1
|
Amirkhosravi L, Khaksari M, Sanjari M, Khorasani P. The nongenomic neuroprotective effects of estrogen, E2-BSA, and G1 following traumatic brain injury: PI3K/Akt and histopathological study. Horm Mol Biol Clin Investig 2024; 45:1-15. [PMID: 38507353 DOI: 10.1515/hmbci-2023-0066] [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: 09/19/2023] [Accepted: 02/28/2024] [Indexed: 03/22/2024]
Abstract
OBJECTIVES Studies suggest that both genomic and nongenomic pathways are involved in mediating the salutary effects of steroids following traumatic brain injury (TBI). This study investigated the nongenomic effects of 17β-estradiol (E2) mediated by the PI3K/p-Akt pathway after TBI. METHODS Ovariectomized rats were apportioned to E2, E2-BSA (E2 conjugated to bovine serum albumin), G1 [G-protein-coupled estrogen receptor agonist (GPER)] or their vehicle was injected following TBI, whereas ICI (classical estrogen receptor antagonist), G15 (GPER antagonist), ICI + G15, and their vehicles were injected before the induction of TBI and injection of drugs. Diffuse TBI was induced by the Marmarou model. Evans blue (EBC, 5 h), brain water contents (BWC), histopathological changes, and brain PI3K and p-Akt protein expressions were measured 24 h after TBI. The veterinary comma scale (VCS) was assessed before and at different times after TBI. RESULTS The results showed a reduction in BWC and EBC and increased VCS in the E2, E2-BSA, and G1 groups. Also, E2, E2-BSA, and G1 reduced brain edema, inflammation, and apoptosis. The ICI and G15 inhibited the beneficial effects of E2, E2-BSA, and G1 on these parameters. All drugs, following TBI, prevented the reduction of brain PI3K/p-Akt expression. The individual or combined use of ICI and G15 eliminated the beneficial effects of E2, E2-BSA, and G1 on PI3K/p-Akt expressions. CONCLUSIONS These findings indicated that PI3K/p-Akt pathway plays a critical role in mediating the salutary effects of estradiol on histopathological changes and neurological outcomes following TBI, suggesting that GPER and classic ERs are involved in regulating the expression of PI3K/p-Akt.
Collapse
Affiliation(s)
- Ladan Amirkhosravi
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Physiology Research Center, Institute of Neuropharmacology, 48463 Kerman University of Medical Sciences , Kerman, Iran
| | - Mojgan Sanjari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Parisa Khorasani
- Pathology and Stem Cell Research Center, 48463 Kerman University of Medical Sciences , Kerman, Iran
| |
Collapse
|
2
|
Choradia A, Bai K, Soni S, Nguyen N, Adhikari S, Rahul DK, Gupta R. Beyond hot flashes: Exploring the role of estrogen therapy in postmenopausal women for myocardial infarction prevention and recovery. BIOMOLECULES & BIOMEDICINE 2024; 24:4-13. [PMID: 37650466 PMCID: PMC10787608 DOI: 10.17305/bb.2023.9535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 09/01/2023]
Abstract
Myocardial infarction (MI) commonly known as "heart attack" results from the blockage of blood flow to the heart. Postmenopausal women face an elevated risk of MI due to declining estrogen levels, a hormone pivotal in maintaining cardiovascular health. It promotes vasodilation, reduce inflammation, and improves lipid profiles. While estrogen therapy shows promise in mitigating MI risk for postmenopausal woman, its efficacy in prevention and recovery remains a subject of debate. This review provides a critical assessment of existing evidence on estrogen therapy's cardioprotective effects for postmenopausal women. It delves into estrogen's role in vascular function enhancement, inflammation reduction, and lipid metabolism modulation. Additionally, it addresses the various forms of estrogen therapy, administration methods, dosage considerations, safety implications, and associated risks. The review highlights the existing controversies and knowledge gaps related to estrogen therapy for MI prevention. It underscores the urgency for in-depth research to decipher the nexus between estrogen therapy and MI risk, especially concerning primary prevention and specific postmenopausal subgroups. Future studies should investigate optimal formulations, doses, and administration routes of estrogen therapy as well as assess treatment timing and duration. Comparative studies and long-term follow-up are necessary to inform clinical decision-making and improve patient care. Addressing these research gaps will empower clinicians to make more judicious choices about estrogen therapy for MI prevention and recovery in postmenopausal women, aiming for enhanced patient outcomes.
Collapse
Affiliation(s)
| | | | - Suha Soni
- University of Texas Health Science Center School of Public Health, Texas, USA
| | | | | | | | | |
Collapse
|
3
|
Ran D, Yan W, Yanhong B, Hong W. Geniposide augments apoptosis in fibroblast-like synoviocytes by restoring hypoxia-enhanced JNK-BNIP3-mediated autophagy. Inflamm Res 2023; 72:1745-1760. [PMID: 37624402 DOI: 10.1007/s00011-023-01782-4] [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: 11/16/2022] [Revised: 07/06/2023] [Accepted: 08/07/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND As the main effector cells of chronic inflammation and hyperplasia of synovium, fibroblast-like synoviocytes (FLSs) show abnormal proliferation and insufficient apoptosis in the hypoxic microenvironment, which is due to the increase of BNIP3-mediated autophagy. This study aimed to explore the mechanism of geniposide (GE) on hypoxia-induced hyper-proliferative FLSs with a focus on autophagy and the JNK-BNIP3 pathway. METHODS The dynamic changes of autophagy, apoptosis, and hypoxia-related proteins in adjuvant arthritis (AA) rats were detected by immunohistochemistry and Western blot. The proliferation, autophagy, apoptosis, and mitochondrial state of FLSs were detected by CCK-8, flow cytometry, immunofluorescence, and transmission electron microscopy, respectively. Western blot, qRT-PCR, and co-immunoprecipitation were used to detect the expression of the JNK-BNIP3 pathway. RESULTS The excessive accumulation of BNIP3 in the synovium of AA rats was accompanied by inhibition of apoptosis and an increase in autophagy. GE inhibited the expression of BNIP3, enhanced apoptosis, decreased autophagy, and improved chronic inflammation and hyperplasia of synovium. The amount of autophagy under different oxygen concentrations was the key to mediating the different survival rates of FLSs, and the inhibition of autophagy triggered apoptosis. GE suppressed the proliferation of FLSs and down-regulated autophagy, leading to the accumulation of ROS and the decrease of mitochondrial membrane potential, induced the increase of apoptosis, and suppressed the accumulation of BNIP3 and the hyperphosphorylation of JNK. CONCLUSION GE inhibited autophagy by restoring the hypoxia-induced activated JNK-BNIP3 pathway, inducing mitochondrial oxidative damage, augmented apoptosis, and decreased survival rate of FLSs.
