1
|
Zhang Y, Yan M, Yu Y, Wang J, Jiao Y, Zheng M, Zhang S. 14-3-3ε: a protein with complex physiology function but promising therapeutic potential in cancer. Cell Commun Signal 2024; 22:72. [PMID: 38279176 PMCID: PMC10811864 DOI: 10.1186/s12964-023-01420-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/02/2023] [Indexed: 01/28/2024] Open
Abstract
Over the past decade, the role of the 14-3-3 protein has received increasing interest. Seven subtypes of 14-3-3 proteins exhibit high homology; however, each subtype maintains its specificity. The 14-3-3ε protein is involved in various physiological processes, including signal transduction, cell proliferation, apoptosis, autophagy, cell cycle regulation, repolarization of cardiac action, cardiac development, intracellular electrolyte homeostasis, neurodevelopment, and innate immunity. It also plays a significant role in the development and progression of various diseases, such as cardiovascular diseases, inflammatory diseases, neurodegenerative disorders, and cancer. These immense and various involvements of 14-3-3ε in diverse processes makes it a promising target for drug development. Although extensive research has been conducted on 14-3-3 dimers, studies on 14-3-3 monomers are limited. This review aimed to provide an overview of recent reports on the molecular mechanisms involved in the regulation of binding partners by 14-3-3ε, focusing on issues that could help advance the frontiers of this field. Video Abstract.
Collapse
Affiliation(s)
- Yue Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Man Yan
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Yongjun Yu
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, 300121, People's Republic of China
| | - Jiangping Wang
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Yuqi Jiao
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People's Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, People's Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071, People's Republic of China.
| |
Collapse
|
2
|
Wu D, Kong X, Zhang W, Di W. Reconstruction of the TGF-β signaling pathway of Fasciola gigantica. Parasitol Res 2023; 123:51. [PMID: 38095703 DOI: 10.1007/s00436-023-08064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023]
Abstract
In the present study, we reconstructed the transforming growth factor beta (TGF-β) signaling pathway for Fasciola gigantica, which is a neglected tropical pathogen. We defined the components involved in the TGF-β signaling pathway and investigated the transcription profiles of these genes for all developmental stages of F. gigantica. In addition, the presence of these components in excretory and secretory products (FgESP) was predicted via signal peptide annotation. The core components of the TGF-β signaling pathway have been detected in F. gigantica; classical and nonclassical single transduction pathways were constructed. Four ligands have been detected, which may mediate the TGF-β signaling pathway and BMP signaling pathway. Two ligand-binding type II receptors were detected, and inhibitory Smad7 was not detected. TLP, BMP-3, BMP-1, and ActRIb showed higher transcription in 42-day juvenile and 70-day juvenile, while ActRIIa, Smad1, ActRIIb, Smad8, KAT2B, and PP2A showed higher transcription in egg. TLM, Ski, Smad6, BMPRI, p70S6K, Smad2, Smad3, TgfβRI, Smad4, and p300 showed higher transcription in metacercariae. Four ligands, 2 receptors and 3 Smads are predicted to be present in the FgESP, suggesting their potential extrinsic function. This study should help to understand signal transduction in the TGF-β signaling pathway in F. gigantica. In addition, this study helps to illustrate the complex mechanisms involved in developmental processes and F. gigantica - host interaction and paves the way for further characterization of the signaling pathway in trematodes.
Collapse
Affiliation(s)
- Dongqi Wu
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, Guangxi, China
| | - Xinping Kong
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, Guangxi, China
| | - Weiyu Zhang
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, Guangxi, China
| | - Wenda Di
- College of Animal Science and Technology, Guangxi University, Nanning, Guangxi, China.
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Guangxi University, Nanning, Guangxi, China.
| |
Collapse
|
3
|
Yang Y, Ye WL, Zhang RN, He XS, Wang JR, Liu YX, Wang Y, Yang XM, Zhang YJ, Gan WJ. The Role of TGF- β Signaling Pathways in Cancer and Its Potential as a Therapeutic Target. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:6675208. [PMID: 34335834 PMCID: PMC8321733 DOI: 10.1155/2021/6675208] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 06/22/2021] [Indexed: 02/08/2023]
Abstract
The transforming growth factor-β (TGF-β) signaling pathway mediates various biological functions, and its dysregulation is closely related to the occurrence of malignant tumors. However, the role of TGF-β signaling in tumorigenesis and development is complex and contradictory. On the one hand, TGF-β signaling can exert antitumor effects by inhibiting proliferation or inducing apoptosis of cancer cells. On the other hand, TGF-β signaling may mediate oncogene effects by promoting metastasis, angiogenesis, and immune escape. This review summarizes the recent findings on molecular mechanisms of TGF-β signaling. Specifically, this review evaluates TGF-β's therapeutic potential as a target by the following perspectives: ligands, receptors, and downstream signaling. We hope this review can trigger new ideas to improve the current clinical strategies to treat tumors related to the TGF-β signaling pathway.
