1
|
Malka M, Czaczkes I, Kashkash S, Shachar S, Bacharach E, Ehrlich M. Inhibition of early EHDV2-Ibaraki infection steps in bovine cells by endosome alkalinization or ikarugamycin, but not by blockage of individual endocytic pathways. Front Cell Infect Microbiol 2025; 15:1494200. [PMID: 39981379 PMCID: PMC11839642 DOI: 10.3389/fcimb.2025.1494200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 01/13/2025] [Indexed: 02/22/2025] Open
Abstract
The Epizootic hemorrhagic disease virus (EHDV), an orbivirus, is the etiological factor of a fatal hemorrhagic disease of wild ruminants. A subset of EHDV serotypes, including the Ibaraki strain of EHDV2 (EHDV2-Ibaraki), infect and cause disease in cattle, thus posing a potential threat to livestock. As a member of the Sedoreoviridae family, the EHDV particle is devoid of a membrane envelope and is predicted to employ endocytic pathways for infection. However, the degree of dependence of EHDV2-Ibaraki on specific internalization pathways while infecting bovine cells (its natural host) is unknown. The endosome alkalinizing agent ammonium chloride blocked EHDV2-Ibaraki infection of Madin-Darby Bovine Kidney (MDBK) cells with dependence on its time of addition, suggesting the criticality of endosomal pH for the completion of early stages of infection. Treatment of cells within the alkalinization-sensitive window (i.e., before endosomal processing) with inhibitors of actin polymerization, macropinocytosis (amiloride), or dynamin GTPase activity (dynasore or dynole), or with the cholesterol-depleting agent methyl-β-cyclodextrin, failed to reduce EHDV2-Ibaraki infection. In contrast, in this same treatment time frame, ikarugamycin potently inhibited infection. Moreover, ikarugamycin inhibited interferon induction in infected cells and induced the accumulation of enlarged Rab7- and lamtor4-decorated vacuoles, suggesting its ability to block viral processing and modify late-endosome compartments. Notably, ikarugamycin treatment at initial infection stages, augmented the infection of MDBK cells with the vesicular stomatitis virus while inhibiting infection with bluetongue virus serotype 8. Together, our results point to differential antiviral effects of ikarugamycin on viruses dependent on distinct sets of endosomes for entry/processing.
Collapse
Affiliation(s)
| | | | | | | | | | - Marcelo Ehrlich
- The Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv-Yafo, Israel
| |
Collapse
|
2
|
Tai SB, Huang CY, Chung CL, Sung PJ, Wen ZH, Chen CL. Prodigiosin Inhibits Transforming Growth Factor β Signaling by Interfering Receptor Recycling and Subcellular Translocation in Epithelial Cells. Mol Pharmacol 2024; 105:286-300. [PMID: 38278554 DOI: 10.1124/molpharm.123.000776] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/05/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024] Open
Abstract
Prodigiosin (PG) is a naturally occurring polypyrrole red pigment produced by numerous microorganisms including some Serratia and Streptomyces strains. PG has exhibited promising anticancer activity; however, the molecular mechanisms of action of PG on malignant cells remain ambiguous. Transforming growth factor-β (TGF-β) is a multifunctional cytokine that governs a wide array of cellular processes in development and tissue homeostasis. Malfunctions of TGF-β signaling are associated with numerous human cancers. Emerging evidence underscores the significance of internalized TGF-β receptors and their intracellular trafficking in initiating signaling cascades. In this study, we identified PG as a potent inhibitor of the TGF-β pathway. PG blocked TGF-β signaling by targeting multiple sites of this pathway, including facilitating the sequestering of TGF-β receptors in the cytoplasm by impeding the recycling of type II TGF-β receptors to the cell surface. Additionally, PG prompts a reduction in the abundance of receptors on the cell surface through the disruption of the receptor glycosylation. In human Caucasian lung carcinoma cells and human hepatocellular cancer cell line cells, nanomolar concentrations of PG substantially diminish TGF-β-triggered phosphorylation of Smad2 protein. This attenuation is further reflected in the suppression of downstream target gene expression, including those encoding fibronectin, plasminogen activator inhibitor-1, and N-cadherin. SIGNIFICANCE STATEMENT: Prodigiosin (PG) emerges from this study as a potent TGF-β pathway inhibitor, disrupting receptor trafficking and glycosylation and reducing TGF-β signaling and downstream gene expression. These findings not only shed light on PG's potential therapeutic role but also present a captivating avenue towards future anti-TGF-β strategies.
Collapse
Affiliation(s)
- Shun-Ban Tai
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Yin Huang
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Ling Chung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ping-Jyun Sung
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Zhi-Hong Wen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chun-Lin Chen
- Departments of Marine Biotechnology and Resources (S.-B.T., Z.-H.W.) and Biological Sciences (C.-L.Chu., C.-L.Che.), National Sun Yat-Sen University, Kaohsiung, Taiwan; Division of Rheumatology, Immunology and Allergy, Department of Internal Medicine, Zuoying Armed Forces General Hospital, Kaohsiung, Taiwan (S.-B.T.); Department of Orthopaedics, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan (C.-y.H.); National Museum of Marine Biology and Aquarium, Pingtung, Taiwan (P.-J.S.); and Department of Biotechnology (C.-L.Che.) and Graduate Institute of Natural Products, College of Pharmacy (C.-L.Che.), Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
3
|
Chaudhary R, Goodman LS, Wang S, Asimakopoulos A, Weiskirchen R, Dooley S, Ehrlich M, Henis YI. Cholesterol modulates type I/II TGF-β receptor complexes and alters the balance between Smad and Akt signaling in hepatocytes. Commun Biol 2024; 7:8. [PMID: 38168942 PMCID: PMC10761706 DOI: 10.1038/s42003-023-05654-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024] Open
Abstract
Cholesterol mediates membrane compartmentalization, affecting signaling via differential distribution of receptors and signaling mediators. While excessive cholesterol and aberrant transforming growth factor-β (TGF-β) signaling characterize multiple liver diseases, their linkage to canonical vs. non-canonical TGF-β signaling remained unclear. Here, we subjected murine hepatocytes to cholesterol depletion (CD) or enrichment (CE), followed by biophysical studies on TGF-β receptor heterocomplex formation, and output to Smad2/3 vs. Akt pathways. Prior to ligand addition, raft-dependent preformed heteromeric receptor complexes were observed. Smad2/3 phosphorylation persisted following CD or CE. CD enhanced phospho-Akt (pAkt) formation by TGF-β or epidermal growth factor (EGF) at 5 min, while reducing it at later time points. Conversely, pAkt formation by TGF-β or EGF was inhibited by CE, suggesting a direct effect on the Akt pathway. The modulation of the balance between TGF-β signaling to Smad2/3 vs. pAkt (by TGF-β or EGF) has potential implications for hepatic diseases and malignancies.
Collapse
Affiliation(s)
- Roohi Chaudhary
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Laureen S Goodman
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel
| | - Sai Wang
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167, Mannheim, Germany
| | - Anastasia Asimakopoulos
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, D-52074, Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH Aachen University Hospital, D-52074, Aachen, Germany
| | - Steven Dooley
- Department of Medicine II, University Medical Center Mannheim, Medical Faculty Mannheim, Heidelberg University, D-68167, Mannheim, Germany
| | - Marcelo Ehrlich
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 6997801, Tel Aviv, Israel.
| |
Collapse
|
4
|
Kolbe N, Hexemer L, Bammert LM, Loewer A, Lukáčová-Medvid’ová M, Legewie S. Data-based stochastic modeling reveals sources of activity bursts in single-cell TGF-β signaling. PLoS Comput Biol 2022; 18:e1010266. [PMID: 35759468 PMCID: PMC9269928 DOI: 10.1371/journal.pcbi.1010266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 07/08/2022] [Accepted: 05/30/2022] [Indexed: 11/30/2022] Open
Abstract
Cells sense their surrounding by employing intracellular signaling pathways that transmit hormonal signals from the cell membrane to the nucleus. TGF-β/SMAD signaling encodes various cell fates, controls tissue homeostasis and is deregulated in diseases such as cancer. The pathway shows strong heterogeneity at the single-cell level, but quantitative insights into mechanisms underlying fluctuations at various time scales are still missing, partly due to inefficiency in the calibration of stochastic models that mechanistically describe signaling processes. In this work we analyze single-cell TGF-β/SMAD signaling and show that it exhibits temporal stochastic bursts which are dose-dependent and whose number and magnitude correlate with cell migration. We propose a stochastic modeling approach to mechanistically describe these pathway fluctuations with high computational efficiency. Employing high-order numerical integration and fitting to burst statistics we enable efficient quantitative parameter estimation and discriminate models that assume noise in different reactions at the receptor level. This modeling approach suggests that stochasticity in the internalization of TGF-β receptors into endosomes plays a key role in the observed temporal bursting. Further, the model predicts the single-cell dynamics of TGF-β/SMAD signaling in untested conditions, e.g., successfully reflects memory effects of signaling noise and cellular sensitivity towards repeated stimulation. Taken together, our computational framework based on burst analysis, noise modeling and path computation scheme is a suitable tool for the data-based modeling of complex signaling pathways, capable of identifying the source of temporal noise.
Collapse
Affiliation(s)
- Niklas Kolbe
- Institute of Geometry and Practical Mathematics, RWTH Aachen University, Aachen, Germany
| | - Lorenz Hexemer
- Institute of Molecular Biology (IMB), Mainz, Germany
- Department of Systems Biology, Institute for Biomedical Genetics (IBMG), University of Stuttgart, Stuttgart, Germany
| | | | - Alexander Loewer
- Systems Biology of the Stress Response, Department of Biology, Technical University of Darmstadt, Darmstadt, Germany
| | | | - Stefan Legewie
- Department of Systems Biology, Institute for Biomedical Genetics (IBMG), University of Stuttgart, Stuttgart, Germany
- Stuttgart Research Center for Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
5
|
Chen SY, Mamai O, Akhurst RJ. TGFβ: Signaling Blockade for Cancer Immunotherapy. ANNUAL REVIEW OF CANCER BIOLOGY 2022; 6:123-146. [PMID: 36382146 PMCID: PMC9645596 DOI: 10.1146/annurev-cancerbio-070620-103554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Discovered over four decades ago, transforming growth factor β (TGFβ) is a potent pleiotropic cytokine that has context-dependent effects on most cell types. It acts as a tumor suppressor in some cancers and/or supports tumor progression and metastasis through its effects on the tumor stroma and immune microenvironment. In TGFβ-responsive tumors it can promote invasion and metastasis through epithelial-mesenchymal transformation, the appearance of cancer stem cell features, and resistance to many drug classes, including checkpoint blockade immunotherapies. Here we consider the biological activities of TGFβ action on different cells of relevance toward improving immunotherapy outcomes for patients, with a focus on the adaptive immune system. We discuss recent advances in the development of drugs that target the TGFβ signaling pathway in a tumor-specific or cell type–specific manner to improve the therapeutic window between response rates and adverse effects.
