1
|
HELVACIOĞLU S, HAMİTOĞLU M, YILDIRIM E, KORKUT ŞVURAL, YABA A, AYDIN A. Protective Effects of Rosmarinic Acid and Epigallocatechin Gallate Against Doxorubicin-Induced Cytotoxicity and Genotoxicity in CHO-K1 Cells. Turk J Pharm Sci 2025; 21:536-543. [PMID: 39801063 PMCID: PMC11730000 DOI: 10.4274/tjps.galenos.2024.80552] [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: 12/28/2023] [Accepted: 02/20/2024] [Indexed: 01/16/2025]
Abstract
Objectives The chemotherapeutic drug doxorubicin (DOX) affects not only cancer cells but also healthy cells in an undesirable manner. The purpose of this study was to investigate the protective roles of rosmarinic acid (RA) and Epigallocatechin gallate (EGCG) alone and in combination against DOX-induced oxidative stress, cytotoxicity, and genotoxicity in healthy cells. In addition, this study evaluated the expression of the mammalian target of rapamycin (mTOR) protein in the Chinese hamster ovary cell line (CHO-K1). Materials and Methods Cell viability was analyzed using the WST-1 cytotoxicity assay. mTOR expression in the CHO-K1 cell line was determined by western blotting. DNA damage was analyzed using a comet assay. Reactive oxygen species (ROS) levels were determined microscopically using the dihydroethidium staining method. Results RA demonstrated superior protective effects against DOX-induced cytotoxicity compared to EGCG. Epigallocatechin gallate and RA did not exert genotoxic effects, but DOX increased genotoxicity in CHO-K1. Neither RA nor EGCG exhibited genotoxic effects; however, DOX significantly increased genotoxicity in CHO-K1 cells. Both RA and EGCG markedly reduced DOX-induced genotoxicity, as confirmed by the comet assay. In the DOX-treated group, the expression of mTOR protein was notably suppressed. EGCG further reduced mTOR protein levels when administered alone or in combination with DOX, whereas RA did not exhibit a similar effect. RA decreased intracellular generation of ROS in CHO-K1 cells. However, at high concentrations, Epigallocatechin gallate did not protect against oxidative stress and cell damage due to its prooxidant properties. Conclusion Epigallocatechin gallate and RA are promising plant-derived active components. Another important point is the evaluation of the safety of herbal products. It should be considered that herbal products may increase the toxicity of chemotherapeutic agents.
Collapse
Affiliation(s)
- Sinem HELVACIOĞLU
- İstinye University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| | - Muhammed HAMİTOĞLU
- Yeditepe University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| | - Ecem YILDIRIM
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Şenay VURAL KORKUT
- Yıldız Technical University Faculty of Arts and Science, Department of Molecular Biology and Genetics, İstanbul, Türkiye
| | - Aylin YABA
- Yeditepe University Faculty of Medicine, Department of Histology and Embryology, İstanbul, Türkiye
| | - Ahmet AYDIN
- Yeditepe University Faculty of Pharmacy, Department of Pharmaceutical Toxicology, İstanbul, Türkiye
| |
Collapse
|
2
|
Scepanovic G, Balaghi N, Rothenberg KE, Fernandez-Gonzalez R. mTor limits autophagy to facilitate cell volume expansion and rapid wound repair in Drosophila embryos. Dev Cell 2025:S1534-5807(24)00778-0. [PMID: 39824179 DOI: 10.1016/j.devcel.2024.12.039] [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: 10/03/2023] [Revised: 07/16/2024] [Accepted: 12/23/2024] [Indexed: 01/20/2025]
Abstract
Embryonic wounds repair rapidly, with no inflammation or scarring. Embryonic wound healing is driven by collective cell movements facilitated by the increase in the volume of the cells adjacent to the wound. The mechanistic target of rapamycin (mTor) complex 1 (TORC1) is associated with cell growth. We found that disrupting TORC1 signaling in Drosophila embryos prevented cell volume increases and slowed down wound repair. Catabolic processes, such as autophagy, can inhibit cell growth. Five-dimensional microscopy demonstrated that the number of autophagosomes decreased during wound repair, suggesting that autophagy must be tightly regulated for rapid wound healing. mTor inhibition increased autophagy, and activating autophagy prevented cell volume expansion and slowed down wound closure. Finally, reducing autophagy in embryos with disrupted TORC1 signaling rescued cell volume changes and rapid wound repair. Together, our results show that TORC1 activation upon wounding negatively regulates autophagy, allowing cells to increase their volumes to facilitate rapid wound healing.
Collapse
Affiliation(s)
- Gordana Scepanovic
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Negar Balaghi
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Katheryn E Rothenberg
- Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada
| | - Rodrigo Fernandez-Gonzalez
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada; Ted Rogers Centre for Heart Research, University of Toronto, Toronto, ON M5G 1M1, Canada; Institute of Biomedical Engineering, University of Toronto, Toronto, ON M5S 3G9, Canada; Developmental and Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.
| |
Collapse
|
3
|
Islam MI, Sultana S, Padmanabhan N, Rashid MU, Siddiqui TJ, Coombs KM, Vitiello PF, Karimi-Abdolrezaee S, Eftekharpour E. Thioredoxin-1 protein interactions in neuronal survival and neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167548. [PMID: 39454970 DOI: 10.1016/j.bbadis.2024.167548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024]
Abstract
Neuronal cell death remains the principal pathophysiologic hallmark of neurodegenerative diseases and the main challenge for treatment strategies. Thioredoxin1 (Trx1) is a major cytoplasmic thiol oxidoreductase protein involved in redox signaling, hence a crucial player in maintaining neuronal health. Trx1 levels are notably reduced in neurodegenerative diseases including Alzheimer's and Parkinson's diseases, however, the impact of this decrease on neuronal physiology remains largely unexplored. This is mainly due to the nature of Trx1 redox regulatory role which is afforded by a rapid electron transfer to its oxidized protein substrates. During this reaction, Trx1 forms a transient bond with the oxidized disulfide bond in the substrate. This is a highly fast reaction which makes the identification of Trx1 substrates a technically challenging task. In this project, we utilized a transgenic mouse model expressing a Flag-tagged mutant form of Trx1 that can form stable disulfide bonds with its substrates, hence allowing identification of the Trx1 target proteins. Autophagy is a vital housekeeping process in neurons that is critical for degradation of damaged proteins under oxidative stress conditions and is interrupted in neurodegenerative diseases. Given Trx1's suggested involvement in autophagy, we aimed to identify potential Trx1 substrates following pharmacologic induction of autophagy in primary cortical neurons. Treatment with rapamycin, an autophagy inducer, significantly reduced neurite outgrowth and caused cytoskeletal alterations. Using immunoprecipitation and mass spectrometry, we have identified 77 Trx1 target proteins associated with a wide range of cellular functions including cytoskeletal organization and neurodegenerative diseases. Focusing on neuronal cytoskeleton organization, we identified a novel interaction between Trx1 and RhoB which was confirmed in genetic models of Trx1 downregulation in primary neuronal cultures and HT22 mouse immortalized hippocampal neurons. The applicability of these findings was also tested against the publicly available proteomic data from Alzheimer's patients. Our study uncovers a novel role for Trx1 in regulating neuronal cytoskeleton organization and provides a mechanistic explanation for its multifaceted role in the physiology and pathology of the nervous system, offering new insights into the molecular mechanisms underlying neurodegeneration.
Collapse
Affiliation(s)
- Md Imamul Islam
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Shakila Sultana
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Nirmala Padmanabhan
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | | | - Tabrez J Siddiqui
- Department of Physiology and Pathophysiology, University of Manitoba, Canada
| | - Kevin M Coombs
- Department of Medical Microbiology, University of Manitoba, Canada
| | - Peter F Vitiello
- Department of Pediatrics, the University of Oklahoma Health Sciences Center, USA
| | | | | |
Collapse
|
4
|
Glorieux C, Enríquez C, Buc Calderon P. The complex interplay between redox dysregulation and mTOR signaling pathway in cancer: A rationale for cancer treatment. Biochem Pharmacol 2024; 232:116729. [PMID: 39709038 DOI: 10.1016/j.bcp.2024.116729] [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: 10/21/2024] [Revised: 12/09/2024] [Accepted: 12/19/2024] [Indexed: 12/23/2024]
Abstract
The mechanistic target of rapamycin (mTOR) is a highly conserved serine/threonine kinase that plays a critical role in regulating cellular processes such as growth, proliferation, and metabolism in healthy cells. Dysregulation of mTOR signaling and oxidative stress have been implicated in various diseases including cancer. This review aims to provide an overview of the current understanding of mTOR and its involvement in cell survival and the regulation of cancer cell metabolism as well as its complex interplay with reactive oxygen species (ROS). On the one hand, ROS can inhibit or activate mTOR pathway in cancer cells through various mechanisms. Conversely, mTOR signaling can induce oxidative stress in tumor cells notably due to the inhibition in the expression of antioxidant enzyme genes. Since mTOR is often activated and plays crucial role in cancer cell survival, the use of mTOR inhibitors, which often induce ROS accumulation, could be an interesting approach for cancer treatment. This review will address the advantages, disadvantages, combination strategies, and limitations associated with therapeutic modulation of mTOR signaling pathway in cancer treatment.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, China.
| | - Cinthya Enríquez
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Programa de Doctorado en Química Medicinal, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, 1100000 Iquique, Chile; Instituto de Química Medicinal, Universidad Arturo Prat, 1100000 Iquique, Chile; Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, 1200 Brussels, Belgium.
| |
Collapse
|
5
|
Kim YS, Kimball SR, Piskounova E, Begley TJ, Hempel N. Stress response regulation of mRNA translation: Implications for antioxidant enzyme expression in cancer. Proc Natl Acad Sci U S A 2024; 121:e2317846121. [PMID: 39495917 PMCID: PMC11572934 DOI: 10.1073/pnas.2317846121] [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] [Indexed: 11/06/2024] Open
Abstract
From tumorigenesis to advanced metastatic stages, tumor cells encounter stress, ranging from limited nutrient and oxygen supply within the tumor microenvironment to extrinsic and intrinsic oxidative stress. Thus, tumor cells seize regulatory pathways to rapidly adapt to distinct physiologic conditions to promote cellular survival, including manipulation of mRNA translation. While it is now well established that metastatic tumor cells must up-regulate their antioxidant capacity to effectively spread and that regulation of antioxidant enzymes is imperative to disease progression, relatively few studies have assessed how translation and the hijacking of RNA systems contribute to antioxidant responses of tumors. Here, we review the major stress signaling pathways involved in translational regulation and discuss how these are affected by oxidative stress to promote prosurvival changes that manipulate antioxidant enzyme expression. We describe how tumors elicit these adaptive responses and detail how stress-induced translation can be regulated by kinases, RNA-binding proteins, RNA species, and RNA modification systems. We also highlight opportunities for further studies focused on the role of mRNA translation and RNA systems in the regulation of antioxidant enzyme expression, which may be of particular importance in the context of metastatic progression and therapeutic resistance.