Collapse
Affiliation(s)
- Yun Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Long Ye
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Ruo-Nan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Xiao-Shun He
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Jing-Ru Wang
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Xuan Liu
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Yi Wang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Xue-Mei Yang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
- Department of Pathology, The First Affiliated Hospital of Soochow University, Soochow University, Suzhou 215006, China
| | - Yu-Juan Zhang
- Department of Pathology, Medical College of Soochow University, Soochow University, Suzhou 215123, China
| | - Wen-Juan Gan
- Department of Pathology, Dushu Lake Hospital Affiliated of Soochow University, Soochow University, Suzhou 215124, China
| |
Collapse
|
4
|
Yang J, Joshi S, Wang Q, Li P, Wang H, Xiong Y, Xiao Y, Wang J, Parker-Thornburg J, Behringer RR, Yu D. 14-3-3ζ loss leads to neonatal lethality by microRNA-126 downregulation-mediated developmental defects in lung vasculature. Cell Biosci 2017; 7:58. [PMID: 29118970 PMCID: PMC5667492 DOI: 10.1186/s13578-017-0186-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/23/2017] [Indexed: 12/30/2022] Open
Abstract
Background The 14-3-3 family of proteins have been reported to play an important role in development in various mouse models, but the context specific developmental functions of 14-3-3ζ remain to be determined. In this study, we identified a context specific developmental function of 14-3-3ζ. Results Targeted deletion of 14-3-3ζ in the C57Bl/6J murine genetic background led to neonatal lethality due to respiratory distress and could be rescued by out-breeding to the CD-1 or backcrossing to the FVB/NJ congenic background. Histological analysis of lung sections from 18.5 days post coitum embryos (dpc) showed that 14-3-3ζ−/− lung development is arrested at the pseudoglandular stage and exhibits vascular defects. The expression of miR-126, an endothelial-specific miRNA known to regulate lung vascular integrity was down-regulated in the lungs of the 14-3-3ζ−/− embryos in the C57Bl/6J background as compared to their wild-type counterparts. Loss of 14-3-3ζ in endothelial cells inhibited the angiogenic capability of the endothelial cells as determined by both trans-well migration assays and tube formation assays and these defects could be rescued by re-expressing miR-126. Mechanistically, loss of 14-3-3ζ led to reduced Erk1/2 phosphorylation resulting in attenuated binding of the transcription factor Ets2 on the miR-126 promoter which ultimately reduced expression of miR-126. Conclusion Our data demonstrates that miR-126 is an important angiogenesis regulator that functions downstream of 14-3-3ζ and downregulation of miR-126 plays a critical role in 14-3-3ζ-loss induced defects in lung vasculature in the C57Bl/6J genetic background. Electronic supplementary material The online version of this article (10.1186/s13578-017-0186-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jun Yang
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA.,University of Texas Health Science Center Graduate School of Biomedical Sciences, Cancer Biology Program, Houston, TX 77030 USA
| | - Sonali Joshi
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Qingfei Wang
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Ping Li
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Hai Wang
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Yan Xiong
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Yi Xiao
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Jinyang Wang
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA
| | - Jan Parker-Thornburg
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Richard R Behringer
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030 USA.,University of Texas Health Science Center Graduate School of Biomedical Sciences, Cancer Biology Program, Houston, TX 77030 USA
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, Unit 108, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030 USA.,University of Texas Health Science Center Graduate School of Biomedical Sciences, Cancer Biology Program, Houston, TX 77030 USA.,Center for Molecular Medicine, China Medical University, Taichung, 40402 Taiwan
| |
Collapse
|
5
|
Gittenberger-de Groot AC, Hoppenbrouwers T, Miquerol L, Kosaka Y, Poelmann RE, Wisse LJ, Yost HJ, Jongbloed MRM, Deruiter MC, Brunelli L. 14-3-3epsilon controls multiple developmental processes in the mouse heart. Dev Dyn 2016; 245:1107-1123. [PMID: 27580238 PMCID: PMC5065397 DOI: 10.1002/dvdy.24440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 08/10/2016] [Accepted: 08/16/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND 14-3-3ε plays an important role in the maturation of the compact ventricular myocardium by modulating the cardiomyocyte cell cycle via p27kip1 . However, additional cardiac defects are possible given the ubiquitous expression pattern of this protein. RESULTS Germ line deletion of 14-3-3ε led to malalignment of both the outflow tract (OFT) and atrioventricular (AV) cushions, with resulting tricuspid stenosis and atresia, mitral valve abnormalities, and perimembranous ventricular septal defects (VSDs). We confirmed myocardial non-compaction and detected a spongy septum with muscular VSDs and blebbing of the epicardium. These defects were associated with abnormal patterning of p27kip1 expression in the subendocardial and possibly the epicardial cell populations. In addition to abnormal pharyngeal arch artery patterning, we found deep endocardial recesses and paucity of intramyocardial coronary vasculature as a result of defective coronary plexus remodeling. CONCLUSIONS The malalignment of both endocardial cushions provides a new explanation for tricuspid and mitral valve defects, while myocardial non-compaction provides the basis for the abnormal coronary vasculature patterning. These abnormalities might arise from p27kip1 dysregulation and a resulting defect in epithelial-to-mesenchymal transformation. These data suggest that 14-3-3ε, in addition to left ventricular non-compaction (LVNC), might be linked to different forms of congenital heart disease (CHD). Developmental Dynamics 245:1107-1123, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Adriana C Gittenberger-de Groot
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands.
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands.
| | - Tamara Hoppenbrouwers
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Yasuhiro Kosaka
- Department of Pediatrics (Neonatology), University of Utah School of Medicine, Salt Lake City, Utah
| | | | - Lambertus J Wisse
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Joseph Yost
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, Utah
| | - Monique R M Jongbloed
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marco C Deruiter
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, The Netherlands
| | - Luca Brunelli
- Department of Pediatrics (Neonatology), University of Utah School of Medicine, Salt Lake City, Utah.
| |
Collapse
|
6
|
Abstract
Transforming growth factor β (TGF-β) and related growth factors are secreted pleiotropic factors that play critical roles in embryogenesis and adult tissue homeostasis by regulating cell proliferation, differentiation, death, and migration. The TGF-β family members signal via heteromeric complexes of type I and type II receptors, which activate members of the Smad family of signal transducers. The main attribute of the TGF-β signaling pathway is context-dependence. Depending on the concentration and type of ligand, target tissue, and developmental stage, TGF-β family members transmit distinct signals. Deregulation of TGF-β signaling contributes to developmental defects and human diseases. More than a decade of studies have revealed the framework by which TGF-βs encode a context-dependent signal, which includes various positive and negative modifiers of the principal elements of the signaling pathway, the receptors, and the Smad proteins. In this review, we first introduce some basic components of the TGF-β signaling pathways and their actions, and then discuss posttranslational modifications and modulatory partners that modify the outcome of the signaling and contribute to its context-dependence, including small noncoding RNAs.
