1
|
Kansal H, Chopra V, Garg K, Sharma S. Role of thioredoxin in chronic obstructive pulmonary disease (COPD): a promising future target. Respir Res 2023; 24:295. [PMID: 38001457 PMCID: PMC10668376 DOI: 10.1186/s12931-023-02574-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
INTRODUCTION Thioredoxin (Trx) is a secretory protein that acts as an antioxidant, redox regulator, anti-allergic, and anti-inflammatory molecule. It has been used to treat dermatitis and inflammation of the digestive tract. In the lungs, Trx has a significant anti-inflammatory impact. On the other hand, Chronic Obstructive Pulmonary Disease (COPD) is one of the significant causes of death in the developed world, with a tremendous individual and socioeconomic impact. Despite new initiatives and endless treatment trials, COPD incidence and death will likely escalate in the coming decades. AREAS COVERED COPD is a chronic inflammatory disease impacting the airways, lung parenchyma, and pulmonary vasculature. Oxidative stress and protease-antiprotease imbalances are thought to be involved in the process. The most popular respiratory inflammatory and allergic disorders therapies are corticosteroids and β-receptor agonists. These medications are helpful but have some drawbacks, such as infection and immunosuppression; thus, addressing Trx signalling treatments may be a viable COPD treatment approach. This review shall cover the pathophysiology of COPD, the pharmacognosy of anti-COPD drugs, including the assets and liabilities of each, and the role and mechanism of Trx in COPD treatment. EXPERT OPINION Limited research has targeted the thioredoxin system as an anti-COPD drug. Spectating the increase in the mortality rates of COPD, this review article would be an interesting one to research.
Collapse
Affiliation(s)
- Heena Kansal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India
| | - Vishal Chopra
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Kranti Garg
- Department of Pulmonary Medicine, Government Medical College, Patiala, India
| | - Siddharth Sharma
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, India.
| |
Collapse
|
2
|
Oberacker T, Kraft L, Schanz M, Latus J, Schricker S. The Importance of Thioredoxin-1 in Health and Disease. Antioxidants (Basel) 2023; 12:antiox12051078. [PMID: 37237944 DOI: 10.3390/antiox12051078] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/06/2023] [Accepted: 05/09/2023] [Indexed: 05/28/2023] Open
Abstract
Thioredoxin-1 (Trx-1) is a multifunctional protein ubiquitously found in the human body. Trx-1 plays an important role in various cellular functions such as maintenance of redox homeostasis, proliferation, and DNA synthesis, but also modulation of transcription factors and control of cell death. Thus, Trx-1 is one of the most important proteins for proper cell and organ function. Therefore, modulation of Trx gene expression or modulation of Trx activity by various mechanisms, including post-translational modifications or protein-protein interactions, could cause a transition from the physiological state of cells and organs to various pathologies such as cancer, and neurodegenerative and cardiovascular diseases. In this review, we not only discuss the current knowledge of Trx in health and disease, but also highlight its potential function as a biomarker.
Collapse
Affiliation(s)
- Tina Oberacker
- Dr. Margarete Fischer-Bosch Institute for Clinical Pharmacology, 70376 Stuttgart, Germany
| | - Leonie Kraft
- Department of Internal Medicine and Nephrology, Robert-Bosch-Hospital Stuttgart, 70376 Stuttgart, Germany
| | - Moritz Schanz
- Department of Internal Medicine and Nephrology, Robert-Bosch-Hospital Stuttgart, 70376 Stuttgart, Germany
| | - Jörg Latus
- Department of Internal Medicine and Nephrology, Robert-Bosch-Hospital Stuttgart, 70376 Stuttgart, Germany
| | - Severin Schricker
- Department of Internal Medicine and Nephrology, Robert-Bosch-Hospital Stuttgart, 70376 Stuttgart, Germany
| |
Collapse
|
3
|
Hanchapola HACR, Liyanage DS, Omeka WKM, Lim C, Kim G, Jeong T, Lee J. Thioredoxin domain-containing protein 12 (TXNDC12) in red spotted grouper (Epinephelus akaara): Molecular characteristics, disulfide reductase activities, and immune responses. FISH & SHELLFISH IMMUNOLOGY 2023; 132:108449. [PMID: 36436687 DOI: 10.1016/j.fsi.2022.11.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/20/2022] [Accepted: 11/21/2022] [Indexed: 06/16/2023]
Abstract
