1
|
Chu K, Crawford AN, Krah BS, Thamilselvan V, Malik A, Aitas NA, Martinez‐Hackert E. Cripto-1 acts as a molecular bridge linking nodal to ALK4 via distinct structural domains. Protein Sci 2025; 34:e70034. [PMID: 39840816 PMCID: PMC11751877 DOI: 10.1002/pro.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2024] [Revised: 11/20/2024] [Accepted: 01/03/2025] [Indexed: 01/23/2025]
Abstract
The TGF-β family ligand Nodal is an essential regulator of embryonic development, orchestrating key processes such as germ layer specification and body axis formation through activation of SMAD2/3-mediated signaling. Significantly, this activation requires the co-receptor Cripto-1. However, despite their essential roles in embryogenesis, the molecular mechanism through which Cripto-1 enables Nodal to activate the SMAD2/3 pathway has remained elusive. Intriguingly, Cripto-1 also has been shown to antagonize other TGF-β family ligands, raising questions about its diverse functions. To clarify how Cripto-1 modulates TGF-β signaling, we integrated AlphaFold3 modeling, surface plasmon resonance (SPR)-based protein-protein interaction analysis, domain-specific anti-Cripto-1 antibodies, and functional studies in NTERA-2 cells. In contrast to canonical TGF-β signaling, where ligands bridge type I and type II receptors for activation, Nodal, bound to the type II receptor, utilizes Cripto-1 to recruit the type I receptor ALK4, forming a unique ternary complex for SMAD2/3 activation. Our molecular characterization of Cripto-1-mediated Nodal signaling clarifies the unique role of this enigmatic co-receptor and advances our understanding of signaling regulation within the TGF-β family. These insights have potential implications for both developmental biology and cancer research.
Collapse
Affiliation(s)
- Kit‐Yee Chu
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMichiganUSA
| | - Amberly N. Crawford
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMichiganUSA
| | - Bradon S. Krah
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMichiganUSA
| | | | - Anjali Malik
- Department of Structural BiologyVan Andel InstituteGrand RapidsMichiganUSA
| | - Nina A. Aitas
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMichiganUSA
| | - Erik Martinez‐Hackert
- Department of Biochemistry and Molecular BiologyMichigan State UniversityEast LansingMichiganUSA
| |
Collapse
|
2
|
Enns CA, Zhang RH, Jue S, Zhang AS. Hepcidin expression is associated with increased γ-secretase-mediated cleavage of neogenin in the liver. J Biol Chem 2024; 300:107927. [PMID: 39454953 PMCID: PMC11599459 DOI: 10.1016/j.jbc.2024.107927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/24/2024] [Revised: 09/26/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Neogenin (NEO1) is a ubiquitously expressed transmembrane protein. It interacts with hemojuvelin (HJV). Both NEO1 and HJV play pivotal roles in iron homeostasis by inducing hepcidin expression in the liver. Our previous studies demonstrated that this process depends on Neo1-Hjv interaction and showed that the Hjv-mediated hepcidin expression is correlated with the accumulation of a truncated and membrane-associated form of Neo1. In this study, we tested whether hepcidin expression is induced by increased γ-secretase-mediated cleavage of Neo1 in the liver. We found that Neo1 underwent cleavage of its ectodomain and intracellular domains by α- and γ-secretases, respectively, in hepatoma cells. Our in vitro studies suggest that γ-secretase is responsible for cleavage and release of the cytoplasmic domain of Neo1 in the Hjv-Neo1 complex. This process was enhanced by the inhibition of α-secretase proteolysis and by co-expression with the Neo1-binding partner, Alk3. Further in vivo studies indicated that Neo1 induction of hepcidin expression required γ-secretase cleavage. Interestingly, neither predicted form of γ-secretase-cleaved Neo1 was able to induce hepcidin when separately expressed in hepatocyte-specific Neo1 KO mice. These results imply that the function of Neo1 requires a de novo γ-secretase proteolysis. Additional studies revealed that in addition to the Hjv-binding domains, the function of Neo1 also required its C-terminal intracellular domain and the N-terminal immunoglobulin-like domains that are involved in Neo1 binding to Alk3. Together, our data support the idea that Neo1 induction of hepcidin is initiated as a full-length form and requires a de novo γ-secretase cleavage of Neo1's cytoplasmic domain.
Collapse
Affiliation(s)
- Caroline A Enns
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Richard H Zhang
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - Shall Jue
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA
| | - An-Sheng Zhang
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon, USA.
| |
Collapse
|
3
|
Zhang J, Jiang Y, Zhang Z, Li S, Fan H, Gu J, Mao R, Xu X. Repulsive guidance molecules b (RGMb): molecular mechanism, function and role in diseases. Expert Rev Mol Med 2024; 26:e24. [PMID: 39375839 PMCID: PMC11488336 DOI: 10.1017/erm.2024.24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/26/2023] [Revised: 12/23/2023] [Accepted: 06/11/2024] [Indexed: 10/09/2024]
Abstract
Repulsive guidance molecule b (RGMb), a glycosylphosphatidylinositol-anchored member of the RGM family, is initially identified as a co-receptor of bone morphogenetic protein (BMP) in the nervous system. The expression of RGMb is transcriptionally regulated by dorsal root ganglion 11 (DRG11), which is a transcription factor expressed in embryonic DRG and dorsal horn neurons and plays an important role in the development of sensory circuits. RGMb is involved in important physiological processes such as embryonic development, immune response, intercellular adhesion and tumorigenesis. Furthermore, RGMb is mainly involved in the regulation of RGMb-neogenin-Rho and BMP signalling pathways. The recent discovery of programmed death-ligand 2 (PD-L2)-RGMb binding reveals that the cell signalling network and functional regulation centred on RGMb are extremely complex. The latest report suggests that down-regulation of the PD-L2-RGMb pathway in the gut microbiota promotes an anti-tumour immune response, which defines a potentially effective immune strategy. However, the biological function of RGMb in a variety of human diseases has not been fully determined, and will remain an active research field. This article reviews the properties and functions of RGMb, focusing on its role under various physiological and pathological conditions.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Yijing Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Zijian Zhang
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Shilin Li
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Haowen Fan
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Jinhua Gu
- Nantong Institute of Genetics and Reproductive Medicine, Affiliated Maternity & Child Healthcare Hospital of Nantong University, Nantong, Jiangsu, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, Jiangsu, People's Republic of China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong University, Nantong, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Yeshaya N, Gupta PK, Dym O, Morgenstern D, Major DT, Fass D. VWD domain stabilization by autocatalytic Asp-Pro cleavage. Protein Sci 2024; 33:e4929. [PMID: 38380729 PMCID: PMC10880436 DOI: 10.1002/pro.4929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/25/2023] [Revised: 01/17/2024] [Accepted: 01/30/2024] [Indexed: 02/22/2024]
Abstract
Domains known as von Willebrand factor type D (VWD) are found in extracellular and cell-surface proteins including von Willebrand factor, mucins, and various signaling molecules and receptors. Many VWD domains have a glycine-aspartate-proline-histidine (GDPH) amino-acid sequence motif, which is hydrolytically cleaved post-translationally between the aspartate (Asp) and proline (Pro). The Fc IgG binding protein (FCGBP), found in intestinal mucus secretions and other extracellular environments, contains 13 VWD domains, 11 of which have a GDPH cleavage site. In this study, we investigated the structural and biophysical consequences of Asp-Pro peptide cleavage in a representative FCGBP VWD domain. We found that endogenous Asp-Pro cleavage increases the resistance of the domain to exogenous proteolytic degradation. Tertiary structural interactions made by the newly generated chain termini, as revealed by a crystal structure of an FCGBP segment containing the VWD domain, may explain this observation. Notably, the Gly-Asp peptide bond, upstream of the cleavage site, assumed the cis configuration in the structure. In addition to these local features of the cleavage site, a global organizational difference was seen when comparing the FCGBP segment structure with the numerous other structures containing the same set of domains. Together, these data illuminate the outcome of GDPH cleavage and demonstrate the plasticity of proteins with VWD domains, which may contribute to their evolution for function in a dynamic extracellular environment.
Collapse
Affiliation(s)
- Noa Yeshaya
- Department of Chemical and Structural BiologyWeizmann Institute of ScienceRehovotIsrael
| | - Prashant Kumar Gupta
- Department of Chemistry and Institute for Nanotechnology & Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Orly Dym
- Department of Life Sciences Core FacilitiesWeizmann Institute of ScienceRehovotIsrael
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized MedicineWeizmann Institute of ScienceRehovotIsrael
| | - Dan Thomas Major
- Department of Chemistry and Institute for Nanotechnology & Advanced MaterialsBar‐Ilan UniversityRamat‐GanIsrael
| | - Deborah Fass
- Department of Chemical and Structural BiologyWeizmann Institute of ScienceRehovotIsrael
| |
Collapse
|
5
|
Meira M, Frey A, Chekkat N, Rybczynska M, Sellam Z, Park JS, Gazzaniga FS, Parmentier A, Le Gall M, Freeman GJ, Kasper DL, Sharpe AH, Rambeaux E, Shamshiev A. Targeting RGMb interactions: Discovery and preclinical characterization of potent anti-RGMb antibodies blocking multiple ligand bindings. MAbs 2024; 16:2432403. [PMID: 39588913 PMCID: PMC11601088 DOI: 10.1080/19420862.2024.2432403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/18/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024] Open
Abstract
Therapeutic efficacy with durable responses has been demonstrated with several antibody drugs that block key immune checkpoint receptors, including PD-1, PD-L1, and CTLA-4. Despite the success of these drugs, a substantial proportion of patients do not benefit. Targeting multiple inhibitory pathways simultaneously to augment anti-tumor immunity has proven to be a promising approach. The emergence of Repulsive Guidance Molecule b (RGMb), a ligand for PD-L2, as a novel co-inhibitory pathway in T cells, together with its regulation by the gut microbiome, encouraged the discovery and development of fully human anti-RGMb antibodies. Here, we describe phage display-derived monoclonal antibodies (mAbs) 2C11 and 5C10 that bind human RGMb with high affinities of 1.4 nM and 0.72 nM, respectively. Both mAbs 2C11 and 5C10 potently inhibited RGMb interaction with PD-L2. MAb 2C11 effectively inhibited RGMb interaction with bone morphogenetic proteins 2 and 4 (BMP2-4), while leaving RGMb interaction with Neogenin 1 (Neo1) unaffected. Conversely, mAb 5C10 disrupted RGMb interaction with Neo1 while maintaining RGMb binding to BMP2-4. These findings map the 2C11 epitope at the membrane-distal N-terminal region of RGMb, which coincides with both PD-L2- and BMP2-4-binding sites. The PD-L2 binding interface is likely positioned between RGMb's N-terminal BMP-binding and C-terminal Neo1-binding regions. The in vivo activity of mAb 2C11 in combination with anti-PD-1 or anti-PD-L1 was tested in MC38 and B16-OVA cancer models and demonstrated synergistic effects by significantly enhancing anti-tumor responses. These properties make mAb 2C11 a promising candidate for therapeutic use to overcome immune checkpoint inhibitor resistances, warranting further exploration in clinical settings.
Collapse
Affiliation(s)
- Maria Meira
- R&D Department, IOME Bio SA, Strasbourg, France
| | - Aurore Frey
- R&D Department, IOME Bio SA, Strasbourg, France
| | | | | | - Zaki Sellam
- R&D Department, IOME Bio SA, Strasbourg, France
| | - Joon Seok Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | - Gordon James Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dennis Lee Kasper
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | - Arlene Helen Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
| | | | | |
Collapse
|
6
|
Zhang K, An X, Zhu Y, Huang L, Yao X, Zeng X, Liang S, Yu J. Netrin-1 inducing antiapoptotic effect of acute myeloid leukemia cells in a concentration-dependent manner through the Unc-5 netrin receptor B-focal adhesion kinase axis. Cancer Biol Ther 2023; 24:2200705. [PMID: 37038247 PMCID: PMC10088980 DOI: 10.1080/15384047.2023.2200705] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 04/12/2023] Open
Abstract
Acute myeloid leukemia (AML) is a hematological malignancy that commonly occurs in children. The prognosis of pediatric AML is relatively poor, thus threatening the patient's survival. The aberrant expression of the axon guidance factor, netrin-1, is observed in various types of malignancies, and it participates in the proliferation and apoptosis of tumor cells. Herein, we aimed to explore the role of netrin-1 in AML cells. Netrin-1 is highly expressed in AML patients. Proliferation and anti-apoptosis were observed in AML cells treated with netrin-1. The interaction between netrin-1 and Unc-5 netrin receptor B (UNC5B) was detected through coimmunoprecipitation, and UNC5B ribonucleic acid interference restrained the influence of netrin-1 on the AML cells. The phosphorylation of focal adhesion kinase-protein kinase B (FAK-Akt) was upregulated in AML cells treated with netrin-1. Both FAK and Akt inhibitors abrogated the effects of netrin-1 on the proliferation and apoptosis of AML cells. In conclusion, netrin-1 could promote the growth and reduce the apoptosis of AML cells in a concentration-dependent manner, and that these effects were mediated by activating the FAK-Akt signaling pathway via the UNC5B.
