1
|
Wen C, Tang J, Wu M, Liu H, Lin X, Fan M, Liu G, Zhang J, Liang L, Liu X, Li Y, Duan Y, Xu X. Preparation, characterization, and stability of pectin-whey protein isolate-based nanoparticles with mitochondrial targeting ability. Int J Biol Macromol 2025; 301:140383. [PMID: 39880250 DOI: 10.1016/j.ijbiomac.2025.140383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 01/13/2025] [Accepted: 01/25/2025] [Indexed: 01/31/2025]
Abstract
Quercetin (Que) is a polyhydroxy flavonoid with strong inhibitory activity against cancer cells. However, the poor water solubility and low bioavailability of Que. limit its application in the functional food industry. In the present study, the nanoparticle loaded with Que. was prepared with whey isolate protein (WPI) stabilized by triphenylphosphonium bromide (TPP) and pectin (P) as wall materials. The formation mechanism, release of Que., and antitumor activity of nanoparticles were investigated. The results showed that the optimal ratio of WPI: TPP: Que.: P in the preparation of nanoparticles (WPI-TPP-Que-P) was 50:8:1:20 (w/w/w/w). The encapsulation rate of Que. in the WPI-TPP-Que-P was 82.64 % with a particle size of 261.7 nm and a zeta potential of -42.1 mV. Compared with WPI-TPP-Que, the retention rate of WPI-TPP-Que-P increased by 4.03 % after in vitro digestion. The release kinetic result indicated that WPI-TPP-Que-P release was dominated by non-Fickian diffusion. In addition, WPI-TPP-Que-P was taken in and achieved intracellular targeting to mitochondria and promoted apoptosis (apoptosis rate: 83.6 %) by decreasing mitochondrial membrane potential and IL-10 content and improving the content of TNF-α in HepG-2 cells. This study highlights the promising application of P-modified mitochondria-targeted nanocarriers for enhanced Que. delivery.
Collapse
Affiliation(s)
- Chaoting Wen
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Jialuo Tang
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Maowei Wu
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Huimin Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Xinying Lin
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Meidi Fan
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Guoyan Liu
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Jixian Zhang
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China.
| | - Li Liang
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Xiaofang Liu
- School of Tourism and Cuisine, Yangzhou University, Yangzhou 225127, China
| | - Youdong Li
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China
| | - Yuqing Duan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Xin Xu
- College of Food Science and Engineering, Yangzhou University, Yang Zhou 225127, China.
| |
Collapse
|
2
|
Yang N, Sun H, Xi L, Zhang L, Lu Y, Wang Q, Cao J, Song J, Tang B, Shang L, Zhou X. Oroxin B Resembles Bisoprolol in Attenuating Beta1-Adrenergic Receptor Autoantibody-Induced Atrial Remodelling via the PTEN/AKT/mTOR Signalling Pathway. Clin Exp Pharmacol Physiol 2025; 52:e70011. [PMID: 39648364 DOI: 10.1111/1440-1681.70011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 10/22/2024] [Accepted: 11/11/2024] [Indexed: 12/10/2024]
Abstract
Beta1-adrenergic receptor autoantibodies (β1-AAbs) promote atrial remodelling and ultimately lead to the development of atrial fibrillation (AF). Oroxin B is a natural flavonoid glycoside with a variety of biological activities, including anti-inflammatory and autophagy-promoting effects, and has therapeutic benefits for a variety of diseases. The aim of this study was to investigate the potential therapeutic role of Oroxin B in the development of β1-AAb-induced atrial fibrillation and to elucidate the underlying mechanisms involved. We established a rat model of β1-AAb-induced atrial fibrillation via active immunisation. The first stage was divided into three groups: the control group, the β1-AAb group and the β1-AAb + bisoprolol group. The second stage was divided into three groups: the control group, the β1-AAb group and the β1-AAb + Oroxin B group. Serum levels of β1-AAbs, atrial tissue levels of cyclic monophosphate (cAMP), atrial electrophysiological parameters, cardiac structure and function, mitochondrial structure, autophagy levels, cardiomyocyte apoptosis and myocardial fibrosis were examined. The results showed that bisoprolol, a β1-blocker, improved β1-AAb-induced atrial electrical remodelling, reduced atrial collagen deposition, ameliorated the increase in LAD and regulated the balance of autophagy and apoptosis in atrial myocytes through the PTEN/AKT/mTOR signalling pathway. Oroxin B, a PTEN agonist, can improve the impairment of autophagy homeostasis and apoptosis in atrial tissue by activating the PTEN/AKT/mTOR signalling pathway, thereby improving atrial structure and electrical remodelling. Moreover, Oroxin B may play a therapeutic role in β1AAb-induced atrial fibrillation. In conclusion, our results demonstrate the potential therapeutic role of Oroxin B in β1AAb-induced atrial fibrillation and the underlying mechanisms, suggesting that Oroxin B may be an effective antiarrhythmic medication.
Collapse
Affiliation(s)
- Na Yang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Huaxin Sun
- Department of Cardiology, the Third People's Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University, Chengdu Cardiovascular Disease Research Institute, Chengdu, Sichuan, China
| | - Linqiang Xi
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Ling Zhang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yanmei Lu
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qianhui Wang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jiaru Cao
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jie Song
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Baopeng Tang
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Luxiang Shang
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, China
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xianhui Zhou
- Xinjiang Key Laboratory of Cardiac Electrophysiology and Remodelling, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Department of Pacing and Electrophysiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
3
|
Santos LD, Walker AL. The Role of Autoantibodies in Companion Animal Cardiac Disease. Vet Clin North Am Small Anim Pract 2023; 53:1367-1377. [PMID: 37423843 DOI: 10.1016/j.cvsm.2023.05.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Clinical studies exploring the role of autoimmune diseases in cardiac dysfunction have become increasingly common in both human and veterinary literature. Autoantibodies (AABs) specific to cardiac receptors have been found in human and canine dilated cardiomyopathy, and circulating autoantibodies have been suggested as a sensitive biomarker for arrhythmogenic right ventricular cardiomyopathy in people and Boxer dogs. In this article, we will summarize recent literature on AABs and their role in cardiac diseases of small animals. Despite the potential for new discoveries in veterinary cardiology, current data in veterinary medicine are limited and further studies are needed.