Thioredoxins are small ubiquitous redox proteins that are involved in many biological processes. Proteins with thiol-disulfide bonds are essential regulators of cellular redox homeostasis and diagnostic markers for redox-dependent diseases. Here, we identified and characterized the thioredoxin domain-containing protein 12 (EaTXNDC12) gene in red spotted grouper (Epinephelus akaara), evaluated transcriptional responses, and investigated the activity of the recombinant protein using functional assays. EaTXNDC12 is a 19.22-kDa endoplasmic reticulum (ER)-resident protein with a 522-bp open reading frame and 173 amino acids, including a signal peptide. We identified a conserved active motif (66WCGAC70) and ER retention motif (170GDEL173) in the EaTXNDC12 amino acid sequence. Relative EaTXNDC12 mRNA expression was analyzed using 12 different tissues, with the highest expression seen in brain tissue, while skin tissue showed the lowest expression level. Furthermore, mRNA expression in response to immune challenges was analyzed in the head kidney, blood, and gill tissues. EaTXNDC12 was significantly modulated in response to bacterial endotoxin lipopolysaccharide (LPS), nervous necrosis virus (NNV), and polyinosinic:polycytidylic acid (poly(I:C)) challenges in all of the tested tissues. Recombinant EaTXNDC12 (rEaTXNDC12) displayed antioxidant ability in an insulin reductase assay, and a capacity for free radical inhibition in a 2,2-diphenyl-1-picryl-hydrazyl-hydrate assay. In addition, a DNA nicking assay revealed that purified rEaTXNDC12 exhibited concentration-dependent DNA protection activity, while results from 2-hydroxyethyl disulfide and L-dehydroascorbic assays indicated that rEaTXNDC12a possesses reducing ability. Furthermore, fathead minnow (FHM) cells transfected with EaTXNDC12-pcDNA demonstrated significantly upregulated cell survival against H2O2-induced apoptosis. Collectively, the results of this study strengthen our knowledge of EaTXNDC12 with respect to cellular redox hemostasis and immune regulation in Epinephelus akaara.
Collapse
Affiliation(s)
- H A C R Hanchapola
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - W K M Omeka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Chaehyeon Lim
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Gaeun Kim
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Taehyug Jeong
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
4
|
Jabbar S, Mathews P, Wang X, Sundaramoorthy P, Chu E, Piryani SO, Ding S, Shen X, Doan PL, Kang Y. Thioredoxin-1 regulates self-renewal and differentiation of murine hematopoietic stem cells through p53 tumor suppressor. Exp Hematol Oncol 2022; 11:83. [PMID: 36316713 PMCID: PMC9624023 DOI: 10.1186/s40164-022-00329-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 09/28/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Thioredoxin-1 (TXN1) is one of the major cellular antioxidants in mammals and is involved in a wide range of physiological cellular responses. However, little is known about the roles and the underlying molecular mechanisms of TXN1 in the regulation of hematopoietic stem/progenitor cells (HSPCs). METHODS TXN1 conditional knockout mice (ROSA-CreER-TXN1fl/fl) and TXN1fl/fl control mice were used. The mice were treated with tamoxifen and the number and biological functions of HSPCs were measured by flow cytometry, PCR and western blot. Limiting dilution competitive transplantation with sorted HSCs and serial transplantations were performed to assess the effects of TXN1 knockout on HSC self-renewal and long-term reconstitutional capacity. RNA sequencing (RNA-seq) was performed to investigate the downstream molecular pathways of TXN1 deletion in murine HSPCs. CRISPR/Cas9 knockout experiments were performed in vitro in EML murine hematopoietic stem/progenitor cell line to investigate the effects of TXN1 and/or TP53 deletion on cell survival, senescence and colony forming units. TP53 protein degradation assay, CHiP PCR and PGL3 firefly/renilla reporter assay were performed. The effects of TXN1 on various molecular pathways relevant to HSC radiation protection were examined in vitro and in vivo. RESULTS TXN1-TP53 tumor suppressor axis regulates HSPC biological fitness. Deletion of TXN1 in HSPCs using in vivo and in vitro models activates TP53 signaling pathway, and attenuates HSPC capacity to reconstitute hematopoiesis. Furthermore, we found that knocking out of TXN1 renders HSPCs more sensitive to radiation and treatment with recombinant TXN1 promotes the proliferation and expansion of HSPCs. CONCLUSIONS Our findings suggest that TXN1-TP53 axis acts as a regulatory mechanism in HSPC biological functions. Additionally, our study demonstrates the clinical potential of TXN1 for enhancing hematopoietic recovery in hematopoietic stem cell transplant and protecting HSPCs from radiation injury.