Collapse
Affiliation(s)
- Akiko Hata
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94143
| | - Ye-Guang Chen
- The State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
7
|
Gadahi JA, Wang S, Bo G, Ehsan M, Yan R, Song X, Xu L, Li X. Proteomic Analysis of the Excretory and Secretory Proteins of Haemonchus contortus (HcESP) Binding to Goat PBMCs In Vivo Revealed Stage-Specific Binding Profiles. PLoS One 2016; 11:e0159796. [PMID: 27467391 PMCID: PMC4965049 DOI: 10.1371/journal.pone.0159796] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 07/09/2016] [Indexed: 02/06/2023] Open
Abstract
Haemonchus contortus is a parasitic gastrointestinal nematode, and its excretory and secretory products (HcESPs) interact extensively with the host cells. In this study, we report the interaction of proteins from HcESPs at different developmental stages to goat peripheral blood mononuclear cells (PBMCs) in vivo using liquid chromatography-tandem mass spectrometry. A total of 407 HcESPs that interacted with goat PBMCs at different time points were identified from a H. contortus protein database using SEQUEST searches. The L4 and L5 stages of H. contortus represented a higher proportion of the identified proteins compared with the early and late adult stages. Both stage-specific interacting proteins and proteins that were common to multiple stages were identified. Forty-seven interacting proteins were shared among all stages. The gene ontology (GO) distributions of the identified goat PBMC-interacting proteins were nearly identical among all developmental stages, with high representation of binding and catalytic activity. Cellular, metabolic and single-organism processes were also annotated as major biological processes, but interestingly, more proteins were annotated as localization processes at the L5 stage than at the L4 and adult stages. Based on the clustering of homologous proteins, we improved the functional annotations of un-annotated proteins identified at different developmental stages. Some unnamed H. contortus ATP-binding cassette proteins, including ADP-ribosylation factor and P-glycoprotein-9, were identified by STRING protein clustering analysis.
Collapse
Affiliation(s)
- Javaid Ali Gadahi
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Shuai Wang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Gao Bo
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - Muhammad Ehsan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - RuoFeng Yan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - XiaoKai Song
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - LiXin Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| | - XiangRui Li
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, PR China
| |
Collapse
|
8
|
Magalhães LG, Lima TC, de Paula RG, Morais ER, Aguiar DP, Gardinassi LG, Garcia GR, Laurentiz RS, Rodrigues V, Bastos JK, Filho AA, Yatsuda AP, Cunha WR, Silva ML. Effects of (−)-6,6′-dinitrohinokinin on adult worms of Schistosoma mansoni: a proteomic analyses. REVISTA BRASILEIRA DE FARMACOGNOSIA-BRAZILIAN JOURNAL OF PHARMACOGNOSY 2016. [DOI: 10.1016/j.bjp.2016.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
9
|
Colitis promotes adaptation of an intestinal nematode: a Heligmosomoides polygyrus mouse model system. PLoS One 2013; 8:e78034. [PMID: 24167594 PMCID: PMC3805612 DOI: 10.1371/journal.pone.0078034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 09/16/2013] [Indexed: 12/13/2022] Open
Abstract
The precise mechanism of the very effective therapeutic effect of gastrointestinal nematodes on some autoimmune diseases is not clearly understood and is currently being intensively investigated. Treatment with living helminths has been initiated to reverse intestinal immune-mediated diseases in humans. However, little attention has been paid to the phenotype of nematodes in the IBD-affected gut and the consequences of nematode adaptation. In the present study, exposure of Heligmosomoides polygyrus larvae to the changed cytokine milieu of the intestine during colitis reduced inflammation in an experimental model of dextran sulphate sodium (DSS)- induced colitis, but increased nematode establishment in the moderate-responder BALB/c mouse strain. We used mass spectrometry in combination with two-dimensional Western blotting to determine changes in protein expression and changes in nematode antigens recognized by IgG1 in mice with colitis. We show that nematode larvae immunogenicity is changed by colitis as soon as 6 days post-infection; IgG1 did not recognize highly conserved proteins Lev-11 (isoform 1 of tropomyosin α1 chain), actin-4 isoform or FTT-2 isoform a (14-3-3 family) protein. These results indicate that changes in the small intestine provoked by colitis directly influence the nematode proteome. The unrecognized proteins seem to be key antigenic epitopes able to induce protective immune responses. The proteome changes were associated with weak immune recognition and increased larval adaptation and worm growth, altered localization in the intestine and increased survival of males but reduced worm fecundity. In this report, the mechanisms influencing nematode survival and the consequences of changed immunogenicity that reflect the immune response at the site colonized by the parasite in mice with colitis are described. The results are relevant to the use of live parasites to ameliorate IBD.
Collapse
|
10
|
Iwata JI, Hacia JG, Suzuki A, Sanchez-Lara PA, Urata M, Chai Y. Modulation of noncanonical TGF-β signaling prevents cleft palate in Tgfbr2 mutant mice. J Clin Invest 2012; 122:873-85. [PMID: 22326956 DOI: 10.1172/jci61498] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 01/04/2012] [Indexed: 11/17/2022] Open
Abstract
Patients with mutations in either TGF-β receptor type I (TGFBR1) or TGF-β receptor type II (TGFBR2), such as those with Loeys-Dietz syndrome, have craniofacial defects and signs of elevated TGF-β signaling. Similarly, mutations in TGF-β receptor gene family members cause craniofacial deformities, such as cleft palate, in mice. However, it is unknown whether TGF-β ligands are able to elicit signals in Tgfbr2 mutant mice. Here, we show that loss of Tgfbr2 in mouse cranial neural crest cells results in elevated expression of TGF-β2 and TGF-β receptor type III (TβRIII); activation of a TβRI/TβRIII-mediated, SMAD-independent, TRAF6/TAK1/p38 signaling pathway; and defective cell proliferation in the palatal mesenchyme. Strikingly, Tgfb2, Tgfbr1 (also known as Alk5), or Tak1 haploinsufficiency disrupted TβRI/TβRIII-mediated signaling and rescued craniofacial deformities in Tgfbr2 mutant mice, indicating that activation of this noncanonical TGF-β signaling pathway was responsible for craniofacial malformations in Tgfbr2 mutant mice. Thus, modulation of TGF-β signaling may be beneficial for the prevention of congenital craniofacial birth defects.
