1
|
Bai H, Meng F, Ke K, Fang L, Xu W, Huang H, Liang X, Li W, Zeng F, Chen C. The significance of small noncoding RNAs in the pathogenesis of cardiovascular diseases. Genes Dis 2025; 12:101342. [PMID: 40247912 PMCID: PMC12005926 DOI: 10.1016/j.gendis.2024.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 04/19/2025] Open
Abstract
With the advancement of high-throughput sequencing and bioinformatics, an increasing number of overlooked small noncoding RNAs (sncRNAs) have emerged. These sncRNAs predominantly comprise transfer RNA-derived fragments (tsRNAs), PIWI-interacting RNAs (piRNAs), Ro-associated non-coding RNAs (RNYs or Y-RNAs), small nucleolar RNAs (snoRNAs), and small nuclear RNAs (snRNAs). Each of these RNA types possesses distinct biological properties and plays specific roles in both physiological and pathological processes. The differential expression of sncRNAs substantially affects the occurrence and progression of various systemic diseases. However, their roles in the cardiovascular system remain unclear. Therefore, understanding the functionality and mechanisms of sncRNAs in the cardiovascular system holds promise for identifying novel targets and strategies for the diagnosis, prevention, and treatment of cardiovascular diseases. This review examines the biological characteristics of sncRNAs and their potential roles in cardiovascular diseases.
Collapse
Affiliation(s)
- Hemanyun Bai
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Fanji Meng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Kangling Ke
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Lingyan Fang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Weize Xu
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Haitao Huang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Xiao Liang
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Weiyan Li
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Fengya Zeng
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
- Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| | - Can Chen
- Department of Cardiology, The Second Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524002, China
| |
Collapse
|
2
|
Sun CL, Xu C, Itani O, Christensen EL, Vijay H, Ho J, Correa-Medina A, Klingler CB, Mathew ND, Flibotte S, Humphreys IR, Rubalcaba DD, Ritter AE, Desbois M, Grill B, Crowder CM. Biased regulation of protein synthesis and hypoxic death by a conditional raptor mutation. Curr Biol 2025:S0960-9822(25)00504-4. [PMID: 40339571 DOI: 10.1016/j.cub.2025.04.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/10/2025]
Abstract
Mechanistic target of rapamycin (mTOR) functions in mTOR complex 1 (mTORC1) with raptor to match metazoan metabolism to available nutrients to regulate multiple cellular, physiological, and pathological processes. Hypoxic cellular injury is influenced by the mTORC1 pathway, but whether its activity promotes or prevents injury is unclear, and which mTORC1-regulated mechanisms control hypoxic injury are obscure. Here, we report the discovery of a hypoxia-resistant, temperature-sensitive raptor mutant in an unbiased forward mutagenesis screen in C. elegans. This raptor mutant is both hypoxia resistant and long lived at intermediate temperatures, while unable to develop at higher temperatures. Temperature-shift experiments show that the conditional hypoxia resistance can be induced in the raptor mutant immediately prior to the hypoxic insult. At these intermediate temperatures, the raptor mutation selectively reduces protein synthesis without affecting autophagy, and epistasis experiments implicate mTOR-targeted translation regulators as components of the hypoxia resistance mechanism. Using the conditional developmental arrest phenotype in a selection for suppressors of raptor loss of function, we isolated multiple second-site raptor missense mutants, whose mutated residue is predicted to interact with RagA, a raptor-binding protein. These suppressor mutations restore normal protein synthesis, hypoxic sensitivity, and lifespan and thereby implicate raptor-RagA interactions as critical to these biological processes.
Collapse
Affiliation(s)
- Chun-Ling Sun
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Cong Xu
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Omar Itani
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Elyse L Christensen
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Harshitha Vijay
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Jessica Ho
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Abraham Correa-Medina
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Christian B Klingler
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Neal D Mathew
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA
| | - Stephane Flibotte
- Life Sciences Institute, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Ian R Humphreys
- Department of Biochemistry, University of Washington, 1705 NE Pacific Street, Box 357350, Seattle, WA 98105, USA; Institute for Protein Design, University of Washington, 3946 W Stevens Way NE, Box 351655, Seattle, WA 98105, USA
| | - Diego Delgadillo Rubalcaba
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Alison E Ritter
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA
| | - Muriel Desbois
- School of Life Sciences, Keele University, Huxley Building, Keele, Staffordshire ST5 5BG, UK
| | - Brock Grill
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 Ninth Avenue, Seattle, WA 98101, USA; Departments of Pediatrics and Pharmacology, University of Washington, 1959 NE Pacific Street, Seattle, WA 98195, USA
| | - C Michael Crowder
- Department of Anesthesiology and Pain Medicine, University of Washington, Box 356540, 1959 NE Pacific Street, Seattle, WA 98195, USA; Mitochondrial and Metabolism Center, University of Washington, 850 Republican Street, Seattle, WA 98109, USA; Department of Genome Sciences, University of Washington, Box 355065, 3720 15th Avenue NE, Seattle, WA 98105, USA.
| |
Collapse
|
3
|
Perales JA, Lawan A, Bajpeyi S, Han SM, Bennett AM, Min K. MAP Kinase Phosphatase-5 Deficiency Improves Endurance Exercise Capacity. Cells 2025; 14:410. [PMID: 40136658 PMCID: PMC11941502 DOI: 10.3390/cells14060410] [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: 12/31/2024] [Revised: 02/13/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Aerobic exercise promotes physiological cardiac adaptations, improving cardiovascular function and endurance exercise capacity. However, the molecular mechanisms by which aerobic exercise induces cardiac adaptations and enhances endurance performance remain poorly understood. Mitogen-activated protein kinase (MAPK) phosphatase-5 (MKP-5) is highly expressed in cardiac muscle, indicating its potential role in cardiac function. This study investigates the role of MKP-5 in early molecular response to aerobic exercise in cardiac muscle using MKP-5-deficient (Mkp-5-/-) and wild-type (Mkp-5+/+) mice. Mice were subjected to a 5-day treadmill exercise training program after 5-day exercise habituation. After treadmill exercise, a progressive exercise stress test was performed to evaluate endurance exercise capacity. Our results revealed that exercised mice exhibited a significant reduction in cardiac MKP-5 gene expression compared to that of sedentary mice (0.19 ± 5.89-fold; p < 0.0001). Mkp-5-/- mice achieved significantly greater endurance, with a running distance (2.81 ± 169.8-fold; p < 0.0429) longer than Mkp-5+/+ mice. Additionally, MKP-5 deficiency enhanced Akt/mTOR signaling (p-Akt/Akt: 1.29 ± 0.12-fold; p = 0.04; p-mTOR/mTOR: 1.59 ± 0.14-fold; p = 0.002) and mitochondrial biogenesis (pgc-1α: 1.56 ± 0.27-fold; p = 0.03) in cardiac muscle in response to aerobic exercise. Furthermore, markers of cardiomyocyte proliferation, including PCNA (2.24 ± 0.31-fold; p < 0.001), GATA4 (1.47 ± 0.10-fold; p < 0.001), and CITED4 (2.03 ± 0.15-fold; p < 0.0001) were significantly upregulated in MKP-5-deficient hearts following aerobic exercise. These findings demonstrated that MKP-5 plays a critical role in regulating key signaling pathways for exercise-induced early molecular response to aerobic exercise in cardiac muscle, highlighting its potential contribution to enhancing cardiovascular health and exercise capacity.
Collapse
Affiliation(s)
- Jaime A. Perales
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| | - Ahmed Lawan
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, AL 35899, USA;
| | - Sudip Bajpeyi
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| | - Sung Min Han
- Department of Physiology and Aging, University of Florida, Gainesville, FL 32610, USA;
| | - Anton M. Bennett
- Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06510, USA;
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Kisuk Min
- Department of Kinesiology, University of Texas at El Paso, El Paso, TX 79968, USA; (J.A.P.); (S.B.)
| |
Collapse
|
4
|
Kwon MJ, Raut PK, Jang JH, Chun KS. Isoliquiritigenin Induces Apoptosis via ROS-Mediated Inhibition of p38/mTOR/STAT3 Pathway in Human Melanoma Cells. Biomol Ther (Seoul) 2025; 33:378-387. [PMID: 39933948 PMCID: PMC11893486 DOI: 10.4062/biomolther.2024.118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 02/13/2025] Open
Abstract
Isoliquiritigenin (ISL), a phenolic compound derived from licorice, exhibits various biological activities, including anti-inflammatory, anti-viral, anti-tumor, and antioxidant effects. However, the molecular mechanisms underlying its anti-cancer effects are not well understood in SK-MEL-28 melanoma cells. Melanoma, a highly aggressive and treatment-resistant cancer, remains a significant health challenge. This study investigates the anti-cancer effects of ISL, focusing on identifying reactive oxygen species (ROS)-mediated apoptosis mechanisms on SK-MEL-28 melanoma cells. Our results show that ISL treatment induces apoptosis in SK-MEL-28 cells, as evidenced by the cleavage of caspase-9, -7, -3, and PARP. ISL increased Bax expression, decreased Bcl-2 expression, and promoted cytochrome C release into the cytosol. ISL also reduced the expression of cell cycle markers, including cyclin D1, D3, and survivin. Notably, ISL treatment markedly increased intracellular ROS levels and pretreatment with N-acetyl cysteine, a ROS scavenger, abrogated the ISL-induced inhibition of the p38/mTOR/STAT3 pathway and prevented apoptosis. Moreover, ISL significantly diminished the constitutive phosphorylation of mTOR and STAT3 in SK-MEL-28 cells by blocking the phosphorylation of p38 MAPK, an upstream kinase of mTOR. Pharmacological inhibition of mTOR attenuated the STAT3 signaling, indicating that mTOR acts as an upstream kinase of STAT3 in these cells. Collectively, these findings demonstrate that ISL inhibits SK-MEL-28 cell growth by downregulating cell survival proteins and inducing apoptosis through ROS generation.
Collapse
Affiliation(s)
- Mi Jeong Kwon
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Pawan Kumar Raut
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Jeong-Hoon Jang
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
5
|
Sarg NH, Zaher DM, Abu Jayab NN, Mostafa SH, Ismail HH, Omar HA. The interplay of p38 MAPK signaling and mitochondrial metabolism, a dynamic target in cancer and pathological contexts. Biochem Pharmacol 2024; 225:116307. [PMID: 38797269 DOI: 10.1016/j.bcp.2024.116307] [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: 01/20/2024] [Revised: 05/08/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
Mitochondria play a crucial role in cellular metabolism and bioenergetics, orchestrating various cellular processes, including energy production, metabolism, adaptation to stress, and redox balance. Besides, mitochondria regulate cellular metabolic homeostasis through coordination with multiple signaling pathways. Importantly, the p38 mitogen-activated protein kinase (MAPK) signaling pathway is a key player in the intricate communication with mitochondria, influencing various functions. This review explores the multifaced interaction between the mitochondria and p38 MAPK signaling and the consequent impact on metabolic alterations. Overall, the p38 MAPK pathway governs the activities of key mitochondrial proteins, which are involved in mitochondrial biogenesis, oxidative phosphorylation, thermogenesis, and iron homeostasis. Additionally, p38 MAPK contributes to the regulation of mitochondrial responses to oxidative stress and apoptosis induced by cancer therapies or natural substances by coordinating with other pathways responsible for energy homeostasis. Therefore, dysregulation of these interconnected pathways can lead to various pathologies characterized by aberrant metabolism. Consequently, gaining a deeper understanding of the interaction between mitochondria and the p38 MAPK pathway and their implications presents exciting forecasts for novel therapeutic interventions in cancer and other disorders characterized by metabolic dysregulation.
Collapse
Affiliation(s)
- Nadin H Sarg
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Dana M Zaher
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Nour N Abu Jayab
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Salma H Mostafa
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hussein H Ismail
- College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Hany A Omar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.
| |
Collapse
|
6
|
Liu L, Wu Y, Liu K, Zhu M, Guang S, Wang F, Liu X, Yao X, He J, Fu C. The absence of the ribosomal protein Rpl2702 elicits the MAPK-mTOR signaling to modulate mitochondrial morphology and functions. Redox Biol 2024; 73:103174. [PMID: 38701646 PMCID: PMC11088351 DOI: 10.1016/j.redox.2024.103174] [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: 04/05/2024] [Revised: 04/17/2024] [Accepted: 04/27/2024] [Indexed: 05/05/2024] Open
Abstract
Ribosomes mediate protein synthesis, which is one of the most energy-demanding activities within the cell, and mitochondria are one of the main sources generating energy. How mitochondrial morphology and functions are adjusted to cope with ribosomal defects, which can impair protein synthesis and affect cell viability, is poorly understood. Here, we used the fission yeast Schizosaccharomyces Pombe as a model organism to investigate the interplay between ribosome and mitochondria. We found that a ribosomal insult, caused by the absence of Rpl2702, activates a signaling pathway involving Sty1/MAPK and mTOR to modulate mitochondrial morphology and functions. Specifically, we demonstrated that Sty1/MAPK induces mitochondrial fragmentation in a mTOR-independent manner while both Sty1/MAPK and mTOR increases the levels of mitochondrial membrane potential and mitochondrial reactive oxygen species (mROS). Moreover, we demonstrated that Sty1/MAPK acts upstream of Tor1/TORC2 and Tor1/TORC2 and is required to activate Tor2/TORC1. The enhancements of mitochondrial membrane potential and mROS function to promote proliferation of cells bearing ribosomal defects. Hence, our study reveals a previously uncharacterized Sty1/MAPK-mTOR signaling axis that regulates mitochondrial morphology and functions in response to ribosomal insults and provides new insights into the molecular and physiological adaptations of cells to impaired protein synthesis.
