1
|
Kim H, Massett MP. Effect of Spermidine on Endothelial Function in Systemic Lupus Erythematosus Mice. Int J Mol Sci 2024; 25:9920. [PMID: 39337408 PMCID: PMC11432455 DOI: 10.3390/ijms25189920] [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: 08/09/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Endothelial dysfunction is common in Systemic Lupus Erythematosus (SLE), even in the absence of cardiovascular disease. Evidence suggests that impaired mitophagy contributes to SLE. Mitochondrial dysfunction is also associated with impaired endothelial function. Spermidine, a natural polyamine, stimulates mitophagy by the PINK1-parkin pathway and counters age-associated endothelial dysfunction. However, the effect of spermidine on mitophagy and vascular function in SLE has not been explored. To address this gap, 9-week-old female lupus-prone (MRL/lpr) and healthy control (MRL/MpJ) mice were randomly assigned to spermidine treatment (lpr_Spermidine and MpJ_Spermidine) for 8 weeks or as control (lpr_Control and MpJ_Control). lpr_Control mice exhibited impaired endothelial function (e.g., decreased relaxation to acetylcholine), increased markers of inflammation, and lower protein content of parkin, a mitophagy marker, in the thoracic aorta. Spermidine treatment prevented endothelial dysfunction in MRL-lpr mice. Furthermore, aortas from lpr_Spermidine mice had lower levels of inflammatory markers and higher levels of parkin. Lupus phenotypes were not affected by spermidine. Collectively, these results demonstrate the beneficial effects of spermidine treatment on endothelial function, inflammation, and mitophagy in SLE mice. These results support future studies of the beneficial effects of spermidine on endothelial dysfunction and cardiovascular disease risk in SLE.
Collapse
Affiliation(s)
| | - Michael P. Massett
- Department of Kinesiology and Sport Management, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
2
|
Zhao P, Jiang WD, Wu P, Liu Y, Ren HM, Jin XW, Feng L, Zhou XQ. Dietary curcumin alleviates intestinal damage induced by ochratoxin A in juvenile grass carp ( Ctenopharyngodon idella): Necroptosis and inflammatory responses. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2024; 18:119-132. [PMID: 39263441 PMCID: PMC11388201 DOI: 10.1016/j.aninu.2024.04.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 11/24/2023] [Accepted: 04/11/2024] [Indexed: 09/13/2024]
Abstract
Ochratoxin A (OTA) is one of the most common pollutants in aquatic feed. As a first line of defense, intestinal barriers could be utilized against OTA in order to prevent disorders. Natural product supplementation is one of the most popular strategies to alleviate toxicity induced by mycotoxins, but there is a lack of knowledge about how it functions in the teleost intestine. In this study, 720 juvenile grass carp of about 11 g were selected and four treatment groups (control group, OTA group, curcumin [Cur] group, and OTA + Cur group) were set up to conduct a 60-day growth test. After the test, the growth performance and intestinal health related indexes of grass carp were investigated. The addition of dietary Cur could have the following main results: (1) inhibit absorption and promote efflux transporters mRNA expression, reducing the residuals of OTA, (2) decrease oxidative stress by reducing oxidative damage and enhancing the expression of antioxidant enzymes, (3) promote mitochondrial fusion proteins to inhibit the expression of mitotic proteins and mitochondrial autophagy proteins and enhance mitochondrial function, (4) reduce necroptosis-related gene expression through inhibiting the tumor necrotic factor receptor-interacting protein kinase/mixed lineage kinase domain-like pathway, (5) reduce the expression of pro-inflammatory factors by inhibiting the Toll-like receptor 4/nuclear factor-κB signaling pathway to alleviate the intestinal inflammatory response. In summary, the results suggested that Cur could alleviate OTA-induced intestinal damage by enhancing antioxidant capacity and mitochondrial function as well as reducing necroptosis and inflammation in the grass carp intestine. This study provided a theoretical basis and production implications for dietary Cur that could improve growth performance and alleviate the intestinal damage induced by OTA in fish.
Collapse
Affiliation(s)
- Piao Zhao
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
| | - Wei-Dan Jiang
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Pei Wu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Yang Liu
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Hong-Mei Ren
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Xiao-Wan Jin
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Lin Feng
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| | - Xiao-Qiu Zhou
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Fish Nutrition and Safety Production University Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 611130, Sichuan, China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Ministry of Agriculture and Rural Affairs, Key Laboratory of Sichuan Province, Sichuan, 611130, China
| |
Collapse
|
3
|
Yan J, Wang X, Fan Z, Xu Y, Zhang Y, Liu Y, Guo L, Liu D. Circ_0098823 binding with IGF2BP3 regulates DNM1L stability to promote metastasis of hepatocellular carcinoma via mitochondrial fission. Apoptosis 2024; 29:709-725. [PMID: 38459420 DOI: 10.1007/s10495-023-01903-8] [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] [Accepted: 10/15/2023] [Indexed: 03/10/2024]
Abstract
Hepatocellular carcinoma (HCC) is highly metastatic and invasive. CircRNA participates in gene regulation of multiple tumor metastases, but little is known whether it is a bystander or an actual player in HCC metastasis. We aim to explore the molecular mechanisms of novel circRNAs in HCC metastasis. RT-qPCR was used to detect the expression of 13 circRNAs derived by the ERBB3 gene. The function of circ_0098823 and DNM1L in HCC cells were estimated by CCK-8, transwell assays, flow cytometry, electron microscope, and in vivo experiments. RNA binding protein of circ_0098823 was confirmed by RNA pull-down, mass spectrometry, and RNA immunoprecipitation. The expression of DNM1L after IGF2BP3 knockdown was detected by RT-qPCR and western blot. Circ_0098823 was significantly up-regulated both in HCC tissues and HGF induced cell lines. Circ_0098823 overexpression significantly enhanced proliferation, migration, and invasion but decreased apoptosis of HCC cells, particularly promoted mitochondrial fission. Compared with the control group, the tumors in the circ_0098823 knockdown mice were significantly smaller and lighter. Circ_0098823 silencing suppressed DNM1L expression, a key molecule for fission, which enhanced proliferation, migration and invasion, and inhibited apoptosis of HCC cell. IGF2BP3 was a binding protein of circ_0098823. The expression and mRNA stability of DNM1L were down-regulated by IGF2BP3 knockdown. IGF2BP3 knockdown significantly alleviated the excessive migration, invasion and apoptosis of HCC cells caused by circ_0098823 overexpression. This study uncovered a novel circ_0098823 with tumor-promoting effect, and the mechanism by which circ_0098823 participates in HCC progression through IGF2BP3-guided DNM1L. Our study broadens molecular understanding of HCC progression.
Collapse
Affiliation(s)
- Jiuliang Yan
- Department of Pancreatic Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Xiaofeng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zongyu Fan
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yiqing Xu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85, Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Yingzi Zhang
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85, Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Yi Liu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85, Wujin Road, Hongkou District, Shanghai, 200080, China
| | - Lei Guo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Fudan University, Shanghai, 200032, China.
| | - Dongli Liu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No.85, Wujin Road, Hongkou District, Shanghai, 200080, China.
| |
Collapse
|
4
|
O'Brien KA, Gu W, Houck JA, Holzner LMW, Yung HW, Armstrong JL, Sowton AP, Baxter R, Darwin PM, Toledo-Jaldin L, Lazo-Vega L, Moreno-Aramayo AE, Miranda-Garrido V, Shortt JA, Matarazzo CJ, Yasini H, Burton GJ, Moore LG, Simonson TS, Murray AJ, Julian CG. Genomic Selection Signals in Andean Highlanders Reveal Adaptive Placental Metabolic Phenotypes That Are Disrupted in Preeclampsia. Hypertension 2024; 81:319-329. [PMID: 38018457 PMCID: PMC10841680 DOI: 10.1161/hypertensionaha.123.21748] [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: 06/28/2023] [Accepted: 10/24/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The chronic hypoxia of high-altitude residence poses challenges for tissue oxygen supply and metabolism. Exposure to high altitude during pregnancy increases the incidence of hypertensive disorders of pregnancy and fetal growth restriction and alters placental metabolism. High-altitude ancestry protects against altitude-associated fetal growth restriction, indicating hypoxia tolerance that is genetic in nature. Yet, not all babies are protected and placental pathologies associated with fetal growth restriction occur in some Andean highlanders. METHODS We examined placental metabolic function in 79 Andeans (18-45 years; 39 preeclamptic and 40 normotensive) living in La Paz, Bolivia (3600-4100 m) delivered by unlabored Cesarean section. Using a selection-nominated approach, we examined links between putatively adaptive genetic variation and phenotypes related to oxygen delivery or placental metabolism. RESULTS Mitochondrial oxidative capacity was associated with fetal oxygen delivery in normotensive but not preeclamptic placenta and was also suppressed in term preeclamptic pregnancy. Maternal haplotypes in or within 200 kb of selection-nominated genes were associated with lower placental mitochondrial respiratory capacity (PTPRD [protein tyrosine phosphatase receptor-δ]), lower maternal plasma erythropoietin (CPT2 [carnitine palmitoyl transferase 2], proopiomelanocortin, and DNMT3 [DNA methyltransferase 3]), and lower VEGF (vascular endothelial growth factor) in umbilical venous plasma (TBX5 [T-box transcription factor 5]). A fetal haplotype within 200 kb of CPT2 was associated with increased placental mitochondrial complex II capacity, placental nitrotyrosine, and GLUT4 (glucose transporter type 4) protein expression. CONCLUSIONS Our findings reveal novel associations between putatively adaptive gene regions and phenotypes linked to oxygen delivery and placental metabolic function in highland Andeans, suggesting that such effects may be of genetic origin. Our findings also demonstrate maladaptive metabolic mechanisms in the context of preeclampsia, including dysregulation of placental oxygen consumption.