Collapse
Affiliation(s)
- Deng Ran
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China
- School of Integrated Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Wang Yan
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China
- Anhui Province Key Laboratory of Research &, Developmentof Chinese Medicine, Hefei, 230012, China
| | - Bu Yanhong
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China
- Anhui Province Key Laboratory of Research &, Developmentof Chinese Medicine, Hefei, 230012, China
| | - Wu Hong
- Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei, 230012, China.
- College of Pharmacy, Anhui University of Chinese Medicine, Qian Jiang Road 1, Hefei, 230012, China.
- Anhui Province Key Laboratory of Research &, Developmentof Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
4
|
Li J, Gong SH, He YL, Cao Y, Chen Y, Huang GH, Wang YF, Zhao M, Cheng X, Zhou YZ, Zhao T, Zhao YQ, Fan M, Wu HT, Zhu LL, Wu LY. Autophagy Is Essential for Neural Stem Cell Proliferation Promoted by Hypoxia. Stem Cells 2023; 41:77-92. [PMID: 36208284 DOI: 10.1093/stmcls/sxac076] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 09/06/2022] [Indexed: 02/02/2023]
Abstract
Hypoxia as a microenvironment or niche stimulates proliferation of neural stem cells (NSCs). However, the underlying mechanisms remain elusive. Autophagy is a protective mechanism by which recycled cellular components and energy are rapidly supplied to the cell under stress. Whether autophagy mediates the proliferation of NSCs under hypoxia and how hypoxia induces autophagy remain unclear. Here, we report that hypoxia facilitates embryonic NSC proliferation through HIF-1/mTORC1 signaling pathway-mediated autophagy. Initially, we found that hypoxia greatly induced autophagy in NSCs, while inhibition of autophagy severely impeded the proliferation of NSCs in hypoxia conditions. Next, we demonstrated that the hypoxia core regulator HIF-1 was necessary and sufficient for autophagy induction in NSCs. Considering that mTORC1 is a key switch that suppresses autophagy, we subsequently analyzed the effect of HIF-1 on mTORC1 activity. Our results showed that the mTORC1 activity was negatively regulated by HIF-1. Finally, we provided evidence that HIF-1 regulated mTORC1 activity via its downstream target gene BNIP3. The increased expression of BNIP3 under hypoxia enhanced autophagy activity and proliferation of NSCs, which was mediated by repressing the activity of mTORC1. We further illustrated that BNIP3 can interact with Rheb, a canonical activator of mTORC1. Thus, we suppose that the interaction of BNIP3 with Rheb reduces the regulation of Rheb toward mTORC1 activity, which relieves the suppression of mTORC1 on autophagy, thereby promoting the rapid proliferation of NSCs. Altogether, this study identified a new HIF-1/BNIP3-Rheb/mTORC1 signaling axis, which regulates the NSC proliferation under hypoxia through induction of autophagy.
Collapse
Affiliation(s)
- Jian Li
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Sheng-Hui Gong
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yun-Ling He
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yan Cao
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Ying Chen
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Guang-Hai Huang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yu-Fei Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China
| | - Ming Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Xiang Cheng
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yan-Zhao Zhou
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Tong Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Yong-Qi Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ming Fan
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| | - Ling-Ling Zhu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China.,Department of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China.,Department of Pharmacology, University of Nanhua, Hengyang, China
| | - Li-Ying Wu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing, People's Republic of China.,Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, People's Republic of China
| |
Collapse
|
5
|
Cho Y, Jeong YJ, Song KH, Chung IK, Magae J, Kwon TK, Choi YH, Kwak JY, Chang YC. 4-O-Methylascochlorin-Mediated BNIP-3 Expression Controls the Balance of Apoptosis and Autophagy in Cervical Carcinoma Cells. Int J Mol Sci 2022; 23:ijms232315138. [PMID: 36499465 PMCID: PMC9736141 DOI: 10.3390/ijms232315138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
4-O-methylascochlorin (MAC) is a 4-fourth carbon-substituted derivative of ascochlorin, a compound extracted from a phytopathogenic fungus Ascochyta viciae. MAC induces apoptosis and autophagy in various cancer cells, but the effects of MAC on apoptosis and autophagy in cervical cancer cells, as well as how the interaction between apoptosis and autophagy mediates the cellular anticancer effects are not known. Here, we investigated that MAC induced apoptotic cell death of cervical cancer cells without regulating the cell cycle and promoted autophagy by inhibiting the phosphorylation of serine-threonine kinase B (Akt), mammalian target of rapamycin (mTOR), and 70-kDa ribosomal protein S6 kinase (p70S6K). Additional investigations suggested that Bcl-2/adenovirus E1B 19 kDa protein-interacting protein 3 (BNIP-3), but not Hypoxia-inducible factor 1 alpha (HIF-1α), is a key regulator of MAC-induced apoptosis and autophagy. BNIP-3 siRNA suppressed MAC-induced increases in cleaved- poly (ADP-ribose) polymerase (PARP) and LC3II expression. The pan-caspase inhibitor Z-VAD-FMK suppressed MAC-induced cell death and enhanced MAC-induced autophagy. The autophagy inhibitor chloroquine (CQ) enhanced MAC-mediated cell death by increasing BNIP-3 expression. These results indicate that MAC induces apoptosis to promote cell death and stimulates autophagy to promote cell survival by increasing BNIP-3 expression. This study also showed that co-treatment of cells with MAC and CQ further enhanced the death of cervical cancer cells.