Collapse
Affiliation(s)
- Kainan Zhang
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Pediatric research institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xizhou An
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Yao Zhu
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Pediatric research institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Lan Huang
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
- Pediatric research institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Xinyuan Yao
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Xing Zeng
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Shaoyan Liang
- Pediatric research institute, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Yu
- Department of Hematology and Oncology, Children's Hospital of Chongqing Medical University; National Clinical Research Center for Child Health and Disorders; Ministry of Education Key Laboratory of Child Development and Disorders; Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
7
|
Sempert K, Shohayeb B, Lanoue V, O'Brien EA, Flores C, Cooper HM. RGMa and Neogenin control dendritic spine morphogenesis via WAVE Regulatory Complex-mediated actin remodeling. Front Mol Neurosci 2023; 16:1253801. [PMID: 37928069 PMCID: PMC10620725 DOI: 10.3389/fnmol.2023.1253801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/06/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
Structural plasticity, the ability of dendritic spines to change their volume in response to synaptic stimulation, is an essential determinant of synaptic strength and long-term potentiation (LTP), the proposed cellular substrate for learning and memory. Branched actin polymerization is a major force driving spine enlargement and sustains structural plasticity. The WAVE Regulatory Complex (WRC), a pivotal branched actin regulator, controls spine morphology and therefore structural plasticity. However, the molecular mechanisms that govern WRC activation during spine enlargement are largely unknown. Here we identify a critical role for Neogenin and its ligand RGMa (Repulsive Guidance Molecule a) in promoting spine enlargement through the activation of WRC-mediated branched actin remodeling. We demonstrate that Neogenin regulates WRC activity by binding to the highly conserved Cyfip/Abi binding pocket within the WRC. We find that after Neogenin or RGMa depletion, the proportions of filopodia and immature thin spines are dramatically increased, and the number of mature mushroom spines concomitantly decreased. Wildtype Neogenin, but not Neogenin bearing mutations in the Cyfip/Abi binding motif, is able to rescue the spine enlargement defect. Furthermore, Neogenin depletion inhibits actin polymerization in the spine head, an effect that is not restored by the mutant. We conclude that RGMa and Neogenin are critical modulators of WRC-mediated branched actin polymerization promoting spine enlargement. This study also provides mechanistic insight into Neogenin's emerging role in LTP induction.
Collapse
Affiliation(s)
- Kai Sempert
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Belal Shohayeb
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Vanessa Lanoue
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Elizabeth A O'Brien
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Cecilia Flores
- Department of Psychiatry, McGill University, Montréal, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada
- Douglas Mental Health University Institute, Montréal, QC, Canada
| | - Helen M Cooper
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Weste J, Houben T, Harder S, Schlüter H, Lücke E, Schreiber J, Hoffmann W. Different Molecular Forms of TFF3 in the Human Respiratory Tract: Heterodimerization with IgG Fc Binding Protein (FCGBP) and Proteolytic Cleavage in Bronchial Secretions. Int J Mol Sci 2022; 23:ijms232315359. [PMID: 36499686 PMCID: PMC9737082 DOI: 10.3390/ijms232315359] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/26/2022] [Revised: 11/25/2022] [Accepted: 11/25/2022] [Indexed: 12/12/2022] Open
Abstract
The polypeptide TFF3 belongs to the trefoil factor family (TFF) of lectins. TFF3 is typically secreted from mucous epithelia together with mucins. Both intestinal and salivary TFF3 mainly exist as disulfide-linked heterodimers with IgG Fc binding protein (FCGBP). Here, we investigated bronchial tissue specimens, bronchial secretions, and bronchoalveolar lavage (BAL) fluid from patients with a chronic obstructive pulmonary disease (COPD) background by fast protein liquid chromatography and proteomics. For the first time, we identified different molecular forms of TFF3 in the lung. The high-molecular mass form represents TFF3-FCGBP oligomers, whereas the low-molecular mass forms are homodimeric and monomeric TFF3 with possibly anti-apoptotic activities. In addition, disulfide-linked TFF3 heterodimers with an Mr of about 60k and 30k were detected in both bronchial secretions and BAL fluid. In these liquids, TFF3 is partly N-terminally truncated probably by neutrophil elastase cleavage. TFF3-FCGBP is likely involved in the mucosal innate immune defense against microbial infections. We discuss a hypothetical model how TFF3 might control FCGBP oligomerization. Furthermore, we did not find indications for interactions of TFF3-FCGBP with DMBT1gp340 or the mucin MUC5AC, glycoproteins involved in mucosal innate immunity. Surprisingly, bronchial MUC5AC appeared to be degraded when compared with gastric MUC5AC.
Collapse
Affiliation(s)
- Jens Weste
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Till Houben
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Sönke Harder
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Hartmut Schlüter
- Section Mass Spectrometry and Proteomics, Diagnostic Center, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Eva Lücke
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Jens Schreiber
- Department of Pneumology, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
| | - Werner Hoffmann
- Institute of Molecular Biology and Medicinal Chemistry, Otto-von-Guericke University Magdeburg, 39120 Magdeburg, Germany
- Correspondence:
| |
Collapse
|
9
|
Pérez-Cruz M, Iliopoulou BP, Hsu K, Wu HH, Erkers T, Swaminathan K, Tang SW, Bader CS, Kambham N, Xie B, Dekruyff RH, Freeman GJ, Meyer E. Immunoregulatory effects of RGMb in gut inflammation. Front Immunol 2022; 13:960329. [PMID: 36420263 PMCID: PMC9676481 DOI: 10.3389/fimmu.2022.960329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/02/2022] [Accepted: 10/19/2022] [Indexed: 11/09/2022] Open
Abstract
Graft-versus-host disease (GvHD) is a major complication after allogeneic hematopoietic cell transplantation (HCT). Current strategies to prevent GvHD with immunosuppressive drugs carry significant morbidity and may affect the graft-versus-tumor (GVT) effect. Inflammatory bowel disease (IBD) is an intestinal inflammatory condition that affects more than 2 million people in the United States. Current strategies to prevent colitis with immunosuppressive drugs carry significant morbidity. Recently, Repulsive Guidance Molecule b (RGMb) has been identified as part of a signaling hub with neogenin and BMP receptors in mice and humans. In addition, RGMb binds BMP-2/4 in mice and humans as well as PD-L2 in mice. RGMb is expressed in the gut epithelium and by antigen presenting cells, and we found significantly increased expression in mouse small intestine after total body irradiation HCT conditioning. We hypothesized that RGMb may play a role in GvHD and IBD pathogenesis by contributing to mucosal inflammation. Using major-mismatched HCT mouse models, treatment with an anti-RGMb monoclonal antibody (mAb) that blocks the interaction with BMP-2/4 and neogenin prevented GvHD and improved survival compared to isotype control (75% versus 30% survival at 60 days after transplantation). The GVT effect was retained in tumor models. Using an inflammatory bowel disease dextran sulfate sodium model, treatment with anti-RGMb blocking monoclonal antibody but not isotype control prevented colitis and improved survival compared to control (73% versus 33% at 21 days after treatment) restoring gut homeostasis. Anti-RGMb mAb (9D1) treatment decreased IFN-γ and significantly increased IL-5 and IL-10 in the gut of the treated mice compared to the isotype control treated mice.
Collapse
Affiliation(s)
- Magdiel Pérez-Cruz
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Bettina P. Iliopoulou
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Katie Hsu
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Hsin-Hsu Wu
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Tom Erkers
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Kavya Swaminathan
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Sai-Wen Tang
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Cameron S. Bader
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Neeraja Kambham
- Department of Developmental biology, Stanford University School of Medicine, Stanford, CA, United States
| | - Bryan Xie
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Rosemarie H. Dekruyff
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, United States
| | - Gordon J. Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Everett Meyer
- Division of Blood and Marrow Transplantation, Department of Medicine, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
10
|
Novak R, Ahmad YA, Timaner M, Bitman-Lotan E, Oknin-Vaisman A, Horwitz R, Hartmann O, Reissland M, Buck V, Rosenfeldt M, Nikomarov D, Diefenbacher ME, Shaked Y, Orian A. RNF4~RGMb~BMP6 axis required for osteogenic differentiation and cancer cell survival. Cell Death Dis 2022; 13:820. [PMID: 36153321 PMCID: PMC9509360 DOI: 10.1038/s41419-022-05262-1] [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] [Academic Contribution Register] [Received: 08/04/2022] [Revised: 09/08/2022] [Accepted: 09/12/2022] [Indexed: 01/23/2023]
Abstract
Molecular understanding of osteogenic differentiation (OD) of human bone marrow-derived mesenchymal stem cells (hBMSCs) is important for regenerative medicine and has direct implications for cancer. We report that the RNF4 ubiquitin ligase is essential for OD of hBMSCs, and that RNF4-deficient hBMSCs remain as stalled progenitors. Remarkably, incubation of RNF4-deficient hBMSCs in conditioned media of differentiating hBMSCs restored OD. Transcriptional analysis of RNF4-dependent gene signatures identified two secreted factors that act downstream of RNF4 promoting OD: (1) BMP6 and (2) the BMP6 co-receptor, RGMb (Dragon). Indeed, knockdown of either RGMb or BMP6 in hBMSCs halted OD, while only the combined co-addition of purified RGMb and BMP6 proteins to RNF4-deficient hBMSCs fully restored OD. Moreover, we found that the RNF4-RGMb-BMP6 axis is essential for survival and tumorigenicity of osteosarcoma and therapy-resistant melanoma cells. Importantly, patient-derived sarcomas such as osteosarcoma, Ewing sarcoma, liposarcomas, and leiomyosarcomas exhibit high levels of RNF4 and BMP6, which are associated with reduced patient survival. Overall, we discovered that the RNF4~BMP6~RGMb axis is required for both OD and tumorigenesis.
Collapse
Affiliation(s)
- Rostislav Novak
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel ,Rambam Health Campus Center, Haifa, 3109610 Israel
| | - Yamen Abu Ahmad
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Michael Timaner
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Eliya Bitman-Lotan
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Avital Oknin-Vaisman
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Roi Horwitz
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Oliver Hartmann
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Michaela Reissland
- grid.8379.50000 0001 1958 8658Protein Stability and Cancer Group, University of Würzburg, Department of Biochemistry and Molecular Biology, Würzburg, Germany
| | - Viktoria Buck
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | - Mathias Rosenfeldt
- grid.8379.50000 0001 1958 8658Department of Pathology, University of Würzburg, Würzburg, Germany
| | | | - Markus Elmar Diefenbacher
- grid.8379.50000 0001 1958 8658Protein Stability and Cancer Group, University of Würzburg, Department of Biochemistry and Molecular Biology, Würzburg, Germany
| | - Yuval Shaked
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| | - Amir Orian
- grid.6451.60000000121102151Rappaport Research Institute and Faculty of Medicine, Technion Integrative Cancer Center Technion- IIT, Haifa, 3109 610 Israel
| |
Collapse
|
11
|
Chaudhari S, Yazdizadeh Shotorbani P, Tao Y, Kasetti R, Zode G, Mathis KW, Ma R. Neogenin pathway positively regulates fibronectin production by glomerular mesangial cells. Am J Physiol Cell Physiol 2022; 323:C226-C235. [PMID: 35704698 DOI: 10.1152/ajpcell.00359.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/22/2022]
Abstract
Neogenin, a transmembrane receptor, was recently found in kidney cells and immune cells. However, the function of neogenin signaling in kidney is not clear. Mesangial cells (MCs) are a major source of extracellular matrix (ECM) proteins in glomerulus. In many kidney diseases, MCs are impaired and manifest myofibroblast phenotype. Over production of ECM by the injured MCs promotes renal injury and accelerates the progression of kidney diseases. The present study was aimed to determine if neogenin receptor was expressed in MCs and if the receptor signaling regulated ECM protein production by MCs. We showed that neogenin was expressed in the glomerular MCs. Deletion of neogenin using CRISPR/Cas9 lentivirus system, significantly reduced the abundance of fibronectin, an ECM protein. Netrin-1, a ligand for neogenin, also significantly decreased fibronectin production by MCs and decreased neogenin protein expression in MCs. Furthermore, treatment of human MCs with high glucose (25 mM) significantly increased the protein abundance of neogenin as early as 8 h. Consistently, neogenin expression in glomerulus significantly increased in the eNOS-/- db/db diabetic mice starting as early as the age of 8 weeks and this increase sustained at least to the age of 24 weeks. We further found that the HG induced increase in neogenin abundance was blunted by antioxidant PEG-catalase and N-acetyl cysteine. Taken together, our results suggest a new mechanism of regulation of fibronectin production by MCs. This previously unrecognized neogenin-fibronectin pathway may contribute to glomerular injury responses during the course of diabetic nephropathy.