Collapse
Affiliation(s)
- Katie A O'Brien
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Wanjun Gu
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
- Herbert Wertheim School of Public Health and Longevity Sciences (W.G.), University of California San Diego, La Jolla, CA
| | - Julie A Houck
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Lorenz M W Holzner
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Hong Wa Yung
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Jenna L Armstrong
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Alice P Sowton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Ruby Baxter
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Paula M Darwin
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lilian Toledo-Jaldin
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Litzi Lazo-Vega
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Any Elena Moreno-Aramayo
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Valquiria Miranda-Garrido
- Department of Obstetrics, Hospital Materno-Infantil, La Paz, Bolivia (L.T.-J., L.L.-V., A.E.M.-M., V.M.-G.)
| | - Jonathan A Shortt
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Christopher J Matarazzo
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Hussna Yasini
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| | - Graham J Burton
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Lorna G Moore
- Department of Obstetrics and Gynecology, Division of Reproductive Sciences (J.A.H., L.G.M.), University of Colorado School of Medicine, Aurora, CO
| | - Tatum S Simonson
- Department of Medicine, Division of Pulmonary, Critical Care, and Sleep Medicine (K.A.O., W.G., T.S.S.), University of California San Diego, La Jolla, CA
| | - Andrew J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, United Kingdom (K.A.O., L.M.W.H., H.W.Y., J.L.A., A.P.S., R.B., P.M.D., G.J.B., A.J.M.)
| | - Colleen G Julian
- Department of Biomedical Informatics (K.A.O., J.A.H., J.A.S., C.J.M., H.Y., C.G.J.), University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
5
|
Ding Y, Lv Z, Cao W, Shi W, He Q, Gao K. Phosphorylation of INF2 by AMPK promotes mitochondrial fission and oncogenic function in endometrial cancer. Cell Death Dis 2024; 15:65. [PMID: 38233384 PMCID: PMC10794193 DOI: 10.1038/s41419-024-06431-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 12/23/2023] [Accepted: 01/03/2024] [Indexed: 01/19/2024]
Abstract
Mitochondria are highly dynamic organelles capable of altering their sizes and shapes to maintain metabolic balance through coordinated fission and fusion processes. In various cancer types, mitochondrial hyperfragmentation has been frequently observed, contributing to the progression of cancer toward metastasis. Inverted formin 2 (INF2), which resides in the endoplasmic reticulum (ER), has been found to accelerate actin polymerization and drive mitochondrial fission. In this study, we demonstrate that INF2 expression is significantly upregulated in endometrial cancer (EC) and is associated with a poor prognosis in EC patients. INF2 promotes anchorage-dependent and independent EC cell growth in part by facilitating mitochondrial fission. Furthermore, in conditions of energy stress, AMP-activated protein kinase (AMPK) phosphorylates INF2 at Ser1077, leading to increased localization of INF2 to the ER and enhanced recruitment of the dynamin-related protein 1 (DRP1) to mitochondria. This AMPK-mediated phosphorylation of INF2 at Ser1077 facilitates mitochondrial division and promotes EC cell growth. Pathological examination using immunohistochemical analyses revealed a positive correlation between AMPK activity and phosphorylated INF2 (Ser1077) in EC specimens. Collectively, our findings uncover novel molecular mechanisms involving the AMPK-INF2 axis, which regulates mitochondrial dynamics and malignant cell growth in EC.
Collapse
Affiliation(s)
- Yan Ding
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Zeheng Lv
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenxin Cao
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Wenming Shi
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, 999077, China.
| | - Qizhi He
- Department of Pathology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, PR China.
| | - Kun Gao
- Department of Clinical Laboratory, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
- Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
6
|
Wang Y, Liu Y, Fang J, Xing X, Wang H, Shi X, Liu X, Niu T, Liu K. Small-molecule agonist AdipoRon alleviates diabetic retinopathy through the AdipoR1/AMPK/EGR4 pathway. J Transl Med 2024; 22:2. [PMID: 38166990 PMCID: PMC10759471 DOI: 10.1186/s12967-023-04783-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) is a progressive disease that involves multiple organs due to increased blood glucose, and diabetic retinopathy (DR) is the main complication of DM in the eyes and causes irreversible vision loss. In the pathogenesis of diabetic vascular disease, oxidative stress caused by hyperglycemia plays an important role in Müller cell impairment. In recent years, AdipoRon, an adiponectin analog that demonstrated important physiological functions in obesity, diabetes, inflammation, and cardiovascular diseases, demonstrated cellular protection from apoptosis and reduced inflammatory damage through a receptor-dependent mechanism. Here, we investigated how AdipoRon reduced oxidative stress and apoptosis in Müller glia in a high glucose environment. RESULTS By binding to adiponectin receptor 1 on Müller glia, AdipoRon activated 5' adenosine monophosphate-activated protein kinase (AMPK)/acetyl-CoA carboxylase phosphorylation downstream, thereby alleviating oxidative stress and eventual apoptosis of cells and tissues. Transcriptome sequencing revealed that AdipoRon promoted the synthesis and expression of early growth response factor 4 (EGR4) and inhibited the cellular protective effects of AdipoRon in a high-glucose environment by reducing the expression of EGR4. This indicated that AdipoRon played a protective role through the EGR4 and classical AMPK pathways. CONCLUSIONS This provides a new target for the early treatment of DR.
Collapse
Affiliation(s)
- Yihan Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
- Department of Ophthalmology, Shanghai Renji Hospital, School of Medicine, Shanghai, 200127, China
| | - Yujuan Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Junwei Fang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xindan Xing
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Hanying Wang
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xin Shi
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Xinyi Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China
| | - Tian Niu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China.
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- National Clinical Research Center for Eye Diseases, Shanghai, 200080, China.
- Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai, 200127, China.
- Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai, 200080, China.
- Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Disease, Shanghai, 200080, China.
| |
Collapse
|
7
|
Jobst M, Hossain M, Kiss E, Bergen J, Marko D, Del Favero G. Autophagy modulation changes mechano-chemical sensitivity of T24 bladder cancer cells. Biomed Pharmacother 2024; 170:115942. [PMID: 38042111 DOI: 10.1016/j.biopha.2023.115942] [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: 07/14/2023] [Revised: 10/27/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
Bladder cancer cells possess unique adaptive capabilities: shaped by their environment, cells face a complex chemical mixture of metabolites and xenobiotics accompanied by physiological mechanical cues. These responses might translate into resistance to chemotherapeutical regimens and can largely rely on autophagy. Considering molecules capable of rewiring tumor plasticity, compounds of natural origin promise to offer valuable options. Fungal derived metabolites, such as bafilomycin and wortmannin are widely acknowledged as autophagy inhibitors. Here, their potential to tune bladder cancer cells´ adaptability to chemical and physical stimuli was assessed. Additionally, dietary occurring mycotoxins were also investigated, namely deoxynivalenol (DON, 0.1-10 µM) and fusaric acid (FA, 0.1-1 mM). Endowing a Janus' face behavior, DON and FA are on the one side described as toxins with detrimental health effects. Concomitantly, they are also explored experimentally for selective pharmacological applications including anticancer activities. In non-cytotoxic concentrations, bafilomycin (BAFI, 1-10 nM) and wortmannin (WORT, 1 µM) modified cell morphology and reduced cancer cell migration. Application of shear stress and inhibition of mechano-gated PIEZO channels reduced cellular sensitivity to BAFI treatment (1 nM). Similarly, for FA (0.5 mM) PIEZO1 expression and inhibition largely aligned with the modulatory potential on cancer cells motility. Additionally, this study highlighted that the activity profile of compounds with similar cytotoxic potential (e.g. co-incubation DON with BAFI or FA with WORT) can diverge substantially in the regulation of cell mechanotransduction. Considering the interdependence between tumor progression and response to mechanical cues, these data promise to provide a novel viewpoint for the study of chemoresistance and associated pathways.
Collapse
Affiliation(s)
- Maximilian Jobst
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Maliha Hossain
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Endre Kiss
- Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Janice Bergen
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; University of Vienna, Vienna Doctoral School in Chemistry (DoSChem), Währinger Str. 42, 1090 Vienna, Austria
| | - Doris Marko
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria
| | - Giorgia Del Favero
- Department of Food Chemistry and Toxicology, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria; Core Facility Multimodal Imaging, University of Vienna Faculty of Chemistry, Währinger Str. 38-40, 1090 Vienna, Austria.
| |
Collapse
|
8
|
Abdellatif M, Rainer PP, Sedej S, Kroemer G. Hallmarks of cardiovascular ageing. Nat Rev Cardiol 2023; 20:754-777. [PMID: 37193857 DOI: 10.1038/s41569-023-00881-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2023] [Indexed: 05/18/2023]
Abstract
Normal circulatory function is a key determinant of disease-free life expectancy (healthspan). Indeed, pathologies affecting the cardiovascular system, which are growing in prevalence, are the leading cause of global morbidity, disability and mortality, whereas the maintenance of cardiovascular health is necessary to promote both organismal healthspan and lifespan. Therefore, cardiovascular ageing might precede or even underlie body-wide, age-related health deterioration. In this Review, we posit that eight molecular hallmarks are common denominators in cardiovascular ageing, namely disabled macroautophagy, loss of proteostasis, genomic instability (in particular, clonal haematopoiesis of indeterminate potential), epigenetic alterations, mitochondrial dysfunction, cell senescence, dysregulated neurohormonal signalling and inflammation. We also propose a hierarchical order that distinguishes primary (upstream) from antagonistic and integrative (downstream) hallmarks of cardiovascular ageing. Finally, we discuss how targeting each of the eight hallmarks might be therapeutically exploited to attenuate residual cardiovascular risk in older individuals.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria.
