1
|
Xu X, Fu J, Yang G, Chen Z, Chen S, Yuan G. Dentin sialoprotein promotes endothelial differentiation of dental pulp stem cells through DSP aa34-50-endoglin-AKT1 axis. J Biol Chem 2025; 301:108380. [PMID: 40049415 PMCID: PMC11997338 DOI: 10.1016/j.jbc.2025.108380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 04/01/2025] Open
Abstract
Dentin sialoprotein (DSP), a major dentin extracellular matrix noncollagenous protein, is well recognized as an important regulator for dentinogenesis. DSP as a secreted protein can interact with membrane receptors, activate intracellular signaling, and initiate the odontoblastic differentiation of dental papilla cells. In a recent study, we have demonstrated that DSP can induce the endothelial differentiation of dental pulp stem cells (DPSCs), a type of tooth pulp-derived multipotent stem cells, dependent on membrane receptor endoglin (ENG). However, the intimate mechanisms by which DSP-ENG association facilitates the endothelial differentiation of DPSCs remain enigmatic. Here, we find that the amino acid (aa) residues 34-50 of DSP (DSPaa34-50) is responsible for its association with ENG using a series of co-immunoprecipitation assays. Immunofluorescent staining and in situ proximity ligation assay demonstrate that overexpressed ENG in human embryonic kidney 293T cells shows codistribution and proximity ligation assay signals to the supplemented DSPaa34-50 protein but not to DSP without aa34-50 (DSPΔ34-50) on cell surfaces. Moreover, the zona pellucida domain of ENG mediates its association with DSPaa34-50. Further experiments indicate that DSPaa34-50 exhibits equivalent effects to the full-length DSP on the migration and endothelial differentiation of DPSCs dependent on ENG but DSPΔ34-50 does not. Mechanistically, DSPaa34-50 activates AKT1 and triggers the expression of blood vessel development-related genes in DPSCs. Multiple experiments demonstrate that AKT1 inhibition suppresses the DSPaa34-50-induced migration and endothelial differentiation of DPSCs. Thus, AKT1 mediates the cellular and molecular functions of DSPaa34-50-ENG association. Collectively, these findings identify that DSP promotes the endothelial differentiation of DPSCs through the DSPaa34-50-ENG-AKT1 signaling axis.
Collapse
Affiliation(s)
- Ximin Xu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei, China
| | - Jing Fu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei, China
| | - Guobin Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Zhi Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China
| | - Shuo Chen
- Department of Developmental Dentistry, School of Dentistry, The University of Texas Health Science Center at San Antonio, San Antonio, Texas, United States
| | - Guohua Yuan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei, China; Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, Hubei, China.
| |
Collapse
|
2
|
Niazi A, Kim JA, Kim DK, Lu D, Sterin I, Park J, Park S. Microvilli control the morphogenesis of the tectorial membrane extracellular matrix. Dev Cell 2025; 60:679-695.e8. [PMID: 39657673 PMCID: PMC11905117 DOI: 10.1016/j.devcel.2024.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024]
Abstract
The apical extracellular matrix (aECM), organized by polarized epithelial cells, exhibits complex structures. The tectorial membrane (TM), an aECM in the cochlea mediating auditory transduction, exhibits highly ordered domain-specific architecture. α-Tectorin (TECTA), a glycosylphosphatidylinositol (GPI)-anchored ECM protein, is essential for TM organization. Here, we identified that α-tectorin is released by distinct modes: proteolytic shedding by TMPRSS2 and GPI-anchor-dependent release from the microvillus tip in mice. In the medial/limbal domain, proteolytically shed α-tectorin forms dense fibers. In contrast, in the lateral/body domain, where supporting cells exhibit dense microvilli, shedding restricts α-tectorin to the microvillus tip, compartmentalizing collagen-binding sites. Tip-localized α-tectorin is released in a GPI-anchor-dependent manner to form collagen-crosslinking fibers, maintaining the spacing and parallel organization of collagen fibrils. Overall, these distinct release modes of α-tectorin determine domain-specific organization, with the microvillus coordinating release modes along its membrane to assemble the higher-order ECM architecture.
Collapse
Affiliation(s)
- Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA; Neuroscience Program, University of Utah, Salt Lake City, UT, USA
| | - Ju Ang Kim
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Dong-Kyu Kim
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Di Lu
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
3
|
Wieteska Ł, Taylor AB, Punch E, Coleman JA, Conway IO, Lin YF, Byeon CH, Hinck CS, Krzysiak T, Ishima R, López-Casillas F, Cherepanov P, Bernard DJ, Hill CS, Hinck AP. Structures of TGF-β with betaglycan and signaling receptors reveal mechanisms of complex assembly and signaling. Nat Commun 2025; 16:1778. [PMID: 40011426 PMCID: PMC11865472 DOI: 10.1038/s41467-025-56796-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 01/30/2025] [Indexed: 02/28/2025] Open
Abstract
Betaglycan (BG) is a transmembrane co-receptor of the transforming growth factor-β (TGF-β) family of signaling ligands. It is essential for embryonic development, tissue homeostasis and fertility in adults. It functions by enabling binding of the three TGF-β isoforms to their signaling receptors and is additionally required for inhibin A (InhA) activity. Despite its requirement for the functions of TGF-βs and InhA in vivo, structural information explaining BG ligand selectivity and its mechanism of action is lacking. Here, we determine the structure of TGF-β bound both to BG and the signaling receptors, TGFBR1 and TGFBR2. We identify key regions responsible for ligand engagement, which has revealed binding interfaces that differ from those described for the closely related co-receptor of the TGF-β family, endoglin, thus demonstrating remarkable evolutionary adaptation to enable ligand selectivity. Finally, we provide a structural explanation for the hand-off mechanism underlying TGF-β signal potentiation.
Collapse
Affiliation(s)
- Łukasz Wieteska
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
- Faculty of Biological Sciences, Astbury Centre for Structural Studies, University of Leeds, Leeds, UK
- Developmental Signalling Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Alexander B Taylor
- Department of Biochemistry & Structural Biology and Greehey Children's Cancer Research Institute, The University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Emma Punch
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Jonathan A Coleman
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Isabella O Conway
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yeu-Farn Lin
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Chang-Hyeock Byeon
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Troy Krzysiak
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fernando López-Casillas
- Departmento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, UNAM, Mexico City, Mexico
| | - Peter Cherepanov
- Chromatin Structure and Mobile DNA Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Daniel J Bernard
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | - Caroline S Hill
- Developmental Signalling Laboratory, The Francis Crick Institute, London, NW1 1AT, UK.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
4
|
Wieteska Ł, Taylor AB, Punch E, Coleman JA, Conway IO, Lin YF, Byeon CH, Hinck CS, Krzysiak T, Ishima R, López-Casillas F, Cherepanov P, Bernard DJ, Hill CS, Hinck AP. Structures of TGF-β with betaglycan and the signaling receptors reveal the mechanism whereby betaglycan potentiates receptor complex assembly and signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.19.604101. [PMID: 39091787 PMCID: PMC11291015 DOI: 10.1101/2024.07.19.604101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Betaglycan (BG) is a transmembrane co-receptor of the transforming growth factor-β (TGF-β) family of signaling ligands. It is essential for embryonic development and tissue homeostasis and fertility in adults. It functions by enabling binding of the three TGF-β isoforms to their signaling receptors and is additionally required for inhibin A (InhA) activity. Despite its requirement for the functions of TGF-βs and InhA in vivo, structural information explaining BG ligand selectivity and its mechanism of action is lacking. Here, we determine the structure of TGF-β bound both to BG and the signaling receptors, TGFBR1 and TGFBR2. We identify key regions responsible for ligand engagement, which has revealed novel binding interfaces that differ from those described for the closely related co-receptor of the TGF-β family, endoglin, thus demonstrating remarkable evolutionary adaptation to enable ligand selectivity. Finally, we provide a structural explanation for the hand-off mechanism underlying TGF-β signal potentiation.