Collapse
Affiliation(s)
- Luís Dos Santos
- Department of Veterinary Clinical Sciences, Purdue University, College of Veterinary Medicine, 625 Harrison Street, West Lafayette, IN 47907, USA.
| | - Ashley L Walker
- William R. Pritchard Veterinary Medical Teaching Hospital, University of California, Davis, 1 Garrod Drive, Davis, CA 9561, USA
| |
Collapse
|
4
|
Huo JY, Jiang WY, Zhang SG, Lyu YT, Geng J, Chen M, Chen YY, Jiang ZX, Shan QJ. Renal denervation ameliorates cardiac metabolic remodeling in diabetic cardiomyopathy rats by suppressing renal SGLT2 expression. J Transl Med 2022; 102:341-351. [PMID: 34775493 DOI: 10.1038/s41374-021-00696-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/16/2021] [Accepted: 10/28/2021] [Indexed: 11/09/2022] Open
Abstract
This study aimed to investigate the effects of renal denervation (RDN) on diabetic cardiomyopathy (DCM) and explore the related mechanisms. Male Sprague-Dawley rats were fed high-fat chow and injected with low-dose streptozotocin to establish a DCM model. Six rats served as controls. The surviving rats were divided into three groups: control group, DCM group and DCM + RDN group. RDN surgery was performed in the fifth week. At the end of the experiment, all rats were subjected to 18F-FDG PET/CT and metabolic cage studies. Cardiac function and structure were evaluated by echocardiography and histology. Myocardial substrate metabolism and mitochondrial function were assessed by multiple methods. In the 13th week, the DCM rats exhibited cardiac hypertrophy and interstitial fibrosis accompanied by diastolic dysfunction. RDN ameliorated DCM-induced cardiac dysfunction (E/A ratio: RDN 1.07 ± 0.18 vs. DCM 0.93 ± 0.12, P < 0.05; E/E' ratio: RDN 10.74 ± 2.48 vs. DCM 13.25 ± 1.99, P < 0.05) and pathological remodeling (collagen volume fraction: RDN 5.05 ± 2.05% vs. DCM 10.62 ± 2.68%, P < 0.05). Abnormal myocardial metabolism in DCM rats was characterized by suppressed glucose metabolism and elevated lipid metabolism. RDN increased myocardial glucose uptake and oxidation while reducing the absorption and utilization of fatty acids. Meanwhile, DCM decreased mitochondrial ATP content, depolarized the membrane potential and inhibited the activity of respiratory chain complexes, but RDN attenuated this mitochondrial damage (ATP: RDN 30.98 ± 7.33 μmol/gprot vs. DCM 22.89 ± 5.90 μmol/gprot, P < 0.05; complexes I, III and IV activity: RDN vs. DCM, P < 0.05). Furthermore, both SGLT2 inhibitor and the combination treatment produced similar effects as RDN alone. Thus, RDN prevented DCM-induced cardiac dysfunction and pathological remodeling, which is related to the improvement of metabolic disorders and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jun-Yu Huo
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wan-Ying Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shi-Geng Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yi-Ting Lyu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jie Geng
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meng Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yuan-Yuan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhi-Xin Jiang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Qi-Jun Shan
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
5
|
Hou W, Hao Y, Sun L, Zhao Y, Zheng X, Song L. The dual roles of autophagy and the GPCRs-mediating autophagy signaling pathway after cerebral ischemic stroke. Mol Brain 2022; 15:14. [PMID: 35109896 PMCID: PMC8812204 DOI: 10.1186/s13041-022-00899-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Ischemic stroke, caused by a lack of blood supply in brain tissues, is the third leading cause of human death and disability worldwide, and usually results in sensory and motor dysfunction, cognitive impairment, and in severe cases, even death. Autophagy is a highly conserved lysosome-dependent process in which eukaryotic cells removal misfolded proteins and damaged organelles in cytoplasm, which is critical for energy metabolism, organelle renewal, and maintenance of intracellular homeostasis. Increasing evidence suggests that autophagy plays important roles in pathophysiological mechanisms under ischemic conditions. However, there are still controversies about whether autophagy plays a neuroprotective or damaging role after ischemia. G-protein-coupled receptors (GPCRs), one of the largest protein receptor superfamilies in mammals, play crucial roles in various physiological and pathological processes. Statistics show that GPCRs are the targets of about one-fifth of drugs known in the world, predicting potential values as targets for drug research. Studies have demonstrated that nutritional deprivation can directly or indirectly activate GPCRs, mediating a series of downstream biological processes, including autophagy. It can be concluded that there are interactions between autophagy and GPCRs signaling pathway, which provides research evidence for regulating GPCRs-mediated autophagy. This review aims to systematically discuss the underlying mechanism and dual roles of autophagy in cerebral ischemia, and describe the GPCRs-mediated autophagy, hoping to probe promising therapeutic targets for ischemic stroke through in-depth exploration of the GPCRs-mediated autophagy signaling pathway.
Collapse
Affiliation(s)
- Weichen Hou
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Li Sun
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Yang Zhao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China
| | - Xiangyu Zheng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| | - Lei Song
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Xinmin Street 71#, Changchun, 130021, China.
| |
Collapse
|
6
|
Sukumaran V, Gurusamy N, Yalcin HC, Venkatesh S. Understanding diabetes-induced cardiomyopathy from the perspective of renin angiotensin aldosterone system. Pflugers Arch 2022; 474:63-81. [PMID: 34967935 PMCID: PMC12120836 DOI: 10.1007/s00424-021-02651-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 12/02/2021] [Accepted: 12/03/2021] [Indexed: 12/31/2022]
Abstract
Experimental and clinical evidence suggests that diabetic subjects are predisposed to a distinct cardiovascular dysfunction, known as diabetic cardiomyopathy (DCM), which could be an autonomous disease independent of concomitant micro and macrovascular disorders. DCM is one of the prominent causes of global morbidity and mortality and is on a rising trend with the increase in the prevalence of diabetes mellitus (DM). DCM is characterized by an early left ventricle diastolic dysfunction associated with the slow progression of cardiomyocyte hypertrophy leading to heart failure, which still has no effective therapy. Although the well-known "Renin Angiotensin Aldosterone System (RAAS)" inhibition is considered a gold-standard treatment in heart failure, its role in DCM is still unclear. At the cellular level of DCM, RAAS induces various secondary mechanisms, adding complications to poor prognosis and treatment of DCM. This review highlights the importance of RAAS signaling and its major secondary mechanisms involving inflammation, oxidative stress, mitochondrial dysfunction, and autophagy, their role in establishing DCM. In addition, studies lacking in the specific area of DCM are also highlighted. Therefore, understanding the complex role of RAAS in DCM may lead to the identification of better prognosis and therapeutic strategies in treating DCM.