Collapse
Affiliation(s)
- Shaima Jabbar
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Parker Mathews
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Xiaobei Wang
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Pasupathi Sundaramoorthy
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Emily Chu
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Sadhna O Piryani
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
| | - Shengli Ding
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27710, USA
| | - Xiling Shen
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC, 27710, USA
| | - Phuong L Doan
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Department of Medicine, School of Medicine, Duke University Medical Center, 2400 Pratt Street, Suite 5000, Durham, NC, DUMC 396127710, USA.
- Duke Cancer Institute, Duke University, Durham, NC, USA.
| |
Collapse
|
5
|
Wei MF, Cheng CH, Wen SY, Lin JC, Chen YH, Wang CW, Lee YH, Kuo SH. Atorvastatin Attenuates Radiotherapy-Induced Intestinal Damage through Activation of Autophagy and Antioxidant Effects. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7957255. [PMID: 36092168 PMCID: PMC9459441 DOI: 10.1155/2022/7957255] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 07/06/2022] [Accepted: 08/12/2022] [Indexed: 11/17/2022]
Abstract
Abdominal or pelvic radiotherapy (RT) often results in small intestinal injury, such as apoptosis of epithelial cells and shortening of the villi. Atorvastatin, a 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor, has many biological effects including cholesterol reduction, protection from cell damage, and autophagy activation. To reduce the extent of radiotherapy- (RT-) induced enteritis, we investigated the protective effects of atorvastatin against RT-induced damage of the intestinal tract. In this study, C57BL/6 mice were randomly distributed into the following groups (n = 8 per group): (1) control group: mice were fed water only, (2) atorvastatin group (Ator): mice were administered atorvastatin, (3) irradiation group (IR): mice received abdominal RT, (4) Ator+IR group: mice received abdominal RT following atorvastatin administration, and (5) Ator+IR+3-MA group: abdominal RT following atorvastatin and 3-methyladenine (an autophagy inhibitor) administration. Based on the assessment of modified Chiu's injury score and villus/crypt ratio, we found that atorvastatin administration significantly reduced intestinal mucosal damage induced by RT. Atorvastatin treatment reduced apoptosis (cleaved caspase-3 and cleaved poly (ADP-ribose) polymerase), DNA damage (γH2AX and 53BP1), oxidative stress (OS, 4-hydroxynonenal), inflammatory molecules (phospho-NF-κB p65 and TGF-β), fibrosis (collagen I and collagen III), barrier leakage (claudin-2 and fluorescein isothiocyanate-dextran), disintegrity (fatty acid-binding protein 2), and dysfunction (lipopolysaccharide) caused by RT in small intestinal tissue. In addition, atorvastatin upregulated the expression of autophagy-active molecules (LC3B), antioxidants (heme oxygenase 1 and thioredoxin 1), and tight junction proteins (occludin and zonula occludens 1). However, the biological functions of atorvastatin in decreasing RT-induced enteritis were reversed after the administration of 3-MA; the function of antioxidant molecules and activity of thioredoxin reductase were independent of autophagy activation. Our results indicate that atorvastatin can effectively relieve RT-induced enteritis through autophagy activation and associated biological functions, including maintaining integrity and function and decreasing apoptosis, DNA damage, inflammation, OS, and fibrosis. It also acts via its antioxidative capabilities.