Collapse
Affiliation(s)
- Yuna Cho
- Research Institute of Biomedical Engineering and Department of Cell Biology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Yun-Jeong Jeong
- Research Institute of Biomedical Engineering and Department of Cell Biology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Kwon-Ho Song
- Research Institute of Biomedical Engineering and Department of Cell Biology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
| | - Il-Kyung Chung
- Department of Biotechnology, Catholic University of Daegu, Gyeongsan-Si 38430, Republic of Korea
| | - Junji Magae
- Magae Bioscience Institute, 49-4 Fujimidai, Tsukuba 300-1263, Japan
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Yung-Hyun Choi
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Jong-Young Kwak
- Department of Pharmacology, School of Medicine, Ajou University, Suwon 16499, Republic of Korea
| | - Young-Chae Chang
- Research Institute of Biomedical Engineering and Department of Cell Biology, School of Medicine, Catholic University of Daegu, Daegu 42472, Republic of Korea
- Correspondence:
| |
Collapse
|
6
|
The Role of Mitochondrial Quality Control in Anthracycline-Induced Cardiotoxicity: From Bench to Bedside. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3659278. [PMID: 36187332 PMCID: PMC9519345 DOI: 10.1155/2022/3659278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022]
Abstract
Cardiotoxicity is the major side effect of anthracyclines (doxorubicin, daunorubicin, epirubicin, and idarubicin), though being the most commonly used chemotherapy drugs and the mainstay of therapy in solid and hematological neoplasms. Advances in the field of cardio-oncology have expanded our understanding of the molecular mechanisms underlying anthracycline-induced cardiotoxicity (AIC). AIC has a complex pathogenesis that includes a variety of aspects such as oxidative stress, autophagy, and inflammation. Emerging evidence has strongly suggested that the loss of mitochondrial quality control (MQC) plays an important role in the progression of AIC. Mitochondria are vital organelles in the cardiomyocytes that serve as the key regulators of reactive oxygen species (ROS) production, energy metabolism, cell death, and calcium buffering. However, as mitochondria are susceptible to damage, the MQC system, including mitochondrial dynamics (fusion/fission), mitophagy, mitochondrial biogenesis, and mitochondrial protein quality control, appears to be crucial in maintaining mitochondrial homeostasis. In this review, we summarize current evidence on the role of MQC in the pathogenesis of AIC and highlight the therapeutic potential of restoring the cardiomyocyte MQC system in the prevention and intervention of AIC.
Collapse
|
7
|
Chen J, Liu Y, Pan D, Xu T, Luo Y, Wu W, Wu P, Zhu H, Li D. Estrogen inhibits endoplasmic reticulum stress and ameliorates myocardial ischemia/reperfusion injury in rats by upregulating SERCA2a. Cell Commun Signal 2022; 20:38. [PMID: 35331264 PMCID: PMC8944077 DOI: 10.1186/s12964-022-00842-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 02/07/2022] [Indexed: 11/10/2022] Open
Abstract
Background The incidence of coronary heart disease (CHD) in premenopausal women is significantly lower than that of men of the same age, suggesting protective roles of estrogen for the cardiovascular system against CHD. This study aimed to confirm the protective effect of estrogen on myocardium during myocardial ischemia/reperfusion (MI/R) injury and explore the underlying mechanisms. Methods Neonatal rat cardiomyocytes and Sprague–Dawley rats were used in this study. Different groups were treated by bilateral ovariectomy, 17β-estradiol (E2), adenoviral infection, or siRNA transfection. The expression of sarcoplasmic reticulum Ca2+ ATPase pump (SERCA2a) and endoplasmic reticulum (ER) stress-related proteins were measured in each group to examine the effect of different E2 levels and determine the relationship between SERCA2a and ER stress. The cell apoptosis, myocardial infarction size, levels of apoptosis and serum cardiac troponin I, ejection fraction, calcium transient, and morphology changes of the myocardium and ER were examined to verify the effects of E2 on the myocardium. Results Bilateral ovariectomy resulted in reduced SERCA2a levels and more severe MI/R injury. E2 treatment increased SERCA2a expression. Both E2 treatment and exogenous SERCA2a overexpression decreased levels of ER stress-related proteins and alleviated myocardial damage. In contrast, SERCA2a knockdown exacerbated ER stress and myocardial damage. Addition of E2 after SERCA2a knockdown did not effectively inhibit ER stress or reduce myocardial injury. Conclusions Our data demonstrate that estrogen inhibits ER stress and attenuates MI/R injury by upregulating SERCA2a. These results provide a new potential target for therapeutic intervention and drug discovery in CHD. Video Abstract
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-022-00842-2.
Collapse
Affiliation(s)
- Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yang Liu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Defeng Pan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Tongda Xu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Wanling Wu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Pei Wu
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Hong Zhu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Beikoghli Kalkhoran S, Kararigas G. Oestrogenic Regulation of Mitochondrial Dynamics. Int J Mol Sci 2022; 23:ijms23031118. [PMID: 35163044 PMCID: PMC8834780 DOI: 10.3390/ijms23031118] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/15/2022] [Accepted: 01/16/2022] [Indexed: 02/04/2023] Open
Abstract
Biological sex influences disease development and progression. The steroid hormone 17β-oestradiol (E2), along with its receptors, is expected to play a major role in the manifestation of sex differences. E2 exerts pleiotropic effects in a system-specific manner. Mitochondria are one of the central targets of E2, and their biogenesis and respiration are known to be modulated by E2. More recently, it has become apparent that E2 also regulates mitochondrial fusion–fission dynamics, thereby affecting cellular metabolism. The aim of this article is to discuss the regulatory pathways by which E2 orchestrates the activity of several components of mitochondrial dynamics in the cardiovascular and nervous systems in health and disease. We conclude that E2 regulates mitochondrial dynamics to maintain the mitochondrial network promoting mitochondrial fusion and attenuating mitochondrial fission in both the cardiovascular and nervous systems.