Collapse
Affiliation(s)
- Jun-ichi Iwata
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry, University of Southern California, Los Angeles, California 90033, USA
| | | | | | | | | | | |
Collapse
|
11
|
Wang X, Chen W, Li X, Zhou C, Deng C, Lv X, Fan Y, Men J, Liang C, Yu X. Identification and molecular characterization of a novel signaling molecule 14-3-3 epsilon in Clonorchis sinensis excretory/secretory products. Parasitol Res 2011; 110:1411-20. [DOI: 10.1007/s00436-011-2642-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2011] [Accepted: 09/02/2011] [Indexed: 11/29/2022]
|
12
|
Ghaffari A, Li Y, Kilani RT, Ghahary A. 14-3-3 sigma associates with cell surface aminopeptidase N in the regulation of matrix metalloproteinase-1. J Cell Sci 2010; 123:2996-3005. [PMID: 20699358 DOI: 10.1242/jcs.069484] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in the degradation of the extracellular matrix during development and tissue repair, as well as in pathological conditions such as tumor invasion and fibrosis. MMP expression by stromal cells is partly regulated by signals from the neighboring epithelial cells. Keratinocyte-releasable 14-3-3sigma, or stratifin, acts as a potent MMP-1-stimulatory factor in fibroblasts. However, its mechanism of transmembrane signaling remains unknown. Ectodomain biotin labeling, serial affinity purification and mass spectroscopy analysis revealed that the stratifin associates with aminopeptidase N (APN), or CD13, at the cell surface. The transient knockdown of APN in fibroblasts eliminated the stratifin-mediated p38 MAP kinase activation and MMP-1 expression, implicating APN in a receptor-mediated transmembrane signaling event. Stratifin deletion studies implicated its C-terminus as a potential APN-binding site. Furthermore, the dephosphorylation of APN ectodomains reduced its binding affinity to the stratifin. The presence of a phosphorylated serine or threonine residue in APN has been implicated. Together, these findings provide evidence that APN is a novel cell surface receptor for stratifin and a potential target in the regulation of MMP-1 expression in epithelial-stromal cell communication.
Collapse
Affiliation(s)
- Abdi Ghaffari
- Department of Surgery, BC Professional Firefighter's Burn and Wound Healing Research Laboratory, University of British Columbia, 344A JBRC, 2660 Oak Street, Vancouver, Canada, BC V6H 3Z6
| | | | | | | |
Collapse
|
13
|
Lu J, Guo H, Treekitkarnmongkol W, Li P, Zhang J, Shi B, Ling C, Zhou X, Chen T, Chiao PJ, Feng X, Seewaldt VL, Muller WJ, Sahin A, Hung MC, Yu D. 14-3-3zeta Cooperates with ErbB2 to promote ductal carcinoma in situ progression to invasive breast cancer by inducing epithelial-mesenchymal transition. Cancer Cell 2009; 16:195-207. [PMID: 19732720 PMCID: PMC2754239 DOI: 10.1016/j.ccr.2009.08.010] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2008] [Revised: 05/20/2009] [Accepted: 08/11/2009] [Indexed: 12/16/2022]
Abstract
ErbB2, a metastasis-promoting oncoprotein, is overexpressed in approximately 25% of invasive/metastatic breast cancers, but in 50%-60% of noninvasive ductal carcinomas in situ (DCIS). It has been puzzling how a subset of ErbB2-overexpressing DCIS develops into invasive breast cancer (IBC). We found that co-overexpression of 14-3-3zeta in ErbB2-overexpressing DCIS conferred a higher risk of progression to IBC. ErbB2 and 14-3-3zeta overexpression, respectively, increased cell migration and decreased cell adhesion, two prerequisites of tumor cell invasion. 14-3-3zeta overexpression reduced cell adhesion by activating the TGF-beta/Smads pathway that led to ZFHX1B/SIP-1 upregulation, E-cadherin loss, and epithelial-mesenchymal transition. Importantly, patients whose breast tumors overexpressed both ErbB2 and 14-3-3zeta had higher rates of metastatic recurrence and death than those whose tumors overexpressed only one.
