1
|
WIN SY, HORIO F, SATO J, MOTAI Y, SEO H, FUJISAWA S, SATO T, OISHI E, HTUN LL, BAWM S, OKAGAWA T, MAEKAWA N, KONNAI S, OHASHI K, MURATA S. Potential of histamine release factor for the utilization as a universal vaccine antigen against poultry red mites, tropical fowl mites, and northern fowl mites. J Vet Med Sci 2025; 87:1-12. [PMID: 39567007 PMCID: PMC11735211 DOI: 10.1292/jvms.24-0186] [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: 05/15/2024] [Accepted: 11/07/2024] [Indexed: 11/22/2024] Open
Abstract
Poultry red mites (PRMs, Dermanyssus gallinae), tropical fowl mites (TFMs, Ornithonyssus bursa), and northern fowl mites (NFMs, Ornithonyssus sylviarum) are hematophagous mites that are distributed worldwide which pose a serious challenge to the poultry industry and negatively impact poultry production and welfare. Vaccines represent a promising approach for controlling avian mites, and the identification of antigens with broad efficacy against multiple avian mite species is advantageous for vaccine control. This study aimed to identify histamine release factor (HRF), which was previously reported as a candidate vaccine antigen against PRMs, from TFMs and NFMs and to analyze its cross-reactivity and acaricidal effects on different avian mite species. The deduced amino acid sequences of the HRFs identified in the TFMs and NFMs were highly homologous to those of the PRMs. We generated recombinant HRF (rHRF) of TFMs, NFMs, and PRMs, and immune plasma against each rHRF was produced by immunization with each antigen. The immune plasma contained antibodies specific to each antigen and showed cross-reactivity with rHRFs from different avian mites. Moreover, PRM nymphs (protonymphs) artificially fed each immune plasma showed higher mortality rates than those fed the control plasma. These results suggest that HRFs can be used as candidate antigens for a universal vaccine with broad efficacy across avian mites.
Collapse
Affiliation(s)
- Shwe Yee WIN
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Fumiya HORIO
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Jumpei SATO
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Yoshinosuke MOTAI
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Hikari SEO
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Sotaro FUJISAWA
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | | | | | - Lat Lat HTUN
- Department of Pharmacology and Parasitology, University of Veterinary Science, Nay Pyi Taw, Myanmar
| | - Saw BAWM
- Department of Pharmacology and Parasitology, University of Veterinary Science, Nay Pyi Taw, Myanmar
- Department of Livestock and Aquaculture Research, Ministry of Agriculture, Livestock and Irrigation, Nay Pyi Taw, Myanmar
| | - Tomohiro OKAGAWA
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Naoya MAEKAWA
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Satoru KONNAI
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Institute for Vaccine Research and Development (GU-IVReD), Hokkaido University, Hokkaido, Japan
| | - Kazuhiko OHASHI
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- International Affairs Office, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| | - Shiro MURATA
- Laboratory of Infectious Diseases, Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
2
|
Miao G, Yang Y, Yang X, Chen D, Liu L, Lei X. The multifaceted potential of TPT1 as biomarker and therapeutic target. Heliyon 2024; 10:e38819. [PMID: 39397949 PMCID: PMC11471257 DOI: 10.1016/j.heliyon.2024.e38819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 09/17/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024] Open
Abstract
Tumor Protein Translationally-Controlled 1 (TPT1) is a highly conserved gene found across eukaryotic species. The protein encoded by TPT1 is ubiquitously expressed both intracellularly and extracellularly across various tissues, and its levels are influenced by various external factors. TPT1 interacts with several key proteins, including p53, MCL1, and immunoglobulins, highlighting its crucial role in cellular processes. The dysregulation of TPT1 expression has been documented in a wide range of diseases, indicating its potential as a valuable biomarker. Additionally, targeting TPT1 presents a promising approach for treating and preventing various conditions. This review will assess the potential of TPT1 as a biomarker and evaluate the effectiveness of current strategies designed to inhibit TPT1 in disease contexts.
Collapse
Affiliation(s)
- Gelan Miao
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Yulian Yang
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Xuelian Yang
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Dexiu Chen
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Li Liu
- Department of Anesthesiology, The First Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| | - Xianying Lei
- Department of Critical Care Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, Sichuan Province, 646000, China
| |
Collapse
|
3
|
Translationally controlled tumor protein restores impaired memory and altered synaptic protein expression in animal models of dementia. Biomed Pharmacother 2023; 160:114357. [PMID: 36738496 DOI: 10.1016/j.biopha.2023.114357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/25/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
This study describes the effects of translationally controlled tumor protein (TCTP) on mice with memory impairment caused by scopolamine (SCO) administration. Specifically, memory functions and expression levels of hippocampal synaptic proteins in 7- to 12-month-old SCO-treated wild-type (WT-SCO) mice were compared to those of TCTP-overexpressing (TG) and TCTP knocked-down (KD) mice similarly treated with SCO. Passive-avoidance tasks were performed with WT, TG, and KD mice for four weeks after intraperitoneal injection of SCO or saline followed by an acquisition test. After completing behavioral studies, hippocampi of all mice groups were collected and their synaptic protein contents were subjected to Western blotting or immunohistochemical analyses, and compared with those of 5x familial Alzheimer's disease (5xFAD) mice and postmortem AD patients. Results of passive avoidance tests revealed that SCO-induced memory impairment was repaired in TCTP-TG mice, but not in TCTP-KD mice. Hippocampal expression levels of synaptophysin, synapsin-1, and PSD-95 were increased in TCTP-TG mice treated with SCO (TG-SCO) but decreased in TCTP-KD mice treated with SCO (KD-SCO). Decreased levels of TCTP, synaptophysin, and PSD-95 were also found in hippocampi of 5xFAD mice and AD patients. Expression levels of p-CREB/CREB and brain-derived neurotrophic factor (BDNF) in TCTP-TG and TG-SCO mice were similar to or increased compared to those in WT mice, but decreased in TCTP-KD and KD-SCO mice. BDNF immunoreactivity was restored in CA1 regions of hippocampi of TG-SCO mice, but not in KD-SCO mice. These results suggest that TCTP can restore damaged memory in mice possibly through restored synaptic protein expression.