Collapse
|
5
|
Niazi A, Kim JA, Kim DK, Lu D, Sterin I, Park J, Park S. Microvilli regulate the release modes of alpha-tectorin to organize the domain-specific matrix architecture of the tectorial membrane. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.04.574255. [PMID: 38260557 PMCID: PMC10802356 DOI: 10.1101/2024.01.04.574255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
The tectorial membrane (TM) is an apical extracellular matrix (ECM) in the cochlea essential for auditory transduction. The TM exhibits highly ordered domain-specific architecture. Alpha-tectorin/TECTA is a glycosylphosphatidylinositol (GPI)-anchored ECM protein essential for TM organization. Here, we identified that TECTA is released by distinct modes: proteolytic shedding by TMPRSS2 and GPI-anchor-dependent release from the microvillus tip. In the medial/limbal domain, proteolytically shed TECTA forms dense fibers. In the lateral/body domain produced by the supporting cells displaying dense microvilli, the proteolytic shedding restricts TECTA to the microvillus tip and compartmentalizes the collagen-binding site. The tip-localized TECTA, in turn, is released in a GPI-anchor-dependent manner to form collagen-crosslinking fibers, required for maintaining the spacing and parallel organization of collagen fibrils. Overall, we showed that distinct release modes of TECTA determine the domain-specific organization pattern, and the microvillus coordinates the release modes along its membrane to organize the higher-order ECM architecture.
Collapse
Affiliation(s)
- Ava Niazi
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Neuroscience Program, University of Utah, Salt Lake City, Utah, USA
| | - Ju Ang Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Current affiliation: Department of Otorhinolaryngology-Head and Neck Surgery, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Dong-Kyu Kim
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
- Current affiliation: Genetics & Genome Biology, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Di Lu
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Igal Sterin
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Joosang Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| | - Sungjin Park
- Department of Neurobiology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
6
|
Borgini M, Wieteska Ł, Hinck CS, Krzysiak T, Hinck AP, Wipf P. Synthesis of 13C-methyl-labeled amino acids and their incorporation into proteins in mammalian cells. Org Biomol Chem 2023; 21:9216-9229. [PMID: 37964666 PMCID: PMC10825848 DOI: 10.1039/d3ob01320k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 11/06/2023] [Indexed: 11/16/2023]
Abstract
Isotopic labeling of methyl-substituted proteinogenic amino acids with 13C has transformed applications of solution-based NMR spectroscopy and allowed the study of much larger and more complex proteins than previously possible with 15N labeling. Procedures are well-established for producing methyl-labeled proteins expressed in bacteria, with efficient incorporation of 13C-methyl labeled metabolic precursors to enable the isotopic labeling of Ile, Val, and Leu methyl groups. Recently, similar methodology has been applied to enable 13C-methyl labeling of Ile, Val, and Leu in yeast, extending the approach to proteins that do not readily fold when produced in bacteria. Mammalian or insect cells are nonetheless preferable for production of many human proteins, yet 13C-methyl labeling using similar metabolic precursors is not feasible as these cells lack the requisite biosynthetic machinery. Herein, we report versatile and high-yielding synthetic routes to 13C methyl-labeled amino acids based on palladium-catalyzed C(sp3)-H functionalization. We demonstrate the efficient incorporation of two of the synthesized amino acids, 13C-γ2-Ile and 13C-γ1,γ2-Val, into human receptor extracellular domains with multiple disulfides using suspension-cultured HEK293 cells. Production costs are reasonable, even at moderate expression levels of 2-3 mg purified protein per liter of medium, and the method can be extended to label other methyl groups, such as 13C-δ1-Ile and 13C-δ1,δ2-Leu. In summary, we demonstrate the cost-effective production of methyl-labeled proteins in mammalian cells by incorporation of 13C methyl-labeled amino acids generated de novo by a versatile synthetic route.
Collapse
Affiliation(s)
- Matteo Borgini
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Łukasz Wieteska
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Troy Krzysiak
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| | - Peter Wipf
- Department of Chemistry, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
7
|
Madamanchi A, Ingle M, Hinck AP, Umulis DM. Computational modeling of TGF-β2:TβRI:TβRII receptor complex assembly as mediated by the TGF-β coreceptor betaglycan. Biophys J 2023; 122:1342-1354. [PMID: 36869592 PMCID: PMC10111353 DOI: 10.1016/j.bpj.2023.02.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/16/2022] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Transforming growth factor-β1, -β2, and -β3 (TGF-β1, -β2, and -β3) are secreted signaling ligands that play essential roles in tissue development, tissue maintenance, immune response, and wound healing. TGF-β ligands form homodimers and signal by assembling a heterotetrameric receptor complex comprised of two type I receptor (TβRI):type II receptor (TβRII) pairs. TGF-β1 and TGF-β3 ligands signal with high potency due to their high affinity for TβRII, which engenders high-affinity binding of TβRI through a composite TGF-β:TβRII binding interface. However, TGF-β2 binds TβRII 200-500 more weakly than TGF-β1 and TGF-β3 and signals with lower potency compared with these ligands. Remarkably, the presence of an additional membrane-bound coreceptor, known as betaglycan, increases TGF-β2 signaling potency to levels similar to TGF-β1 and -β3. The mediating effect of betaglycan occurs even though it is displaced from and not present in the heterotetrameric receptor complex through which TGF-β2 signals. Published biophysics studies have experimentally established the kinetic rates of the individual ligand-receptor and receptor-receptor interactions that initiate heterotetrameric receptor complex assembly and signaling in the TGF-β system; however, current experimental approaches are not able to directly measure kinetic rates for the intermediate and latter steps of assembly. To characterize these steps in the TGF-β system and determine the mechanism of betaglycan in the potentiation of TGF-β2 signaling, we developed deterministic computational models with different modes of betaglycan binding and varying cooperativity between receptor subtypes. The models identified conditions for selective enhancement of TGF-β2 signaling. The models provide support for additional receptor binding cooperativity that has been hypothesized but not evaluated in the literature. The models further showed that betaglycan binding to the TGF-β2 ligand through two domains provides an effective mechanism for transfer to the signaling receptors that has been tuned to efficiently promote assembly of the TGF-β2(TβRII)2(TβRI)2 signaling complex.
Collapse
Affiliation(s)
- Aasakiran Madamanchi
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana
| | - Michelle Ingle
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - David M Umulis
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, Indiana; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, Indiana.
| |
Collapse
|
8
|
Hsu YJ, Yin YJ, Tsai KF, Jian CC, Liang ZW, Hsu CY, Wang CC. TGFBR3 supports anoikis through suppressing ATF4 signaling. J Cell Sci 2022; 135:276173. [PMID: 35912788 DOI: 10.1242/jcs.258396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 07/18/2022] [Indexed: 11/20/2022] Open
Abstract
Epithelial morphogenesis and oncogenic transformation can cause loss of cell adhesion, and detached cells are eliminated by anoikis. Here, we reveal that transforming growth factor beta receptor 3 (TGFBR3) acts as an anoikis mediator through the coordination of activating transcription factor 4 (ATF4). In breast cancer, TGFBR3 is progressively lost, but elevated TGFBR3 is associated with a histologic subtype characterized by cellular adhesion defects. Dissecting the impact of extracellular matrix (ECM) deprivation, we demonstrate that ECM loss promotes TGFBR3 expression, which in turn differentiates cell aggregates to a prosurvival phenotype and drives the intrinsic apoptotic pathway. We demonstrate that inhibition of TGFBR3 impairs epithelial anoikis by activating ATF4 signaling. These preclinical findings provide a rationale for therapeutic inhibition of ATF4 in the subgroup of breast cancer patients with low TGFBR3 expression.
Collapse
Affiliation(s)
- Yu-Jhen Hsu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yih-Jia Yin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan.,Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Kai-Feng Tsai
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Cian-Chun Jian
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Zi-Wen Liang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chien-Yu Hsu
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chun-Chao Wang
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan.,Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| |
Collapse
|
9
|
Hou M, Zhu L, Jiang J, Liu Z, Li Z, Jia W, Hu J, Zhou X, Zhang D, Luo Y, Peng X, Xi Q, Jin L, Zhang X. Novel Heterozygous Mutations in ZP2 Cause Abnormal Zona Pellucida and Female Infertility. Reprod Sci 2022; 29:3047-3054. [PMID: 35595959 DOI: 10.1007/s43032-022-00958-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 04/23/2022] [Indexed: 11/24/2022]
Abstract
Zona pellucida (ZP) which is an extracellular matrix consisting of ZP1, ZP2, ZP3, and ZP4 plays a vital role in oocyte maturity, early embryonic development, and fertilization process. Any alterations of structure or function may lead to the abnormal formation of ZP and female infertility. Two novel heterozygous mutations c.1859G > A (p.Cys620Tyr) and c.1421 T > C (p.Leu474Pro) in ZP2 gene were recognized in three patients from two unrelated families with abnormal ZP and female infertility in this study. The expression constructs carrying wild-type ZP2 gene, c.1859G > A (p.Cys620Tyr) mutant ZP2 gene, and c.1421 T > C (p.Leu474Pro) mutant ZP2 gene were transfected into CHO cells respectively. There was a remarkable decrease in the expression of p.Cys620Tyr mutant protein with western blot. In addition, secretion of p.Leu474Pro mutant protein in the culture medium reduced markedly compared with that of wild-type ZP2 protein. Furthermore, co-immunoprecipitation showed that the p.Leu474Pro mutation affected the interaction between ZP2 and ZP3. Prediction of three-dimensional (3D) structure of the proteins showed that p.Cys620Tyr mutation altered the disulfide bond of ZP2 protein and may affect its function. These findings extend the ranges of mutations of ZP2 gene. Meanwhile, it will be helpful to the precise diagnosis of abnormal ZP.