Collapse
Affiliation(s)
- Szu-Ying Chen
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Ons Mamai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Rosemary J. Akhurst
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
- Department of Anatomy, University of California, San Francisco, California, USA
| |
Collapse
|
6
|
Exploring ITM2A as a new potential target for brain delivery. Fluids Barriers CNS 2022; 19:25. [PMID: 35313913 PMCID: PMC8935840 DOI: 10.1186/s12987-022-00321-3] [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: 11/29/2021] [Accepted: 03/04/2022] [Indexed: 12/22/2022] Open
Abstract
Background Integral membrane protein 2A (ITM2A) is a transmembrane protein expressed in a variety of tissues; little is known about its function, particularly in the brain. ITM2A was found to be highly enriched in human brain versus peripheral endothelial cells by transcriptomic and proteomic studies conducted within the European Collaboration on the Optimization of Macromolecular Pharmaceutical (COMPACT) Innovative Medicines Initiative (IMI) consortium. Here, we report the work that was undertaken to determine whether ITM2A could represent a potential target for delivering drugs to the brain. Methods A series of ITM2A constructs, cell lines and specific anti-human and mouse ITM2A antibodies were generated. Binding and internalization studies in Human Embryonic Kidney 293 (HEK293) cells overexpressing ITM2A and in brain microvascular endothelial cells from mouse and non-human primate (NHP) were performed with these tools. The best ITM2A antibody was evaluated in an in vitro human blood brain barrier (BBB) model and in an in vivo mouse pharmacokinetic study to investigate its ability to cross the BBB. Results Antibodies specifically recognizing extracellular parts of ITM2A or tags inserted in its extracellular domain showed selective binding and uptake in ITM2A-overexpressing cells. However, despite high RNA expression in mouse and human microvessels, the ITM2A protein was rapidly downregulated when endothelial cells were grown in culture, probably explaining why transcytosis could not be observed in vitro. An attempt to directly demonstrate in vivo transcytosis in mice was inconclusive, using either a cross-reactive anti-ITM2A antibody or in vivo phage panning of an anti-ITM2A phage library. Conclusions The present work describes our efforts to explore the potential of ITM2A as a target mediating transcytosis through the BBB, and highlights the multiple challenges linked to the identification of new brain delivery targets. Our data provide evidence that antibodies against ITM2A are internalized in ITM2A-overexpressing HEK293 cells, and that ITM2A is expressed in brain microvessels, but further investigations will be needed to demonstrate that ITM2A is a potential target for brain delivery. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00321-3.
Collapse
|
7
|
Szilágyi SS, Gutman O, Henis YI. Complex Formation Among TGF-β Receptors in Live Cell Membranes Measured by Patch-FRAP. Methods Mol Biol 2022; 2488:23-34. [PMID: 35347680 DOI: 10.1007/978-1-0716-2277-3_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Signaling by receptors from the transforming growth factor-β (TGF-β) superfamily plays critical roles in multiple physiological and pathological processes. Their signaling requires complex formation between type I and type II receptors with Ser/Thr kinase activity, whereby the type II receptor phosphorylates and activates the relevant type I receptor(s), which transduces downstream signaling. It is therefore important to study complex formation among receptors from this family. In the current chapter, we use the type I (ALK5) and type II TGF-β receptors (TβRI and TβRII) as an example for measuring complex formation among cell-surface receptors in live cells by patch-FRAP, a variation of fluorescence recovery after photobleaching (FRAP).
Collapse
Affiliation(s)
- Szabina Szófia Szilágyi
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Orit Gutman
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
8
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
9
|
Characterization of DLBCL with a PMBL gene expression signature. Blood 2021; 138:136-148. [PMID: 33684939 DOI: 10.1182/blood.2020007683] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 02/12/2021] [Indexed: 12/13/2022] Open
Abstract
Primary mediastinal large B-cell lymphoma (PMBL) is a type of aggressive B-cell lymphoma that typically affects young adults, characterized by presence of a bulky anterior mediastinal mass. Lymphomas with gene expression features of PMBL have been described in nonmediastinal sites, raising questions about how these tumors should be classified. Here, we investigated whether these nonmediastinal lymphomas are indeed PMBLs or instead represent a distinct group within diffuse large B-cell lymphoma (DLBCL). From a cohort of 325 de novo DLBCL cases, we identified tumors from patients without evidence of anterior mediastinal involvement that expressed a PMBL expression signature (nm-PMBLsig+; n = 16; 5%). A majority of these tumors expressed MAL and CD23, proteins typically observed in bona fide PMBL (bf-PMBL). Evaluation of clinical features of nm-PMBLsig+ cases revealed close associations with DLBCL, and a majority displayed a germinal center B cell-like cell of origin (GCB). In contrast to patients with bf-PMBL, patients with nm-PMBLsig+ presented at an older age and did not show pleural disease, and bone/bone marrow involvement was observed in 3 cases. However, although clinically distinct from bf-PMBL, nm-PMBLsig+ tumors resembled bf-PMBL at the molecular level, with upregulation of immune response, JAK-STAT, and NF-κB signatures. Mutational analysis revealed frequent somatic gene mutations in SOCS1, IL4R, ITPKB, and STAT6, as well as CD83 and BIRC3, with the latter genes significantly more frequently affected than in GCB DLBCL or bf-PMBL. Our data establish nm-PMBLsig+ lymphomas as a group within DLBCL with distinct phenotypic and genetic features. These findings may have implications for gene expression- and mutation-based subtyping of aggressive B-cell lymphomas and related targeted therapies.
Collapse
|
10
|
Hackland J. Top-Down Inhibition (TDi) and Baseline Activation (BLa): Controlling Signal Transduction When Endogenous Cytokines are Ruining Your Differentiation. ACTA ACUST UNITED AC 2019; 51:e98. [PMID: 31756052 DOI: 10.1002/cpsc.98] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the 20 years since the first human pluripotent stem cell (hPSC) lines were established, there have been a plethora of protocols developed that allow us to generate a wide range of human cell types in vitro. Efforts to achieve a greater degree of specificity and efficiency in generating desired cell types have resulted in increasingly complex approaches. The magnitude and timing of signals has become key, and the concept of a "fully defined" system is a forever sought-after goal with shifting goalposts. This overview discusses two related approaches that can be used to deliver a tightly regulated, intermediate-strength signal, and which can also manage the impact of endogenous signaling variation and enable a switch away from bovine serum albumin-containing medium to a better-defined system without suffering a subsequent loss of robustness or efficiency. The approaches, referred to as top-down inhibition and baseline activation, were developed to deliver intermediate levels of BMP and WNT signaling during neural crest induction from hPSC, but could be applied to a variety of other signals and differentiation systems. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- James Hackland
- Developmental Biology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
11
|
Shapira KE, Ehrlich M, Henis YI. Cholesterol depletion enhances TGF-β Smad signaling by increasing c-Jun expression through a PKR-dependent mechanism. Mol Biol Cell 2018; 29:2494-2507. [PMID: 30091670 PMCID: PMC6233055 DOI: 10.1091/mbc.e18-03-0175] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 07/16/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor-β (TGF-β) plays critical roles in numerous physiological and pathological responses. Cholesterol, a major plasma membrane component, can have pronounced effects on signaling responses. Cells continually monitor cholesterol content and activate multilayered transcriptional and translational signaling programs, following perturbations to cholesterol homeostasis (e.g., statins, the commonly used cholesterol-reducing drugs). However, the cross-talk of such programs with ligand-induced signaling responses (e.g., TGF-β signaling) remained unknown. Here, we studied the effects of a mild reduction in free (membrane-associated) cholesterol on distinct components of TGF-β-signaling pathways. Our findings reveal a new regulatory mechanism that enhances TGF-β-signaling responses by acting downstream from receptor activation. Reduced cholesterol results in PKR-dependent eIF2α phosphorylation, which enhances c-Jun translation, leading in turn to higher levels of JNK-mediated c-Jun phosphorylation. Activated c-Jun enhances transcription and expression of Smad2/3. This leads to enhanced sensitivity to TGF-β stimulation, due to increased Smad2/3 expression and phosphorylation. The phospho/total Smad2/3 ratio remains unchanged, indicating that the effect is not due to altered receptor activity. We propose that cholesterol depletion induces overactivation of PKR, JNK, and TGF-β signaling, which together may contribute to the side effects of statins in diverse disease settings.
Collapse
Affiliation(s)
- Keren E. Shapira
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I. Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
12
|
Chung CL, Wang SW, Sun WC, Shu CW, Kao YC, Shiao MS, Chen CL. Sorafenib suppresses TGF-β responses by inducing caveolae/lipid raft-mediated internalization/degradation of cell-surface type II TGF-β receptors: Implications in development of effective adjunctive therapy for hepatocellular carcinoma. Biochem Pharmacol 2018; 154:39-53. [PMID: 29678520 DOI: 10.1016/j.bcp.2018.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/13/2018] [Indexed: 12/31/2022]
Abstract
Sorafenib is the only FDA approved drug for the treatment of advanced hepatocellular carcinoma (HCC) and other malignancies. Studies indicate that TGF-β signalling is associated with tumour progression in HCC. Autocrine and paracrine TGF-β promotes tumour growth and malignancy by inducing epithelial-mesenchymal transition (EMT). Sorafenib is believed to antagonize tumour progression by inhibiting TGF-β-induced EMT. It improves survival of patients but HCC later develops resistance and relapses. The underlying mechanism of resistance is unknown. Understanding of the molecular mechanism of sorafenib inhibition of TGF-β-induced signalling or responses in HCC may lead to development of adjunctive effective therapy for HCC. In this study, we demonstrate that sorafenib suppresses TGF-β responsiveness in hepatoma cells, hepatocytes, and animal liver, mainly by downregulating cell-surface type II TGF-β receptors (TβRII) localized in caveolae/lipid rafts and non-lipid raft microdomains via caveolae/lipid rafts-mediated internalization and degradation. Furthermore, sorafenib-induced downregulation and degradation of cell-surface TβRII is prevented by simultaneous treatment with a caveolae disruptor or lysosomal inhibitors. On the other hand, sorafenib only downregulates cell-surface TβRII localized in caveolae/lipid rafts but not localized in non-lipid raft microdomains in hepatic stellate cells. These results suggest that sorafenib inhibits TGF-β signalling mainly by inducing caveolae/lipid raft-mediated internalization and degradation of cell-surface TβR-II in target cells. They may also imply that treatment with agents which promote formation of caveolae/lipid rafts, TGF-β receptor kinase inhibitors (e.g., LY2157299) or TGF-β peptide antagonists (by liver-targeting delivery) may be considered as effective adjunct therapy with sorafenib for HCC.
Collapse
Affiliation(s)
- Chih-Ling Chung
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Shih-Wei Wang
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Wei-Chih Sun
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Chih-Wen Shu
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 81326, Taiwan, ROC
| | - Yu-Chen Kao
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC
| | - Meng-Shin Shiao
- Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10400, Thailand
| | - Chun-Lin Chen
- Department of Biological Sciences, National Sun Yat-sen University, Kaohsiung 80424, Taiwan, ROC; Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung 80424, Taiwan, ROC.
| |
Collapse
|
13
|
Yakymovych I, Yakymovych M, Heldin CH. Intracellular trafficking of transforming growth factor β receptors. Acta Biochim Biophys Sin (Shanghai) 2018; 50:3-11. [PMID: 29186283 DOI: 10.1093/abbs/gmx119] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Indexed: 02/06/2023] Open
Abstract
Transforming growth factor β (TGFβ) family members signal via heterotetrameric complexes of type I (TβRI) and type II (TβRII) dual specificity kinase receptors. The availability of the receptors on the cell surface is controlled by several mechanisms. Newly synthesized TβRI and TβRII are delivered from the Golgi apparatus to the cell surface via separate routes. On the cell surface, TGFβ receptors are distributed between different microdomains of the plasma membrane and can be internalized via clathrin- and caveolae-mediated endocytic mechanisms. Although receptor endocytosis is not essential for TGFβ signaling, localization of the activated receptor complexes on the early endosomes promotes TGFβ-induced Smad activation. Caveolae-mediated endocytosis, which is widely regarded as a mechanism that facilitates the degradation of TGFβ receptors, has been shown to be required for TGFβ signaling via non-Smad pathways. The importance of proper control of TGFβ receptor intracellular trafficking is emphasized by clinical data, as mislocalization of receptors has been described in connection with several human diseases. Thus, control of intracellular trafficking of the TGFβ receptors together with the regulation of their expression, posttranslational modifications and down-regulation, ensure proper regulation of TGFβ signaling.