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- BioTechMed Graz, Graz, Austria.
| | - Peter P Rainer
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Guido Kroemer
- Centre de Recherche des Cordeliers, Equipe labellisée par la Ligue contre le cancer, Université de Paris, Sorbonne Université, Inserm U1138, Institut Universitaire de France, Paris, France.
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France.
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.
| |
Collapse
|
9
|
Tong W, Leng L, Wang Y, Guo J, Owusu FB, Zhang Y, Wang F, Li R, Li Y, Chang Y, Wang Y, Wang Q. Buyang huanwu decoction inhibits diabetes-accelerated atherosclerosis via reduction of AMPK-Drp1-mitochondrial fission axis. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116432. [PMID: 37003404 DOI: 10.1016/j.jep.2023.116432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/26/2023] [Accepted: 03/22/2023] [Indexed: 05/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional Chinese drugs, including Buyang Huanwu decoction (BYHWD), have been used in traditional practice to manage cardiovascular and cerebrovascular diseases. However, the effect and mechanisms by which this decoction alleviates diabetes-accelerated atherosclerosis are unknown and require exploration. AIM OF THE STUDY This study aims to investigate the pharmacological effects of BYHWD on preventing diabetes-accelerated atherosclerosis, and elucidate its underlying mechanism. MATERIALS AND METHODS Streptozotocin (STZ)-induced diabetic ApoE-/- mice were treated with BYHWD. Atherosclerotic aortic lesions, endothelial function, mitochondrial morphology, and mitochondrial dynamics-related proteins were evaluated in isolated aortas. High glucose-exposed human umbilical endothelial cells (HUVECs) were treated with BYHWD and its components. AMPK siRNA transfection, Drp1 molecular docking, Drp1 enzyme activity measurement, and so on were used to explore and verify the mechanism. RESULT BYHWD treatment inhibited the worsening of diabetes-accelerated atherosclerosis by lessening atherosclerotic lesions in diabetic ApoE-/- mice, by impeding endothelial dysfunction under diabetic conditions, and by inhibiting mitochondrial fragmentation by lowering protein expression levels of Drp1 and mitochondrial fission-1 protein (Fis1) in diabetic aortic endothelium. In high glucose-exposed HUVECs, BYHWD treatment also downgraded reactive oxygen species, promoted nitric oxide levels, and abated mitochondrial fission by reducing protein expression levels of Drp1 and fis1, but not mitofusin-1 and optic atrophy-1. Interestingly, we found that BYHWD's protective effect against mitochondrial fission is mediated by AMPK activation-dependent reduction of Drp1 levels. The main serum chemical components of BYHWD, ferulic acid, and calycosin-7-glucoside, can reduce the expression of Drp1 by regulating AMPK, and can inhibit the activity of GTPase of Drp1. CONCLUSION The above findings support the conclusion that BYHWD suppresses diabetes-accelerated atherosclerosis by reducing mitochondrial fission through modulation of the AMPK/Drp1 pathway.
Collapse
Affiliation(s)
- Wanyu Tong
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ling Leng
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yucheng Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jingwen Guo
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Felix Boahen Owusu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yue Zhang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Fang Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ruiqiao Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China
| | - Yanxu Chang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China
| | - Yuefei Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China; State Key Laboratory of Component-Based Chinese Medicine, Ministry of Education, Tianjin, 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
10
|
Tian L, Wu Y, Choi HJ, Sui X, Li X, Sofi MH, Kassir MF, Chen X, Mehrotra S, Ogretmen B, Yu XZ. S1P/S1PR1 signaling differentially regulates the allogeneic response of CD4 and CD8 T cells by modulating mitochondrial fission. Cell Mol Immunol 2022; 19:1235-1250. [PMID: 36071219 PMCID: PMC9622814 DOI: 10.1038/s41423-022-00921-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 08/15/2022] [Indexed: 01/27/2023] Open
Abstract
Graft-versus-host disease (GVHD) significantly contributes to patient morbidity and mortality after allogeneic hematopoietic cell transplantation (allo-HSCT). Sphingosine-1-phosphate (S1P) signaling is involved in the biogenetic processes of different immune cells. In the current study, we demonstrated that recipient sphingosine kinase 1 (Sphk1), but not Sphk2, was required for optimal S1PR1-dependent donor T-cell allogeneic responses by secreting S1P. Using genetic and pharmacologic approaches, we demonstrated that inhibition of Sphk1 or S1PR1 substantially attenuated acute GVHD (aGVHD) while retaining the graft-versus-leukemia (GVL) effect. At the cellular level, the Sphk1/S1P/S1PR1 pathway differentially modulated the alloreactivity of CD4+ and CD8+ T cells; it facilitated T-cell differentiation into Th1/Th17 cells but not Tregs and promoted CD4+ T-cell infiltration into GVHD target organs but was dispensable for the CTL activity of allogeneic CD8+ T cells. At the molecular level, the Sphk1/S1P/S1PR1 pathway augmented mitochondrial fission and increased mitochondrial mass in allogeneic CD4+ but not CD8+ T cells by activating the AMPK/AKT/mTOR/Drp1 pathway, providing a mechanistic basis for GVL maintenance when S1P signaling was inhibited. For translational purposes, we detected the regulatory efficacy of pharmacologic inhibitors of Sphk1 and S1PR1 in GVHD induced by human T cells in a xenograft model. Our study provides novel mechanistic insight into how the Sphk1/S1P/S1PR1 pathway modulates T-cell alloreactivity and validates Sphk1 or S1PR1 as a therapeutic target for the prevention of GVHD and leukemia relapse. This novel strategy may be readily translated into the clinic to benefit patients with hematologic malignancies and disorders.
Collapse
Affiliation(s)
- Linlu Tian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yongxia Wu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hee-Jin Choi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaohui Sui
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Xinlei Li
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Hanief Sofi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Mohamed Faisal Kassir
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiao Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
- The Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
11
|
Tian L, Wu Y, Choi HJ, Sui X, Li X, Sofi MH, Kassir MF, Chen X, Mehrotra S, Ogretmen B, Yu XZ. S1P/S1PR1 signaling differentially regulates the allogeneic response of CD4 and CD8 T cells by modulating mitochondrial fission. Cell Mol Immunol 2022. [PMID: 36071219 DOI: 10.1038/s41423-022-00921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023] Open
Abstract
Graft-versus-host disease (GVHD) significantly contributes to patient morbidity and mortality after allogeneic hematopoietic cell transplantation (allo-HSCT). Sphingosine-1-phosphate (S1P) signaling is involved in the biogenetic processes of different immune cells. In the current study, we demonstrated that recipient sphingosine kinase 1 (Sphk1), but not Sphk2, was required for optimal S1PR1-dependent donor T-cell allogeneic responses by secreting S1P. Using genetic and pharmacologic approaches, we demonstrated that inhibition of Sphk1 or S1PR1 substantially attenuated acute GVHD (aGVHD) while retaining the graft-versus-leukemia (GVL) effect. At the cellular level, the Sphk1/S1P/S1PR1 pathway differentially modulated the alloreactivity of CD4+ and CD8+ T cells; it facilitated T-cell differentiation into Th1/Th17 cells but not Tregs and promoted CD4+ T-cell infiltration into GVHD target organs but was dispensable for the CTL activity of allogeneic CD8+ T cells. At the molecular level, the Sphk1/S1P/S1PR1 pathway augmented mitochondrial fission and increased mitochondrial mass in allogeneic CD4+ but not CD8+ T cells by activating the AMPK/AKT/mTOR/Drp1 pathway, providing a mechanistic basis for GVL maintenance when S1P signaling was inhibited. For translational purposes, we detected the regulatory efficacy of pharmacologic inhibitors of Sphk1 and S1PR1 in GVHD induced by human T cells in a xenograft model. Our study provides novel mechanistic insight into how the Sphk1/S1P/S1PR1 pathway modulates T-cell alloreactivity and validates Sphk1 or S1PR1 as a therapeutic target for the prevention of GVHD and leukemia relapse. This novel strategy may be readily translated into the clinic to benefit patients with hematologic malignancies and disorders.
Collapse
Affiliation(s)
- Linlu Tian
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yongxia Wu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hee-Jin Choi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Xiaohui Sui
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Xinlei Li
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - M Hanief Sofi
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA
| | - Mohamed Faisal Kassir
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Xiao Chen
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Xue-Zhong Yu
- Department of Microbiology & Immunology, Medical University of South Carolina, Charleston, SC, USA.
- Department of Microbiology & Immunology, Medical College of Wisconsin, Milwaukee, WI, USA.
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA.