Collapse
Affiliation(s)
- Meiqi Hou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Lixia Zhu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jinghang Jiang
- Reproductive Medicine Center, Jingmen No. 2 People's Hospital, Jingmen, Hubei, China
| | - Zhenxing Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Zhou Li
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Weimin Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Juan Hu
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaopei Zhou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Dazhi Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yalin Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xuejie Peng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Qingsong Xi
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Jin
- Reproductive Medicine Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xianqin Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.
| |
Collapse
|
10
|
Rivera AM, Wilburn DB, Swanson WJ. Domain Expansion and Functional Diversification in Vertebrate Reproductive Proteins. Mol Biol Evol 2022; 39:msac105. [PMID: 35587583 PMCID: PMC9154058 DOI: 10.1093/molbev/msac105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The rapid evolution of fertilization proteins has generated remarkable diversity in molecular structure and function. Glycoproteins of vertebrate egg coats contain multiple zona pellucida (ZP)-N domains (1-6 copies) that facilitate multiple reproductive functions, including species-specific sperm recognition. In this report, we integrate phylogenetics and machine learning to investigate how ZP-N domains diversify in structure and function. The most C-terminal ZP-N domain of each paralog is associated with another domain type (ZP-C), which together form a "ZP module." All modular ZP-N domains are phylogenetically distinct from nonmodular or free ZP-N domains. Machine learning-based classification identifies eight residues that form a stabilizing network in modular ZP-N domains that is absent in free domains. Positive selection is identified in some free ZP-N domains. Our findings support that strong purifying selection has conserved an essential structural core in modular ZP-N domains, with the relaxation of this structural constraint allowing free N-terminal domains to functionally diversify.
Collapse
Affiliation(s)
- Alberto M. Rivera
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Damien B. Wilburn
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA
| | - Willie J. Swanson
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| |
Collapse
|
11
|
Rivera AM, Swanson WJ. The Importance of Gene Duplication and Domain Repeat Expansion for the Function and Evolution of Fertilization Proteins. Front Cell Dev Biol 2022; 10:827454. [PMID: 35155436 PMCID: PMC8830517 DOI: 10.3389/fcell.2022.827454] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/12/2022] [Indexed: 11/13/2022] Open
Abstract
The process of gene duplication followed by gene loss or evolution of new functions has been studied extensively, yet the role gene duplication plays in the function and evolution of fertilization proteins is underappreciated. Gene duplication is observed in many fertilization protein families including Izumo, DCST, ZP, and the TFP superfamily. Molecules mediating fertilization are part of larger gene families expressed in a variety of tissues, but gene duplication followed by structural modifications has often facilitated their cooption into a fertilization function. Repeat expansions of functional domains within a gene also provide opportunities for the evolution of novel fertilization protein. ZP proteins with domain repeat expansions are linked to species-specificity in fertilization and TFP proteins that experienced domain duplications were coopted into a novel sperm function. This review outlines the importance of gene duplications and repeat domain expansions in the evolution of fertilization proteins.
Collapse
Affiliation(s)
- Alberto M. Rivera
- Department of Genome Sciences, University of Washington, Seattle, WA, United States
| | | |
Collapse
|
12
|
Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, Duca L, Durbeej M, Afratis NA, Troeberg L, Franchi M, Masola V, Onisto M. A guide to the composition and functions of the extracellular matrix. FEBS J 2021; 288:6850-6912. [PMID: 33605520 DOI: 10.1111/febs.15776] [Citation(s) in RCA: 505] [Impact Index Per Article: 126.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/13/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022]
Abstract
Extracellular matrix (ECM) is a dynamic 3-dimensional network of macromolecules that provides structural support for the cells and tissues. Accumulated knowledge clearly demonstrated over the last decade that ECM plays key regulatory roles since it orchestrates cell signaling, functions, properties and morphology. Extracellularly secreted as well as cell-bound factors are among the major members of the ECM family. Proteins/glycoproteins, such as collagens, elastin, laminins and tenascins, proteoglycans and glycosaminoglycans, hyaluronan, and their cell receptors such as CD44 and integrins, responsible for cell adhesion, comprise a well-organized functional network with significant roles in health and disease. On the other hand, enzymes such as matrix metalloproteinases and specific glycosidases including heparanase and hyaluronidases contribute to matrix remodeling and affect human health. Several cell processes and functions, among them cell proliferation and survival, migration, differentiation, autophagy, angiogenesis, and immunity regulation are affected by certain matrix components. Structural alterations have been also well associated with disease progression. This guide on the composition and functions of the ECM gives a broad overview of the matrisome, the major ECM macromolecules, and their interaction networks within the ECM and with the cell surface, summarizes their main structural features and their roles in tissue organization and cell functions, and emphasizes the importance of specific ECM constituents in disease development and progression as well as the advances in molecular targeting of ECM to design new therapeutic strategies.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Achilleas D Theocharis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH)/Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Dimitra Manou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Spyros S Skandalis
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Demitrios H Vynios
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Greece
| | - Véronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems- Functional Molecular Systems, Eggenstein-Leopoldshafen, Germany
| | - Sylvie Ricard-Blum
- University of Lyon, UMR 5246, ICBMS, Université Lyon 1, CNRS, Villeurbanne Cedex, France
| | - Christian E H Schmelzer
- Fraunhofer Institute for Microstructure of Materials and Systems IMWS, Halle (Saale), Germany
- Institute of Pharmacy, Faculty of Natural Sciences I, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Laurent Duca
- UMR CNRS 7369 Matrice Extracellulaire et Dynamique Cellulaire (MEDyC), Team 2: Matrix Aging and Vascular Remodelling, Université de Reims Champagne Ardenne (URCA), UFR Sciences Exactes et Naturelles, Reims, France
| | - Madeleine Durbeej
- Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, Sweden
| | - Nikolaos A Afratis
- Department Biological Regulation, Weizmann Institute of Science, Rehovot, Israel
| | - Linda Troeberg
- Norwich Medical School, University of East Anglia, Bob Champion Research and Education Building, Norwich, UK
| | - Marco Franchi
- Department for Life Quality Study, University of Bologna, Rimini, Italy
| | | | - Maurizio Onisto
- Department of Biomedical Sciences, University of Padova, Italy
| |
Collapse
|
13
|
Zakrzewski PK. Canonical TGFβ Signaling and Its Contribution to Endometrial Cancer Development and Progression-Underestimated Target of Anticancer Strategies. J Clin Med 2021; 10:3900. [PMID: 34501347 PMCID: PMC8432036 DOI: 10.3390/jcm10173900] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 08/24/2021] [Accepted: 08/26/2021] [Indexed: 02/08/2023] Open
Abstract
Endometrial cancer is one of the leading gynecological cancers diagnosed among women in their menopausal and postmenopausal age. Despite the progress in molecular biology and medicine, no efficient and powerful diagnostic and prognostic marker is dedicated to endometrial carcinogenesis. The canonical TGFβ pathway is a pleiotropic signaling cascade orchestrating a variety of cellular and molecular processes, whose alterations are responsible for carcinogenesis that originates from different tissue types. This review covers the current knowledge concerning the canonical TGFβ pathway (Smad-dependent) induced by prototypical TGFβ isoforms and the involvement of pathway alterations in the development and progression of endometrial neoplastic lesions. Since Smad-dependent signalization governs opposed cellular processes, such as growth arrest, apoptosis, tumor cells growth and differentiation, as well as angiogenesis and metastasis, TGFβ cascade may act both as a tumor suppressor or tumor promoter. However, the final effect of TGFβ signaling on endometrial cancer cells depends on the cancer disease stage. The multifunctional role of the TGFβ pathway indicates the possible utilization of alterations in the TGFβ cascade as a potential target of novel anticancer strategies.