Collapse
Affiliation(s)
- Ihor Yakymovych
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 75123, Sweden
| | - Mariya Yakymovych
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 75123, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala 75123, Sweden
| |
Collapse
|
14
|
Carrasco-Ramírez P, Greening DW, Andrés G, Gopal SK, Martín-Villar E, Renart J, Simpson RJ, Quintanilla M. Podoplanin is a component of extracellular vesicles that reprograms cell-derived exosomal proteins and modulates lymphatic vessel formation. Oncotarget 2017; 7:16070-89. [PMID: 26893367 PMCID: PMC4941298 DOI: 10.18632/oncotarget.7445] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 02/10/2016] [Indexed: 12/16/2022] Open
Abstract
Podoplanin (PDPN) is a transmembrane glycoprotein that plays crucial roles in embryonic development, the immune response, and malignant progression. Here, we report that cells ectopically or endogenously expressing PDPN release extracellular vesicles (EVs) that contain PDPN mRNA and protein. PDPN incorporates into membrane shed microvesicles (MVs) and endosomal-derived exosomes (EXOs), where it was found to colocalize with the canonical EV marker CD63 by immunoelectron microscopy. We have previously found that expression of PDPN in MDCK cells induces an epithelial-mesenchymal transition (EMT). Proteomic profiling of MDCK-PDPN cells compared to control cells shows that PDPN-induced EMT is associated with upregulation of oncogenic proteins and diminished expression of tumor suppressors. Proteomic analysis of exosomes reveals that MDCK-PDPN EXOs were enriched in protein cargos involved in cell adhesion, cytoskeletal remodeling, signal transduction and, importantly, intracellular trafficking and EV biogenesis. Indeed, expression of PDPN in MDCK cells stimulated both EXO and MV production, while knockdown of endogenous PDPN in human HN5 squamous carcinoma cells reduced EXO production and inhibited tumorigenesis. EXOs released from MDCK-PDPN and control cells both stimulated in vitro angiogenesis, but only EXOs containing PDPN were shown to promote lymphatic vessel formation. This effect was mediated by PDPN on the surface of EXOs, as demonstrated by a neutralizing specific monoclonal antibody. These results contribute to our understanding of PDPN-induced EMT in association to tumor progression, and suggest an important role for PDPN in EV biogenesis and/or release and for PDPN-EXOs in modulating lymphangiogenesis.
Collapse
Affiliation(s)
- Patricia Carrasco-Ramírez
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - David W Greening
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Germán Andrés
- Electron Microscopy Unit, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Shashi K Gopal
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Ester Martín-Villar
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Jaime Renart
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | - Richard J Simpson
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Miguel Quintanilla
- Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas (CSIC) - Universidad Autónoma de Madrid (UAM), Madrid, Spain
| |
Collapse
|
15
|
Abstract
Transforming growth factor β (TGF-β) and related ligands have potent effects on an enormous diversity of biological functions in all animals examined. Because of the strong conservation of TGF-β family ligand functions and signaling mechanisms, studies from multiple animal systems have yielded complementary and synergistic insights. In the nematode Caenorhabditis elegans, early studies were instrumental in the elucidation of TGF-β family signaling mechanisms. Current studies in C. elegans continue to identify new functions for the TGF-β family in this organism as well as new conserved mechanisms of regulation.
Collapse
Affiliation(s)
- Cathy Savage-Dunn
- Department of Biology, Queens College, and the Graduate Center, New York, New York 11367
| | - Richard W Padgett
- Waksman Institute, Department of Molecular Biology and Biochemistry, Cancer Institute of New Jersey, Rutgers University, Piscataway, New Jersey 08854-8020
| |
Collapse
|
16
|
Varadaraj A, Jenkins LM, Singh P, Chanda A, Snider J, Lee NY, Amsalem-Zafran AR, Ehrlich M, Henis YI, Mythreye K. TGF-β triggers rapid fibrillogenesis via a novel TβRII-dependent fibronectin-trafficking mechanism. Mol Biol Cell 2017; 28:1195-1207. [PMID: 28298487 PMCID: PMC5415016 DOI: 10.1091/mbc.e16-08-0601] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 02/22/2017] [Accepted: 02/27/2017] [Indexed: 02/02/2023] Open
Abstract
There is increased recycling of soluble fibronectin from the cell surface for fibrillogenesis. This recycling is regulated by TGF-β in a transcription- and SMAD-independent manner via specific TβRII and integrin α5β1 interactions. The recycling of fibronectin is Rab11 dependent and is required for TGF-β–induced cell migration. Fibronectin (FN) is a critical regulator of extracellular matrix (ECM) remodeling through its availability and stepwise polymerization for fibrillogenesis. Availability of FN is regulated by its synthesis and turnover, and fibrillogenesis is a multistep, integrin-dependent process essential for cell migration, proliferation, and tissue function. Transforming growth factor β (TGF-β) is an established regulator of ECM remodeling via transcriptional control of ECM proteins. Here we show that TGF-β, through increased FN trafficking in a transcription- and SMAD-independent manner, is a direct and rapid inducer of the fibrillogenesis required for TGF-β–induced cell migration. Whereas TGF-β signaling is dispensable for rapid fibrillogenesis, stable interactions between the cytoplasmic domain of the type II TGF-β receptor (TβRII) and the FN receptor (α5β1 integrin) are required. We find that, in response to TGF-β, cell surface–internalized FN is not degraded by the lysosome but instead undergoes recycling and incorporation into fibrils, a process dependent on TβRII. These findings are the first to show direct use of trafficked and recycled FN for fibrillogenesis, with a striking role for TGF-β in this process. Given the significant physiological consequences associated with FN availability and polymerization, our findings provide new insights into the regulation of fibrillogenesis for cellular homeostasis.
Collapse
Affiliation(s)
- Archana Varadaraj
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Laura M Jenkins
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Priyanka Singh
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - Anindya Chanda
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC 29201
| | - John Snider
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208
| | - N Y Lee
- Division of Pharmacology, College of Pharmacy, Ohio State University, Columbus, OH 43210
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC 29208 .,Department of Drug Discovery and Biomedical Sciences, University of South Carolina, Columbia, SC 29208
| |
Collapse
|
17
|
Bergeron JJM, Di Guglielmo GM, Dahan S, Dominguez M, Posner BI. Spatial and Temporal Regulation of Receptor Tyrosine Kinase Activation and Intracellular Signal Transduction. Annu Rev Biochem 2016; 85:573-97. [PMID: 27023845 DOI: 10.1146/annurev-biochem-060815-014659] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Epidermal growth factor (EGF) and insulin receptor tyrosine kinases (RTKs) exemplify how receptor location is coupled to signal transduction. Extracellular binding of ligands to these RTKs triggers their concentration into vesicles that bud off from the cell surface to generate intracellular signaling endosomes. On the exposed cytosolic surface of these endosomes, RTK autophosphorylation selects the downstream signaling proteins and lipids to effect growth factor and polypeptide hormone action. This selection is followed by the recruitment of protein tyrosine phosphatases that inactivate the RTKs and deliver them by membrane fusion and fission to late endosomes. Coincidentally, proteinases inside the endosome cleave the EGF and insulin ligands. Subsequent inward budding of the endosomal membrane generates multivesicular endosomes. Fusion with lysosomes then results in RTK degradation and downregulation. Through the spatial positioning of RTKs in target cells for EGF and insulin action, the temporal extent of signaling, attenuation, and downregulation is regulated.
Collapse
Affiliation(s)
- John J M Bergeron
- Department of Medicine, McGill University Hospital Research Institute, Montreal, Quebec, Canada H4A 3J1; , , ,
| | - Gianni M Di Guglielmo
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada N6A 5C1;
| | - Sophie Dahan
- Department of Medicine, McGill University Hospital Research Institute, Montreal, Quebec, Canada H4A 3J1; , , ,
| | - Michel Dominguez
- Department of Medicine, McGill University Hospital Research Institute, Montreal, Quebec, Canada H4A 3J1; , , ,
| | - Barry I Posner
- Department of Medicine, McGill University Hospital Research Institute, Montreal, Quebec, Canada H4A 3J1; , , ,
| |
Collapse
|
18
|
Amsalem AR, Marom B, Shapira KE, Hirschhorn T, Preisler L, Paarmann P, Knaus P, Henis YI, Ehrlich M. Differential regulation of translation and endocytosis of alternatively spliced forms of the type II bone morphogenetic protein (BMP) receptor. Mol Biol Cell 2016; 27:716-30. [PMID: 26739752 PMCID: PMC4750929 DOI: 10.1091/mbc.e15-08-0547] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 12/24/2015] [Indexed: 12/22/2022] Open
Abstract
The cytoplasmic extension of the long-form isoform of BMPRII, unique among TGF-β superfamily receptors, is found to regulate the translation of BMPRII and its clathrin-mediated endocytosis. Both processes reduce its cell surface levels. The higher expression of BMPRII-SF at the plasma membrane results in enhanced activation of Smad signaling. The expression and function of transforming growth factor-β superfamily receptors are regulated by multiple molecular mechanisms. The type II BMP receptor (BMPRII) is expressed as two alternatively spliced forms, a long and a short form (BMPRII-LF and –SF, respectively), which differ by an ∼500 amino acid C-terminal extension, unique among TGF-β superfamily receptors. Whereas this extension was proposed to modulate BMPRII signaling output, its contribution to the regulation of receptor expression was not addressed. To map regulatory determinants of BMPRII expression, we compared synthesis, degradation, distribution, and endocytic trafficking of BMPRII isoforms and mutants. We identified translational regulation of BMPRII expression and the contribution of a 3’ terminal coding sequence to this process. BMPRII-LF and -SF differed also in their steady-state levels, kinetics of degradation, intracellular distribution, and internalization rates. A single dileucine signal in the C-terminal extension of BMPRII-LF accounted for its faster clathrin-mediated endocytosis relative to BMPRII-SF, accompanied by mildly faster degradation. Higher expression of BMPRII-SF at the plasma membrane resulted in enhanced activation of Smad signaling, stressing the potential importance of the multilayered regulation of BMPRII expression at the plasma membrane.