Collapse
Affiliation(s)
- Ming-Feng Wei
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Ching-Hsueh Cheng
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Shu-Yu Wen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jui-Chueh Lin
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yu-Hsuan Chen
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital HsinChu Branch, HsinChu, Taiwan
| | - Chun-Wei Wang
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Sung-Hsin Kuo
- Division of Radiation Oncology, Department of Oncology, National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
- Cancer Research Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Department of Radiation Oncology, National Taiwan University Cancer Center, National Taiwan University College of Medicine, Taipei, Taiwan
- Medical Physics Program, College of Nuclear Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
6
|
Hasan AA, Kalinina E, Tatarskiy V, Shtil A. The Thioredoxin System of Mammalian Cells and Its Modulators. Biomedicines 2022; 10:biomedicines10071757. [PMID: 35885063 PMCID: PMC9313168 DOI: 10.3390/biomedicines10071757] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/16/2022] [Accepted: 07/19/2022] [Indexed: 12/21/2022] Open
Abstract
Oxidative stress involves the increased production and accumulation of free radicals, peroxides, and other metabolites that are collectively termed reactive oxygen species (ROS), which are produced as by-products of aerobic respiration. ROS play a significant role in cell homeostasis through redox signaling and are capable of eliciting damage to macromolecules. Multiple antioxidant defense systems have evolved to prevent dangerous ROS accumulation in the body, with the glutathione and thioredoxin/thioredoxin reductase (Trx/TrxR) systems being the most important. The Trx/TrxR system has been used as a target to treat cancer through the thiol–disulfide exchange reaction mechanism that results in the reduction of a wide range of target proteins and the generation of oxidized Trx. The TrxR maintains reduced Trx levels using NADPH as a co-substrate; therefore, the system efficiently maintains cell homeostasis. Being a master regulator of oxidation–reduction processes, the Trx-dependent system is associated with cell proliferation and survival. Herein, we review the structure and catalytic properties of the Trx/TrxR system, its role in cellular signaling in connection with other redox systems, and the factors that modulate the Trx system.
Collapse
Affiliation(s)
- Aseel Ali Hasan
- T.T. Berezov Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia;
| | - Elena Kalinina
- T.T. Berezov Department of Biochemistry, Peoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, 117198 Moscow, Russia;
- Correspondence: ; Tel.: +7-495-434-62-05
| | - Victor Tatarskiy
- Laboratory of Molecular Oncobiology, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia;
| | - Alexander Shtil
- Laboratory of Tumor Cell Death, Blokhin National Medical Research Center of Oncology, 24 Kashirskoye Shosse, 115478 Moscow, Russia;
| |
Collapse
|
7
|
Liu Y, Xue N, Zhang B, Lv H, Li S. Role of Thioredoxin-1 and its inducers in human health and diseases. Eur J Pharmacol 2022; 919:174756. [PMID: 35032486 DOI: 10.1016/j.ejphar.2022.174756] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/08/2021] [Accepted: 01/06/2022] [Indexed: 12/27/2022]
Abstract
Thioredoxin-1 (Trx-1) is a small redox-active protein normally found in mammalian cells that responds to the changing redox environment by contributing electrons or regulating related proteins. There is growing evidence that Trx-1 has multiple functions, including cytoprotective, anti-apoptotic, antioxidant and anti-inflammatory effects. To date, researchers have found that Trx-1 deficiency leads to severe damage in various disease models, such as atherosclerosis, cerebral ischemia, diabetes and tumors. Conversely, activation of Trx-1 has a protective effect against these diseases. Accordingly, a variety of Trx-1 inducers have been widely used in the clinic with significant therapeutic value. In this paper, we summarize the pathogenesis of Trx-1 involvement in the above-mentioned diseases and describe the protective effects of Trx-1 inducers on them.
Collapse
Affiliation(s)
- Yuanyuan Liu
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Nianyu Xue
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Boxi Zhang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China
| | - Hongming Lv
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China.
| | - Shize Li
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, 163319, Heilongjiang Bayi, China.