Collapse
|
9
|
Zhang S, Ou K, Huang J, Fang L, Wang C. In utero exposure to mixed PAHs causes heart mass reduction in adult male mice. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112804. [PMID: 34555720 DOI: 10.1016/j.ecoenv.2021.112804] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 06/13/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are a risk factor for the occurrence of cardiac diseases. The present study was conducted to investigate the influence of prenatal exposure to a mixed PAHs on heart and the underlying mechanism. Pregnant mice were orally administered with a mixture of 8 kinds of PAHs (0, 5, 50, 500 μg/kg body weight) once every 2 days for a total of 8 dosages. The mixed PAHs contained naphthalene, acenaphthylene, phenanthrene, fluoranthene, pyrene, benzo[a]pyrene, dibenzo[a,h]anthracene and benzo[g,h,i]perylene at a weight ratio of 10: 10: 10: 10: 10: 1: 1: 1. The adult males, not females, showed significantly decreased heart/body weight ratio, which was attributed to the loss of cardiac fiber and the increase of cell apoptosis. The protein expression of transforming growth factor β1 and its downstream transcription factors, Smad3 and Smad4, was significantly downregulated, which caused the loss of cardiac fiber. The downregulated phosphatidylinositol 3-kinase and AKT led to increased expression of caspase3, caspase9, BAX and reduced expression of Bcl-2, which was responsible for the increased cell apoptosis. Different levels of aromatic hydrocarbon receptor and sex hormone receptors between males and females were associated with the distinct effect on heart.
Collapse
Affiliation(s)
- Shenli Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jie Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
10
|
Zhang S, Ou K, Huang J, Fang L, Wang C, Wang Q. Prenatal EGCG exposure-induced heart mass reduction in adult male mice and underlying mechanisms. Food Chem Toxicol 2021; 157:112588. [PMID: 34600025 DOI: 10.1016/j.fct.2021.112588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), which is a major polyphenol in tea, has an unclear effect on cardiac development. In the present study, mice (C57BL/6) were exposed in utero to EGCG dissolved in drinking water (3 μg/ml) for 16 days. A significant decrease in the heart/body weight ratio was observed in adult males but not in adult females. The protein expression levels of TGF-β1 and its downstream transcription factors SMAD3 and SMAD4 were significantly decreased in male hearts. The PI3K/AKT signaling pathway was inhibited, the expression of proapoptotic proteins, such as BAX, Cleaved Caspase3 and Cleaved Caspase9, was elevated, and the level of antiapoptotic proteins, such as BCL-2, was decreased. A reduced heart/body weight ratio may be associated with the loss of cardiac fibers and an increase in myocardial apoptosis. The cardiac levels of aromatic hydrocarbon receptor and androgen receptor were elevated only in males, which may explain the sexual dimorphism in the effects. The promoter methylation levels of pik3r1, tgf-β, smad4 were elevated, and those of ahr were reduced, explaining the mechanism underlying the cardiac histological alteration caused by prenatal exposure to EGCG. The results suggest that ingestion of EGCG during pregnancy may be a risk factor for cardiac development in offspring.
Collapse
Affiliation(s)
- Shenli Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jie Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
11
|
Hajializadeh Z, Khaksari M. The protective effects of 17-β estradiol and SIRT1 against cardiac hypertrophy: a review. Heart Fail Rev 2021; 27:725-738. [PMID: 34537933 DOI: 10.1007/s10741-021-10171-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2021] [Indexed: 12/27/2022]
Abstract
One of the major causes of morbidity and mortality worldwide is cardiac hypertrophy (CH), which leads to heart failure. Sex differences in CH can be caused by sex hormones or their receptors. The incidence of CH increases in postmenopausal women due to the decrease in female sex hormone 17-β estradiol (E2) during menopause. E2 and its receptors inhibit CH in humans and animal models. Silent information regulator 1 (SIRT1) is a NAD+-dependent HDAC (histone deacetylase) and plays a major role in biological processes, such as inflammation, apoptosis, and oxidative stress responses. Probably SIRT1 because of these effects, is one of the main suppressors of CH and has a cardioprotective effect. On the other hand, estrogen and its agonists are highly efficient in modulating SIRT1 expression. In the present study, we review the protective effects of E2 and SIRT1 against CH.
Collapse
Affiliation(s)
- Zahra Hajializadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Khaksari
- Endocrinology and Metabolism Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
12
|
Kampaengsri T, Ponpuak M, Wattanapermpool J, Bupha-Intr T. Deficit of female sex hormones desensitizes rat cardiac mitophagy. CHINESE J PHYSIOL 2021; 64:72-79. [PMID: 33938817 DOI: 10.4103/cjp.cjp_102_20] [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: 11/04/2022] Open
Abstract
Long-term deprivation of female sex hormones has been shown to mediate accumulation of damaged mitochondria in ventricular muscle leading to cardiovascular dysfunction. Therefore, the roles of female sex hormones in mitochondrial quality control are closely focused. In the present study, depletion of female sex hormones impairing mitochondrial autophagy in the heart was hypothesized. Cardiac mitophagy was therefore investigated in the heart of 10-week ovariectomized (OVX) and sham-operated (SHAM) rats. By using isolated mitochondria preparation, results demonstrated an increase in mitochondrial PTEN-induced kinase 1 accumulation in the sample of OVX rats indicating mitochondrial outer membrane dysfunction. However, no change in p62 and LC3-II translocation to mitochondria was observed between two groups indicating unresponsiveness of mitophagosome formation in the OVX rat heart. This loss might be resulted from significant decreases in Parkin and Bcl2l13 expression, but not Bnip3 activation. In summary, results suggest that mitochondrial abnormality in the heart after deprivation of female sex hormones could consequently be due to desensitization of mitophagy process.