Collapse
Affiliation(s)
- Sarika Chaudhari
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | | | - Yu Tao
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Ramesh Kasetti
- The North Texas Eye Research Institute and Dept. of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, United States
| | - Gulab Zode
- The North Texas Eye Research Institute and Dept. of Pharmacology and Neuroscience, University of North Texas Health Science Center, Fort Worth, United States
| | - Keisa W Mathis
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| | - Rong Ma
- Dept. of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
12
|
Copola AGL, Dos Santos ÍGD, Coutinho LL, Del-Bem LEV, de Almeida Campos-Junior PH, da Conceição IMCA, Nogueira JM, do Carmo Costa A, Silva GAB, Jorge EC. Transcriptomic characterization of the molecular mechanisms induced by RGMa during skeletal muscle nuclei accretion and hypertrophy. BMC Genomics 2022; 23:188. [PMID: 35255809 PMCID: PMC8902710 DOI: 10.1186/s12864-022-08396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/22/2021] [Accepted: 02/15/2022] [Indexed: 12/02/2022] Open
Abstract
Background The repulsive guidance molecule a (RGMa) is a GPI-anchor axon guidance molecule first found to play important roles during neuronal development. RGMa expression patterns and signaling pathways via Neogenin and/or as BMP coreceptors indicated that this axon guidance molecule could also be working in other processes and diseases, including during myogenesis. Previous works from our research group have consistently shown that RGMa is expressed in skeletal muscle cells and that its overexpression induces both nuclei accretion and hypertrophy in muscle cell lineages. However, the cellular components and molecular mechanisms induced by RGMa during the differentiation of skeletal muscle cells are poorly understood. In this work, the global transcription expression profile of RGMa-treated C2C12 myoblasts during the differentiation stage, obtained by RNA-seq, were reported. Results RGMa treatment could modulate the expression pattern of 2,195 transcripts in C2C12 skeletal muscle, with 943 upregulated and 1,252 downregulated. Among them, RGMa interfered with the expression of several RNA types, including categories related to the regulation of RNA splicing and degradation. The data also suggested that nuclei accretion induced by RGMa could be due to their capacity to induce the expression of transcripts related to ‘adherens junsctions’ and ‘extracellular-cell adhesion’, while RGMa effects on muscle hypertrophy might be due to (i) the activation of the mTOR-Akt independent axis and (ii) the regulation of the expression of transcripts related to atrophy. Finally, RGMa induced the expression of transcripts that encode skeletal muscle structural proteins, especially from sarcolemma and also those associated with striated muscle cell differentiation. Conclusions These results provide comprehensive knowledge of skeletal muscle transcript changes and pathways in response to RGMa. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-022-08396-w.
Collapse
Affiliation(s)
- Aline Gonçalves Lio Copola
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Íria Gabriela Dias Dos Santos
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Luiz Lehmann Coutinho
- Departamento de Zootecnia, Escola Superior de Agricultura Luiz de Queiroz, Universidade de São Paulo, Piracicaba, Brasil
| | - Luiz Eduardo Vieira Del-Bem
- Departamento de Botânica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brasil
| | | | | | - Júlia Meireles Nogueira
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Alinne do Carmo Costa
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Gerluza Aparecida Borges Silva
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil
| | - Erika Cristina Jorge
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31.270-901, Brasil.
| |
Collapse
|
13
|
Huang L, Fung E, Bose S, Popp A, Böser P, Memmott J, Kutskova YA, Miller R, Tarcsa E, Klein C, Veldman GM, Mueller BK, Cui YF. Elezanumab, a clinical stage human monoclonal antibody that selectively targets repulsive guidance molecule A to promote neuroregeneration and neuroprotection in neuronal injury and demyelination models. Neurobiol Dis 2021; 159:105492. [PMID: 34478849 DOI: 10.1016/j.nbd.2021.105492] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/19/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022] Open
Abstract
Repulsive guidance molecule A (RGMa) is a potent inhibitor of axonal growth and a regulator of neuronal cell death. It is up-regulated following neuronal injury and accumulates in chronic neurodegenerative diseases. Neutralizing RGMa has the potential to promote neuroregeneration and neuroprotection. Previously we reported that a rat anti-N terminal RGMa (N-RGMa) antibody r5F9 and its humanized version h5F9 (ABT-207) promote neuroprotection and neuroregeneration in preclinical neurodegenerative disease models. However, due to its cross-reactivity to RGMc/hemojuvelin, ABT-207 causes iron accumulation in vivo, which could present a safety liability. Here we report the generation and characterization of a novel RGMa-selective anti-N-RGMa antibody elezanumab, which is currently under Phase 2 clinical evaluation in multiple disease indications. Elezanumab, a human monoclonal antibody generated by in vitro PROfusion mRNA display technology, competes with ABT-207 in binding to N-RGMa but lacks RGMc cross-reactivity with no impact on iron metabolism. It neutralizes repulsive activity of soluble RGMa in vitro and blocks membrane RGMa mediated BMP signaling. In the optic nerve crush and optic neuritis models, elezanumab promotes axonal regeneration and prevents retinal nerve fiber layer degeneration. In the spinal targeted experimental autoimmune encephalomyelitis (EAE) model, elezanumab promotes axonal regeneration and remyelination, decreases inflammatory lesion area and improves functional recovery. Finally, in the mouse cuprizone model, elezanumab reduces demyelination, which is consistent with its inhibitory effect on BMP signaling. Taken together, these preclinical data demonstrate that elezanumab has neuroregenerative and neuroprotective activities without impact on iron metabolism, thus providing a compelling rationale for its clinical development in neurodegenerative diseases.
Collapse
Affiliation(s)
- Lili Huang
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Emma Fung
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Sahana Bose
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Andreas Popp
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - Preethne Böser
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - John Memmott
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Yuliya A Kutskova
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Renee Miller
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Edit Tarcsa
- AbbVie Bioresearch Center, 100 Research Drive, Worcester, MA 01605, USA.
| | - Corinna Klein
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | | | - Bernhard K Mueller
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| | - Yi-Fang Cui
- AbbVie Deutschland GmbH & Co. KG, Knollstrasse, 67061, Ludwigshafen 67061, Germany.
| |
Collapse
|
14
|
Phan TL, Kim HJ, Lee SJ, Choi MC, Kim SH. Elevated RGMA Expression Predicts Poor Prognosis in Patients with Glioblastoma. Onco Targets Ther 2021; 14:4867-4878. [PMID: 34588781 PMCID: PMC8473061 DOI: 10.2147/ott.s317285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/23/2021] [Accepted: 09/10/2021] [Indexed: 11/23/2022] Open
Abstract
Background Glioblastoma (GBM) is the most aggressive type of human brain tumor with a poor prognosis and a low survival rate. Secreted proteins from tumors are recently considered as important modulators to promote tumorigenesis by communicating with microenvironments. Repulsive guidance molecule A (RGMA) was initially characterized as an axon guidance molecule after secretion in the brain during embryogenesis but has not been studied in GBM. In this study, we investigated secreted gene expression patterns and the correlation between RGMA expression and prognosis in GBM using in silico analysis. Methods RGMA mRNA levels in normal human astrocyte (NHA), human glioma cells, and GBM patient-derived glioma stem cells (GSCs) were assessed by qRT‐PCR. Patient survival analysis was performed with the Kaplan–Meier curve and univariate and multivariate analyses using publicly available datasets. The predictive roles of RGMA in progressive malignancy were evaluated using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA). Results RGMA mRNA expression was elevated in glioma cells and GSCs compared with NHA and correlated with unfavorable prognosis in glioma patients. Thus, RGMA could serve as an independent predictive factor for GBM. Furthermore, the increased levels of RGMA expression and its putative receptor, neogenin (NEO1), were associated with poor patient survival rates in GBM. Conclusion We identified RGMA as an independent prognostic biomarker for progressive malignancy in glioblastoma and address the possibilities to develop novel therapeutic strategies against glioblastoma.
Collapse
Affiliation(s)
- Thi Le Phan
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Jin Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Suk Jun Lee
- Department of Biomedical Laboratory Science, College of Health & Medical Sciences, Cheongju University, Chungbuk, 28503, Republic of Korea
| | - Moon-Chang Choi
- Department of Biomedical Science, Chosun University, Gwangju, 61452, Republic of Korea
| | - Sung-Hak Kim
- Department of Animal Science, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, Republic of Korea
| |
Collapse
|
15
|
Enns CA, Jue S, Zhang AS. Hepatocyte neogenin is required for hemojuvelin-mediated hepcidin expression and iron homeostasis in mice. Blood 2021; 138:486-499. [PMID: 33824974 PMCID: PMC8370464 DOI: 10.1182/blood.2020009485] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/08/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022] Open
Abstract
Neogenin (NEO1) is a ubiquitously expressed multifunctional transmembrane protein. It interacts with hemojuvelin (HJV), a BMP coreceptor that plays a pivotal role in hepatic hepcidin expression. Earlier studies suggest that the function of HJV relies on its interaction with NEO1. However, the role of NEO1 in iron homeostasis remains controversial because of the lack of an appropriate animal model. Here, we generated a hepatocyte-specific Neo1 knockout (Neo1fl/fl;Alb-Cre+) mouse model that circumvented the developmental and lethality issues of the global Neo1 mutant. Results show that ablation of hepatocyte Neo1 decreased hepcidin expression and caused iron overload. This iron overload did not result from altered iron utilization by erythropoiesis. Replacement studies revealed that expression of the Neo1L1046E mutant that does not interact with Hjv, was unable to correct the decreased hepcidin expression and high serum iron in Neo1fl/fl;Alb-Cre+ mice. In Hjv-/- mice, expression of HjvA183R mutant that has reduced interaction with Neo1, also displayed a blunted induction of hepcidin expression. These observations indicate that Neo1-Hjv interaction is essential for hepcidin expression. Further analyses suggest that the Hjv binding triggered the cleavage of the Neo1 cytoplasmic domain by a protease, which resulted in accumulation of truncated Neo1 on the plasma membrane. Additional studies did not support that Neo1 functions by inhibiting Hjv shedding as previously proposed. Together, our data favor a model in which Neo1 interaction with Hjv leads to accumulation of cleaved Neo1 on the plasma membrane, where Neo1 acts as a scaffold to induce the Bmp signaling and hepcidin expression.
Collapse
Affiliation(s)
- Caroline A Enns
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| | - Shall Jue
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| | - An-Sheng Zhang
- Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, OR
| |
Collapse
|
16
|
The IgGFc-binding protein FCGBP is secreted with all GDPH sequences cleaved but maintained by interfragment disulfide bonds. J Biol Chem 2021; 297:100871. [PMID: 34126068 PMCID: PMC8267560 DOI: 10.1016/j.jbc.2021.100871] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/25/2021] [Revised: 05/30/2021] [Accepted: 06/10/2021] [Indexed: 11/23/2022] Open
Abstract
Mucus forms an important protective barrier that minimizes bacterial contact with the colonic epithelium. Intestinal mucus is organized in a complex network with several specific proteins, including the mucin-2 (MUC2) and the abundant IgGFc-binding protein, FCGBP. FCGBP is expressed in all intestinal goblet cells and is secreted into the mucus. It is comprised of repeated von Willebrand D (vWD) domain assemblies, most of which have a GDPH amino acid sequence that can be autocatalytically cleaved, as previously observed in the mucins MUC2 and mucin-5AC. However, the functions of FCGBP in the mucus are not understood. We show that all vWD domains of FCGBP with a GDPH sequence are cleaved and that these cleavages occur early during biosynthesis in the endoplasmic reticulum. All cleaved fragments, however, remain connected via a disulfide bond within each vWD domain. This cleavage generates a C-terminal-reactive Asp-anhydride that could react with other molecules, such as MUC2, but this was not observed. Quantitative analyses by MS showed that FCGBP was mainly soluble in chaotropic solutions, whereas MUC2 was insoluble, and most of the secreted FCGBP was not covalently bound to MUC2. Although FCGBP has been suggested to bind immunoglobulin G, we were unable to reproduce this binding in vitro using purified proteins. In conclusion, while the function of FCGBP is still unknown, our results suggest that it does not contribute to covalent crosslinking in the mucus, nor incorporate immunoglobulin G into mucus, instead the single disulfide bond linking each fragment could mediate controlled dissociation.
Collapse
|
17
|
Robinson RA, Griffiths SC, van de Haar LL, Malinauskas T, van Battum EY, Zelina P, Schwab RA, Karia D, Malinauskaite L, Brignani S, van den Munkhof MH, Düdükcü Ö, De Ruiter AA, Van den Heuvel DMA, Bishop B, Elegheert J, Aricescu AR, Pasterkamp RJ, Siebold C. Simultaneous binding of Guidance Cues NET1 and RGM blocks extracellular NEO1 signaling. Cell 2021; 184:2103-2120.e31. [PMID: 33740419 PMCID: PMC8063088 DOI: 10.1016/j.cell.2021.02.045] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/13/2020] [Revised: 01/15/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022]
Abstract
During cell migration or differentiation, cell surface receptors are simultaneously exposed to different ligands. However, it is often unclear how these extracellular signals are integrated. Neogenin (NEO1) acts as an attractive guidance receptor when the Netrin-1 (NET1) ligand binds, but it mediates repulsion via repulsive guidance molecule (RGM) ligands. Here, we show that signal integration occurs through the formation of a ternary NEO1-NET1-RGM complex, which triggers reciprocal silencing of downstream signaling. Our NEO1-NET1-RGM structures reveal a "trimer-of-trimers" super-assembly, which exists in the cell membrane. Super-assembly formation results in inhibition of RGMA-NEO1-mediated growth cone collapse and RGMA- or NET1-NEO1-mediated neuron migration, by preventing formation of signaling-compatible RGM-NEO1 complexes and NET1-induced NEO1 ectodomain clustering. These results illustrate how simultaneous binding of ligands with opposing functions, to a single receptor, does not lead to competition for binding, but to formation of a super-complex that diminishes their functional outputs.
Collapse
Affiliation(s)
- Ross A Robinson
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Samuel C Griffiths
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lieke L van de Haar
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Eljo Y van Battum
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Pavol Zelina
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Rebekka A Schwab
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Dimple Karia
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Lina Malinauskaite
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Sara Brignani
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Marleen H van den Munkhof
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Özge Düdükcü
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Anna A De Ruiter
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Dianne M A Van den Heuvel
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Jonathan Elegheert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK; MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center, Utrecht University, Universiteitsweg 100, 3584 CG Utrecht, the Netherlands.