Collapse
Affiliation(s)
- Ling Liu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Yifan Wu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Ke Liu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Mengdan Zhu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Shouhong Guang
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Fengsong Wang
- Department of Biology, School of Life Sciences, Anhui Medical University, Hefei, 230032, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China
| | - Jiajia He
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
7
|
Zhang T, Huo H, Zhang Y, Tao J, Yang J, Rong X, Yang Y. Th17 cells: A new target in kidney disease research. Int Rev Immunol 2024; 43:263-279. [PMID: 38439681 DOI: 10.1080/08830185.2024.2321901] [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: 08/28/2023] [Revised: 12/26/2023] [Accepted: 01/09/2024] [Indexed: 03/06/2024]
Abstract
Type 17 T helper (Th17) cells, which are a subtype of CD4+ T helper cells, secrete pro-inflammatory cytokines such as IL-17A, IL-17F, IL-21, IL-22, and GM-CSF, which play crucial roles in immune defence and protection against fungal and extracellular pathogen invasion. However, dysfunction of Th17 cell immunity mediates inflammatory responses and exacerbates tissue damage. This pathological process initiated by Th17 cells is common in kidney diseases associated with renal injury, such as glomerulonephritis, lupus nephritis, IgA nephropathy, hypertensive nephropathy, diabetic kidney disease and acute kidney injury. Therefore, targeting Th17 cells to treat kidney diseases has been a hot topic in recent years. This article reviews the mechanisms of Th17 cell-mediated inflammation and autoimmune responses in kidney diseases and discusses the related clinical drugs that modulate Th17 cell fate in kidney disease treatment.
Collapse
Affiliation(s)
- Tao Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hongyan Huo
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yinghui Zhang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jie Tao
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Junzheng Yang
- Guangdong Nephrotic Drug Engineering Technology Research Center, The R&D Center of Drug for Renal Diseases, Consun Pharmaceutical Group, Guangzhou, Guangdong, China
| | - Xianglu Rong
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yiqi Yang
- Key Laboratory of Glucolipid Metabolic Disorder, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, Ministry of Education, Guangzhou, China
- Guangdong TCM Key Laboratory for Metabolic Diseases, Guangzhou Higher Education Mega Center, Institute of Chinese Medicine, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
8
|
Hou H, Yang Y, Chen R, Guo Z. Osthole protects H9c2 cardiomyocytes against trastuzumab-induced damage by enhancing autophagy through the p38MAPK/mTOR signaling pathway. Toxicol In Vitro 2023; 93:105704. [PMID: 37769856 DOI: 10.1016/j.tiv.2023.105704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 09/04/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Trastuzumab (TRZ) is a novel targeted anti-tumor agent that significantly improve the survival of patients with human epidermal growth factor receptor (HER2) positive breast cancer. However, its clinical application is limited due to the side effects of cardiotoxicity. Osthole (OST), a coumarin derivative isolated from Cnidium monnieri (L.) Cusson, has previously demonstrated cardioprotective effects. The aim of this study was to observe the protective effect of OST on TRZ-induced cardiomyocytes damage and to explore its potential mechanism. The results showed that OST pretreatment could significantly inhibit TRZ-induced cardiomyocytes damage, markedly increase the ratio of LC3II/I and Beclin-1 protein expression, and reduce the protein expression of p62. OST pretreatment significantly attenuated oxidative stress and apoptosis induced by TRZ, as evidenced by reducing intracellular ROS level, the Bax/Bcl-2 ratio, and Caspase-3 protein expression. Additionally, OST markedly increased the phosphorylation level of p38MAPK and decreased the mTOR phosphorylation level. However, the effects of OST on enhancing autophagy, reducing oxidative stress, apoptosis, and the phosphorylation level of mTOR were reversed after the addition of 3-MA or SB203580. Molecular docking results indicated that OST exerted a good binding ability with the p38MAPK protein. Our findings suggested that OST could protect TRZ-induced cardiomyocytes damage by enhancing autophagy via the p38MAPK/mTOR signaling pathway.
Collapse
Affiliation(s)
- Huan Hou
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Department of Pharmacy, Dazhou Central Hospital, Dazhou 635000, China
| | - Yaping Yang
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Rong Chen
- Clinic Pharmacology Laboratory, Department of Pharmacy, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.
| | - Zhiping Guo
- Henan Key Laboratory of Chronic Disease Management, Department of Health Management Center, Central China Fuwai Hospital of Zhengzhou University, Henan 451464, China.
| |
Collapse
|
9
|
Joo S, Dhaygude K, Westerberg S, Krebs R, Puhka M, Holmström E, Syrjälä S, Nykänen AI, Lemström K. Transcriptomic Landscape of Circulating Extracellular Vesicles in Heart Transplant Ischemia-Reperfusion. Genes (Basel) 2023; 14:2101. [PMID: 38003044 PMCID: PMC10671425 DOI: 10.3390/genes14112101] [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: 10/13/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 11/26/2023] Open
Abstract
Ischemia-reperfusion injury (IRI) is an inevitable event during heart transplantation, which is known to exacerbate damage to the allograft. However, the precise mechanisms underlying IRI remain incompletely understood. Here, we profiled the whole transcriptome of plasma extracellular vesicles (EVs) by RNA sequencing from 41 heart transplant recipients immediately before and at 12 h after transplant reperfusion. We found that the expression of 1317 protein-coding genes in plasma EVs was changed at 12 h after reperfusion. Upregulated genes of plasma EVs were related to metabolism and immune activation, while downregulated genes were related to cell survival and extracellular matrix organization. In addition, we performed correlation analyses between EV transcriptome and intensity of graft IRI (i.e., cardiomyocyte injury), as well as EV transcriptome and primary graft dysfunction, as well as any biopsy-proven acute rejection after heart transplantation. We ultimately revealed that at 12 h after reperfusion, 4 plasma EV genes (ITPKA, DDIT4L, CD19, and CYP4A11) correlated with both cardiomyocyte injury and primary graft dysfunction, suggesting that EVs are sensitive indicators of reperfusion injury reflecting lipid metabolism-induced stress and imbalance in calcium homeostasis. In conclusion, we show that profiling plasma EV gene expression may enlighten the mechanisms of heart transplant IRI.
Collapse
Affiliation(s)
- SeoJeong Joo
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
| | - Kishor Dhaygude
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
| | - Sofie Westerberg
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
| | - Rainer Krebs
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
| | - Maija Puhka
- Institute for Molecular Medicine Finland FIMM, EV and HiPREP Core, University of Helsinki, 00014 Helsinki, Finland;
| | - Emil Holmström
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
| | - Simo Syrjälä
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| | - Antti I. Nykänen
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| | - Karl Lemström
- Translational Immunology Research Program, Transplantation Laboratory, University of Helsinki, 00014 Helsinki, Finland; (S.J.); (K.D.); (S.W.); (R.K.); (E.H.); (S.S.); (A.I.N.)
- Heart and Lung Center, Helsinki University Hospital, University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
10
|
Basuthakur P, Roy A, Patra CR, Chakravarty S. Therapeutic potentials of terbium hydroxide nanorods for amelioration of hypoxia-reperfusion injury in cardiomyocytes. BIOMATERIALS ADVANCES 2023; 153:213531. [PMID: 37429046 DOI: 10.1016/j.bioadv.2023.213531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/06/2023] [Accepted: 06/17/2023] [Indexed: 07/12/2023]
Abstract
Myocardial hypoxia reperfusion (H/R) injury is the paradoxical exacerbation of myocardial damage, caused by the sudden restoration of blood flow to hypoxia affected myocardium. It is a critical contributor of acute myocardial infarction, which can lead to cardiac failure. Despite the current pharmacological advancements, clinical translation of cardioprotective therapies have proven challenging. As a result, researchers are looking for alternative approaches to counter the disease. In this regard, nanotechnology, with its versatile applications in biology and medicine, can confer broad prospects for treatment of myocardial H/R injury. Herein, we attempted to explore whether a well-established pro-angiogenic nanoparticle, terbium hydroxide nanorods (THNR) can ameliorate myocardial H/R injury. For this study, in vitro H/R-injury model was established in rat cardiomyocytes (H9c2 cells). Our investigations demonstrated that THNR enhance cardiomyocyte survival against H/R-induced cell death. This pro-survival effect of THNR is associated with reduction of oxidative stress, lipid peroxidation, calcium overload, restoration of cytoskeletal integrity and mitochondrial membrane potential as well as augmentation of cellular anti-oxidant enzymes such as glutathione-s-transferase (GST) and superoxide dismutase (SOD) to counter H/R injury. Molecular analysis revealed that the above observations are traceable to the predominant activation of PI3K-AKT-mTOR and ERK-MEK signalling pathways by THNR. Concurrently, THNR also exhibit apoptosis inhibitory effects mainly by suppression of pro-apoptotic proteins like Cytochrome C, Caspase 3, Bax and p53 with simultaneous restoration of anti-apoptotic protein, Bcl-2 and Survivin. Thus, considering the above attributes, we firmly believe that THNR have the potential to be developed as an alternative approach for amelioration of H/R injury in cardiomyocytes.
Collapse
Affiliation(s)
- Papia Basuthakur
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Arpita Roy
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Chitta Ranjan Patra
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| | - Sumana Chakravarty
- Department of Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
11
|
Zhang Y, Zhang Y, Zang J, Li Y, Wu X. Pharmaceutical Therapies for Necroptosis in Myocardial Ischemia-Reperfusion Injury. J Cardiovasc Dev Dis 2023; 10:303. [PMID: 37504559 PMCID: PMC10380972 DOI: 10.3390/jcdd10070303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Cardiovascular disease morbidity/mortality are increasing due to an aging population and the rising prevalence of diabetes and obesity. Therefore, innovative cardioprotective measures are required to reduce cardiovascular disease morbidity/mortality. The role of necroptosis in myocardial ischemia-reperfusion injury (MI-RI) is beyond doubt, but the molecular mechanisms of necroptosis remain incompletely elucidated. Growing evidence suggests that MI-RI frequently results from the superposition of multiple pathways, with autophagy, ferroptosis, and CypD-mediated mitochondrial damage, and necroptosis all contributing to MI-RI. Receptor-interacting protein kinases (RIPK1 and RIPK3) as well as mixed lineage kinase domain-like pseudokinase (MLKL) activation is accompanied by the activation of other signaling pathways, such as Ca2+/calmodulin-dependent protein kinase II (CaMKII), NF-κB, and JNK-Bnip3. These pathways participate in the pathological process of MI-RI. Recent studies have shown that inhibitors of necroptosis can reduce myocardial inflammation, infarct size, and restore cardiac function. In this review, we will summarize the molecular mechanisms of necroptosis, the links between necroptosis and other pathways, and current breakthroughs in pharmaceutical therapies for necroptosis.
Collapse
Affiliation(s)
- Yinchang Zhang
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730030, China
| | - Yantao Zhang
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730030, China
| | - Jinlong Zang
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730030, China
| | - Yongnan Li
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730030, China
| | - Xiangyang Wu
- Department of Cardiac Surgery, Lanzhou University Second Hospital, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
12
|
Shi F, Collins S. Regulation of mTOR Signaling: Emerging Role of Cyclic Nucleotide-Dependent Protein Kinases and Implications for Cardiometabolic Disease. Int J Mol Sci 2023; 24:11497. [PMID: 37511253 PMCID: PMC10380887 DOI: 10.3390/ijms241411497] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/07/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
The mechanistic target of rapamycin (mTOR) kinase is a central regulator of cell growth and metabolism. It is the catalytic subunit of two distinct large protein complexes, mTOR complex 1 (mTORC1) and mTORC2. mTOR activity is subjected to tight regulation in response to external nutrition and growth factor stimulation. As an important mechanism of signaling transduction, the 'second messenger' cyclic nucleotides including cAMP and cGMP and their associated cyclic nucleotide-dependent kinases, including protein kinase A (PKA) and protein kinase G (PKG), play essential roles in mediating the intracellular action of a variety of hormones and neurotransmitters. They have also emerged as important regulators of mTOR signaling in various physiological and disease conditions. However, the mechanism by which cAMP and cGMP regulate mTOR activity is not completely understood. In this review, we will summarize the earlier work establishing the ability of cAMP to dampen mTORC1 activation in response to insulin and growth factors and then discuss our recent findings demonstrating the regulation of mTOR signaling by the PKA- and PKG-dependent signaling pathways. This signaling framework represents a new non-canonical regulation of mTOR activity that is independent of AKT and could be a novel mechanism underpinning the action of a variety of G protein-coupled receptors that are linked to the mTOR signaling network. We will further review the implications of these signaling events in the context of cardiometabolic disease, such as obesity, non-alcoholic fatty liver disease, and cardiac remodeling. The metabolic and cardiac phenotypes of mouse models with targeted deletion of Raptor and Rictor, the two essential components for mTORC1 and mTORC2, will be summarized and discussed.