Collapse
Affiliation(s)
- Jing Lu
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hua Guo
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Warapen Treekitkarnmongkol
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ping Li
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jian Zhang
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Bin Shi
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Chen Ling
- Molecular Oncology Group, McGill University Health Center, Montreal, Quebec, H3A 1A1, Canada
| | - Xiaoyan Zhou
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Tongzhen Chen
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Paul J. Chiao
- Department of Surgical Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xinhua Feng
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - William J. Muller
- Molecular Oncology Group, McGill University Health Center, Montreal, Quebec, H3A 1A1, Canada
| | - Aysegul Sahin
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Mien-Chie Hung
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
- China Medical University & Hospital, Taichung, 404, Taiwan
| | - Dihua Yu
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
- To whom correspondence should be addressed. , TEL: 713-792-3636, FAX: 713-792-4454
| |
Collapse
|
14
|
Interaction of hookworm 14-3-3 with the forkhead transcription factor DAF-16 requires intact Akt phosphorylation sites. Parasit Vectors 2009; 2:21. [PMID: 19393088 PMCID: PMC2683825 DOI: 10.1186/1756-3305-2-21] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2009] [Accepted: 04/24/2009] [Indexed: 01/29/2023] Open
Abstract
Background Third-stage infective larvae (L3) of hookworms are in an obligatory state of developmental arrest that ends upon entering the definitive host, where they receive a signal that re-activates development. Recovery from the developmentally arrested dauer stage of Caenorhabditis elegans is analogous to the resumption of development during hookworm infection. Insulin-like signaling (ILS) mediates recovery from arrest in C. elegans and activation of hookworm dauer L3. In C. elegans, phosphorylation of the forkhead transcription factor DAF-16 in response to ILS creates binding cites for the 14-3-3 protein Ce-FTT-2, which translocates DAF-16 out of the nucleus, resulting in resumption of reproductive development. Results To determine if hookworm 14-3-3 proteins play a similar role in L3 activation, hookworm FTT-2 was identified and tested for its ability to interact with A. caninum DAF-16 in vitro. The Ac-FTT-2 amino acid sequence was 91% identical to the Ce-FTT-2, and was most closely related to FTT-2 from other nematodes. Ac-FTT-2 was expressed in HEK 293T cells, and was recognized by an antibody against human 14-3-3β isoform. Reciprocal co-immunoprecipitations using anti-epitope tag antibodies indicated that Ac-FTT-2 interacts with Ac-DAF-16 when co-expressed in serum-stimulated HEK 293T cells. This interaction requires intact Akt consensus phosphorylation sites at serine107 and threonine312, but not serine381. Ac-FTT-2 was undetectable by Western blot in excretory/secretory products from serum-stimulated (activated) L3 or adult A. caninum. Conclusion The results indicate that Ac-FTT-2 interacts with DAF-16 in a phosphorylation-site dependent manner, and suggests that Ac-FTT-2 mediates activation of L3 by binding Ac-DAF-16 during hookworm infection.
Collapse
|
15
|
Fibroblast growth factor receptor 2 phosphorylation on serine 779 couples to 14-3-3 and regulates cell survival and proliferation. Mol Cell Biol 2008; 28:3372-85. [PMID: 18332103 DOI: 10.1128/mcb.01837-07] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The fibroblast growth factors (FGFs) exert their diverse (or pleiotropic) biological responses through the binding and activation of specific cell surface receptors (FGFRs). While FGFRs are known to initiate intracellular signaling through receptor tyrosine phosphorylation, the precise mechanisms by which the FGFRs regulate pleiotropic biological responses remain unclear. We now identify a new mechanism by which FGFR2 is able to regulate intracellular signaling and cellular responses. We show that FGFR2 is phosphorylated on serine 779 (S779) in response to FGF2. S779, which lies adjacent to the phospholipase Cgamma binding site at Y766, provides a docking site for the 14-3-3 phosphoserine-binding proteins and is essential for the full activation of the phosphatidylinositol 3-kinase and Ras/mitogen-activated protein kinase pathways. Furthermore, S779 signaling is essential for promoting cell survival and proliferation in both Ba/F3 cells and BALB/c 3T3 fibroblasts. This new mode of FGFR2 phosphoserine signaling via the 14-3-3 proteins may provide an increased repertoire of signaling outputs to allow the regulation of pleiotropic biological responses. In this regard, we have identified conserved putative phosphotyrosine/phosphoserine motifs in the cytoplasmic domains of diverse cell surface receptors, suggesting that they may perform important functional roles beyond the FGFRs.
Collapse
|
16
|
Bahia D, Andrade LF, Ludolf F, Mortara RA, Oliveira G. Protein tyrosine kinases in Schistosoma mansoni. Mem Inst Oswaldo Cruz 2008; 101 Suppl 1:137-43. [PMID: 17308761 DOI: 10.1590/s0074-02762006000900022] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Accepted: 06/26/2006] [Indexed: 11/22/2022] Open
Abstract
The identification and description of signal transduction molecules and mechanisms are essential to elucidate Schistosoma mansoni host-parasite interactions and parasite biology. This mini review focuses on recent advancements in the study of signalling molecules and transduction mechanisms in S. mansoni, drawing special attention to the recently identified and characterised protein tyrosine kinases of S. mansoni.
Collapse
Affiliation(s)
- Diana Bahia
- Centro de Pesquisas René Rachou-Fiocruz, Av. Augusto de Lima 1715, Barro Preto, 31190-002 Belo Horizonte, MG, Brazil.
| | | | | | | | | |
Collapse
|
17
|
Siles-Lucas M, Merli M, Gottstein B. 14-3-3 proteins in Echinococcus: their role and potential as protective antigens. Exp Parasitol 2008; 119:516-523. [PMID: 18316081 DOI: 10.1016/j.exppara.2008.01.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2007] [Revised: 01/22/2008] [Accepted: 01/28/2008] [Indexed: 10/22/2022]
Abstract
14-3-3 Proteins are a family of highly conserved proteins among all eukaryotic organisms studied so far. As basically intracellular proteins, they play a key role in basic cellular events related to cellular proliferation, including signal transduction, cell-cycle control, cell differentiation and cell survival. The 14-3-3 proteins have been described and characterized in several parasites, and mostly studied in Echinococcus granulosus and Echinococcus multilocularis. Here, we review the discoveries regarding this protein family in the genus Echinococcus, describing new data about specific aspects related with their implication in the parasite biology and immunology in the frame of the host-parasite relationship.