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK.
| |
Collapse
|
18
|
Hepatocyte neogenin, another key actor in iron homeostasis. Blood 2021; 138:423-425. [PMID: 34383040 DOI: 10.1182/blood.2021011936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 04/12/2021] [Indexed: 11/20/2022] Open
|
19
|
Gipson GR, Goebel EJ, Hart KN, Kappes EC, Kattamuri C, McCoy JC, Thompson TB. Structural perspective of BMP ligands and signaling. Bone 2020; 140:115549. [PMID: 32730927 PMCID: PMC7502536 DOI: 10.1016/j.bone.2020.115549] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/17/2020] [Revised: 06/30/2020] [Accepted: 07/03/2020] [Indexed: 12/13/2022]
Abstract
The Bone Morphogenetic Proteins (BMPs) are the largest class signaling molecules within the greater Transforming Growth Factor Beta (TGFβ) family, and are responsible for a wide array of biological functions, including dorsal-ventral patterning, skeletal development and maintenance, as well as cell homeostasis. As such, dysregulation of BMPs results in a number of diseases, including fibrodysplasia ossificans progressiva (FOP) and pulmonary arterial hypertension (PAH). Therefore, understanding BMP signaling and regulation at the molecular level is essential for targeted therapeutic intervention. This review discusses the recent advances in the structural and biochemical characterization of BMPs, from canonical ligand-receptor interactions to co-receptors and antagonists. This work aims to highlight how BMPs differ from other members of the TGFβ family, and how that information can be used to further advance the field. Lastly, this review discusses several gaps in the current understanding of BMP structures, with the aim that discussion of these gaps will lead to advancements in the field.
Collapse
Affiliation(s)
- Gregory R Gipson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Erich J Goebel
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Kaitlin N Hart
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Emily C Kappes
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Chandramohan Kattamuri
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Jason C McCoy
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA
| | - Thomas B Thompson
- Department of Molecular Genetics, Biochemistry, and Microbiology, University of Cincinnati, Medical Sciences Building, Cincinnati, OH 45267, USA.
| |
Collapse
|
20
|
Malinauskas T, Peer TV, Bishop B, Mueller TD, Siebold C. Repulsive guidance molecules lock growth differentiation factor 5 in an inhibitory complex. Proc Natl Acad Sci U S A 2020; 117:15620-15631. [PMID: 32576689 PMCID: PMC7354924 DOI: 10.1073/pnas.2000561117] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/05/2023] Open
Abstract
Repulsive guidance molecules (RGMs) are cell surface proteins that regulate the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. They control fundamental biological processes, such as migration and differentiation by direct interaction with the Neogenin (NEO1) receptor and function as coreceptors for the bone morphogenetic protein (BMP)/growth differentiation factor (GDF) family. We determined crystal structures of all three human RGM family members in complex with GDF5, as well as the ternary NEO1-RGMB-GDF5 assembly. Surprisingly, we show that all three RGMs inhibit GDF5 signaling, which is in stark contrast to RGM-mediated enhancement of signaling observed for other BMPs, like BMP2. Despite their opposite effect on GDF5 signaling, RGMs occupy the BMP type 1 receptor binding site similar to the observed interactions in RGM-BMP2 complexes. In the NEO1-RGMB-GDF5 complex, RGMB physically bridges NEO1 and GDF5, suggesting cross-talk between the GDF5 and NEO1 signaling pathways. Our crystal structures, combined with structure-guided mutagenesis of RGMs and BMP ligands, binding studies, and cellular assays suggest that RGMs inhibit GDF5 signaling by competing with GDF5 type 1 receptors. While our crystal structure analysis and in vitro binding data initially pointed towards a simple competition mechanism between RGMs and type 1 receptors as a possible basis for RGM-mediated GDF5 inhibition, further experiments utilizing BMP2-mimicking GDF5 variants clearly indicate a more complex mechanism that explains how RGMs can act as a functionality-changing switch for two structurally and biochemically similar signaling molecules.
Collapse
Affiliation(s)
- Tomas Malinauskas
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN Oxford, United Kingdom;
| | - Tina V Peer
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute, University of Würzburg, 97082 Würzburg, Germany
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN Oxford, United Kingdom
| | - Thomas D Mueller
- Department of Molecular Plant Physiology and Biophysics, Julius-von-Sachs Institute, University of Würzburg, 97082 Würzburg, Germany
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN Oxford, United Kingdom;
| |
Collapse
|
21
|
Isaksen TJ, Fujita Y, Yamashita T. Repulsive Guidance Molecule A Suppresses Adult Neurogenesis. Stem Cell Reports 2020; 14:677-691. [PMID: 32243839 PMCID: PMC7160374 DOI: 10.1016/j.stemcr.2020.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2019] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 01/17/2023] Open
Abstract
Repulsive guidance molecule A (RGMa) is a glycosylphosphatidylinositol-anchored glycoprotein that exhibits repulsive neurite guidance and regulates neuronal differentiation and survival during brain development. However, the function of RGMa in the adult brain is unknown. Here, we show that RGMa is expressed in the adult hippocampus and provide evidence that RGMa signaling suppresses adult neurogenesis. Knockdown of RGMa in the dentate gyrus increased the number of surviving newborn neurons; however, these cells failed to properly migrate into the granular cell layer. In vitro, RGMa stimulation of adult neural stem cells suppressed neurite outgrowth of newborn neurons, which could be prevented by knockdown of the multifunctional receptor neogenin, as well as pharmacological inhibition of the downstream target Rho-associated protein kinase. These findings present a function for RGMa in the adult brain and add to the intricate molecular network that regulates adult brain plasticity. RGMa suppress survival and growth of newborn neurons in the adult dentate gyrus RGMa signaling depends on neogenin for the regulation of adult neurogenesis RGMa induces RhoA/ROCK activation in adult neuronal stem cells
Collapse
Affiliation(s)
- Toke Jost Isaksen
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; WPI Immunology Frontier Research Center, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan; Graduate School of Frontier Bioscience, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan.
| |
Collapse
|
22
|
Huyghe A, Furlan G, Ozmadenci D, Galonska C, Charlton J, Gaume X, Combémorel N, Riemenschneider C, Allègre N, Zhang J, Wajda P, Rama N, Vieugué P, Durand I, Brevet M, Gadot N, Imhof T, Merrill BJ, Koch M, Mehlen P, Chazaud C, Meissner A, Lavial F. Netrin-1 promotes naive pluripotency through Neo1 and Unc5b co-regulation of Wnt and MAPK signalling. Nat Cell Biol 2020; 22:389-400. [PMID: 32231305 DOI: 10.1038/s41556-020-0483-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/03/2017] [Accepted: 02/13/2020] [Indexed: 12/20/2022]
Abstract
In mouse embryonic stem cells (mESCs), chemical blockade of Gsk3α/β and Mek1/2 (2i) instructs a self-renewing ground state whose endogenous inducers are unknown. Here we show that the axon guidance cue Netrin-1 promotes naive pluripotency by triggering profound signalling, transcriptomic and epigenetic changes in mESCs. Furthermore, we demonstrate that Netrin-1 can substitute for blockade of Gsk3α/β and Mek1/2 to sustain self-renewal of mESCs in combination with leukaemia inhibitory factor and regulates the formation of the mouse pluripotent blastocyst. Mechanistically, we reveal how Netrin-1 and the balance of its receptors Neo1 and Unc5B co-regulate Wnt and MAPK pathways in both mouse and human ESCs. Netrin-1 induces Fak kinase to inactivate Gsk3α/β and stabilize β-catenin while increasing the phosphatase activity of a Ppp2r2c-containing Pp2a complex to reduce Erk1/2 activity. Collectively, this work identifies Netrin-1 as a regulator of pluripotency and reveals that it mediates different effects in mESCs depending on its receptor dosage, opening perspectives for balancing self-renewal and lineage commitment.
Collapse
Affiliation(s)
- Aurélia Huyghe
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Giacomo Furlan
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Duygu Ozmadenci
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Christina Galonska
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Jocelyn Charlton
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Xavier Gaume
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Noémie Combémorel
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | | | - Nicolas Allègre
- GReD, Université Clermont Auvergne, CNRS, INSERM, BP38, Clermont-Ferrand, France
| | - Jenny Zhang
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Pauline Wajda
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Nicolas Rama
- Apoptosis, Cancer and Development Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Pauline Vieugué
- Apoptosis, Cancer and Development Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France
| | - Isabelle Durand
- Cytometry Facility, Université de Lyon, Université Claude Bernard Lyon 1, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, INSERM 1052, CNRS 5286, Lyon, France
| | - Marie Brevet
- Research Pathology platform, Department of translational research and innovation, Centre Léon Bérard, Lyon, France
| | - Nicolas Gadot
- Research Pathology platform, Department of translational research and innovation, Centre Léon Bérard, Lyon, France
| | - Thomas Imhof
- Institute for Dental Research and Oral Musculoskeletal Research, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Bradley J Merrill
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Manuel Koch
- Institute for Dental Research and Oral Musculoskeletal Research, Center for Biochemistry, University of Cologne, Cologne, Germany
| | - Patrick Mehlen
- Apoptosis, Cancer and Development Laboratory, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France.,Department of Translational Research and Innovation, Centre Léon Bérard, Lyon, France
| | - Claire Chazaud
- GReD, Université Clermont Auvergne, CNRS, INSERM, BP38, Clermont-Ferrand, France
| | - Alexander Meissner
- Department of Genome Regulation, Max Planck Institute for Molecular Genetics, Berlin, Germany.,Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Harvard Stem Cell Institute, Cambridge, MA, USA.,Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Fabrice Lavial
- Cellular Reprogramming and Oncogenesis Laboratory, Equipe labellisée la Ligue contre le cancer, Labex DEVweCAN, Université de Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon, France.
| |
Collapse
|
23
|
Yu S, Leung KM, Kim HY, Umetsu SE, Xiao Y, Albacker LA, Lee HJ, Umetsu DT, Freeman GJ, DeKruyff RH. Blockade of RGMb inhibits allergen-induced airways disease. J Allergy Clin Immunol 2019; 144:94-108.e11. [PMID: 30703386 PMCID: PMC8088837 DOI: 10.1016/j.jaci.2018.12.1022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/13/2017] [Revised: 11/20/2018] [Accepted: 12/07/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Allergic asthma causes morbidity in many subjects, and novel precision-directed treatments would be valuable. OBJECTIVE We sought to examine the role of a novel innate molecule, repulsive guidance molecule b (RGMb), in murine models of allergic asthma. METHODS In models of allergic asthma using ovalbumin or cockroach allergen, mice were treated with anti-RGMb or control mAb and examined for airway inflammation and airway hyperreactivity (AHR), a cardinal feature of asthma. The mechanisms by which RGMb causes airways disease were also examined. RESULTS We found that blockade of RGMb by treatment with anti-RGMb mAb effectively blocked the development of airway inflammation and AHR. Importantly, blockade of RGMb completely blocked the development of airway inflammation and AHR, even if treatment occurred only during the challenge (effector) phase. IL-25 played an important role in these models of asthma because IL-25 receptor-deficient mice did not develop disease after sensitization and challenge with allergen. RGMb was expressed primarily by innate cells in the lungs, including bronchial epithelial cells (known producers of IL-25), activated eosinophils, and interstitial macrophages, which in the inflamed lung expressed the IL-25 receptor and produced IL-5 and IL-13. We also found that neogenin, the canonical receptor for RGMb, was expressed by interstitial macrophages and bronchial epithelial cells in the inflamed lung, suggesting that an innate RGMb-neogenin axis might modulate allergic asthma. CONCLUSIONS These results demonstrate an important role for a novel innate pathway in regulating type 2 inflammation in patients with allergic asthma involving RGMb and RGMb-expressing cells, such as interstitial macrophages and bronchial epithelial cells. Moreover, targeting this previously unappreciated innate pathway might provide an important treatment option for allergic asthma.
Collapse
Affiliation(s)
- Sanhong Yu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Mass; Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Krystle M Leung
- Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Hye-Young Kim
- Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Sarah E Umetsu
- Department of Pathology, University of California, San Francisco, Calif
| | - Yanping Xiao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Mass
| | - Lee A Albacker
- Boston Children's Hospital, Harvard Medical School, Boston, Mass; Immunology Program, Harvard Medical School, Boston, Mass
| | - Hyun-Jun Lee
- Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Dale T Umetsu
- Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Department of Medicine, Harvard Medical School, Boston, Mass
| | - Rosemarie H DeKruyff
- Boston Children's Hospital, Harvard Medical School, Boston, Mass; Sean N Parker Center for Allergy and Asthma Research, Department of Medicine, Stanford University, Stanford, Calif.
| |
Collapse
|
24
|
Sekiya T, Takaki S. RGMB enhances the suppressive activity of the monomeric secreted form of CTLA-4. Sci Rep 2019; 9:6984. [PMID: 31061392 PMCID: PMC6502797 DOI: 10.1038/s41598-019-43068-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/19/2018] [Accepted: 03/15/2019] [Indexed: 12/17/2022] Open
Abstract
The immunoregulatory molecule CTLA-4 plays a crucial role in the maintenance of immune homeostasis. CTLA-4-neutralizing antibodies are now approved for the treatment of advanced melanoma, and are in development for treating other cancers as well. However, a thorough understanding of CTLA-4 function at the molecular level is necessary in order to develop strategies to prevent the unintended autoimmunity that is frequently associated with systemic blockade of CTLA-4 activity. Here, we describe an extracellular molecule, repulsive guidance molecule B (RGMB) as a novel binding partner of CTLA-4. RGMB expression was detected at high levels in dendritic cell subsets that have been suggested to have tolerogenic capabilities. RGMB binds an extracellular domain of CTLA-4, and specifically strengthens the binding of the monomeric, soluble form of CTLA-4 (sCTLA-4) to CD80, enhancing CTLA-4's suppressive effect on co-stimulation. Examination of expression data from tumor tissues revealed a negative correlation between RGMB expression and immune activation status in the majority of non-hematologic tumor tissues. These findings advance our understanding of CTLA-4 activity, as well as identify the RGMB/CTLA-4 binding interface as a potential target for the development of novel immune checkpoint blockade therapies.