Collapse
Affiliation(s)
- Piotr K Zakrzewski
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland
| |
Collapse
|
14
|
Zona Pellucida Genes and Proteins: Essential Players in Mammalian Oogenesis and Fertility. Genes (Basel) 2021; 12:genes12081266. [PMID: 34440440 PMCID: PMC8391237 DOI: 10.3390/genes12081266] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/03/2021] [Accepted: 08/10/2021] [Indexed: 12/17/2022] Open
Abstract
All mammalian oocytes and eggs are surrounded by a relatively thick extracellular matrix (ECM), the zona pellucida (ZP), that plays vital roles during oogenesis, fertilization, and preimplantation development. Unlike ECM surrounding somatic cells, the ZP is composed of only a few glycosylated proteins, ZP1–4, that are unique to oocytes and eggs. ZP1–4 have a large region of polypeptide, the ZP domain (ZPD), consisting of two subdomains, ZP-N and ZP-C, separated by a short linker region, that plays an essential role in polymerization of nascent ZP proteins into crosslinked fibrils. Both subdomains adopt immunoglobulin (Ig)-like folds for their 3-dimensional structure. Mouse and human ZP genes are encoded by single-copy genes located on different chromosomes and are highly expressed in the ovary by growing oocytes during late stages of oogenesis. Genes encoding ZP proteins are conserved among mammals, and their expression is regulated by cis-acting sequences located close to the transcription start-site and by the same/similar trans-acting factors. Nascent ZP proteins are synthesized, packaged into vesicles, secreted into the extracellular space, and assembled into long, crosslinked fibrils that have a structural repeat, a ZP2-ZP3 dimer, and constitute the ZP matrix. Fibrils are oriented differently with respect to the oolemma in the inner and outer layers of the ZP. Sequence elements in the ZPD and the carboxy-terminal propeptide of ZP1–4 regulate secretion and assembly of nascent ZP proteins. The presence of both ZP2 and ZP3 is required to assemble ZP fibrils and ZP1 and ZP4 are used to crosslink the fibrils. Inactivation of mouse ZP genes by gene targeting has a detrimental effect on ZP formation around growing oocytes and female fertility. Gene sequence variations in human ZP genes due to point, missense, or frameshift mutations also have a detrimental effect on ZP formation and female fertility. The latter mutations provide additional support for the role of ZPD subdomains and other regions of ZP polypeptide in polymerization of human ZP proteins into fibrils and matrix.
Collapse
|
15
|
Weadick CJ. Molecular Evolutionary Analysis of Nematode Zona Pellucida (ZP) Modules Reveals Disulfide-Bond Reshuffling and Standalone ZP-C Domains. Genome Biol Evol 2021; 12:1240-1255. [PMID: 32426804 PMCID: PMC7456536 DOI: 10.1093/gbe/evaa095] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2020] [Indexed: 12/11/2022] Open
Abstract
Zona pellucida (ZP) modules mediate extracellular protein-protein interactions and contribute to important biological processes including syngamy and cellular morphogenesis. Although some biomedically relevant ZP modules are well studied, little is known about the protein family's broad-scale diversity and evolution. The increasing availability of sequenced genomes from "nonmodel" systems provides a valuable opportunity to address this issue and to use comparative approaches to gain new insights into ZP module biology. Here, through phylogenetic and structural exploration of ZP module diversity across the nematode phylum, I report evidence that speaks to two important aspects of ZP module biology. First, I show that ZP-C domains-which in some modules act as regulators of ZP-N domain-mediated polymerization activity, and which have never before been found in isolation-can indeed be found as standalone domains. These standalone ZP-C domain proteins originated in independent (paralogous) lineages prior to the diversification of extant nematodes, after which they evolved under strong stabilizing selection, suggesting the presence of ZP-N domain-independent functionality. Second, I provide a much-needed phylogenetic perspective on disulfide bond variability, uncovering evidence for both convergent evolution and disulfide-bond reshuffling. This result has implications for our evolutionary understanding and classification of ZP module structural diversity and highlights the usefulness of phylogenetics and diverse sampling for protein structural biology. All told, these findings set the stage for broad-scale (cross-phyla) evolutionary analysis of ZP modules and position Caenorhabditis elegans and other nematodes as important experimental systems for exploring the evolution of ZP modules and their constituent domains.
Collapse
|
16
|
Kschonsak M, Rougé L, Arthur CP, Hoangdung H, Patel N, Kim I, Johnson MC, Kraft E, Rohou AL, Gill A, Martinez-Martin N, Payandeh J, Ciferri C. Structures of HCMV Trimer reveal the basis for receptor recognition and cell entry. Cell 2021; 184:1232-1244.e16. [PMID: 33626330 DOI: 10.1016/j.cell.2021.01.036] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/10/2021] [Accepted: 01/21/2021] [Indexed: 01/19/2023]
Abstract
Human cytomegalovirus (HCMV) infects the majority of the human population and represents the leading viral cause of congenital birth defects. HCMV utilizes the glycoproteins gHgLgO (Trimer) to bind to platelet-derived growth factor receptor alpha (PDGFRα) and transforming growth factor beta receptor 3 (TGFβR3) to gain entry into multiple cell types. This complex is targeted by potent neutralizing antibodies and represents an important candidate for therapeutics against HCMV. Here, we determine three cryogenic electron microscopy (cryo-EM) structures of the trimer and the details of its interactions with four binding partners: the receptor proteins PDGFRα and TGFβR3 as well as two broadly neutralizing antibodies. Trimer binding to PDGFRα and TGFβR3 is mutually exclusive, suggesting that they function as independent entry receptors. In addition, Trimer-PDGFRα interaction has an inhibitory effect on PDGFRα signaling. Our results provide a framework for understanding HCMV receptor engagement, neutralization, and the development of anti-viral strategies against HCMV.
Collapse
Affiliation(s)
- Marc Kschonsak
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA.
| | - Lionel Rougé
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | | | - Ho Hoangdung
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Nidhi Patel
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Ingrid Kim
- Department of Antibody Engineering, Genentech, South San Francisco, CA 94080, USA
| | - Matthew C Johnson
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Edward Kraft
- Department of BioMolecular Resources, Genentech, South San Francisco, CA 94080, USA
| | - Alexis L Rohou
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA
| | - Avinash Gill
- Department of Antibody Engineering, Genentech, South San Francisco, CA 94080, USA
| | - Nadia Martinez-Martin
- Department of Microchemistry, Proteomics and Lipidomics Department, Genentech, South San Francisco, CA 94080, USA.
| | - Jian Payandeh
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA; Department of Antibody Engineering, Genentech, South San Francisco, CA 94080, USA.
| | - Claudio Ciferri
- Department of Structural Biology, Genentech, South San Francisco, CA 94080, USA.
| |
Collapse
|
17
|
Cohen JD, Bermudez JG, Good MC, Sundaram MV. A C. elegans Zona Pellucida domain protein functions via its ZPc domain. PLoS Genet 2020; 16:e1009188. [PMID: 33141826 PMCID: PMC7665627 DOI: 10.1371/journal.pgen.1009188] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 11/13/2020] [Accepted: 10/12/2020] [Indexed: 01/24/2023] Open
Abstract
Zona Pellucida domain (ZP) proteins are critical components of the body's external-most protective layers, apical extracellular matrices (aECMs). Although their loss or dysfunction is associated with many diseases, it remains unclear how ZP proteins assemble in aECMs. Current models suggest that ZP proteins polymerize via their ZPn subdomains, while ZPc subdomains modulate ZPn behavior. Using the model organism C. elegans, we investigated the aECM assembly of one ZP protein, LET-653, which shapes several tubes. Contrary to prevailing models, we find that LET-653 localizes and functions via its ZPc domain. Furthermore, we show that ZPc domain function requires cleavage at the LET-653 C-terminus, likely in part to relieve inhibition of the ZPc by the ZPn domain, but also to promote some other aspect of ZPc domain function. In vitro, the ZPc, but not ZPn, domain bound crystalline aggregates. These data offer a new model for ZP function whereby the ZPc domain is primarily responsible for matrix incorporation and tissue shaping.