Collapse
Affiliation(s)
- Yeon Soo Kim
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey, PA17033
| | - Scot R. Kimball
- Department of Cellular and Molecular Physiology, College of Medicine, Pennsylvania State University, Hershey, PA17033
| | - Elena Piskounova
- Department of Dermatology, Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Thomas J. Begley
- The RNA Institute and Department of Biological Sciences, University at Albany, Albany, NY12222
| | - Nadine Hempel
- Department of Medicine, Division of Hematology/Oncology, University of Pittsburgh School of Medicine, Pittsburgh, PA15213
| |
Collapse
|
6
|
Duong LD, West JD, Morano KA. Redox regulation of proteostasis. J Biol Chem 2024; 300:107977. [PMID: 39522946 DOI: 10.1016/j.jbc.2024.107977] [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: 07/02/2024] [Revised: 10/22/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Oxidants produced through endogenous metabolism or encountered in the environment react directly with reactive sites in biological macromolecules. Many proteins, in particular, are susceptible to oxidative damage, which can lead to their altered structure and function. Such structural and functional changes trigger a cascade of events that influence key components of the proteostasis network. Here, we highlight recent advances in our understanding of how cells respond to the challenges of protein folding and metabolic alterations that occur during oxidative stress. Immediately after an oxidative insult, cells selectively block the translation of most new proteins and shift molecular chaperones from folding to a holding role to prevent wholesale protein aggregation. At the same time, adaptive responses in gene expression are induced, allowing for increased expression of antioxidant enzymes, enzymes that carry out the reduction of oxidized proteins, and molecular chaperones, all of which serve to mitigate oxidative damage and rebalance proteostasis. Likewise, concomitant activation of protein clearance mechanisms, namely proteasomal degradation and particular autophagic pathways, promotes the degradation of irreparably damaged proteins. As oxidative stress is associated with inflammation, aging, and numerous age-related disorders, the molecular events described herein are therefore major determinants of health and disease.
Collapse
Affiliation(s)
- Long Duy Duong
- Department of Microbiology & Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - James D West
- Biochemistry & Molecular Biology Program, Departments of Biology and Chemistry, The College of Wooster, Wooster, Ohio, USA.
| | - Kevin A Morano
- Department of Microbiology & Molecular Genetics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.
| |
Collapse
|
7
|
Yu Y, Poulsen SA, Di Trapani G, Tonissen KF. Exploring the Redox and pH Dimension of Carbonic Anhydrases in Cancer: A Focus on Carbonic Anhydrase 3. Antioxid Redox Signal 2024; 41:957-975. [PMID: 38970427 DOI: 10.1089/ars.2024.0693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/08/2024]
Abstract
Significance: Both redox and pH are important regulatory processes that underpin cell physiological functions, in addition to influencing cancer cell development and tumor progression. The thioredoxin (Trx) and glutathione redox systems and the carbonic anhydrase (CA) proteins are considered key regulators of cellular redox and pH, respectively, with components of the Trx system and CAs regarded as cancer therapeutic targets. However, the redox and pH axis in cancer cells is an underexplored topic of research. Recent Advances: Structural studies of a CA family member, CA3, localized two of its five cysteine residues to the protein surface. Redox-regulated modifications to CA3 have been identified, including glutathionylation. CA3 has been shown to bind to other proteins, including B cell lymphoma-2-associated athanogene 3, and squalene epoxidase, which can modulate autophagy and proinflammatory signaling, respectively, in cancer cells. Critical Issues: CA3 has also been associated with epithelial-mesenchymal transition processes, which promote cancer cell metastasis, whereas CA3 overexpression activates the phosphatidylinositol-3 kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway, which upregulates cell growth and inhibits autophagy. It is not yet known if CA3 modulates cancer progression through its reported antioxidant functions. Future Directions: CA3 is one of the least studied CA isozymes. Further studies are required to assess the cellular antioxidant role of CA3 and its impact on cancer progression. Identification of other binding partners is also required, including whether CA3 binds to Trx in human cells. The development of specific CA3 inhibitors will facilitate these functional studies and allow CA3 to be investigated as a cancer therapeutic target. Antioxid. Redox Signal. 41, 957-975.
Collapse
Affiliation(s)
- Yezhou Yu
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Australia
- School of Environment and Science, Griffith University, Nathan, Australia
| | - Sally-Ann Poulsen
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Australia
- School of Environment and Science, Griffith University, Nathan, Australia
| | | | - Kathryn F Tonissen
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Australia
- School of Environment and Science, Griffith University, Nathan, Australia
| |
Collapse
|
8
|
Cui M, Chen F, Shao L, Wei C, Zhang W, Sun W, Wang J. Mesenchymal stem cells and ferroptosis: Clinical opportunities and challenges. Heliyon 2024; 10:e25251. [PMID: 38356500 PMCID: PMC10864896 DOI: 10.1016/j.heliyon.2024.e25251] [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: 05/20/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Objective This review discusses recent experimental and clinical findings related to ferroptosis, with a focus on the role of MSCs. Therapeutic efficacy and current applications of MSC-based ferroptosis therapies are also discussed. Background Ferroptosis is a type of programmed cell death that differs from apoptosis, necrosis, and autophagy; it involves iron metabolism and is related to the pathogenesis of many diseases, such as Parkinson's disease, cancers, and liver diseases. In recent years, the use of mesenchymal stem cells (MSCs) and MSC-derived exosomes has become a trend in cell-free therapies. MSCs are a heterogeneous cell population isolated from a diverse range of human tissues that exhibit immunomodulatory functions, regulate cell growth, and repair damaged tissues. In addition, accumulating evidence indicates that MSC-derived exosomes play an important role, mainly by carrying a variety of bioactive substances that affect recipient cells. The potential mechanism by which MSC-derived exosomes mediate the effects of MSCs on ferroptosis has been previously demonstrated. This review provides the first overview of the current knowledge on ferroptosis, MSCs, and MSC-derived exosomes and highlights the potential application of MSCs exosomes in the treatment of ferroptotic conditions. It summarizes their mechanisms of action and techniques for enhancing MSC functionality. Results obtained from a large number of experimental studies revealed that both local and systemic administration of MSCs effectively suppressed ferroptosis in injured hepatocytes, neurons, cardiomyocytes, and nucleus pulposus cells and promoted the survival and regeneration of injured organs. Methods We reviewed the role of ferroptosis in related tissues and organs, focusing on its characteristics in different diseases. Additionally, the effects of MSCs and MSC-derived exosomes on ferroptosis-related pathways in various organs were reviewed, and the mechanism of action was elucidated. MSCs were shown to improve the disease course by regulating ferroptosis.
Collapse
Affiliation(s)
- Mengling Cui
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Fukun Chen
- Department of Radiology, Kunming Medical University & the Third Affiliated Hospital, Kunming, Yunnan, 650101, PR China
| | - Lishi Shao
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Chanyan Wei
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Weihu Zhang
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Wenmei Sun
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| | - Jiaping Wang
- Department of Radiology, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, 650101, PR China
| |
Collapse
|
9
|
Nicco C, Thomas M, Guillermet J, Havard M, Laurent-Tchenio F, Doridot L, Dautry F, Batteux F, Tchenio T. Mechanistic target of rapamycin (mTOR) regulates self-sustained quiescence, tumor indolence, and late clinical metastasis in a Beclin-1-dependent manner. Cell Cycle 2023; 22:542-564. [PMID: 36123968 PMCID: PMC9928463 DOI: 10.1080/15384101.2022.2123187] [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] [Indexed: 01/10/2023] Open
Abstract
Self-sustained quiescence (SSQ) has been characterized as a stable but reversible non-proliferative cellular state that limits the cloning of cultured cancer cells. By developing refined clonogenic assays, we showed here that cancer cells in SSQ can be selected with anticancer agents and that culture at low cell density induced SSQ in pancreas and prostate adenocarcinoma cells. Pre-culture of cells in 3D or their pretreatment with pharmacological inhibitors of mechanistic target of rapamycin (mTOR) synergize with low cell density for induction of SSQ in a Beclin-1-dependent manner. Dissociated pancreatic adenocarcinoma (PAAD) cells rendered defective for SSQ by down-regulating Beclin-1 expression exhibit higher tumor growth rate when injected subcutaneously into mice. Conversely, dissociated PAAD cells in SSQ promote the formation of small indolent tumors that eventually transitioned to a rapid growth phase. Ex vivo clonogenic assays showed that up to 40% of clonogenic cancer cells enzymatically dissociated from resected fast-growing tumors could enter SSQ, suggesting that SSQ could significantly impact the proliferation of cancer cells that are naturally dispersed from tumors. Remarkably, the kinetics of clinical metastatic recurrence in 124 patients with pancreatic adenocarcinoma included in the TGCA-PAAD project could be predicted from Beclin-1 and Cyclin-A2 mRNA levels in their primary tumor, Cyclin A2 mRNA being a marker of both cell proliferation and mTOR complex 1 activity. Overall, our data show that SSQ is likely to promote the late development of clinical metastases and suggest that identifying new agents targeting cancer cells in SSQ could help improve patient survival.