Collapse
|
4
|
Maeng J, Lee K. Inhibitors of dimerized translationally controlled tumor protein, a histamine releasing factor, may serve as anti-allergic drug candidates. Biochimie 2023; 211:141-152. [PMID: 36963558 DOI: 10.1016/j.biochi.2023.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 03/26/2023]
Abstract
It has been established that translationally controlled tumor protein (TCTP), also called histamine releasing factor (HRF), exhibits cytokine-like activities associated with initiation of allergic responses only after forming dimers (dTCTP). Agents that inhibit dTCTP by preventing its dimerization or otherwise block its function, also block development of allergic reactions, thereby serving as potential drugs to treat allergic diseases. Several lines of evidence have proven that peptides and antibodies that specifically inhibit the interactions between dTCTP and either its putative receptor or immunoglobulins exhibit significant in vivo efficacy as potential anti-inflammatory agents in murine models of allergic inflammatory diseases. This review highlights the development of several inhibitors targeting dTCTP and discusses how they affect the pathophysiologic processes of allergic and inflammatory diseases in several animal models and offers new perspectives on anti-allergic drug discovery.
Collapse
Affiliation(s)
- Jeehye Maeng
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea
| | - Kyunglim Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Seo H, Bae HD, Pyun H, Kim BG, Lee SI, Song JS, Lee K. PEGylation improves the therapeutic potential of dimerized translationally controlled tumor protein blocking peptide in ovalbumin-induced mouse model of airway inflammation. Drug Deliv 2022; 29:2320-2329. [PMID: 35850571 PMCID: PMC9302014 DOI: 10.1080/10717544.2022.2100511] [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] [Indexed: 11/17/2022] Open
Abstract
Dimerized translationally controlled tumor protein (dTCTP) initiates a variety of allergic responses in mouse models and that dTCTP-binding peptide 2 (dTBP2) attenuates the allergic inflammation by targeting dTCTP. However, the usefulness of peptide-based drugs is often limited due to their short half-lives, rapid degradation, and high levels of clearance after systemic administration. In this study, we chemically conjugated dTBP2 with 10 kDa polyethylene glycol (PEG) to improve its therapeutic potential. N-terminal mono-PEGylated dTBP2 (PEG-dTBP2) was characterized by in vitro bioactivity assay, pharmacokinetics study, and in vivo efficacy. When compared to the unmodified dTBP2, PEG-dTBP2 reduced proinflammatory cytokine IL-8 secretion in human bronchial cells by 10 to 15% and increased plasma half-life by approximately 2.5-fold in mice. This study specifically demonstrated that PEG-dTBP2 shows higher inhibitory action against ovalbumin (OVA)-induced airway inflammation in mice compared to dTBP2. Importantly, PEG-dTBP2, when administered once at 1 mg/kg, significantly reduced the migration of inflammatory cells and the levels of cytokines in the bronchoalveolar lavage fluids as well as OVA-specific IgE levels in serum. In addition, the degree of goblet cell hyperplasia and mucus secretion were significantly attenuated in the PEG-dTBP2 group compared with the control group. These results suggest that PEG-dTBP2 can be considered a potential candidate drug for regulating allergic inflammation.
Collapse
Affiliation(s)
- Hyeran Seo
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Hae-Duck Bae
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Haejun Pyun
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| | - Bo-Gyu Kim
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Sang-Il Lee
- Department of Internal Medicine and Institute of Health Science, Gyeongsang National University School of Medicine and Hospital, Jinju, Republic of Korea
| | - Jin-Sook Song
- Data Convergence Drug Research Center, Therapeutics & Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, Korea
| | - Kyunglim Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, Korea
| |
Collapse
|
6
|
Cantrell MS, Wall JD, Pu X, Turner M, Woodbury L, Fujise K, McDougal OM, Warner LR. Expression and purification of a cleavable recombinant fortilin from Escherichia coli for structure activity studies. Protein Expr Purif 2022; 189:105989. [PMID: 34626801 PMCID: PMC8557625 DOI: 10.1016/j.pep.2021.105989] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 01/03/2023]
Abstract
Complications related to atherosclerosis account for approximately 1 in 4 deaths in the United States and treatment has focused on lowering serum LDL-cholesterol levels with statins. However, approximately 50% of those diagnosed with atherosclerosis have blood cholesterol levels within normal parameters. Human fortilin is an anti-apoptotic protein and a factor in macrophage-mediated atherosclerosis and is hypothesized to protect inflammatory macrophages from apoptosis, leading to subsequent cardiac pathogenesis. Fortilin is unique because it provides a novel drug target for atherosclerosis that goes beyond lowering cholesterol and utilization of a solution nuclear magnetic resonance (NMR) spectroscopy, structure-based drug discovery approach requires milligram quantities of pure, bioactive, recombinant fortilin. Here, we designed expression constructs with different affinity tags and protease cleavage sites to find optimal conditions to obtain the quantity and purity of protein necessary for structure activity relationship studies. Plasmids encoding fortilin with maltose binding protein (MBP), 6-histidine (6His) and glutathione-S-transferase (GST), N- terminal affinity tags were expressed and purified from Escherichia coli (E. coli). Cleavage sites with tobacco etch virus (TEV) protease and human rhinovirus (HRV) 3C protease were assessed. Despite high levels of expression of soluble protein, the fusion constructs were resistant to proteinases without the inclusion of amino acids between the cleavage site and N-terminus. We surveyed constructs with increasing lengths of glycine/serine (GGS) linkers between the cleavage site and fortilin and found that inclusion of at least one GGS insert led to successful protease cleavage and pure fortilin with conserved binding to calcium as measured by NMR.