Collapse
Affiliation(s)
| | - Marisa Ponpuak
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | | | - Tepmanas Bupha-Intr
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
13
|
Yan J, Cao J, Chen Z. Mining prognostic markers of Asian hepatocellular carcinoma patients based on the apoptosis-related genes. BMC Cancer 2021; 21:175. [PMID: 33602168 PMCID: PMC7891020 DOI: 10.1186/s12885-021-07886-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Background Apoptosis-related genes(Args)play an essential role in the occurrence and progression of hepatocellular carcinoma(HCC). However, few studies have focused on the prognostic significance of Args in HCC. In the study, we aim to explore an efficient prognostic model of Asian HCC patients based on the Args. Methods We downloaded mRNA expression profiles and corresponding clinical data of Asian HCC patients from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. The Args were collected from Deathbase, a database related to cell death, combined with the research results of GeneCards、National Center for Biotechnology Information (NCBI) databases and a lot of literature. We used Wilcoxon-test and univariate Cox analysis to screen the differential expressed genes (DEGs) and the prognostic related genes (PRGs) of HCC. The intersection genes of DEGs and PGGs were seen as crucial Args of HCC. The prognostic model of Asian HCC patients was constructed by least absolute shrinkage and selection operator (lasso)- proportional hazards model (Cox) regression analysis. Kaplan-Meier curve, Principal Component Analysis (PCA) analysis, t-distributed Stochastic Neighbor Embedding (t-SNE) analysis, risk score curve, receiver operating characteristic (ROC) curve, and the HCC data of ICGC database and the data of Asian HCC patients of Kaplan-Meier plotter database were used to verify the model. Results A total of 20 of 56 Args were differentially expressed between HCC and adjacent normal tissues (p < 0.05). Univariate Cox regression analysis showed that 10 of 56 Args were associated with survival time and survival status of HCC patients (p < 0.05). There are seven overlapping genes of these 20 and 10 genes, including BAK1, BAX, BNIP3, CRADD, CSE1L, FAS, and SH3GLB1. Through Lasso-Cox analysis, an HCC prognostic model composed of BAK1, BNIP3, CSE1L, and FAS was constructed. Kaplan-Meier curve, PCA, t-SNE analysis, risk score curve, ROC curve, and secondary verification of ICGC database and Kaplan-Meier plotter database all support the reliability of the model. Conclusions Lasso-Cox regression analysis identified a 4-gene prognostic model, which integrates clinical and gene expression and has a good effect. The expression of Args is related to the prognosis of HCC patients, but the specific mechanism remains to be further verified. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-07886-6.
Collapse
Affiliation(s)
- Junbin Yan
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Jielu Cao
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China
| | - Zhiyun Chen
- The Second Central Laboratory, Key Lab of Integrative Chinese and Western medicine for the Diagnosis and Treatment of Circulatory Diseases of Zhejiang Province, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310006, China.
| |
Collapse
|
14
|
Ness H, Ljones K, Pinho M, Høydal M. Acute high-intensity aerobic exercise increases gene expression of calcium-related proteins and activates endoplasmic reticulum stress responses in diabetic hearts. COMPARATIVE EXERCISE PHYSIOLOGY 2021. [DOI: 10.3920/cep200022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Regular aerobic exercise training has a wide range of beneficial cardiac effects, but recent data also show that acute very strenuous aerobic exercise may impose a transient cardiac exhaustion. The aim of this study was to assess the response to acute high-intensity aerobic exercise on properties of mitochondrial respiration, cardiomyocyte contractile function, Ca2+ handling and transcriptional changes for key proteins facilitating Ca2+ handling and endoplasmic reticulum (ER) stress responses in type 2 diabetic mice. Diabetic mice were assigned to either sedentary control or an acute bout of exercise, consisting of a 10×4 minutes high-intensity interval treadmill run. Mitochondrial respiration, contractile and Ca2+ handling properties of cardiomyocytes were analysed 1 hour after completion of exercise. Gene expression levels of key Ca2+ handling and ER stress response proteins were measured in cardiac tissue samples harvested 1 hour and 24 hours after exercise. We found no significant changes in mitochondrial respiration, cardiomyocyte contractile function or Ca2+ handling 1 hour after the acute exercise. However, gene expression of Atp2a2, Slc8a1 and Ryr2, encoding proteins involved in cardiomyocyte Ca2+ handling, were all significantly upregulated 24 hours after the acute exercise bout. Acute exercise also altered gene expression of several key proteins in ER stress response and unfolded protein response, including Grp94, total Xbp1, Gadd34, and Atf6. The present results show that despite no significant alterations in functional properties of cardiomyocyte function, Ca2+ handling or mitochondrial respiration following one bout of high intensity aerobic exercise training, the expression of genes involved in Ca2+ handling and key components in ER stress and the unfolded protein response were changed. These transcriptional changes may constitute important steps in initiating adaptive remodelling to exercise training in type 2 diabetes.
Collapse
Affiliation(s)
- H.O. Ness
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - K. Ljones
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - M. Pinho
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| | - M.A. Høydal
- NTNU, Norwegian University of Technology and Science (NTNU), Faculty of Medicine and Health, Department of Circulation and Medical Imaging, Group of Molecular and Cellular Cardiology, Prinsesse Kristinas gate 9, Trondheim, 7489, Norway
| |
Collapse
|
15
|
Mycobacterium tuberculosis Rv0580c Impedes the Intracellular Survival of Recombinant Mycobacteria, Manipulates the Cytokines, and Induces ER Stress and Apoptosis in Host Macrophages via NF-κB and p38/JNK Signaling. Pathogens 2021; 10:pathogens10020143. [PMID: 33535567 PMCID: PMC7912736 DOI: 10.3390/pathogens10020143] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/11/2022] Open
Abstract
The Mycobacterium tuberculosis (M. tb) genome encodes a large number of hypothetical proteins, which need to investigate their role in physiology, virulence, pathogenesis, and host interaction. To explore the role of hypothetical protein Rv0580c, we constructed the recombinant Mycobacterium smegmatis (M. smegmatis) strain, which expressed the Rv0580c protein heterologously. We observed that Rv0580c expressing M. smegmatis strain (Ms_Rv0580c) altered the colony morphology and increased the cell wall permeability, leading to this recombinant strain becoming susceptible to acidic stress, oxidative stress, cell wall-perturbing stress, and multiple antibiotics. The intracellular survival of Ms_Rv0580c was reduced in THP-1 macrophages. Ms_Rv0580c up-regulated the IFN-γ expression via NF-κB and JNK signaling, and down-regulated IL-10 expression via NF-κB signaling in THP-1 macrophages as compared to control. Moreover, Ms_Rv0580c up-regulated the expression of HIF-1α and ER stress marker genes via the NF-κB/JNK axis and JNK/p38 axis, respectively, and boosted the mitochondria-independent apoptosis in macrophages, which might be lead to eliminate the intracellular bacilli. This study explores the crucial role of Rv0580c protein in the physiology and novel host-pathogen interactions of mycobacteria.