Collapse
Affiliation(s)
- Ayelet R Amsalem
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Barak Marom
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Keren E Shapira
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Livia Preisler
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Pia Paarmann
- Institute for Chemistry and Biochemistry, Freie Univesitaet Berlin, 1495 Berlin, Germany
| | - Petra Knaus
- Institute for Chemistry and Biochemistry, Freie Univesitaet Berlin, 1495 Berlin, Germany
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
19
|
Abstract
In cells responding to extracellular polypeptide ligands, regulatory mechanisms at the level of cell surface receptors are increasingly seen to define the nature of the ligand-induced signaling responses. Processes that govern the levels of receptors at the plasma membrane, including posttranslational modifications, are crucial to ensure receptor function and specify the downstream signals. Indeed, extracellular posttranslational modifications of the receptors help define stability and ligand binding, while intracellular modifications mediate interactions with signaling mediators and accessory proteins that help define the nature of the signaling response. The use of various molecular biology and biochemistry techniques, based on chemical crosslinking, e.g., biotin or radioactive labeling, immunofluorescence to label membrane receptors and flow cytometry, allows for quantification of changes of cell surface receptor presentation. Here, we discuss recent progress in our understanding of the regulation of TGF-β receptors, i.e., the type I (TβRI) and type II (TβRII) TGF-β receptors, and describe basic methods to identify and quantify TGF-β cell surface receptors.
Collapse
Affiliation(s)
- Erine H Budi
- Department of Cell and Tissue Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Programs in Cell Biology, and Developmental and Stem Cell Biology, University of California, San Francisco, CA, USA
| | - Jian Xu
- Center for Craniofacial Molecular Biology, Ostrow School of Dentistry of USC, University of Southern California, Los Angeles, CA, USA
| | - Rik Derynck
- Department of Cell and Tissue Biology, Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, Programs in Cell Biology, and Developmental and Stem Cell Biology, University of California, San Francisco, CA, USA.
| |
Collapse
|
20
|
Ehrlich M. Endocytosis and trafficking of BMP receptors: Regulatory mechanisms for fine-tuning the signaling response in different cellular contexts. Cytokine Growth Factor Rev 2015; 27:35-42. [PMID: 26776724 DOI: 10.1016/j.cytogfr.2015.12.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Signaling by bone morphogenetic protein (BMP) receptors is regulated at multiple levels in order to ensure proper interpretation of BMP stimuli in different cellular settings. As with other signaling receptors, regulation of the amount of exposed and signaling-competent BMP receptors at the plasma-membrane is predicted to be a key mechanism in governing their signaling output. Currently, the endocytosis of BMP receptors is thought to resemble that of the structurally related transforming growth factor-β (TGF-β) receptors, as BMP receptors are constitutively internalized (independently of ligand binding), with moderate kinetics, and mostly via clathrin-mediated endocytosis. Also similar to TGF-β receptors, BMP receptors are able to signal from the plasma membrane, while internalization to endosomes may have a signal modulating effect. When at the plasma membrane, BMP receptors localize to different membrane domains including cholesterol rich domains and caveolae, suggesting a complex interplay between membrane distribution and internalization. An additional layer of complexity stems from the putative regulatory influence on the signaling and trafficking of BMP receptors exerted by ligand traps and/or co-receptors. Furthermore, the trafficking and signaling of BMP receptors are subject to alterations in cellular context. For example, genetic diseases involving changes in the expression of auxiliary factors of endocytic pathways hamper retrograde BMP signals in neurons, and perturb the regulation of synapse formation. This review summarizes current understanding of the trafficking of BMP receptors and discusses the role of trafficking in regulation of BMP signals.
Collapse
Affiliation(s)
- Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
21
|
Akhurst RJ, Padgett RW. Matters of context guide future research in TGFβ superfamily signaling. Sci Signal 2015; 8:re10. [PMID: 26486175 DOI: 10.1126/scisignal.aad0416] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The highly conserved wiring of the SMAD-dependent transforming growth factor β (TGFβ) superfamily signaling pathway has been mapped over the last 20 years after molecular discovery of its component parts. Numerous alternative TGFβ-activated signaling pathways that elicit SMAD-independent biological responses also exist. However, the molecular mechanisms responsible for the renowned context dependency of TGFβ signaling output remains an active and often confounding area of research, providing a prototype relevant to regulation of other signaling pathways. Highlighting discoveries presented at the 9th FASEB meeting, The TGFβ Superfamily: Signaling in Development and Disease (July 12-17th 2015 in Snowmass, Colorado), this Review outlines research into the rich contextual nature of TGFβ signaling output and offers clues for therapeutic advances.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Helen Diller Family Comprehensive Cancer Center and Department of Anatomy, University of California at San Francisco, San Francisco, CA 94158-9001, USA.
| | - Richard W Padgett
- Waksman Institute, Department of Molecular Biology and Biochemistry, and Cancer Institute of New Jersey, Rutgers University, Piscataway, NJ 08854-8020, USA
| |
Collapse
|
22
|
Huang SS, Chen CL, Huang FW, Johnson FE, Huang JS. Ethanol Enhances TGF-β Activity by Recruiting TGF-β Receptors From Intracellular Vesicles/Lipid Rafts/Caveolae to Non-Lipid Raft Microdomains. J Cell Biochem 2015; 117:860-71. [PMID: 26419316 DOI: 10.1002/jcb.25389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Regular consumption of moderate amounts of ethanol has important health benefits on atherosclerotic cardiovascular disease (ASCVD). Overindulgence can cause many diseases, particularly alcoholic liver disease (ALD). The mechanisms by which ethanol causes both beneficial and harmful effects on human health are poorly understood. Here we demonstrate that ethanol enhances TGF-β-stimulated luciferase activity with a maximum of 0.5-1% (v/v) in Mv1Lu cells stably expressing a luciferase reporter gene containing Smad2-dependent elements. In Mv1Lu cells, 0.5% ethanol increases the level of P-Smad2, a canonical TGF-β signaling sensor, by ∼ 2-3-fold. Ethanol (0.5%) increases cell-surface expression of the type II TGF-β receptor (TβR-II) by ∼ 2-3-fold from its intracellular pool, as determined by I(125) -TGF-β-cross-linking/Western blot analysis. Sucrose density gradient ultracentrifugation and indirect immunofluorescence staining analyses reveal that ethanol (0.5% and 1%) also displaces cell-surface TβR-I and TβR-II from lipid rafts/caveolae and facilitates translocation of these receptors to non-lipid raft microdomains where canonical signaling occurs. These results suggest that ethanol enhances canonical TGF-β signaling by increasing non-lipid raft microdomain localization of the TGF-β receptors. Since TGF-β plays a protective role in ASCVD but can also cause ALD, the TGF-β enhancer activity of ethanol at low and high doses appears to be responsible for both beneficial and harmful effects. Ethanol also disrupts the location of lipid raft/caveolae of other membrane proteins (e.g., neurotransmitter, growth factor/cytokine, and G protein-coupled receptors) which utilize lipid rafts/caveolae as signaling platforms. Displacement of these membrane proteins induced by ethanol may result in a variety of pathologies in nerve, heart and other tissues.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02115.,Harvard Medical School, Boston, Massachusetts, 02115
| | - Frank E Johnson
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, Missouri, 63104
| |
Collapse
|
23
|
Crupi MJF, Yoganathan P, Bone LN, Lian E, Fetz A, Antonescu CN, Mulligan LM. Distinct Temporal Regulation of RET Isoform Internalization: Roles of Clathrin and AP2. Traffic 2015; 16:1155-73. [DOI: 10.1111/tra.12315] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 08/19/2015] [Accepted: 08/19/2015] [Indexed: 01/08/2023]
Affiliation(s)
- Mathieu J. F. Crupi
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Piriya Yoganathan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Leslie N. Bone
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B 2K3 Canada
| | - Eric Lian
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Andrew Fetz
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| | - Costin N. Antonescu
- Department of Chemistry and Biology; Ryerson University; Toronto Ontario M5B 2K3 Canada
| | - Lois M. Mulligan
- Division of Cancer Biology and Genetics, Cancer Research Institute and Department of Pathology & Molecular Medicine; Queen's University; Kingston Ontario K7L 3N6 Canada
| |
Collapse
|
24
|
Tazat K, Hector-Greene M, Blobe GC, Henis YI. TβRIII independently binds type I and type II TGF-β receptors to inhibit TGF-β signaling. Mol Biol Cell 2015; 26:3535-45. [PMID: 26269580 PMCID: PMC4591696 DOI: 10.1091/mbc.e15-04-0203] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Accepted: 08/05/2015] [Indexed: 02/01/2023] Open
Abstract
Study of the TβRIII interaction with the signaling TGF-β receptors shows that TβRIII homo-oligomerization is indirect, depending largely on interactions with GIPC scaffolds. TβRI and II bind independently to TβRIII, competing with TβRI-TβRII complex formation and inhibiting Smad2/3 signaling by a mechanism independent of TβRIII ectodomain shedding. Transforming growth factor-β (TGF-β) receptor oligomerization has important roles in signaling. Complex formation among type I and type II (TβRI and TβRII) TGF-β receptors is well characterized and is essential for signal transduction. However, studies on their interactions with the type III TGF-β coreceptor (TβRIII) in live cells and their effects on TGF-β signaling are lacking. Here we investigated the homomeric and heteromeric interactions of TβRIII with TβRI and TβRII in live cells by combining IgG-mediated patching/immobilization of a given TGF-β receptor with fluorescence recovery after photobleaching studies on the lateral diffusion of a coexpressed receptor. Our studies demonstrate that TβRIII homo-oligomerization is indirect and depends on its cytoplasmic domain interactions with scaffold proteins (mainly GIPC). We show that TβRII and TβRI bind independently to TβRIII, whereas TβRIII augments TβRI/TβRII association, suggesting that TβRI and TβRII bind to TβRIII simultaneously but not as a complex. TβRIII expression inhibited TGF-β–mediated Smad2/3 signaling in MDA-MB-231 cell lines, an effect that depended on the TβRIII cytoplasmic domain and did not require TβRIII ectodomain shedding. We propose that independent binding of TβRI and TβRII to TβRIII competes with TβRI/TβRII signaling complex formation, thus inhibiting TGF-β–mediated Smad signaling.
Collapse
Affiliation(s)
- Keren Tazat
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC 27708 Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27708
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel )
| |
Collapse
|
25
|
Pomeraniec L, Hector-Greene M, Ehrlich M, Blobe GC, Henis YI. Regulation of TGF-β receptor hetero-oligomerization and signaling by endoglin. Mol Biol Cell 2015; 26:3117-27. [PMID: 26157163 PMCID: PMC4551323 DOI: 10.1091/mbc.e15-02-0069] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 06/30/2015] [Indexed: 11/23/2022] Open
Abstract
Endoglin is a modulator of TGF-β signaling in endothelial cells. We show that it forms stable homodimers serving as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind endoglin differentially, with TβRII recruiting ALK5. Signaling data indicate a role for this receptor complex in balancing TGF-β signaling between Smad1/5/8 and Smad2/3. Complex formation among transforming growth factor-β (TGF-β) receptors and its modulation by coreceptors represent an important level of regulation for TGF-β signaling. Oligomerization of ALK5 and the type II TGF-β receptor (TβRII) has been thoroughly investigated, both in vitro and in intact cells. However, such studies, especially in live cells, are missing for the endothelial cell coreceptor endoglin and for the ALK1 type I receptor, which enables endothelial cells to respond to TGF-β by activation of both Smad2/3 and Smad1/5/8. Here we combined immunoglobulin G–mediated immobilization of one cell-surface receptor with lateral mobility studies of a coexpressed receptor by fluorescence recovery after photobleaching (FRAP) to demonstrate that endoglin forms stable homodimers that function as a scaffold for binding TβRII, ALK5, and ALK1. ALK1 and ALK5 bind to endoglin with differential dependence on TβRII, which plays a major role in recruiting ALK5 to the complex. Signaling data indicate a role for the quaternary receptor complex in regulating the balance between TGF-β signaling to Smad1/5/8 and to Smad2/3.