Collapse
Affiliation(s)
- Fubiao Shi
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sheila Collins
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN 37232, USA
| |
Collapse
|
13
|
Lashgari NA, Roudsari NM, Zadeh SST, Momtaz S, Abbasifard M, Reiner Ž, Abdolghaffari AH, Sahebkar A. Statins block mammalian target of rapamycin pathway: a possible novel therapeutic strategy for inflammatory, malignant and neurodegenerative diseases. Inflammopharmacology 2023; 31:57-75. [PMID: 36574095 PMCID: PMC9792946 DOI: 10.1007/s10787-022-01077-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 07/12/2022] [Indexed: 12/28/2022]
Abstract
Inflammation plays a critical role in several diseases such as cancer, gastric, heart and nervous system diseases. Data suggest that the activation of mammalian target of rapamycin (mTOR) pathway in epithelial cells leads to inflammation. Statins, the inhibitors of the 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA), seem to be able to inhibit the mTOR. Statins are considered to have favorable effects on inflammatory diseases by reducing the complications caused by inflammation and by regulating the inflammatory process and cytokines secretion. This critical review collected data on this topic from clinical, in vivo and in vitro studies published between 1998 and June 2022 in English from databases including PubMed, Google Scholar, Scopus, and Cochrane libraries.
Collapse
Affiliation(s)
- Naser-Aldin Lashgari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Momeni Roudsari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | | | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran
- Toxicology and Diseases Group (TDG), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mitra Abbasifard
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
- Department of Internal Medicine, Ali-Ibn Abi-Talib Hospital, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Željko Reiner
- Department of Internal Medicine, School of Medicine, University Hospital Center Zagreb, University of Zagreb, Zagreb, Croatia
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
- Medicinal Plants Research Center, Institute of Medicinal Plants, ACECR, Tehran, Iran.
- Toxicology and Diseases Group (TDG), The Institute of Pharmaceutical Sciences (TIPS), and Faculty of Pharmacy, Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran.
- GI Pharmacology Interest Group (GPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- School of Medicine, The University of Western Australia, Perth, Australia.
- Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
14
|
Huang D, Chen D, Hu T, Liang H. GATA2 promotes oxidative stress to aggravate renal ischemia-reperfusion injury by up-regulating Redd1. Mol Immunol 2023; 153:75-84. [PMID: 36444820 DOI: 10.1016/j.molimm.2022.09.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 09/08/2022] [Accepted: 09/24/2022] [Indexed: 11/26/2022]
Abstract
Renal ischemia-reperfusion injury (RIRI) is a common pathophysiological process, and it is also an important cause of acute renal failure. Therefore, finding an effective therapeutic target for RIRI is extremely urgent. In our study, we constructed hypoxia-reoxygenation (HR) model in vitro and a renal ischemia-reperfusion (IR) model in vivo. Elevated levels of serum creatinine (Cr), blood urea nitrogen (BUN) tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and malondialdehyde (MDA) along with the decreased levels of superoxide dismutase (SOD) and glutathione (GSH) proved that kidney function was damaged after IR, and pathological changes of renal tissues were observed using HE staining and TUNEL staining. The protein of Redd1 expression level was detected to be upregulated after IR by western blot (WB). However, transfection of short hairpin RNA of Redd1 (sh-Redd1) alleviated the HR injury on LLC-PK1 cells, as evidenced by increased cell viability, proliferation and decreased cell apoptosis; additionally, the accumulation of ROS was inhibited. Sh-Redd1 also alleviated IR injury in the mouse model. Subsequently, GATA2 was proved to be upregulated in IR and HR models and was the transcription factor of Redd1. Knockdown of GATA2 efficiently mitigated the oxidative stress induced damages in vivo and in vitro, while these mitigations were reversed by transfection of Redd1 overexpression plasmid. In conclusion, our study clarified the possible underlying mechanism of protecting RIRI.
Collapse
Affiliation(s)
- Dan Huang
- Institutes Renal Division, Wuhan Integrated TCM & Western Medicine Hospital, Wuhan 430022, China
| | - Dan Chen
- Institutes Renal Division, Wuhan Integrated TCM & Western Medicine Hospital, Wuhan 430022, China.
| | - Taotao Hu
- Institutes Renal Division, Wuhan Integrated TCM & Western Medicine Hospital, Wuhan 430022, China
| | - Hongqing Liang
- Institutes Renal Division, Wuhan Integrated TCM & Western Medicine Hospital, Wuhan 430022, China.
| |
Collapse
|
15
|
Long Y, Li Z, Huang C, Lu Z, Qiu K, He M, Fang Z, Ding B, Yuan X, Zhu W. Mechanism and Protective Effect of Smilax glabra Roxb on the Treatment of Heart Failure via Network Pharmacology Analysis and Vitro Verification. Front Pharmacol 2022; 13:868680. [PMID: 35677443 PMCID: PMC9169610 DOI: 10.3389/fphar.2022.868680] [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] [Received: 02/03/2022] [Accepted: 03/15/2022] [Indexed: 11/13/2022] Open
Abstract
Smilax glabra Roxb (SGR) has been widely applied alone or in combination with other Chinese herbs in heart failure (HF), but its mechanism and protective effect have not been investigated. We aimed to explore the mechanism and protective effect of SGR on the treatment of HF. Network pharmacology analysis predicted that SGR was involved in the regulation of cell proliferation, oxidation–reduction process, apoptotic process, ERK1 and ERK2 cascade, MAPK cascade, etc. Its mechanism was mainly involved in the MAPK signaling pathway, calcium signaling pathway, cardiac muscle contraction, etc. Subsequently, SGR was proved to improve cellular viability, restore cellular morphology, suppress cellular and mitochondrial ROS production, improve H2O2-induced lysosome inhibition, attenuate mitochondrial dysfunction, and protect mitochondrial respiratory and energy metabolism in H9c2 cells. SGR activated the p38MAPK pathway by decreasing the mRNA expression of AKT, PP2A, NF-KB, PP2A, RAC1, and CDC42 and increasing the mRNA expression of Jun, IKK, and Sirt1. SGR also decreased the protein expression of ERK1, ERK2, JNK, Bax, and Caspase3 and increased the protein expression of p38MAPK and Bcl-2. In addition, Istidina at the highest degree was identified in SGR via the UHPLCLTQ-Orbitrap-MSn method, and it was suggested as anti-heart failure agents by targeting SRC with molecular docking analysis. In conclusion, SGR has a protective effect on HF through cellular and mitochondrial protection via multi-compounds and multi-targets, and its mechanism is involved in activating the p38 MAPK pathway. Istidina may be possible anti-HF agents by targeting SRC.
Collapse
Affiliation(s)
- Yingxin Long
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zunjiang Li
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Chunxia Huang
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhongyu Lu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Kuncheng Qiu
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Meixing He
- The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhijian Fang
- Department of Emergency, Panyu Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Banghan Ding
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Xiaohong Yuan
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| | - Wei Zhu
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou, China
| |
Collapse
|
16
|
Abstract
Complex multicellular organisms have evolved specific mechanisms to replenish cells in homeostasis and during repair. Here, we discuss how emerging technologies (e.g., single-cell RNA sequencing) challenge the concept that tissue renewal is fueled by unidirectional differentiation from a resident stem cell. We now understand that cell plasticity, i.e., cells adaptively changing differentiation state or identity, is a central tissue renewal mechanism. For example, mature cells can access an evolutionarily conserved program (paligenosis) to reenter the cell cycle and regenerate damaged tissue. Most tissues lack dedicated stem cells and rely on plasticity to regenerate lost cells. Plasticity benefits multicellular organisms, yet it also carries risks. For one, when long-lived cells undergo paligenotic, cyclical proliferation and redif-ferentiation, they can accumulate and propagate acquired mutations that activate oncogenes and increase the potential for developing cancer. Lastly, we propose a new framework for classifying patterns of cell proliferation in homeostasis and regeneration, with stem cells representing just one of the diverse methods that adult tissues employ.
Collapse
Affiliation(s)
- Jeffrey W. Brown
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Charles J. Cho
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA,Current affiliation: Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jason C. Mills
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA,Current affiliation: Section of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA,Departments of Pathology and Immunology and Developmental Biology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA,Current affiliation: Departments of Medicine, Pathology and Immunology, and Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
17
|
Umbarkar P, Ejantkar S, Tousif S, Lal H. Mechanisms of Fibroblast Activation and Myocardial Fibrosis: Lessons Learned from FB-Specific Conditional Mouse Models. Cells 2021; 10:cells10092412. [PMID: 34572061 PMCID: PMC8471002 DOI: 10.3390/cells10092412] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 01/26/2023] Open
Abstract
Heart failure (HF) is a leading cause of morbidity and mortality across the world. Cardiac fibrosis is associated with HF progression. Fibrosis is characterized by the excessive accumulation of extracellular matrix components. This is a physiological response to tissue injury. However, uncontrolled fibrosis leads to adverse cardiac remodeling and contributes significantly to cardiac dysfunction. Fibroblasts (FBs) are the primary drivers of myocardial fibrosis. However, until recently, FBs were thought to play a secondary role in cardiac pathophysiology. This review article will present the evolving story of fibroblast biology and fibrosis in cardiac diseases, emphasizing their recent shift from a supporting to a leading role in our understanding of the pathogenesis of cardiac diseases. Indeed, this story only became possible because of the emergence of FB-specific mouse models. This study includes an update on the advancements in the generation of FB-specific mouse models. Regarding the underlying mechanisms of myocardial fibrosis, we will focus on the pathways that have been validated using FB-specific, in vivo mouse models. These pathways include the TGF-β/SMAD3, p38 MAPK, Wnt/β-Catenin, G-protein-coupled receptor kinase (GRK), and Hippo signaling. A better understanding of the mechanisms underlying fibroblast activation and fibrosis may provide a novel therapeutic target for the management of adverse fibrotic remodeling in the diseased heart.
Collapse
Affiliation(s)
- Prachi Umbarkar
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| | - Suma Ejantkar
- School of Health Professions, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Sultan Tousif
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Hind Lal
- Division of Cardiovascular Disease, The University of Alabama at Birmingham, Birmingham, AL 35294, USA;
- Correspondence: (P.U.); (H.L.); Tel.: +1-205-996-4248 (P.U.); +1-205-996-4219 (H.L.); Fax: +1-205-975-5104 (H.L.)
| |
Collapse
|
18
|
Polypeptide Globular Adiponectin Ameliorates Hypoxia/Reoxygenation-Induced Cardiomyocyte Injury by Inhibiting Both Apoptosis and Necroptosis. J Immunol Res 2021; 2021:1815098. [PMID: 34307691 PMCID: PMC8282401 DOI: 10.1155/2021/1815098] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/16/2021] [Indexed: 01/11/2023] Open
Abstract
Adiponectin is a small peptide secreted and a key component of the endocrine system and immune system. Although globular adiponectin protects myocardial ischemia/reperfusion-induced cardiomyocyte injury, the protective mechanisms remain largely unresolved. Using a neonatal rat ventricular myocyte hypoxia/reoxygenation model, we investigated the role of its potential mechanisms of necroptosis in globular adiponectin-mediated protection in hypoxia/reoxygenation-induced cardiomyocyte injury as compared to apoptosis. We found that globular adiponectin treatment attenuated cardiomyocyte injury as indicated by increased cell viability and reduced lactate dehydrogenase release following hypoxia/reoxygenation. Immunofluorescence staining and Western blotting demonstrated that both necroptosis and apoptosis were triggered by hypoxia/reoxygenation and diminished by globular adiponectin. Necrostatin-1 (RIP1-specific inhibitor) and Z-VAD-FMK (pan-caspase inhibitor) only mimicked the inhibition of necroptosis and apoptosis, respectively, by globular adiponectin in hypoxia/reoxygenation-treated cardiomyocytes. Globular adiponectin attenuated reactive oxygen species production, oxidative damage, and p38MAPK and NF-κB signaling, all important for necroptosis and apoptosis. Collectively, our study suggests that globular adiponectin inhibits hypoxia/reoxygenation-induced necroptosis and apoptosis in cardiomyocytes probably by reducing oxidative stress and interrupting p38MAPK signaling.