| |
Collapse
|
8
|
Jastrząb A, Skrzydlewska E. Regulacja układu zależnego od tioredoksyny jako element farmakoterapii w chorobach z zaburzeniami równowagi redoks. POSTEP HIG MED DOSW 2021. [DOI: 10.5604/01.3001.0014.6952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Streszczenie
Działanie wielu czynników egzogennych, a także zaburzone procesy metaboliczne komórek przyczyniają się do nasilonego wytwarzania oksydantów, a to zaburza równowagę redoks, wywołując zmiany metaboliczne, w tym śmierci lub transformacji nowotworowej komórek. Jednak każda komórka zawiera antyoksydanty, które mają zapobiegać tego typu sytuacjom. Jednym z układów antyoksydacyjnych, funkcjonujących w komórkach, jest układ zależny od tioredoksyny, w skład którego wchodzą: tioredoksyna (Trx), reduktaza tioredoksyny (TrxR) oraz peroksydaza tioredoksyny (TPx), które mogą redukować utlenione składniki komórek kosztem fosforanu dinukleotydu nikotynoamidoadeninowego (NADPH). Działanie takie wynika z budowy przestrzennej Trx oraz TrxR, która umożliwia wytworzenie wewnątrzcząsteczkowego mostka disulfidowego w obrębie cząsteczki tioredoksyny oraz dwóch międzycząsteczkowych mostków selenosulfidowych w obrębie dimeru reduktazy tioredoksyny. Inną, równie istotną funkcją układu zależnego od tioredoksyny jest regulowanie ekspresji wielu białek za pośrednictwem takich czynników jak czynnik transkrypcyjnego NF-κB oraz kinaza regulująca apoptozę (ASK-1), które uruchamiają kaskady przemian metabolicznych prowadzących ostatecznie do proliferacji lub apoptozy komórek. Wzrost ekspresji/aktywności składników systemu zależnego od Trx obserwuje się w rozwoju wielu nowotworów. Dlatego też poszukiwanie selektywnych inhibitorów tioredoksyny lub reduktazy tioredoksyny jest obecnie jednym z głównych kierunków badań w farmakoterapii nowotworów. Wykazano, że wiele naturalnie występujących związków polifenolowych pochodzenia naturalnego o działaniu antyoksydacyjnym (np. kwercetyna czy kurkumina) powoduje inaktywację układu Trx-TrxR. Jednocześnie wiele syntetycznych związków, w tym związki kompleksowe, które stosowane są w terapii przeciwnowotworowej (np. cisplatyna, auranofina, moteksafina gadolinu), również hamują działanie układu zależnego od Trx.
Collapse
Affiliation(s)
- Anna Jastrząb
- Zakład Chemii Nieorganicznej i Analitycznej , Uniwersytet Medyczny w Białymstoku
| | | |
Collapse
|
9
|
Ethanol extract of Liriodendron chinense (Hemsl.) Sarg barks attenuates hyperuricemic nephropathy by inhibiting renal fibrosis and inflammation in mice. JOURNAL OF ETHNOPHARMACOLOGY 2020; 264:113278. [PMID: 32841699 DOI: 10.1016/j.jep.2020.113278] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/30/2020] [Accepted: 08/11/2020] [Indexed: 02/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Liriodendron chinense (Hemsl.) Sarg, known as the Chinese tulip tree, has a long history of cultivation and utilization in many Asia countries, especially in China to use in traditional Chinese medicine for expelling "wind and dampness", a term corresponding to rheumatic fever and rheumatoid arthritis. Interestingly, the barks of Liriodendron chinense (Hemsl.) Sarg was also found in folk to treat gout. However, further experimental studies remained to confirm its uric acid-lowering effects. AIM OF THE STUDY The aim of the study was to evaluate the protective effect of ethanol extract of the barks of Liriodendron chinense (Hemsl.) Sarg (EELC) in a mouse model of hyperuricemic nephropathy (HN) and the involved mechanisms. MATERIALS AND METHODS EELC at a respective dose of 250 mg/kg/d or 500 mg/kg/d were orally administered to HN mice induced by a mixture of adenine (160 mg/kg/d)/potassium oxonate (2.4 g/kg/d) for 21 days. At the end of the treatment, serum uric acid, kidney functions (serum creatinine, blood urea nitrogen and urine microalbumin), 24-h urine uric acid excretion, as well as kidney pathological changes were investigated by biochemical assay, histopathological score, immunofluorescence and histochemistry, RT-qPCR, and western blotting analysis. RESULTS AND DISCUSSION Oral administration of EELC significantly lowered serum uric acid level at 500 mg/kg (185.75 ± 15.49 μmol/L of EELC vs. 238.28 ± 20.97 μmol/L of HN model, p < 0.01) in HN mice. EELC at 500 mg/kg also remarkably reduced the levels of serum creatinine (82.92 ± 7.86 μmol/L of EELC vs. 92.08 ± 6.13 μmol/L of HN model, p < 0.0001), blood urea nitrogen (21.50 ± 1.87 mmol/L of EELC vs. 29.40 ± 3.95 mmol/L of HN model, p < 0.001) and urine microalbumin (4.25 ± 0.40 mg/L of EELC vs. 5.95 ± 0.33 mg/L of HN model, p < 0.001) to improve renal function. It also attenuated renal fibrosis, especially the high-dose of EELC. Furthermore, EELC could inhibit the activation of NF-κB, ASK1/JNK/c-Jun, JAK2/STAT3 signaling pathways and reduce the release of pro-inflammatory cytokine TNF-α in the kidneys of HN mice. Additionally, EELC remarkably increased urine uric acid excretion of HN mice, which may be achieved by the upregulation of organic anion transporter 1 (OAT1), OAT3 and ATP-binding cassette subfamily G member 2 (ABCG2) proteins. CONCLUSIONS EELC alleviated the progression of HN by suppressing the activation of NF-κB, ASK1/JNK/c-Jun and JAK2/STAT3 signaling pathway, reducing the infiltration of inflammatory factors and uric acid accumulation in the kidney.