Collapse
Affiliation(s)
- Carole Nicco
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Marine Thomas
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Julie Guillermet
- Centre de Recherches en Cancérologie de Toulouse (CRCT), Inserm U1037, CNRS U5071, Université Toulouse III, Toulouse, France
| | - Maryline Havard
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Fanny Laurent-Tchenio
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Ludivine Doridot
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - François Dautry
- Laboratory of Biology and Applied Pharmacology (LBPA), CNRS UMR8113, IDA FR3242, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - Frédéric Batteux
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| | - Thierry Tchenio
- Institut Cochin, INSERM U1016/CNRS UMR 8104, Université de Paris, Paris, France
| |
Collapse
|
10
|
Nagarajan N, Oka SI, Nah J, Wu C, Zhai P, Mukai R, Xu X, Kashyap S, Huang CY, Sung EA, Mizushima W, Titus AS, Takayama K, Mourad Y, Francisco J, Liu T, Chen T, Li H, Sadoshima J. Thioredoxin 1 promotes autophagy through transnitrosylation of Atg7 during myocardial ischemia. J Clin Invest 2023; 133:e162326. [PMID: 36480290 PMCID: PMC9888389 DOI: 10.1172/jci162326] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Modification of cysteine residues by oxidative and nitrosative stress affects structure and function of proteins, thereby contributing to the pathogenesis of cardiovascular disease. Although the major function of thioredoxin 1 (Trx1) is to reduce disulfide bonds, it can also act as either a denitrosylase or transnitrosylase in a context-dependent manner. Here we show that Trx1 transnitrosylates Atg7, an E1-like enzyme, thereby stimulating autophagy. During ischemia, Trx1 was oxidized at Cys32-Cys35 of the oxidoreductase catalytic center and S-nitrosylated at Cys73. Unexpectedly, Atg7 Cys545-Cys548 reduced the disulfide bond in Trx1 at Cys32-Cys35 through thiol-disulfide exchange and this then allowed NO to be released from Cys73 in Trx1 and transferred to Atg7 at Cys402. Experiments conducted with Atg7 C402S-knockin mice showed that S-nitrosylation of Atg7 at Cys402 promotes autophagy by stimulating E1-like activity, thereby protecting the heart against ischemia. These results suggest that the thiol-disulfide exchange and the NO transfer are functionally coupled, allowing oxidized Trx1 to mediate a salutary effect during myocardial ischemia through transnitrosylation of Atg7 and stimulation of autophagy.
Collapse
Affiliation(s)
- Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Changgong Wu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Koichiro Takayama
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Tong Chen
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
11
|
Yu Y, Wu T, Lu Y, Zhao W, Zhang J, Chen Q, Ge G, Hua Y, Chen K, Ullah I, Zhang F. Exosomal thioredoxin-1 from hypoxic human umbilical cord mesenchymal stem cells inhibits ferroptosis in doxorubicin-induced cardiotoxicity via mTORC1 signaling. Free Radic Biol Med 2022; 193:108-121. [PMID: 36241072 DOI: 10.1016/j.freeradbiomed.2022.10.268] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/13/2022]
Abstract
Doxorubicin (DOX), a clinical chemotherapeutic drug, is often annoyed by its cardiotoxicity which involves ferroptosis in its pathological progress. Human umbilical cord mesenchymal stem cells (HucMSCs)-derived exosomes (HucMSCs-Exo) are proven effective in treating cardiovascular diseases. This study aimed to compare the therapeutic effects between normoxic HucMSCs-Exo (Exo) and hypoxic HucMSCs-Exo (Hypo-Exo) on DOX-induced ferroptosis and explore the underlying mechanisms. An acute cardiotoxicity model was successfully constructed by administrating two doses intraperitoneal injections of DOX (25 mg/kg in total). Exo and Hypo-Exo were extracted by ultracentrifugation and characterized. Compared with Exo, Hypo-Exo and Ferrostatin-1 (Fer-1) exerted superior effects on inhibiting DOX-induced ferroptosis, as evidenced by decreasing malondialdehyde (MDA), iron content and increasing glutathione (GSH) level as well as ferroptosis-related genes expression including prostaglandin-endoperoxide synthase 2 (Ptgs2) mRNA level and glutathione peroxidase 4 (GPX4) protein level. Based on quantitative proteomics analysis, we found that thioredoxin1 (Trx1) was remarkably upregulated in Hypo-Exo and exhibited anti-ferroptosis activity via activating the mechanistic target of rapamycin complex 1 (mTORC1) in neonatal rat cardiomyocytes (NRCMs). Trx1 knockdown and rapamycin (an mTORC1 inhibitor) partially abolished the protective effects of Hypo-Exo. Furthermore, our data indicated that solute carrier family 7 member 11 (SLC7A11) was critical for GPX4 protein synthesis. In conclusion, Hypo-Exo exhibited a better suppression of ferroptosis in DOX-induced cardiotoxicity. Trx1-mediated mTORC1 activation is critical for the Hypo-Exo anti-ferroptosis process, which involves increased GPX4 protein synthesis and decreased iron overload. This study indicated that Hypo-Exo may present a potential strategy against ferroptosis in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yue Yu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianyu Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yao Lu
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Nanjing Medical University, Xuzhou, 221009, Jiangsu, China
| | - Wei Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qiushi Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Gaoyuan Ge
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yan Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Kaiyan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Inam Ullah
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Fengxiang Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
12
|
Thioredoxin deficiency increases oxidative stress and causes bilateral symmetrical degeneration in rat midbrain. Neurobiol Dis 2022; 175:105921. [DOI: 10.1016/j.nbd.2022.105921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/26/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
|
13
|
Zhu J, Wang H, Jiang X. mTORC1 beyond anabolic metabolism: Regulation of cell death. J Biophys Biochem Cytol 2022; 221:213609. [PMID: 36282248 PMCID: PMC9606688 DOI: 10.1083/jcb.202208103] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1), a multi-subunit protein kinase complex, interrogates growth factor signaling with cellular nutrient and energy status to control metabolic homeostasis. Activation of mTORC1 promotes biosynthesis of macromolecules, including proteins, lipids, and nucleic acids, and simultaneously suppresses catabolic processes such as lysosomal degradation of self-constituents and extracellular components. Metabolic regulation has emerged as a critical determinant of various cellular death programs, including apoptosis, pyroptosis, and ferroptosis. In this article, we review the expanding knowledge on how mTORC1 coordinates metabolic pathways to impinge on cell death regulation. We focus on the current understanding on how nutrient status and cellular signaling pathways connect mTORC1 activity with ferroptosis, an iron-dependent cell death program that has been implicated in a plethora of human diseases. In-depth understanding of the principles governing the interaction between mTORC1 and cell death pathways can ultimately guide the development of novel therapies for the treatment of relevant pathological conditions.
Collapse
Affiliation(s)
- Jiajun Zhu
- Department of Basic Medical Sciences, School of Medicine, Tsinghua University, Beijing, China,Tsinghua-Peking Center for Life Sciences, Beijing, China,Correspondence to Jiajun Zhu:
| | - Hua Wang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY,Xuejun Jiang:
| |
Collapse
|
14
|
Morris G, Gevezova M, Sarafian V, Maes M. Redox regulation of the immune response. Cell Mol Immunol 2022; 19:1079-1101. [PMID: 36056148 PMCID: PMC9508259 DOI: 10.1038/s41423-022-00902-0] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/29/2022] [Indexed: 12/20/2022] Open
Abstract
AbstractThe immune-inflammatory response is associated with increased nitro-oxidative stress. The aim of this mechanistic review is to examine: (a) the role of redox-sensitive transcription factors and enzymes, ROS/RNS production, and the activity of cellular antioxidants in the activation and performance of macrophages, dendritic cells, neutrophils, T-cells, B-cells, and natural killer cells; (b) the involvement of high-density lipoprotein (HDL), apolipoprotein A1 (ApoA1), paraoxonase-1 (PON1), and oxidized phospholipids in regulating the immune response; and (c) the detrimental effects of hypernitrosylation and chronic nitro-oxidative stress on the immune response. The redox changes during immune-inflammatory responses are orchestrated by the actions of nuclear factor-κB, HIF1α, the mechanistic target of rapamycin, the phosphatidylinositol 3-kinase/protein kinase B signaling pathway, mitogen-activated protein kinases, 5' AMP-activated protein kinase, and peroxisome proliferator-activated receptor. The performance and survival of individual immune cells is under redox control and depends on intracellular and extracellular levels of ROS/RNS. They are heavily influenced by cellular antioxidants including the glutathione and thioredoxin systems, nuclear factor erythroid 2-related factor 2, and the HDL/ApoA1/PON1 complex. Chronic nitro-oxidative stress and hypernitrosylation inhibit the activity of those antioxidant systems, the tricarboxylic acid cycle, mitochondrial functions, and the metabolism of immune cells. In conclusion, redox-associated mechanisms modulate metabolic reprogramming of immune cells, macrophage and T helper cell polarization, phagocytosis, production of pro- versus anti-inflammatory cytokines, immune training and tolerance, chemotaxis, pathogen sensing, antiviral and antibacterial effects, Toll-like receptor activity, and endotoxin tolerance.