Collapse
Affiliation(s)
- Maranda S Cantrell
- Biomolecular Sciences Ph.D. Program, Boise State University, Boise, ID, 83725, USA; Department of Chemistry and Biochemistry, Boise State University, Boise, ID, 83725, USA
| | - Jackson D Wall
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, 83725, USA
| | - Xinzhu Pu
- Biomolecular Research Center, Boise State University, Boise, ID, 83725, USA
| | - Matthew Turner
- Biomolecular Research Center, Boise State University, Boise, ID, 83725, USA
| | - Luke Woodbury
- Biomolecular Research Center, Boise State University, Boise, ID, 83725, USA
| | - Ken Fujise
- Harborview Medical Center, University of Washington, Seattle, WA, 98104-2499, USA
| | - Owen M McDougal
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, 83725, USA
| | - Lisa R Warner
- Department of Chemistry and Biochemistry, Boise State University, Boise, ID, 83725, USA.
| |
Collapse
|
7
|
Cho H, Kim HK, Oh A, Jeong MG, Song J, Lee K, Hwang ES. dTBP2 attenuates severe airway inflammation by blocking inflammatory cellular network mediated by dTCTP. Biomed Pharmacother 2021; 144:112316. [PMID: 34628164 DOI: 10.1016/j.biopha.2021.112316] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/18/2021] [Accepted: 10/05/2021] [Indexed: 11/28/2022] Open
Abstract
Dimeric translationally controlled tumor protein (dTCTP), also known as histamine-releasing factor, amplifies allergic responses and its production has been shown to increase in inflammatory diseases such as allergic asthma. Despite the critical role of dTCTP in allergic inflammation, little is known about its production pathways, associated cellular networks, and underlying molecular mechanisms. In this study, we explored the dTCTP-mediated inflammatory networks and molecular mechanisms of dTCTP associated with lipopolysaccharides (LPS)-induced severe asthma. LPS stimulation increased dTCTP production by mast cells and dTCTP secretion during degranulation, and extracellular dTCTP subsequently increased the production of pro-inflammatory molecules, including IL-8, by airway epithelial cells without affecting mast cell activation. Furthermore, dimeric TCTP-binding peptide 2 (dTBP2), a dTCTP inhibitor peptide, selectively blocked the dTCTP-mediated signaling network from mast cells to epithelial cells and decreased IL-8 production through IkB induction and nuclear p65 export in airway epithelial cells. More importantly, dTBP2 efficiently attenuated LPS-induced severe airway inflammation in vivo, resulting in decreased immune cell infiltration and IL-17 production and attenuated dTCTP secretion. These results suggest that dTCTP produced by mast cells exacerbates airway inflammation through activation of airway epithelial cells in a paracrine signaling manner, and that dTBP2 is beneficial in the treatment of severe airway inflammation by blocking the dTCTP-mediated inflammatory cellular network.
Collapse
Affiliation(s)
- Hyunsoo Cho
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Hyo Kyeong Kim
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Areum Oh
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Mi Gyeong Jeong
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Jiseo Song
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea
| | - Kyunglim Lee
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea.
| | - Eun Sook Hwang
- College of Pharmacy and Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, South Korea.
| |
Collapse
|
8
|
Cho H, Park J, Kim HK, Hwang ES, Lee K. Dimerized Translationally Controlled Tumor Protein-Binding Peptide 2 Attenuates Systemic Anaphylactic Reactions Through Direct Suppression of Mast Cell Degranulation. Front Pharmacol 2021; 12:764321. [PMID: 34737708 PMCID: PMC8560797 DOI: 10.3389/fphar.2021.764321] [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: 08/25/2021] [Accepted: 10/04/2021] [Indexed: 11/13/2022] Open
Abstract
Dimerized translationally controlled tumor protein (dTCTP) amplifies allergic responses through activation of several types of immune cells and release of inflammatory mediators. In particular, dTCTP plays an important role in histamine release by triggering mast cells and has been proposed as a target in the treatment of allergic diseases. dTCTP-binding peptide 2 (dTBP2) is known to attenuate severe allergic rhinitis and asthma through inhibition of dTCTP activity on airway epithelial cells and T cells; however, it is unclear whether dTBP2 affects mast cell function and mast cell disease. In this study, we explored the effects of dTBP2 on mast cell degranulation and allergen-induced anaphylactic reactions. We found that bacterial product lipopolysaccharide increased the expression of dTCTP in mast cells and rapidly released dTCTP by the mast cell stimulator compound 48/80. Interestingly, the released dTCTP further promoted mast cell degranulation in an autocrine activation manner and increased calcium mobilization in mast cells, which is essential for degranulation. Furthermore, dTBP2 directly and dose-dependently inhibited in vitro mast cell degranulation enhanced by compound 48/80, suggesting a direct and potent anti-anaphylactic activity of dTBP2. dTBP2 also significantly suppressed the dTCTP-induced degranulation and histamine release through inhibition of the p38 MAPK signaling pathway and suppression of lysosomal expansion and calcium mobilization in mast cells. More importantly, in vivo administration of dTBP2 decreased mortality and significantly attenuated histamine release and inflammatory cytokine production in compound 48/80-induced systemic anaphylactic reactions. These results suggest that dTBP2 is beneficial for the control of anaphylaxis with increased dTCTP.