- The Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
12
|
NFKB1 Gene Mutant Was Associated with Prognosis of Coronary Artery Disease and Exacerbated Endothelial Mitochondrial Fission and Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9494926. [PMID: 36317060 PMCID: PMC9617727 DOI: 10.1155/2022/9494926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 08/11/2022] [Accepted: 09/21/2022] [Indexed: 11/18/2022]
Abstract
Endothelial apoptosis is the core pathological change in atherosclerotic cardiovascular disease, including coronary artery disease (CAD). Determining the molecular mechanisms underlying endothelial apoptosis is important. Nuclear factor kappa B (NF-κB) is a crucial transcription factor for controlling apoptosis. Our previous study demonstrated that the -94 ATTG ins/del mutant in the promoter of NFKB1 gene (rs28362491) is a risk factor for CAD. In the present study, we found that NFKB1 rs28362491 polymorphism was positively associated with increased major adverse cardiac and cerebrovascular events (MACCEs) in CAD patients. After adjusting for confounding factors including age, smoking, hypertension, glucose, and low-density lipoprotein cholesterol, the mutant DD genotype was an independent predictor of MACCEs (OR = 2.578, 95%CI = 1.64–4.05, P = 0.003). The in vitro study showed that mutant human umbilical vein endothelial cells (DD-mutant HUVECs) were more susceptible to high-glucose/palmitate-induced apoptosis, which was accompanied by decreased p50 expression and increased expression of cleaved caspase-3, Cytochrome c, and phospho-p65 (P < 0.05). The mitochondrial membrane potential was significantly lower, while increasing levels of mtROS and more opening of the mPTP were observed in DD-mutant HUVECs (P < 0.05). Furthermore, the percentage of cells with fragmented or spherical mitochondria was significantly higher in DD-mutant HUVECs than in wild-type cells (genotype II HUVECs) (P < 0.05). In addition, after stimulation with high glucose/palmitate, the NFKB1 gene mutant significantly increased the expression of Drp1, which indicated that the NFKB1 gene mutant affected the expression of mitochondrial morphology-related proteins, leading to excessive mitochondrial fission. In conclusion, the mutant DD genotype of the NFKB1 gene was an independent predictor of worse long-term prognosis for CAD patients. DD-mutant HUVECs exhibited abnormal activation of the NF-κB pathway and increased Drp1 expression, which caused excessive mitochondrial fission and dysfunction, ultimately leading to increased apoptosis.
Collapse
|
13
|
Xue R, Li S, Wei Z, Zhang Z, Cao Y. Melatonin attenuates di-(2-ethylhexyl) phthalate-induced apoptosis of human granulosa cells by inhibiting mitochondrial fission. Reprod Toxicol 2022; 113:18-29. [PMID: 35952901 DOI: 10.1016/j.reprotox.2022.08.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/19/2022] [Accepted: 08/06/2022] [Indexed: 10/15/2022]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) is one of the most used plasticizers which have contaminated environment widely, and its extensive use causes female reproductive injury. Melatonin has a substantial protective effect against female reproductive toxicity. This study was undertaken to investigate the influence of melatonin on DEHP-induced damage of human granulosa cells (GCs) in vitro and explore the potential mechanisms. Here, we found that melatonin treatment alleviated DEHP-induced human GCs apoptosis and improved mitochondrial function via inhibiting dynamin-related protein 1 (Drp1) mediated mitochondrial fission. Melatonin inhibited the expression, activation and oligomerization of Drp1, which decreased translocation of Drp1 to mitochondria in DEHP-exposed human GCs. Inhibition of mitochondrial fission reduced intracellular reactive oxygen species (ROS) production, sustained mitochondrial membrane potential and decreased cytochrome c release. Further research showed that AMPK-PGC-1α signal pathway was involved in the inhibition of melatonin on Drp1 expression and activation. Melatonin treatment promoted AMPK activation suppressed by DEHP, and activated AMPK recovered the balance of Drp1 phosphorylation at Ser616 and Ser637 sites and enhanced PGC-1α expression. Moreover, PGC-1α could prevent mitochondrial fission by decreasing Drp1 expression directly via binding to its promoter. In contrast, blocking of AMPK or PGC-1α with specific inhibitor negated the protective effects of melatonin on mitochondrial homeostasis and GCs apoptosis. In summary, our results indicated the protective effects of melatonin on improving mitochondrial function and attenuating cells injury in DEHP-exposed human GCs. Melatonin treatment may be a promising therapeutic approach against DEHP-induced reproductive disorder.
Collapse
Affiliation(s)
- Rufeng Xue
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China.
| | - Shuhang Li
- Department of Oncology of The First Affiliated Hospital, the CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui, China; Hefei National Laboratory for Physical Sciences at Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhaolian Wei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Reproductive Health and Genetics, Hefei, Anhui, China; Biopreservation and Artificial Organs, Anhui Provincial Engineering Research Center, Anhui Medical University, Hefei, Anhui, China
| | - Zhiguo Zhang
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China.
| | - Yunxia Cao
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China; NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract (Anhui Medical University), Hefei, Anhui, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, Hefei, Anhui, China.
| |
Collapse
|
14
|
Diao H, Wu K, Lan D, Wang D, Zhao J, Huang B, Shao X, Wang R, Tan H, Tang X, Yan M, Zhang Y. BAG3 Alleviates Atherosclerosis by Inhibiting Endothelial-to-Mesenchymal Transition via Autophagy Activation. Genes (Basel) 2022; 13:genes13081338. [PMID: 35893075 PMCID: PMC9332509 DOI: 10.3390/genes13081338] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 07/23/2022] [Accepted: 07/24/2022] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a chronic systemic inflammatory disease that causes severe cardiovascular events. B cell lymphoma 2-associated athanogene (BAG3) was proven to participate in the regulation of tumor angiogenesis, neurodegenerative diseases, and cardiac diseases, but its role in atherosclerosis remains unclear. Here, we aim to investigate the role of BAG3 in atherosclerosis and elucidate the potential molecular mechanism. In this study, ApoE-/- mice were given a tail-vein injection of BAG3-overexpressing lentivirus and fed a 12-week high-fat diet (HFD) to investigate the role of BAG3 in atherosclerosis. The overexpression of BAG3 reduced plaque areas and improved atherosclerosis in ApoE-/- mice. Our research proves that BAG3 promotes autophagy in vitro, contributing to the suppression of EndMT in human umbilical vein endothelial cells (HUVECs). Mechanically, autophagy activation is mediated by BAG3 via the interaction between BAG3 and its chaperones HSP70 and HSPB8. In conclusion, BAG3 facilitates autophagy activation via the formation of the chaperone-assisted selective autophagy (CASA) complex interacting with HSP70 and HSPB8, leading to the inhibition of EndMT during the progression of atherosclerosis and indicating that BAG3 is a potential therapeutic target for atherosclerosis.
Collapse
Affiliation(s)
- Hongtao Diao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Kaili Wu
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Dingming Lan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Dongwei Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Jingjing Zhao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Bingying Huang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Xiaoqi Shao
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Ruonan Wang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Huiling Tan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Xinyuan Tang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Meiling Yan
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
| | - Yue Zhang
- Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Guangdong TCM Key Laboratory for Metabolic Diseases, Guangdong Pharmaceutical University, Guangzhou 510006, China; (H.D.); (K.W.); (D.L.); (D.W.); (J.Z.); (B.H.); (X.S.); (R.W.); (H.T.); (X.T.); (M.Y.)
- Key Laboratory of Glucolipid Metabolic Disorder, Ministry of Education of China, Guangzhou 510006, China
- Correspondence:
| |
Collapse
|
15
|
Abdellatif M, Bugger H, Kroemer G, Sedej S. NAD + and Vascular Dysfunction: From Mechanisms to Therapeutic Opportunities. J Lipid Atheroscler 2022; 11:111-132. [PMID: 35656147 PMCID: PMC9133775 DOI: 10.12997/jla.2022.11.2.111] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 11/09/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is an essential and pleiotropic coenzyme involved not only in cellular energy metabolism, but also in cell signaling, epigenetic regulation, and post-translational protein modifications. Vascular disease risk factors are associated with aberrant NAD+ metabolism. Conversely, the therapeutic increase of NAD+ levels through the administration of NAD+ precursors or inhibitors of NAD+-consuming enzymes reduces chronic low-grade inflammation, reactivates autophagy and mitochondrial biogenesis, and enhances oxidative metabolism in vascular cells of humans and rodents with vascular pathologies. As such, NAD+ has emerged as a potential target for combatting age-related cardiovascular and cerebrovascular disorders. This review discusses NAD+-regulated mechanisms critical for vascular health and summarizes new advances in NAD+ research directly related to vascular aging and disease, including hypertension, atherosclerosis, coronary artery disease, and aortic aneurysms. Finally, we enumerate challenges and opportunities for NAD+ repletion therapy while anticipating the future of this exciting research field, which will have a major impact on vascular medicine.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- Department of Cardiology, Medical University of Graz, Graz, Austria
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
| | - Heiko Bugger
- Department of Cardiology, Medical University of Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Centre de Recherche des Cordeliers, Equipe Labellisée par la Ligue Contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Institut Universitaire de France, Paris, France
- Institut du Cancer Paris CARPEM, Department of Biology, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
- Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| |
Collapse
|
16
|
Mao RW, He SP, Lan JG, Zhu WZ. Honokiol ameliorates cisplatin-induced acute kidney injury via inhibition of mitochondrial fission. Br J Pharmacol 2022; 179:3886-3904. [PMID: 35297042 DOI: 10.1111/bph.15837] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 02/07/2022] [Accepted: 02/13/2022] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND PURPOSE Mitochondrial damage and oxidative stress are the crucial contributors to the tubular cell injury and death in acute kidney injury (AKI). Novel therapeutic strategies targeting mitochondria protection and halting the progression of AKI are urgently needed. Honokiol (HKL) is a small-molecule polyphenol that exhibits extraordinary cytoprotective effects, such as anti-inflammatory and anti-oxidative properties. Thus, we wonder whether HKL could ameliorate cisplatin-induced AKI via preventing mitochondrial dysfunction. EXPERIMENTAL APPROACH AKI was induced by cisplatin administration. Biochemical and histological analysis were applied to determine kidney injury. The effect of HKL on mitochondrial function and morphology were evaluated by immunohistochemistry, transmission electron microscopy, immunoblot and immunofluorescence. To investigate the mechanism of HKL in mitochondrial dynamics remodeling and resistance to apoptosis, we did transfection experiments, immunoblot, immunoprecipitation and flow cytometry assay. KEY RESULTS We demonstrated that the prominent mitochondrial fragmentation occurred in experimental models of cisplatin-induced nephrotoxicity, which was coupled with radical oxygen species (ROS) overproduction, deterioration of mitochondrial function, release of apoptogenic factors, and consequent apoptosis. HKL treatment exhibited notable renoprotection and attenuated these perturbations. Mechanically, we show that HKL treatment recovered the expression of SIRT3 and improved AMPK activity in tubular cells exposure to cisplatin, which preserved the Drp1 phosphorylation at Ser637 and blocked its translocation to mitochondria, consequently preventing mitochondrial fragmentation and subsequent cell injury and death. CONCLUSIONS AND IMPLICATIONS Our results indicate that HKL may protect against cisplatin-induced AKI by preserving mitochondrial integrity and fitness through a mechanism of SIRT3/AMPK-dependent mitochondrial dynamics remodeling.