Collapse
Affiliation(s)
- Jennifer D. Cohen
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Jessica G. Bermudez
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Matthew C. Good
- Department of Bioengineering, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Cell and Developmental Biology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Meera V. Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
18
|
Yan W. An Interview with Dr. Teresa K Woodruff. Biol Reprod 2020. [DOI: 10.1093/biolre/ioaa094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Wei Yan
- The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
19
|
Abstract
The zona pellucida (ZP) is an extracellular matrix that surrounds all mammalian oocytes, eggs, and early embryos and plays vital roles during oogenesis, fertilization, and preimplantation development. The ZP is composed of three or four glycosylated proteins, ZP1–4, that are synthesized, processed, secreted, and assembled into long, cross-linked fibrils by growing oocytes. ZP proteins have an immunoglobulin-like three-dimensional structure and a ZP domain that consists of two subdomains, ZP-N and ZP-C, with ZP-N of ZP2 and ZP3 required for fibril assembly. A ZP2–ZP3 dimer is located periodically along ZP fibrils that are cross-linked by ZP1, a protein with a proline-rich N terminus. Fibrils in the inner and outer regions of the ZP are oriented perpendicular and parallel to the oolemma, respectively, giving the ZP a multilayered appearance. Upon fertilization of eggs, modification of ZP2 and ZP3 results in changes in the ZP's physical and biological properties that have important consequences. Certain structural features of ZP proteins suggest that they may be amyloid-like proteins.
Collapse
Affiliation(s)
- Eveline S. Litscher
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;,
| | - Paul M. Wassarman
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;,
| |
Collapse
|
20
|
Woodruff TK. Lessons from bioengineering the ovarian follicle: a personal perspective. Reproduction 2020; 158:F113-F126. [PMID: 31846436 DOI: 10.1530/rep-19-0190] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 07/15/2019] [Indexed: 12/14/2022]
Abstract
The ovarian follicle and its maturation captivated my imagination and inspired my scientific journey - what we know now about this remarkable structure is captured in this invited review. In the past decade, our knowledge of the ovarian follicle expanded dramatically as cross-disciplinary collaborations brought new perspectives to bear, ultimately leading to the development of extragonadal follicles as model systems with significant clinical implications. Follicle maturation in vitro in an 'artificial' ovary became possible by learning what the follicle is fundamentally and autonomously capable of - which turns out to be quite a lot. Progress in understanding and harnessing follicle biology has been aided by engineers and materials scientists who created hardware that enables tissue function for extended periods of time. The EVATAR system supports extracorporeal ovarian function in an engineered environment that mimics the endocrine environment of the reproductive tract. Finally, applying the tools of inorganic chemistry, we discovered that oocytes require zinc to mature over time - a truly new aspect of follicle biology with no antecedent other than the presence of zinc in sperm. Drawing on the tools and ideas from the fields of bioengineering, materials science and chemistry unlocked follicle biology in ways that we could not have known or even predicted. Similarly, how today's basic science discoveries regarding ovarian follicle maturation are translated to improve the experience of tomorrow's patients is yet to be determined.
Collapse
Affiliation(s)
- Teresa K Woodruff
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
21
|
Kim SK, Henen MA, Hinck AP. Structural biology of betaglycan and endoglin, membrane-bound co-receptors of the TGF-beta family. Exp Biol Med (Maywood) 2019; 244:1547-1558. [PMID: 31601110 PMCID: PMC6920675 DOI: 10.1177/1535370219881160] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Betaglycan and endoglin, membrane-bound co-receptors of the TGF-β family, are required to mediate the signaling of a select subset of TGF-β family ligands, TGF-β2 and InhA, and BMP-9 and BMP-10, respectively. Previous biochemical and biophysical methods suggested alternative modes of ligand binding might be responsible for these co-receptors to selectively recognize and potentiate the functions of their ligands, yet the molecular details were lacking. Recent progress determining structures of betaglycan and endoglin, both alone and as bound to their cognate ligands, is presented herein. The structures reveal relatively minor, but very significant structural differences that lead to entirely different modes of ligand binding. The different modes of binding nonetheless share certain commonalities, such as multivalency, which imparts the co-receptors with very high affinity for their cognate ligands, but at the same time provides a mechanism for release by stepwise binding of the signaling receptors, both of which are essential for their functions.
Collapse
Affiliation(s)
- Sun Kyung Kim
- Department of Structural Biology, University of Pittsburgh,
Pittsburgh, PA 15260, USA
- Department of Biochemistry and Biophysics, University California
San Francisco, San Francisco, CA 94158, USA
| | - Morkos A Henen
- Department of Structural Biology, University of Pittsburgh,
Pittsburgh, PA 15260, USA
- Faculty of Pharmacy, Mansoura University, Mansoura 35516,
Egypt
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh,
Pittsburgh, PA 15260, USA
| |
Collapse
|
22
|
Kim DK, Kim JA, Park J, Niazi A, Almishaal A, Park S. The release of surface-anchored α-tectorin, an apical extracellular matrix protein, mediates tectorial membrane organization. SCIENCE ADVANCES 2019; 5:eaay6300. [PMID: 31807709 PMCID: PMC6881170 DOI: 10.1126/sciadv.aay6300] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 10/02/2019] [Indexed: 06/10/2023]
Abstract
The tectorial membrane (TM) is an apical extracellular matrix (ECM) that hovers over the cochlear sensory epithelium and plays an essential role in auditory transduction. The TM forms facing the luminal endolymph-filled space and exhibits complex ultrastructure. Contrary to the current extracellular assembly model, which posits that secreted collagen fibrils and ECM components self-arrange in the extracellular space, we show that surface tethering of α-tectorin (TECTA) via a glycosylphosphatidylinositol anchor is essential to prevent diffusion of secreted TM components. In the absence of surface-tethered TECTA, collagen fibrils aggregate randomly and fail to recruit TM glycoproteins. Conversely, conversion of TECTA into a transmembrane form results in a layer of collagens on the epithelial surface that fails to form a multilayered structure. We propose a three-dimensional printing model for TM morphogenesis: A new layer of ECM is printed on the cell surface concomitant with the release of a preestablished layer to generate the multilayered TM.
Collapse
Affiliation(s)
- Dong-Kyu Kim
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ju Ang Kim
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Joosang Park
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ava Niazi
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| | - Ali Almishaal
- Department of Communication Sciences and Disorders, University of Utah, Salt Lake City, UT 84112, USA
| | - Sungjin Park
- Department of Neurobiology and Anatomy, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
| |
Collapse
|
23
|
Nopvichai C, Pongkorpsakol P, Wongkrasant P, Wangpaiboon K, Charoenwongpaiboon T, Ito K, Muanprasat C, Pichyangkura R. Galactomannan Pentasaccharide Produced from Copra Meal Enhances Tight Junction Integration of Epithelial Tissue through Activation of AMPK. Biomedicines 2019; 7:biomedicines7040081. [PMID: 31614968 PMCID: PMC6966651 DOI: 10.3390/biomedicines7040081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/09/2019] [Accepted: 10/11/2019] [Indexed: 12/24/2022] Open
Abstract
Mannan oligosaccharide (MOS) is well-known as an effective fed supplement for livestock to increase their nutrients absorption and health status. Pentasaccharide of mannan (MOS5) was reported as a molecule that possesses the ability to increase tight junction of epithelial tissue, but the structure and mechanism of action remains undetermined. In this study, the mechanism of action and structure of MOS5 were investigated. T84 cells were cultured and treated with MOS5 compared with vehicle and compound C, a 5′-adenosine monophosphate-activated protein kinase (AMPK) inhibitor. The results demonstrated that the ability of MOS5 to increase tight junction integration was inhibited in the presence of dorsomorphine (compound C). Phosphorylation level of AMPK was elevated in MOS5 treated group as determined by Western blot analysis. Determination of MOS5 structure was performed using enzymatic mapping together with 1H, 13C NMR, and 2D-NMR analysis. The results demonstrated that the structure of MOS5 is a β-(1,4)-mannotetraose with α-(1,6)-galactose attached at the second mannose unit from non-reducing end.