Collapse
Affiliation(s)
- Hyunsoo Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Jiyoung Park
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea.,Fluorescence Core Imaging Center, Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Hyo Kyeong Kim
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Eun Sook Hwang
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| | - Kyunglim Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul, South Korea
| |
Collapse
|
9
|
Gremski LH, Matsubara FH, Polli NLC, Antunes BC, Schluga PHDC, da Justa HC, Minozzo JC, Wille ACM, Senff-Ribeiro A, Veiga SS. Prospective Use of Brown Spider Venom Toxins as Therapeutic and Biotechnological Inputs. Front Mol Biosci 2021; 8:706704. [PMID: 34222343 PMCID: PMC8247472 DOI: 10.3389/fmolb.2021.706704] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 06/04/2021] [Indexed: 11/20/2022] Open
Abstract
Brown spider (genus Loxosceles) venoms are mainly composed of protein toxins used for predation and defense. Bites of these spiders most commonly produce a local dermonecrotic lesion with gravitational spread, edema and hemorrhage, which together are defined as cutaneous loxoscelism. Systemic loxoscelism, such as hematological abnormalities and renal injury, are less frequent but more lethal. Some Loxosceles venom toxins have already been isolated and extensively studied, such as phospholipases D (PLDs), which have been recombinantly expressed and were proven to reproduce toxic activities associated to the whole venom. PLDs have a notable potential to be engineered and converted in non-toxic antigens to produce a new generation of antivenoms or vaccines. PLDs also can serve as tools to discover inhibitors to be used as therapeutic agents. Other Loxosceles toxins have been identified and functionally characterized, such as hyaluronidases, allergen factor, serpin, TCTP and knottins (ICK peptides). All these toxins were produced as recombinant molecules and are biologically active molecules that can be used as tools for the potential development of chemical candidates to tackle many medical and biological threats, acting, for instance, as antitumoral, insecticides, analgesic, antigens for allergy tests and biochemical reagents for cell studies. In addition, these recombinant toxins may be useful to develop a rational therapy for loxoscelism. This review summarizes the main candidates for the development of drugs and biotechnological inputs that have been described in Brown spider venoms.
Collapse
Affiliation(s)
| | | | | | - Bruno Cesar Antunes
- Department of Cell Biology, Federal University of Paraná, Curitiba, Brazil.,Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | | | | | - João Carlos Minozzo
- Production and Research Center of Immunobiological Products, State Department of Health, Piraquara, Brazil
| | - Ana Carolina Martins Wille
- Department of Structural, Molecular Biology and Genetics, State University of Ponta Grossa, Ponta Grossa, Brazil
| | | | | |
Collapse
|
10
|
Boia-Ferreira M, Moreno KG, Basílio ABC, da Silva LP, Vuitika L, Soley B, Wille ACM, Donatti L, Barbaro KC, Chaim OM, Gremski LH, Veiga SS, Senff-Ribeiro A. TCTP from Loxosceles Intermedia (Brown Spider) Venom Contributes to the Allergic and Inflammatory Response of Cutaneous Loxoscelism. Cells 2019; 8:E1489. [PMID: 31766608 PMCID: PMC6953063 DOI: 10.3390/cells8121489] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 11/07/2019] [Accepted: 11/10/2019] [Indexed: 12/24/2022] Open
Abstract
LiTCTP is a toxin from the Translationally Controlled Tumor Protein (TCTP) family identified in Loxosceles brown spider venoms. These proteins are known as histamine-releasing factors (HRF). TCTPs participate in allergic and anaphylactic reactions, which suggest their potential role as therapeutic targets. The histaminergic effect of TCTP is related to its pro-inflammatory functions. An initial characterization of LiTCTP in animal models showed that this toxin can increase the microvascular permeability of skin vessels and induce paw edema in a dose-dependent manner. We evaluated the role of LiTCTP in vitro and in vivo in the inflammatory and allergic aspects that undergo the biological responses observed in Loxoscelism, the clinical condition after an accident with Loxosceles spiders. Our results showed LiTCTP recombinant toxin (LiRecTCTP) as an essential synergistic factor for the dermonecrotic toxin actions (LiRecDT1, known as the main toxin in the pathophysiology of Loxoscelism), revealing its contribution to the exacerbated inflammatory response clinically observed in envenomated patients.
Collapse
Affiliation(s)
- Marianna Boia-Ferreira
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Kamila G. Moreno
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Alana B. C. Basílio
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Lucas P. da Silva
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Larissa Vuitika
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Bruna Soley
- Department of Pharmacology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil;
| | - Ana Carolina M. Wille
- Department of Structural and Molecular Biology, State University of Ponta Grossa, Ponta Grossa 84030-900, PR, Brazil;
| | - Lucélia Donatti
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Katia C. Barbaro
- Laboratory of Immunopathology, Butantan Institute, São Paulo 05503-900, SP, Brazil;
| | - Olga M. Chaim
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Luiza Helena Gremski
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Silvio S. Veiga
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| | - Andrea Senff-Ribeiro
- Department of Cell Biology, Federal University of Paraná, Curitiba 81531-980, PR, Brazil; (M.B.-F.); (K.G.M.); (A.B.C.B.); (L.P.d.S.); (L.V.); (L.D.); or (L.H.G.); (S.S.V.)
| |
Collapse
|
11
|
Branco R, Masle J. Systemic signalling through translationally controlled tumour protein controls lateral root formation in Arabidopsis. JOURNAL OF EXPERIMENTAL BOTANY 2019; 70:3927-3940. [PMID: 31037291 PMCID: PMC6685649 DOI: 10.1093/jxb/erz204] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/06/2019] [Indexed: 05/05/2023]
Abstract
The plant body plan and primary organs are established during embryogenesis. However, in contrast to animals, plants have the ability to generate new organs throughout their whole life. These give them an extraordinary developmental plasticity to modulate their size and architecture according to environmental constraints and opportunities. How this plasticity is regulated at the whole-organism level is elusive. Here we provide evidence for a role for translationally controlled tumour protein (TCTP) in regulating the iterative formation of lateral roots in Arabidopsis. AtTCTP1 modulates root system architecture through a dual function: as a general constitutive growth promoter enhancing root elongation and as a systemic signalling agent via mobility in the vasculature. AtTCTP1 encodes mRNAs with long-distance mobility between the shoot and roots. Mobile shoot-derived TCTP1 gene products act specifically to enhance the frequency of lateral root initiation and emergence sites along the primary root pericycle, while root elongation is controlled by local constitutive TCTP1 expression and scion size. These findings uncover a novel type for an integrative signal in the control of lateral root initiation and the compromise for roots between branching more profusely or elongating further. They also provide the first evidence in plants of an extracellular function of the vital, highly expressed ubiquitous TCTP1.