Collapse
Affiliation(s)
- Rui-Wen Mao
- Department of Research Center for Molecular Metabolomics, Xiangya Hospital Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital Central South University, Changsha, China
| | - Shan-Ping He
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Jun-Gang Lan
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| | - Wu-Zheng Zhu
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Laboratory of Animal Nutrition and Human Health, College of Life Sciences, Hunan Normal University, Changsha, P.R. China
| |
Collapse
|
17
|
Koutouroushis C, Sarkar O. Role of Autophagy in Cardiovascular Disease and Aging. Cureus 2021; 13:e20042. [PMID: 34873555 PMCID: PMC8631374 DOI: 10.7759/cureus.20042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is expected to further increase as people continue to live even longer. Although the life span of the general population is increasing, the con of such a prolonged life span is that aging has certain detrimental effects on the molecular, structural, and functional elements of the cardiovascular system. This review will discuss various molecular pathways linked to longevity, most notably autophagy and its associated mechanisms, and how these pathways can be targeted to promote cardiovascular health through the process of aging. It is to be noted that the process of autophagy decreases with aging; hence, this review concludes that the promotion of autophagy, through implementation of caloric restriction, intermittent fasting, and pharmacologic agents, has proven to be an efficacious means of stimulating cardiovascular health. Therefore, autophagy is an important target for prevention and procrastination of cardiovascular pathologies in the geriatric population.
Collapse
Affiliation(s)
| | - Oiendrila Sarkar
- General Internal Medicine, St. Mary's Hospital, Isle of Wight NHS Trust, Newport, GBR
| |
Collapse
|
18
|
Li D, Yang S, Xing Y, Pan L, Zhao R, Zhao Y, Liu L, Wu M. Novel Insights and Current Evidence for Mechanisms of Atherosclerosis: Mitochondrial Dynamics as a Potential Therapeutic Target. Front Cell Dev Biol 2021; 9:673839. [PMID: 34307357 PMCID: PMC8293691 DOI: 10.3389/fcell.2021.673839] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular disease (CVD) is the main cause of death worldwide. Atherosclerosis is the underlying pathological basis of CVD. Mitochondrial homeostasis is maintained through the dynamic processes of fusion and fission. Mitochondria are involved in many cellular processes, such as steroid biosynthesis, calcium homeostasis, immune cell activation, redox signaling, apoptosis, and inflammation, among others. Under stress conditions, mitochondrial dynamics, mitochondrial cristae remodeling, and mitochondrial ROS (mitoROS) production increase, mitochondrial membrane potential (MMP) decreases, calcium homeostasis is imbalanced, and mitochondrial permeability transition pore open (mPTP) and release of mitochondrial DNA (mtDNA) are activated. mtDNA recognized by TLR9 can lead to NF-κB pathway activation and pro-inflammatory factor expression. At the same time, TLR9 can also activate NLRP3 inflammasomes and release interleukin, an event that eventually leads to tissue damage and inflammatory responses. In addition, mitochondrial dysfunction may amplify the activation of NLRP3 through the production of mitochondrial ROS, which together aggravate accumulating mitochondrial damage. In addition, mtDNA defects or gene mutation can lead to mitochondrial oxidative stress. Finally, obesity, diabetes, hypertension and aging are risk factors for the progression of CVD, which are closely related to mitochondrial dynamics. Mitochondrial dynamics may represent a new target in the treatment of atherosclerosis. Antioxidants, mitochondrial inhibitors, and various new therapies to correct mitochondrial dysfunction represent a few directions for future research on therapeutic intervention and amelioration of atherosclerosis.
Collapse
Affiliation(s)
- Dan Li
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shengjie Yang
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanwei Xing
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Limin Pan
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ran Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yixi Zhao
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Longtao Liu
- Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Guang'an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
19
|
Kong Q, Quan Y, Tian G, Zhou J, Liu X. Purinergic P2 Receptors: Novel Mediators of Mechanotransduction. Front Pharmacol 2021; 12:671809. [PMID: 34025431 PMCID: PMC8138185 DOI: 10.3389/fphar.2021.671809] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/26/2021] [Indexed: 02/05/2023] Open
Abstract
Mechanosensing and mechanotransduction are vital processes in mechanobiology and play critical roles in regulating cellular behavior and fate. There is increasing evidence that purinergic P2 receptors, members of the purinergic family, play a crucial role in cellular mechanotransduction. Thus, information on the specific mechanism of P2 receptor-mediated mechanotransduction would be valuable. In this review, we focus on purinergic P2 receptor signaling pathways and describe in detail the interaction of P2 receptors with other mechanosensitive molecules, including transient receptor potential channels, integrins, caveolae-associated proteins and hemichannels. In addition, we review the activation of purinergic P2 receptors and the role of various P2 receptors in the regulation of various pathophysiological processes induced by mechanical stimuli.
Collapse
Affiliation(s)
- Qihang Kong
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Quan
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Geer Tian
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junteng Zhou
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaojing Liu
- Laboratory of Cardiovascular Diseases, Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Autophagy and Mitophagy as Essential Components of Atherosclerosis. Cells 2021; 10:cells10020443. [PMID: 33669743 PMCID: PMC7922388 DOI: 10.3390/cells10020443] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) is one of the greatest health problems affecting people worldwide. Atherosclerosis, in turn, is one of the most common causes of cardiovascular disease. Due to the high mortality rate from cardiovascular diseases, prevention and treatment at the earliest stages become especially important. This requires developing a deep understanding of the mechanisms underlying the development of atherosclerosis. It is well-known that atherogenesis is a complex multi-component process that includes lipid metabolism disorders, inflammation, oxidative stress, autophagy disorders and mitochondrial dysfunction. Autophagy is a cellular control mechanism that is critical to maintaining health and survival. One of the specific forms of autophagy is mitophagy, which aims to control and remove defective mitochondria from the cell. Particularly defective mitophagy has been shown to be associated with atherogenesis. In this review, we consider the role of autophagy, focusing on a special type of it—mitophagy—in the context of its role in the development of atherosclerosis.
Collapse
|
21
|
Yan C, Tian X, Li J, Liu D, Ye D, Xie Z, Han Y, Zou MH. A High-Fat Diet Attenuates AMPK α1 in Adipocytes to Induce Exosome Shedding and Nonalcoholic Fatty Liver Development In Vivo. Diabetes 2021; 70:577-588. [PMID: 33262120 PMCID: PMC7881856 DOI: 10.2337/db20-0146] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 11/19/2020] [Indexed: 12/13/2022]
Abstract
Exosomes are important for intercellular communication, but the role of exosomes in the communication between adipose tissue (AT) and the liver remains unknown. The aim of this study is to determine the contribution of AT-derived exosomes in nonalcoholic fatty liver disease (NAFLD). Exosome components, liver fat content, and liver function were monitored in AT in mice fed a high-fat diet (HFD) or treated with metformin or GW4869 and with AMPKα1-floxed (Prkaα1 fl/fl/wild-type [WT]), Prkaα1 -/-, liver tissue-specific Prkaα1 -/-, or AT-specific Prkaα1 -/- modification. In cultured adipocytes and white AT, the absence of AMPKα1 increased exosome release and exosomal proteins by elevating tumor susceptibility gene 101 (TSG101)-mediated exosome biogenesis. In adipocytes treated with palmitic acid, TSG101 facilitated scavenger receptor class B (CD36) sorting into exosomes. CD36-containing exosomes were then endocytosed by hepatocytes to induce lipid accumulation and inflammation. Consistently, an HFD induced more severe lipid accumulation and cell death in Prkaα1 -/- and AT-specific Prkaα1 -/- mice than in WT and liver-specific Prkaα1 -/- mice. AMPK activation by metformin reduced adipocyte-mediated exosome release and mitigated fatty liver development in WT and liver-specific Prkaα1 -/- mice. Moreover, administration of the exosome inhibitor GW4869 blocked exosome secretion and alleviated HFD-induced fatty livers in Prkaα1 -/- and adipocyte-specific Prkaα1 -/- mice. We conclude that HFD-mediated AMPKα1 inhibition promotes NAFLD by increasing numbers of AT CD36-containing exosomes.