Collapse
|
15
|
Huang CY, Oka SI, Xu X, Chen CF, Tung CY, Chang YY, Mourad Y, Vehra O, Ivessa A, Yehia G, Romanienko P, Hsu CP, Sadoshima J. PERM1 regulates genes involved in fatty acid metabolism in the heart by interacting with PPARα and PGC-1α. Sci Rep 2022; 12:14576. [PMID: 36028747 PMCID: PMC9418182 DOI: 10.1038/s41598-022-18885-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 08/22/2022] [Indexed: 11/09/2022] Open
Abstract
PERM1 (PGC-1/ERR-induced regulator in muscle 1) is a muscle-specific protein induced by PGC-1 and ERRs. Previous studies have shown that PERM1 promotes mitochondrial biogenesis and metabolism in cardiomyocytes in vitro. However, the role of endogenous PERM1 in the heart remains to be investigated with loss-of-function studies in vivo. We report the generation and characterization of systemic Perm1 knockout (KO) mice. The baseline cardiac phenotype of the homozygous Perm1 KO mice appeared normal. However, RNA-sequencing and unbiased pathway analyses showed that homozygous downregulation of PERM1 leads to downregulation of genes involved in fatty acid and carbohydrate metabolism in the heart. Transcription factor binding site analyses suggested that PPARα and PGC-1α are involved in changes in the gene expression profile. Chromatin immunoprecipitation assays showed that PERM1 interacts with the proximal regions of PPAR response elements (PPREs) in endogenous promoters of genes involved in fatty acid oxidation. Co-immunoprecipitation and reporter gene assays showed that PERM1 promoted transcription via the PPRE, partly in a PPARα and PGC-1α dependent manner. These results suggest that endogenous PERM1 is involved in the transcription of genes involved in fatty acid oxidation through physical interaction with PPARα and PGC-1α in the heart in vivo.
Collapse
Affiliation(s)
- Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.,Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.,Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Chian-Feng Chen
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Chien-Yi Tung
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Ya-Yuan Chang
- Cancer Progression Research Center, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Omair Vehra
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Andreas Ivessa
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA
| | - Ghassan Yehia
- Genome Editing Core Facility, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Peter Romanienko
- Genome Editing Core Facility, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, 08901, USA
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Medicine, School of Medicine, National Yang-Ming Chiao-Tung University, Taipei, Taiwan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, 185 South Orange Ave., MSB G609, Newark, NJ, 07103, USA.
| |
Collapse
|
16
|
Inflammation Promotes Oxidative and Nitrosative Stress in Chronic Myelogenous Leukemia. Biomolecules 2022; 12:biom12020247. [PMID: 35204748 PMCID: PMC8961589 DOI: 10.3390/biom12020247] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 11/16/2022] Open
Abstract
Chronic inflammation is characterized by the production of reactive oxygen species (ROS), reactive nitrogen species, and inflammatory cytokines in myeloproliferative neoplasms (MPNs). In addition to these parameters, the aim of this study was to analyze the influence of ROS on the proliferation-related AKT/mTOR signaling pathway and the relationship with inflammatory factors in chronic myelogenous leukemia (CML). The activity of the antioxidant enzymes superoxide dismutase, glutathione peroxidase, and catalase is reduced in erythrocytes while levels of the oxidative stress markers malondialdehyde and protein carbonyl are elevated in the plasma of patients with CML. In addition, nitrogen species (nitrotyrosine, iNOS, eNOS) and inflammation markers (IL-6, NFkB, and S100 protein) were increased in granulocytes of CML while anti-inflammatory levels of IL-10 were decreased in plasma. CML granulocytes exhibited greater resistance to cytotoxic H2O2 activity compared to healthy subjects. Moreover, phosphorylation of the apoptotic p53 protein was reduced while the activity of the AKT/mTOR signaling pathway was increased, which was further enhanced by oxidative stress (H2O2) in granulocytes and erythroleukemic K562 cells. IL-6 caused oxidative stress and DNA damage that was mitigated using antioxidant or inhibition of inflammatory NFkB transcription factor in K562 cells. We demonstrated the presence of oxidative and nitrosative stress in CML, with the former mediated by AKT/mTOR signaling and stimulated by inflammation.
Collapse
|
17
|
Autophagy Is Involved in the Viability of Overexpressing Thioredoxin o1 Tobacco BY-2 Cells under Oxidative Conditions. Antioxidants (Basel) 2021; 10:antiox10121884. [PMID: 34942987 PMCID: PMC8698322 DOI: 10.3390/antiox10121884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 11/17/2021] [Accepted: 11/22/2021] [Indexed: 01/06/2023] Open
Abstract
Autophagy is an essential process for the degradation of non-useful components, although the mechanism involved in its regulation is less known in plants than in animal systems. Redox regulation of autophagy components is emerging as a possible key mechanism with thioredoxins (TRXs) proposed as involved candidates. In this work, using overexpressing PsTRXo1 tobacco cells (OEX), which present higher viability than non-overexpressing cells after H2O2 treatment, we examine the functional interaction of autophagy and PsTRXo1 in a collaborative response. OEX cells present higher gene expression of the ATG (Autophagy related) marker ATG4 and higher protein content of ATG4, ATG8, and lipidated ATG8 as well as higher ATG4 activity than control cells, supporting the involvement of autophagy in their response to H2O2. In this oxidative situation, autophagy occurs in OEX cells as is evident from an accumulation of autolysosomes and ATG8 immunolocalization when the E-64d autophagy inhibitor is used. Interestingly, cell viability decreases in the presence of the inhibitor, pointing to autophagy as being involved in cell survival. The in vitro interaction of ATG4 and PsTRXo1 proteins is confirmed by dot-blot and co-immunoprecipitation assays as well as the redox regulation of ATG4 activity by PsTRXo1. These findings extend the role of TRXs in mediating the redox regulation of the autophagy process in plant cells.
Collapse
|
18
|
Oka SI, Tang F, Chin A, Ralda G, Xu X, Hu C, Yang Z, Abdellatif M, Sadoshima J. β-Hydroxybutyrate, a Ketone Body, Potentiates the Antioxidant Defense via Thioredoxin 1 Upregulation in Cardiomyocytes. Antioxidants (Basel) 2021; 10:antiox10071153. [PMID: 34356388 PMCID: PMC8301070 DOI: 10.3390/antiox10071153] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 07/12/2021] [Accepted: 07/16/2021] [Indexed: 12/21/2022] Open
Abstract
Thioredoxin 1 (Trx1) is a major antioxidant that acts adaptively to protect the heart during the development of diabetic cardiomyopathy. The molecular mechanism(s) responsible for regulating the Trx1 level and/or activity during diabetic cardiomyopathy is unknown. β-hydroxybutyrate (βHB), a major ketone body in mammals, acts as an alternative energy source in cardiomyocytes under stress, but it also appears to be involved in additional mechanisms that protect the heart against stress. βHB upregulated Trx1 in primary cultured cardiomyocytes in a dose- and a time-dependent manner and a ketogenic diet upregulated Trx1 in the heart. βHB protected cardiomyocytes against H2O2-induced death, an effect that was abolished in the presence of Trx1 knockdown. βHB also alleviated the H2O2-induced inhibition of mTOR and AMPK, known targets of Trx1, in a Trx1-dependent manner, suggesting that βHB potentiates Trx1 function. It has been shown that βHB is a natural inhibitor of HDAC1 and knockdown of HDAC1 upregulated Trx1 in cardiomyocytes, suggesting that βHB may upregulate Trx1 through HDAC inhibition. βHB induced Trx1 acetylation and inhibited Trx1 degradation, suggesting that βHB-induced inhibition of HDAC1 may stabilize Trx1 through protein acetylation. These results suggest that βHB potentiates the antioxidant defense in cardiomyocytes through the inhibition of HDAC1 and the increased acetylation and consequent stabilization of Trx1. Thus, modest upregulation of ketone bodies in diabetic hearts may protect the heart through the upregulation of Trx1.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Fan Tang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Adave Chin
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
- Department of Cardiovascular Disease, Ningbo Medical Treatment Centre Li Huili Hospital, Ningbo 315000, China
| | - Chengchen Hu
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Zhi Yang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Maha Abdellatif
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07103, USA; (S.-i.O.); (F.T.); (A.C.); (G.R.); (X.X.); (C.H.); (Z.Y.); (M.A.)
- Correspondence: ; Tel.: +1-97-3972-8619
| |
Collapse
|
19
|
Oka SI, Byun J, Huang CY, Imai N, Ralda G, Zhai P, Xu X, Kashyap S, Warren JS, Alan Maschek J, Tippetts TS, Tong M, Venkatesh S, Ikeda Y, Mizushima W, Kashihara T, Sadoshima J. Nampt Potentiates Antioxidant Defense in Diabetic Cardiomyopathy. Circ Res 2021; 129:114-130. [PMID: 33928788 PMCID: PMC8513534 DOI: 10.1161/circresaha.120.317943] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Antioxidants/metabolism
- Apoptosis
- Autophagy
- Cells, Cultured
- Cytokines/genetics
- Cytokines/metabolism
- Diabetic Cardiomyopathies/enzymology
- Diabetic Cardiomyopathies/genetics
- Diabetic Cardiomyopathies/pathology
- Diet, High-Fat
- Disease Models, Animal
- Fibrosis
- Glutathione/metabolism
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria, Heart/enzymology
- Mitochondria, Heart/genetics
- Mitochondria, Heart/pathology
- Mitophagy
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- NAD/metabolism
- NADP/metabolism
- Nicotinamide Phosphoribosyltransferase/genetics
- Nicotinamide Phosphoribosyltransferase/metabolism
- Oxidative Stress
- Rats, Wistar
- Sirtuins/genetics
- Sirtuins/metabolism
- Thioredoxins/metabolism
- Mice
- Rats
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taiwan (C.-y.H.)
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan (C.-y.H.)
| | - Nobushige Imai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Zhejiang, China (X.X.)