Collapse
Affiliation(s)
- Takashi Sekiya
- Section of Immune Response Modification, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan.
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan.
| | - Satoshi Takaki
- Department of Immune Regulation, The Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, 1-7-1 Kohnodai, Ichikawa, Chiba, 272-8516, Japan
| |
Collapse
|
25
|
Dong X, Leksa NC, Chhabra ES, Arndt JW, Lu Q, Knockenhauer KE, Peters RT, Springer TA. The von Willebrand factor D'D3 assembly and structural principles for factor VIII binding and concatemer biogenesis. Blood 2019; 133:1523-1533. [PMID: 30642920 PMCID: PMC6450429 DOI: 10.1182/blood-2018-10-876300] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/10/2018] [Accepted: 12/28/2018] [Indexed: 12/21/2022] Open
Abstract
D assemblies make up half of the von Willebrand factor (VWF), yet are of unknown structure. D1 and D2 in the prodomain and D'D3 in mature VWF at Golgi pH form helical VWF tubules in Weibel Palade bodies and template dimerization of D3 through disulfides to form ultralong VWF concatemers. D'D3 forms the binding site for factor VIII. The crystal structure of monomeric D'D3 with cysteine residues required for dimerization mutated to alanine was determined at an endoplasmic reticulum (ER)-like pH. The smaller C8-3, TIL3 (trypsin inhibitor-like 3), and E3 modules pack through specific interfaces as they wind around the larger, N-terminal, Ca2+-binding von Willebrand D domain (VWD) 3 module to form a wedge shape. D' with its TIL' and E' modules projects away from D3. The 2 mutated cysteines implicated in D3 dimerization are buried, providing a mechanism for protecting them against premature disulfide linkage in the ER, where intrachain disulfide linkages are formed. D3 dimerization requires co-association with D1 and D2, Ca2+, and Golgi-like acidic pH. Associated structural rearrangements in the C8-3 and TIL3 modules are required to expose cysteine residues for disulfide linkage. Our structure provides insight into many von Willebrand disease mutations, including those that diminish factor VIII binding, which suggest that factor VIII binds not only to the N-terminal TIL' domain of D' distal from D3 but also extends across 1 side of D3. The organizing principle for the D3 assembly has implications for other D assemblies and the construction of higher-order, disulfide-linked assemblies in the Golgi in both VWF and mucins.
Collapse
Affiliation(s)
- Xianchi Dong
- Children's Hospital Boston, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| | | | | | | | - Qi Lu
- Bioverativ, a Sanofi company, Waltham, MA; and
| | | | | | - Timothy A Springer
- Children's Hospital Boston, Boston, MA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA
| |
Collapse
|
26
|
Lockhart-Cairns MP, Lim KTW, Zuk A, Godwin ARF, Cain SA, Sengle G, Baldock C. Internal cleavage and synergy with twisted gastrulation enhance BMP inhibition by BMPER. Matrix Biol 2019; 77:73-86. [PMID: 30125619 PMCID: PMC6456722 DOI: 10.1016/j.matbio.2018.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/23/2018] [Revised: 08/13/2018] [Accepted: 08/16/2018] [Indexed: 12/13/2022]
Abstract
Bone morphogenetic proteins (BMPs) are essential signalling molecules involved in developmental and pathological processes and are regulated in the matrix by secreted glycoproteins. One such regulator is BMP-binding endothelial cell precursor-derived regulator (BMPER) which can both inhibit and enhance BMP signalling in a context and concentration-dependent manner. Twisted gastrulation (Tsg) can also promote or ablate BMP activity but it is unclear whether Tsg and BMPER directly interact and thereby exert a synergistic function on BMP signalling. Here, we show that human BMPER binds to Tsg through the N-terminal BMP-binding region which alone more potently inhibits BMP-4 signalling than full-length BMPER. Additionally, BMPER and Tsg cooperatively inhibit BMP-4 signalling suggesting a synergistic function to dampen BMP activity. Furthermore, full-length BMPER is targeted to the plasma membrane via binding of its C-terminal region to cell surface heparan sulphate proteoglycans but the active cleavage fragment is diffusible. Small-angle X-ray scattering and electron microscopy show that BMPER has an elongated conformation allowing the N-terminal BMP-binding and C-terminal cell-interactive regions to be spatially separated. To gain insight into the regulation of BMPER bioavailability by internal cleavage, a disease-causing BMPER point mutation, P370L, previously identified in the acid-catalysed cleavage site, was introduced. The mutated protein was secreted but the mutation prevented intracellular cleavage resulting in a lack of bioactive cleavage fragment. Furthermore, mutant BMPER was extracellularly cleaved at a downstream site presumably becoming available due to the mutation. This susceptibility to extracellular proteases and loss of bioactive N-terminal cleavage fragment may result in loss of BMPER function in disease.
Collapse
Affiliation(s)
- Michael P Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, UK; Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - Karen Tzia Wei Lim
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Alexandra Zuk
- Center for Biochemistry, Medical Faculty, University of Cologne, Germany
| | - Alan R F Godwin
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Stuart A Cain
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, UK
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Germany; Center for Molecular Medicine, University of Cologne, Germany
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, UK.
| |
Collapse
|
27
|
Ren T, Zheng B, Huang Y, Wang S, Bao X, Liu K, Guo W. Osteosarcoma cell intrinsic PD-L2 signals promote invasion and metastasis via the RhoA-ROCK-LIMK2 and autophagy pathways. Cell Death Dis 2019; 10:261. [PMID: 30886151 PMCID: PMC6423010 DOI: 10.1038/s41419-019-1497-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/21/2018] [Revised: 01/29/2019] [Accepted: 03/04/2019] [Indexed: 12/15/2022]
Abstract
Known as co-stimulatory molecule, programmed death ligand-2 (PD-L2) contributes to T-cell exhaustion by interaction with programmed death-1 (PD-1) receptor, but its tumor cell-intrinsic signal effects have been little investigated. PD-L2 expression was detected by immunohistochemistry in 18 pairs of primary osteosarcoma tissues and matching lung metastasis tissues. We also investigated the effects of PD-L2 knockdown on osteosarcoma both in vitro and in vivo. In our study, PD-L2 expression was elevated in lung metastases compared with primary osteosarcoma according to an immunohistochemistry assay. Wound-healing and transwell assays revealed that PD-L2 knockdown leaded to inhibition of migration and invasion of human osteosarcoma cells in vitro. Mechanistically, we demonstrated that PD-L2 knockdown attenuated migration and invasion by inactivating RhoA-ROCK-LIMK2 signaling, suppressing epithelial–mesenchymal transition (EMT), and inhibiting autophagy by decreasing beclin-1 expression. In support of these observations, beclin-1 knockdown also inhibited activation of the RhoA-ROCK-LIMK2 pathway, leading to autophagy inhibition-induced blockade of migration and invasion. Depletion of PD-L2 in KHOS cells markedly weakens pulmonary metastatic potential in vivo by orthotopic transplantation of nude mice. Our study reveals a pro-metastatic functional mechanism for PD-L2 in osteosarcoma. Furthermore, we demonstrate a regulatory role for PD-L2 on autophagy, as well as a relationship between autophagy and metastasis in osteosarcoma, which may represent a potential therapeutic target for osteosarcoma.
Collapse
Affiliation(s)
- Tingting Ren
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Bingxin Zheng
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.,Department of Orthopedic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, People's Republic of China
| | - Yi Huang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Shidong Wang
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Xing Bao
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Kuisheng Liu
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China
| | - Wei Guo
- Musculoskeletal Tumor Center, Peking University People's Hospital, Beijing, People's Republic of China. .,Beijing Key Laboratory of Musculoskeletal Tumor, Beijing, People's Republic of China.
| |
Collapse
|
28
|
Branching mechanisms shaping dendrite architecture. Dev Biol 2018; 451:16-24. [PMID: 30550882 DOI: 10.1016/j.ydbio.2018.12.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/08/2018] [Revised: 11/19/2018] [Accepted: 12/05/2018] [Indexed: 12/11/2022]
Abstract
A neuron's contribution to the information flow within a neural circuit is governed by the structure of its dendritic arbor. The geometry of the dendritic arbor directly determines synaptic density and the size of the receptive field, both of which influence the firing pattern of the neuron. Importantly, the position of individual dendritic branches determines the identity of the neuron's presynaptic partner and thus the nature of the incoming sensory information. To generate the unique stereotypic architecture of a given neuronal subtype, nascent branches must emerge from the dendritic shaft at preprogramed branch points. Subsequently, a complex array of extrinsic factors regulates the degree and orientation of branch expansion to ensure maximum coverage of the receptive field whilst constraining growth within predetermined territories. In this review we focus on studies that best illustrate how environmental cues such as the Wnts and Netrins and their receptors sculpt the dendritic arbor. We emphasize the pivotal role played by the actin cytoskeleton and its upstream regulators in branch initiation, outgrowth and navigation. Finally, we discuss how protocadherin and DSCAM contact-mediated repulsion prevents inappropriate synapse formation between sister dendrites or dendrites and the axon from the same neuron. Together these studies highlight the clever ways evolution has solved the problem of constructing complex branch geometries.
Collapse
|
29
|
Elegheert J, Behiels E, Bishop B, Scott S, Woolley RE, Griffiths SC, Byrne EFX, Chang VT, Stuart DI, Jones EY, Siebold C, Aricescu AR. Lentiviral transduction of mammalian cells for fast, scalable and high-level production of soluble and membrane proteins. Nat Protoc 2018; 13:2991-3017. [PMID: 30455477 PMCID: PMC6364805 DOI: 10.1038/s41596-018-0075-9] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/01/2023]
Abstract
Structural, biochemical and biophysical studies of eukaryotic soluble and membrane proteins require their production in milligram quantities. Although large-scale protein expression strategies based on transient or stable transfection of mammalian cells are well established, they are associated with high consumable costs, limited transfection efficiency or long and tedious selection of clonal cell lines. Lentiviral transduction is an efficient method for the delivery of transgenes to mammalian cells and unifies the ease of use and speed of transient transfection with the robust expression of stable cell lines. In this protocol, we describe the design and step-by-step application of a lentiviral plasmid suite, termed pHR-CMV-TetO2, for the constitutive or inducible large-scale production of soluble and membrane proteins in HEK293 cell lines. Optional features include bicistronic co-expression of fluorescent marker proteins for enrichment of co-transduced cells using cell sorting and of biotin ligase for in vivo biotinylation. We demonstrate the efficacy of the method for a set of soluble proteins and for the G-protein-coupled receptor (GPCR) Smoothened (SMO). We further compare this method with baculovirus transduction of mammalian cells (BacMam), using the type-A γ-aminobutyric acid receptor (GABAAR) β3 homopentamer as a test case. The protocols described here are optimized for simplicity, speed and affordability; lead to a stable polyclonal cell line and milligram-scale amounts of protein in 3-4 weeks; and routinely achieve an approximately three- to tenfold improvement in protein production yield per cell as compared to transient transduction or transfection.
Collapse
Affiliation(s)
- Jonathan Elegheert
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France.
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France.
| | - Ester Behiels
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Interdisciplinary Institute for Neuroscience, University of Bordeaux, Bordeaux, France
- Interdisciplinary Institute for Neuroscience, CNRS UMR 5297, Bordeaux, France
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Suzanne Scott
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Rachel E Woolley
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Samuel C Griffiths
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Eamon F X Byrne
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Veronica T Chang
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - David I Stuart
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - E Yvonne Jones
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| | - A Radu Aricescu
- Division of Structural Biology, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
30
|
RGMb protects against acute kidney injury by inhibiting tubular cell necroptosis via an MLKL-dependent mechanism. Proc Natl Acad Sci U S A 2018; 115:E1475-E1484. [PMID: 29382757 DOI: 10.1073/pnas.1716959115] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022] Open
Abstract
Tubular cell necrosis is a key histological feature of acute kidney injury (AKI). Necroptosis is a type of programed necrosis, which is executed by mixed lineage kinase domain-like protein (MLKL) upon its binding to the plasma membrane. Emerging evidence indicates that necroptosis plays a critical role in the development of AKI. However, it is unclear whether renal tubular cells undergo necroptosis in vivo and how the necroptotic pathway is regulated during AKI. Repulsive guidance molecule (RGM)-b is a member of the RGM family. Our previous study demonstrated that RGMb is highly expressed in kidney tubular epithelial cells, but its biological role in the kidney has not been well characterized. In the present study, we found that RGMb reduced membrane-associated MLKL levels and inhibited necroptosis in cultured cells. During ischemia/reperfusion injury (IRI) or oxalate nephropathy, MLKL was induced to express on the apical membrane of proximal tubular (PT) cells. Specific knockout of Rgmb in tubular cells (Rgmb cKO) increased MLKL expression at the apical membrane of PT cells and induced more tubular cell death and more severe renal dysfunction compared with wild-type mice. Treatment with the necroptosis inhibitor Necrostatin-1 or GSK'963 reduced MLKL expression on the apical membrane of PT cells and ameliorated renal function impairment after IRI in both wild-type and Rgmb cKO mice. Taken together, our results suggest that proximal tubular cell necroptosis plays an important role in AKI, and that RGMb protects against AKI by inhibiting MLKL membrane association and necroptosis in proximal tubular cells.