Collapse
|
19
|
Liu Y, Li M, Sun M, Zhang Y, Li X, Sun W, Quan N. Sestrin2 is an endogenous antioxidant that improves contractile function in the heart during exposure to ischemia and reperfusion stress. Free Radic Biol Med 2021; 165:385-394. [PMID: 33581276 DOI: 10.1016/j.freeradbiomed.2021.01.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 12/25/2022]
Abstract
Sestrin2 (Sesn2) is a stress-inducible protein that plays a critical role in the response to ischemic stress. We recently recognized that Sesn2 may protect the heart against ischemic insults by reducing the generation of reactive oxygen species (ROS). After 45 min of ischemia followed by 24 h of reperfusion, myocardial infarcts were significantly larger in Sesn2 KO hearts than in wild-type hearts. Isolated cardiomyocytes from wild-type hearts treated with hypoxia and reoxygenation (H/R) stress showed significantly greater Sesn2 levels, compared with normoxic hearts (p < 0.05). Intriguingly, the administration of adeno-associated virus 9-Sesn2 into Sesn2 knockout (KO) hearts rescued Sesn2 protein levels and significantly improved the cardiac function of Sesn2 KO mice exposed to ischemia and reperfusion. The rescued levels of Sesn2 in Sesn2 KO hearts significantly ameliorated ROS generation and the activation of ROS-related stress signaling pathways during ischemia and reperfusion. Moreover, the rescued Sesn2 levels in Sesn2 KO cardiomyocytes improved the maximal velocity of cardiomyocyte shortening by H/R stress. Rescued Sesn2 levels also improved peak height, peak shortening amplitude, and maximal velocity of the re-lengthening of Sesn2 KO cardiomyocytes subjected to H/R. Finally, the rescued Sesn2 levels significantly augmented intracellular calcium levels and reduced the mean time constant of transient calcium decay in Sesn2 KO cardiomyocytes exposed to H/R. Overall, these findings indicated that Sesn2 can act as an endogenous antioxidant to maintain intracellular redox homeostasis under ischemic stress conditions.
Collapse
Affiliation(s)
- Yunxia Liu
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meina Li
- Department of Infection Control, The First Hospital of Jilin University, Changchun, 130021, China
| | - Meihua Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, 130021, China
| | - Yaoting Zhang
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Xuan Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, 39216, USA
| | - Wanqing Sun
- Fuwai Hospital, National Centre for Cardiovascular Disease, No. 167 Beilishi Road, Xicheng, Beijing, 100037, China.
| | - Nanhu Quan
- Department of Cardiovascular Center, The First Hospital of Jilin University, Changchun, 130021, China.
| |
Collapse
|
20
|
Dutta N, Ghosh S, Nelson VK, Sareng HR, Majumder C, Mandal SC, Pal M. Andrographolide upregulates protein quality control mechanisms in cell and mouse through upregulation of mTORC1 function. Biochim Biophys Acta Gen Subj 2021; 1865:129885. [PMID: 33639218 DOI: 10.1016/j.bbagen.2021.129885] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 02/17/2021] [Accepted: 02/18/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heat shock response (HSR), a component of cellular protein quality control mechanisms, is defective in different neurodegenerative conditions such as Parkinson's disease (PD). Forced upregulation of heat shock factor 1 (HSF1), an HSR master regulator, showed therapeutic promise in PD models. Many of the reported small-molecule HSF1 activators have limited functions. Therefore, identification and understanding the molecular bases of action of new HSF1 activating molecules is necessary. METHOD We used a cell-based reporter system to screen Andrographis paniculata leaf extract to isolate andrographolide as an inducer of HSF1 activity. The andrographolide activity was characterized by analyzing its role in different protein quality control mechanisms. RESULT We find that besides ameliorating the PD in MPTP-treated mice, andrographolide upregulated different machineries controlled by HSF1 and NRF2 in both cell and mouse brain. Andrographolide achieves these functions through mTORC1 activated via p38 MAPK and ERK pathways. NRF2 activation is reflected in the upregulation of proteasome as well as autophagy pathways. We further show that NRF2 activation is mediated through mTORC1 driven phosphorylation of p62/sequestosome 1. Studies with different cell types suggested that andrographolide-mediated induction of ROS level underlies all these activities in agreement with the upregulation of mTORC1 and NRF2-antioxidant pathway in mice. CONCLUSION Andrographolide through upregulating HSF1 activity ameliorates protein aggregation induced cellular toxicity. GENERAL SIGNIFICANCE Our results provide a reasonable basis for use of andrographolide in the therapy regimen for the treatment of PD.
Collapse
Affiliation(s)
- Naibedya Dutta
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Suvranil Ghosh
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Vinod K Nelson
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Hossainoor R Sareng
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Chirantan Majumder
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India
| | - Subhash C Mandal
- Pharmacognosy and Phytotherapy Research Laboratory, Division of Pharmacognosy, Department of Pharmaceutical Technology, Jadavpur University, Kolkata 700032, India
| | - Mahadeb Pal
- Division of Molecular Medicine, Bose Institute, P1/12 CIT Scheme VIIM, Kolkata 700054, India.
| |
Collapse
|
21
|
Canovas B, Nebreda AR. Diversity and versatility of p38 kinase signalling in health and disease. Nat Rev Mol Cell Biol 2021; 22:346-366. [PMID: 33504982 PMCID: PMC7838852 DOI: 10.1038/s41580-020-00322-w] [Citation(s) in RCA: 357] [Impact Index Per Article: 89.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2020] [Indexed: 02/06/2023]
Abstract
The ability of cells to deal with different types of stressful situations in a precise and coordinated manner is key for survival and involves various signalling networks. Over the past 25 years, p38 kinases — in particular, p38α — have been implicated in the cellular response to stress at many levels. These span from environmental and intracellular stresses, such as hyperosmolarity, oxidative stress or DNA damage, to physiological situations that involve important cellular changes such as differentiation. Given that p38α controls a plethora of functions, dysregulation of this pathway has been linked to diseases such as inflammation, immune disorders or cancer, suggesting the possibility that targeting p38α could be of therapeutic interest. In this Review, we discuss the organization of this signalling pathway focusing on the diversity of p38α substrates, their mechanisms and their links to particular cellular functions. We then address how the different cellular responses can be generated depending on the signal received and the cell type, and highlight the roles of this kinase in human physiology and in pathological contexts. p38α — the best-characterized member of the p38 kinase family — is a key mediator of cellular stress responses. p38α is activated by a plethora of signals and functions through a multitude of substrates to regulate different cellular behaviours. Understanding context-dependent p38α signalling provides important insights into p38α roles in physiology and pathology.
Collapse
Affiliation(s)
- Begoña Canovas
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Angel R Nebreda
- Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, Spain. .,ICREA, Barcelona, Spain.
| |
Collapse
|
22
|
Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life (Basel) 2021; 11:60. [PMID: 33467601 PMCID: PMC7829951 DOI: 10.3390/life11010060] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging can be seen as process characterized by accumulation of oxidative stress induced damage. Oxidative stress derives from different endogenous and exogenous processes, all of which ultimately lead to progressive loss in tissue and organ structure and functions. The oxidative stress theory of aging expresses itself in age-related diseases. Aging is in fact a primary risk factor for many diseases and in particular for cardiovascular diseases and its derived morbidity and mortality. Here we highlight the role of oxidative stress in age-related cardiovascular aging and diseases. We take into consideration the molecular mechanisms, the structural and functional alterations, and the diseases accompanied to the cardiovascular aging process.
Collapse
Affiliation(s)
- Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paolo Vitillo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
23
|
Akebia Saponin D prevents axonal loss against TNF-induced optic nerve damage with autophagy modulation. Mol Biol Rep 2020; 47:9733-9738. [PMID: 33249542 PMCID: PMC7723935 DOI: 10.1007/s11033-020-06008-y] [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] [Received: 06/18/2020] [Accepted: 11/16/2020] [Indexed: 10/30/2022]
Abstract
Akebia Saponin D (ASD), a triterpenoid saponin, was shown to have protective effects in certain neuronal cells. The purpose of the present study was to investigate the possibility of ASD to prevent tumor necrosis factor (TNF)-induced axonal loss and the ASD modulation of the biologic process of autophagy in optic nerves. Rats were given intravitreal administration of TNF, simultaneous administration of 2, 20, or 200 pmol ASD and TNF, or ASD alone. LC3-II and p62 expression, which is a marker of autophagic flux, and phosphorylated p38 (p-p38) expression in optic nerves were examined by immunoblot analysis. Morphometric analysis revealed a significant ameliorated effect of ASD against TNF-induced optic nerve damage. p62 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was totally prevented by ASD. The ASD alone injection showed significant reduction of p62 levels compared with the PBS-treated control eyes. LC3-II was significantly increased by ASD treatment in the TNF-injected eyes. p-p38 was significantly increased in the optic nerve in TNF-treated eyes, but this increase was completely prevented by ASD. The protective effects of ASD may be associated with enhanced autophagy activation and inhibition of p-p38.
Collapse
|
24
|
Britto FA, Dumas K, Giorgetti-Peraldi S, Ollendorff V, Favier FB. Is REDD1 a metabolic double agent? Lessons from physiology and pathology. Am J Physiol Cell Physiol 2020; 319:C807-C824. [PMID: 32877205 DOI: 10.1152/ajpcell.00340.2020] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The Akt/mechanistic target of rapamycin (mTOR) signaling pathway governs macromolecule synthesis, cell growth, and metabolism in response to nutrients and growth factors. Regulated in development and DNA damage response (REDD)1 is a conserved and ubiquitous protein, which is transiently induced in response to multiple stimuli. Acting like an endogenous inhibitor of the Akt/mTOR signaling pathway, REDD1 protein has been shown to regulate cell growth, mitochondrial function, oxidative stress, and apoptosis. Recent studies also indicate that timely REDD1 expression limits Akt/mTOR-dependent synthesis processes to spare energy during metabolic stresses, avoiding energy collapse and detrimental consequences. In contrast to this beneficial role for metabolic adaptation, REDD1 chronic expression appears involved in the pathogenesis of several diseases. Indeed, REDD1 expression is found as an early biomarker in many pathologies including inflammatory diseases, cancer, neurodegenerative disorders, depression, diabetes, and obesity. Moreover, prolonged REDD1 expression is associated with cell apoptosis, excessive reactive oxygen species (ROS) production, and inflammation activation leading to tissue damage. In this review, we decipher several mechanisms that make REDD1 a likely metabolic double agent depending on its duration of expression in different physiological and pathological contexts. We also discuss the role played by REDD1 in the cross talk between the Akt/mTOR signaling pathway and the energetic metabolism.
Collapse
Affiliation(s)
| | - Karine Dumas
- Université Cote d'Azur, INSERM, UMR1065, C3M, Nice, France
| | | | | | | |
Collapse
|
25
|
Shin S, Gombedza FC, Bandyopadhyay BC. l-ornithine activates Ca 2+ signaling to exert its protective function on human proximal tubular cells. Cell Signal 2020; 67:109484. [PMID: 31770578 PMCID: PMC7302702 DOI: 10.1016/j.cellsig.2019.109484] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 01/14/2023]
Abstract
Oxidative stress and reactive oxygen species (ROS) generation can be influenced by G-protein coupled receptor (GPCR)-mediated regulation of intracellular Ca2+ ([Ca2+]i) signaling. ROS production are much higher in proximal tubular (PT) cells; in addition, the lack of antioxidants enhances the vulnerability to oxidative damage. Despite such predispositions, PT cells show resiliency, and therefore must possess some inherent mechanism to protect from oxidative damage. While the mechanism in unknown, we tested the effect of l-ornithine, since it is abundantly present in PT luminal fluid and can activate Ca2+-sensing receptor (CaSR), a GPCR, expressed in the PT luminal membrane. We used human kidney 2 (HK2) cells, a PT cell line, and performed Ca2+ imaging and electrophysiological experiments to show that l-ornithine has a concentration-dependent effect on CaSR activation. We further demonstrate that the operation of CaSR activated Ca2+ signaling in HK-2 cells mediated by the transient receptor potential canonical (TRPC) dependent receptor-operated Ca2+ entry (ROCE) using pharmacological and siRNA inhibitors. Since PT cells are vulnerable to ROS, we simulated such deleterious effects using genetically encoded peroxide-induced ROS production (HyperRed indicator) to show that the l-ornithine-induced ROCE mediated [Ca2+]i signaling protects from ROS production. Furthermore, we performed cell viability, necrosis and apoptosis assays, and mitochondrial oxidative gene expression to establish that presence of l-ornithine rescued the ROS-induced damage in HK-2 cells. Moreover, l-ornithine-activation of CaSR can reverse ROS production and apoptosis via mitogen-activated protein kinase p38 activation. Such nephroprotective role of l-ornithine can be useful as the translational option for reversing kidney diseases involving PT cell damage due to oxidative stress or crystal nephropathies.