Collapse
Affiliation(s)
- Mar Siles-Lucas
- Unidad de Patología Animal, Instituto de Recursos Naturales y Agrobiología de Salamanca (IRNASA), Consejo Superior de Investigaciones Científicas (CSIC), Cordel de Merinas 40-52, 37008 Salamanca, Spain.
| | | | | |
Collapse
|
18
|
Knobloch J, Beckmann S, Burmeister C, Quack T, Grevelding CG. Tyrosine kinase and cooperative TGFβ signaling in the reproductive organs of Schistosoma mansoni. Exp Parasitol 2007; 117:318-36. [PMID: 17553494 DOI: 10.1016/j.exppara.2007.04.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 04/06/2007] [Accepted: 04/16/2007] [Indexed: 10/23/2022]
Abstract
Drug-induced suppression of female schistosome sexual maturation is an auspicious strategy to combat schistosomiasis since the eggs are the causative agent. The establishment of drug targets requires knowledge about the molecular mechanisms that regulate the development of the female reproductive organs, which include vitellarium and ovary. This review summarizes recent studies suggesting tyrosine kinases as important factors for the regulation of female gonad development. In this context, especially cytoplasmatic tyrosine kinases of the Src class seem to play dominant roles. Moreover, experimental data and theoretical concepts are provided supporting a crosstalk between tyrosine kinase and TGFbeta signaling in the production of vitellocytes. Finally, we take advantage from the schistosome genome project to propose a model for the regulation of vitelline-cell production and differentiation.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Institute for Animal Developmental and Molecular Biology, Heinrich-Heine-University, D-40225 Düsseldorf, Germany.
| | | | | | | | | |
Collapse
|
19
|
Kusel JR, Al-Adhami BH, Doenhoff MJ. The schistosome in the mammalian host: understanding the mechanisms of adaptation. Parasitology 2007; 134:1477-526. [PMID: 17572930 DOI: 10.1017/s0031182007002971] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
SUMMARYIn this review, we envisage the host environment, not as a hostile one, since the schistosome thrives there, but as one in which the relationship between the two organisms consists of constant communication, through signalling mechanisms involving sense organs, surface glycocalyx, surface membrane and internal organs of the parasite, with host fluids and cells. The surface and secretions of the schistosome egg have very different properties from those of other parasite stages, but adapted for the dispersal of the eggs and for the preservation of host liver function. We draw from studies of mammalian cells and other organisms to indicate how further work might be carried out on the signalling function of the surface glycocalyx, the raft structure of the surface and existence of pores in the surface membrane, the repair of the surface membrane, the role of the membrane structure in ion channel function (including recent work on the actin cytoskeleton and calcium channels) and the possible role of P-glycoproteins in the adaptation of the parasite to its environment. We are speculative in some areas, such as the suggestions that variability in surface properties of schistosomes may relate to the existence of membrane rafts and that parasite communities may exhibit quorum sensing. This speculative approach is adopted with the hope that future work on the whole organisms and their interactions will be encouraged.
Collapse
Affiliation(s)
- J R Kusel
- Glasgow Biomedical Research Centre, University of Glasgow, Glasgow G12 8TA, UK.
| | | | | |
Collapse
|
20
|
Loverde PT, Osman A, Hinck A. Schistosoma mansoni: TGF-beta signaling pathways. Exp Parasitol 2007; 117:304-17. [PMID: 17643432 PMCID: PMC2149906 DOI: 10.1016/j.exppara.2007.06.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2007] [Revised: 05/25/2007] [Accepted: 06/05/2007] [Indexed: 10/23/2022]
Abstract
Schistosome parasites have co-evolved an intricate relationship with their human and snail hosts as well as a novel interplay between the adult male and female parasites. We review the role of the TGF-beta signaling pathway in parasite development, host-parasite interactions and male-female interactions. The data to date support multiple roles for the TGF-beta signaling pathway throughout schistosome development, in particular, in the tegument which is at the interface with the host and between the male and female schistosome, development of vitelline cells in female worms whose genes and development are regulated by a stimulus from the male schistosome and embryogenesis of the egg. The human ligand TGF-beta1 has been demonstrated to regulate the expression of a schistosome target gene that encodes a gynecophoric canal protein in the schistosome worm itself. Studies on signaling in schistosomes opens a new era for investigation of host-parasite and male-female interactions.
Collapse
Affiliation(s)
- Philip T Loverde
- Department of Biochemistry, University of Texas Health Science Center, San Antonio, TX 78229-3900, USA.
| | | | | |
Collapse
|
21
|
Uribe N, Siles-Lucas M, López-Abán J, Esteban A, Suarez L, Martínez-Fernández A, del Olmo E, Muro A. The Sb14-3-3ζ recombinant protein protects against Schistosoma bovis in BALB/c mice. Vaccine 2007; 25:4533-9. [PMID: 17485147 DOI: 10.1016/j.vaccine.2007.04.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Revised: 03/05/2007] [Accepted: 04/02/2007] [Indexed: 11/20/2022]
Abstract
Schistosoma bovis is a trematode parasite mainly affecting cattle and sheep. Evidences about the arise of drug resistance and the high rates of re-infection of animals in endemic areas have pointed out the need of developing new control tools, e.g., effective vaccines. Schistosomes 14-3-3 proteins have been defined as vaccine candidates against respective infections. We have therefore investigated the protective capacity of the 14-3-3 protein from S. bovis - Sb14zeta - against S. bovis in mice. In addition, we have addressed the influence of the co-administration of four different immunomodulators with the 14-3-3 polypeptide. The values of protection against S. bovis were statistically significant when the Sb14zeta was combined in two independent experiments with the AA0029 (61.0% and 40.31%), AA2829 (49% and 36.3%) and PAL (49% and 40.075%) immunomodulatory molecules. Immune responses from vaccinated animals showed that the highest protection rates do not necessarily match with a dominant Th1-type response.
Collapse
Affiliation(s)
- N Uribe
- Laboratorio de Parasitología, Universidad de Salamanca, Avda Campo Charro, s/n, 37007 Salamanca, Spain
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Vermeire JJ, Humphries JE, Yoshino TP. Signal transduction in larval trematodes: putative systems associated with regulating larval motility and behaviour. Parasitology 2006; 131 Suppl:S57-70. [PMID: 16569293 DOI: 10.1017/s0031182005008358] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The multi-host lifestyle of parasitic trematodes necessitates their ability to communicate with their external environment in order to invade and navigate within their hosts' internal environment. Through recent EST and genome sequencing efforts, it has become clear that members of the Trematoda possess many of the elaborate signal transduction systems that have been delineated in other invertebrate model systems like Drosophila melanogaster and Caenorhabditis elegans. Gene homologues representing several well-described signal receptor families including receptor tyrosine kinases, receptor serine tyrosine kinases, G protein-coupled receptors and elements of their downstream signalling systems have been identified in larval trematodes. A majority of this work has focused on the blood flukes, Schistosoma spp. and therefore represents a narrow sampling of the diverse digenean helminth taxon. Despite this fact and given the substantial evidence supporting the existence of such signalling systems, the question then becomes, how are these systems employed by larval trematodes to aid them in interpreting signals received from their immediate environment to initiate appropriate responses in cells and tissues comprising the developing parasite stages? High-throughput, genome-wide analysis tools now allow us to begin to functionally characterize genes differentially expressed throughout the development of trematode larvae. Investigation of the systems used by these parasites to receive and transduce external signals may facilitate the creation of technologies for achieving control of intramolluscan schistosome infections and also continue to yield valuable insights into the basic mechanisms regulating motility and behaviour in this important group of helminths.