Collapse
Affiliation(s)
- Chatchai Nopvichai
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | - Pawin Pongkorpsakol
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Preedajit Wongkrasant
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | - Karan Wangpaiboon
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| | | | - Kazuo Ito
- Graduate School of Science, Osaka City University, Osaka 558-8585, Japan.
| | - Chatchai Muanprasat
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
- Division of Preclinical Sciences, Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok 10540, Thailand.
| | - Rath Pichyangkura
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
24
|
Kim SK, Whitley MJ, Krzysiak TC, Hinck CS, Taylor AB, Zwieb C, Byeon CH, Zhou X, Mendoza V, López-Casillas F, Furey W, Hinck AP. Structural Adaptation in Its Orphan Domain Engenders Betaglycan with an Alternate Mode of Growth Factor Binding Relative to Endoglin. Structure 2019; 27:1427-1442.e4. [PMID: 31327662 DOI: 10.1016/j.str.2019.06.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/11/2019] [Accepted: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Betaglycan (BG) and endoglin (ENG), homologous co-receptors of the TGF-β family, potentiate the signaling activity of TGF-β2 and inhibin A, and BMP-9 and BMP-10, respectively. BG exists as monomer and forms 1:1 growth factor (GF) complexes, while ENG exists as a dimer and forms 2:1 GF complexes. Herein, the structure of the BG orphan domain (BGO) reveals an insertion that blocks the region that the endoglin orphan domain (ENGO) uses to bind BMP-9, preventing it from binding in the same manner. Using binding studies with domain-deleted forms of TGF-β and BGO, as well as small-angle X-ray scattering data, BGO is shown to bind its cognate GF in an entirely different manner compared with ENGO. The alternative interfaces likely engender BG and ENG with the ability to selectively bind and target their cognate GFs in a unique temporal-spatial manner, without interfering with one another or other TGF-β family GFs.
Collapse
Affiliation(s)
- Sun Kyung Kim
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA; Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Matthew J Whitley
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Troy C Krzysiak
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Cynthia S Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Alexander B Taylor
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA; X-ray Crystallography Core Laboratory, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Christian Zwieb
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | - Chang-Hyeock Byeon
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Xiaohong Zhou
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA
| | - Valentín Mendoza
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - Fernando López-Casillas
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ciudad de México 04510, México
| | - William Furey
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Andrew P Hinck
- Department of Structural Biology, University of Pittsburgh School of Medicine, Biomedical Science Tower 3, Room 2051, 3501 Fifth Avenue, Pittsburgh, PA 15260, USA.
| |
Collapse
|
25
|
Henen MA, Mahlawat P, Zwieb C, Kodali RB, Hinck CS, Hanna RD, Krzysiak TC, Ilangovan U, Cano KE, Hinck G, Vonberg M, McCabe M, Hinck AP. TGF-β2 uses the concave surface of its extended finger region to bind betaglycan's ZP domain via three residues specific to TGF-β and inhibin-α. J Biol Chem 2019; 294:3065-3080. [PMID: 30598510 PMCID: PMC6398128 DOI: 10.1074/jbc.ra118.005210] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/04/2018] [Indexed: 01/17/2023] Open
Abstract
Betaglycan (BG) is a membrane-bound co-receptor of the TGF-β family that selectively binds transforming growth factor-β (TGF-β) isoforms and inhibin A (InhA) to enable temporal-spatial patterns of signaling essential for their functions in vivo Here, using NMR titrations of methyl-labeled TGF-β2 with BG's C-terminal binding domain, BGZP-C, and surface plasmon resonance binding measurements with TGF-β2 variants, we found that the BGZP-C-binding site on TGF-β2 is located on the inner surface of its extended finger region. Included in this binding site are Ile-92, Lys-97, and Glu-99, which are entirely or mostly specific to the TGF-β isoforms and the InhA α-subunit, but they are unconserved in other TGF-β family growth factors (GFs). In accord with the proposed specificity-determining role of these residues, BG bound bone morphogenetic protein 2 (BMP-2) weakly or not at all, and TGF-β2 variants with the corresponding residues from BMP-2 bound BGZP-C more weakly than corresponding alanine variants. The BGZP-C-binding site on InhA previously was reported to be located on the outside of the extended finger region, yet at the same time to include Ser-112 and Lys-119, homologous to TGF-β2 Ile-92 and Lys-97, on the inside of the fingers. Therefore, it is likely that both TGF-β2 and InhA bind BGZP-C through a site on the inside of their extended finger regions. Overall, these results identify the BGZP-C-binding site on TGF-β2 and shed light on the specificity of BG for select TGF-β-type GFs and the mechanisms by which BG influences their signaling.
Collapse
Affiliation(s)
- Morkos A Henen
- From the Departments of Structural Biology and
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Pardeep Mahlawat
- From the Departments of Structural Biology and
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Christian Zwieb
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | | | - Cynthia S Hinck
- From the Departments of Structural Biology and
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Ramsey D Hanna
- Chemistry, University of Pittsburgh, Pittsburgh, Pennsylvania 15260 and
| | | | - Udayar Ilangovan
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Kristin E Cano
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Garrett Hinck
- From the Departments of Structural Biology and
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Machell Vonberg
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Megan McCabe
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| | - Andrew P Hinck
- From the Departments of Structural Biology and
- the Department of Biochemistry and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229-3900
| |
Collapse
|
26
|
Cohen JD, Flatt KM, Schroeder NE, Sundaram MV. Epithelial Shaping by Diverse Apical Extracellular Matrices Requires the Nidogen Domain Protein DEX-1 in Caenorhabditis elegans. Genetics 2019; 211:185-200. [PMID: 30409789 PMCID: PMC6325709 DOI: 10.1534/genetics.118.301752] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/05/2018] [Indexed: 02/06/2023] Open
Abstract
The body's external surfaces and the insides of biological tubes, like the vascular system, are lined by a lipid-, glycoprotein-, and glycosaminoglycan-rich apical extracellular matrix (aECM). aECMs are the body's first line of defense against infectious agents and promote tissue integrity and morphogenesis, but are poorly described relative to basement membranes and stromal ECMs. While some aECM components, such as zona pellucida (ZP) domain proteins, have been identified, little is known regarding the overall composition of the aECM or the mechanisms by which different aECM components work together to shape epithelial tissues. In Caenorhabditis elegans, external epithelia develop in the context of an ill-defined ZP-containing aECM that precedes secretion of the collagenous cuticle. C. elegans has 43 genes that encode at least 65 unique ZP proteins, and we show that some of these comprise distinct precuticle aECMs in the embryo. Previously, the nidogen- and EGF-domain protein DEX-1 was shown to anchor dendrites to the C. elegans nose tip in concert with the ZP protein DYF-7 Here, we identified a new, strong loss-of-function allele of dex-1, cs201dex-1 mutants die as L1 larvae and have a variety of tissue distortion phenotypes, including excretory defects, pharyngeal ingression, alae defects, and a short and fat body shape, that strongly resemble those of genes encoding ZP proteins. DEX-1 localizes to ZP-containing aECMs in the tissues that show defects in dex-1 mutants. Our studies suggest that DEX-1 is a component of multiple distinct embryonic aECMs that shape developing epithelia, and a potential partner of multiple ZP proteins.
Collapse
Affiliation(s)
- Jennifer D Cohen
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| | - Kristen M Flatt
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Illinois 61801-4730
| | - Nathan E Schroeder
- Program in Neuroscience, University of Illinois at Urbana-Champaign, Illinois 61801-4730
- Department of Crop Sciences, University of Illinois at Urbana-Champaign, Illinois 61801-4730
| | - Meera V Sundaram
- Department of Genetics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104
| |
Collapse
|
27
|
Abstract
The egg coat, an extracellular matrix made up of glycoprotein filaments, plays a key role in animal fertilization by acting as a gatekeeper for sperm. Egg coat components polymerize using a common zona pellucida (ZP) "domain" module that consists of two related immunoglobulin-like domains, called ZP-N and ZP-C. The ZP module has also been recognized in a large number of other secreted proteins with different biological functions, whose mutations are linked to severe human diseases. During the last decade, tremendous progress has been made toward understanding the atomic architecture of the ZP module and the structural basis of its polymerization. Moreover, sperm-binding regions at the N-terminus of mollusk and mammalian egg coat subunits were found to consist of domain repeats that also adopt a ZP-N fold. This discovery revealed an unexpected link between invertebrate and vertebrate fertilization and led to the first structure of an egg coat-sperm protein recognition complex. In this review we summarize these exciting findings, discuss their functional implications, and outline future challenges that must be addressed in order to develop a comprehensive view of this family of biomedically important extracellular molecules.