Collapse
Affiliation(s)
- Leslie Pomeraniec
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerard C Blobe
- Department of Medicine, Duke University Medical Center, Durham, NC 27708
| | - Yoav I Henis
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
26
|
Tu K, Li J, Verma VK, Liu C, Billadeau DD, Lamprecht G, Xiang X, Guo L, Dhanasekaran R, Roberts LR, Shah VH, Kang N. Vasodilator-stimulated phosphoprotein promotes activation of hepatic stellate cells by regulating Rab11-dependent plasma membrane targeting of transforming growth factor beta receptors. Hepatology 2015; 61:361-74. [PMID: 24917558 PMCID: PMC4262723 DOI: 10.1002/hep.27251] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/23/2014] [Indexed: 12/22/2022]
Abstract
UNLABELLED Liver microenvironment is a critical determinant for development and progression of liver metastasis. Under transforming growth factor beta (TGF-β) stimulation, hepatic stellate cells (HSCs), which are liver-specific pericytes, transdifferentiate into tumor-associated myofibroblasts that promote tumor implantation (TI) and growth in the liver. However, the regulation of this HSC activation process remains poorly understood. In this study, we tested whether vasodilator-stimulated phosphoprotein (VASP) of HSCs regulated the TGF-β-mediated HSC activation process and tumor growth. In both an experimental liver metastasis mouse model and cancer patients, colorectal cancer cells reaching liver sinusoids induced up-regulation of VASP and alpha-smooth muscle actin (α-SMA) in adjacent HSCs. VASP knockdown in HSCs inhibited TGF-β-mediated myofibroblastic activation of HSCs, TI, and growth in mice. Mechanistically, VASP formed protein complexes with TGF-β receptor II (TβRII) and Rab11, a Ras-like small GTPase and key regulator of recycling endosomes. VASP knockdown impaired Rab11 activity and Rab11-dependent targeting of TβRII to the plasma membrane, thereby desensitizing HSCs to TGF-β1 stimulation. CONCLUSIONS Our study demonstrates a requirement of VASP for TGF-β-mediated HSC activation in the tumor microenvironment by regulating Rab11-dependent recycling of TβRII to the plasma membrane. VASP and its effector, Rab11, in the tumor microenvironment thus present therapeutic targets for reducing TI and metastatic growth in the liver.
Collapse
Affiliation(s)
- Kangsheng Tu
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Hepatobillary Surgery, the 1 Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710061, China
| | - Jiachu Li
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
- Department of Oncology, the 1 Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Vikas K Verma
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Chunsheng Liu
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
| | | | - Georg Lamprecht
- Division of Gastroenterology and Endocrinology, University of Rostock, Rostock, 18057, Germany
| | - Xiaoyu Xiang
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Luyang Guo
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | | | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Vijay H. Shah
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ningling Kang
- GI Research Unit and Cancer Cell Biology Program, Mayo Clinic, Rochester, MN, 55905, USA
- Tumor Microenvironment and Metastasis Section, the Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| |
Collapse
|
27
|
Dynamic scaling of morphogen gradients on growing domains. Nat Commun 2014; 5:5077. [PMID: 25295831 DOI: 10.1038/ncomms6077] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 08/26/2014] [Indexed: 11/08/2022] Open
Abstract
Developmental mechanisms are highly conserved, yet act in embryos of very different sizes. How scaling is achieved has remained elusive. Here we identify a generally applicable mechanism for dynamic scaling on growing domains and show that it quantitatively agrees with data from the Drosophila wing imaginal disc. We show that for the measured parameter ranges, the Dpp gradient does not reach steady state during Drosophila wing development. We further show that both, pre-steady-state dynamics and advection of cell-bound ligand in a growing tissue can, in principle, enable scaling, even for non-uniform tissue growth. For the parameter values that have been established for the Dpp morphogen in the Drosophila wing imaginal disc, we show that scaling is mainly a result of the pre-steady-state dynamics. Pre-steady-state dynamics are pervasive in morphogen-controlled systems, thus making this a probable general mechanism for dynamic scaling of morphogen gradients in growing developmental systems.
Collapse
|
28
|
Shapira KE, Hirschhorn T, Barzilay L, Smorodinsky NI, Henis YI, Ehrlich M. Dab2 inhibits the cholesterol-dependent activation of JNK by TGF-β. Mol Biol Cell 2014; 25:1620-8. [PMID: 24648493 PMCID: PMC4019493 DOI: 10.1091/mbc.e13-09-0537] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
TGF-β signals through Smad-dependent and non-Smad pathways, depending on cell context. In ovarian cancer cells, the clathrin adaptor Dab2 enhances internalization of the type I TGF-β receptor, restricts its lateral mobility, and inhibits TGF-β–mediated, cholesterol-dependent JNK activation. Transforming growth factor-β (TGF-β) ligands activate Smad-mediated and noncanonical signaling pathways in a cell context–dependent manner. Localization of signaling receptors to distinct membrane domains is a potential source of signaling output diversity. The tumor suppressor/endocytic adaptor protein disabled-2 (Dab2) was proposed as a modulator of TGF-β signaling. However, the molecular mechanism(s) involved in the regulation of TGF-β signaling by Dab2 were not known. Here we investigate these issues by combining biophysical studies of the lateral mobility and endocytosis of the type I TGF-β receptor (TβRI) with TGF-β phosphoprotein signaling assays. Our findings demonstrate that Dab2 interacts with TβRI to restrict its lateral diffusion at the plasma membrane and enhance its clathrin-mediated endocytosis. Small interfering RNA–mediated knockdown of Dab2 or Dab2 overexpression shows that Dab2 negatively regulates TGF-β–induced c-Jun N-terminal kinase (JNK) activation, whereas activation of the Smad pathway is unaffected. Moreover, activation of JNK by TGF-β in the absence of Dab2 is disrupted by cholesterol depletion. These data support a model in which Dab2 regulates the domain localization of TβRI in the membrane, balancing TGF-β signaling via the Smad and JNK pathways.
Collapse
Affiliation(s)
- Keren E Shapira
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Tal Hirschhorn
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Lior Barzilay
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nechama I Smorodinsky
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yoav I Henis
- Department of Neurobiology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marcelo Ehrlich
- Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
29
|
Vizán P, Miller DSJ, Gori I, Das D, Schmierer B, Hill CS. Controlling long-term signaling: receptor dynamics determine attenuation and refractory behavior of the TGF-β pathway. Sci Signal 2013; 6:ra106. [PMID: 24327760 DOI: 10.1126/scisignal.2004416] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Understanding the complex dynamics of growth factor signaling requires both mechanistic and kinetic information. Although signaling dynamics have been studied for pathways downstream of receptor tyrosine kinases and G protein (heterotrimeric guanine nucleotide-binding protein)-coupled receptors, they have not been investigated for the transforming growth factor-β (TGF-β) superfamily pathways. Using an integrative experimental and mathematical modeling approach, we dissected the dynamic behavior of the TGF-β to Smad pathway, which is mediated by type I and type II receptor serine/threonine kinases, in response to acute, chronic, and repeated ligand stimulations. TGF-β exposure produced a transient response that attenuated over time, resulting in desensitized cells that were refractory to further acute stimulation. This loss of signaling competence depended on ligand binding, but not on receptor activity, and was restored only after the ligand had been depleted. Furthermore, TGF-β binding triggered the rapid depletion of signaling-competent receptors from the cell surface, with the type I and type II receptors exhibiting different degradation and trafficking kinetics. A computational model of TGF-β signal transduction from the membrane to the nucleus that incorporates our experimental findings predicts that autocrine signaling, such as that associated with tumorigenesis, severely compromises the TGF-β response, which we confirmed experimentally. Thus, we have shown that the long-term signaling behavior of the TGF-β pathway is determined by receptor dynamics, does not require TGF-β-induced gene expression, and influences context-dependent responses in vivo.
Collapse
Affiliation(s)
- Pedro Vizán
- 1Developmental Signalling Laboratory, Cancer Research UK London Research Institute, 44 Lincoln's Inn Fields, London WC2A 3LY, UK
| | | | | | | | | | | |
Collapse
|
30
|
Kaiser O, Paasche G, Stöver T, Ernst S, Lenarz T, Kral A, Warnecke A. TGF-beta superfamily member activin A acts with BDNF and erythropoietin to improve survival of spiral ganglion neurons in vitro. Neuropharmacology 2013; 75:416-25. [PMID: 23973291 DOI: 10.1016/j.neuropharm.2013.08.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 07/03/2013] [Accepted: 08/08/2013] [Indexed: 01/15/2023]
Abstract
Activins are regulators of embryogenesis, osteogenesis, hormones and neuronal survival. Even though activin receptor type II has been detected in spiral ganglion neurons (SGN), little is known about the role of activins in the inner ear. An activin-mediated neuroprotection is of considerable clinical interest since SGN are targets of electrical stimulation with cochlear implants in hearing impaired patients. Thus, the presence of activin type-I and type-II receptors was demonstrated immunocytochemically and the individual and combined effects of activin A, erythropoietin (EPO) and brain-derived neurotrophic factor (BDNF) on SGN were examined in vitro. SGN isolated from neonatal rats (P 3-5) were cultured in serum-free medium supplemented with activin A, BDNF and EPO. Compared to the negative control, survival rates of SGN were significantly improved when cultivated individually with activin A (p<0.001) and in combination with BDNF (p<0.001). Neither neurite outgrowth nor neuronal survival was influenced by the addition of EPO to activin A-treated neurons. However, when all three factors were added, a significantly (p<0.001) improved neuronal survival was observed (61.2±3.6%) compared to activin A (25.4±2.1%), BDNF (22.8±3.3%) and BDNF+EPO (19.2±1.5%). Under the influence of the EPO-inhibitors, this increase in neuronal survival was blocked. Acting with BDNF and EPO to promote neuronal survival in vitro, activin A presents an interesting factor for pharmacological intervention in the inner ear. The present study demonstrates a synergetic effect of a combined therapy with several trophic factors.
Collapse
Affiliation(s)
- Odett Kaiser
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| | - Gerrit Paasche
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Timo Stöver
- Department of Otolaryngology, University Hospital, Johann Wolfgang Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Stefanie Ernst
- Institute for Biometry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Thomas Lenarz
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Andrej Kral
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Athanasia Warnecke
- Department of Otolaryngology, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany.
| |
Collapse
|
31
|
Di Rubbo S, Irani NG, Kim SY, Xu ZY, Gadeyne A, Dejonghe W, Vanhoutte I, Persiau G, Eeckhout D, Simon S, Song K, Kleine-Vehn J, Friml J, De Jaeger G, Van Damme D, Hwang I, Russinova E. The clathrin adaptor complex AP-2 mediates endocytosis of brassinosteroid insensitive1 in Arabidopsis. THE PLANT CELL 2013; 25:2986-97. [PMID: 23975899 PMCID: PMC3784593 DOI: 10.1105/tpc.113.114058] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/22/2013] [Accepted: 08/06/2013] [Indexed: 05/18/2023]
Abstract
Clathrin-mediated endocytosis (CME) regulates many aspects of plant development, including hormone signaling and responses to environmental stresses. Despite the importance of this process, the machinery that regulates CME in plants is largely unknown. In mammals, the heterotetrameric adaptor protein complex-2 (AP-2) is required for the formation of clathrin-coated vesicles at the plasma membrane (PM). Although the existence of AP-2 has been predicted in Arabidopsis thaliana, the biochemistry and functionality of the complex is still uncharacterized. Here, we identified all the subunits of the Arabidopsis AP-2 by tandem affinity purification and found that one of the large AP-2 subunits, AP2A1, localized at the PM and interacted with clathrin. Furthermore, endocytosis of the leucine-rich repeat receptor kinase, brassinosteroid insensitive1 (BRI1), was shown to depend on AP-2. Knockdown of the two Arabidopsis AP2A genes or overexpression of a dominant-negative version of the medium AP-2 subunit, AP2M, impaired BRI1 endocytosis and enhanced the brassinosteroid signaling. Our data reveal that the CME machinery in Arabidopsis is evolutionarily conserved and that AP-2 functions in receptor-mediated endocytosis.