Collapse
Affiliation(s)
- Chenghui Yan
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Xiaoxiang Tian
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jiayin Li
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Dan Liu
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Ding Ye
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Zhonglin Xie
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| | - Yaling Han
- Cardiovascular Research Institute and Department of Cardiology, General Hospital of Northern Theater Command, Shenyang, China
| | - Ming-Hui Zou
- Center for Molecular and Translational Medicine, Georgia State University, Atlanta, GA
| |
Collapse
|
22
|
RSK2 protects human breast cancer cells under endoplasmic reticulum stress through activating AMPKα2-mediated autophagy. Oncogene 2020; 39:6704-6718. [PMID: 32958832 DOI: 10.1038/s41388-020-01447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/14/2020] [Accepted: 08/24/2020] [Indexed: 01/07/2023]
Abstract
Autophagy can protect stressed cancer cell by degradation of damaged proteins and organelles. However, the regulatory mechanisms behind this cellular process remain incompletely understood. Here, we demonstrate that RSK2 (p90 ribosomal S6 kinase 2) plays a critical role in ER stress-induced autophagy in breast cancer cells. We demonstrated that the promotive effect of RSK2 on autophagy resulted from directly binding of AMPKα2 in nucleus and phosphorylating it at Thr172 residue. IRE1α, an ER membrane-associated protein mediating unfolded protein response (UPR), is required for transducing the signal for activation of ERK1/2-RSK2 under ER stress. Suppression of autophagy by knockdown of RSK2 enhanced the sensitivity of breast cancer cells to ER stress both in vitro and in vivo. Furthermore, we demonstrated that inhibition of RSK2-mediated autophagy rendered breast cancer cells more sensitive to paclitaxel, a chemotherapeutic agent that induces ER stress-mediated cell death. This study identifies RSK2 as a novel controller of autophagy in tumor cells and suggests that targeting RSK2 can be exploited as an approach to reinforce the efficacy of ER stress-inducing agents against cancer.
Collapse
|
23
|
Zhu J, Cheng M, Zhao X. A tRNA-derived fragment (tRF-3001b) aggravates the development of nonalcoholic fatty liver disease by inhibiting autophagy. Life Sci 2020; 257:118125. [PMID: 32702444 DOI: 10.1016/j.lfs.2020.118125] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 07/16/2020] [Accepted: 07/16/2020] [Indexed: 02/07/2023]
Abstract
AIM Nonalcoholic fatty liver disease (NAFLD) is a growing health problem worldwide. Impaired autophagy has been linked to NAFLD pathogenesis. Whether transfer RNA (tRNA)-derived fragments (tRFs) regulate the progression of NAFLD via autophagy is not clear. Here, we aimed to identify autophagy- or adipogenesis-related tRFs and investigate their roles in NAFLD. METHODS Small RNA sequencing was performed on NAFLD and control mice, and candidate tRFs were validated using quantitative reverse transcription PCR (qRT-PCR). The role of a key tRF was investigated using Oil red O staining, western blotting, qRT-PCR and a luciferase reporter assay. KEY FINDINGS In NAFLD mice, the expression of p62 was increased and the ratio of LC3B-II/LC3-I was decreased compared to control mice. We identified nine differentially expressed tRFs, among which tRF-3001b was found to be significantly upregulated in NAFLD mice compared to the control liver tissues. Autophagy was decreased in FA (fatty acids)-induced LO2 cells, while silencing of tRF-3001b significantly abrogated the decrease in autophagy and increase in lipid formation. Moreover, chloroquine (CQ) dramatically abrogated the effect of tRF-3001b inhibition on lipid formation. Mechanistically, tRF-3001b targeted and inhibited the expression of the autophagy-related gene Prkaa1. In vivo, tRF-3001b silencing significantly improved pathology and decreased the levels of triglycerides and cholesterol in NAFLD mice, while CQ dramatically abrogated the effect of tRF-3001b deficiency. SIGNIFICANCE tRF-3001b may aggravate the development of NAFLD by inhibiting autophagy via targeting Prkaa1.
Collapse
Affiliation(s)
- Juanjuan Zhu
- Department of infectious disease, The Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang 550001, Guizhou, China
| | - Mingliang Cheng
- Department of infectious disease, The Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang 550001, Guizhou, China.
| | - Xueke Zhao
- Department of infectious disease, The Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang 550001, Guizhou, China
| |
Collapse
|
24
|
AMPK, Mitochondrial Function, and Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21144987. [PMID: 32679729 PMCID: PMC7404275 DOI: 10.3390/ijms21144987] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is in charge of numerous catabolic and anabolic signaling pathways to sustain appropriate intracellular adenosine triphosphate levels in response to energetic and/or cellular stress. In addition to its conventional roles as an intracellular energy switch or fuel gauge, emerging research has shown that AMPK is also a redox sensor and modulator, playing pivotal roles in maintaining cardiovascular processes and inhibiting disease progression. Pharmacological reagents, including statins, metformin, berberine, polyphenol, and resveratrol, all of which are widely used therapeutics for cardiovascular disorders, appear to deliver their protective/therapeutic effects partially via AMPK signaling modulation. The functions of AMPK during health and disease are far from clear. Accumulating studies have demonstrated crosstalk between AMPK and mitochondria, such as AMPK regulation of mitochondrial homeostasis and mitochondrial dysfunction causing abnormal AMPK activity. In this review, we begin with the description of AMPK structure and regulation, and then focus on the recent advances toward understanding how mitochondrial dysfunction controls AMPK and how AMPK, as a central mediator of the cellular response to energetic stress, maintains mitochondrial homeostasis. Finally, we systemically review how dysfunctional AMPK contributes to the initiation and progression of cardiovascular diseases via the impact on mitochondrial function.
Collapse
|
25
|
Lusini M, Nenna A, Chello C, Greco SM, Gagliardi I, Nappi F, Chello M. Role of autophagy in aneurysm and dissection of the ascending aorta. Future Cardiol 2020; 16:517-526. [PMID: 32524854 DOI: 10.2217/fca-2019-0076] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Maintenance of physiologically balanced levels of autophagy is crucial for cellular homeostasis and in the normal vessel wall, balanced autophagy can be considered a cytoprotective mechanism that preserves endothelial function and prevents cardiovascular disease. Recent studies pointed out the importance of the modulation of the autophagic flux in the pathogenesis of aortic dissection and aneurysms of the ascending aorta. Notably, shear stress (and its receptor p62), IL-6, Rab7 and Atg5/IRE1α pathways of autophagy may be considered the novel super-selective therapeutic target for the preventive and postoperative treatment of aortic aneurysm and aortic dissection. This review intends to summarize current evidences in this field trying to enlighten new avenues for future researches.
Collapse
Affiliation(s)
- Mario Lusini
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Antonio Nenna
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Camilla Chello
- Department of Dermatology, Università La Sapienza di Roma, Rome, Italy
| | | | - Ilaria Gagliardi
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Francesco Nappi
- Department of Cardiac Surgery, Centre Cardiologique du Nord, Paris, France
| | - Massimo Chello
- Department of Cardiovascular Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
26
|
Abdellatif M, Ljubojevic-Holzer S, Madeo F, Sedej S. Autophagy in cardiovascular health and disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 172:87-106. [PMID: 32620252 DOI: 10.1016/bs.pmbts.2020.04.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Autophagy is a cellular housekeeping and quality control mechanism that is essential for homeostasis and survival. By virtue of this role, any perturbations to the flow of this process in cardiac or vascular cells can elicit harmful effects on the cardiovascular system, and subsequently affect whole organismal health. In this chapter, we summarize the preclinical evidence supporting the role of autophagy in sustaining cardiovascular health during homeostasis and disease. Furthermore, we discuss how autophagy activation by dietary, genetic and pharmaceutical interventions can be exploited to counteract common cardiovascular disorders, including atherosclerosis, coronary artery disease, diabetic cardiomyopathy, arrhythmia, chemotherapy-induced cardiotoxicity and heart failure.
Collapse
Affiliation(s)
| | - Senka Ljubojevic-Holzer
- Department of Cardiology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria
| | - Frank Madeo
- BioTechMed Graz, Graz, Austria; Institute of Molecular Biosciences, NAWI Graz, University of Graz, Graz, Austria
| | - Simon Sedej
- Department of Cardiology, Medical University of Graz, Graz, Austria; BioTechMed Graz, Graz, Austria; Institute of Physiology, Faculty of Medicine, University of Maribor, Maribor, Slovenia.
| |
Collapse
|
27
|
Plewes MR, Hou X, Talbott HA, Zhang P, Wood JR, Cupp AS, Davis JS. Luteinizing hormone regulates the phosphorylation and localization of the mitochondrial effector dynamin-related protein-1 (DRP1) and steroidogenesis in the bovine corpus luteum. FASEB J 2020; 34:5299-5316. [PMID: 32077149 DOI: 10.1096/fj.201902958r] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/04/2020] [Accepted: 02/04/2020] [Indexed: 12/31/2022]
Abstract
The corpus luteum is an endocrine gland that synthesizes and secretes progesterone. Luteinizing hormone (LH) activates protein kinase A (PKA) signaling in luteal cells, increasing delivery of substrate to mitochondria for progesterone production. Mitochondria maintain a highly regulated equilibrium between fusion and fission in order to sustain biological function. Dynamin-related protein 1 (DRP1), is a key mediator of mitochondrial fission. The mechanism by which DRP1 is regulated in the ovary is largely unknown. We hypothesize that LH via PKA differentially regulates the phosphorylation of DRP1 on Ser616 and Ser637 in bovine luteal cells. In primary cultures of steroidogenic small luteal cells (SLCs), LH, and forskolin stimulated phosphorylation of DRP1 (Ser 637), and inhibited phosphorylation of DRP1 (Ser 616). Overexpression of a PKA inhibitor blocked the effects of LH and forskolin on DRP1 phosphorylation. In addition, LH decreased the association of DRP1 with the mitochondria. Genetic knockdown of the DRP1 mitochondria receptor, and a small molecule inhibitor of DRP1 increased basal and LH-induced progesterone production. Studies with a general Dynamin inhibitor and siRNA knockdown of DRP1 showed that DRP1 is required for optimal LH-induced progesterone biosynthesis. Taken together, the findings place DRP1 as an important target downstream of PKA in steroidogenic luteal cells.