Collapse
|
10
|
Zheng Z, Fan S, Zheng J, Huang W, Gasparetto C, Chao NJ, Hu J, Kang Y. Inhibition of thioredoxin activates mitophagy and overcomes adaptive bortezomib resistance in multiple myeloma. J Hematol Oncol 2018; 11:29. [PMID: 29482577 PMCID: PMC5828316 DOI: 10.1186/s13045-018-0575-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/12/2018] [Indexed: 11/25/2022] Open
Abstract
Background Although current chemotherapy using bortezomib (Velcade) against multiple myeloma in adults has achieved significant responses and even remission, a majority of patients will develop acquired resistance to bortezomib. Increased thioredoxin level has been reported to be associated with carcinogenesis; however, the role of thioredoxin in bortezomib drug resistance of myeloma remains unclear. Methods We generated several bortezomib-resistant myeloma cell lines by serially passaging with increased concentrations of bortezomib over a period of 1.5 years. Thioredoxin expression was measured by real-time PCR and western blot. Results The role of thioredoxin in the survival of bortezomib-resistant myeloma cells was determined by specific shRNA knockdown in vitro and in vivo. Thioredoxin inhibitor (PX12) was used to determine the effectiveness of thioredoxin inhibition in the treatment of bortezomib-resistant myeloma cells. The effect of thioredoxin inhibition on mitophagy induction was examined. The correlation of thioredoxin expression with patient overall survival was interrogated. Thioredoxin expression was significantly upregulated in bortezomib-resistant myeloma cells and the change correlated with the increase of bortezomib concentration. Thioredoxin gene knockdown using specific shRNA sensitized bortezomib-resistant myeloma cells to bortezomib efficiency in vitro and in vivo. Similarly, pharmacological inhibition with PX12 inhibited the growth of bortezomib-resistant myeloma cells and overcame bortezomib resistance in vitro and in vivo. Furthermore, inhibition of thioredoxin resulted in the activation of mitophagy and blockage of mitophagy prevented the effects of PX12 on bortezomib-resistant myeloma cells, indicating that mitophagy is the important molecular mechanism for the induction of cell death in bortezomib-resistant myeloma cells by PX12. Moreover, inhibition of thioredoxin resulted in downregulation of phosphorylated mTOR and ERK1/2. Finally, thioredoxin was overexpressed in primary myeloma cells isolated from bortezomib-resistant myeloma patients and overexpression of thioredoxin correlated with poor overall survival in patients with multiple myeloma. Conclusions Our findings demonstrated that increased thioredoxin plays a critical role in bortezomib resistance in multiple myeloma through mitophagy inactivation and increased mTOR and ERK1/2 phosphorylation. Thioredoxin provides a potential target for clinical therapeutics against multiple myeloma, particularly for bortezomib-resistant/refractory myeloma patients. Electronic supplementary material The online version of this article (10.1186/s13045-018-0575-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhihong Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.,Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Shengjun Fan
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jing Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei Huang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Cristina Gasparetto
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Nelson J Chao
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jianda Hu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA.