Collapse
Affiliation(s)
- Simone Di Rubbo
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Niloufer G. Irani
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Soo Youn Kim
- Division of Molecules and Life Sciences and Center for Plant Intracellular Trafficking, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Zheng-Yi Xu
- Division of Molecules and Life Sciences and Center for Plant Intracellular Trafficking, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Astrid Gadeyne
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Wim Dejonghe
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Isabelle Vanhoutte
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Geert Persiau
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Dominique Eeckhout
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Sibu Simon
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Kyungyoung Song
- Division of Molecules and Life Sciences and Center for Plant Intracellular Trafficking, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Jürgen Kleine-Vehn
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Jiří Friml
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Geert De Jaeger
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Daniël Van Damme
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
| | - Inhwan Hwang
- Division of Molecules and Life Sciences and Center for Plant Intracellular Trafficking, Pohang University of Science and Technology, Pohang 790-784, Korea
| | - Eugenia Russinova
- Department of Plant Systems Biology, VIB, 9052 Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052 Ghent, Belgium
- Address correspondence to
| |
Collapse
|
32
|
Rejon CA, Ho CC, Wang Y, Zhou X, Bernard DJ, Hébert TE. Cycloheximide inhibits follicle-stimulating hormone β subunit transcription by blocking de novo synthesis of the labile activin type II receptor in gonadotrope cells. Cell Signal 2013; 25:1403-12. [PMID: 23499904 DOI: 10.1016/j.cellsig.2013.03.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Accepted: 03/04/2013] [Indexed: 01/08/2023]
Abstract
The pituitary gonadotropins, follicle-stimulating hormone (FSH) and luteinizing hormone (LH), play essential roles in the regulation of vertebrate reproduction. Activins and inhibins have opposing actions on FSH (but not LH) synthesis, either inducing or inhibiting transcription of the FSHβ subunit gene (Fshb). The translational inhibitor cycloheximide (CHX) produces inhibin-like effects in cultured pituitary cells, selectively suppressing FSH production. Using the murine gonadotrope-like cell line, LβT2, as a model, we tested the hypothesis that a component of the activin pathway is highly labile in gonadotrope cells and that its rapid loss following CHX treatment impairs activin-stimulated Fshb transcription. Treatment of cells with CHX for 6h, but not 1h, blocked activin A-stimulated Fshb transcription. Pre-treatment of LβT2 cells with CHX for as few as 2-3h inhibited activin A-stimulated SMAD2/3 phosphorylation without altering total SMAD2/3 protein levels. These data indicated that CHX affects activin signalling upstream of SMAD proteins, most likely at the receptor level. Indeed, CHX rapidly reduced activin A binding to LβT2 cells. We went on to show that activin A signals via the type II receptor ACVR2, rather than ACVR2B, to regulate Fshb transcription and that the receptor has a half life of ~2h in LβT2 cells. The mechanism of ACVR2 turnover remains undefined, but appears to be ligand-, proteasome-, and lysosome-independent. Collectively, these data indicate that CHX produces inhibin-like effects in gonadotropes by preventing de novo synthesis of the highly labile ACVR2, thereby blocking activin signaling to the Fshb promoter.
Collapse
Affiliation(s)
- Carlis A Rejon
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada
| | | | | | | | | | | |
Collapse
|
33
|
Liu C, Billadeau DD, Abdelhakim H, Leof E, Kaibuchi K, Bernabeu C, Bloom GS, Yang L, Boardman L, Shah VH, Kang N. IQGAP1 suppresses TβRII-mediated myofibroblastic activation and metastatic growth in liver. J Clin Invest 2013; 123:1138-56. [PMID: 23454766 PMCID: PMC3582119 DOI: 10.1172/jci63836] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Accepted: 12/06/2012] [Indexed: 01/11/2023] Open
Abstract
In the tumor microenvironment, TGF-β induces transdifferentiation of quiescent pericytes and related stromal cells into myofibroblasts that promote tumor growth and metastasis. The mechanisms governing myofibroblastic activation remain poorly understood, and its role in the tumor microenvironment has not been explored. Here, we demonstrate that IQ motif containing GTPase activating protein 1 (IQGAP1) binds to TGF-β receptor II (TβRII) and suppresses TβRII-mediated signaling in pericytes to prevent myofibroblastic differentiation in the tumor microenvironment. We found that TGF-β1 recruited IQGAP1 to TβRII in hepatic stellate cells (HSCs), the resident liver pericytes. Iqgap1 knockdown inhibited the targeting of the E3 ubiquitin ligase SMAD ubiquitination regulatory factor 1 (SMURF1) to the plasma membrane and TβRII ubiquitination and degradation. Thus, Iqgap1 knockdown stabilized TβRII and potentiated TGF-β1 transdifferentiation of pericytes into myofibroblasts in vitro. Iqgap1 deficiency in HSCs promoted myofibroblast activation, tumor implantation, and metastatic growth in mice via upregulation of paracrine signaling molecules. Additionally, we found that IQGAP1 expression was downregulated in myofibroblasts associated with human colorectal liver metastases. Taken together, our studies demonstrate that IQGAP1 in the tumor microenvironment suppresses TβRII and TGF-β dependent myofibroblastic differentiation to constrain tumor growth.
Collapse
Affiliation(s)
- Chunsheng Liu
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Daniel D. Billadeau
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Haitham Abdelhakim
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Edward Leof
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Kozo Kaibuchi
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Carmelo Bernabeu
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - George S. Bloom
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Liu Yang
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Lisa Boardman
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Vijay H. Shah
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| | - Ningling Kang
- GI Research Unit and Cancer Cell Biology Program,
Department of Immunology, and
Thoracic Diseases Research Unit, Mayo Clinic, Rochester, Minnesota, USA.
Department of Cell Pharmacology, Nagoya University Graduate School of Medicine, Nagoya, Japan.
Centro de Investigaciones Biologicas, Consejo Superior de Investigaciones Cientificas (CSIC), and Centro de Investigacion Biomedica en Red de Enfermedades Raras (CIBERER), Madrid, Spain.
Department of Biology and Cell Biology, University of Virginia, Charlottesville, Virginia, USA
| |
Collapse
|
34
|
Shapira KE, Gross A, Ehrlich M, Henis YI. Coated pit-mediated endocytosis of the type I transforming growth factor-β (TGF-β) receptor depends on a di-leucine family signal and is not required for signaling. J Biol Chem 2012; 287:26876-89. [PMID: 22707720 DOI: 10.1074/jbc.m112.362848] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The roles of transforming growth factor-β (TGF-β) receptor endocytosis in signaling have been investigated in numerous studies, mainly through the use of endocytosis inhibitory treatments, yielding conflicting results. Two potential sources for these discrepancies were the pleiotropic effects of a general blockade of specific internalization pathways and the scarce information on the regulation of the endocytosis of the signal-transducing type I TGF-β receptor (TβRI). Here, we employed extracellularly tagged myc-TβRI (wild type, truncation mutants, and a series of endocytosis-defective and endocytosis-enhanced mutants) to directly investigate the relationship between TβRI endocytosis and signaling. Our findings indicate that TβRI is targeted for constitutive clathrin-mediated endocytosis via a di-leucine (Leu(180)-Ile(181)) signal and an acidic cluster motif. Using Smad-dependent transcriptional activation assays and following Smad2/3 nuclear translocation in response to TGF-β stimulation, we show that TβRI endocytosis is dispensable for TGF-β signaling and may play a role in signal termination. Alanine replacement of Leu(180)-Ile(181) led to partial constitutive activation of TβRI, resulting in part from its retention at the plasma membrane and in part from potential alterations of TβRI regulatory interactions in the vicinity of the mutated residues.
Collapse
Affiliation(s)
- Keren E Shapira
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | |
Collapse
|
35
|
Direct sensing of endothelial oxidants by vascular endothelial growth factor receptor-2 and c-Src. PLoS One 2011; 6:e28454. [PMID: 22145046 PMCID: PMC3228784 DOI: 10.1371/journal.pone.0028454] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Accepted: 11/08/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND ADPH oxidase-derived reactive oxygen species (ROS) play important roles in redox homeostasis and signal transduction in endothelial cells (ECs). We previously demonstrated that c-Src plays a key role in VEGF-induced, ROS-dependent selective activation of PI3K-Akt but not PLCγ-1-ERK1/2 signaling pathways. The aim of the present study was to understand how VEGFR-2-c-Src signaling axis 'senses' NADPH oxidase-derived ROS levels and couples VEGF activation of c-Src to the redox state of ECs. METHODOLOGY/PRINCIPAL FINDINGS Using biotinylated probe that detects oxidation of cysteine thiol (cys-OH) in intracellular proteins, we demonstrate that VEGF induced oxidative modification in c-Src and VEGFR-2, and that reduction in ROS levels using siRNA against p47(phox) subunit of Rac1-dependent NADPH oxidase inhibited this phenomenon. Co-immunoprecipitation studies using human coronary artery ECs (HCAEC) showed that VEGF-induced ROS-dependent interaction between VEGFR-2 and c-Src correlated with their thiol oxidation status. Immunofluorescence studies using antibodies against internalized VEGFR-2 and c-Src demonstrated that VEGF-induced subcellular co-localization of these tyrosine kinases were also dependent on NADPH oxidsase-derived ROS. CONCLUSION/SIGNIFICANCE These results demonstrate that VEGF induces cysteine oxidation in VEGFR-2 and c-Src in an NADPH oxidase-derived ROS-dependent manner, suggesting that VEGFR-2 and c-Src can 'sense' redox levels in ECs. The data also suggest that thiol oxidation status of VEGFR-2 and c-Src correlates with their ability to physically interact with each other and c-Src activation. Taken together, these findings suggest that prior to activating downstream c-Src-PI3K-Akt signaling pathway, VEGFR-2-c-Src axis requires an NADPH oxidase-derived ROS threshold in ECs.