Collapse
Affiliation(s)
- Marcel Bokhove
- Department of Biosciences and Nutrition & Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Luca Jovine
- Department of Biosciences and Nutrition & Center for Innovative Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
28
|
Abstract
An ovulated egg of vertebrates is surrounded by unique extracellular matrix, the egg coat or zona pellucida, playing important roles in fertilization and early development. The vertebrate egg coat is composed of two to six zona pellucida (ZP) glycoproteins that are characterized by the evolutionarily conserved ZP-domain module and classified into six subfamilies based on phylogenetic analyses. Interestingly, investigations of biochemical and functional features of the ZP glycoproteins show that the roles of each ZP-glycoprotein family member in the egg-coat formation and the egg-sperm interactions seemingly vary across vertebrates. This might be one reason why comprehensive understandings of the molecular basis of either architecture or physiological functions of egg coat still remain elusive despite more than 3 decades of intensive investigations. In this chapter, an overview of avian egg focusing on the oogenesis are provided in the first section, and unique features of avian egg coat, i.e., perivitelline layer, including the morphology, biogenesis pathway, and physiological functions are discussed mainly on chicken and quail in terms of the characteristics of ZP glycoproteins in the following sections. In addition, these features of avian egg coat are compared to mammalian zona pellucida, from the viewpoint that the structural and functional varieties of ZP glycoproteins might be associated with the evolutionary adaptation to their reproductive strategies. By comparing the egg coat of birds and mammals whose reproductive strategies are largely different, new insights into the molecular mechanisms of vertebrate egg-sperm interactions might be provided.
Collapse
Affiliation(s)
- Hiroki Okumura
- Department of Applied Biological Chemistry, Faculty of Agriculture, Meijo University, Nagoya, Japan.
| |
Collapse
|
29
|
Saito T, Bokhove M, Croci R, Zamora-Caballero S, Han L, Letarte M, de Sanctis D, Jovine L. Structural Basis of the Human Endoglin-BMP9 Interaction: Insights into BMP Signaling and HHT1. Cell Rep 2018; 19:1917-1928. [PMID: 28564608 PMCID: PMC5464963 DOI: 10.1016/j.celrep.2017.05.011] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/30/2017] [Accepted: 05/02/2017] [Indexed: 11/25/2022] Open
Abstract
Endoglin (ENG)/CD105 is an essential endothelial cell co-receptor of the transforming growth factor β (TGF-β) superfamily, mutated in hereditary hemorrhagic telangiectasia type 1 (HHT1) and involved in tumor angiogenesis and preeclampsia. Here, we present crystal structures of the ectodomain of human ENG and its complex with the ligand bone morphogenetic protein 9 (BMP9). BMP9 interacts with a hydrophobic surface of the N-terminal orphan domain of ENG, which adopts a new duplicated fold generated by circular permutation. The interface involves residues mutated in HHT1 and overlaps with the epitope of tumor-suppressing anti-ENG monoclonal TRC105. The structure of the C-terminal zona pellucida module suggests how two copies of ENG embrace homodimeric BMP9, whose binding is compatible with ligand recognition by type I but not type II receptors. These findings shed light on the molecular basis of the BMP signaling cascade, with implications for future therapeutic interventions in this fundamental pathway. Crystal structures of human ENG and its complex with BMP9 were determined The orphan domain of ENG adopts a fold that explains the effect of HHT1 mutations ZP module-mediated dimerization of ENG creates a clamp that secures homodimeric BMP9 ENG-bound BMP9 can interact with the ALK1 receptor but not the ActRIIB receptor
Collapse
Affiliation(s)
- Takako Saito
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Marcel Bokhove
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Romina Croci
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Sara Zamora-Caballero
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Ling Han
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden
| | - Michelle Letarte
- Molecular Medicine, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, University of Toronto, Toronto, ON M5G 0A4, Canada; Department of Immunology, University of Toronto, Toronto, ON M5G 0A4, Canada
| | | | - Luca Jovine
- Department of Biosciences and Nutrition and Center for Innovative Medicine, Karolinska Institutet, Huddinge 14183, Sweden.
| |
Collapse
|
30
|
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: 143] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar 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
|
31
|
Jenkins LM, Horst B, Lancaster CL, Mythreye K. Dually modified transmembrane proteoglycans in development and disease. Cytokine Growth Factor Rev 2017; 39:124-136. [PMID: 29291930 DOI: 10.1016/j.cytogfr.2017.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Accepted: 12/20/2017] [Indexed: 12/11/2022]
Abstract
Aberrant cell signaling in response to secreted growth factors has been linked to the development of multiple diseases, including cancer. As such, understanding mechanisms that control growth factor availability and receptor-growth factor interaction is vital. Dually modified transmembrane proteoglycans (DMTPs), which are classified as cell surface macromolecules composed of a core protein decorated with covalently linked heparan sulfated (HS) and/or chondroitin sulfated (CS) glycosaminoglycan (GAG) chains, provide one type of regulatory mechanism. Specifically, DMTPs betaglycan and syndecan-1 (SDC1) play crucial roles in modulating key cell signaling pathways, such as Wnt, transforming growth factor-β and fibroblast growth factor signaling, to affect epithelial cell biology and cancer progression. This review outlines current and potential functions for betaglycan and SDC1, with an emphasis on comparing individual roles for HS and CS modified DMTPs. We highlight the mutual dependence of DMTPs' GAG chains and core proteins and provide comprehensive knowledge on how these DMTPs, through regulation of ligand availability and receptor internalization, control cell signaling pathways involved in development and disease.
Collapse
Affiliation(s)
- Laura M Jenkins
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA.
| | - Ben Horst
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA.
| | - Carly L Lancaster
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA.
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, USA; Department of Drug Discovery and Biomedical Sciences, South Carolina College of Pharmacy, University of South Carolina, Columbia, SC, 29208, USA.
| |
Collapse
|
32
|
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: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar 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
|
33
|
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
|
34
|
Gill HK, Cohen JD, Ayala-Figueroa J, Forman-Rubinsky R, Poggioli C, Bickard K, Parry JM, Pu P, Hall DH, Sundaram MV. Integrity of Narrow Epithelial Tubes in the C. elegans Excretory System Requires a Transient Luminal Matrix. PLoS Genet 2016; 12:e1006205. [PMID: 27482894 PMCID: PMC4970718 DOI: 10.1371/journal.pgen.1006205] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 06/28/2016] [Indexed: 02/07/2023] Open
Abstract
Most epithelial cells secrete a glycoprotein-rich apical extracellular matrix that can have diverse but still poorly understood roles in development and physiology. Zona Pellucida (ZP) domain glycoproteins are common constituents of these matrices, and their loss in humans is associated with a number of diseases. Understanding of the functions, organization and regulation of apical matrices has been hampered by difficulties in imaging them both in vivo and ex vivo. We identified the PAN-Apple, mucin and ZP domain glycoprotein LET-653 as an early and transient apical matrix component that shapes developing epithelia in C. elegans. LET-653 has modest effects on shaping of the vulva and epidermis, but is essential to prevent lumen fragmentation in the very narrow, unicellular excretory duct tube. We were able to image the transient LET-653 matrix by both live confocal imaging and transmission electron microscopy. Structure/function and fluorescence recovery after photobleaching studies revealed that LET-653 exists in two separate luminal matrix pools, a loose fibrillar matrix in the central core of the lumen, to which it binds dynamically via its PAN domains, and an apical-membrane-associated matrix, to which it binds stably via its ZP domain. The PAN domains are both necessary and sufficient to confer a cyclic pattern of duct lumen localization that precedes each molt, while the ZP domain is required for lumen integrity. Ectopic expression of full-length LET-653, but not the PAN domains alone, could expand lumen diameter in the developing gut tube, where LET-653 is not normally expressed. Together, these data support a model in which the PAN domains regulate the ability of the LET-653 ZP domain to interact with other factors at the apical membrane, and this ZP domain interaction promotes expansion and maintenance of lumen diameter. These data identify a transient apical matrix component present prior to cuticle secretion in C. elegans, demonstrate critical roles for this matrix component in supporting lumen integrity within narrow bore tubes such as those found in the mammalian microvasculature, and reveal functional importance of the evolutionarily conserved ZP domain in this tube protecting activity. Most organs in the body are made up of networks of tubes that transport fluids or gases. These tubes come in many different sizes and shapes, with some narrow capillaries being only one cell in diameter. As tubes develop and take their final shapes, they secrete various glycoproteins into their hollow interior or lumen. The functions of these luminal proteins are not well understood, but there is increasing evidence that they are important for lumen shaping and that their loss can contribute to diseases such as cardiovascular disease and chronic kidney disease. Through studies of the nematode C. elegans, we identified a luminal glycoprotein, LET-653, that is transiently expressed in multiple developing tube types but is particularly critical to maintain integrity of the narrowest, unicellular tubes. We identified protein domains that direct LET-653 to specific apical matrix compartments and mediate its oscillatory pattern of lumen localization. Furthermore, we showed that the LET-653 tube-protecting activity depends on a Zona Pellucida (ZP) domain similar to that found in the mammalian egg-coat and in many other luminal or sensory matrix proteins involved in human disease.