Collapse
|
31
|
Nickel J, Ten Dijke P, Mueller TD. TGF-β family co-receptor function and signaling. Acta Biochim Biophys Sin (Shanghai) 2018; 50:12-36. [PMID: 29293886 DOI: 10.1093/abbs/gmx126] [Citation(s) in RCA: 131] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2017] [Accepted: 11/08/2017] [Indexed: 01/04/2023] Open
Abstract
Transforming growth factor-β (TGF-β) family members, which include TGF-βs, activins and bone morphogenetic proteins, are pleiotropic cytokines that elicit cell type-specific effects in a highly context-dependent manner in many different tissues. These secreted protein ligands signal via single-transmembrane Type I and Type II serine/threonine kinase receptors and intracellular SMAD transcription factors. Deregulation in signaling has been implicated in a broad array of diseases, and implicate the need for intricate fine tuning in cellular signaling responses. One important emerging mechanism by which TGF-β family receptor signaling intensity, duration, specificity and diversity are regulated and/or mediated is through cell surface co-receptors. Here, we provide an overview of the co-receptors that have been identified for TGF-β family members. While some appear to be specific to TGF-β family members, others are shared with other pathways and provide possible ways for signal integration. This review focuses on novel functions of TGF-β family co-receptors, which continue to be discovered.
Collapse
Affiliation(s)
- Joachim Nickel
- Universitätsklinikum Würzburg, Lehrstuhl für Tissue Engineering und Regenerative Medizin und Fraunhofer Institut für Silicatforschung (ISC), Translationszentrum "Regenerative Therapien", Röntgenring 11, D-97070 Würzburg, Germany
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology and Cancer Genomics Centre Netherlands, Leiden University Medical Center, Einthovenweg 20, 2300 RC Leiden, The Netherlands
| | - Thomas D Mueller
- Lehrstuhl für molekulare Pflanzenphysiologie und Biophysik, Julius-von-Sachs Institut für Biowissenschaften, Universität Würzburg, Julius-von-Sachs-Platz 2, D-97082 Würzburg, Germany
| |
Collapse
|
32
|
Zabkiewicz C, Resaul J, Hargest R, Jiang WG, Ye L. Bone morphogenetic proteins, breast cancer, and bone metastases: striking the right balance. Endocr Relat Cancer 2017; 24:R349-R366. [PMID: 28733469 PMCID: PMC5574206 DOI: 10.1530/erc-17-0139] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 06/29/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β super family, and are essential for the regulation of foetal development, tissue differentiation and homeostasis and a multitude of cellular functions. Naturally, this has led to the exploration of aberrance in this highly regulated system as a key factor in tumourigenesis. Originally identified for their role in osteogenesis and bone turnover, attention has been turned to the potential role of BMPs in tumour metastases to, and progression within, the bone niche. This is particularly pertinent to breast cancer, which commonly metastasises to bone, and in which studies have revealed aberrations of both BMP expression and signalling, which correlate clinically with breast cancer progression. Ultimately a BMP profile could provide new prognostic disease markers. As the evidence suggests a role for BMPs in regulating breast tumour cellular function, in particular interactions with tumour stroma and the bone metastatic microenvironment, there may be novel therapeutic potential in targeting BMP signalling in breast cancer. This review provides an update on the current knowledge of BMP abnormalities and their implication in the development and progression of breast cancer, particularly in the disease-specific bone metastasis.
Collapse
Affiliation(s)
- Catherine Zabkiewicz
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Jeyna Resaul
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Rachel Hargest
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Wen Guo Jiang
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Lin Ye
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
33
|
Abstract
Iron fulfils a central role in many essential biochemical processes in human physiology; thus, proper processing of iron is crucial. Although iron metabolism is subject to relatively strict physiological control, numerous disorders, such as cancer and neurodegenerative diseases, have recently been linked to deregulated iron homeostasis. Consequently, iron metabolism constitutes a promising and largely unexploited therapeutic target for the development of new pharmacological treatments for these diseases. Several iron metabolism-targeted therapies are already under clinical evaluation for haematological disorders, and these and newly developed therapeutic agents are likely to have substantial benefit in the clinical management of iron metabolism-associated diseases, for which few efficacious treatments are currently available.
Collapse
Affiliation(s)
- Bart J. Crielaard
- Department of Polymer Chemistry and Bioengineering, Zernike Institute for Advanced Materials, Faculty of Mathematics and Natural Sciences, University of Groningen, Groningen, The Netherlands
- W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, Groningen, The Netherlands
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, University Clinic and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen, Germany
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
- Department of Targeted Therapeutics, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands
| | - Stefano Rivella
- Children’s Hospital of Philadelphia, Abramson Research Center, Philadelphia, PA, United States of America
| |
Collapse
|
34
|
B7-DC (PD-L2) costimulation of CD4 + T-helper 1 response via RGMb. Cell Mol Immunol 2017; 15:888-897. [PMID: 28479601 DOI: 10.1038/cmi.2017.17] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 02/08/2017] [Indexed: 12/20/2022] Open
Abstract
The role of B7-DC in T-cell responses remains controversial because both coinhibitory and costimulatory functions have been reported in various experimental systems in vitro and in vivo. In addition to interacting with the coinhibitory receptor PD-1, B7-DC has also been shown to bind repulsive guidance molecule b (RGMb). The functional consequences of the B7-DC/RGMb interaction, however, remain unclear. More than a decade ago, we reported that replacement of a murine B7-DC mutant lysine with serine (K113S) at positive 113 resulted in a loss of binding capacity to PD-1. Nevertheless, K113S remained costimulatory for T cells in vitro, implicating a dual functionality for B7-DC in T-cell responses. Here we show that recombinant K113S protein interacts with RGMb with a similar affinity to wild-type B7-DC. More importantly, K113S costimulates CD4+ T-cell responses via RGMb and promotes Th1 polarization. RGMb is expressed on the surface of naive mouse T cells, macrophages, neutrophils and dendritic cells. Finally, K113S/RGMb costimulation suppresses Th2-mediated asthma and ameliorates small airway inflammation and lung pathology in an experimental mouse model. Our findings indicate that RGMb is a costimulatory receptor for B7-DC. These findings from the K113S variant provide not only a possible explanation for the B7-DC-triggered contradictory effects on T-cell responses, but also a novel approach to investigate the B7-DC/PD-1/RGMb axis. Recombinant K113S or its derivatives could potentially be developed as an agonist for RGMb to costimulate the Th1 response without triggering PD-1-mediated T-cell inhibition.
Collapse
|
35
|
Siebold C, Yamashita T, Monnier PP, Mueller BK, Pasterkamp RJ. RGMs: Structural Insights, Molecular Regulation, and Downstream Signaling. Trends Cell Biol 2017; 27:365-378. [PMID: 28007423 PMCID: PMC5404723 DOI: 10.1016/j.tcb.2016.11.009] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/10/2016] [Revised: 11/27/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022]
Abstract
Although originally discovered as neuronal growth cone-collapsing factors, repulsive guidance molecules (RGMs) are now known as key players in many fundamental processes, such as cell migration, differentiation, iron homeostasis, and apoptosis, during the development and homeostasis of many tissues and organs, including the nervous, skeletal, and immune systems. Furthermore, three RGMs (RGMa, RGMb/DRAGON, and RGMc/hemojuvelin) have been linked to the pathogenesis of various disorders ranging from multiple sclerosis (MS) to cancer and juvenile hemochromatosis (JHH). While the molecular details of these (patho)biological effects and signaling modes have long remained unknown, recent studies unveil several exciting and novel aspects of RGM processing, ligand-receptor interactions, and downstream signaling. In this review, we highlight recent advances in the mechanisms-of-action and function of RGM proteins.
Collapse
Affiliation(s)
- Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, 2-2, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Philippe P Monnier
- Krembil Research Institute, 60 Leonard Street, M5T 2S8, Toronto, ONT, Canada
| | - Bernhard K Mueller
- Neuroscience Discovery Research, Abbvie, Knollstrasse 50, 67061 Ludwigshafen, Germany
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Mercati O, Huguet G, Danckaert A, André-Leroux G, Maruani A, Bellinzoni M, Rolland T, Gouder L, Mathieu A, Buratti J, Amsellem F, Benabou M, Van-Gils J, Beggiato A, Konyukh M, Bourgeois JP, Gazzellone MJ, Yuen RKC, Walker S, Delépine M, Boland A, Régnault B, Francois M, Van Den Abbeele T, Mosca-Boidron AL, Faivre L, Shimoda Y, Watanabe K, Bonneau D, Rastam M, Leboyer M, Scherer SW, Gillberg C, Delorme R, Cloëz-Tayarani I, Bourgeron T. CNTN6 mutations are risk factors for abnormal auditory sensory perception in autism spectrum disorders. Mol Psychiatry 2017; 22:625-633. [PMID: 27166760 PMCID: PMC5378808 DOI: 10.1038/mp.2016.61] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/18/2015] [Revised: 02/12/2016] [Accepted: 02/17/2016] [Indexed: 12/11/2022]
Abstract
Contactin genes CNTN5 and CNTN6 code for neuronal cell adhesion molecules that promote neurite outgrowth in sensory-motor neuronal pathways. Mutations of CNTN5 and CNTN6 have previously been reported in individuals with autism spectrum disorders (ASDs), but very little is known on their prevalence and clinical impact. In this study, we identified CNTN5 and CNTN6 deleterious variants in individuals with ASD. Among the carriers, a girl with ASD and attention-deficit/hyperactivity disorder was carrying five copies of CNTN5. For CNTN6, both deletions (6/1534 ASD vs 1/8936 controls; P=0.00006) and private coding sequence variants (18/501 ASD vs 535/33480 controls; P=0.0005) were enriched in individuals with ASD. Among the rare CNTN6 variants, two deletions were transmitted by fathers diagnosed with ASD, one stop mutation CNTN6W923X was transmitted by a mother to her two sons with ASD and one variant CNTN6P770L was found de novo in a boy with ASD. Clinical investigations of the patients carrying CNTN5 or CNTN6 variants showed that they were hypersensitive to sounds (a condition called hyperacusis) and displayed changes in wave latency within the auditory pathway. These results reinforce the hypothesis of abnormal neuronal connectivity in the pathophysiology of ASD and shed new light on the genes that increase risk for abnormal sensory perception in ASD.
Collapse
Affiliation(s)
- O Mercati
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - G Huguet
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - A Danckaert
- Imagopole, Citech, Institut Pasteur, Paris, France
| | - G André-Leroux
- Institut Pasteur, Unité de Microbiologie Structurale, Paris, France
- CNRS UMR 3528, Paris, France
- INRA, Unité MaIAGE, UR1404, Jouy-en-Josas, France
| | - A Maruani
- Assistance Publique-Hôpitaux de Paris, Child and Adolescent Psychiatry Department, Robert Debré Hospital, Paris, France
| | - M Bellinzoni
- Institut Pasteur, Unité de Microbiologie Structurale, Paris, France
- CNRS UMR 3528, Paris, France
| | - T Rolland
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - L Gouder
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - A Mathieu
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - J Buratti
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - F Amsellem
- Assistance Publique-Hôpitaux de Paris, Child and Adolescent Psychiatry Department, Robert Debré Hospital, Paris, France
| | - M Benabou
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - J Van-Gils
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - A Beggiato
- Assistance Publique-Hôpitaux de Paris, Child and Adolescent Psychiatry Department, Robert Debré Hospital, Paris, France
| | - M Konyukh
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - J-P Bourgeois
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - M J Gazzellone
- Centre for Applied Genomics, Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - R K C Yuen
- Centre for Applied Genomics, Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - S Walker
- Centre for Applied Genomics, Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
| | - M Delépine
- Centre National de Génotypage, Evry, France
| | - A Boland
- Centre National de Génotypage, Evry, France
| | - B Régnault
- Eukaryote Genotyping Platform, Genopole, Institut Pasteur, Paris, France
| | - M Francois
- Assistance Publique-Hôpitaux de Paris, ENT and Head and Neck Surgery Department, Robert Debré Hospital, Paris-VII University, Paris, France
| | - T Van Den Abbeele
- Assistance Publique-Hôpitaux de Paris, ENT and Head and Neck Surgery Department, Robert Debré Hospital, Paris-VII University, Paris, France
| | - A L Mosca-Boidron
- Département de Génétique, CHU Dijon et Université de Bourgogne, Dijon, France
| | - L Faivre
- Département de Génétique, CHU Dijon et Université de Bourgogne, Dijon, France
| | - Y Shimoda
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Japan
| | - K Watanabe
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Japan
| | - D Bonneau
- Département de Biochimie et Génétique, Centre Hospitalier Universitaire, Angers, France
| | - M Rastam
- Department of Clinical Sciences in Lund, Lund University, Lund, Sweden
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Gothenburg, Sweden
| | - M Leboyer
- INSERM U955, Psychiatrie Translationnelle, Créteil, France
- Université Paris Est, Faculté de Médecine, Créteil, France
- Assistance Publique-Hôpitaux de Paris, DHU Pe-PSY, H. Mondor Hospital, Department of Psychiatry, Créteil, France
- FondaMental Foundation, Créteil, France
| | - S W Scherer
- Centre for Applied Genomics, Program in Genetics and Genome Biology, Hospital for Sick Children, Toronto, ON, Canada
- McLaughlin Centre, Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - C Gillberg
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Gothenburg, Sweden
| | - R Delorme
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
- Assistance Publique-Hôpitaux de Paris, Child and Adolescent Psychiatry Department, Robert Debré Hospital, Paris, France
| | - I Cloëz-Tayarani
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - T Bourgeron
- Human Genetics and Cognitive Functions Unit, Institut Pasteur, Paris, France
- CNRS UMR 3571: Genes, Synapses and Cognition, Institut Pasteur, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
- Gillberg Neuropsychiatry Centre, University of Gothenburg, Gothenburg, Sweden
- FondaMental Foundation, Créteil, France
| |
Collapse
|
37
|
Villarreal MM, Kim SK, Barron L, Kodali R, Baardsnes J, Hinck CS, Krzysiak TC, Henen MA, Pakhomova O, Mendoza V, O'Connor-McCourt MD, Lafer EM, López-Casillas F, Hinck AP. Binding Properties of the Transforming Growth Factor-β Coreceptor Betaglycan: Proposed Mechanism for Potentiation of Receptor Complex Assembly and Signaling. Biochemistry 2016; 55:6880-6896. [PMID: 27951653 PMCID: PMC5551644 DOI: 10.1021/acs.biochem.6b00566] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/03/2023]
Abstract
![]()
Transforming
growth factor (TGF) β1, β2, and β3
(TGF-β1–TGF-β3, respectively) are small secreted
signaling proteins that each signal through the TGF-β type I
and type II receptors (TβRI and TβRII, respectively).