Collapse
Affiliation(s)
- Rémi Branco
- The Australian National University, College of Science, Research School of Biology, Canberra ACT, Australia
| | - Josette Masle
- The Australian National University, College of Science, Research School of Biology, Canberra ACT, Australia
- Correspondence:
| |
Collapse
|
12
|
Brown Spider ( Loxosceles) Venom Toxins as Potential Biotools for the Development of Novel Therapeutics. Toxins (Basel) 2019; 11:toxins11060355. [PMID: 31248109 PMCID: PMC6628458 DOI: 10.3390/toxins11060355] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 06/03/2019] [Accepted: 06/04/2019] [Indexed: 11/16/2022] Open
Abstract
Brown spider envenomation results in dermonecrosis with gravitational spreading characterized by a marked inflammatory reaction and with lower prevalence of systemic manifestations such as renal failure and hematological disturbances. Several toxins make up the venom of these species, and they are mainly peptides and proteins ranging from 5–40 kDa. The venoms have three major families of toxins: phospholipases-D, astacin-like metalloproteases, and the inhibitor cystine knot (ICK) peptides. Serine proteases, serpins, hyaluronidases, venom allergens, and a translationally controlled tumor protein (TCTP) are also present. Toxins hold essential biological properties that enable interactions with a range of distinct molecular targets. Therefore, the application of toxins as research tools and clinical products motivates repurposing their uses of interest. This review aims to discuss possibilities for brown spider venom toxins as putative models for designing molecules likely for therapeutics based on the status quo of brown spider venoms. Herein, we explore new possibilities for the venom components in the context of their biochemical and biological features, likewise their cellular targets, three-dimensional structures, and mechanisms of action.
Collapse
|
13
|
Ulambayar B, Lee H, Yang EM, Park HS, Lee K, Ye YM. Dimerized, Not Monomeric, Translationally Controlled Tumor Protein Induces Basophil Activation and Mast Cell Degranulation in Chronic Urticaria. Immune Netw 2019; 19:e20. [PMID: 31281717 PMCID: PMC6597440 DOI: 10.4110/in.2019.19.e20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/31/2019] [Accepted: 04/05/2019] [Indexed: 01/01/2023] Open
Abstract
Translationally controlled tumor protein (TCTP) is also known as histamine releasing factor as it has the ability to activate mast cells. To investigate the role of TCTP in the pathogenesis of chronic spontaneous urticaria (CSU), we evaluated serum level of TCTP and effect of TCTP on basophil and mast cell degranulation. TCTP levels in the sera from 116 CSU patients and 70 normal healthy controls (NCs) were measured by ELISA. CD203c expression on basophils from CSU patients and β-hexosaminidase release from Laboratory of Allergic Disease 2 mast cells were measured upon stimulation monomeric and dimeric TCTP. Non-reducing Western blot analysis was used for detecting dimeric TCTP. No difference was observed in serum TCTP levels between CSU patients and NCs (p=0.676). However, dimeric TCTP intensity on Western blot was stronger in CSU patients than in NCs. TCTP levels were higher in patients with severe CSU (p=0.049) and with IgG positivity to FcɛRIα (p=0.038). A significant positive correlation was observed between TCTP and eosinophil cationic protein levels (Spearman's rho=0.341; p=0.001). Both basophil and mast cell degranulation were significantly increased after stimulation with dimeric TCTP, but not with monomic TCTP. The ability of TCTP to activate basophil and mast cells is dependent on dimerization, suggesting that the inhibition of TCTP dimerization can be a therapeutic option for CSU. Association between TCTP levels and the presence of IgG to high affinity Fc epsilon receptor I alpha subunit in CSU patients indicates that autoimmune mechanisms may be involved in the dimerization of TCTP.
Collapse
Affiliation(s)
- Bastsetseg Ulambayar
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Heewon Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Eun-Mi Yang
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea
| | - Kyunglim Lee
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Womans University, Seoul 03760, Korea
| | - Young-Min Ye
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon 16499, Korea
| |
Collapse
|
14
|
Matsuura K, Otani M, Takano M, Kadoyama K, Matsuyama S. Proteomic Analysis of Hippocampus and Cortex in Streptozotocin-Induced Diabetic Model Mice Showing Dementia. J Diabetes Res 2018; 2018:8953015. [PMID: 29850612 PMCID: PMC5907478 DOI: 10.1155/2018/8953015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/24/2018] [Accepted: 02/21/2018] [Indexed: 11/18/2022] Open
Abstract
AIM Diabetes with its associated hyperglycemia induces various type of peripheral damage and also impairs the central nervous system (CNS). This study is aimed at clarifying the precise mechanism of diabetes-induced dementia as an impairment of CNS. METHODS The proteomic analysis of the hippocampus and cortex in streptozotocin- (STZ-) treated mouse diabetic model showing dementia was performed using two-dimensional gel electrophoresis (2-DE) followed by mass spectrometry (n = 3/group). RESULTS Significant changes in the expression of 32 proteins and 7 phosphoproteins were observed in the hippocampus and cortex. These identified proteins and phosphoproteins could be functionally classified as cytoskeletal protein, oxidoreductase, protein deubiquitination, energy metabolism, GTPase activation, heme binding, hydrolase, iron storage, neurotransmitter release, protease inhibitor, transcription, glycolysis, antiapoptosis, calcium ion binding, heme metabolic process, protein degradation, vesicular transport, and unknown in the hippocampus or cortex. Additionally, Western blotting validated the changes in translationally controlled tumor protein, ATP-specific succinyl-CoA synthetase beta subunit, and gamma-enolase isoform 1. CONCLUSIONS These findings showed that STZ-induced diabetes changed the expression of proteins and phosphoproteins in the hippocampus and cortex. We propose that alterations in expression levels of these proteins play an important role in diabetes-induced dementia.