Collapse
|
16
|
Raw and salt-processed Achyranthes bidentata attenuate LPS-induced acute kidney injury by inhibiting ROS and apoptosis via an estrogen-like pathway. Biomed Pharmacother 2020; 129:110403. [PMID: 32574970 DOI: 10.1016/j.biopha.2020.110403] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Traditional Chinese medicine suggests that Radix Achyranthis Bidentatae nourishes and protects the kidneys, the effect of which is enhanced following a salt treatment. Raw and salt-processed Achyranthes bidentata are produced via different processing techniques from the same crude Achyranthes root. The anti-inflammatory and immunomodulatory properties of this plant have been verified earlier. However, there is a scarcity of experimental evidence for the renal-protective effects. AIM The purpose of present study is to compare the protective effects of raw and salt-processed Achyranthes on lipopolysaccharide (LPS) - induced acute kidney injury in mice and chemically characterize their extracts. METHOD The monomer components of raw and salt-processed Achyranthes extracts were analyzed using high performance liquid chromatography (HPLC). The aggregation and distribution of 2-Deoxy-D-glucose (2-DG) near infrared fluorescence probe in mice was examined with a small animal imaging systems. The pathological and morphological changes of kidneys were observed by H&E staining, and the serum urea nitrogen (BUN) and serum creatinine (Scr) levels were used to evaluate the renal function. The levels of cytokines in serum were detected by cytometric bead array. Flow cytometry assay was performed to assess the apoptosis and reactive oxygen species (ROS) in the kidney cells, and cell surface marker expression including CD45+, F4/80+, and Ly-6G+. The estrogenic activities of the raw and salt-processed Achyranthes were observed by uterine weight gain test in sexually immature mice. Western blot was used to detect the protein expression levels in the kidney. RESULTS Chemical analysis showed that the salt-processed Achyranthes contained more ginsenoside Ro and chikusetsusaponin Ⅳa than the raw Achyranthes, but there was no difference in the contents of β-ecdysterone, 25R-inokosterone, and 25S-inokosterone.in vivo near-infrared fluorescence imaging showed a significant reduced inflammation in the AKI mice. Histological studies showed that the raw and salt-processed Achyranthes markedly decreased the inflammatory infiltration, swelling and vacuolar degeneration in renal tissues and the Scr and BUN. Importantly, the raw and salt-processed Achyranthes extracts demonstrated different degrees of inhibition on the LPS-induced AKI, with salt-processed Achyranthes showing better inhibition. Results of flow cytometry showed a significant inhibition of IFN-γ, TNF-α, and IL-2, and promoted IL-10, along with reduced macrophages (CD45 + F4/80+), neutrophils (CD45+ Ly-6G+) and phagocytes. Furthermore, the extracts reduced the accumulation of ROS and apoptosis in the kidney, and also regulated the expression of apoptosis marker proteins TLR4, Bcl-2, Bax, cleaved caspase 3 and cleaved caspase 9 levels. Notably, they increased ERα, ERβ, and GPR30 in the renal tissues of AKI mice and LPS non-treated mice. In the subsequent experiments, it was found that the raw and salt-processed Achyranthes extracts increased the uterine coefficient in sexually immature mice, improved the LPS-induced decrease in NRK52e cell viability, and reduced the apoptosis, which could be antagonized by ICI182, 780 (estrogen receptor-unspecific antagonist, Faslodex). CONCLUSIONS The renal-protective effect of raw and salt-processed Achyranthes was exhibited through antiapoptotic and antioxidant mechanisms via an estrogen-like pathway, along with a modulation of the inflammatory response by regulating immune cells. Ginsenoside Ro and Chikusetsu saponin IVa were found to be the key factors to enhance the protective effect of salt-processed Achyranthes.
Collapse
|
17
|
Jia J, Yang X, Zhao Q, Ying F, Cai E, Sun S, He X. BNIP3 contributes to cisplatin-induced apoptosis in ovarian cancer cells. FEBS Open Bio 2020; 10:1463-1473. [PMID: 32412667 PMCID: PMC7396446 DOI: 10.1002/2211-5463.12881] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 04/15/2020] [Accepted: 05/11/2020] [Indexed: 12/11/2022] Open
Abstract
BNIP3 is a proapoptotic protein that mediates apoptosis, necrosis and autophagy. However, the involvement of BNIP3 in cisplatin‐induced apoptosis in ovarian cancer is not clear. In this study, we examined the role of BNIP3 in ovarian cancer during cisplatin treatment and its correlation with clinical outcomes. We first measured cisplatin cytotoxicity and BNIP3 levels before and after cisplatin exposure for ovarian cancer cell lines A2780, SKOV3, OVCAR4, OV2008, ES2 and HO8910. BNIP3 was observed to be differentially expressed in these cell lines, and cisplatin induced a significant increase in BNIP3 levels in A2780 and OVCAR4. BNIP3 knockdown with siRNA in A2780 and OVCAR4 significantly reduced cisplatin cytotoxicity in these two cell lines and alleviated cisplatin‐induced apoptosis. We searched the online databases Gene Expression Omnibus and The Cancer Genome Atlas to analyze the correlation between BNIP3 level and overall survival and progression‐free survival in patients with ovarian cancer. Pooled analyses showed that higher BNIP3 level was correlated with poorer overall survival (95% confidence intervals; hazard ratio = 1.18, 1.04–1.34; P = 0.013) and progression‐free survival (95% confidence intervals; hazard ratio = 1.26, 1.10–1.43; P = 0.00049). However, the results of individual datasets and stratification analyses of histology, FIGO (Federation Internationale de Gynecolgie et d’Obstetrique) stage, chemotherapy regimen and P53 mutation status varied. These findings indicate that cisplatin‐induced apoptosis is dependent on BNIP3 level in ovarian cancer cell lines. Targeting BNIP3 may therefore be a potential way of restoring cisplatin sensitivity.