Collapse
Affiliation(s)
| | - Narasimman Gurusamy
- Department of Bioscience Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Huseyin C Yalcin
- Biomedical Research Center, Qatar University, Al-Tarfa, 2371, Doha, Qatar
| | - Sundararajan Venkatesh
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers-New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
7
|
Sun C, Lu J, Long Y, Guo S, Jia W, Ning N, Hao H, Wang X, Bian Y, Liu H, Wang L. Adiponectin up-regulates the decrease of myocardial autophagic flux induced by β 1 -adrenergic receptor autoantibody partly dependent on AMPK. J Cell Mol Med 2021; 25:8464-8478. [PMID: 34322993 PMCID: PMC8419161 DOI: 10.1111/jcmm.16807] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/28/2021] [Accepted: 07/05/2021] [Indexed: 12/20/2022] Open
Abstract
Cardiomyocytes autophagy is essential for maintaining cardiac function. Our previous studies have found that β1‐adrenergic receptor autoantibody (β1‐AA) induced the decreased myocardial autophagic flux, which resulted in cardiomyocyte death and cardiac dysfunction. And other studies demonstrated that β1‐AA induced the decrease of AMPK phosphorylation, the key hub of autophagy pathway, while adiponectin up‐regulated autophagic flux mediated by AMPK. However, it is not clear whether adiponectin improves the inhibition of myocardial autophagic flux induced by β1‐AA by up‐regulating the level of AMPK phosphorylation. In this study, it has been confirmed that β1‐AA induced the decrease of AMPK phosphorylation level in both vivo and vitro. Moreover, pretreatment of cardiomyocytes with AMPK inhibitor Compound C could further reduce the autophagic flux induced by β1‐AA. Adiponectin deficiency could aggravate the decrease of myocardial AMPK phosphorylation level, autophagic flux and cardiac function induced by β1‐AA. Further, exogenous adiponectin could reverse the decline of AMPK phosphorylation level and autophagic flux induced by β1‐AA and even reduce cardiomyocyte death. While pretreated with the Compound C, the adiponectin treatment did not improve the decreased autophagosome formation, but still improved the decreased autophagosome clearance induced by β1‐AA in cardiomyocytes. This study is the first time to confirm that β1‐AA could inhibit myocardial autophagic flux by down‐regulating AMPK phosphorylation level. Adiponectin could improve the inhibition of myocardial autophagic flux induced by β1‐AA partly dependent on AMPK, so as to provide an experimental basis for the treatment of patients with β1‐AA‐positive cardiac dysfunction.
Collapse
Affiliation(s)
- Cong Sun
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China.,Department of Pathology, Linfen Central Hospital, Linfen, China
| | - Jiebei Lu
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Yaolin Long
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Shuai Guo
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Weiwei Jia
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Na Ning
- Department of Pathology, Shanxi Medical University, Taiyuan, China
| | - Haihu Hao
- Department of Orthopedics, Shanxi Bethune Hospital & Shanxi Academy of Medical Sciences, Taiyuan, China
| | - Xiaohui Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| | - Yunfei Bian
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Li Wang
- Department of Pathology, Shanxi Medical University, Taiyuan, China.,Key Laboratory of Cellular Physiology (Shanxi Medical University), Ministry of Education, Taiyuan, China
| |
Collapse
|
8
|
Jiang X, Shao M, Liu X, Liu X, Zhang X, Wang Y, Yin K, Wang S, Hu Y, Jose PA, Zhou Z, Xu F, Yang Z. Reversible Treatment of Pressure Overload-Induced Left Ventricular Hypertrophy through Drd5 Nucleic Acid Delivery Mediated by Functional Polyaminoglycoside. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2003706. [PMID: 33717857 PMCID: PMC7927605 DOI: 10.1002/advs.202003706] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Indexed: 05/12/2023]
Abstract
Left ventricular hypertrophy and fibrosis are major risk factors for heart failure, which require timely and effective treatment. Genetic therapy has been shown to ameliorate hypertrophic cardiac damage. In this study, it is found that in mice, the dopamine D5 receptor (D5R) expression in the left ventricle (LV) progressively decreases with worsening of transverse aortic constriction-induced left ventricular hypertrophy. Then, a reversible treatment of left ventricular hypertrophy with Drd5 nucleic acids delivered by tobramycin-based hyperbranched polyaminoglycoside (SS-HPT) is studied. The heart-specific increase in D5R expression by SS-HPT/Drd5 plasmid in the early stage of left ventricular hypertrophy attenuates cardiac hypertrophy and fibrosis by preventing oxidative and endoplasmic reticulum (ER) stress and ameliorating autophagic dysregulation. By contrast, SS-HPT/Drd5 siRNA promotes the progression of left ventricular hypertrophy and accelerates the deterioration of myocardial function into heart failure. The reduction in cardiac D5R expression and dysregulated autophagy are observed in patients with hypertrophic cardiomyopathy and heart failure. The data show a cardiac-specific beneficial effect of SS-HPT/Drd5 plasmid on myocardial remodeling and dysfunction, which may provide an effective therapy of patients with left ventricular hypertrophy and heart failure.