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junco S Warren
- Nora Eccles Harrison Cardiovascular Research and Training Institute (J.S.W.), University of Utah, Salt Lake City
| | - John Alan Maschek
- Metabolomics, Proteomics, and Mass Spectrometry Cores (J.A.M.), University of Utah, Salt Lake City
- Department of Nutrition and Integrative Physiology (J.A.M.), University of Utah, Salt Lake City
| | - Trevor S Tippetts
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Sundararajan Venkatesh
- Department of Microbiology, Biochemistry, and Molecular Genetics (S.V.), Rutgers New Jersey Medical School, Newark
- Department of Nutrition and Integrative Physiology and the Diabetes and Metabolism Research Center (T.S.T., S.V.), University of Utah, Salt Lake City
| | - Yoshiyuki Ikeda
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Toshihide Kashihara
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine (S.-i.O., J.B., C.-y.H., N.I., G.R., P.Z., X.X., S.K., M.T., Y.I., W.M., T.K., J.S.), Rutgers New Jersey Medical School, Newark
| |
Collapse
|
20
|
Meng J, Fu L, Liu K, Tian C, Wu Z, Jung Y, Ferreira RB, Carroll KS, Blackwell TK, Yang J. Global profiling of distinct cysteine redox forms reveals wide-ranging redox regulation in C. elegans. Nat Commun 2021; 12:1415. [PMID: 33658510 PMCID: PMC7930113 DOI: 10.1038/s41467-021-21686-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Post-translational changes in the redox state of cysteine residues can rapidly and reversibly alter protein functions, thereby modulating biological processes. The nematode C. elegans is an ideal model organism for studying cysteine-mediated redox signaling at a network level. Here we present a comprehensive, quantitative, and site-specific profile of the intrinsic reactivity of the cysteinome in wild-type C. elegans. We also describe a global characterization of the C. elegans redoxome in which we measured changes in three major cysteine redox forms after H2O2 treatment. Our data revealed redox-sensitive events in translation, growth signaling, and stress response pathways, and identified redox-regulated cysteines that are important for signaling through the p38 MAP kinase (MAPK) pathway. Our in-depth proteomic dataset provides a molecular basis for understanding redox signaling in vivo, and will serve as a valuable and rich resource for the field of redox biology. Reversible cysteine oxidative modifications have emerged as important mechanisms that alter protein function. Here the authors globally assess the cysteine reactivity and an array of cysteine oxidative modifications in C. elegans, providing insights into redox signaling at the organismal level.
Collapse
Affiliation(s)
- Jin Meng
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Ling Fu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,Innovation Institute of Medical School, Medical College, Qingdao University, Qingdao, China
| | - Keke Liu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Caiping Tian
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China.,School of Medicine, Tsinghua University, Beijing, China
| | - Ziyun Wu
- Research Division, Joslin Diabetes Center, Boston, MA, USA.,Department of Genetics, Harvard Medical School, Boston, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Youngeun Jung
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Renan B Ferreira
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - Kate S Carroll
- Department of Chemistry, The Scripps Research Institute, Jupiter, FL, USA
| | - T Keith Blackwell
- Research Division, Joslin Diabetes Center, Boston, MA, USA. .,Department of Genetics, Harvard Medical School, Boston, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA.
| | - Jing Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China. .,Innovation Institute of Medical School, Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
21
|
Akkaya H. Kisspeptin-10 Administration Regulates
mTOR and AKT Activities and Oxidative Stress in Mouse Cardiac Tissue. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021020095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Sciarretta S, Forte M, Frati G, Sadoshima J. The complex network of mTOR signaling in the heart. Cardiovasc Res 2021; 118:424-439. [PMID: 33512477 DOI: 10.1093/cvr/cvab033] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 12/13/2020] [Accepted: 01/26/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) integrates several intracellular and extracellular signals involved in the regulation of anabolic and catabolic processes. mTOR assembles into two macromolecular complexes, named mTORC1 and mTORC2, which have different regulators, substrates and functions. Studies of gain- and loss-of-function animal models of mTOR signaling revealed that mTORC1/2 elicit both adaptive and maladaptive functions in the cardiovascular system. Both mTORC1 and mTORC2 are indispensable for driving cardiac development and cardiac adaption to stress, such as pressure overload. However, persistent and deregulated mTORC1 activation in the heart is detrimental during stress and contributes to the development and progression of cardiac remodeling and genetic and metabolic cardiomyopathies. In this review, we discuss the latest findings regarding the role of mTOR in the cardiovascular system, both under basal conditions and during stress, such as pressure overload, ischemia and metabolic stress. Current data suggest that mTOR modulation may represent a potential therapeutic strategy for the treatment of cardiac diseases.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | | | - Giacomo Frati
- Department of Medical and Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy.,IRCCS Neuromed, Pozzilli (IS), Italy
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
23
|
Autophagy and Redox Homeostasis in Parkinson's: A Crucial Balancing Act. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8865611. [PMID: 33224433 PMCID: PMC7671810 DOI: 10.1155/2020/8865611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated primarily from endogenous biochemical reactions in mitochondria, endoplasmic reticulum (ER), and peroxisomes. Typically, ROS/RNS correlate with oxidative damage and cell death; however, free radicals are also crucial for normal cellular functions, including supporting neuronal homeostasis. ROS/RNS levels influence and are influenced by antioxidant systems, including the catabolic autophagy pathways. Autophagy is an intracellular lysosomal degradation process by which invasive, damaged, or redundant cytoplasmic components, including microorganisms and defunct organelles, are removed to maintain cellular homeostasis. This process is particularly important in neurons that are required to cope with prolonged and sustained operational stress. Consequently, autophagy is a primary line of protection against neurodegenerative diseases. Parkinson's is caused by the loss of midbrain dopaminergic neurons (mDANs), resulting in progressive disruption of the nigrostriatal pathway, leading to motor, behavioural, and cognitive impairments. Mitochondrial dysfunction, with associated increases in oxidative stress, and declining proteostasis control, are key contributors during mDAN demise in Parkinson's. In this review, we analyse the crosstalk between autophagy and redoxtasis, including the molecular mechanisms involved and the detrimental effect of an imbalance in the pathogenesis of Parkinson's.
Collapse
|
24
|
Role of Muscle-Specific Histone Methyltransferase (Smyd1) in Exercise-Induced Cardioprotection against Pathological Remodeling after Myocardial Infarction. Int J Mol Sci 2020; 21:ijms21197010. [PMID: 32977624 PMCID: PMC7582695 DOI: 10.3390/ijms21197010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/07/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022] Open
Abstract
Pathological remodeling is the main detrimental complication after myocardial infarction (MI). Overproduction of reactive oxygen species (ROS) in infarcted myocardium may contribute to this process. Adequate exercise training after MI may reduce oxidative stress-induced cardiac tissue damage and remodeling. SET and MYND domain containing 1 (Smyd1) is a muscle-specific histone methyltransferase which is upregulated by resistance training, may strengthen sarcomere assembly and myofiber folding, and may promote skeletal muscles growth and hypertrophy. However, it remains elusive if Smyd1 has similar functions in post-MI cardiac muscle and participates in exercise-induced cardioprotection. Accordingly, we investigated the effects of interval treadmill exercise on cardiac function, ROS generation, Smyd1 expression, and sarcomere assembly of F-actin in normal and infarcted hearts. Adult male rats were randomly divided into five groups (n = 10/group): control (C), exercise alone (EX), sham-operated (S), MI induced by permanent ligation of left anterior descending coronary artery (MI), and MI with interval exercise training (MI + EX). Exercise training significantly improved post-MI cardiac function and sarcomere assembly of F-actin. The cardioprotective effects were associated with increased Smyd1, Trx1, cTnI, and α-actinin expression as well as upregulated ratio of phosphorylated AMP-activated protein kinase (AMPK)/AMPK, whereas Hsp90, MuRF1, brain natriuretic peptide (BNP) expression, ROS generation, and myocardial fibrosis were attenuated. The improved post-MI cardiac function was associated with increased Smyd1 expression. In cultured H9C2 cardiomyoblasts, in vitro treatment with H2O2 (50 µmol/L) or AMP-activated protein kinase (AMPK) agonist (AICAR, 1 mmol/L) or their combination for 4 h simulated the effects of exercise on levels of ROS and Smyd1. In conclusion, we demonstrated a novel role of Smyd1 in association with post-MI exercise-induced cardioprotection. The moderate level of ROS-induced upregulation of Smyd1 may be an important target for modulating post-MI cardiac function and remodeling.