Collapse
Affiliation(s)
- Kenji Matsuura
- Faculty of Pharmacy, Osaka-Ohtani University, Tondabayashi 584-8540, Japan
| | - Mieko Otani
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Masaoki Takano
- Department of Life Sciences Pharmacy, School of Pharmaceutical Sciences, Kobe Gakuin University, Kobe 650-8586, Japan
| | - Keiichi Kadoyama
- Department of Pharmaceutical Health Care, Faculty of Pharmaceutical Sciences, Himeji Dokkyo University, Himeji 670-8524, Japan
| | - Shogo Matsuyama
- Biosignal Research Center, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
15
|
Karaki S, Benizri S, Mejías R, Baylot V, Branger N, Nguyen T, Vialet B, Oumzil K, Barthélémy P, Rocchi P. Lipid-oligonucleotide conjugates improve cellular uptake and efficiency of TCTP-antisense in castration-resistant prostate cancer. J Control Release 2017; 258:1-9. [PMID: 28472637 DOI: 10.1016/j.jconrel.2017.04.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/25/2017] [Accepted: 04/29/2017] [Indexed: 01/08/2023]
Abstract
Translationally controlled tumor protein (TCTP) has been implicated in a plethora of important cellular processes related to cell growth, cell cycle progression, malignant transformation and inhibition of apoptosis. Therefore, TCTP is now recognized as a potential therapeutic target in several cancers including prostate, breast and lung cancers. We previously showed that TCTP is overexpressed in castration-resistant prostate cancer (CRPC), and it has been implicated resistance to treatment. Recently, we developed TCTP antisense oligonucleotides (ASOs) to inhibit TCTP expression. However, the intracellular delivery and silencing activity of these oligonucleotides remains a challenge, and depend on the use of transfection agents and delivery systems. Here we show that lipid-modified ASO (LASOs) has improved penetration and efficiency in inhibiting TCTP expression in the absence of additional transfection agents, both in vitro and in vivo. Transfection with TCTP-LASO led to rapid and prolonged internalization via macropinocytosis, TCTP downregulation and significant decreased cell viability. We also show that lipid-modification led to delayed tumor progression in CRPC xenografts models, with no significant toxic effects observed.
Collapse
Affiliation(s)
- Sara Karaki
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Sebastien Benizri
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Raquel Mejías
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Virginie Baylot
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Nicolas Branger
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Tan Nguyen
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France
| | - Brune Vialet
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Khalid Oumzil
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France
| | - Philippe Barthélémy
- ARNA Laboratory, University of Bordeaux, F-33076 Bordeaux, France; INSERM U1212, F-33076, Bordeaux, France; UMR CNRS 5320, F-33076, Bordeaux, France
| | - Palma Rocchi
- Centre de Recherche en Cancérologie de Marseille (CRCM), INSERM UMR1068, 27 Bd. Lei Roure BP30059, 13273 Marseille, France; Institut Paoli-Calmettes, 13273 Marseille, France; Aix-Marseille Université, 13284 Marseille, France; CNRS UMR7258, 13009 Marseille, France.
| |
Collapse
|
16
|
Zhang J, Shim G, de Toledo SM, Azzam EI. The Translationally Controlled Tumor Protein and the Cellular Response to Ionizing Radiation-Induced DNA Damage. Results Probl Cell Differ 2017; 64:227-253. [DOI: 10.1007/978-3-319-67591-6_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2023]
|
17
|
Bommer UA. The Translational Controlled Tumour Protein TCTP: Biological Functions and Regulation. Results Probl Cell Differ 2017; 64:69-126. [PMID: 29149404 DOI: 10.1007/978-3-319-67591-6_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
The Translational Controlled Tumour Protein TCTP (gene symbol TPT1, also called P21, P23, Q23, fortilin or histamine-releasing factor, HRF) is a highly conserved protein present in essentially all eukaryotic organisms and involved in many fundamental cell biological and disease processes. It was first discovered about 35 years ago, and it took an extended period of time for its multiple functions to be revealed, and even today we do not yet fully understand all the details. Having witnessed most of this history, in this chapter, I give a brief overview and review the current knowledge on the structure, biological functions, disease involvements and cellular regulation of this protein.TCTP is able to interact with a large number of other proteins and is therefore involved in many core cell biological processes, predominantly in the response to cellular stresses, such as oxidative stress, heat shock, genotoxic stress, imbalance of ion metabolism as well as other conditions. Mechanistically, TCTP acts as an anti-apoptotic protein, and it is involved in DNA-damage repair and in cellular autophagy. Thus, broadly speaking, TCTP can be considered a cytoprotective protein. In addition, TCTP facilitates cell division through stabilising the mitotic spindle and cell growth through modulating growth signalling pathways and through its interaction with the proteosynthetic machinery of the cell. Due to its activities, both as an anti-apoptotic protein and in promoting cell growth and division, TCTP is also essential in the early development of both animals and plants.Apart from its involvement in various biological processes at the cellular level, TCTP can also act as an extracellular protein and as such has been involved in modulating whole-body defence processes, namely in the mammalian immune system. Extracellular TCTP, typically in its dimerised form, is able to induce the release of cytokines and other signalling molecules from various types of immune cells. There are also several examples, where TCTP was shown to be involved in antiviral/antibacterial defence in lower animals. In plants, the protein appears to have a protective effect against phytotoxic stresses, such as flooding, draught, too high or low temperature, salt stress or exposure to heavy metals. The finding for the latter stress condition is corroborated by earlier reports that TCTP levels are considerably up-regulated upon exposure of earthworms to high levels of heavy metals.Given the involvement of TCTP in many biological processes aimed at maintaining cellular or whole-body homeostasis, it is not surprising that dysregulation of TCTP levels may promote a range of disease processes, foremost cancer. Indeed a large body of evidence now supports a role of TCTP in at least the most predominant types of human cancers. Typically, this can be ascribed to both the anti-apoptotic activity of the protein and to its function in promoting cell growth and division. However, TCTP also appears to be involved in the later stages of cancer progression, such as invasion and metastasis. Hence, high TCTP levels in tumour tissues are often associated with a poor patient outcome. Due to its multiple roles in cancer progression, TCTP has been proposed as a potential target for the development of new anti-cancer strategies in recent pilot studies. Apart from its role in cancer, TCTP dysregulation has been reported to contribute to certain processes in the development of diabetes, as well as in diseases associated with the cardiovascular system.Since cellular TCTP levels are highly regulated, e.g. in response to cell stress or to growth signalling, and because deregulation of this protein contributes to many disease processes, a detailed understanding of regulatory processes that impinge on TCTP levels is required. The last section of this chapter summarises our current knowledge on the mechanisms that may be involved in the regulation of TCTP levels. Essentially, expression of the TPT1 gene is regulated at both the transcriptional and the translational level, the latter being particularly advantageous when a rapid adjustment of cellular TCTP levels is required, for example in cell stress responses. Other regulatory mechanisms, such as protein stability regulation, may also contribute to the regulation of overall TCTP levels.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- School of Medicine, Graduate Medicine, University of Wollongong, Wollongong, NSW, 2522, Australia.