Collapse
Affiliation(s)
- Xiaoliang Jiang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Meiyu Shao
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Xue Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xing Liu
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| | - Xu Zhang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Yuming Wang
- Department of Hepato‐Biliary‐Pancreatic SurgeryHenan Provincial People's HospitalPeople's Hospital of Zhengzhou UniversityZhengzhouHenan450003P. R. China
| | - Kunlun Yin
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Shuiyun Wang
- Department of Cardiovascular SurgeryState Key Laboratory of Cardiovascular DiseaseFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Yang Hu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Pedro A Jose
- Department of Pharmacology and PhysiologyThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
- Department of MedicineDivision of Kidney Diseases & HypertensionThe George Washington University School of Medicine & Health SciencesWashingtonDC20052USA
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular DiseaseBeijing Key Laboratory for Molecular Diagnostics of Cardiovascular DiseasesDiagnostic Laboratory ServiceFuwai HospitalNational Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037P. R. China
| | - Fu‐Jian Xu
- Key Lab of Biomedical Materials of Natural MacromoleculesMinistry of EducationBeijing Laboratory of Biomedical MaterialsBeijing Advanced Innovation Center for Soft Matter Science and EngineeringBeijing University of Chemical TechnologyBeijing100029P. R. China
| | - Zhiwei Yang
- NHC Key Laboratory of Human Disease Comparative Medicine (The Institute of Laboratory Animal Sciences, CAMS & PUMC), and Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases5 Pan Jia Yuan Nan Li, Chaoyang DistrictBeijing100021P. R. China
| |
Collapse
|
9
|
Chen G, Jin X, Luo D, Ai J, Xiao K, Lai J, He Q, Li H, Wang K. β-Adrenoceptor regulates contraction and inflammatory cytokine expression of human bladder smooth muscle cells via autophagy under pathological hydrostatic pressure. Neurourol Urodyn 2020; 39:2128-2138. [PMID: 32949194 DOI: 10.1002/nau.24517] [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: 07/29/2020] [Revised: 08/30/2020] [Accepted: 09/08/2020] [Indexed: 02/05/2023]
Abstract
AIMS Abnormal intravesical pressure created by partial bladder outlet obstruction (PBOO) triggered the progression from chronic inflammation to fibrosis, initiating structural and functional alterations of bladder. To elucidate the underlying mechanisms of contraction and inflammatory response, we investigated the isolated human bladder smooth muscle cells (hBSMC) under pathological hydrostatic pressure (HP) mimicking the in vivo PBOO condition. METHODS hBSMCs were subjected to HP of 200 cm H2 O to explore the contraction and inflammatory cytokine expression of hBSMC treated with β-adrenoceptors (ADRBs) and/or autophagy signaling pathway agonists and/or antagonists. RESULTS We showed that pathological HP induced the release of the proinflammatory cytokines, including monocyte chemotactic protein-1, regulated upon activation normal T cell expressed and secreted factor, and interleukin-6. HP downregulated ADRB2 and ADRB3 expression, which was consistent with the results of the PBOO rat model. ADRB2 or autophagy activation repressed pathological HP-induced proinflammatory cytokine production. ADRB2, ADRB3 or autophagy activation ameliorated the HP-enhanced contraction. The increased contraction and autophagy activity by ADRB2 agonist under HP conditions were reversed by pretreatment with antagonists of adenosine monophosphate-activated protein kinase (AMPK). CONCLUSION The present study provides evidence that the ADRB3 agonist suppresses hBSMC contraction under pathological HP conditions. Moreover, the ADRB2 agonist negatively regulates the contraction and inflammatory response of hBSMCs through AMPK/mTOR-mediated autophagy under pathological HP. These findings provide a theoretical basis for potential therapeutic strategies for patients with PBOO.
Collapse
Affiliation(s)
- Guo Chen
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China.,Department of Surgery, Division of Urology, No.4 West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xi Jin
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | | | - Jianzhong Ai
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kaiwen Xiao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Junyu Lai
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qin He
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Li
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kunjie Wang
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Rangarajan N, Kapoor I, Li S, Drossopoulos P, White KK, Madden VJ, Dohlman HG. Potassium starvation induces autophagy in yeast. J Biol Chem 2020; 295:14189-14202. [PMID: 32788210 DOI: 10.1074/jbc.ra120.014687] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/02/2020] [Indexed: 01/04/2023] Open
Abstract
Autophagy is a conserved process that recycles cellular contents to promote survival. Although nitrogen limitation is the canonical inducer of autophagy, recent studies have revealed several other nutrients important to this process. In this study, we used a quantitative, high-throughput assay to identify potassium starvation as a new and potent inducer of autophagy in the yeast Saccharomyces cerevisiae We found that potassium-dependent autophagy requires the core pathway kinases Atg1, Atg5, and Vps34, and other components of the phosphatidylinositol 3-kinase complex. Transmission EM revealed abundant autophagosome formation in response to both stimuli. RNA-Seq indicated distinct transcriptional responses: nitrogen affects transport of ions such as copper, whereas potassium targets the organization of other cellular components. Thus, nitrogen and potassium share the ability to influence molecular supply and demand but do so in different ways. Both inputs promote catabolism through bulk autophagy, but result in distinct mechanisms of cellular remodeling and synthesis.
Collapse
Affiliation(s)
- Nambirajan Rangarajan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ishani Kapoor
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Shuang Li
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Peter Drossopoulos
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Kristen K White
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria J Madden
- Microscopy Services Laboratory, Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Henrik G Dohlman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
11
|
Peng Z, Zhan H, Shao Y, Xiong Y, Zeng L, Zhang C, Liu Z, Huang Z, Su H, Yang Z. 13-Methylberberine improves endothelial dysfunction by inhibiting NLRP3 inflammasome activation via autophagy induction in human umbilical vein endothelial cells. Chin Med 2020; 15:8. [PMID: 31993073 PMCID: PMC6977264 DOI: 10.1186/s13020-020-0286-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Accepted: 01/09/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atherosclerosis, the underlying cause of the majority of cardiovascular diseases, is a lipid-driven, inflammatory disease of the large arteries. Atherosclerotic cardiovascular disease (ASCVD) threatens human lives due to high morbidity and mortality. Many studies have demonstrated that atherosclerosis is accelerated via activation of the NLRP3 inflammasome. The NLRP3 inflammasome plays a critical role in the development of vascular inflammation and atherosclerosis. In atherosclerotic plaques, excessive generation of reactive oxygen species (ROS) activates the NLRP3 inflammasome. 13-Methylberberine (13-MB) is a newly synthesized compound used in traditional Chinese medicine that has outstanding antibacterial, antitumor, and antiobesity activities, especially anti-inflammatory activity. However, the role of 13-MB in atherosclerosis needs to be explored. METHODS CCK-8 assays and flow cytometry were conducted to determine the cell viability and apoptotic profiles of human umbilical vein endothelial cells (HUVECs) treated with 13-MB. Carboxy-DCFH-DA and JC-10 assays were used to measure ROS and determine mitochondrial membrane potential. Western blot analysis was performed to investigate proteins that are associated with the NLRP3 inflammasome and autophagy. ELISA was used to detect and quantify inflammatory cytokines related to the NLRP3 inflammasome. Transfection and confocal microscopy were conducted to observe autophagy. RESULTS Pretreatment with 13-MB markedly reduced cytotoxicity and apoptosis, as well as intracellular ROS production, in H2O2-induced HUVECs. Moreover, 13-MB showed a protective effect in maintaining mitochondrial membrane potential. 13-MB also suppressed NLRP3 inflammasome activation and promoted autophagy induction in HUVECs. CONCLUSION 13-MB exerts cytoprotective effects in an H2O2-induced cell injury model by inhibiting NLRP3 inflammasome activation via autophagy induction in HUVECs. These anti-inflammatory and autophagy induction activities may provide valuable evidence for further investigating the potential role of 13-MB in atherosclerosis.