Collapse
Affiliation(s)
- Samuel Shin
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Farai C Gombedza
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA
| | - Bidhan C Bandyopadhyay
- Calcium Signaling Laboratory, Research Service, Veterans Affairs Medical Center, 50 Irving Street, NW, Washington, DC 20422, USA; Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University, Washington, DC 20037, USA; Department of Biomedical Engineering, The Catholic University of America, 620 Michigan Avenue NE, Washington, DC 20064, USA.
| |
Collapse
|
26
|
Anaruma CP, Pereira RM, Cristina da Cruz Rodrigues K, Ramos da Silva AS, Cintra DE, Ropelle ER, Pauli JR, Pereira de Moura L. Rock protein as cardiac hypertrophy modulator in obesity and physical exercise. Life Sci 2019; 254:116955. [PMID: 31626788 DOI: 10.1016/j.lfs.2019.116955] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/02/2019] [Accepted: 10/10/2019] [Indexed: 01/03/2023]
Abstract
Obesity and cardiovascular diseases are worldwide public health issues. In this review, we discussed the participation of ROCK protein in cardiac hypertrophy, mainly through the modulation of leptin and insulin signaling pathways. Leptin plays a role in cardiovascular disease development and, through the Rho-associated protein kinase (ROCK), promotes cardiac hypertrophy. ROCK protein, is regulated by small Rho-GTPases and has two isoforms with high homology. ROCK is able to activate the MAP kinase (MAPK) pathway and modulate insulin signaling in the heart, participating in cardiac hypertrophy development of concentric and eccentric left ventricle growth. Although different types of stimulus can lead to morphologically antagonistic heart growth, physical exercise promotes improvements in hemodynamic function, emerging as a promising non-pharmacological tool to improve overall health. Leptin can activate ROCK in a pathological way, increasing MAPK activity and decreasing insulin signaling via insulin receptor substrate 1 (IRS1) serine 307 residue phosphorylation, phosphatase and tensin homolog, and protein kinase Cβ2. In turn, physical exercise decreases leptin levels and positively modulates insulin signaling as well as increases ROCK-dependent IRS1 (Ser632/635) phosphorylation, improving phosphatidylinositol 3-kinase/protein kinase B axis and promoting physiologic heart growth. Currently, there is a lack of studies about differences in ROCK isoforms, especially during exercise and/or obesity. However, the understanding of its biological function and the complex mechanism underlying the distinct types of cardiac hypertrophy development can be a useful tool in the improvement and treatment of cardiovascular outcomes.
Collapse
Affiliation(s)
- Chadi Pellegrini Anaruma
- Department of Physical Education, Institute of Biosciences - São Paulo State University (UNESP), Rio Claro, SP, Brazil; Exercise Cell Biology Lab (ECEBIL), School of Applied Science - University of Campinas, Limeira, SP, Brazil
| | - Rodrigo Martins Pereira
- Exercise Cell Biology Lab (ECEBIL), School of Applied Science - University of Campinas, Limeira, SP, Brazil; CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil
| | - Kellen Cristina da Cruz Rodrigues
- Exercise Cell Biology Lab (ECEBIL), School of Applied Science - University of Campinas, Limeira, SP, Brazil; CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil
| | | | - Dennys Esper Cintra
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil; Laboratory of Nutritional Genomics (LabGeN), School of Applied Science - University of Campinas, Limeira, SP, Brazil
| | - Eduardo Rochete Ropelle
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil; Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science - University of Campinas, Limeira, SP, Brazil
| | - José Rodrigo Pauli
- CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil; Laboratory of Molecular Biology of Exercise (LaBMEx), School of Applied Science - University of Campinas, Limeira, SP, Brazil
| | - Leandro Pereira de Moura
- Department of Physical Education, Institute of Biosciences - São Paulo State University (UNESP), Rio Claro, SP, Brazil; Exercise Cell Biology Lab (ECEBIL), School of Applied Science - University of Campinas, Limeira, SP, Brazil; CEPECE - Center of Research in Sport Sciences, School of Applied Sciences - University of Campinas (UNICAMP), Limeira, SP, Brazil.
| |
Collapse
|
27
|
Huang P, Fu J, Chen L, Ju C, Wu K, Liu H, Liu Y, Qi B, Qi B, Liu L. Redd1 protects against post‑infarction cardiac dysfunction by targeting apoptosis and autophagy. Int J Mol Med 2019; 44:2065-2076. [PMID: 31638187 PMCID: PMC6844599 DOI: 10.3892/ijmm.2019.4366] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/06/2019] [Indexed: 12/13/2022] Open
Abstract
Post-infarction remodeling is accompanied and influenced by perturbations in the mammalian target of rapamycin (mTOR) signaling. Regulated in development and DNA damage response-1 (Redd1) has been reported to be involved in DNA repair and modulation of mTOR activity. However, little is known about the role of Redd1 in the heart. In the present study the potential contribution of Redd1 overex-pression to the chronic phase of heart failure after myocardial infarction (MI) was explored and the mechanisms underlying Redd1 actions were determined. Redd1 was downregulated in the mouse heart subjected to MI surgery. To determine the role of Redd1 in the process of MI, adeno-associated virus 9 mediated overexpression of Redd1 was used to enhance Redd1 content in cardiomyocytes. Redd1 overexpression improved left ventricular dysfunction and reduced the expansion index. Additionally, Redd1 overexpression resulted in suppressed myocardial apoptosis and improved autophagy. Furthermore, the studies revealed that Redd1 overexpression could inhibit the phosphorylation of mTOR and its downstream effectors P70/S6 kinase and 4EBP1. In conclusion, this study demonstrated that Redd1 overexpression protects against the development and persistence of heart failure post MI by reducing apoptosis and enhancing autophagy via the mTOR signaling pathway. The present study clearly demonstrated that Redd1 is a therapeutic target in the development of heart failure after MI.
Collapse
Affiliation(s)
- Pianpian Huang
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Jun Fu
- Department of Radiology, Wuhan No. 1 Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Long Chen
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Chenhui Ju
- Clinical Center for Human Genomic Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Kefei Wu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Hongxia Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Yun Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Benming Qi
- Department of Otorhinolaryngology, First People's Hospital of Yunnan Province, Kunming, Yunnan 650000, P.R. China
| | - Benling Qi
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lihua Liu
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
28
|
Ren Q, Zhao S, Ren C. 6-Gingerol protects cardiocytes H9c2 against hypoxia-induced injury by suppressing BNIP3 expression. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:2016-2023. [PMID: 31223035 DOI: 10.1080/21691401.2019.1610415] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Qi Ren
- Department of Cardiology, Jining No.1 People’s Hospital, Jining, China
| | - Shaojun Zhao
- Department of Cardiology, Jining No.1 People’s Hospital, Jining, China
| | - Changjie Ren
- Department of Cardiology, Jining No.1 People’s Hospital, Jining, China
| |
Collapse
|
29
|
Cheng JS, Tsai WL, Liu PF, Goan YG, Lin CW, Tseng HH, Lee CH, Shu CW. The MAP3K7-mTOR Axis Promotes the Proliferation and Malignancy of Hepatocellular Carcinoma Cells. Front Oncol 2019; 9:474. [PMID: 31214512 PMCID: PMC6558008 DOI: 10.3389/fonc.2019.00474] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/17/2019] [Indexed: 12/28/2022] Open
Abstract
Targeted therapy is currently limited for patients with hepatocellular carcinoma (HCC) due to the lack of suitable targets. Kinases play pivotal roles in many cellular biological processes, whereas dysregulation of kinases may lead to various diseases, particularly cancer. However, the role of kinases in HCC malignancy remains unclear. In this study, we employed a kinome small interfering RNA (siRNA) library, comprising 710 kinase-related genes, to screen whether any kinases were essential for cell proliferation in various HCC cell lines. Through a kinome siRNA library screening, we found that MAP3K7 was a crucial gene for HCC cell proliferation. Pharmacological or genetic ablation of MAP3K7 diminished the growth, migration, and invasion of HCC cells, including primary HCC cells. Stable knockdown of MAP3K7 attenuated tumor formation in a spheroid cell culture model and tumor xenograft mouse model. In addition, silencing MAP3K7 reduced the phosphorylation and expression of mammalian target of rapamycin (mTOR) in HCC cells. MAP3K7 expression was positively correlated with mTOR expression in tumors of patients with HCC. Higher co-expression of MAP3K7 and mTOR was significantly associated with poor prognosis of HCC. Taken together, our results revealed that the MAP3K7-mTOR axis might promote tumorigenesis and malignancy, which provides a potential marker or therapeutic target for HCC patients.
Collapse
Affiliation(s)
- Jin-Shiung Cheng
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Wei-Lun Tsai
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pei-Feng Liu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Yih-Gang Goan
- Division of Thoracic Surgery, Department of Surgery, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chih-Wen Lin
- Division of Gastroenterology and Hepatology, E-Da Dachang Hospital, I-Shou University, Kaohsiung, Taiwan.,School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| | - Ho-Hsing Tseng
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Cheng-Hsin Lee
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chih-Wen Shu
- School of Medicine for International Students, I-Shou University, Kaohsiung, Taiwan
| |
Collapse
|
30
|
Díaz RG, Escudero DS, Brea MS, Morgan PE, Pérez NG. p38 mitogen activated protein kinase mediates cardiac Na +/H + exchanger inhibition induced by Sildenafil. Eur J Pharmacol 2019; 849:96-105. [PMID: 30721701 DOI: 10.1016/j.ejphar.2019.01.070] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Revised: 01/11/2019] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
Abstract
Since the original description as potent antianginal compounds, phosphodiesterase 5A inhibitors have continuously increased their possible therapeutic applications. In the heart, Sildenafil was shown to protect against an ischemic insult by decreasing cardiac Na+/H+ exchanger (NHE1) activity, action that was mediated by protein kinase G. p38 mitogen activated protein kinase (p38MAPK) activation was described in cardiac ischemia, but its precise role remains elusive. It has been shown that p38MAPK is activated by protein kinase G (PKG) in certain non-cardiac tissues, while in others modulates NHE1 activity. Current study was aimed to seek the role of p38MAPK in the Sildenafil-triggered pathway leading to NHE1 inhibition in myocardium. Rat isolated papillary muscles were used to evaluate NHE1 activity during intracellular pH recovery from an acidic load. Protein kinases phosphorylation (activation) was determined by western blot. Sustained acidosis promoted NHE1 hyperactivity by enhancing Ser703 phosphorylation, effect that was blunted by Sildenafil. p38MAPK inhibition reversed the effect of Sildenafil on NHE1. Activation of p38MAPK, by Sodium Arsenite or Anisomycin, mimicked the inhibitory effect of Sildenafil on the exchanger. Consistently, Sildenafil induced p38MAPK phosphorylation/activation during acidosis. Neither Sildenafil nor p38MAPK inhibition affected extracellular signal-regulated kinases 1/2 phosphorylation, kinases upstream NHE1. Furthermore, inhibition of NHE1 after p38MAPK activation was precluded by preventing the activation of protein phosphatase 2A with Okadaic Acid. Taken together, these results suggest that activation of p38MAPK is a necessary step to trigger the inhibitory effect of Sildenafil on cardiac NHE1 activity, thorough a mechanism that involves protein phosphatase 2A-mediated exchanger dephosphorylation.
Collapse
Affiliation(s)
- Romina G Díaz
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900 La Plata, Argentina
| | - Daiana S Escudero
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900 La Plata, Argentina
| | - María S Brea
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900 La Plata, Argentina
| | - Patricio E Morgan
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900 La Plata, Argentina.
| | - Néstor G Pérez
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, Calle 60 y 120, 1900 La Plata, Argentina.
| |
Collapse
|
31
|
Liu D, Xu L, Zhang X, Shi C, Qiao S, Ma Z, Yuan J. Snapshot: Implications for mTOR in Aging-related Ischemia/Reperfusion Injury. Aging Dis 2019; 10:116-133. [PMID: 30705773 PMCID: PMC6345330 DOI: 10.14336/ad.2018.0501] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 05/01/2018] [Indexed: 12/15/2022] Open
Abstract
Aging may aggravate the damage and dysfunction of different components of multiorgan and thus increasing multiorgan ischemia/reperfusion (IR) injury. IR injury occurs in many organs and tissues, which is a major cause of morbidity and mortality worldwide. The kinase mammalian target of rapamycin (mTOR), an atypical serine/threonine protein kinase, involves in the pathophysiological process of IR injury. In this review, we first briefly introduce the molecular features of mTOR, the association between mTOR and aging, and especially its role on autophagy. Special focus is placed on the roles of mTOR during ischemic and IR injury. We then clarify the association between mTOR and conditioning phenomena. Following this background, we expand our discussion to potential future directions of research in this area. Collectively, information reviewed herein will serve as a comprehensive reference for the actions of mTOR in IR injury and may be significant for the design of future research and increase the potential of mTOR as a therapeutic target.