| |
Collapse
|
18
|
Abstract
Proteins from TCTP/HRF family were identified as venom toxins of spiders from different genus. We have found a TCTP toxin in the venom gland of Loxosceles intermedia, a venomous spider very common in South Brazil. TCTP from L. intermedia, named LiTCTP, was cloned, produced in a heterologous prokaryotic system, and the recombinant toxin was biochemically characterized. Our results point that LiTCTP is involved in the inflammatory events of Loxocelism, the clinical signs triggered after Loxosceles sp. bite, which include intense inflammatory reaction at the bite site followed by local necrosis. TCTP toxins were also identified in spiders from different genus. There are very few articles about TCTP toxins in other venomous animals in the literature, although a NCBI database search on the protein sequences reveals TCTP on snake's venom glands transcriptomic and genomic studies. Studies on TCTP as a venom toxin are very few and its biological role as a venom component in prey capture is still unknown.
Collapse
|
19
|
Bommer UA, Iadevaia V, Chen J, Knoch B, Engel M, Proud CG. Growth-factor dependent expression of the translationally controlled tumour protein TCTP is regulated through the PI3-K/Akt/mTORC1 signalling pathway. Cell Signal 2015; 27:1557-68. [PMID: 25936523 DOI: 10.1016/j.cellsig.2015.04.011] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/21/2015] [Accepted: 04/25/2015] [Indexed: 11/21/2022]
Abstract
Translationally controlled tumour protein TCTP (gene symbol: TPT1) is a highly-conserved, cyto-protective protein implicated in many physiological and disease processes, in particular cancer, where it is associated with poor patient outcomes. To understand the mechanisms underlying the accumulation of high TCTP levels in cancer cells, we studied the signalling pathways that control translation of TCTP mRNA, which contains a 5'-terminal oligopyrimidine tract (5'-TOP). In HT29 colon cancer cells and in HeLa cells, serum increases the expression of TCTP two- and four-fold, respectively, and this is inhibited by rapamycin or mTOR kinase inhibitors. Polysome profiling and mRNA quantification indicate that these effects occur at the level of mRNA translation. Blocking this pathway upstream of mTOR complex 1 (mTORC1) by inhibiting Akt also prevented increases in TCTP levels in both HeLa and HT29 colon cancer cells, whereas knockout of TSC2, a negative regulator of mTORC1, led to derepression of TCTP synthesis under serum starvation. Overexpression of eIF4E enhanced the polysomal association of the TCTP mRNA, although it did not protect its translation from inhibition by rapamycin. Conversely, expression of a constitutively-active mutant of the eIF4E inhibitor 4E-BP1, which is normally inactivated by mTORC1, inhibited TCTP mRNA translation in HEK293 cells. Our results demonstrate that TCTP mRNA translation is regulated by signalling through the PI3-K/Akt/mTORC1 pathway. This explains why TCTP levels are frequently increased in cancers, since mTORC1 signalling is hyperactive in ~80% of tumours.
Collapse
Affiliation(s)
- Ulrich-Axel Bommer
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia; Graduate School of Medicine, Faculty of Science, Medicine and Health, University of Wollongong, Wollongong 2522 NSW, Australia.
| | | | - Jiezhong Chen
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | - Bianca Knoch
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | - Martin Engel
- Illawarra Health and Medical Research Institute, University of Wollongong, Wollongong 2522 NSW, Australia
| | | |
Collapse
|
20
|
The influence of chronic ibuprofen treatment on proteins expressed in the mouse hippocampus. Eur J Pharmacol 2015; 752:61-8. [PMID: 25659514 DOI: 10.1016/j.ejphar.2015.01.047] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 01/13/2015] [Accepted: 01/28/2015] [Indexed: 11/22/2022]
Abstract
Ibuprofen is a nonsteroidal anti-inflammatory drug (NSAID), treatment with which has been shown to delay the onset, slows the cognitive decline, and decreases the incidence of Alzheimer׳s disease (AD) in epidemiological and clinical studies. However, a comprehensive understanding of its mechanism of action remains unclear. To elucidate the prophylactic effect of ibuprofen on the onset of the learning and memory disturbances of AD, we performed proteomic analysis of the hippocampus of chronic ibuprofen-treated mice using two-dimensional gel electrophoresis (2-DE) followed by mass spectrometry. Twenty-eight proteins and seven phosphoproteins were identified to be significantly changed in the hippocampus of chronic ibuprofen-treated mice: translationally controlled tumor protein, thioredoxin-dependent peroxide reductase, and peroxiredoxin 6 were increased, and glial fibrillary acidic protein, dihydropyrimidinase-related protein 2, EF-hand domain-containing protein D2, and 14-3-3ζ were decreased. These identified proteins and phosphoproteins could be classified as cytoskeletal, neuronal development, chaperone, metabolic, apoptosis, neurotransmitter release, ATP synthase, deubiquitination, proteasome, NOS inhibitor, adapter, vesicle transport, signal transduction, antioxidant enzyme, proton transport, synaptogenesis, and serine/threonine phosphatase types. Western blot analysis showed the changes in dihydropyrimidinase-related protein 2, heat shock protein 8, ubiquitin carboxyl-terminal hydrolase PGP9.5, and γ-enolase levels in the hippocampus of chronic ibuprofen-treated mice. These findings showed that the chronic treatment with ibuprofen changed the levels of some proteins and phosphoproteins in the hippocampus. We propose that these identified proteins and phosphoproteins play an important role in decreasing the incidence of AD, especially impaired learning and memory functions.