Collapse
Affiliation(s)
- Jinghui Jia
- Department of Obstetrics and Gynecology, Air Force General Hospital, PLA, Beijing, China.,Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoxin Yang
- Department of Obstetrics and Gynecology, Tongren Hospital of WuHan University (Wuhan Third Hospital), Wuhan, China
| | - Qing Zhao
- Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China.,Department of Obstetrics and Gynecology, Tongji Medical College, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Feiquan Ying
- Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - E Cai
- Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Si Sun
- Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoqi He
- Department of Obstetrics and Gynecology, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Kondakova IV, Shashova EE, Sidenko EA, Astakhova TM, Zakharova LA, Sharova NP. Estrogen Receptors and Ubiquitin Proteasome System: Mutual Regulation. Biomolecules 2020; 10:biom10040500. [PMID: 32224970 PMCID: PMC7226411 DOI: 10.3390/biom10040500] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 03/21/2020] [Accepted: 03/25/2020] [Indexed: 12/11/2022] Open
Abstract
This review provides information on the structure of estrogen receptors (ERs), their localization and functions in mammalian cells. Additionally, the structure of proteasomes and mechanisms of protein ubiquitination and cleavage are described. According to the modern concept, the ubiquitin proteasome system (UPS) is involved in the regulation of the activity of ERs in several ways. First, UPS performs the ubiquitination of ERs with a change in their functional activity. Second, UPS degrades ERs and their transcriptional regulators. Third, UPS affects the expression of ER genes. In addition, the opportunity of the regulation of proteasome functioning by ERs—in particular, the expression of immune proteasomes—is discussed. Understanding the complex mechanisms underlying the regulation of ERs and proteasomes has great prospects for the development of new therapeutic agents that can make a significant contribution to the treatment of diseases associated with the impaired function of these biomolecules.
Collapse
Affiliation(s)
- Irina V. Kondakova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Elena E. Shashova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Evgenia A. Sidenko
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 5 Kooperativny Street, 634009 Tomsk, Russia; (I.V.K.); (E.E.S.); (E.A.S.)
| | - Tatiana M. Astakhova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
| | - Liudmila A. Zakharova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
| | - Natalia P. Sharova
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, 26 Vavilov Street, 119334 Moscow, Russia; (T.M.A.); (L.A.Z.)
- Correspondence: ; Tel.: +7-499-135-7674; Fax: +7-499-135-3322
| |
Collapse
|
19
|
Direct Peritoneal Resuscitation with Pyruvate Protects the Spinal Cord and Induces Autophagy via Regulating PHD2 in a Rat Model of Spinal Cord Ischemia-Reperfusion Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4909103. [PMID: 31998438 PMCID: PMC6969651 DOI: 10.1155/2020/4909103] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/17/2019] [Accepted: 12/03/2019] [Indexed: 01/18/2023]
Abstract
Direct peritoneal resuscitation with pyruvate (Pyr-PDS) has emerged as an interesting candidate to alleviate injury in diverse organs, while the potential mechanism has yet to be fully elucidated. To explore the effect of autophagy in the spinal cord ischemia-reperfusion (SCIR) injury and the underlying mechanism, we established a model of SCIR in vivo and in vitro. In vivo, male SD rats underwent aortic occlusion for 60 min and then followed by intraperitoneally infused with 20 mL of pyruvate or normal saline for 30 min, and the spinal cords were removed for analysis after 48 h of reperfusion. The functional and morphological results showed that Pyr-PDS alleviated SCIR injury; meanwhile, the expression of autophagy-related genes and transmission electron microscopy displayed autophagy was activated by SCIR injury, and Pyr-PDS treatment could further upregulate the degree of autophagy which plays a protective part in the SCIR injury, while there is no significant difference after treatment with saline. In addition, SCIR injury inhibited expression of PHD2, which results to activate its downstream HIF-1α/BNIP3 pathway to promote autophagy. In the Pyr-PDS, the results revealed PHD2 was further inhibited compared to the SCIR group, which could further activate the HIF-1α/BNIP3 signaling pathway. Additionally, oxygen-glucose deprivation and reoxygenation were applied to SH-SY5Y cells to mimic anoxic conditions in vitro, and the expression of autophagy-related genes, PHD2, and its downstream HIF-1α/BNIP3 pathway showed the same trend as the results in vivo. Besides, IOX2, a specific inhibitor of PHD2 was also treated to SH-SY5Y cells during reoxygenation, in which the result is as same as the pyruvate group. Then, we observed the expression of autophagy-related genes and the HIF-1α signal pathway in the process of reoxygenation; the results showed that as the reoxygenation goes, the expression of the HIF-1α signal pathway and degree of autophagy came to decrease gradually, while treated with pyruvate could maintain autophagy high and stable through keeping PHD2 at a lower level during reoxygenation, and the latter was observed downregulated during reoxygenation process from 0 to 24 hours in a time-effect way. The above results indicated that direct peritoneal resuscitation with pyruvate showed effective protection to ischemia-reperfusion of the spinal cord through activating autophagy via acting on PHD2 and its downstream HIF-1α/BNIP3 pathway.