Collapse
Affiliation(s)
- Zhihua Peng
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Hong Zhan
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Yijia Shao
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
| | - Yan Xiong
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Lijin Zeng
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Cong Zhang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhihao Liu
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Zhenhua Huang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| | - Huanxing Su
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Zhen Yang
- Division of Emergency Medicine, Department of Emergency Intensive Care Unit, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
- Department of Cardiology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 Guangdong China
- Key Laboratory on Assisted Circulation, Ministry of Health, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080 China
| |
Collapse
|
12
|
Rangarajan N, Gordy CL, Askew L, Bevill SM, Elston TC, Errede B, Hurst JH, Kelley JB, Sheetz JB, Suzuki SK, Valentin NH, Young E, Dohlman HG. Systematic analysis of F-box proteins reveals a new branch of the yeast mating pathway. J Biol Chem 2019; 294:14717-14731. [PMID: 31399514 DOI: 10.1074/jbc.ra119.010063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/06/2019] [Indexed: 11/06/2022] Open
Abstract
The mating pathway in yeast Saccharomyces cerevisiae has long been used to reveal new mechanisms of signal transduction. The pathway comprises a pheromone receptor, a heterotrimeric G protein, and intracellular effectors of morphogenesis and transcription. Polarized cell growth, in the direction of a potential mating partner, is accomplished by the G-protein βγ subunits and the small G-protein Cdc42. Transcription induction, needed for cell-cell fusion, is mediated by Gβγ and the mitogen-activated protein kinase (MAPK) scaffold protein Ste5. A potential third pathway is initiated by the G-protein α subunit Gpa1. Gpa1 signaling was shown previously to involve the F-box adaptor protein Dia2 and an endosomal effector protein, the phosphatidylinositol 3-kinase Vps34. Vps34 is also required for proper vacuolar sorting and autophagy. Here, using a panel of reporter assays, we demonstrate that mating pheromone stimulates vacuolar targeting of a cytoplasmic reporter protein and that this process depends on Vps34. Through a systematic analysis of F-box deletion mutants, we show that Dia2 is required to sustain pheromone-induced vacuolar targeting. We also found that other F-box proteins selectively regulate morphogenesis (Ydr306, renamed Pfu1) and transcription (Ucc1). These findings point to the existence of a new and distinct branch of the pheromone-signaling pathway, one that likely leads to vacuolar engulfment of cytoplasmic proteins and recycling of cellular contents in preparation for mating.
Collapse
Affiliation(s)
- Nambirajan Rangarajan
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Claire L Gordy
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Lauren Askew
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Samantha M Bevill
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Timothy C Elston
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Beverly Errede
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Jillian H Hurst
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Kelley
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Joshua B Sheetz
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Sara Kimiko Suzuki
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalie H Valentin
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Everett Young
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Henrik G Dohlman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
13
|
Chia LY, Evans BA, Mukaida S, Bengtsson T, Hutchinson DS, Sato M. Adrenoceptor regulation of the mechanistic target of rapamycin in muscle and adipose tissue. Br J Pharmacol 2019; 176:2433-2448. [PMID: 30740664 PMCID: PMC6592864 DOI: 10.1111/bph.14616] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 01/08/2019] [Accepted: 01/21/2019] [Indexed: 12/16/2022] Open
Abstract
A vital role of adrenoceptors in metabolism and energy balance has been well documented in the heart, skeletal muscle, and adipose tissue. It has been only recently demonstrated, however, that activation of the mechanistic target of rapamycin (mTOR) makes a significant contribution to various metabolic and physiological responses to adrenoceptor agonists. mTOR exists as two distinct complexes named mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) and has been shown to play a critical role in protein synthesis, cell proliferation, hypertrophy, mitochondrial function, and glucose uptake. This review will describe the physiological significance of mTORC1 and 2 as a novel paradigm of adrenoceptor signalling in the heart, skeletal muscle, and adipose tissue. Understanding the detailed signalling cascades of adrenoceptors and how they regulate physiological responses is important for identifying new therapeutic targets and identifying novel therapeutic interventions. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Ling Yeong Chia
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Bronwyn A. Evans
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Saori Mukaida
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Tore Bengtsson
- Department of Molecular Biosciences, The Wenner‐Gren InstituteStockholm UniversityStockholmSweden
| | - Dana S. Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Masaaki Sato
- Drug Discovery Biology, Monash Institute of Pharmaceutical SciencesMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
14
|
Honokiol Protects against Anti- β1-Adrenergic Receptor Autoantibody-Induced Myocardial Dysfunction via Activation of Autophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1640804. [PMID: 30116474 PMCID: PMC6079338 DOI: 10.1155/2018/1640804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 05/04/2018] [Accepted: 05/30/2018] [Indexed: 12/19/2022]
Abstract
Myocardial diseases are prevalent syndromes with high mortality rate. The exploration of effective interference is important. Anti-β1-adrenergic receptor autoantibody (β1-AAB) is highly correlated with myocardial dysfunction. The actions and underlying mechanisms of honokiol (HNK) in β1-AAB-positive patients await to be unraveled. In this study, we established a rat model of β1-AAB positive with myocardial dysfunction. Cardiac function following β1-AR-ECII administration was analyzed using the VisualSonics Vevo 770 High-Resolution In Vivo Imaging System. The levels of autophagy-related proteins were detected by Western blotting. Our data revealed that HNK reversed β1-AAB-induced effects and protected myocardial tissues from dysfunction. After HNK treatment, the cardiac contractile ability increased and the LDH activity decreased. HNK attenuated myocardial degeneration. In addition, HNK promoted the activation of the AMP-dependent protein kinase/Unc-51-like autophagy activating kinase (AMPK/ULK) pathway and activated autophagy. These results suggest that HNK protects against β1-AAB-induced myocardial dysfunction via activation of autophagy and it may be a potentially therapeutic compound for β1-AAB-positive myocardial diseases.
Collapse
|
15
|
Abstract
It has been recognized that myocardial apoptosis is one major factor in the development of heart dysfunction and autophagy has been shown to influence the apoptosis. In previous studies, we reported that anti-β1-adrenergic receptor autoantibodies (β1-AABs) decreased myocardial autophagy, but the role of decreased autophagy in cardiomyocyte apoptosis remains unclear. In the present study, we used a β1-AAB-immunized rat model to investigate the role of decreased autophagy in cardiomyocyte apoptosis. We reported that the level of autophagic flux increased early and then decreased in an actively β1-AAB-immunized rat model. Rapamycin, an mTOR inhibitor, restored myocardial apoptosis in the presence of β1-AABs. Further, we found that the early increase of autophagy was an adaptive stress response that is possibly unrelated to β1-AR, and the activation of the β1-AR and PKA contributed to late decreased autophagy. Then, after upregulating or inhibiting autophagy with rapamycin, Atg5 overexpression adenovirus or 3-methyladenine in cultured primary neonatal rat cardiomyocytes, we found that autophagy decline promoted myocardial apoptosis effectively through the mitochondrial apoptotic pathway. In conclusion, the reduction of apoptosis through the proper regulation of autophagy may be important for treating patients with β1-AAB-positive heart dysfunction.