Collapse
|
36
|
Marom B, Heining E, Knaus P, Henis YI. Formation of stable homomeric and transient heteromeric bone morphogenetic protein (BMP) receptor complexes regulates Smad protein signaling. J Biol Chem 2011; 286:19287-96. [PMID: 21471205 DOI: 10.1074/jbc.m110.210377] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The type I and type II bone morphogenetic protein receptors (BMPRI and BMPRII) are present at the plasma membrane as monomers and homomeric and heteromeric complexes, which are modulated by ligand binding. The complexes of their extracellular domains with ligand were shown to form heterotetramers. However, the dynamics of the oligomeric interactions among the full-length receptors in live cell membranes were not explored, and the roles of BMP receptor homodimerization were unknown. Here, we investigated these issues by combining patching/immobilization of an epitope-tagged BMP receptor at the cell surface with measurements of the lateral diffusion of a co-expressed, differently tagged BMP receptor by fluorescence recovery after photobleaching (FRAP). These studies led to several novel conclusions. (a) All homomeric complexes (without or with BMP-2) were stable on the patch/FRAP time scale (minutes), whereas the heterocomplexes were transient, a difference that may affect signaling. (b) Patch/FRAP between HA- and myc-tagged BMPRII combined with competition by untagged BMPRIb showed that the heterocomplexes form at the expense of homodimers. (c) Stabilization of BMPRII·BMPRIb heterocomplexes (but not homomeric complexes) by IgG binding to same-tag receptors elevated phospho-Smad formation both without and with BMP-2. These findings suggest two mechanisms that may suppress the tendency of preformed BMP receptor hetero-oligomers to signal without ligand: (a) competition between homo- and heterocomplex formation, which reduces the steady-state level of the latter, and (b) the transient nature of the heterocomplexes, which limits the time during which BMPRI can be phosphorylated by BMPRII in the heterocomplex.
Collapse
Affiliation(s)
- Barak Marom
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
37
|
SNX25 regulates TGF-β signaling by enhancing the receptor degradation. Cell Signal 2011; 23:935-46. [PMID: 21266196 DOI: 10.1016/j.cellsig.2011.01.022] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2011] [Accepted: 01/19/2011] [Indexed: 11/20/2022]
Abstract
SNXs (sorting nexin), a family of proteins playing roles in cargo sorting and signaling from compartments within the endocytic network, regulate traffic of membrane proteins including TGF-β receptors. Here we report that the full length human and mouse SNX25, a SNX member with PX, PXA and RGS domains, co-localizes with TGF-β receptors, and forms internalized cytosolic punctae upon treatment with TGF-β. While overexpression of SNX25 inhibits TGF-β induced luciferase reporter activity, knocking down endogenous SNX25 by siRNA in NIH3T3 cells elevates the TGF-β receptor levels and facilitates TGF-β signaling. Immunoprecipitation experiments demonstrate that SNX25 interacts with TβRI. Western blot analyses indicate that SNX25 enhances the degradation of TGF-β receptors. SNX25 induced TGF-β receptor degradation is shown via the clathrin dependent endocytosis pathway into lysosome. We have characterized that PXA domain of SNX25 is required for the degradation of TβRI. Our findings demonstrate that SNX25 negatively regulates TGF-β signaling by enhancing the receptor degradation through lysosome pathway.
Collapse
|
38
|
Horbelt D, Guo G, Robinson PN, Knaus P. Quantitative analysis of TGFBR2 mutations in Marfan-syndrome-related disorders suggests a correlation between phenotypic severity and Smad signaling activity. J Cell Sci 2010; 123:4340-50. [PMID: 21098638 DOI: 10.1242/jcs.074773] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Mutations in the gene encoding transforming growth factor-beta receptor type II (TGFBR2) have been described in patients with Loeys-Dietz syndrome (LDS), Marfan syndrome type 2 (MFS2) and familial thoracic aortic aneurysms and dissections (TAAD). Here, we present a comprehensive and quantitative analysis of TGFBR2 expression, turnover and TGF-β-induced Smad and ERK signaling activity for nine mutations identified in patients with LDS, MFS2 and TAAD. The mutations had different effects on protein stability, internalization and signaling. A dominant-negative effect was demonstrated for mutations associated with LDS and MFS2. No mutation showed evidence of an immediate cell-autonomous paradoxical activation of TGF-β signaling. There were no cell biological differences between mutations described in patients with LDS and MFS2. By contrast, R460C, which has been found in familial TAAD but not in MFS2 or LDS, showed a less-severe dominant-negative effect and retained residual Smad phosphorylation and transcriptional activity. TAAD is characterized primarily by thoracic aortic aneurysms or dissections. By contrast, MFS2 is characterized by numerous skeletal abnormalities, and patients with LDS additionally can display craniofacial and other abnormalities. Therefore, our findings suggest that the balance between defects in Smad and ERK signaling might be an important determinant of phenotypic severity in disorders related to mutations in TGFBR2.
Collapse
Affiliation(s)
- Daniel Horbelt
- Institute for Chemistry-Biochemistry, Freie Universität Berlin, Berlin, 14195, Germany
| | | | | | | |
Collapse
|
39
|
TGF beta (transforming growth factor beta) receptor type III directs clathrin-mediated endocytosis of TGF beta receptor types I and II. Biochem J 2010; 429:137-45. [PMID: 20406198 DOI: 10.1042/bj20091598] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The TGFbeta (transforming growth factor beta) pathway is an essential cell signalling pathway that is implicated in both normal developmental processes, such as organogenesis, and pathological disorders, such as cancer and fibrosis. There are three prototypical TbetaRs (TGFbeta receptors): TbetaRI (TbetaR type I), TGbetaRII (TbetaR type II) and TGFbetaRIII (TbetaR type III, also known as betaglycan). Whereas the role of TbetaRII and TbetaRI in TGFbeta signal propagation has been established, the contribution of TbetaRIII to TGFbeta signalling is less well understood. At the cell surface, TbetaRI and TbetaRII receptors can be internalized by clathrin-mediated endocytosis and clathrin-independent membrane-raft-dependent endocytosis. Interestingly, the endocytic route of the receptors plays a direct role in TGFbeta-dependent Smad signal transduction; receptors endocytosed via clathrin-mediated endocytosis activate Smad signalling, whereas receptors endocytosed via membrane rafts are targeted for degradation. The objective of the present study was to evaluate the contribution of TbetaRIII to TbetaRII and TbetaRI membrane partitioning, receptor half-life and signalling. Using sucrose-density ultracentrifugation to isolate membrane-raft fractions, we show that TbetaRIII recruits both TbetaRII and TbetaRI to non-raft membrane fractions. Immunofluorescence microscopy analysis demonstrated that overexpression of TbetaRIII affects intracellular trafficking of TbetaRII by recruiting TbetaRII to EEA1 (early endosome antigen 1)- and Rab5-positive early endosomes. Using 125I-labelled TGFbeta1 to follow cell-surface receptor degradation we show that overexpression of TbetaRIII also extends the receptor half-life of the TbetaRII-TbetaRI complex. Interestingly, we also show, using a luciferase reporter assay, that TbetaRIII increases basal TGFbeta signalling. As numerous pathologies show aberrant activation of TGFbeta signalling, the present study illustrates that TbetaRIII may represent a novel therapeutic target.
Collapse
|
40
|
TGF-beta receptors, in a Smad-independent manner, are required for terminal skeletal muscle differentiation. Exp Cell Res 2010; 316:2487-503. [PMID: 20471380 DOI: 10.1016/j.yexcr.2010.04.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2010] [Revised: 04/28/2010] [Accepted: 04/30/2010] [Indexed: 11/22/2022]
Abstract
Skeletal muscle differentiation is strongly inhibited by transforming growth factor type beta (TGF-beta), although muscle formation as well as regeneration normally occurs in an environment rich in this growth factor. In this study, we evaluated the role of intracellular regulatory Smads proteins as well as TGF-beta-receptors (TGF-beta-Rs) during skeletal muscle differentiation. We found a decrease of TGF-beta signaling during differentiation. This phenomenon is explained by a decline in the levels of the regulatory proteins Smad-2, -3, and -4, a decrease in the phosphorylation of Smad-2 and lost of nuclear translocation of Smad-3 and -4 in response to TGF-beta. No change in the levels and inhibitory function of Smad-7 was observed. In contrast, we found that TGF-beta-R type I (TGF-beta-RI) and type II (TGF-beta-RII) increased on the cell surface during skeletal muscle differentiation. To analyze the direct role of the serine/threonine kinase activities of TGF-beta-Rs, we used the specific inhibitor SB 431542 and the dominant-negative form of TGF-beta-RII lacking the cytoplasmic domain. The TGF-beta-Rs were important for successful muscle formation, determined by the induction of myogenin, creatine kinase activity, and myosin. Silencing of Smad-2/3 expression by specific siRNA treatments accelerated myogenin, myosin expression, and myotube formation; although when SB 431542 was present inhibition in myosin induction and myotube formation was observed, suggesting that these last steps of skeletal muscle differentiation require active TGF-beta-Rs. These results suggest that both down-regulation of Smad regulatory proteins and cell signaling through the TGF-beta receptors independent of Smad proteins are essential for skeletal muscle differentiation.
Collapse
|
41
|
Li W, Puertollano R, Bonifacino JS, Overbeek PA, Everett ET. Disruption of the murine Ap2β1 gene causes nonsyndromic cleft palate. Cleft Palate Craniofac J 2010; 47:566-73. [PMID: 20500056 DOI: 10.1597/09-145] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Development of the secondary palate in mammals is a complex process that can be easily perturbed, leading to the common and distressing birth defect cleft palate. Animal models are particularly useful tools for dissecting underlying genetic components of cleft palate. We describe a new cleft palate model resulting from a transgene insertion mutation. Transgene insertional mutagenesis disrupts the genomic organization and expression of the Ap2β1 gene located on chromosome 11. This gene encodes the β2-adaptin subunit of the heterotetrameric adaptor protein 2 complex involved in clathrin-dependent endocytosis. Homozygous cleft palate mutant mice express no Ap2β1 messenger RNA or β2-adaptin protein and die during the perinatal period. Heterozygous mice are phenotypically normal despite expressing diminished β2-adaptin messenger RNA and protein compared with wildtype. Remarkably, the paralogous β1-adaptin subunit of the adaptor protein 1 complex partially substitutes for the missing β2-adaptin in embryonic fibroblasts from homozygous mutant mice, resulting in assembly of reduced levels of an adaptor protein 2 complex bearing β1-adaptin. This variant adaptor protein 2 complex is, therefore, apparently capable of maintaining viability of the homozygous mutant embryos until birth but insufficient to support palatogenesis. Nonsyndromic cleft palate in an animal model is associated with disruption of the Ap2β1 gene.
Collapse
Affiliation(s)
- Wei Li
- Department of Oral Facial Development, Indiana University School of Dentistry, Indianapolis, Indiana, USA
| | | | | | | | | |
Collapse
|
42
|
Kang JS, Liu C, Derynck R. New regulatory mechanisms of TGF-beta receptor function. Trends Cell Biol 2009; 19:385-94. [PMID: 19648010 DOI: 10.1016/j.tcb.2009.05.008] [Citation(s) in RCA: 276] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2009] [Revised: 05/05/2009] [Accepted: 05/08/2009] [Indexed: 12/24/2022]
Abstract
Transforming growth factor-beta (TGF-beta) regulates cell proliferation, differentiation and apoptosis, and TGF-beta-related proteins have key roles in development, tissue homeostasis and disease. Upon binding to their cell surface receptors, TGF-beta family proteins signal through Smads to induce changes in gene expression. TGF-beta-induced Smad signaling and additional non-Smad pathways have been studied extensively in an effort to understand the complex and versatile responses to TGF-beta family proteins. Recently, it has become increasingly apparent that the signaling responses are also extensively defined by regulatory mechanisms at the level of the receptors themselves. Here, we discuss recent insights into the effects of post-translational modifications, protein associations and mode of internalization on the functions of the TGF-beta receptors and their signaling responses.