Collapse
Affiliation(s)
- Michele R Plewes
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA.,Nebraska Western Iowa Veterans Health Care System, Omaha, NE, USA
| | - Xiaoying Hou
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Heather A Talbott
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Pan Zhang
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA
| | - Jennifer R Wood
- Department of Animal Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Andrea S Cupp
- Department of Animal Sciences, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - John S Davis
- Olson Center for Women's Health, Department of Obstetrics and Gynecology, University of Nebraska Medical Center, 983255 Nebraska Medical Center, Omaha, 68198-3255, NE, USA.,Nebraska Western Iowa Veterans Health Care System, Omaha, NE, USA
| |
Collapse
|
28
|
p38 MAPK-DRP1 signaling is involved in mitochondrial dysfunction and cell death in mutant A53T α-synuclein model of Parkinson's disease. Toxicol Appl Pharmacol 2020; 388:114874. [DOI: 10.1016/j.taap.2019.114874] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 12/18/2019] [Accepted: 12/23/2019] [Indexed: 01/05/2023]
|
29
|
Si L, Fu J, Liu W, Hayashi T, Mizuno K, Hattori S, Fujisaki H, Onodera S, Ikejima T. Silibinin-induced mitochondria fission leads to mitophagy, which attenuates silibinin-induced apoptosis in MCF-7 and MDA-MB-231 cells. Arch Biochem Biophys 2020; 685:108284. [PMID: 32014401 DOI: 10.1016/j.abb.2020.108284] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/19/2020] [Accepted: 01/27/2020] [Indexed: 02/08/2023]
Abstract
We reported previously that higher doses (150-250 μM) of silibinin enhanced fission and inhibited fusion of mitochondria, accompanying apoptosis of double-positive breast cancer cell line MCF-7 cells and triple-negative breast cancer cell line MDA-MB-231 cells. We report here three important questions yet unclarified in the previous study; 1) Whether enhanced fission of mitochondria by the treatment of silibinin leads to mitophagy, 2) Whether mitophagy positively contributes to apoptosis and 3) Whether estrogen receptor-positive (ER+) MCF-7 cells and estrogen receptor-negative (ER-) MDA-MB-231 cells are affected in a different way by silibinin treatment, since silibinin often works through ERs signaling pathway. Mitophagy driven by Pink1/Parkin signaling, plays an important role in eliminating damaged mitochondria. Indeed, increased expression of Pink1 and the recruitment of Parkin and LC3-II to mitochondria by the treatment with silibinin account for silibinin induction of mitophagy. In this study, the effects of mitochondrial division inhibitor 1 (mdivi-1) and small interfering RNA targeting dynamin-related protein 1 (DRP1) were examined to reveal the effect of mitochondrial fission on mitophagy. As expected, mdivi-1 or siRNA targeting DRP1 reversed silibinin-induced mitochondrial fission due to down-regulation in the expression of DRP1. Inhibition of mitochondrial fission by mdivi-1 prevented induction of mitophagy as well as autophagy in both MCF-7 and MDA-MB-231 cells, indicating that silibinin-induced mitochondrial fission leads to mitophagy. Inhibition of mitochondrial fission efficiently prevented silibinin-induced apoptosis in MCF-7 and MDA-MB-231 cells in our previous work, and the second point of the present study, inhibition of mitophagy by Pink1 or Parkin knockdown increased silibinin-induced apoptosis of these cells, respectively, suggesting that the mitophagy induced by silibinin treatment serves as a cytoprotective effect, resulting in reduction of apoptosis of cancer cells in both cells. In the third point, we studied whether estrogen receptors (ERs) played a role in silibinin-induced mitophagy and apoptosis in MCF-7 and MDA-MB-231 cells. ERα and ERβ are not involved in silibinin-induced mitophagic process in MCF-7 and MDA-MB-231 cells. These findings demonstrated that silibinin induced mitochondria fission leads to mitophagy, which attenuates silibinin-induced apoptosis not through ERs-Pink1 or -Parkin pathway in MCF-7 and MDA-MB-231.
Collapse
Affiliation(s)
- Lingling Si
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Jianing Fu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China
| | - Toshihiko Hayashi
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China; Department of Chemistry and Life Science, School of Advanced Engineering, Kogakuin University, 2665-1, Nakanomachi, Hachioji, Tokyo, 192-0015, Japan
| | - Kazunori Mizuno
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Toride, Ibaraki, 302-0017, Japan
| | - Satoshi Onodera
- Medical Research Institute of Curing Mibyo, 1-6-28 Narusedai Machida Tokyo, 194-0042, Japan
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China; Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, 110016, Liaoning, PR China.
| |
Collapse
|
30
|
Abstract
Cardiovascular diseases are the most prominent maladies in aging societies. Indeed, aging promotes the structural and functional declines of both the heart and the blood circulation system. In this review, we revise the contribution of known longevity pathways to cardiovascular health and delineate the possibilities to interfere with them. In particular, we evaluate autophagy, the intracellular catabolic recycling system associated with life- and health-span extension. We present genetic models, pharmacological interventions, and dietary strategies that block, reduce, or enhance autophagy upon age-related cardiovascular deterioration. Caloric restriction or caloric restriction mimetics like metformin, spermidine, and rapamycin (all of which trigger autophagy) are among the most promising cardioprotective interventions during aging. We conclude that autophagy is a fundamental process to ensure cardiac and vascular health during aging and outline its putative therapeutic importance.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.)
| | - Simon Sedej
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,BioTechMed Graz, Austria (S.S., D.C.-G., F.M.)
| | - Didac Carmona-Gutierrez
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Frank Madeo
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France (G.K.).,Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France (G.K.).,INSERM, U1138, Paris, France (G.K.).,Université Paris Descartes, Sorbonne Paris Cité, France (G.K.).,Université Pierre et Marie Curie, Paris, France (G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France (G.K.).,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (G.K.)
| |
Collapse
|
31
|
Pescatore LA, Gamarra LF, Liberman M. Multifaceted Mechanisms of Vascular Calcification in Aging. Arterioscler Thromb Vasc Biol 2019; 39:1307-1316. [DOI: https:/doi.org/10.1161/atvbaha.118.311576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 05/12/2019] [Indexed: 08/30/2023]
Abstract
Approximately 20% of the world’s population will be around or above 65 years of age by the next decade. Out of these, 40% are suspected to have cardiovascular diseases as a cause of mortality. Arteriosclerosis, characterized by increased vascular calcification, impairing Windkessel effect and tissue perfusion, and determining end-organ damage, is a hallmark of vascular pathology in the elderly population. Risk factors accumulated during aging affect the normal physiological and vascular aging process, which contributes to the progression of arteriosclerosis. Traditional risk factors, age-associated diseases, and respective regulating mechanisms influencing vascular calcification and vascular stiffness have been extensively studied for many years. Despite the well-known fact that aging alone can induce vascular damage, specific mechanisms that implicate physiological aging in vascular calcification, contributing to vascular stiffness, are poorly understood. This review focuses on mechanisms activated during normal aging, for example, cellular senescence, autophagy, extracellular vesicles secretion, and oxidative stress, along with the convergence of premature aging models’ pathophysiology, such as Hutchinson-Gilford Progeria (prelamin accumulation) and Klotho deficiency, to understand vascular calcification in aging. Understanding the mechanisms of vascular damage in aging that intersect with age-associated diseases and risk factors is crucial to foster innovative therapeutic targets to mitigate cardiovascular disease.
Visual Overview—
An online visual overview is available for this article.
Collapse
Affiliation(s)
- Luciana A. Pescatore
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
- Laboratório de Biologia Vascular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brazil (L.A.P.)
| | - Lionel F. Gamarra
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| | - Marcel Liberman
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| |
Collapse
|
32
|
Pescatore LA, Gamarra LF, Liberman M. Multifaceted Mechanisms of Vascular Calcification in Aging. Arterioscler Thromb Vasc Biol 2019; 39:1307-1316. [PMID: 31144990 DOI: 10.1161/atvbaha.118.311576] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Approximately 20% of the world's population will be around or above 65 years of age by the next decade. Out of these, 40% are suspected to have cardiovascular diseases as a cause of mortality. Arteriosclerosis, characterized by increased vascular calcification, impairing Windkessel effect and tissue perfusion, and determining end-organ damage, is a hallmark of vascular pathology in the elderly population. Risk factors accumulated during aging affect the normal physiological and vascular aging process, which contributes to the progression of arteriosclerosis. Traditional risk factors, age-associated diseases, and respective regulating mechanisms influencing vascular calcification and vascular stiffness have been extensively studied for many years. Despite the well-known fact that aging alone can induce vascular damage, specific mechanisms that implicate physiological aging in vascular calcification, contributing to vascular stiffness, are poorly understood. This review focuses on mechanisms activated during normal aging, for example, cellular senescence, autophagy, extracellular vesicles secretion, and oxidative stress, along with the convergence of premature aging models' pathophysiology, such as Hutchinson-Gilford Progeria (prelamin accumulation) and Klotho deficiency, to understand vascular calcification in aging. Understanding the mechanisms of vascular damage in aging that intersect with age-associated diseases and risk factors is crucial to foster innovative therapeutic targets to mitigate cardiovascular disease. Visual Overview- An online visual overview is available for this article.