Collapse
|
25
|
Affiliation(s)
- Francesco Perone
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli (IS), Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli (IS), Italy.,Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
26
|
Oka SI, Chin A, Park JY, Ikeda S, Mizushima W, Ralda G, Zhai P, Tong M, Byun J, Tang F, Einaga Y, Huang CY, Kashihara T, Zhao M, Nah J, Tian B, Hirabayashi Y, Yodoi J, Sadoshima J. Thioredoxin-1 maintains mitochondrial function via mechanistic target of rapamycin signalling in the heart. Cardiovasc Res 2020; 116:1742-1755. [PMID: 31584633 PMCID: PMC7825501 DOI: 10.1093/cvr/cvz251] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 08/29/2019] [Accepted: 09/26/2019] [Indexed: 12/12/2022] Open
Abstract
AIMS Thioredoxin 1 (Trx1) is an evolutionarily conserved oxidoreductase that cleaves disulphide bonds in oxidized substrate proteins such as mechanistic target of rapamycin (mTOR) and maintains nuclear-encoded mitochondrial gene expression. The cardioprotective effect of Trx1 has been demonstrated via cardiac-specific overexpression of Trx1 and dominant negative Trx1. However, the pathophysiological role of endogenous Trx1 has not been defined with a loss-of-function model. To address this, we have generated cardiac-specific Trx1 knockout (Trx1cKO) mice. METHODS AND RESULTS Trx1cKO mice were viable but died with a median survival age of 25.5 days. They developed heart failure, evidenced by contractile dysfunction, hypertrophy, and increased fibrosis and apoptotic cell death. Multiple markers consistently indicated increased oxidative stress and RNA-sequencing revealed downregulation of genes involved in energy production in Trx1cKO mice. Mitochondrial morphological abnormality was evident in these mice. Although heterozygous Trx1cKO mice did not show any significant baseline phenotype, pressure-overload-induced cardiac dysfunction, and downregulation of metabolic genes were exacerbated in these mice. mTOR was more oxidized and phosphorylation of mTOR substrates such as S6K and 4EBP1 was impaired in Trx1cKO mice. In cultured cardiomyocytes, Trx1 knockdown inhibited mitochondrial respiration and metabolic gene promoter activity, suggesting that Trx1 maintains mitochondrial function in a cell autonomous manner. Importantly, mTOR-C1483F, an oxidation-resistant mutation, prevented Trx1 knockdown-induced mTOR oxidation and inhibition and attenuated suppression of metabolic gene promoter activity. CONCLUSION Endogenous Trx1 is essential for maintaining cardiac function and metabolism, partly through mTOR regulation via Cys1483.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Adave Chin
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Ji Yeon Park
- Seoul National University Biomedical Informatics, Division of Biomedical Informatics, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Shohei Ikeda
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Guersom Ralda
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Mingming Tong
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Jaemin Byun
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Fan Tang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Yudai Einaga
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan
| | - Toshihide Kashihara
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Mengyuan Zhao
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Bin Tian
- Department of Biochemistry & Molecular Biology, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Yoko Hirabayashi
- Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Junji Yodoi
- Department of Biological Responses, Laboratory of Infection and Prevention, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| |
Collapse
|
27
|
Oeing CU, Nakamura T, Pan S, Mishra S, Dunkerly-Eyring BL, Kokkonen-Simon KM, Lin BL, Chen A, Zhu G, Bedja D, Lee DI, Kass DA, Ranek MJ. PKG1α Cysteine-42 Redox State Controls mTORC1 Activation in Pathological Cardiac Hypertrophy. Circ Res 2020; 127:522-533. [PMID: 32393148 PMCID: PMC7416445 DOI: 10.1161/circresaha.119.315714] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
RATIONALE Stimulated PKG1α (protein kinase G-1α) phosphorylates TSC2 (tuberous sclerosis complex 2) at serine 1365, potently suppressing mTORC1 (mechanistic [mammalian] target of rapamycin complex 1) activation by neurohormonal and hemodynamic stress. This reduces pathological hypertrophy and dysfunction and increases autophagy. PKG1α oxidation at cysteine-42 is also induced by these stressors, which blunts its cardioprotective effects. OBJECTIVE We tested the dependence of mTORC1 activation on PKG1α C42 oxidation and its capacity to suppress such activation by soluble GC-1 (guanylyl cyclase 1) activation. METHODS AND RESULTS Cardiomyocytes expressing wild-type (WT) PKG1α (PKG1αWT) or cysteine-42 to serine mutation redox-dead (PKG1αCS/CS) were exposed to ET-1 (endothelin 1). Cells expressing PKG1αWT exhibited substantial mTORC1 activation (p70 S6K [p70 S6 kinase], 4EBP1 [elF4E binding protein-1], and Ulk1 [Unc-51-like kinase 1] phosphorylation), reduced autophagy/autophagic flux, and abnormal protein aggregation; all were markedly reversed by PKG1αCS/CS expression. Mice with global knock-in of PKG1αCS/CS subjected to pressure overload (PO) also displayed markedly reduced mTORC1 activation, protein aggregation, hypertrophy, and ventricular dysfunction versus PO in PKG1αWT mice. Cardioprotection against PO was equalized between groups by co-treatment with the mTORC1 inhibitor everolimus. TSC2-S1365 phosphorylation increased in PKG1αCS/CS more than PKG1αWT myocardium following PO. TSC2S1365A/S1365A (TSC2 S1365 phospho-null, created by a serine to alanine mutation) knock-in mice lack TSC2 phosphorylation by PKG1α, and when genetically crossed with PKG1αCS/CS mice, protection against PO-induced mTORC1 activation, cardiodepression, and mortality in PKG1αCS/CS mice was lost. Direct stimulation of GC-1 (BAY-602770) offset disparate mTORC1 activation between PKG1αWT and PKG1αCS/CS after PO and blocked ET-1 stimulated mTORC1 in TSC2S1365A-expressing myocytes. CONCLUSIONS Oxidation of PKG1α at C42 reduces its phosphorylation of TSC2, resulting in amplified PO-stimulated mTORC1 activity and associated hypertrophy, dysfunction, and depressed autophagy. This is ameliorated by direct GC-1 stimulation.
Collapse
Affiliation(s)
- Christian U. Oeing
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Taishi Nakamura
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
- Department of Cardiovascular Medicine, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shi Pan
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Sumita Mishra
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | | | - Kristen M. Kokkonen-Simon
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Brian L. Lin
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Anna Chen
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Guangshuo Zhu
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - Dong Ik. Lee
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| | - David A. Kass
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University, Baltimore, MD 21205
| | - Mark J. Ranek
- Division of Cardiology, Department of Medicine, The Johns Hopkins Medical Institutions, Baltimore, MD 21205
| |
Collapse
|
28
|
Iida K, Naiki T, Naiki-Ito A, Suzuki S, Kato H, Nozaki S, Nagai T, Etani T, Nagayasu Y, Ando R, Kawai N, Yasui T, Takahashi S. Luteolin suppresses bladder cancer growth via regulation of mechanistic target of rapamycin pathway. Cancer Sci 2020; 111:1165-1179. [PMID: 31994822 PMCID: PMC7156788 DOI: 10.1111/cas.14334] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/24/2022] Open
Abstract
Luteolin is a natural flavonoid with strong anti–oxidative properties that is reported to have an anti–cancer effect in several malignancies other than bladder cancer. In this study, we describe the effect of luteolin on a human bladder cancer cell line, T24, in the context of the regulation of p21, thioredoxin‐1 (TRX1) and the mechanistic target of rapamycin (mTOR) pathway. Luteolin inhibited cell survival and induced G2/M cell‐cycle arrest, p21 upregulation and downregulation of phospho(p)‐S6, which is downstream of mTOR signaling. Luteolin also upregulated TRX1 and reduced intracellular reactive oxygen species production. In a subcutaneous xenograft mouse model using the rat bladder cancer cell line, BC31, tumor volumes were significantly decreased in mice orally administered luteolin compared to control. Immunohistochemical analysis revealed that increased p21 and decreased p‐S6 expression were induced in the luteolin treatment group. Moreover, in another in vivo N‐butyl‐N‐(4‐hydroxybutyl) nitrosamine (BBN)‐induced rat bladder cancer model, the oral administration of luteolin led to a trend of decreased bladder tumor dimension and significantly decreased the Ki67‐labeling index and p‐S6 expression. Furthermore, the major findings on the metabolism of luteolin suggest that both plasma and urine luteolin‐3ʹ‐O‐glucuronide concentrations are strongly associated with the inhibition of cell proliferation and mTOR signaling. Moreover, a significant decrease in the squamous differentiation of bladder cancer is attributed to plasma luteolin‐3ʹ‐glucuronide concentration. In conclusion, luteolin, and in particular its metabolized product, may represent another natural product‐derived therapeutic agent that acts against bladder cancer by upregulating p21 and inhibiting mTOR signaling.
Collapse
Affiliation(s)
- Keitaro Iida
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Nozaki
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takashi Nagai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuko Nagayasu
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Ryosuke Ando
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
29
|
Garza-Lombó C, Pappa A, Panayiotidis MI, Franco R. Redox homeostasis, oxidative stress and mitophagy. Mitochondrion 2020; 51:105-117. [PMID: 31972372 DOI: 10.1016/j.mito.2020.01.002] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 12/21/2019] [Accepted: 01/03/2020] [Indexed: 02/07/2023]
Abstract
Autophagy is a ubiquitous homeostatic mechanism for the degradation or turnover of cellular components. Degradation of mitochondria via autophagy (mitophagy) is involved in a number of physiological processes including cellular homeostasis, differentiation and aging. Upon stress or injury, mitophagy prevents the accumulation of damaged mitochondria and the increased steady state levels of reactive oxygen species leading to oxidative stress and cell death. A number of human diseases, particularly neurodegenerative disorders, have been linked to the dysregulation of mitophagy. In this mini-review, we aimed to review the molecular mechanisms involved in the regulation of mitophagy and their relationship with redox signaling and oxidative stress.
Collapse
Affiliation(s)
- Carla Garza-Lombó
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| | - Aglaia Pappa
- Department of Molecular Biology & Genetics, Democritus University of Thrace, Alexandroupolis 68100, Greece
| | | | - Rodrigo Franco
- Redox Biology Center and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, United States.
| |
Collapse
|
30
|
Wipperman MF, Montrose DC, Gotto AM, Hajjar DP. Mammalian Target of Rapamycin: A Metabolic Rheostat for Regulating Adipose Tissue Function and Cardiovascular Health. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:492-501. [PMID: 30803496 DOI: 10.1016/j.ajpath.2018.11.013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 11/03/2018] [Accepted: 11/28/2018] [Indexed: 12/31/2022]
Abstract
The complex relationship between diet and metabolism is an important contributor to cellular metabolism and health. Over the past few decades, a central role for mammalian target of rapamycin (mTOR) in the regulation of multiple cellular processes, including the response to food intake, maintaining homeostasis, and the pathogenesis of disease, has been shown. Herein, we first review our current understanding of the biochemical functions of mTOR and its response to fluctuations in hormone levels, like insulin. Second, we highlight the role of mTOR in lipogenesis, adipogenesis, β-oxidation of lipids, and ketosis of carbohydrates, lipids, and proteins. Special attention is paid to recent advances in mTOR signaling in white versus brown adipose tissues. Finally, we review how mTOR regulates cardiovascular health and disease. Together, these insights define a clearer picture of the connection between mTOR signaling, metabolic health, and disease.