Collapse
|
20
|
Wei Y, Huang J. Role of estrogen and its receptors mediated-autophagy in cell fate and human diseases. J Steroid Biochem Mol Biol 2019; 191:105380. [PMID: 31078693 DOI: 10.1016/j.jsbmb.2019.105380] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/11/2022]
Abstract
Studies have shown that morbidity of several diseases varies between males and females. This difference likely arises due to sex-related hormones. Estrogen, a primary female sex steroid hormone, plays a critical role in mediating many of the physiological functions like growth, differentiation, metabolism, and cell death. Recently, it has been demonstrated that estrogen mediates autophagy through its receptors (ERs) namely ERα, ERβ, and G-protein coupled estrogen receptor (GPER). However, the specific role of estrogen and its receptors mediated-autophagy in cell fate and human diseases such as cancers, cardiovascular disease and nervous system disease remains unclear. In this review, we comprehensively summarize the complex role of estrogen and its receptors-mediated autophagy in different cell lines and human diseases. In addition, we further discuss the key signaling molecules governing the role of ERs in autophagy. This review will serve as the basis for a proposed model of autophagy constituting a new frontier in estrogen-related human diseases. Here, we discuss the dual role of ERα in classical and non-classical autophagy through B-cell lymphoma 2 (BCL2)-associated athanogene 3 (BAG3). Next, we review the role of ERβ in pro-survival pathways through the promotion of autophagy under stress conditions. We further discuss activation of GPER via estrogen often mediates autophagy or mitophagy suppression, respectively. In summary, we believe that understanding the relationship between estrogen and its receptors mediated-autophagy on cell fate and human diseases will provide insightful knowledge for future therapeutic implications.
Collapse
Affiliation(s)
- Yong Wei
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China
| | - Jian Huang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, Hubei, PR China.
| |
Collapse
|
21
|
Mahmoodzadeh S, Dworatzek E. The Role of 17β-Estradiol and Estrogen Receptors in Regulation of Ca 2+ Channels and Mitochondrial Function in Cardiomyocytes. Front Endocrinol (Lausanne) 2019; 10:310. [PMID: 31156557 PMCID: PMC6529529 DOI: 10.3389/fendo.2019.00310] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 04/30/2019] [Indexed: 11/13/2022] Open
Abstract
Numerous epidemiological, clinical, and animal studies showed that cardiac function and manifestation of cardiovascular diseases (CVDs) are different between males and females. The underlying reasons for these sex differences are definitely multifactorial, but major evidence points to a causal role of the sex steroid hormone 17β-estradiol (E2) and its receptors (ER) in the physiology and pathophysiology of the heart. Interestingly, it has been shown that cardiac calcium (Ca2+) ion channels and mitochondrial function are regulated in a sex-specific manner. Accurate mitochondrial function and Ca2+ signaling are of utmost importance for adequate heart function and crucial to maintaining the cardiovascular health. Due to the highly sensitive nature of these processes in the heart, this review article highlights the current knowledge regarding sex dimorphisms in the heart implicating the importance of E2 and ERs in the regulation of cardiac mitochondrial function and Ca2+ ion channels, thus the contractility. In particular, we provide an overview of in-vitro and in-vivo studies using either E2 deficiency; ER deficiency or selective ER activation, which suggest that E2 and ERs are strongly involved in these processes. In this context, this review also discusses the divergent E2-responses resulting from the activation of different ER subtypes in these processes. Detailed understanding of the E2 and ER-mediated molecular and cellular mechanisms in the heart under physiological and pathological conditions may help to design more specifically targeted drugs for the management of CVDs in men and women.
Collapse
Affiliation(s)
- Shokoufeh Mahmoodzadeh
- Department of Molecular Muscle Physiology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- *Correspondence: Shokoufeh Mahmoodzadeh
| | - Elke Dworatzek
- Department of Molecular Muscle Physiology, Max-Delbrueck-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Institute of Gender in Medicine, Charité Universitaetsmedizin, Berlin, Germany
| |
Collapse
|
22
|
Damous LL, de Carvalho AETS, Nakamuta JS, Shiroma ME, Louzada ACS, Soares-Jr JM, Krieger JE, Baracat EC. Cell-free therapy with the secretome of adipose tissue-derived stem cells in rats' frozen-thawed ovarian grafts. Stem Cell Res Ther 2018; 9:323. [PMID: 30463630 PMCID: PMC6249760 DOI: 10.1186/s13287-018-1054-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 09/29/2018] [Accepted: 10/18/2018] [Indexed: 02/06/2023] Open
Abstract
The use of secretome may be a new strand of cell therapy, which is equal to or even superior to the injection of live cells, called cell-free therapy. In ovarian transplantation, this approach may be a therapeutic possibility for the ovarian graft in hypoxia. We designed the present study to evaluate whether the cell-free therapy with the secretome of adipose tissue-derived stem cells (ASCs) in rat frozen-thawed ovarian grafts could protect a graft against ischemic injury. A single dose of rat ASCs secretome or vehicle was injected into the bilateral frozen-thawed ovaries of 18 adult female rats immediately after an autologous transplant. Nine animals were used to control the cryopreservation protocol and were evaluated before and after the cryopreservation process. Daily vaginal smears were performed for estrous cycle evaluation until euthanasia on postoperative day 30. Follicle viability by trypan blue, graft morphology by HE, and apoptosis by TUNEL and cleaved-caspase-3 were assessed. No differences were found with respect to estrous cycle resumption and follicle viability (p > 0.05). However, compared with the vehicle-treated grafts, the morphology of the secretome-treated grafts was impaired, showing reduced follicular population and increased apoptosis (p < 0.05). ASC secretome impaired the rat frozen-thawed ovarian graft from ischemic injury. However, more studies are needed to evaluate the factors involved and the possibility of applying the secretome in scaffolds to optimize its use.
Collapse
Affiliation(s)
- Luciana Lamarão Damous
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil. .,Baturite St, 120. Ap 91., Aclimação, São Paulo, 01530-030, Brazil.
| | - Ana Elisa Teófilo Saturi de Carvalho
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Juliana Sanajotti Nakamuta
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Marcos Eiji Shiroma
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - Andressa Cristina Sposato Louzada
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - José Maria Soares-Jr
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), Faculdade de Medicina da Universidade de São Paulo, Dr Enéas de Carvalho Aguiar Av 44, 10th floor, Cerqueira Cesar, São Paulo, 05403-000, Brazil
| | - Edmund C Baracat
- Disciplina de Ginecologia, Laboratório de Biologia Estrutural e Molecular (LIM58), Faculdade de Medicina da Universidade de São Paulo, Dr Arnaldo av 455, 4nf floor, room 4119, Pacaembu, São Paulo, 01246-903, Brazil
| |
Collapse
|