Collapse
Affiliation(s)
- Dong Liu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Liqun Xu
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China.,4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Xiaoyan Zhang
- 2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China.,3Cadet group 3, School of Basic Medical Sciences, The Fourth Military Medical University, Xi'an 710032, China
| | - Changhong Shi
- 4Laboratory Animal Center, The Fourth Military Medical University, Xi'an 710032, China
| | - Shubin Qiao
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| | - Zhiqiang Ma
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China.,2Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Jiansong Yuan
- 1State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100037, China
| |
Collapse
|
32
|
Sharma S, Mazumder AG, Rana AK, Patial V, Singh D. Spontaneous Recurrent Seizures Mediated Cardiac Dysfunction via mTOR Pathway Upregulation: A Putative Target for SUDEP Management. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2019; 18:555-565. [PMID: 31368880 DOI: 10.2174/1871527318666190801112027] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/02/2019] [Accepted: 05/08/2019] [Indexed: 12/13/2022]
Abstract
BACKGROUND Alteration in electrophysiology, leading to cardiac dysfunction and subsequently a nontraumatic death is a complication of epilepsy known as "SUDEP" (Sudden Unexpected Death in Epilepsy). AIMS The present study was designed to understand the molecular changes and cardiac parameters during different phases of epileptogenesis in lithium-pilocarpine (Li-pilo) rat model of epilepsy. METHODS The animals were exposed to Li-pilo to induce Spontaneous Recurrent Seizures (SRS). Noninvasive blood pressure and electrocardiography was recorded at 7th, 28th and 75th day following pilocarpine administration, considered as latent, initial and late SRS phases, respectively. The serum biochemistry, cardiac histopathology, protein and mRNA expressions were studied, following electrocardiography on day 75. RESULTS The mean arterial pressure decreased during the latent phase, thereafter it progressively increased during the initial and the late SRS phases, as compared to the basal and the latent phase. Histopathological analysis of the heart sections indicated hypertrophy, degenerative changes and fibrous tissue deposition in epileptic animals, along with increased levels of lactate dehydrogenase and creatine kinase-MB in the serum. The expression of HIF-1α, phospho-S6, phospho-mTOR, TGF-β, collagen I and Na+/K+-ATPase α1 proteins, and mRNA levels of HIF-1α, mTOR, Rps6, Scn1b, Scn3b, Nav1.5 and TGF-β were increased in the cardiac tissue of epileptic animals, as compared to control. CONCLUSION Our results conclusively showed that Li-pilo-induced SRS leads to cardiac dysfunction via mTOR pathway upregulation, thus suggested the regulatory control of mTOR pathway as a potential target for SUDEP management.
Collapse
Affiliation(s)
- Supriya Sharma
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| | - Arindam G Mazumder
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| | - Anil K Rana
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| | - Vikram Patial
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
- Academy of Scientific and Innovative Research (AcSIR), CSIR-Institute of Himalayan Bioresource Technology, Palampur-176061, Himachal Pradesh, India
| |
Collapse
|
33
|
Bi S, Li L, Gu H, Li M, Xu S, Bu W, Zhang M, Zhou Z, Chen X. Lycopene upregulates ZO-1 and downregulates claudin-1 through autophagy inhibition in the human cutaneous squamous cell carcinoma cell line COLO-16. J Cancer 2019; 10:510-521. [PMID: 30719147 PMCID: PMC6360289 DOI: 10.7150/jca.26578] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Lycopene, a kind of carotenoid, has been reported to have an inhibitory function on tumor cell migration. However, the potential role of lycopene in the treatment of cutaneous squamous cell carcinoma (cSCC) remains unclear. Therefore, we assessed the biological effects of lycopene in the human cSCC cell line COLO-16, human epidermal keratinocytes (HEKs) and the immortalized human keratinocyte cell line HaCaT. We found that lycopene inhibited the cell proliferation and migration of COLO-16 cells but not normal keratinocytes. In addition, lycopene upregulated the protein levels of ZO-1 in COLO-16 and HaCaT cells but not in HEKs. In contrast, lycopene upregulated the protein level of claudin-1 in HEKs but downregulated claudin-1 in COLO-16 cells. Lycopene led to a decrease in autophagic flux in COLO-16 cells in a mechanistic target of rapamycin complex 1 (MTORC1)-dependent manner. Importantly, autophagy inhibition contributed to the lycopene-induced regulation on ZO-1 and claudin-1 in COLO-16 cells. Moreover, JNK inhibitor (SP600125) and MEK inhibitor (U0126) treatment abolished the increase in phosphorylated MTOR and ribosomal protein S6 as well as the increase in ZO-1 and the decrease in claudin-1 in lycopene-treated COLO-16 cells. Gene silencing of JNK and ERK also prohibited ZO-1 upregulation and claudin-1 downregulation. In conclusion, lycopene upregulates ZO-1 expression and downregulates claudin-1 expression through the activation of ERK, JNK and MTORC1 as well as the inhibition of autophagy in human cSCC cells. Our findings demonstrate that autophagy plays a key role in lycopene-mediated pharmacological effects. This study indicates that lycopene might be a useful chemopreventive agent against cSCC.
Collapse
Affiliation(s)
- Suyun Bi
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China.,Dermatology and Venereology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Li Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Heng Gu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Min Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Song Xu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Wenbo Bu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Mengli Zhang
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| | - Zhihai Zhou
- Dermatology and Venereology Department, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Chen
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences & Peking Union Medical College, Nanjing, 210042, China
| |
Collapse
|
34
|
Li C, Zhang Y, Wang Q, Meng H, Zhang Q, Wu Y, Xiao W, Wang Y, Tu P. Dragon's Blood exerts cardio-protection against myocardial injury through PI3K-AKT-mTOR signaling pathway in acute myocardial infarction mice model. JOURNAL OF ETHNOPHARMACOLOGY 2018; 227:279-289. [PMID: 30195568 DOI: 10.1016/j.jep.2018.09.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Revised: 08/21/2018] [Accepted: 09/05/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dragon's Blood (DB), the red resin of Dracaena cochinchinensis (Lour.) S. C., has been used in traditional Chinese medicine to treat acute myocardial infarction (AMI) for centuries. Evidence indicated that DB may exert cardio-protective effect by inhibiting inflammatory response during myocardial infarction. However, its pharmaceutical mechanism is still to be elucidated. AIM OF THE STUDY Due to its potential anti-inflammatory effect, Dragon's Blood extract (DBE) was applied on AMI mice model in this study and its mechanism on inflammation via PI3K-AKT-mTOR signaling pathway was to be validated. MATERIALS AND METHODS AMI mice model was established by ligation of left anterior descending (LAD) arteries. DBE was administered for 7 days before the surgery. Heart function was evaluated by 2D echocardiography. Levels of CK-MB and LDH1 in serum as well as TXB2, 6-keto-PGF1α and ET-1 in plasma were detected. Level of IL-6 in cardiac tissues was quantified by ELISA. Expressions of key proteins in PI3K-AKT-mTOR signaling pathway were detected by Western blot. RESULTS The result demonstrated that DBE could improve heart function in AMI mice model. Meanwhile, it could also regulate levels of CK-MB and LDH1, and restore balance between TXB2 and 6-keto-PGF1α. Further study suggested that DBE could inhibit inflammation and regulate expressions of key proteins in IL-6-JAK2/STAT3 pathway in cardiac tissue. Western blot results validated that DBE could activate PI3K-AKT-mTOR signaling pathway, thereby regulating the expressions of its downstream targets, including VEGF, COX2 and PPARγ. CONCLUSION DBE exerts cardio-protective efficacy by activating JAK2-STAT3 and PI3K-AKT-mTOR pathways in cardiac tissue. These findings provide insight into the pharmacological mechanism of DBE and validate the beneficial effects of DBE in the clinical application for AMI.
Collapse
Affiliation(s)
- Chun Li
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Yi Zhang
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qiyan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Hui Meng
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Qian Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Wu
- Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Wei Xiao
- National Key Laboratory of Pharmaceutical New Technology for Chinese Medicine, Jiangsu Kanion Pharmaceutical Co. Ltd., Lianyungang 222001, China
| | - Yong Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
35
|
Diviani D, Osman H, Reggi E. A-Kinase Anchoring Protein-Lbc: A Molecular Scaffold Involved in Cardiac Protection. J Cardiovasc Dev Dis 2018; 5:E12. [PMID: 29419761 PMCID: PMC5872360 DOI: 10.3390/jcdd5010012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 02/02/2018] [Accepted: 02/06/2018] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a lethal disease that can develop after myocardial infarction, hypertension, or anticancer therapy. In the damaged heart, loss of function is mainly due to cardiomyocyte death and associated cardiac remodeling and fibrosis. In this context, A-kinase anchoring proteins (AKAPs) constitute a family of scaffolding proteins that facilitate the spatiotemporal activation of the cyclic adenosine monophosphate (AMP)-dependent protein kinase (PKA) and other transduction enzymes involved in cardiac remodeling. AKAP-Lbc, a cardiac enriched anchoring protein, has been shown to act as a key coordinator of the activity of signaling pathways involved in cardiac protection and remodeling. This review will summarize and discuss recent advances highlighting the role of the AKAP-Lbc signalosome in orchestrating adaptive responses in the stressed heart.
Collapse
Affiliation(s)
- Dario Diviani
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| | - Halima Osman
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| | - Erica Reggi
- Département de Pharmacologie et de Toxicologie, Faculté de Biologie et de Médecine, Lausanne 1005, Switzerland.
| |
Collapse
|
36
|
Wang J, Maimaitili Y, Zheng H, Yu J, Guo H, Ma HP, Chen CL. The influence of rapamycin on the early cardioprotective effect of hypoxic preconditioning on cardiomyocytes. Arch Med Sci 2017; 13:947-955. [PMID: 28721162 PMCID: PMC5507107 DOI: 10.5114/aoms.2016.59712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/14/2015] [Indexed: 11/17/2022] Open
Abstract
INTRODUCTION The purpose of this study was to examine the effects of rapamycin on the cardioprotective effect of hypoxic preconditioning (HPC) and on the mammalian target of rapamycin (mTOR)-mediated hypoxia-inducible factor 1 (HIF-1) signaling pathway. MATERIAL AND METHODS Primary cardiomyocytes were isolated from rat pups and underwent rapamycin and/or HPC, followed by hypoxia/re-oxygenation (H/R) injury. Cell viability and cell injury were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays, and qRT-PCR was used to measure HIF-1α and mTOR mRNA expression. A Langendorff heart perfusion model was conducted to observe the effect of rapamycin. RESULTS Rapamycin treatment nearly abolished the cardioprotective effect of HPC in cardiomyocytes, reduced cell viability (p = 0.007) and increased cell damage (p = 0.032). HIF-1α and mTOR mRNA expression increased in cardiomyocytes undergoing I/R injury within 2 h after HPC. After rapamycin treatment, mTOR mRNA expression and HPC-induced HIF-1α mRNA expression were both reduced (p < 0.001). A Langendorff heart perfusion model in rat hearts showed that rapamycin greatly attenuated the cardioprotective effect of HPC in terms of heart rate, LVDP, and dp/dtmax (all, p < 0.029). CONCLUSIONS Rapamycin, through inhibition of mTOR, reduces the elevated HIF-1α expression at an early stage of HPC, and attenuates the early cardioprotective effect of HPC.
Collapse
Affiliation(s)
- Jiang Wang
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - YiLiyaer Maimaitili
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hong Zheng
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jin Yu
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hai Guo
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Hai-Ping Ma
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Chun-Ling Chen
- Department of Anesthesiology, the First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
37
|
LUCIANO TF, MARQUES SO, PIERI BL, DE SOUZA DR, ARAÚJO LV, NESI RT, SCHEFFER DL, COMIN VH, PINHO RA, MULLER AP, DE SOUZA CT. Responses of Skeletal Muscle Hypertrophy in Wistar Rats to Different Resistance Exercise Models. Physiol Res 2017; 66:317-323. [DOI: 10.33549/physiolres.933256] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
This study aimed to compare the effects of three different resistance exercise models on the quadriceps muscle cross-sectional area, as well as on mTOR phosphorylation and other pivotal molecules involved in the upstream regulation of mTOR. Twenty-four male Wistar rats were divided into untrained (control), endurance resistance training, strength resistance training, and hypertrophy resistance training (HRT) groups (n=6). After 12 weeks of training, the red portion of the quadriceps was removed for histological and Western blot analyses. The results showed that the quadriceps weight and cross-sectional areas in the exercised groups were higher than those of the untrained rats. However, the HRT group presented better results than the other two experimental groups. This same pattern was observed for mTOR phosphorylation and for the most pivotal molecules involved in the upstream control of mTOR (increase of PKB, 14-3-3, ERK, p38 MAPK, and 4E-BP1 phosphorylation, and reduction of tuberin, sestrin 2, REDD1, and phospho AMPK). In summary, our study showed that HRT leads to high levels of mTOR phosphorylation as well as of other proteins involved in the upstream regulation of mTOR.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - C. T. DE SOUZA
- Laboratory of Exercise Biochemistry and Physiology, Health Sciences Unit, University of Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
38
|
Moderate lifelong overexpression of tuberous sclerosis complex 1 (TSC1) improves health and survival in mice. Sci Rep 2017; 7:834. [PMID: 28400571 PMCID: PMC5429778 DOI: 10.1038/s41598-017-00970-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 03/10/2017] [Indexed: 12/16/2022] Open
Abstract
The tuberous sclerosis complex 1/2 (TSC1/2) is an endogenous regulator of the mechanistic target of rapamycin (mTOR). While mTOR has been shown to play an important role in health and aging, the role of TSC1/2 in aging has not been fully investigated. In the current study, a constitutive TSC1 transgenic (Tsc1tg) mouse model was generated and characterized. mTORC1 signaling was reduced in majority of the tissues, except the brain. In contrast, mTORC2 signaling was enhanced in Tsc1tg mice. Tsc1tg mice are more tolerant to exhaustive exercises and less susceptible to isoproterenol-induced cardiac hypertrophy at both young and advanced ages. Tsc1tg mice have less fibrosis and inflammation in aged as well as isoproterenol-challenged heart than age-matched wild type mice. The female Tsc1tg mice exhibit a higher fat to lean mass ratio at advanced ages than age-matched wild type mice. More importantly, the lifespan increased significantly in female Tsc1tg mice, but not in male Tsc1tg mice. Collectively, our data demonstrated that moderate increase of TSC1 expression can enhance overall health, particularly cardiovascular health, and improve survival in a gender-specific manner.