However, TGF-β2, which is well-known to bind TβRII several
hundred-fold more weakly than TGF-β1 and TGF-β3, has an
additional requirement for betaglycan, a membrane-anchored nonsignaling
receptor. Betaglycan has two domains that bind TGF-β2 at independent
sites, but how it binds TGF-β2 to potentiate TβRII binding
and how the complex with TGF-β, TβRII, and betaglycan
undergoes the transition to the signaling complex with TGF-β,
TβRII, and TβRI are not understood. To investigate the
mechanism, the binding of the TGF-βs to the betaglycan extracellular
domain, as well as its two independent binding domains, either directly
or in combination with the TβRI and TβRII ectodomains,
was studied using surface plasmon resonance, isothermal titration
calorimetry, and size-exclusion chromatography. These studies show
that betaglycan binds TGF-β homodimers with a 1:1 stoichiometry
in a manner that allows one molecule of TβRII to bind. These
studies further show that betaglycan modestly potentiates the binding
of TβRII and must be displaced to allow TβRI to bind.
These findings suggest that betaglycan functions to bind and concentrate
TGF-β2 on the cell surface and thus promote the binding of TβRII
by both membrane-localization effects and allostery. These studies
further suggest that the transition to the signaling complex is mediated
by the recruitment of TβRI, which simultaneously displaces betaglycan
and stabilizes the bound TβRII by direct receptor–receptor
contact.
Collapse
Affiliation(s)
| | | | | | - Ravi Kodali
- Department of Structural Biology, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | - Jason Baardsnes
- National Research Council, Human Health Therapeutics Portfolio , Montréal, Quebec, Canada
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | - Troy C Krzysiak
- Department of Structural Biology, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | - Morkos A Henen
- Department of Structural Biology, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| | | | - Valentín Mendoza
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México, Mexico
| | | | | | - Fernando López-Casillas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México , Ciudad de México, Mexico
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh , Pittsburgh, Pennsylvania 15261, United States
| |
Collapse
|
38
|
Abstract
We review the evolution and structure of members of the transforming growth factor β (TGF-β) family, antagonistic or agonistic modulators, and receptors that regulate TGF-β signaling in extracellular environments. The growth factor (GF) domain common to all family members and many of their antagonists evolved from a common cystine knot growth factor (CKGF) domain. The CKGF superfamily comprises six distinct families in primitive metazoans, including the TGF-β and Dan families. Compared with Wnt/Frizzled and Notch/Delta families that also specify body axes, cell fate, tissues, and other families that contain CKGF domains that evolved in parallel, the TGF-β family was the most fruitful in evolution. Complexes between the prodomains and GFs of the TGF-β family suggest a new paradigm for regulating GF release by conversion from closed- to open-arm procomplex conformations. Ternary complexes of the final step in extracellular signaling show how TGF-β GF dimers bind type I and type II receptors on the cell surface, and enable understanding of much of the specificity and promiscuity in extracellular signaling. However, structures suggest that when GFs bind repulsive guidance molecule (RGM) family coreceptors, type I receptors do not bind until reaching an intracellular, membrane-enveloped compartment, blurring the line between extra- and intracellular signaling. Modulator protein structures show how structurally diverse antagonists including follistatins, noggin, and members of the chordin family bind GFs to regulate signaling; complexes with the Dan family remain elusive. Much work is needed to understand how these molecular components assemble to form signaling hubs in extracellular environments in vivo.
Collapse
Affiliation(s)
- Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15260
| | - Thomas D Mueller
- Department of Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, D-97082 Wuerzburg, Germany
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine and Division of Hematology, Department of Medicine, Boston Children's Hospital, Boston, Massachusetts 02115
- Department of Biological Chemistry and Pharmacology, Harvard Medical School, Boston, Massachusetts 02115
| |
Collapse
|
39
|
Liu J, Wang W, Liu M, Su L, Zhou H, Xia Y, Ran J, Lin HY, Yang B. Repulsive guidance molecule b inhibits renal cyst development through the bone morphogenetic protein signaling pathway. Cell Signal 2016; 28:1842-1851. [DOI: 10.1016/j.cellsig.2016.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/15/2016] [Revised: 08/25/2016] [Accepted: 08/25/2016] [Indexed: 02/04/2023]
|
40
|
Zhao N, Maxson JE, Zhang RH, Wahedi M, Enns CA, Zhang AS. Neogenin Facilitates the Induction of Hepcidin Expression by Hemojuvelin in the Liver. J Biol Chem 2016; 291:12322-35. [PMID: 27072365 DOI: 10.1074/jbc.m116.721191] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/10/2016] [Indexed: 01/24/2023] Open
Abstract
Hemojuvelin (HJV) regulates iron homeostasis by direct interaction with bone morphogenetic protein (BMP) ligands to induce hepcidin expression through the BMP signaling pathway in the liver. Crystallography studies indicate that HJV can simultaneously bind to both BMP2 and the ubiquitously expressed cell surface receptor neogenin. However, the role of the neogenin-HJV interaction in the function of HJV is unknown. Here we identify a mutation in HJV that specifically lowers its interaction with neogenin. Expression of this mutant Hjv in the liver of Hjv(-/-) mice dramatically attenuated its induction of BMP signaling and hepcidin mRNA, suggesting that interaction with neogenin is critical for the iron regulatory function of HJV. Further studies revealed that neogenin co-immunoprecipitated with ALK3, an essential type-I BMP receptor for hepatic hepcidin expression. Neogenin has also been shown to facilitate the cleavage of HJV by furin in transfected cells. Surprisingly, although cleavage of HJV by furin has been implicated in the regulation of HJV function in cell culture models and furin-cleaved soluble Hjv is detectable in the serum of mice, mutating the furin cleavage site did not alter the stimulation of hepcidin expression by Hjv in mice. In vivo studies validated the important role of HJV-BMP interaction for Hjv stimulation of BMP signaling and hepcidin expression. Together these data support a model in which neogenin acts as a scaffold to facilitate assembly of the HJV·BMP·BMP receptor complex to induce hepcidin expression.
Collapse
Affiliation(s)
- Ningning Zhao
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| | - Julia E Maxson
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| | - Richard H Zhang
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| | - Mastura Wahedi
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| | - Caroline A Enns
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| | - An-Sheng Zhang
- From the Department of Cell, Developmental, and Cancer Biology, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
41
|
Meng C, Liu W, Huang H, Wang Y, Chen B, Freeman GJ, Schneyer A, Lin HY, Xia Y. Repulsive Guidance Molecule b (RGMb) Is Dispensable for Normal Gonadal Function in Mice. Biol Reprod 2016; 94:78. [PMID: 26911425 DOI: 10.1095/biolreprod.115.135921] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/01/2015] [Accepted: 02/11/2016] [Indexed: 12/22/2022] Open
Abstract
Bone morphogenetic protein (BMP) signaling plays an important role in spermatogenesis and follicle development. Our previous studies have shown that repulsive guidance molecule b (RGMb, also known as Dragon) is a coreceptor that enhances BMP2 and BMP4 signaling in several cell types and that RGMb is expressed in spermatocytes and spermatids in the testis and in oocytes of the secondary follicles in the ovary. Here, we demonstrated that specific deletion of Rgmb in germ cells in the testis and ovary did not alter Smad1/5/8 phosphorylation, gonadal structures, and fertility. In addition, ovaries from postnatal global Rgmb knockout mice showed similar structures to the wild-type ovaries. Our results suggest that RGMb is not essential for normal gonadal function.
Collapse
Affiliation(s)
- Chenling Meng
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenjing Liu
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Huihui Huang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Yang Wang
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Binbin Chen
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Alan Schneyer
- Pioneer Valley Life Science Institute and Department of Veterinary and Animal Science, University of Massachusetts-Amherst, Amherst, Massachusetts
| | - Herbert Y Lin
- Program in Membrane Biology, Center for Systems Biology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Yin Xia
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China School of Biomedical Sciences Core Laboratory, The Chinese University of Hong Kong Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
42
|
van Erp S, van den Heuvel DMA, Fujita Y, Robinson RA, Hellemons AJCGM, Adolfs Y, Van Battum EY, Blokhuis AM, Kuijpers M, Demmers JAA, Hedman H, Hoogenraad CC, Siebold C, Yamashita T, Pasterkamp RJ. Lrig2 Negatively Regulates Ectodomain Shedding of Axon Guidance Receptors by ADAM Proteases. Dev Cell 2015; 35:537-552. [PMID: 26651291 DOI: 10.1016/j.devcel.2015.11.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/04/2014] [Revised: 10/02/2015] [Accepted: 11/09/2015] [Indexed: 12/11/2022]
Abstract
Many guidance receptors are proteolytically cleaved by membrane-associated metalloproteases of the ADAM family, leading to the shedding of their ectodomains. Ectodomain shedding is crucial for receptor signaling and function, but how this process is controlled in neurons remains poorly understood. Here, we show that the transmembrane protein Lrig2 negatively regulates ADAM-mediated guidance receptor proteolysis in neurons. Lrig2 binds Neogenin, a receptor for repulsive guidance molecules (RGMs), and prevents premature Neogenin shedding by ADAM17 (TACE). RGMa reduces Lrig2-Neogenin interactions, providing ADAM17 access to Neogenin and allowing this protease to induce ectodomain shedding. Regulation of ADAM17-mediated Neogenin cleavage by Lrig2 is required for neurite growth inhibition by RGMa in vitro and for cortical neuron migration in vivo. Furthermore, knockdown of Lrig2 significantly improves CNS axon regeneration. Together, our data identify a unique ligand-gated mechanism to control receptor shedding by ADAMs and reveal functions for Lrigs in neuron migration and regenerative failure.
Collapse
Affiliation(s)
- Susan van Erp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Dianne M A van den Heuvel
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Yuki Fujita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Ross A Robinson
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Anita J C G M Hellemons
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Youri Adolfs
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Eljo Y Van Battum
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Anna M Blokhuis
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands
| | - Marijn Kuijpers
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Jeroen A A Demmers
- Proteomics Centre and Department of Cell Biology, Erasmus University Medical Centre, Dr Molewaterplein 50, 3015 GE Rotterdam, the Netherlands
| | - Håkan Hedman
- Oncology Research Laboratory, Department of Radiation Sciences, Umeå University, 90187 Umeå, Sweden
| | - Casper C Hoogenraad
- Cell Biology, Faculty of Science, Utrecht University, 3584 CH Utrecht, the Netherlands
| | - Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University 2-2, Yamadaoka, Suita, Osaka 565-0871, Japan
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
43
|
Liu J, Gao HY, Wang XF. The role of the Rho/ROCK signaling pathway in inhibiting axonal regeneration in the central nervous system. Neural Regen Res 2015; 10:1892-6. [PMID: 26807132 PMCID: PMC4705809 DOI: 10.4103/1673-5374.170325] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 02/05/2023] Open
Abstract
The Rho/Rho-associated coiled-coil containing protein kinase (Rho/ROCK) pathway is a major signaling pathway in the central nervous system, transducing inhibitory signals to block regeneration. After central nervous system damage, the main cause of impaired regeneration is the presence of factors that strongly inhibit regeneration in the surrounding microenvironment. These factors signal through the Rho/ROCK signaling pathway to inhibit regeneration. Therefore, a thorough understanding of the Rho/ROCK signaling pathway is crucial for advancing studies on regeneration and repair of the injured central nervous system.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
- Correspondence to: Jing Liu, .
| | - Hong-yan Gao
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
| | - Xiao-feng Wang
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
- Department of Neonatology, People's Hospital of Rizhao, Rizhao, Shangdong Province, China
| |
Collapse
|
44
|
Healey EG, Bishop B, Elegheert J, Bell CH, Padilla-Parra S, Siebold C. Repulsive guidance molecule is a structural bridge between neogenin and bone morphogenetic protein. Nat Struct Mol Biol 2015; 22:458-65. [PMID: 25938661 PMCID: PMC4456160 DOI: 10.1038/nsmb.3016] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/17/2014] [Accepted: 03/31/2015] [Indexed: 02/07/2023]
Abstract
Repulsive guidance molecules (RGMs) control crucial processes including cell motility, adhesion, immune-cell regulation and systemic iron metabolism. RGMs signal via the neogenin (NEO1) and the bone morphogenetic protein (BMP) pathways. Here, we report crystal structures of the N-terminal domains of all human RGM family members in complex with the BMP ligand BMP2, revealing a new protein fold and a conserved BMP-binding mode. Our structural and functional data suggest a pH-linked mechanism for RGM-activated BMP signaling and offer a rationale for RGM mutations causing juvenile hemochromatosis. We also determined the crystal structure of the ternary BMP2-RGM-NEO1 complex, which, along with solution scattering and live-cell super-resolution fluorescence microscopy, indicates BMP-induced clustering of the RGM-NEO1 complex. Our results show how RGM acts as the central hub that links BMP and NEO1 and physically connects these fundamental signaling pathways.