Collapse
Affiliation(s)
- J J Vermeire
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, 2115 Observatory Drive, Madison, WI 53706, USA
| | | | | |
Collapse
|
23
|
Knobloch J, Rossi A, Osman A, LoVerde PT, Klinkert MQ, Grevelding CG. Cytological and biochemical evidence for a gonad-preferential interplay of SmFKBP12 and SmTβR-I in Schistosoma mansoni. Mol Biochem Parasitol 2004; 138:227-36. [PMID: 15555734 DOI: 10.1016/j.molbiopara.2004.09.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2004] [Revised: 09/20/2004] [Accepted: 09/30/2004] [Indexed: 11/19/2022]
Abstract
In eukaryotes, FK506-binding proteins with a molecular weight of 12 kDa (FKBP12s) influence a variety of signal transduction pathways that regulate cell division, differentiation, and ion homeostasis. Amongst these, TGFbeta signaling and calcineurin (CN) phosphatase activity is modulated by FKBP12 via binding to TGFbeta-family type I receptors (TbetaR-Is) or to the CN subunit A, respectively. In this work, we demonstrate the tissue-specific expression of the Schistosoma mansoni FKBP12 homologue (SmFKBP12) in the gonads of female parasites as well as in the tegument of both genders. Components of the TGFbeta pathway have been characterized in schistosomes and their roles in mediating host-parasite or male-female interactions proposed. We show that a schistosome TGFbeta-family type I receptor (SmTbetaR-I, SmRK-1) is expressed in the female gonads, suggesting that SmFKBP12 may regulate its activity in this tissue. This hypothesis is supported by yeast two-hybrid analyses showing a direct binding of SmFKBP12 and SmTbetaR-I, which was specifically inhibited by the drug FK506. Our data provide the first evidence for the activity of a transmembrane receptor in the vitellarium of schistosome females and indicate that FKBP12-meditated regulation of the TGFbeta pathway is evolutionarily conserved in a primitive metazoan such as Schistosoma. Furthermore, we show that the schistosome CN (SmCN) is not expressed in the female gonads, but co-localizes with SmFKBP12 only in the tegument. From these data we conclude an SmFKBP12/SmTbetaR-I, but not an SmCN/SmFKBP12 interplay in the female gonads.
Collapse
Affiliation(s)
- Jürgen Knobloch
- Institut für Genetik, Genetische Parasitologie und Biologisch-Medizinisches-Forschungszentrum, Heinrich-Heine-Universität, 40225 Düsseldorf, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Abstract
Transforming growth factor-betas (TGF-betas) regulate pivotal cellular processes such as proliferation, differentiation and apoptosis. After ligand binding, the signals are transmitted by two types of transmembrane serine/threonine kinase receptors. The type I receptor phosphorylates Smad proteins, intracellular effectors which upon oligomerization enter the nucleus to regulate transcription following assembly with transcriptional co-factors and co-modulators. The cellular distribution of TGF-beta receptors along with their oligomerization mode and their complex formation with different cell surface receptors represent crucial steps in determining the initiation of distinct signalling cascades. In addition, the broad array of intracellular proteins that influence the TGF-beta pathway demonstrates that signal transduction does not proceed in a linear fashion but rather comprises a complex network of cascades that mutually influence each other. The present review describes the intricate control of TGF-beta signal transduction on various levels of the cascade with particular focus (i) on the assembly of different receptor subtypes and (ii) on the multitude of crosstalk with signal transducers from other pathways. Integration of the TGF-beta/Smad pathway into the signalling network has taken on added importance as it substantially contributes to elicit the plethora of cell- and tissue-specific effects of TGF-beta.
Collapse
Affiliation(s)
- Marion Lutz
- Department of Physiological Chemistry II, Biocenter, University of Würzburg, 97074 Würzburg, Germany
| | | |
Collapse
|
25
|
McGonigle S, Loschiavo M, Pearce EJ. 14-3-3 proteins in Schistosoma mansoni; identification of a second epsilon isoform. Int J Parasitol 2002; 32:685-93. [PMID: 12062487 DOI: 10.1016/s0020-7519(01)00323-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
A new member of the 14-3-3 protein family in Schistosoma mansoni has been identified. Sequence analysis demonstrated that this protein is a member of the epsilon sub-group and is the orthologue of Schistosoma japonicum 14-3-3epsilon. Since we had previously identified a 14-3-3epsilon protein from S. mansoni, we termed the original protein 14-3-3epsilon-1 and this second epsilon protein 14-3-3epsilon-2. Schistosoma mansoni encodes at least four different 14-3-3 isoforms: the two epsilon proteins and 14-3-3 protein 1 and protein 2, which are zeta-like isoforms. Phylogenetic analysis demonstrated the early divergence of the epsilon isoforms, and that schistosome proteins 1 and 2 are among the oldest non-epsilon 14-3-3 proteins yet identified. Schistosoma mansoni 14-3-3epsilon-1, 14-3-3epsilon-2, and protein 1 are stage specifically expressed in a similar manner, being absent in cercariae and schistosomula, and abundant in lung stage and adult male and female worms. Protein 2 transcript was not detected at any of the life cycle stages examined. All three detected 14-3-3 isoforms elicit an immune response during infection, with the greatest response directed against protein 1. Binding studies with S. mansoni receptor kinase-1 (SmRK1) and human Raf kinase revealed that the three 14-3-3epsilon isoforms exhibit a preference for target protein binding. Although all three isoforms do bind to both targets, 14-3-3 protein 1 interacts most strongly with Raf, whereas the 14-3-3-1 isoform binds SmRK1 preferentially. These results suggest that the individual 14-3-3 proteins may have evolved to play isoform-specific roles in the development and survival of S. mansoni within its host.