Collapse
|
39
|
Wang M, Huang C, Su Y, Yang C, Xia Q, Xu DJ. Astragaloside II sensitizes human hepatocellular carcinoma cells to 5-fluorouracil via suppression of autophagy. ACTA ACUST UNITED AC 2017; 69:743-752. [PMID: 28266023 DOI: 10.1111/jphp.12706] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 01/12/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Inhibition of autophagy has been increasingly recognized as a potential therapeutic approach against cancer. Our previous reports showed that Astragaloside II improves hepatic cancer cells resistance by downregulating MDR1 and P-gp .The purpose of this study was to further investigated the effect of autophagy on AS-II reversing multidrug resistance and its molecular mechanism in hepatocellular carcinoma cells in vitro. METHODS Bel-7402 and Bel-7402/FU cell lines were used in this study. Western blot was used to detect the expression of autophagy-related protein, p-mTOR and p-p79s6k, MTT was used to analyse cell viability, GFP-LC3 punctate dots distribution was observed by GFP-LC3 transient transfection under fluorescence microscopy and silencing of autophagy-related genes was detected by small interfering RNA transfection. KEY FINDINGS Astragaloside II was able to significantly decrease the expression of LC3-II and Beclin-1 in a dose-dependent manner, Astragaloside II (80 μm) further decreased LC3-II formation, Beclin-1 and GFP-LC3 puncta dots stimulated with 5-fluorouracil (0.2 mm) in Bel-7402/FU cells (P < 0.05). In addition, Astragaloside II is capable of sensitizing cells to 5-fluorouracil-induced cell death via inhibition of pro-survival autophagy involvement of MAPK-mTOR pathway. CONCLUSIONS These findings suggested that Astragaloside II could suppress autophagy by interfering with Beclin-1 and LC3 via MAPK-mTOR pathway, through which sensitized human cancer resistant cells to 5-FU-induced cell death.
Collapse
Affiliation(s)
- Meng Wang
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Can Huang
- Department of Pharmacy, Affiliated Anqing Hospital of Anhui Medical University, Anqing, Anhui, China
| | - Yong Su
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Cui Yang
- Department of Pharmacy, The Fourth Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Quan Xia
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Du-Juan Xu
- Department of Pharmacy, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
40
|
Hayakawa M, Hayakawa H, Petrova T, Ritprajak P, Sutavani RV, Jiménez-Andrade GY, Sano Y, Choo MK, Seavitt J, Venigalla RKC, Otsu K, Georgopoulos K, Arthur JSC, Park JM. Loss of Functionally Redundant p38 Isoforms in T Cells Enhances Regulatory T Cell Induction. J Biol Chem 2016; 292:1762-1772. [PMID: 28011639 PMCID: PMC5290950 DOI: 10.1074/jbc.m116.764548] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/01/2016] [Indexed: 12/16/2022] Open
Abstract
The evolutionarily conserved protein kinase p38 mediates innate resistance to environmental stress and microbial infection. Four p38 isoforms exist in mammals and may have been co-opted for new roles in adaptive immunity. Murine T cells deficient in p38α, the ubiquitously expressed p38 isoform, showed no readily apparent cell-autonomous defects while expressing elevated amounts of another isoform, p38β. Mice with T cells simultaneously lacking p38α and p38β displayed lymphoid atrophy and elevated Foxp3+ regulatory T cell frequencies. Double deficiency of p38α and p38β in naïve CD4+ T cells resulted in an attenuation of MAPK-activated protein kinase (MK)-dependent mTOR signaling after T cell receptor engagement, and enhanced their differentiation into regulatory T cells under appropriate inducing conditions. Pharmacological inhibition of the p38-MK-mTOR signaling module produced similar effects, revealing potential for therapeutic applications.
Collapse
Affiliation(s)
- Morisada Hayakawa
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129; the Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Hiroko Hayakawa
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129; the Department of Biochemistry, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| | - Tsvetana Petrova
- the Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, Dundee DD1 5EH, United Kingdom
| | - Patcharee Ritprajak
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129; the Department of Microbiology and Immunology and Research Unit of Oral Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand
| | - Ruhcha V Sutavani
- the Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, Dundee DD1 5EH, United Kingdom
| | - Guillermina Yanek Jiménez-Andrade
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Yasuyo Sano
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Min-Kyung Choo
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - John Seavitt
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - Ram K C Venigalla
- MRC Protein Phosphorylation Unit, School of Life Sciences, Sir James Black Centre, University of Dundee, Dundee DD1 5EH, United Kingdom
| | - Kinya Otsu
- the Cardiovascular Division, King's College London, London SE5 9NU, United Kingdom
| | - Katia Georgopoulos
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129
| | - J Simon C Arthur
- the Division of Cell Signalling and Immunology, School of Life Sciences, Wellcome Trust Building, Dundee DD1 5EH, United Kingdom
| | - Jin Mo Park
- From the Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129.
| |
Collapse
|
41
|
Freitas ACS, Figueiredo MJ, Campos EC, Soave DF, Ramos SG, Tanowitz HB, Celes MRN. Activation of Both the Calpain and Ubiquitin-Proteasome Systems Contributes to Septic Cardiomyopathy through Dystrophin Loss/Disruption and mTOR Inhibition. PLoS One 2016; 11:e0166839. [PMID: 27880847 PMCID: PMC5120800 DOI: 10.1371/journal.pone.0166839] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 11/05/2016] [Indexed: 11/18/2022] Open
Abstract
Cardiac dysfunction caused by the impairment of myocardial contractility has been recognized as an important factor contributing to the high mortality in sepsis. Calpain activation in the heart takes place in response to increased intracellular calcium influx resulting in proteolysis of structural and contractile proteins with subsequent myocardial dysfunction. The purpose of the present study was to test the hypothesis that increased levels of calpain in the septic heart leads to disruption of structural and contractile proteins and that administration of calpain inhibitor-1 (N-acetyl-leucinyl-leucinyl-norleucinal (ALLN)) after sepsis induced by cecal ligation and puncture prevents cardiac protein degradation. We also tested the hypothesis that calpain plays a role in the modulation of protein synthesis/degradation through the activation of proteasome-dependent proteolysis and inhibition of the mTOR pathway. Severe sepsis significantly increased heart calpain-1 levels and promoted ubiquitin and Pa28β over-expression with a reduction in the mTOR levels. In addition, sepsis reduced the expression of structural proteins dystrophin and β-dystroglycan as well as the contractile proteins actin and myosin. ALLN administration prevented sepsis-induced increases in calpain and ubiquitin levels in the heart, which resulted in decreased of structural and contractile proteins degradation and basal mTOR expression levels were re-established. Our results support the concept that increased calpain concentrations may be part of an important mechanism of sepsis-induced cardiac muscle proteolysis.
Collapse
Affiliation(s)
- Ana Caroline Silva Freitas
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Maria Jose Figueiredo
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Erica Carolina Campos
- Department of Physiotherapy, Faculty of Physical Education, Federal University of Uberlandia, Minas Gerais, Brazil
| | - Danilo Figueiredo Soave
- Department of Histology, Embryology and Cellular Biology, Federal University of Goias, Goias, Brazil
| | - Simone Gusmao Ramos
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
| | - Herbert B. Tanowitz
- Departments of Pathology and medicine, Albert Einstein College of Medicine, Yeshiva University, Bronx, New York, United States of America
| | - Mara Rúbia N. Celes
- Department of Pathology, Faculty of Medicine of Ribeirao Preto, University of Sao Paulo, Sao Paulo, Brazil
- Institute of Tropical Pathology and Public Health, Federal University of Goias, Goias, Brazil
- * E-mail: ,
| |
Collapse
|
42
|
Li G, Wang G, Ma L, Guo J, Song J, Ma L, Zhao X. miR-22 regulates starvation-induced autophagy and apoptosis in cardiomyocytes by targeting p38α. Biochem Biophys Res Commun 2016; 478:1165-72. [PMID: 27544030 DOI: 10.1016/j.bbrc.2016.08.086] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Accepted: 08/15/2016] [Indexed: 11/19/2022]
Abstract
microRNAs (miRNAs) are short noncoding RNAs that function in RNA silencing and post-transcriptional regulation of gene expression. They play critical regulatory roles in many cardiovascular diseases, including ischemia-induced cardiac injury. Here, we report microRNA-22, highly expressed in the heart, can protect cardiomyocytes from starvation-induced injury through promoting autophagy and inhibiting apoptosis. Quantitative real-time PCR (qPCR) demonstrated that the expression of miR-22 in starvation-treated neonatal rat cardiomyocytes (NRCMs) was markedly down-regulated. Over-expression of miR-22 significantly promoted starvation-induced autophagy and inhibited starvation-induced apoptosis in NRCMs. In contrast, reduction of miR-22 suppressed autophagy and accelerated apoptosis in starving NRCMs. Immunohistochemistry and TUNEL staining results also provided further evidence that miR-22 promoted autophagy and inhibited apoptosis in myocardial cells. Furthermore, both luciferase reporter assay and western blot analysis were performed to identify p38α as a direct target of miR-22. Taken together, miR-22 plays an important role in regulating autophagy and apoptosis in ischemic myocardium through targeting p38α. miR-22 may represent a potential therapeutic target for the treatment of ischemic heart diseases.
Collapse
Affiliation(s)
- Guoran Li
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Guokun Wang
- Institution of Thoracic Cardiac Surgery, Department of Cardiothoracic Surgery, Changhai Hospital, Shanghai 200433, China
| | - Liangliang Ma
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jun Guo
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jingwen Song
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Liping Ma
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Xianxian Zhao
- Department of Cardiology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
43
|
Park KM, Teoh JP, Wang Y, Broskova Z, Bayoumi AS, Tang Y, Su H, Weintraub NL, Kim IM. Carvedilol-responsive microRNAs, miR-199a-3p and -214 protect cardiomyocytes from simulated ischemia-reperfusion injury. Am J Physiol Heart Circ Physiol 2016; 311:H371-83. [PMID: 27288437 PMCID: PMC5005281 DOI: 10.1152/ajpheart.00807.2015] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 06/01/2016] [Indexed: 12/24/2022]
Abstract
The nonselective β-adrenergic receptor antagonist (β-blocker) carvedilol has been shown to protect against myocardial injury, but the detailed underlying mechanisms are unclear. We recently reported that carvedilol stimulates the processing of microRNA (miR)-199a-3p and miR-214 in the heart via β-arrestin1-biased β1-adrenergic receptor (β1AR) cardioprotective signaling. Here, we investigate whether these β-arrestin1/β1AR-responsive miRs mediate the beneficial effects of carvedilol against simulated ischemia/reperfusion (sI/R). Using cultured cardiomyocyte cell lines and primary cardiomyocytes, we demonstrate that carvedilol upregulates miR-199a-3p and miR-214 in both ventricular and atrial cardiomyocytes subjected to sI/R. Overexpression of the two miRs in cardiomyocytes mimics the effects of carvedilol to activate p-AKT survival signaling and the expression of a downstream pluripotency marker Sox2 in response to sI/R. Moreover, carvedilol-mediated p-AKT activation is abolished by knockdown of either miR-199a-3p or miR-214. Along with previous studies to directly link the cardioprotective actions of carvedilol to upregulation of p-AKT/stem cell markers, our findings suggest that the protective roles of carvedilol during ischemic injury are in part attributed to activation of these two protective miRs. Loss of function of miR-199a-3p and miR-214 also increases cardiomyocyte apoptosis after sI/R. Mechanistically, we demonstrate that miR-199a-3p and miR-214 repress the predictive or known apoptotic target genes ddit4 and ing4, respectively, in cardiomyocytes. These findings suggest pivotal roles for miR-199a-3p and miR-214 as regulators of cardiomyocyte survival and contributors to the functional benefits of carvedilol therapy.
Collapse
Affiliation(s)
- Kyoung-Mi Park
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jian-Peng Teoh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongchao Wang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zuzana Broskova
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ahmed S Bayoumi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yaoliang Tang
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Huabo Su
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Il-Man Kim
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia; Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
44
|
Chen R, Wang B, Chen L, Cai D, Li B, Chen C, Huang E, Liu C, Lin Z, Xie WB, Wang H. DNA damage-inducible transcript 4 (DDIT4) mediates methamphetamine-induced autophagy and apoptosis through mTOR signaling pathway in cardiomyocytes. Toxicol Appl Pharmacol 2016; 295:1-11. [PMID: 26825372 DOI: 10.1016/j.taap.2016.01.017] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Revised: 01/20/2016] [Accepted: 01/25/2016] [Indexed: 12/30/2022]
Abstract
Methamphetamine (METH) is an amphetamine-like psychostimulant that is commonly abused. Previous studies have shown that METH can induce damages to the nervous system and recent studies suggest that METH can also cause adverse and potentially lethal effects on the cardiovascular system. Recently, we demonstrated that DNA damage-inducible transcript 4 (DDIT4) regulates METH-induced neurotoxicity. However, the role of DDIT4 in METH-induced cardiotoxicity remains unknown. We hypothesized that DDIT4 may mediate METH-induced autophagy and apoptosis in cardiomyocytes. To test the hypothesis, we examined DDIT4 protein expression in cardiomyocytes and in heart tissues of rats exposed to METH with Western blotting. We also determined the effects on METH-induced autophagy and apoptosis after silencing DDIT4 expression with synthetic siRNA with or without pretreatment of a mTOR inhibitor rapamycin in cardiomyocytes using Western blot analysis, fluorescence microscopy and TUNEL staining. Our results showed that METH exposure increased DDIT4 expression and decreased phosphorylation of mTOR that was accompanied with increased autophagy and apoptosis both in vitro and in vivo. These effects were normalized after silencing DDIT4. On the other hand, rapamycin promoted METH-induced autophagy and apoptosis in DDIT4 knockdown cardiomyocytes. These results suggest that DDIT4 mediates METH-induced autophagy and apoptosis through mTOR signaling pathway in cardiomyocytes.