Collapse
|
21
|
Chen C, Deng Y, Hua M, Xi Q, Liu R, Yang S, Liu J, Zhong J, Tang M, Lu S, Zhang Z, Min X, Tang C, Wang Y. Expression and clinical role of TCTP in epithelial ovarian cancer. J Mol Histol 2015; 46:145-56. [PMID: 25564355 DOI: 10.1007/s10735-014-9607-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 12/30/2014] [Indexed: 11/28/2022]
Abstract
The aim of this study is to investigate the potential role and prognostic significance of translationally controlled tumor protein (TCTP) in human epithelial ovarian cancer (EOC). Western blot was used to evaluate the expression of TCTP in eight fresh EOC tissues. Immunohistochemistry was performed on formalin-fixed paraffin-embedded sections of 119 cases of ovarian cancers. Kaplan-Meier method indicated the relation between TCTP and EOC patients' overall survival rate. Starvation and re-feeding was used to assess cell cycle. Knocking down of TCTP and CCK8 assay showed the role of TCTP in HO8910 cell cycle. We found that TCTP was overexpressed in carcinoma tissues compared with normal tissues. Immunohistochemistry revealed that TCTP expression was significantly associated with clinicopathologic variables. Kaplan-Meier analysis revealed that high TCTP expression was significantly related to poor prognosis of the patients. Starvation and re-feeding suggested TCTP played a critical role in HO8910 cell proliferation. Interference of TCTP and CCK8 assay showed that the TCTP-siRNA treated HO8910 cells grew more slowly than the control group. CCK-8 assays and terminal-deoxynucleoitidyl transferase mediated nick end labeling assays were also performed to demonstrate the cisplatin could inhibit the survival of HO8910 cells and promote their apoptosis. All the experiments we have done showed that TCTP could promote the progression of EOC and reduce the sensitiveness of HO8910 cells to cisplatin.
Collapse
Affiliation(s)
- Chen Chen
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Nantong University, Nantong, 226001, Jiangsu Province, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Mathieu C, Demarta-Gatsi C, Porcherie A, Brega S, Thiberge S, Ronce K, Smith L, Peronet R, Amino R, Ménard R, Mécheri S. Plasmodium bergheihistamine-releasing factor favours liver-stage development via inhibition of IL-6 production and associates with a severe outcome of disease. Cell Microbiol 2014; 17:542-58. [DOI: 10.1111/cmi.12382] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/11/2014] [Accepted: 10/15/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Cédric Mathieu
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Claudia Demarta-Gatsi
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Adeline Porcherie
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Sara Brega
- Institut Pasteur; Unité de Biologie et Génétique du Paludisme; Paris F-75015 France
| | - Sabine Thiberge
- Institut Pasteur; Unité de Biologie et Génétique du Paludisme; Paris F-75015 France
| | - Karine Ronce
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Leanna Smith
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Roger Peronet
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| | - Rogerio Amino
- Institut Pasteur; Unité de Biologie et Génétique du Paludisme; Paris F-75015 France
| | - Robert Ménard
- Institut Pasteur; Unité de Biologie et Génétique du Paludisme; Paris F-75015 France
| | - Salaheddine Mécheri
- Institut Pasteur; Unité de Biologie des Interactions Hôte Parasites; Paris F-75015 France
- Centre National de la Recherche Scientifique; Unité de Recherche Associée 2581 CEDEX 15 Paris F-75724 France
| |
Collapse
|
23
|
Kaarbø M, Storm ML, Qu S, Wæhre H, Risberg B, Danielsen HE, Saatcioglu F. TCTP is an androgen-regulated gene implicated in prostate cancer. PLoS One 2013; 8:e69398. [PMID: 23894469 PMCID: PMC3718683 DOI: 10.1371/journal.pone.0069398] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2013] [Accepted: 06/10/2013] [Indexed: 11/19/2022] Open
Abstract
TCTP has been implicated in a plethora of important cellular processes related to cell growth, cell cycle progression, malignant transformation and inhibition of apoptosis. In addition to these intracellular functions, TCTP has extracellular functions and plays an important role in immune cells. TCTP expression was previously shown to be deregulated in prostate cancer, but its function in prostate cancer cells is largely unknown. Here we show that TCTP expression is regulated by androgens in LNCaP prostate cancer cells in vitro as well as human prostate cancer xenografts in vivo. Knockdown of TCTP reduced colony formation and increased apoptosis in LNCaP cells, implicating it as an important factor for prostate cancer cell growth. Global gene expression profiling in TCTP knockdown LNCaP cells showed that several interferon regulated genes are regulated by TCTP, suggesting that it may have a role in regulating immune function in prostate cancer. In addition, recombinant TCTP treatment increased colony formation in LNCaP cells suggesting that secreted TCTP may function as a proliferative factor in prostate cancer. These results suggest that TCTP may have a role in prostate cancer development.
Collapse
Affiliation(s)
- Mari Kaarbø
- Department of Biosciences, University of Oslo, Oslo, Norway
| | | | - Su Qu
- Department of Biosciences, University of Oslo, Oslo, Norway
| | - Håkon Wæhre
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| | - Bjørn Risberg
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
- Division of Pathology, Oslo University Hospital, Oslo, Norway
| | - Håvard E. Danielsen
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
- Center for Cancer Biomedicine, Oslo University Hospital, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| | - Fahri Saatcioglu
- Department of Biosciences, University of Oslo, Oslo, Norway
- Institute for Medical Informatics, Oslo University Hospital, Oslo, Norway
| |
Collapse
|