine University of Manitoba, Shantou, China
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China
- Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|