Collapse
Affiliation(s)
- Matthew F Wipperman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York; Clinical and Translational Science Center, Weill Cornell Medicine, Cornell University, New York
| | - David C Montrose
- Department of Pathology, Stony Brook Medicine, Stony Brook, New York
| | - Antonio M Gotto
- Department of Medicine, Weill Cornell Medicine, Cornell University, New York
| | - David P Hajjar
- Department of Pathology and Biochemistry, Weill Cornell Medicine, Cornell University, New York.
| |
Collapse
|
31
|
Emerging role of innate B1 cells in the pathophysiology of autoimmune and neuroimmune diseases: Association with inflammation, oxidative and nitrosative stress and autoimmune responses. Pharmacol Res 2019; 148:104408. [PMID: 31454534 DOI: 10.1016/j.phrs.2019.104408] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022]
|
32
|
Ford MM, Smythers AL, McConnell EW, Lowery SC, Kolling DRJ, Hicks LM. Inhibition of TOR in Chlamydomonas reinhardtii Leads to Rapid Cysteine Oxidation Reflecting Sustained Physiological Changes. Cells 2019; 8:cells8101171. [PMID: 31569396 PMCID: PMC6829209 DOI: 10.3390/cells8101171] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/16/2019] [Accepted: 09/26/2019] [Indexed: 12/16/2022] Open
Abstract
The target of rapamycin (TOR) kinase is a master metabolic regulator with roles in nutritional sensing, protein translation, and autophagy. In Chlamydomonas reinhardtii, a unicellular green alga, TOR has been linked to the regulation of increased triacylglycerol (TAG) accumulation, suggesting that TOR or a downstream target(s) is responsible for the elusive “lipid switch” in control of increasing TAG accumulation under nutrient limitation. However, while TOR has been well characterized in mammalian systems, it is still poorly understood in photosynthetic systems, and little work has been done to show the role of oxidative signaling in TOR regulation. In this study, the TOR inhibitor AZD8055 was used to relate reversible thiol oxidation to the physiological changes seen under TOR inhibition, including increased TAG content. Using oxidized cysteine resin-assisted capture enrichment coupled with label-free quantitative proteomics, 401 proteins were determined to have significant changes in oxidation following TOR inhibition. These oxidative changes mirrored characterized physiological modifications, supporting the role of reversible thiol oxidation in TOR regulation of TAG production, protein translation, carbohydrate catabolism, and photosynthesis through the use of reversible thiol oxidation. The delineation of redox-controlled proteins under TOR inhibition provides a framework for further characterization of the TOR pathway in photosynthetic eukaryotes.
Collapse
Affiliation(s)
- Megan M Ford
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Amanda L Smythers
- Department of Chemistry, Marshall University, Huntington, WV 25755, USA.
| | - Evan W McConnell
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Sarah C Lowery
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | | | - Leslie M Hicks
- Department of Chemistry, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
33
|
Babu H, Ambikan AT, Gabriel EE, Svensson Akusjärvi S, Palaniappan AN, Sundaraj V, Mupanni NR, Sperk M, Cheedarla N, Sridhar R, Tripathy SP, Nowak P, Hanna LE, Neogi U. Systemic Inflammation and the Increased Risk of Inflamm-Aging and Age-Associated Diseases in People Living With HIV on Long Term Suppressive Antiretroviral Therapy. Front Immunol 2019; 10:1965. [PMID: 31507593 PMCID: PMC6718454 DOI: 10.3389/fimmu.2019.01965] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/05/2019] [Indexed: 12/19/2022] Open
Abstract
The ART program in low- and middle-income countries (LMIC) like India, follows a public health approach with a standardized regimen for all people living with HIV (PLHIV). Based on the evidence from high-income countries (HIC), the risk of an enhanced, and accentuated onset of premature-aging or age-related diseases has been observed in PLHIV. However, very limited data is available on residual inflammation and immune activation in the populations who are on first-generation anti-HIV drugs like zidovudine and lamivudine that have more toxic side effects. Therefore, the aim of the present study was to evaluate the levels of systemic inflammation and understand the risk of age-associated diseases in PLHIV on long-term suppressive ART using a large number of biomarkers of inflammation and immune activation. Blood samples were obtained from therapy naïve PLHIV (Pre-ART, n = 43), PLHIV on ART for >5 years (ART, n = 53), and HIV-negative healthy controls (HIVNC, n = 41). Samples were analyzed for 92 markers of inflammation, sCD14, sCD163, and telomere length. Several statistical tests were performed to compare the groups under study. Multivariate linear regression was used to investigate the associations. Despite a median duration of 8 years of successful ART, sCD14 (p < 0.001) and sCD163 (p = 0.04) levels continued to be significantly elevated in ART group as compared to HIVNC. Eleven inflammatory markers, including 4E-BP1, ADA, CCL23, CD5, CD8A, CST5, MMP1, NT3, SLAMF1, TRAIL, and TRANCE, were found to be significantly different (p < 0.05) between the groups. Many of these markers are associated with age-related co-morbidities including cardiovascular disease, neurocognitive decline and some of these markers are being reported for the first time in the context of HIV-induced inflammation. Linear regression analysis showed a significant negative association between HIV-1-positivity and telomere length (p < 0.0001). In ART-group CXCL1 (p = 0.048) and TGF-α (p = 0.026) showed a significant association with the increased telomere length and IL-10RA was significantly associated with decreased telomere length (p = 0.042). This observation warrants further mechanistic studies to generate evidence to highlight the need for enhanced treatment monitoring and special interventions in HIV-infected individuals.
Collapse
Affiliation(s)
- Hemalatha Babu
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anoop T. Ambikan
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Erin E. Gabriel
- Department of Medical Epidemiology and Biostatistics, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sara Svensson Akusjärvi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | | | - Naveen Reddy Mupanni
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Maike Sperk
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Narayanaiah Cheedarla
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | | | - Srikanth P. Tripathy
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Piotr Nowak
- Unit of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Luke Elizabeth Hanna
- Department of HIV/AIDS, National Institute for Research in Tuberculosis (ICMR), Chennai, India
| | - Ujjwal Neogi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
Sciarretta S, Forte M, Frati G, Sadoshima J. New Insights Into the Role of mTOR Signaling in the Cardiovascular System. Circ Res 2019; 122:489-505. [PMID: 29420210 DOI: 10.1161/circresaha.117.311147] [Citation(s) in RCA: 327] [Impact Index Per Article: 54.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The mTOR (mechanistic target of rapamycin) is a master regulator of several crucial cellular processes, including protein synthesis, cellular growth, proliferation, autophagy, lysosomal function, and cell metabolism. mTOR interacts with specific adaptor proteins to form 2 multiprotein complexes, called mTORC1 (mTOR complex 1) and mTORC2 (mTOR complex 2). In the cardiovascular system, the mTOR pathway regulates both physiological and pathological processes in the heart. It is needed for embryonic cardiovascular development and for maintaining cardiac homeostasis in postnatal life. Studies involving mTOR loss-of-function models revealed that mTORC1 activation is indispensable for the development of adaptive cardiac hypertrophy in response to mechanical overload. mTORC2 is also required for normal cardiac physiology and ensures cardiomyocyte survival in response to pressure overload. However, partial genetic or pharmacological inhibition of mTORC1 reduces cardiac remodeling and heart failure in response to pressure overload and chronic myocardial infarction. In addition, mTORC1 blockade reduces cardiac derangements induced by genetic and metabolic disorders and has been reported to extend life span in mice. These studies suggest that pharmacological targeting of mTOR may represent a therapeutic strategy to confer cardioprotection, although clinical evidence in support of this notion is still scarce. This review summarizes and discusses the new evidence on the pathophysiological role of mTOR signaling in the cardiovascular system.
Collapse
Affiliation(s)
- Sebastiano Sciarretta
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Maurizio Forte
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Giacomo Frati
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.)
| | - Junichi Sadoshima
- From the Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy (S.S., G.F.); Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S., M.F., G.F.); and Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark (J.S.).
| |
Collapse
|
35
|
Yarana C, Thompson H, Chaiswing L, Butterfield DA, Weiss H, Bondada S, Alhakeem S, Sukati S, St Clair DK. Extracellular vesicle-mediated macrophage activation: An insight into the mechanism of thioredoxin-mediated immune activation. Redox Biol 2019; 26:101237. [PMID: 31276937 PMCID: PMC6612011 DOI: 10.1016/j.redox.2019.101237] [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: 02/01/2019] [Revised: 05/28/2019] [Accepted: 05/31/2019] [Indexed: 12/21/2022] Open
Abstract
Extracellular vesicles (EVs) generated from redox active anticancer drugs are released into the extracellular environment. These EVs contain oxidized molecules and trigger inflammatory responses by macrophages. Using a mouse model of doxorubicin (DOX)-induced tissue injury, we previously found that the major sources of circulating EVs are from heart and liver, organs that are differentially affected by DOX. Here, we investigated the effects of EVs from cardiomyocytes and those from hepatocytes on macrophage activation. EVs from H9c2 rat cardiomyocytes (H9c2 EVs) and EVs from FL83b mouse hepatocytes (FL83 b EVs) have different levels of protein-bound 4-hydroxynonenal and thus different immunostimulatory effects on mouse RAW264.7 macrophages. H9c2 EVs but not FL83 b EVs induced both pro-inflammatory and anti-inflammatory macrophage activation, mediated by NFκB and Nrf-2 pathways, respectively. DOX enhanced the effects of H9c2 EVs but not FL83 b EVs. While EVs from DOX-treated H9c2 cells (H9c2 DOXEVs) suppressed mitochondrial respiration and increased glycolysis of macrophages, EVs from DOX-treated FL83b cells (FL83b DOXEVs) enhanced mitochondrial reserve capacity. Mechanistically, the different immunostimulatory functions of H9c2 EVs and FL83 b EVs are regulated, in part, by the redox status of the cytoplasmic thioredoxin 1 (Trx1) of macrophages. H9c2 DOXEVs lowered the level of reduced Trx1 in cytoplasm while FL83b DOXEVs did the opposite. Trx1 overexpression alleviated the effect of H9c2 DOXEVs on NFκB and Nrf-2 activation and prevented the upregulation of their target genes. Our findings identify EVs as a novel Trx1-mediated redox mediator of immune response, which greatly enhances our understanding of innate immune responses during cancer therapy.