Collapse
|
16
|
Lin FH, Zhang WL, Li H, Tian XD, Zhang J, Li X, Li CY, Tan JH. Role of autophagy in modulating post-maturation aging of mouse oocytes. Cell Death Dis 2018; 9:308. [PMID: 29472597 PMCID: PMC5833823 DOI: 10.1038/s41419-018-0368-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/29/2018] [Accepted: 01/30/2018] [Indexed: 11/30/2022]
Abstract
Mechanisms for post-maturation oocyte aging (PMOA) are not fully understood, and whether autophagy plays any role in PMOA is unknown. To explore the role of autophagy in PMOA, expression of autophagosomes and effects of the autophagy (macro-autophagy) activity on PMOA were observed in mouse oocytes. Oocyte activation rates and active caspase-3 levels increased continuously from 0 to 18 h of in vitro aging. While levels of microtubule-associated protein light chain 3 (LC3)-II increased up to 12 h and decreased thereafter, contents of p62 decreased from 0 to 12 h and then elevated to basal level by 18 h. However, the LC3-II/I ratio remained unchanged following aging in different media or for different times. During in vitro aging up to 12 h, upregulating autophagy with rapamycin or lithium chloride decreased activation susceptibility, cytoplasmic calcium, p62 contents, oxidative stress, caspase-3 activation and cytoplasmic fragmentation while increasing developmental competence, LC3-II contents, LC3-II/I ratio, mitochondrial membrane potential, spindle/chromosome integrity and normal cortical granule distribution. Downregulating autophagy with 3-methyladenine (3-MA) produced opposite effects on all these parameters except cytoplasmic fragmentation. After 12 h of aging culture, however, regulating autophagy with either rapamycin/lithium chloride or 3-MA had no impact on oocyte activation susceptibility. It is concluded that autophagy plays an important role in regulating PMOA. Thus, during the early stage of PMOA, autophagy increases as an adaptive response to prevent further apoptosis, but by the late stage of PMOA, the activation of more caspases blocks the autophagic process leading to severer apoptosis.
Collapse
Affiliation(s)
- Fei-Hu Lin
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Wei-Ling Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Hong Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Xiao-Dan Tian
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Jie Zhang
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Xiao Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Chuan-Yong Li
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China
| | - Jing-He Tan
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an City, 271018, P. R. China.
| |
Collapse
|
17
|
Guo Y, Li Z, Shi C, Li J, Yao M, Chen X. Trichostatin A attenuates oxidative stress-mediated myocardial injury through the FoxO3a signaling pathway. Int J Mol Med 2017; 40:999-1008. [PMID: 28849190 PMCID: PMC5593460 DOI: 10.3892/ijmm.2017.3101] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Accepted: 08/03/2017] [Indexed: 11/06/2022] Open
Abstract
Trichostatin A (TSA), a histone deacetylase inhibitor, is widely used as an anticancer drug. Recently, TSA has been shown to exert a protective effect on ischemia/reperfusion (I/R) injury; however, the underlying mechanisms remain unclear. Forkhead box O3a (FoxO3a), a unique FoxO family member, has been shown to attenuate myocardial injury by increasing resistance to oxidative stress in mice. The present study aimed to investigate whether TSA exerts its cardioprotective effects through the FoxO3a signaling pathway. For this purpose, healthy male Wistar rats were pre-treated with TSA for 5 days before they were subjected to ligation/relaxation of the left anterior descending branch of the coronary artery and to 30 min of ischemia, followed by 24 h of reperfusion. The activities of creatine kinase (CK), lactate dehydrogenase (LDH), aspartate aminotransferase (AST) and superoxide diamutase (SOD), as well as the malondialdehyde (MDA) levels were examined. The H9c2 rat myocardial cell line was cultured in 10% FBS-containing DMEM for 24 h. The cells were incubated with/without TSA (50 nmol/l) for 1 h and then incubated with/without H2O2 (400 µM) for 2 h. Reactive oxygen species (ROS) and mitochondrial membrane potential (Δψm) were measured by probe staining in the H9c2 cells. The expression of FoxO3a, mitochondrial SOD2 and catalase was quantified by western blot analysis. The levels of H3 and H4 acetylation of the FoxO3a promoter region were examined by chromatin immunoprecipitation assay. TSA significantly reduced the myocardial infarct size and the activities of serum LDH, AST and CK in the rats. TSA also decreased the levels of MDA and increased the activities of SOD in the myocardial tissue of the rats. Consistent with the reduced injury to the TSA-treated rats, TSA significantly reduced the H2O2-induced levels of ROS and increased Δψm. In addition, TSA increased the expression of FoxO3a, SOD2 and catalase, which may be related to increasing the level of H4 acetylation of the FoxO3a promoter region. Our results thus revealed that TSA protected the myocardium from oxidative stress-mediated damage by increasing H4 acetylation of the FoxO3a promoter region, and the expression of FoxO3a, SOD2 and catalase.
Collapse
Affiliation(s)
- Yunhui Guo
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Zhiping Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Canxia Shi
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jia Li
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Meng Yao
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xia Chen
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
18
|
Sinha RA, Singh BK, Yen PM. Reciprocal Crosstalk Between Autophagic and Endocrine Signaling in Metabolic Homeostasis. Endocr Rev 2017; 38:69-102. [PMID: 27901588 DOI: 10.1210/er.2016-1103] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 11/28/2016] [Indexed: 12/19/2022]
Abstract
Autophagy is a cellular quality control and energy-providing process that is under strict control by intra- and extracellular stimuli. Recently, there has been an exponential increase in autophagy research and its implications for mammalian physiology. Autophagy deregulation is now being implicated in many human diseases, and its modulation has shown promising results in several preclinical studies. However, despite the initial discovery of autophagy as a hormone-regulated process by De Duve in the early 1960s, endocrine regulation of autophagy still remains poorly understood. In this review, we provide a critical summary of our present understanding of the basic mechanism of autophagy, its regulation by endocrine hormones, and its contribution to endocrine and metabolic homeostasis under physiological and pathological settings. Understanding the cross-regulation of hormones and autophagy on endocrine cell signaling and function will provide new insight into mammalian physiology as well as promote the development of new therapeutic strategies involving modulation of autophagy in endocrine and metabolic disorders.