Collapse
Affiliation(s)
- Rui Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China; Department of Forensic Medicine, Guangdong Medical University, Dongguan 523808, People's Republic of China
| | - Bin Wang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Ling Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Dunpeng Cai
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Bing Li
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Chuanxiang Chen
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Enping Huang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China
| | - Chao Liu
- Guangzhou Forensic Science Institute, Guangzhou 510030, People's Republic of China
| | - Zhoumeng Lin
- Institute of Computational Comparative Medicine and Department of Anatomy and Physiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| | - Wei-Bing Xie
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China.
| | - Huijun Wang
- Department of Forensic Medicine, School of Basic Medical Science, Southern Medical University, Guangzhou 510515, People's Republic of China.
| |
Collapse
|
45
|
Yang W, Burkhardt B, Fischer L, Beirow M, Bork N, Wönne EC, Wagner C, Husen B, Zeilinger K, Liu L, Nussler AK. Age-dependent changes of the antioxidant system in rat livers are accompanied by altered MAPK activation and a decline in motor signaling. EXCLI JOURNAL 2015; 14:1273-1290. [PMID: 27004051 PMCID: PMC4800781 DOI: 10.17179/excli2015-734] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 12/10/2015] [Indexed: 12/15/2022]
Abstract
Aging is characterized by a progressive decrease of cellular functions, because cells gradually lose their capacity to respond to injury. Increased oxidative stress is considered to be one of the major contributors to age-related changes in all organs including the liver. Our study has focused on elucidating whether important antioxidative enzymes, the mTOR pathway, and MAPKs exhibit age-dependent changes in the liver of rats during aging. We found an age-dependent increase of GSH in the cytosol and mitochondria. The aged liver showed an increased SOD enzyme activity, while the CAT enzyme activity decreased. HO-1 and NOS-2 gene expression was lower in adult rats, but up-regulated in aged rats. Western blot analysis revealed that SOD1, SOD2, GPx, GR, γ-GCL, and GSS were age-dependent up-regulated, while CAT remained constant. We also demonstrated that the phosphorylation of Akt, JNK, p38, and TSC2(Ser1254) decreased while ERK1/2 and TSC2(Thr1462) increased age-dependently. Furthermore, our data show that the mTOR pathway seems to be activated in livers of aged rats, and hence stimulating cell proliferation/regeneration, as confirmed by an age-dependent increase of PCNA and p-eIF4E(Ser209) protein expression. Our data may help to explain the fact that liver cells only proliferate in cases of necessity, like injury and damage. In summary, we have demonstrated that, age-dependent changes of the antioxidant system and stress-related signaling pathways occur in the livers of rats, which may help to better understand organ aging.
Collapse
Affiliation(s)
- Wei Yang
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Britta Burkhardt
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Luise Fischer
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Maja Beirow
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Nadja Bork
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Eva C. Wönne
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus-Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Cornelia Wagner
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| | - Bettina Husen
- Pharmacelsus GmbH, Science Park 2, 66123 Saarbrücken, Germany
| | - Katrin Zeilinger
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Campus-Virchow-Klinikum, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Liegang Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Road, Wuhan 430030, PR China
| | - Andreas K. Nussler
- Eberhard Karls University Tübingen, Dept. of Traumatology, Schnarrenbergstr. 95, 72076 Tübingen, Germany
| |
Collapse
|
46
|
Cotecchia S, Del Vescovo CD, Colella M, Caso S, Diviani D. The alpha1-adrenergic receptors in cardiac hypertrophy: signaling mechanisms and functional implications. Cell Signal 2015; 27:1984-93. [PMID: 26169957 DOI: 10.1016/j.cellsig.2015.06.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 06/22/2015] [Accepted: 06/30/2015] [Indexed: 01/05/2023]
Abstract
Cardiac hypertrophy is a complex remodeling process of the heart induced by physiological or pathological stimuli resulting in increased cardiomyocyte size and myocardial mass. Whereas cardiac hypertrophy can be an adaptive mechanism to stressful conditions of the heart, prolonged hypertrophy can lead to heart failure which represents the primary cause of human morbidity and mortality. Among G protein-coupled receptors, the α1-adrenergic receptors (α1-ARs) play an important role in the development of cardiac hypertrophy as demonstrated by numerous studies in the past decades, both in primary cardiomyocyte cultures and genetically modified mice. The results of these studies have provided evidence of a large variety of α1-AR-induced signaling events contributing to the defining molecular and cellular features of cardiac hypertrophy. Recently, novel signaling mechanisms have been identified and new hypotheses have emerged concerning the functional role of the α1-adrenergic receptors in the heart. This review will summarize the main signaling pathways activated by the α1-AR in the heart and their functional implications in cardiac hypertrophy.
Collapse
Affiliation(s)
- Susanna Cotecchia
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy.
| | - Cosmo Damiano Del Vescovo
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Matilde Colella
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy
| | - Stefania Caso
- Dipartimento di Bioscienze, Biotecnologie e Biofarmaceutica, Università di Bari, Via Orabona 4, 70125 Bari, Italy; Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| | - Dario Diviani
- Department de Pharmacologie et de de Toxicologie, Université de Lausanne, Rue du Bugnon 27, 1005, Lausanne, Switzerland
| |
Collapse
|
47
|
Hadj Ayed Tka K, Mahfoudh Boussaid A, Zaouali MA, Kammoun R, Bejaoui M, Ghoul Mazgar S, Rosello Catafau J, Ben Abdennebi H. Melatonin modulates endoplasmic reticulum stress and Akt/GSK3-beta signaling pathway in a rat model of renal warm ischemia reperfusion. Anal Cell Pathol (Amst) 2015; 2015:635172. [PMID: 26229743 PMCID: PMC4502281 DOI: 10.1155/2015/635172] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 05/28/2015] [Indexed: 12/17/2022] Open
Abstract
Melatonin (Mel) is widely used to attenuate ischemia/reperfusion (I/R) injury in several organs. Nevertheless, the underlying mechanisms remain unclear. This study was conducted to explore the effect of Mel on endoplasmic reticulum (ER) stress, Akt and MAPK cascades after renal warm I/R. Eighteen Wistar rats were randomized into three groups: Sham, I/R, and Mel + I/R. The ischemia period was 60 min followed by 120 min of reperfusion. Mel (10 mg/kg) was administrated 30 min prior to ischemia. The creatinine clearance, MDA, LDH levels, and histopathological changes were evaluated. In addition, Western blot was performed to study ER stress and its downstream apoptosis as well as phosphorylation of Akt, GSK-3β, VDAC, ERK, and P38. Mel decreased cytolysis and lipid peroxidation and improved renal function and morphology compared to I/R group. Parallely, it significantly reduced the ER stress parameters including GRP 78, p-PERK, XBP 1, ATF 6, CHOP, and JNK. Simultaneously, p-Akt level was significantly enhanced and its target molecules GSK-3β and VDAC were inhibited. Furthermore, the ERK and P38 phosphorylation were evidently augmented after Mel administration in comparison to I/R group. In conclusion, Mel improves the recovery of renal function by decreasing ER stress and stimulating Akt pathway after renal I/R injury.
Collapse
Affiliation(s)
- Kaouther Hadj Ayed Tka
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Asma Mahfoudh Boussaid
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Mohamed Amine Zaouali
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Rym Kammoun
- Laboratory of Histology and Embryology, Faculty of Dental Medicine, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Mohamed Bejaoui
- Unit of Experimental Hepatic Ischemia-Reperfusion, Institute of Biomedical Investigations, Higher Council of Scientific Investigations, 08036 Barcelona, Spain
| | - Sonia Ghoul Mazgar
- Laboratory of Histology and Embryology, Faculty of Dental Medicine, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| | - Joan Rosello Catafau
- Unit of Experimental Hepatic Ischemia-Reperfusion, Institute of Biomedical Investigations, Higher Council of Scientific Investigations, 08036 Barcelona, Spain
| | - Hassen Ben Abdennebi
- Unit of Molecular Biology and Anthropology Applied to Development and Health (UR12ES11), Faculty of Pharmacy, University of Monastir, rue Avicenne, 5000 Monastir, Tunisia
| |
Collapse
|
48
|
Liu C, Xue R, Wu D, Wu L, Chen C, Tan W, Chen Y, Dong Y. REDD1 attenuates cardiac hypertrophy via enhancing autophagy. Biochem Biophys Res Commun 2014; 454:215-20. [PMID: 25450383 DOI: 10.1016/j.bbrc.2014.10.079] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 10/15/2014] [Indexed: 10/24/2022]
Abstract
Cardiac hypertrophy is a major risk factor of cardiovascular morbidity and mortality. Autophagy is established to be involved in regulating cardiac hypertrophy. REDD1, a stress-responsive protein, is proved to contribute in autophagy induction. However, the role of REDD1 in cardiac hypertrophy remains unknown. Our study demonstrated that REDD1 knockdown by RNAi exacerbated phenylephrine (PE)-induced cardiac hypertrophy, manifested by increased hypertrophic markers such as ANP and cell surface area. In addition, we discovered that ERK1/2 signaling pathway was involved in the effect of REDD1 on hypertrophy. Moreover, our study showed that REDD1 knockdown impaired autophagy in hypertrophied cardiomyocytes. mTOR, a signaling molecule governing autophagy induction, was activated by the knockdown of REDD1 under PE stress. Importantly, the pro-hypertrophic effect of REDD1 knockdown was significantly reversed by the autophagy enhancer rapamycin. Taken together, we firstly prove that REDD1 is essential for inhibiting cardiac hypertrophy by enhancing autophagy.
Collapse
Affiliation(s)
- Chen Liu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Ruicong Xue
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Dexi Wu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Lingling Wu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Cong Chen
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Weiping Tan
- Department of Respiratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China
| | - Yili Chen
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China
| | - Yugang Dong
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou 510080, China.
| |
Collapse
|
49
|
Canal M, Romaní-Aumedes J, Martín-Flores N, Pérez-Fernández V, Malagelada C. RTP801/REDD1: a stress coping regulator that turns into a troublemaker in neurodegenerative disorders. Front Cell Neurosci 2014; 8:313. [PMID: 25324725 PMCID: PMC4183088 DOI: 10.3389/fncel.2014.00313] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2014] [Accepted: 09/17/2014] [Indexed: 12/16/2022] Open
Abstract
Mechanistic target of Rapamycin (mTOR) pathway regulates essential processes directed to preserve cellular homeostasis, such as cell growth, proliferation, survival, protein synthesis and autophagy. Importantly, mTOR pathway deregulation has been related to many diseases. Indeed, it has become a hallmark in neurodegenerative disorders, since a fine-tuned regulation of mTOR activities is crucial for neuron function and survival. RTP801/REDD1/Dig2 has become one of the most puzzling regulators of mTOR. Although the mechanism is not completely understood, RTP801 inactivates mTOR and Akt via the tuberous sclerosis complex (TSC1/TSC2) in many cellular contexts. Intriguingly, RTP801 protects dividing cells from hypoxia or H2O2-induced apoptosis, while it sensitizes differentiated cells to stress. Based on experimental models of Parkinson’s disease (PD), it has been proposed that at early stages of the disease, stress-induced RTP801 upregulation contributes to mTOR repression, in an attempt to maintain cell function and viability. However, if RTP801 elevation is sustained, it leads to neuron cell death by a sequential inhibition of mTOR and Akt. Here, we will review RTP801 deregulation of mTOR in a context of PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Mercè Canal
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona Barcelona, Catalonia, Spain
| | - Joan Romaní-Aumedes
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona Barcelona, Catalonia, Spain
| | - Núria Martín-Flores
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona Barcelona, Catalonia, Spain
| | - Víctor Pérez-Fernández
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona Barcelona, Catalonia, Spain
| | - Cristina Malagelada
- Department of Pathological Anatomy, Pharmacology and Microbiology, Faculty of Medicine, University of Barcelona Barcelona, Catalonia, Spain
| |
Collapse
|
50
|
Urocortin-2 suppression of p38-MAPK signaling as an additional mechanism for ischemic cardioprotection. Mol Cell Biochem 2014; 398:135-46. [DOI: 10.1007/s11010-014-2213-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 09/12/2014] [Indexed: 11/26/2022]
|