Collapse
Affiliation(s)
- Chontida Yarana
- Center for Research and Innovation, Faculty of Medical Technology, Mahidol University, Salaya, 73170, Thailand
| | - Hannah Thompson
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA
| | - D Allan Butterfield
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA; Department of Chemistry, University of Kentucky, Lexington, KY, 40506, USA
| | - Heidi Weiss
- Markey Cancer Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Subbarao Bondada
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, 40536, USA
| | - Sara Alhakeem
- Department of Microbiology, Immunology & Molecular Genetics, University of Kentucky, Lexington, KY, 40536, USA
| | - Suriyan Sukati
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA; School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY, 40536, USA.
| |
Collapse
|
36
|
Signorelli S, Tarkowski ŁP, Van den Ende W, Bassham DC. Linking Autophagy to Abiotic and Biotic Stress Responses. TRENDS IN PLANT SCIENCE 2019; 24:413-430. [PMID: 30824355 PMCID: PMC6475611 DOI: 10.1016/j.tplants.2019.02.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 05/05/2023]
Abstract
Autophagy is a process in which cellular components are delivered to lytic vacuoles to be recycled and has been demonstrated to promote abiotic/biotic stress tolerance. Here, we review how the responses triggered by stress conditions can affect autophagy and its signaling pathways. Besides the role of SNF-related kinase 1 (SnRK1) and TOR kinases in the regulation of autophagy, abscisic acid (ABA) and its signaling kinase SnRK2 have emerged as key players in the induction of autophagy under stress conditions. Furthermore, an interplay between reactive oxygen species (ROS) and autophagy is observed, ROS being able to induce autophagy and autophagy able to reduce ROS production. We also highlight the importance of osmotic adjustment for the successful performance of autophagy and discuss the potential role of GABA in plant survival and ethylene (ET)-induced autophagy.
Collapse
Affiliation(s)
- Santiago Signorelli
- Laboratory of Molecular Plant Biology, KU Leuven, Leuven, Belgium; Departamento de Biología Vegetal, Facultad de Agronomía, Universidad de la República, Montevideo 12900, Uruguay.
| | | | - Wim Van den Ende
- Laboratory of Molecular Plant Biology, KU Leuven, Leuven, Belgium
| | - Diane C Bassham
- Department of Genetics, Development and Cell Biology, Iowa State University, Ames, IA 50011, USA
| |
Collapse
|
37
|
Miyamoto S. Autophagy and cardiac aging. Cell Death Differ 2019; 26:653-664. [PMID: 30692640 PMCID: PMC6460392 DOI: 10.1038/s41418-019-0286-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death and the prevalence of CVD dramatically increases with age. Cardiac aging is associated with hypertrophy, fibrosis, inflammation, and decreased contractility. Autophagy, a bulk degradation/recycling system, is essential to maintain cellular homeostasis. Cardiac autophagy is decreased with age, and misfolded proteins and dysfunctional mitochondria are accumulated in the aging heart. Inhibition of autophagy leads to exacerbated cardiac aging, while stimulation of autophagy improves cardiac function and also increases lifespan in many organisms. Thus autophagy represents a potential therapeutic target for aging-related cardiac dysfunction. This review discusses recent progress in our understanding of the role and regulation of autophagy in the aging heart.
Collapse
Affiliation(s)
- Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
38
|
Janse van Rensburg HC, Van den Ende W, Signorelli S. Autophagy in Plants: Both a Puppet and a Puppet Master of Sugars. FRONTIERS IN PLANT SCIENCE 2019; 10:14. [PMID: 30723485 PMCID: PMC6349728 DOI: 10.3389/fpls.2019.00014] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/07/2019] [Indexed: 05/20/2023]
Abstract
Autophagy is a major pathway that recycles cellular components in eukaryotic cells both under stressed and non-stressed conditions. Sugars participate both metabolically and as signaling molecules in development and response to various environmental and nutritional conditions. It is therefore essential to maintain metabolic homeostasis of sugars during non-stressed conditions in cells, not only to provide energy, but also to ensure effective signaling when exposed to stress. In both plants and animals, autophagy is activated by the energy sensor SnRK1/AMPK and inhibited by TOR kinase. SnRK1/AMPK and TOR kinases are both important regulators of cellular metabolism and are controlled to a large extent by the availability of sugars and sugar-phosphates in plants whereas in animals AMP/ATP indirectly translate sugar status. In plants, during nutrient and sugar deficiency, SnRK1 is activated, and TOR is inhibited to allow activation of autophagy which in turn recycles cellular components in an attempt to provide stress relief. Autophagy is thus indirectly regulated by the nutrient/sugar status of cells, but also regulates the level of nutrients/sugars by recycling cellular components. In both plants and animals sugars such as trehalose induce autophagy and in animals this is independent of the TOR pathway. The glucose-activated G-protein signaling pathway has also been demonstrated to activate autophagy, although the exact mechanism is not completely clear. This mini-review will focus on the interplay between sugar signaling and autophagy.
Collapse
Affiliation(s)
| | - Wim Van den Ende
- Laboratory of Molecular Plant Biology, KU Leuven, Leuven, Belgium
| | - Santiago Signorelli
- Laboratory of Molecular Plant Biology, KU Leuven, Leuven, Belgium
- Departamento de Biologiía Vegetal, Facultad de Agronomía, Universidad de la Repuíblica, Montevideo, Uruguay
| |
Collapse
|
39
|
Elmazoglu Z, Yar Saglam AS, Sonmez C, Karasu C. Luteolin protects microglia against rotenone-induced toxicity in a hormetic manner through targeting oxidative stress response, genes associated with Parkinson’s disease and inflammatory pathways. Drug Chem Toxicol 2018; 43:96-103. [PMID: 30207190 DOI: 10.1080/01480545.2018.1504961] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Zubeyir Elmazoglu
- Cellular Stress Response and Signal Transduction Research Laboratory, Department of Medical Pharmacology, Gazi University, Ankara, Turkey
| | | | - Can Sonmez
- Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Cimen Karasu
- Cellular Stress Response and Signal Transduction Research Laboratory, Department of Medical Pharmacology, Gazi University, Ankara, Turkey
| |
Collapse
|
40
|
Emerging Role of mTOR Signaling-Related miRNAs in Cardiovascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6141902. [PMID: 30305865 PMCID: PMC6165581 DOI: 10.1155/2018/6141902] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 07/04/2018] [Indexed: 12/21/2022]
Abstract
Mechanistic/mammalian target of rapamycin (mTOR), an atypical serine/threonine kinase of the phosphoinositide 3-kinase- (PI3K-) related kinase family, elicits a vital role in diverse cellular processes, including cellular growth, proliferation, survival, protein synthesis, autophagy, and metabolism. In the cardiovascular system, the mTOR signaling pathway integrates both intracellular and extracellular signals and serves as a central regulator of both physiological and pathological processes. MicroRNAs (miRs), a class of short noncoding RNA, are an emerging intricate posttranscriptional modulator of critical gene expression for the development and maintenance of homeostasis across a wide array of tissues, including the cardiovascular system. Over the last decade, numerous studies have revealed an interplay between miRNAs and the mTOR signaling circuit in the different cardiovascular pathophysiology, like myocardial infarction, hypertrophy, fibrosis, heart failure, arrhythmia, inflammation, and atherosclerosis. In this review, we provide a comprehensive state of the current knowledge regarding the mechanisms of interactions between the mTOR signaling pathway and miRs. We have also highlighted the latest advances on mTOR-targeted therapy in clinical trials and the new perspective therapeutic strategies with mTOR-targeting miRs in cardiovascular diseases.
Collapse
|
41
|
Dagnell M, Schmidt EE, Arnér ESJ. The A to Z of modulated cell patterning by mammalian thioredoxin reductases. Free Radic Biol Med 2018; 115:484-496. [PMID: 29278740 PMCID: PMC5771652 DOI: 10.1016/j.freeradbiomed.2017.12.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
Mammalian thioredoxin reductases (TrxRs) are selenocysteine-containing proteins (selenoproteins) that propel a large number of functions through reduction of several substrates including the active site disulfide of thioredoxins (Trxs). Well-known enzymatic systems that in turn are supported by Trxs and TrxRs include deoxyribonucleotide synthesis through ribonucleotide reductase, antioxidant defense through peroxiredoxins and methionine sulfoxide reductases, and redox modulation of a number of transcription factors. Although these functions may be essential for cells due to crucial roles in maintenance of cell viability and proliferation, findings during the last decade reveal that mammals have major redundancy in their cellular reductive systems. The synthesis of glutathione (GSH) and reductive functions of GSH-dependent pathways typically act in parallel with Trx-dependent pathways, with only one of these systems often being sufficient to support viability. Importantly, this does not imply that a modulation of the Trx system will remain without consequences, even when GSH-dependent pathways remain functional. As suggested by several recent findings, the Trx system in general and the TrxRs in particular, function as key regulators of signaling pathways. In this review article we will discuss findings that collectively suggest that modulation in mammalian systems of cytosolic TrxR1 (TXNRD1) or mitochondrial TrxR2 (TXNRD2) influence cell patterning and cellular stress responses. Effects of lower activities include increased adipogenesis, insulin responsiveness, glycogen accumulation, hyperproliferation, and distorted embryonic development, while increased activities correlate with decreased proliferation and extended lifespan, as well as worse cancer prognosis. The molecular mechanisms that underlie these diverse effects, involving regulation of protein phosphorylation cascades and of key transcription factors that guide cellular differentiation pathways, will be discussed. We conclude that the selenium-dependent oxidoreductases TrxR1 and TrxR2 should be considered as key components of signaling pathways that control cell differentiation and cellular stress responses.
Collapse
Affiliation(s)
- Markus Dagnell
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Edward E Schmidt
- Microbiology & Immunology, Montana State University, Bozeman, MT 59718, USA
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
| |
Collapse
|