Collapse
Affiliation(s)
- Rohit A Sinha
- Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School Singapore, Singapore 169016
| | - Brijesh K Singh
- Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School Singapore, Singapore 169016
| | - Paul M Yen
- Program of Cardiovascular and Metabolic Disorders, Duke-National University of Singapore Medical School Singapore, Singapore 169016
| |
Collapse
|
19
|
Tang B, Shen F, Wan D, Guo BH, Wang YJ, Yi QY, Liu YJ. DNA-binding, molecular docking studies and biological activity studies of ruthenium(ii) polypyridyl complexes. RSC Adv 2017. [DOI: 10.1039/c7ra05103d] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Three new Ru(ii) complexes [Ru(N–N)2(PTCP)]2+ were synthesized and characterized. The DNA-binding, in vitro cytotoxicity, apoptosis, autophagy and western blot analysis were investigated.
Collapse
Affiliation(s)
- Bing Tang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Fang Shen
- Department of Applied Chemistry
- South China Agricultural University
- Guangzhou 510642
- P. R. China
| | - Dan Wan
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Bo-Hong Guo
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Yang-Jie Wang
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Qiao-Yan Yi
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
| | - Yun-Jun Liu
- School of Pharmacy
- Guangdong Pharmaceutical University
- Guangzhou
- P. R. China
- Guangdong Cosmetics Engineering & Technology Research Center
| |
Collapse
|
20
|
Hou Z, Deng K, Li C, Deng X, Lian H, Cheng Z, Jin D, Lin J. 808 nm Light-triggered and hyaluronic acid-targeted dual-photosensitizers nanoplatform by fully utilizing Nd3+-sensitized upconversion emission with enhanced anti-tumor efficacy. Biomaterials 2016; 101:32-46. [DOI: 10.1016/j.biomaterials.2016.05.024] [Citation(s) in RCA: 140] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Revised: 05/18/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022]
|
21
|
Lv T, Du Y, Cao N, Zhang S, Gong Y, Bai Y, Wang W, Liu H. Proliferation in cardiac fibroblasts induced by β1-adrenoceptor autoantibody and the underlying mechanisms. Sci Rep 2016; 6:32430. [PMID: 27577254 PMCID: PMC5006240 DOI: 10.1038/srep32430] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 08/09/2016] [Indexed: 12/25/2022] Open
Abstract
Chronic sustained stimulation of β-adrenoceptor is closely related to cardiac fibrosis which is bad for cardiac function. Growing evidence showed that the high prevalence of β1-adrenoceptor autoantibody (β1-AA) in the sera of patients with various types of cardiovascular diseases decreased cardiac function. In the current study, we demonstrated that β1-AA impaired the cardiac function evaluated by echocardiography and that β1-AA triggered cardiac fibrosis in terms of increased expression of α-smooth muscle actin as the marker of myofibroblast and collagen deposition in a passive β1-AA immunized mice model during 16 weeks. Further, we showed that β1-AA activated β1-AR/cAMP/PKA pathway and promoted proliferation in primary cardiac fibroblasts through specific binding to β1-AR but not to β2-AR. Moreover, β1-AA was also likely to promote proliferation in cardiac fibroblasts through activating p38MAPK and ERK1/2 as p38MAPK inhibitor SB203580 and ERK1/2 inhibitor PD98059 partially reversed the proliferative effect. The persistent activating signalling of PKA and P38MAPK in 1 h induced by β1-AA was associated with lacking agonist-induced desensitization phenomena. The conditioned medium from β1-AA-stimulated cardiac fibroblasts induced cardiomyocyte apoptosis, which indicated that β1-AA changed the secretion of cardiac fibroblasts contributing to cardiac injury. These findings will contribute to our understanding of the pathological mechanisms of β1-AA.
Collapse
Affiliation(s)
- Tingting Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yunhui Du
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Ning Cao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yulin Gong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Yan Bai
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China
| | - Wen Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China.,Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| |
Collapse
|
22
|
Decreased autophagy: a major factor for cardiomyocyte death induced by β1-adrenoceptor autoantibodies. Cell Death Dis 2015; 6:e1862. [PMID: 26313913 PMCID: PMC4558518 DOI: 10.1038/cddis.2015.237] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 06/27/2015] [Accepted: 07/20/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyocyte death is one major factor in the development of heart dysfunction, thus, understanding its mechanism may help with the prevention and treatment of this disease. Previously, we reported that anti-β1-adrenergic receptor autoantibodies (β1-AABs) decreased myocardial autophagy, but the role of these in cardiac function and cardiomyocyte death is unclear. We report that rapamycin, an mTOR inhibitor, restored cardiac function in a passively β1-AAB-immunized rat model with decreased cardiac function and myocardial autophagic flux. Next, after upregulating or inhibiting autophagy with Beclin-1 overexpression/rapamycin or RNA interference (RNAi)-mediated expression of Beclin-1/3-methyladenine, β1-AAB-induced autophagy was an initial protective stress response before apoptosis. Then, decreased autophagy contributed to cardiomyocyte death followed by decreases in cardiac function. In conclusion, proper regulation of autophagy may be important for treating patients with β1-AAB-positive heart dysfunction.
Collapse
|
23
|
Zhang S, He Z, Wang J, Wang L, Wu Y, Wang J, Lv T, Liu H. Mitochondrial Ultrastructural Alterations and Declined M2 Receptor Density Were Involved in Cardiac Dysfunction in Rats after Long Term Treatment with Autoantibodies against M2 Muscarinic Receptor. PLoS One 2015; 10:e0129563. [PMID: 26086781 PMCID: PMC4472961 DOI: 10.1371/journal.pone.0129563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2014] [Accepted: 05/11/2015] [Indexed: 12/03/2022] Open
Abstract
Background Previous studies showed that autoantibodies (M2-AA) against the second extracellular loop of M2 muscarinic receptor (M2AChR-el2) from dilated cardiomyopathy (DCM) serum could induce DCM-like morphological changes in mice hearts. However, the effects of M2-AA on the cardiac function during the process of DCM and the potential mechanisms are not fully known. The present study was designed to dynamically observe the cardiac function, mitochondrial changes, and M2 receptor binding characteristics in rats long-term stimulated with M2-AA in vivo. Methods M2-AA-positive model was established by actively immunizing healthy male Wistar rats with synthetic M2AChR-el2 peptide for 18 months. Meanwhile, vehicle group rats were administrated with physiological saline. The change of mitochondrial membrane potential (ΔΨm) was detected by radionuclide imaging. The ultrastructure of mitochondria was observed under electron microscopy. The M2 receptor binding characteristics were determined by radioactive ligand binding assay. Results After immunization for 12 months, compared with vehicle group, M2AChR-el2-immunized rats showed decreased myocardial contractility and cardiac diastolic function evidenced by declined maximal rate of rise of ventricular pressure and increased left ventricular end-diastolic pressure, respectively. Additionally, mitochondrial swelling and vacuolation were observed. At 18 months, M2AChR-el2-immunized rats manifested significant decreased cardiac systolic and diastolic function and pathological changes such as enlargement of right ventricular cavity and wall thinning; and the mitochondrial damage was aggravated. Furthermore, the M2 receptor maximum binding capacity (Bmax) of the M2AChR-el2-immunized rats significantly decreased, while the M2 receptor dissociation constant (Kd) was increased. Conclusions Our study suggested that long-term stimulation with M2-AA leaded to the ventricular dilatation and gradual deterioration of cardiac dysfunction. Mitochondrial damage and the down-regulation of M2 receptor density and affinity may be involved in the process.