Collapse
Affiliation(s)
- Sharon McGonigle
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, NY 14853, USA
| | | | | |
Collapse
|
26
|
McGonigle S, Beall MJ, Pearce EJ. Eukaryotic initiation factor 2 alpha subunit associates with TGF beta receptors and 14-3-3 epsilon and acts as a modulator of the TGF beta response. Biochemistry 2002; 41:579-87. [PMID: 11781097 DOI: 10.1021/bi011407z] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Schistosoma mansoni receptor kinase 1 (SmRK1) is a divergent member of the TGF beta receptor family. Intracellular proteins that associate with these receptors are likely to play an important role in signaling. 14-3-3 epsilon is a previously described cytoplasmic protein, which associates with both SmRK1 and the human type I TGF beta receptor (T beta RI); overexpression of 14-3-3 epsilon leads to enhanced TGF beta-mediated signaling by T beta RI. We now describe the identification of S. mansoni eukaryotic translation initiation factor 2 alpha subunit (eIF2 alpha), through its interaction with SmRK1 in a yeast two-hybrid assay. S. mansoni eIF2 alpha also interacts with human TGF beta receptors. Strongest association was demonstrated with kinase inactive receptors, particularly the type II TGF beta receptor (T beta RII). Both T beta RI and T beta RII phosphorylate eIF2 alpha in vitro, at sites other than the previously described eIF2 alpha phosphorylation sites. EIF2 alpha also modulates signaling by TGF beta receptors; however, in contrast to 14-3-3 epsilon, eIF2 alpha overexpression inhibits the TGF beta-driven response. These data suggest a novel function for eIF2 alpha in the TGF beta signaling pathway. In addition, we have demonstrated an independent interaction between eIF2 alpha and 14-3-3 epsilon. Coexpression of 14-3-3 epsilon with eIF2 alpha leads to the abrogation of the inhibitory effect of eIF2 alpha on TGF beta-mediated signaling. The interaction of these two regulatory proteins with each other and with the TGF beta receptors and their relative expression levels are likely to be important in fine-tuning the regulation of TGF beta signal transduction.
Collapse
Affiliation(s)
- Sharon McGonigle
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | | | | |
Collapse
|
27
|
Abstract
Smad proteins transduce signals from transforming growth factor-β (TGF-β) superfamily ligands that regulate cell proliferation, differentiation and death through activation of receptor serine/threonine kinases. Phosphorylation of receptor-activated Smads (R-Smads) leads to formation of complexes with the common mediator Smad (Co-Smad), which are imported to the nucleus. Nuclear Smad oligomers bind to DNA and associate with transcription factors to regulate expression of target genes. Alternatively, nuclear R-Smads associate with ubiquitin ligases and promote degradation of transcriptional repressors, thus facilitating target gene regulation by TGF-β. Smads themselves can also become ubiquitinated and are degraded by proteasomes. Finally, the inhibitory Smads (I-Smads) block phosphorylation of R-Smads by the receptors and promote ubiquitination and degradation of receptor complexes, thus inhibiting signalling.
Collapse
Affiliation(s)
- A Moustakas
- Ludwig Institute for Cancer Research, Box 595, SE-751 24 Uppsala, Sweden.
| | | | | |
Collapse
|
28
|
Beall MJ, Pearce EJ. Human transforming growth factor-beta activates a receptor serine/threonine kinase from the intravascular parasite Schistosoma mansoni. J Biol Chem 2001; 276:31613-9. [PMID: 11406634 DOI: 10.1074/jbc.m104685200] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The biology of the helminth parasite Schistosoma mansoni is closely integrated with that of its mammalian host. SmRK1, a divergent type I transforming growth factor-beta (TGF-beta) receptor of unknown ligand specificity, was previously identified as a candidate for a receptor that allows schistosomes to respond to host-derived growth factors. The TGF-beta family includes activin, bone morphogenetic proteins (BMPs), and TGF-beta, all of which can play crucial roles in metazoan development. The downstream signaling protein of receptors that respond to TGF-beta and activin is Smad2, whereas the receptors that respond to BMPs signal via Smad1. When a constitutively active mutant of SmRK1 was overexpressed with either schistosome Smad1 (SmSmad1) or SmSmad2, a receptor-dependent modulation of SmSmad phosphorylation and luciferase reporter activity occurred only with SmSmad2. To evaluate potential ligand activators of SmRK1, a chimeric receptor containing the extracellular domain of SmRK1 joined to the intracellular domain of the human type I TGF-beta receptor was used. The chimeric receptor bound radiolabeled TGF-beta and could activate a luciferase reporter gene in response to both TGF-beta 1 and TGF-beta 3 but not BMP7. Confirmatory results were obtained using full-length SmRK1. These experiments implicate TGF-beta as a ligand for SmRK1 and as a potential host-derived regulator of parasite growth and development.
Collapse
Affiliation(s)
- M J Beall
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, USA
| | | |
Collapse
|
29
|
McGonigle S, Feeney EL, Beall MJ, Pearce EJ. SIP, a novel SH3 domain-containing protein, interacts with Schistosoma mansoni receptor kinase 1. Mol Biochem Parasitol 2001; 114:119-23. [PMID: 11356521 DOI: 10.1016/s0166-6851(01)00232-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- S McGonigle
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | |
Collapse
|