| |
Collapse
|
11
|
Abstract
During sepsis, the alarmin HMGB1 is released from tissues and promotes systemic inflammation that results in multi-organ damage, with the kidney particularly susceptible to injury. The severity of inflammation and pro-damage signaling mediated by HMGB1 appears to be dependent on the alarmin's redox state. Therefore, we examined HMGB1 redox in kidney cells during sepsis. Using intravital microscopy, CellROX labeling of kidneys in live mice indicated increased ROS generation in the kidney perivascular endothelium and tubules during lipopolysaccharide (LPS)-induced sepsis. Subsequent CellROX and MitoSOX labeling of LPS-stressed endothelial and kidney proximal tubule cells demonstrated increased ROS generation in these cells as sepsis worsens. Consequently, HMGB1 oxidation increased in the cytoplasm of kidney cells during its translocation from the nucleus to the circulation, with the degree of oxidation dependent on the severity of sepsis, as measured in in vivo mouse samples using a thiol assay and mass spectrometry (LC-MS/MS). The greater the oxidation of HMGB1, the greater the ability of the alarmin to stimulate pro-inflammatory cyto-/chemokine release (measured by Luminex Multiplex) and alter mitochondrial ATP generation (Luminescent ATP Detection Assay). Administration of glutathione and thioredoxin inhibitors to cell cultures enhanced HMGB1 oxidation during sepsis in endothelial and proximal tubule cells, respectively. In conclusion, as sepsis worsens, ROS generation and HMGB1 oxidation increases in kidney cells, which enhances HMGB1's pro-inflammatory signaling. Conversely, the glutathione and thioredoxin systems work to maintain the protein in its reduced state. Endotoxins (LPS) increase cellular oxidative stress during sepsis. During its translocation, HMGB1 gets oxidized in the cytoplasm. Thioredoxin and glutathione keep HMGB1 in a reduced redox state during sepsis. HMGB1 oxidation enhances its stimulation of inflammatory cyto-/chemokine release.
Collapse
|
12
|
Kaplan O, Zárubová J, Mikulová B, Filová E, Bártová J, Bačáková L, Brynda E. Enhanced Mitogenic Activity of Recombinant Human Vascular Endothelial Growth Factor VEGF121 Expressed in E. coli Origami B (DE3) with Molecular Chaperones. PLoS One 2016; 11:e0163697. [PMID: 27716773 PMCID: PMC5055331 DOI: 10.1371/journal.pone.0163697] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 09/13/2016] [Indexed: 12/22/2022] Open
Abstract
We describe the production of a highly-active mutant VEGF variant, α2-PI1-8-VEGF121, which contains a substrate sequence for factor XIIIa at the aminoterminus designed for incorporation into a fibrin gel. The α2-PI1-8-VEGF121 gene was synthesized, cloned into a pET-32a(+) vector and expressed in Escherichia coli Origami B (DE3) host cells. To increase the protein folding and the solubility, the resulting thioredoxin-α2-PI1-8-VEGF121 fusion protein was co-expressed with recombinant molecular chaperones GroES/EL encoded by independent plasmid pGro7. The fusion protein was purified from the soluble fraction of cytoplasmic proteins using affinity chromatography. After cleavage of the thioredoxin fusion part with thrombin, the target protein was purified by a second round of affinity chromatography. The yield of purified α2-PI1-8-VEGF121 was 1.4 mg per liter of the cell culture. The α2-PI1-8-VEGF121 expressed in this work increased the proliferation of endothelial cells 3.9-8.7 times in comparison with commercially-available recombinant VEGF121. This very high mitogenic activity may be caused by co-expression of the growth factor with molecular chaperones not previously used in VEGF production. At the same time, α2-PI1-8-VEGF121 did not elicit considerable inflammatory activation of human endothelial HUVEC cells and human monocyte-like THP-1 cells.
Collapse
Affiliation(s)
- Ondřej Kaplan
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, CZ-162 06, Prague, Czech Republic
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
- * E-mail:
| | - Jana Zárubová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Barbora Mikulová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
- Faculty of Science, Charles University in Prague, CZ-128 40, Prague, Czech Republic
| | - Elena Filová
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Jiřina Bártová
- School of Dental Medicine, General University Hospital in Prague, CZ-128 08, Prague, Czech Republic
| | - Lucie Bačáková
- Institute of Physiology, Czech Academy of Sciences, CZ-142 20, Prague, Czech Republic
| | - Eduard Brynda
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, CZ-162 06, Prague, Czech Republic
| |
Collapse
|
13
|
Lennicke C, Rahn J, Lichtenfels R, Wessjohann LA, Seliger B. Hydrogen peroxide - production, fate and role in redox signaling of tumor cells. Cell Commun Signal 2015; 13:39. [PMID: 26369938 PMCID: PMC4570748 DOI: 10.1186/s12964-015-0118-6] [Citation(s) in RCA: 373] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 09/08/2015] [Indexed: 02/07/2023] Open
Abstract
Hydrogen peroxide (H2O2) is involved in various signal transduction pathways and cell fate decisions. The mechanism of the so called “redox signaling” includes the H2O2-mediated reversible oxidation of redox sensitive cysteine residues in enzymes and transcription factors thereby altering their activities. Depending on its intracellular concentration and localization, H2O2 exhibits either pro- or anti-apoptotic activities. In comparison to normal cells, cancer cells are characterized by an increased H2O2 production rate and an impaired redox balance thereby affecting the microenvironment as well as the anti-tumoral immune response. This article reviews the current knowledge about the intracellular production of H2O2 along with redox signaling pathways mediating either the growth or apoptosis of tumor cells. In addition it will be discussed how the targeting of H2O2-linked sources and/or signaling components involved in tumor progression and survival might lead to novel therapeutic targets.