Collapse
Affiliation(s)
- Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing, 100069, P. R. China
| | - Zhongmei He
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, P. R. China
| | - Jin Wang
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, P. R. China
| | - Li Wang
- Department of Pathology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, P. R. China
| | - Ye Wu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
- The Key Laboratory of Remodeling-related Cardiovascular Diseases, Capital Medical University, Ministry of Education, Beijing, 100069, P. R. China
| | - Jie Wang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, P. R. China
| | - Tingting Lv
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, P. R. China
- Department of Physiology, School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, 030001, P. R. China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Diseases, Capital Medical University, Beijing, 100069, P. R. China
- * E-mail:
| |
Collapse
|
24
|
Hou Z, Zhang Y, Deng K, Chen Y, Li X, Deng X, Cheng Z, Lian H, Li C, Lin J. UV-emitting upconversion-based TiO2 photosensitizing nanoplatform: near-infrared light mediated in vivo photodynamic therapy via mitochondria-involved apoptosis pathway. ACS NANO 2015; 9:2584-99. [PMID: 25692960 DOI: 10.1021/nn506107c] [Citation(s) in RCA: 361] [Impact Index Per Article: 36.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Photodynamic therapy (PDT) is a promising antitumor treatment that is based on the photosensitizers that inhibit cancer cells by yielding reactive oxygen species (ROS) after irradiation of light with specific wavelengths. As a potential photosensitizer, titanium dioxide (TiO2) exhibits minimal dark cytotoxicity and excellent ultraviolet (UV) light triggered cytotoxicity, but is challenged by the limited tissue penetration of UV light. Herein, a novel near-infrared (NIR) light activated photosensitizer for PDT based on TiO2-coated upconversion nanoparticle (UCNP) core/shell nanocomposites (UCNPs@TiO2 NCs) is designed. NaYF4:Yb(3+),Tm(3+)@NaGdF4:Yb(3+) core/shell UCNPs can efficiently convert NIR light to UV emission that matches well with the absorption of TiO2 shells. The UCNPs@TiO2 NCs endocytosed by cancer cells are able to generate intracellular ROS under NIR irradiation, decreasing the mitochondrial membrane potential to release cytochrome c into the cytosol and then activating caspase 3 to induce cancer cell apoptosis. NIR light triggered PDT of tumor-bearing mice with UCNPs@TiO2 as photosensitizers can suppress tumor growth efficiently due to the better tissue penetration than UV irradiation. On the basis of the evidence of in vitro and in vivo results, UCNPs@TiO2 NCs could serve as an effective photosensitizer for NIR light mediated PDT in antitumor therapy.
Collapse
Affiliation(s)
- Zhiyao Hou
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Yuanxin Zhang
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Kerong Deng
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
- ‡University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Yinyin Chen
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
- ‡University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Xuejiao Li
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Xiaoran Deng
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
- ‡University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Ziyong Cheng
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Hongzhou Lian
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Chunxia Li
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| | - Jun Lin
- †State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, People's Republic of China
| |
Collapse
|
25
|
Jiang L, Yuan F, Yin X, Dong J. Responses and adaptations of intervertebral disc cells to microenvironmental stress: a possible central role of autophagy in the adaptive mechanism. Connect Tissue Res 2014; 55:311-21. [PMID: 25000273 DOI: 10.3109/03008207.2014.942419] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Intervertebral discs comprise the largest avascular cartilaginous organ in the body, and its nutrient condition can be impaired by degeneration, aging and even metabolic disease. The unique microenvironment brings special stresses to various disc cell types, including nucleus pulposus cells, notochordal cells, annulus fibrosus cells and endplate chondrocytes. These cells experience nutrient starvation, acidic stress, hypoxic stress, hyperglycemic stress, osmotic stress and mechanical stress. Understanding the detailed responses and complex adaptive mechanisms of disc cells to various stresses might provide some clues to guide therapy for disc degeneration. By reviewing the published literatures describing disc cells under different hostile microenvironments, we conclude that these cells exhibit different responses to microenvironmental stresses with different mechanisms. Moreover, the interaction and combination of these stresses create a complex environment that synergistically increase or decrease influences on disc cells, compared with the effects of a single stress. Interestingly, most of these stresses activate autophagy, a self-protective mechanism by which dysfunctional protein and organelles are degraded. It is becoming clear that autophagy facilitates the cellular adaptation to stresses and might play a central role in regulating the adaptation of disc cells under stress. Therefore, autophagy modulation might be a potential therapeutic method to treat disc degeneration.
Collapse
Affiliation(s)
- Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University , Shanghai , China
| | | | | | | |
Collapse
|
26
|
Wauson EM, Dbouk HA, Ghosh AB, Cobb MH. G protein-coupled receptors and the regulation of autophagy. Trends Endocrinol Metab 2014; 25:274-82. [PMID: 24751357 PMCID: PMC4082244 DOI: 10.1016/j.tem.2014.03.006] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 01/06/2023]
Abstract
Autophagy is an important catabolic cellular process that eliminates damaged and unnecessary cytoplasmic proteins and organelles. Basal autophagy occurs during normal physiological conditions, but the activity of this process can be significantly altered in human diseases. Thus, defining the regulatory inputs and signals that control autophagy is essential. Nutrients are key modulators of autophagy. Although autophagy is generally accepted to be regulated in a cell-autonomous fashion, recent studies suggest that nutrients can modulate autophagy in a systemic manner by inducing the secretion of hormones and neurotransmitters that regulate G protein-coupled receptors (GPCRs). Emerging studies show that GPCRs also regulate autophagy by directly detecting extracellular nutrients. We review the role of GPCRs in autophagy regulation, highlighting their potential as therapeutic drug targets.
Collapse
Affiliation(s)
- Eric M Wauson
- Department of Physiology and Pharmacology, Des Moines University, Des Moines, IA 50312, USA.
| | - Hashem A Dbouk
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA
| | - Anwesha B Ghosh
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9041, USA.
| |
Collapse
|