Collapse
Affiliation(s)
- Eleanor G Healey
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Benjamin Bishop
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jonathan Elegheert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christian H Bell
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Sergi Padilla-Parra
- 1] Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK. [2] Cellular Imaging Core, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Christian Siebold
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
45
|
Rausa M, Ghitti M, Pagani A, Nai A, Campanella A, Musco G, Camaschella C, Silvestri L. Identification of TMPRSS6 cleavage sites of hemojuvelin. J Cell Mol Med 2015; 19:879-88. [PMID: 25704252 PMCID: PMC4395201 DOI: 10.1111/jcmm.12462] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/18/2014] [Accepted: 09/19/2014] [Indexed: 12/11/2022] Open
Abstract
Hemojuvelin (HJV), the coreceptor of the BMP-SMAD pathway that up-regulates hepcidin transcription, is a repulsive guidance molecule (RGMc) which undergoes a complex intracellular processing. Following autoproteolysis, it is exported to the cell surface both as a full-length and a heterodimeric protein. In vitro membrane HJV (m-HJV) is cleaved by the transmembrane serine protease TMPRSS6 to attenuate signalling and to inhibit hepcidin expression. In this study, we investigated the number and position of HJV cleavage sites by mutagenizing arginine residues (R), potential TMPRSS6 targets, to alanine (A). We analysed translation and membrane expression of HJV R mutants and the pattern of fragments they release in the culture media in the presence of TMPRSS6. Abnormal fragments were observed for mutants at arginine 121, 176, 218, 288 and 326. Considering that all variants, except HJVR121A, lack autoproteolytic activity and some (HJVR176A and HJVR288A) are expressed at reduced levels on cell surface, we identified the fragments originating from either full-length or heterodimeric proteins and defined the residues 121 and 326 as the TMPRSS6 cleavage sites in both isoforms. Using the N-terminal FLAG-tagged HJV, we showed that residue 121 is critical also in the rearrangement of the N-terminal heterodimeric HJV. Exploiting the recently reported RGMb crystallographic structure, we generated a model of HJV that was used as input structure for all-atoms molecular dynamics simulation in explicit solvent. As assessed by in silico studies, we concluded that some arginines in the von Willebrand domain appear TMPRSS6 insensitive, likely because of partial protein structure destabilization.
Collapse
Affiliation(s)
- Marco Rausa
- Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy; Vita Salute University, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Coles CH, Mitakidis N, Zhang P, Elegheert J, Lu W, Stoker AW, Nakagawa T, Craig AM, Jones EY, Aricescu AR. Structural basis for extracellular cis and trans RPTPσ signal competition in synaptogenesis. Nat Commun 2014; 5:5209. [PMID: 25385546 PMCID: PMC4239663 DOI: 10.1038/ncomms6209] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/30/2014] [Accepted: 09/09/2014] [Indexed: 01/26/2023] Open
Abstract
Receptor protein tyrosine phosphatase sigma (RPTPσ) regulates neuronal extension and acts as a presynaptic nexus for multiple protein and proteoglycan interactions during synaptogenesis. Unknown mechanisms govern the shift in RPTPσ function, from outgrowth promotion to synaptic organization. Here, we report crystallographic, electron microscopic and small-angle X-ray scattering analyses, which reveal sufficient inter-domain flexibility in the RPTPσ extracellular region for interaction with both cis (same cell) and trans (opposite cell) ligands. Crystal structures of RPTPσ bound to its postsynaptic ligand TrkC detail an interaction surface partially overlapping the glycosaminoglycan-binding site. Accordingly, heparan sulphate and heparin oligomers compete with TrkC for RPTPσ binding in vitro and disrupt TrkC-dependent synaptic differentiation in neuronal co-culture assays. We propose that transient RPTPσ ectodomain emergence from the presynaptic proteoglycan layer allows capture by TrkC to form a trans-synaptic complex, the consequent reduction in RPTPσ flexibility potentiating interactions with additional ligands to orchestrate excitatory synapse formation.
Collapse
Affiliation(s)
- Charlotte H. Coles
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Nikolaos Mitakidis
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Peng Zhang
- Brain Research Centre and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - Jonathan Elegheert
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Weixian Lu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Andrew W. Stoker
- Cancer Section, Institute of Child Health, University College London, 30 Guilford Street, London WC1N 1EH, UK
| | - Terunaga Nakagawa
- Department of Molecular Physiology and Biophysics, Vanderbilt University, School of Medicine, 702 Light Hall (0615), Nashville, Tennessee 37232-0615, USA
| | - Ann Marie Craig
- Brain Research Centre and Department of Psychiatry, University of British Columbia, Vancouver, British Columbia, Canada V6T 2B5
| | - E. Yvonne Jones
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - A. Radu Aricescu
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| |
Collapse
|
47
|
Finci LI, Krüger N, Sun X, Zhang J, Chegkazi M, Wu Y, Schenk G, Mertens HDT, Svergun DI, Zhang Y, Wang JH, Meijers R. The crystal structure of netrin-1 in complex with DCC reveals the bifunctionality of netrin-1 as a guidance cue. Neuron 2014; 83:839-849. [PMID: 25123307 DOI: 10.1016/j.neuron.2014.07.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 07/07/2014] [Indexed: 01/01/2023]
Abstract
Netrin-1 is a guidance cue that can trigger either attraction or repulsion effects on migrating axons of neurons, depending on the repertoire of receptors available on the growth cone. How a single chemotropic molecule can act in such contradictory ways has long been a puzzle at the molecular level. Here we present the crystal structure of netrin-1 in complex with the Deleted in Colorectal Cancer (DCC) receptor. We show that one netrin-1 molecule can simultaneously bind to two DCC molecules through a DCC-specific site and through a unique generic receptor binding site, where sulfate ions staple together positively charged patches on both DCC and netrin-1. Furthermore, we demonstrate that UNC5A can replace DCC on the generic receptor binding site to switch the response from attraction to repulsion. We propose that the modularity of binding allows for the association of other netrin receptors at the generic binding site, eliciting alternative turning responses.
Collapse
Affiliation(s)
- Lorenzo I Finci
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA. 02215, USA
| | - Nina Krüger
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| | - Xiaqin Sun
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Jie Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Magda Chegkazi
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| | - Yu Wu
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China
| | - Gundolf Schenk
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| | - Haydyn D T Mertens
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| | - Dmitri I Svergun
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| | - Yan Zhang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, 100871, China
| | - Jia-Huai Wang
- State Key Laboratory of Biomembrane and Membrane Biotechnology, College of Life Sciences, Peking University, Beijing, 100871, China.,Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA. 02215, USA
| | - Rob Meijers
- European Molecular Biology Laboratory (EMBL), Hamburg Outstation, Notkestrasse 85, 22607, Hamburg, Germany
| |
Collapse
|
48
|
Xu K, Wu Z, Renier N, Antipenko A, Tzvetkova-Robev D, Xu Y, Minchenko M, Nardi-Dei V, Rajashankar KR, Himanen J, Tessier-Lavigne M, Nikolov DB. Neural migration. Structures of netrin-1 bound to two receptors provide insight into its axon guidance mechanism. Science 2014; 344:1275-9. [PMID: 24876346 PMCID: PMC4369087 DOI: 10.1126/science.1255149] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023]
Abstract
Netrins are secreted proteins that regulate axon guidance and neuronal migration. Deleted in colorectal cancer (DCC) is a well-established netrin-1 receptor mediating attractive responses. We provide evidence that its close relative neogenin is also a functional netrin-1 receptor that acts with DCC to mediate guidance in vivo. We determined the structures of a functional netrin-1 region, alone and in complexes with neogenin or DCC. Netrin-1 has a rigid elongated structure containing two receptor-binding sites at opposite ends through which it brings together receptor molecules. The ligand/receptor complexes reveal two distinct architectures: a 2:2 heterotetramer and a continuous ligand/receptor assembly. The differences result from different lengths of the linker connecting receptor domains fibronectin type III domain 4 (FN4) and FN5, which differs among DCC and neogenin splice variants, providing a basis for diverse signaling outcomes.
Collapse
Affiliation(s)
- Kai Xu
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Zhuhao Wu
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York 10065
| | - Nicolas Renier
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York 10065
| | - Alexander Antipenko
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | | | - Yan Xu
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Maria Minchenko
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Vincenzo Nardi-Dei
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Kanagalaghatta R. Rajashankar
- Department of Chemistry and Chemical Biology, Cornell University & NE-CAT, Advanced Photon Source, Argonne, IL 60439
| | - Juha Himanen
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065
| | - Marc Tessier-Lavigne
- Laboratory of Brain Development and Repair, The Rockefeller University, New York, New York 10065,Correspondence to: or
| | - Dimitar B. Nikolov
- Structural Biology Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065,Correspondence to: or
| |
Collapse
|
49
|
Barak R, Lahmi R, Gevorkyan-Airapetov L, Levy E, Tzur A, Opatowsky Y. Crystal structure of the extracellular juxtamembrane region of Robo1. J Struct Biol 2014; 186:283-91. [PMID: 24607414 DOI: 10.1016/j.jsb.2014.02.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/27/2013] [Revised: 02/26/2014] [Accepted: 02/28/2014] [Indexed: 12/17/2022]
Abstract
Robo receptors play pivotal roles in neurodevelopment, and their deregulation is implicated in several neuropathological conditions and cancers. To date, the mechanism of Robo activation and regulation remains obscure. Here we present the crystal structure of the juxtamembrane (JM) domains of human Robo1. The structure exhibits unexpectedly high backbone similarity to the netrin and RGM binding region of neogenin and DCC, which are functionally related receptors of Robo1. Comparison of these structures reveals a conserved surface that overlaps with a cluster of oncogenic and neuropathological mutations found in all Robo isoforms. The structure also reveals the intricate folding of the JM linker, which points to its role in Robo1 activation. Further experiments with cultured cells demonstrate that exposure or relief of the folded JM linker results in enhanced shedding of the Robo1 ectodomain.
Collapse
Affiliation(s)
- Reut Barak
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Roxane Lahmi
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Lada Gevorkyan-Airapetov
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Eliad Levy
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Amit Tzur
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel; Advanced Materials and Nanotechnology Institute, Bar-Ilan University, Ramat-Gan 5290002, Israel
| | - Yarden Opatowsky
- The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
50
|
Xiao Y, Yu S, Zhu B, Bedoret D, Bu X, Francisco LM, Hua P, Duke-Cohan JS, Umetsu DT, Sharpe AH, DeKruyff RH, Freeman GJ. RGMb is a novel binding partner for PD-L2 and its engagement with PD-L2 promotes respiratory tolerance. ACTA ACUST UNITED AC 2014; 211:943-59. [PMID: 24752301 PMCID: PMC4010901 DOI: 10.1084/jem.20130790] [Citation(s) in RCA: 253] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/30/2022]
Abstract
Interaction between the inhibitory molecule PD-L2 on dendritic cells and repulsive guidance molecule b (RGMb) on lung macrophages is required to establish respiratory tolerance. We report that programmed death ligand 2 (PD-L2), a known ligand of PD-1, also binds to repulsive guidance molecule b (RGMb), which was originally identified in the nervous system as a co-receptor for bone morphogenetic proteins (BMPs). PD-L2 and BMP-2/4 bind to distinct sites on RGMb. Normal resting lung interstitial macrophages and alveolar epithelial cells express high levels of RGMb mRNA, whereas lung dendritic cells express PD-L2. Blockade of the RGMb–PD-L2 interaction markedly impaired the development of respiratory tolerance by interfering with the initial T cell expansion required for respiratory tolerance. Experiments with PD-L2–deficient mice showed that PD-L2 expression on non–T cells was critical for respiratory tolerance, but expression on T cells was not required. Because PD-L2 binds to both PD-1, which inhibits antitumor immunity, and to RGMb, which regulates respiratory immunity, targeting the PD-L2 pathway has therapeutic potential for asthma, cancer, and other immune-mediated disorders. Understanding this pathway may provide insights into how to optimally modulate the PD-1 pathway in cancer immunotherapy while minimizing adverse events.
Collapse
Affiliation(s)
- Yanping Xiao
- Department of Medical Oncology, Dana-Farber Cancer Institute; 2 Division of Immunology and Department of Pediatrics, Boston Children's Hospital; 3 Department of Microbiology and Immunobiology and 4 Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|