Collapse
Affiliation(s)
- Jong Seok Kang
- Department of Cell and Tissue Biology, University of California - San Francisco, San Francisco, CA 94143, USA
| | | | | |
Collapse
|
43
|
The extracellular domain of the TGFβ type II receptor regulates membrane raft partitioning. Biochem J 2009; 421:119-31. [DOI: 10.1042/bj20081131] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cell-surface TGFβ (transforming growth factor β) receptors partition into membrane rafts and the caveolin-positive endocytic compartment by an unknown mechanism. In the present study, we investigated the determinant in the TGFβ type II receptor (TβRII) that is necessary for membrane raft/caveolar targeting. Using subcellular fractionation and immunofluorescence microscopy techniques, we demonstrated that the extracellular domain of TβRII mediates receptor partitioning into raft and caveolin-positive membrane domains. Pharmacological perturbation of glycosylation using tunicamycin or the mutation of Mgat5 [mannosyl(α-1,6)-glycoprotein β-1,6-N-acetylglucosaminyltransferase V] activity interfered with the raft partitioning of TβRII. However, this was not due to the glycosylation state of TβRII, as a non-glycosylated TβRII mutant remained enriched in membrane rafts. This suggested that other cell-surface glycoproteins associate with the extracellular domain of TβRII and direct their partitioning in membrane raft domains. To test this we analysed a GMCSF (granulocyte/macrophage colony-stimulating factor)–TβRII chimaeric receptor, which contains a glycosylated GMCSF extracellular domain fused to the transmembrane and intracellular domains of TβRII. This chimaeric receptor was found to be largely excluded from membrane rafts and caveolin-positive structures. Our results indicate that the extracellular domain of TβRII mediates receptor partitioning into membrane rafts and efficient entrance into caveolin-positive endosomes.
Collapse
|
44
|
Kardassis D, Murphy C, Fotsis T, Moustakas A, Stournaras C. Control of transforming growth factor β signal transduction by small GTPases. FEBS J 2009; 276:2947-65. [DOI: 10.1111/j.1742-4658.2009.07031.x] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
45
|
Endocytosis of adiponectin receptor 1 through a clathrin- and Rab5-dependent pathway. Cell Res 2009; 19:317-27. [PMID: 18982021 DOI: 10.1038/cr.2008.299] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In eukaryotic cells, receptor endocytosis is a key event regulating signaling transduction. Adiponectin receptors belong to a new receptor family that is distinct from G-protein-coupled receptors and has critical roles in the pathogenesis of diabetes and metabolic syndrome. Here, we analyzed the endocytosis of adiponectin and adiponectin receptor 1 (AdipoR1) and found that they are both internalized into transferrin-positive compartments that follow similar traffic routes. Blocking clathrin-mediated endocytosis by expressing Eps15 mutants or depleting K(+) trapped AdipoR1 at the plasma membrane, and K(+) depletion abolished adiponectin internalization, indicating that the endocytosis of AdipoR1 and adiponectin is clathrin-dependent. Depletion of K(+) and overexpression of Eps15 mutants enhance adiponectin-stimulated AMP-activated protein kinase phosphorylation, suggesting that the endocytosis of AdipoR1 might downregulate adiponectin signaling. In addition, AdipoR1 colocalizes with the small GTPase Rab5, and a dominant negative Rab5 abrogates AdipoR1 endocytosis. These data indicate that AdipoR1 is internalized through a clathrin- and Rab5-dependent pathway and that endocytosis may play a role in the regulation of adiponectin signaling.
Collapse
|
46
|
Abstract
The recruitment of clathrin to the membrane and its assembly into coated pits results from its interaction with endocytic adaptors and other regulatory proteins in the context of a specific lipid microenvironment. Dab2 (disabled 2) is a mitotic phosphoprotein and a monomeric adaptor for clathrin-mediated endocytosis. In the present study, we employed GFP (green fluorescent protein) fusion constructs of different isoforms and mutants of rat Dab2 and characterized their effect on the size, distribution and dynamics of clathrin assemblies. Enhanced levels of expression of the p82 isoform of Dab2 in COS7 cells induced enlarged clathrin assemblies at the plasma membrane. p82-clathrin assemblies, which concentrate additional endocytic proteins, such as AP2 (adaptor protein 2) and epsin, are dynamic structures in which both p82 and clathrin exchange actively between the membrane-bound and cytosolic sub-populations. The ability of p82 to induce enlarged clathrin assemblies is dependent on the presence of a functional PTB domain (phosphotyrosine-binding domain), on binding to clathrin and phospholipids, and on a newly identified and evolutionarily conserved poly-lysine stretch which precedes the PTB domain. These same molecular features are required for Dab2 to enhance the spreading of COS7 cells on fibronectin. The ability of the p82 isoform of Dab2 to enhance cell spreading was confirmed in both HeLa cells and HBL cells (human breast epithelial cells). COS7 cells expressing GFP-p82 and plated on to fibronectin concentrate the beta1 integrin into clathrin-p82 assemblies. Furthermore, during cell spreading, p82-clathrin assemblies concentrate at the site of the initial cell-matrix contact and are absent from regions of intense membrane ruffling. We propose a role for Dab2 and clathrin in integrin-mediated cell spreading.
Collapse
|
47
|
Abstract
Transforming growth factor-beta (TGF-beta) signaling is tightly regulated to ensure its proper physiological functions in different cells and tissues. Like other cell surface receptors, TGF-beta receptors are internalized into the cell, and this process plays an important regulatory role in TGF-beta signaling. It is well documented that TGF-beta receptors are endocytosed via clathrin-coated vesicles as TGF-beta endocytosis can be blocked by potassium depletion and the GTPase-deficient dynamin K44A mutant. TGF-beta receptors may also enter cells via cholesterol-rich membrane microdomain lipid rafts/caveolae and are found in caveolin-1-positive vesicles. Although receptor endocytosis is not essential for TGF-beta signaling, clathrin-mediated endocytosis has been shown to promote TGF-beta-induced Smad activation and transcriptional responses. Lipid rafts/caveolae are widely regarded as signaling centers for G protein-coupled receptors and tyrosine kinase receptors, but they are indicated to facilitate the degradation of TGF-beta receptors and therefore turnoff of TGF-beta signaling. This review summarizes current understanding of TGF-beta receptor endocytosis, the possible mechanisms underlying this process, and the role of endocytosis in modulation of TGF-beta signaling.
Collapse
Affiliation(s)
- Ye-Guang Chen
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
48
|
Rechtman MM, Nakaryakov A, Shapira KE, Ehrlich M, Henis YI. Different domains regulate homomeric and heteromeric complex formation among type I and type II transforming growth factor-beta receptors. J Biol Chem 2009; 284:7843-52. [PMID: 19147499 DOI: 10.1074/jbc.m809215200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Transforming growth factor-beta (TGF-beta) binds to and signals via two serine-threonine kinase receptors, type I (TbetaRI) and type II (TbetaRII). The oligomerization of TGF-beta receptors modulates ligand binding and receptor trafficking and may contribute to signal diversification. However, numerous features of the molecular domains that determine the homo- and hetero-oligomerization of full-length receptors at the cell surface and the mode of these interactions remain unclear. Here, we address these questions through computerized immunofluorescence co-patching and patch/fluorescence recovery after photobleaching measurements of different combinations of epitope-tagged receptors and their mutants in live cells. We show that TbetaRI and TbetaRII are present on the plasma membrane both as monomers and homo- and hetero-oligomers. The homodimerization of TbetaRII depends on a cytoplasmic juxtamembrane region (amino acid residues 200-220). In contrast, the cytoplasmic domain of TbetaRI is dispensable for its homodimerization. TbetaRI.TbetaRII hetero-oligomerization depends on the cytoplasmic domain of TbetaRI and on a C-terminal region of TbetaRII (residues 419-565). TGF-beta1 elevates TbetaRII homodimerization to some degree and strongly enhances TbetaRI.TbetaRII heteromeric complex formation. Both ligand-induced effects depend on the region encompassed between residues 200-242 of TbetaRII. Furthermore, the kinase activity of TbetaRI is also necessary for the latter effect. All forms of the homo- and hetero-oligomers, whether constitutively present on the membrane or formed upon TGF-beta1 stimulation, were stable in the time-scale of our patch/FRAP measurements. We suggest that the different forms of receptor oligomerization may serve as a basis for the heterogeneity of TGF-beta signaling responses.
Collapse
Affiliation(s)
- Maya Mouler Rechtman
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | | | | | | | | |
Collapse
|
49
|
Finger EC, Lee NY, You HJ, Blobe GC. Endocytosis of the type III transforming growth factor-beta (TGF-beta) receptor through the clathrin-independent/lipid raft pathway regulates TGF-beta signaling and receptor down-regulation. J Biol Chem 2008; 283:34808-18. [PMID: 18845534 PMCID: PMC2596377 DOI: 10.1074/jbc.m804741200] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2008] [Revised: 09/26/2008] [Indexed: 11/06/2022] Open
Abstract
Transforming growth factor-beta (TGF-beta) signals through three highly conserved cell surface receptors, the type III TGF-beta receptor (T beta RIII), the type II TGF-beta receptor (T beta RII), and the type I TGF-beta receptor (T beta RI) to regulate diverse cellular processes including cell proliferation, differentiation, migration, and apoptosis. Although T beta RI and T beta RII undergo ligand-independent endocytosis by both clathrin-mediated endocytosis, resulting in enhanced signaling, and clathrin-independent endocytosis, resulting in receptor degradation, the mechanism and function of T beta RIII endocytosis is poorly understood. T beta RIII is a heparan sulfate proteoglycan with a short cytoplasmic tail that functions as a TGF-beta superfamily co-receptor, contributing to TGF-beta signaling through mechanisms yet to be fully defined. We have reported previously that T beta RIII endocytosis, mediated by a novel interaction with beta arrestin-2, results in decreased TGF-beta signaling. Here we demonstrate that T beta RIII undergoes endocytosis in a ligand and glycosaminoglycan modification-independent and cytoplasmic domain-dependent manner, with the interaction of Thr-841 in the cytoplasmic domain of T beta RIII with beta-arrestin2 enhancing T beta RIII endocytosis. T beta RIII undergoes both clathrin-mediated and clathrin-independent endocytosis. Importantly, inhibition of the clathrin-independent, lipid raft pathway, but not of the clathrin-dependent pathway, results in decreased TGF-beta1 induced Smad2 and p38 phosphorylation, supporting a specific role for clathrin-independent endocytosis of T beta RIII in regulating both Smad-dependent and Smad-independent TGF-beta signaling.
Collapse
Affiliation(s)
- Elizabeth C. Finger
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Nam Y. Lee
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Hye-jin You
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| | - Gerard C. Blobe
- Departments of Pharmacology and Cancer
Biology and Medicine, Duke University Medical
Center, Durham, North Carolina 27708
| |
Collapse
|
50
|
Zwaagstra JC, Collins C, Langlois MJ, O'Connor-McCourt MD. Analysis of the contribution of receptor subdomains to the cooperative binding and internalization of transforming growth factor-β (TGF-β) type I and type II receptors. Exp Cell Res 2008; 314:2553-68. [DOI: 10.1016/j.yexcr.2008.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 06/03/2008] [Accepted: 06/04/2008] [Indexed: 10/22/2022]
|