Collapse
Affiliation(s)
- Claudia Lennicke
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Jette Rahn
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Rudolf Lichtenfels
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany
| | - Ludger A Wessjohann
- Leibniz-Institute of Plant Biochemistry, Weinberg 3, 06120, Halle /Saale, Germany
| | - Barbara Seliger
- Institute of Medical Immunology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 2, 06112, Halle/Saale, Germany.
| |
Collapse
|
14
|
Matchimakul P, Rinaldi G, Suttiprapa S, Mann VH, Popratiloff A, Laha T, Pimenta RN, Cochran CJ, Kaewkes S, Sripa B, Brindley PJ. Apoptosis of cholangiocytes modulated by thioredoxin of carcinogenic liver fluke. Int J Biochem Cell Biol 2015; 65:72-80. [PMID: 26007234 DOI: 10.1016/j.biocel.2015.05.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/27/2015] [Accepted: 05/14/2015] [Indexed: 02/07/2023]
Abstract
Chronic infection with the food-borne liver fluke, Opisthorchis viverrini, frequently induces cancer of the bile ducts, cholangiocarcinoma. Opisthorchiasis is endemic in Thailand, Lao PDR, Cambodia and Vietnam, where eating undercooked freshwater fish carrying the juvenile stage of this pathogen leads to human infection. Because inhibition of apoptosis facilitates carcinogenesis, this study investigated modulation by thioredoxin from O. viverrini of apoptosis of bile duct epithelial cells, cholangiocytes. Cells of a cholangiocyte line were incubated with the parasite enzyme after which they were exposed hydrogen peroxide. Oxidative stress-induced apoptosis was monitored using flow cytometry, growth in real time and imaging of living cells using laser confocal microscopy. Immunolocalization revealed liver fluke thioredoxin within cholangiocytes. Cells exposed to thioredoxin downregulated apoptotic genes in the mitogen activated protein kinases pathway and upregulated anti-apoptosis-related genes including apoptosis signaling kinase 1, caspase 9, caspase 8, caspase 3, survivin and others. Western blots of immunoprecipitates of cell lysates revealed binding of thioredoxin to apoptosis signaling kinase 1. Together the findings indicated that thioredoxin from O. viverrini inhibited oxidative stress-induced apoptosis of bile duct epithelial cells, which supports a role for this liver fluke oxidoreductase in opisthorchiasis-induced cholangiocarcinogenesis.
Collapse
Affiliation(s)
- Pitchaya Matchimakul
- Biomedical Sciences Program, Graduate School, Khon Kaen University, Khon Kaen 40002, Thailand; WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Gabriel Rinaldi
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA; Parasite Genomics, Wellcome Trust Sanger Institute, Genome Campus, Hinxton CB10 1SA, UK
| | - Sutas Suttiprapa
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA; Department of Microbiology, Faculty of Science, Mahidol University, Rachthewee, Bangkok 10400, Thailand
| | - Victoria H Mann
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Anastas Popratiloff
- Center for Microscopy & Image Analysis, and Department of Anatomy & Regenerative Biology, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Thewarach Laha
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Rafael N Pimenta
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Christina J Cochran
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA
| | - Sasithorn Kaewkes
- Department of Parasitology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Banchob Sripa
- WHO Collaborating Centre for Research and Control of Opisthorchiasis (Southeast Asian Liver Fluke Disease), Tropical Disease Research Laboratory, Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand.
| | - Paul J Brindley
- Department of Microbiology, Immunology & Tropical Medicine, and Research Center for Neglected Diseases of Poverty, School of Medicine & Health Sciences, George Washington University, Washington, DC 20037, USA.
| |
Collapse
|