Collapse
Affiliation(s)
- Luciana A Pescatore
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.).,Laboratório de Biologia Vascular, Instituto do Coração (InCor), Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, SP, Brazil (L.A.P.)
| | - Lionel F Gamarra
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| | - Marcel Liberman
- From the Hospital Israelita Albert Einstein, São Paulo, SP, Brazil (L.A.P., L.F.G., M.L.)
| |
Collapse
|
33
|
Wang YS, Yu P, Wang Y, Zhang J, Hang W, Yin ZX, Liu G, Chen J, Werle KD, Quan CS, Gao H, Zeng Q, Cui R, Liang J, Ding Q, Li YL, Xu ZX. AMP-activated protein kinase protects against necroptosis via regulation of Keap1-PGAM5 complex. Int J Cardiol 2018; 259:153-162. [PMID: 29579593 DOI: 10.1016/j.ijcard.2018.01.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/19/2017] [Accepted: 01/10/2018] [Indexed: 01/24/2023]
Abstract
BACKGROUND The AMP-activated protein kinase (AMPK) plays critical roles in growth regulation and metabolism reprogramming. AMPK activation protects cells against apoptosis from injury in different cell and animal models. However, its function in necroptosis remains largely unclear. METHODS AND RESULTS In the current study, we demonstrated that AMPK was activated upon necroptosis induction and protected mouse embryonic fibroblasts (MEFs) and cardiomyocytes from N-methyl-N'-nitro-N-nitrosoguanidine (MNNG) and reactive oxygen species (ROS) induced necroptosis. Activation of AMPK with chemicals A-769662, 2-deoxyglucose (2-DG), and metformin or constitutively active (CA) AMPK markedly decreased necroptosis and cytotoxicity induced by MNNG. In contrast, AMPK inhibitor compound C, dominant negative (DN) AMPK, as well as AMPK shRNAs increased necroptosis and cytotoxicity induced by MNNG. We further showed that AMPK physically associated with a protein complex containing PGAM5 and Keap1 whereby facilitating Keap1-mediated PGAM5 ubiquitination upon necroptosis induction. The AMPK agonist metformin ameliorated myocardial ischemia and reperfusion (IR) injury and reduced necroptosis through down-regulating the expression of PGAM5 in the Langendorff-perfused rat hearts. CONCLUSION Activation of AMPK protects against necroptosis via promoting Keap1-mediated PGAM5 degradation. Metformin may act as a valuable agent for the protection of myocardial ischemia and reperfusion injury by activating AMPK and reducing necroptosis.
Collapse
Affiliation(s)
- Yi-Shu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yong Wang
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Hang
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Otorhinolaryngology Head and Neck Surgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Zhi-Xian Yin
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Gang Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Jianfeng Chen
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kaitlin D Werle
- Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Cheng-Shi Quan
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Hang Gao
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Qinghua Zeng
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China
| | - Rutao Cui
- Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiyong Liang
- Department of Systems Biology, UT MD Anderson Cancer Center, TX, USA
| | - Qiang Ding
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Alabama Birmingham, Birmingham, AL, USA
| | - Yu-Lin Li
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China.
| | - Zhi-Xiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Norman Bethune College of Medicine, Jilin University, Changchun, China; Division of Hematology and Oncology, Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
34
|
Gao F, Chen J, Zhu H. A potential strategy for treating atherosclerosis: improving endothelial function via AMP-activated protein kinase. SCIENCE CHINA-LIFE SCIENCES 2018; 61:1024-1029. [PMID: 29675553 DOI: 10.1007/s11427-017-9285-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 03/12/2018] [Indexed: 12/11/2022]
Abstract
Endothelial dysfunction is caused by many factors, such as dyslipidemia, endoplasmic reticulum (ER) stress, and inflammation. It has been demonstrated that endothelial dysfunction is the initial process of atherosclerosis. AMP-activated protein kinase (AMPK) is an important metabolic switch that plays a crucial role in lipid metabolism and inflammation. However, recent evidence indicates that AMPK could be a target for atherosclerosis by improving endothelial function. For instance, activation of AMPK inhibits the production of reactive oxygen species induced by mitochondrial dysfunction, ER stress, and NADPH oxidase. Moreover, activation of AMPK inhibits the production of pro-inflammatory factors induced by dyslipidemia and hyperglycemia and restrains production of perivascular adipose tissue-released adipokines. AMPK activation prevents endothelial dysfunction by increasing the bioavailability of nitric oxide. Therefore, we focused on the primary risk factors involved in endothelial dysfunction, and summarize the features of AMPK in the protection of endothelial function, by providing signaling pathways thought to be important in the pathological progress of risk factors.
Collapse
Affiliation(s)
- Feng Gao
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiemei Chen
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Haibo Zhu
- State Key Laboratory for Bioactive Substances and Functions of Natural Medicines, Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
35
|
Antagonistic effects of lycopene on cadmium-induced hippocampal dysfunctions in autophagy, calcium homeostatis and redox. Oncotarget 2018; 8:44720-44731. [PMID: 28615536 PMCID: PMC5546513 DOI: 10.18632/oncotarget.18249] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 04/26/2017] [Indexed: 01/03/2023] Open
Abstract
Cadmium (Cd), a widely existed environmental contaminant, was shown to trigger neurotoxicity by regulating autophagy, ion homeostasis and redox. Lycopene (LYC) is a natural substance with potent antioxidant capacity. Nevertheless, little is known about i) the relationship of Cd-induced neurotoxicity and autophagy, ion homeostasis as well as redox in the hippocampus; ii) the role of LYC in the regulation of hippocampal autophagy, ionic balance and antioxidant capacity during Cd exposure. Therefore, this study sought to investigate the Cd exposure-induced hippocampal dysfunctions for neurotoxicity, and the preventive potential of LYC on the hippocampus impairment by reversing the dysfunctions during the exposure. In vivo study with mice model demonstrated that Cd exposure increased gene expression of a wide spectrum of autophagy-related gene (ATG) and gene regulating autophagy in hippocampus. This suggests the activation of hippocampal autophagy mediated by Cd. Cd exposure also decreased Ca2+-ATPase activity, thus increasing intracellular Ca2+ concentration in hippocampus, indicating the possibility that Cd-induced autophagy requires the Ca2+ signaling. Moreover, Cd exposure triggered redox stress in hippocampus cells, as antioxidant enzyme activities were decreased while oxidative productions were promoted. Cd exposure led to severe cytotoxicity in hippocampus cells. Of important note, all the hippocampal dysfunctions upon Cd exposure were reversed by LYC treatment to normal situations, and exposure-induced neurotoxicity was abrogated. The in vivo findings were recapitulated relevantly in the mouse hippocampal neuronal cell line, TH22. In all, the above data imply that LYC could be a potent therapeutic agent in treating Cd-triggered hippocampal dysfunctions and subsequent cell damage.
Collapse
|
36
|
Zhou H, Zhang Y, Hu S, Shi C, Zhu P, Ma Q, Jin Q, Cao F, Tian F, Chen Y. Melatonin protects cardiac microvasculature against ischemia/reperfusion injury via suppression of mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis. J Pineal Res 2017; 63:e12413. [PMID: 28398674 PMCID: PMC5518188 DOI: 10.1111/jpi.12413] [Citation(s) in RCA: 292] [Impact Index Per Article: 41.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/07/2017] [Indexed: 12/18/2022]
Abstract
The cardiac microvascular system, which is primarily composed of monolayer endothelial cells, is the site of blood supply and nutrient exchange to cardiomyocytes. However, microvascular ischemia/reperfusion injury (IRI) following percutaneous coronary intervention is a woefully neglected topic, and few strategies are available to reverse such pathologies. Here, we studied the effects of melatonin on microcirculation IRI and elucidated the underlying mechanism. Melatonin markedly reduced infarcted area, improved cardiac function, restored blood flow, and lower microcirculation perfusion defects. Histological analysis showed that cardiac microcirculation endothelial cells (CMEC) in melatonin-treated mice had an unbroken endothelial barrier, increased endothelial nitric oxide synthase expression, unobstructed lumen, reduced inflammatory cell infiltration, and less endothelial damage. In contrast, AMP-activated protein kinase α (AMPKα) deficiency abolished the beneficial effects of melatonin on microvasculature. In vitro, IRI activated dynamin-related protein 1 (Drp1)-dependent mitochondrial fission, which subsequently induced voltage-dependent anion channel 1 (VDAC1) oligomerization, hexokinase 2 (HK2) liberation, mitochondrial permeability transition pore (mPTP) opening, PINK1/Parkin upregulation, and ultimately mitophagy-mediated CMEC death. However, melatonin strengthened CMEC survival via activation of AMPKα, followed by p-Drp1S616 downregulation and p-Drp1S37 upregulation, which blunted Drp1-dependent mitochondrial fission. Suppression of mitochondrial fission by melatonin recovered VDAC1-HK2 interaction that prevented mPTP opening and PINK1/Parkin activation, eventually blocking mitophagy-mediated cellular death. In summary, this study confirmed that melatonin protects cardiac microvasculature against IRI. The underlying mechanism may be attributed to the inhibitory effects of melatonin on mitochondrial fission-VDAC1-HK2-mPTP-mitophagy axis via activation of AMPKα.
Collapse
Affiliation(s)
- Hao Zhou
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Ying Zhang
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Shunying Hu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Chen Shi
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing)Department of Radiation OncologyPeking University Cancer Hospital and InstituteBeijingChina
| | - Pingjun Zhu
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Qiang Ma
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Qinhua Jin
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Feng Cao
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Feng Tian
- Department of CardiologyChinese PLA General HospitalBeijingChina
| | - Yundai Chen
- Department of CardiologyChinese PLA General HospitalBeijingChina
| |
Collapse
|