Collapse
Affiliation(s)
- Hasreet K. Gill
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jennifer D. Cohen
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jesus Ayala-Figueroa
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Rachel Forman-Rubinsky
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Corey Poggioli
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Kevin Bickard
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Jean M. Parry
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- Department of Biology, Georgian Court University, Lakewood, New Jersey, United States of America
| | - Pu Pu
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - David H. Hall
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Meera V. Sundaram
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
35
|
A structured interdomain linker directs self-polymerization of human uromodulin. Proc Natl Acad Sci U S A 2016; 113:1552-7. [PMID: 26811476 DOI: 10.1073/pnas.1519803113] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Uromodulin (UMOD)/Tamm-Horsfall protein, the most abundant human urinary protein, plays a key role in chronic kidney diseases and is a promising therapeutic target for hypertension. Via its bipartite zona pellucida module (ZP-N/ZP-C), UMOD forms extracellular filaments that regulate kidney electrolyte balance and innate immunity, as well as protect against renal stones. Moreover, salt-dependent aggregation of UMOD filaments in the urine generates a soluble molecular net that captures uropathogenic bacteria and facilitates their clearance. Despite the functional importance of its homopolymers, no structural information is available on UMOD and how it self-assembles into filaments. Here, we report the crystal structures of polymerization regions of human UMOD and mouse ZP2, an essential sperm receptor protein that is structurally related to UMOD but forms heteropolymers. The structure of UMOD reveals that an extensive hydrophobic interface mediates ZP-N domain homodimerization. This arrangement is required for filament formation and is directed by an ordered ZP-N/ZP-C linker that is not observed in ZP2 but is conserved in the sequence of deafness/Crohn's disease-associated homopolymeric glycoproteins α-tectorin (TECTA) and glycoprotein 2 (GP2). Our data provide an example of how interdomain linker plasticity can modulate the function of structurally similar multidomain proteins. Moreover, the architecture of UMOD rationalizes numerous pathogenic mutations in both UMOD and TECTA genes.
Collapse
|
36
|
Jin Y, Wang H, Han W, Lu J, Chu P, Han S, Ni X, Ning B, Yu D, Guo Y. Single nucleotide polymorphism rs11669203 in TGFBR3L is associated with the risk of neuroblastoma in a Chinese population. Tumour Biol 2015; 37:3739-47. [PMID: 26468016 DOI: 10.1007/s13277-015-4192-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022] Open
Abstract
With a primary mortality, neuroblastoma (NB) is the most common extracranial solid tumor in childhood. Amplification of the MYCN (v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog) oncogene is observed in 20-30 % of NB cases, a feature which also characterizes a highly aggressive subtype of the disease. However, the systematic study of association between single nucleotide polymorphisms (SNPs) in MYCN-regulated genes and the risk of NB has not been investigated. In the current study, we scanned a set of 16 SNPs located within known or predicted MYCN binding sites in a cohort of 247 patients of Chinese origin with neuroblastic family tumors, including neuroblastoma (NB), ganglioneuroma (GN), and ganglioneuroblastoma (GNB), and in 290 cancer-free controls to determine whether any of the tested SNPs are associated with neuroblastic family tumors. We found that the rs11669203 G>C polymorphism, located in TGFBR3L promoter, is significantly associated with the risk of NB. Further, we found that this association is site specific to adrenal NB compared to non-adrenal NB. In addition, transcriptome analysis indicated that increased expression of TGFBR3L is strongly correlated with poor survival. The SNP rs11669203 located at the MYCN binding site of TGFBR3L is significantly associated with elevated risk of NB, and abnormal MYCN-regulated TGFBR3L expression may contribute to NB oncogenesis.
Collapse
Affiliation(s)
- Yaqiong Jin
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Huanmin Wang
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Wei Han
- Department of Surgical Oncology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Ping Chu
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Shujing Han
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Xin Ni
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
- Department of Head and Neck Surgery, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Baitang Ning
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, USA
| | - Dianke Yu
- National Center for Toxicological Research, US Food and Drug Administration, Jefferson, USA.
| | - Yongli Guo
- Beijing Key Laboratory for Pediatric Diseases of Otolaryngology, Head and Neck Surgery, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
37
|
Zhu J, Mishra RK, Schiltz GE, Makanji Y, Scheidt KA, Mazar AP, Woodruff TK. Virtual High-Throughput Screening To Identify Novel Activin Antagonists. J Med Chem 2015; 58:5637-48. [PMID: 26098096 DOI: 10.1021/acs.jmedchem.5b00753] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Activin belongs to the TGFβ superfamily, which is associated with several disease conditions, including cancer-related cachexia, preterm labor with delivery, and osteoporosis. Targeting activin and its related signaling pathways holds promise as a therapeutic approach to these diseases. A small-molecule ligand-binding groove was identified in the interface between the two activin βA subunits and was used for a virtual high-throughput in silico screening of the ZINC database to identify hits. Thirty-nine compounds without significant toxicity were tested in two well-established activin assays: FSHβ transcription and HepG2 cell apoptosis. This screening workflow resulted in two lead compounds: NUCC-474 and NUCC-555. These potential activin antagonists were then shown to inhibit activin A-mediated cell proliferation in ex vivo ovary cultures. In vivo testing showed that our most potent compound (NUCC-555) caused a dose-dependent decrease in FSH levels in ovariectomized mice. The Blitz competition binding assay confirmed target binding of NUCC-555 to the activin A:ActRII that disrupts the activin A:ActRII complex's binding with ALK4-ECD-Fc in a dose-dependent manner. The NUCC-555 also specifically binds to activin A compared with other TGFβ superfamily member myostatin (GDF8). These data demonstrate a new in silico-based strategy for identifying small-molecule activin antagonists. Our approach is the first to identify a first-in-class small-molecule antagonist of activin binding to ALK4, which opens a completely new approach to inhibiting the activity of TGFβ receptor superfamily members. in addition, the lead compound can serve as a starting point for lead optimization toward the goal of a compound that may be effective in activin-mediated diseases.
Collapse
Affiliation(s)
- Jie Zhu
- †Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 East Superior Street, Lurie 10-250, Chicago, Illinois 60611, United States.,‡Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208, United States
| | - Rama K Mishra
- §Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Gary E Schiltz
- §Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Yogeshwar Makanji
- †Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 East Superior Street, Lurie 10-250, Chicago, Illinois 60611, United States
| | - Karl A Scheidt
- §Center for Molecular Innovation and Drug Discovery, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States.,⊥Department of Chemistry, Northwestern University, Evanston, 60208, Illinois, United States.,∥Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, United States
| | - Andrew P Mazar
- ∥Department of Pharmacology, Northwestern University, Chicago, Illinois 60611, United States.,□Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| | - Teresa K Woodruff
- †Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, 303 East Superior Street, Lurie 10-250, Chicago, Illinois 60611, United States.,‡Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208, United States.,□Chemistry of Life Processes Institute, Northwestern University, 2170 Campus Drive, Evanston, Illinois 60208, United States
| |
Collapse
|
38
|
Okumura H, Sato T, Sakuma R, Fukushima H, Matsuda T, Ujita M. Identification of distinctive interdomain interactions among ZP-N, ZP-C and other domains of zona pellucida glycoproteins underlying association of chicken egg-coat matrix. FEBS Open Bio 2015; 5:454-65. [PMID: 26106520 PMCID: PMC4475693 DOI: 10.1016/j.fob.2015.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 05/15/2015] [Accepted: 05/22/2015] [Indexed: 12/12/2022] Open
Abstract
Chicken ZP1 and ZP3 assemble through strong interactions between their ZP-C domains. ZP-C domains of chicken ZP1 and ZP3 are deeply embedded in the egg-coat matrix. Chicken ZP1 forms a homocomplex through non-covalent interaction between repeat domains. Chicken ZPD is deposited on the interstices of ZP1–ZP3 matrix in the egg coat. We propose a model for the architecture of chicken egg-coat matrix from these results.
The vertebrate egg coat, including mammalian zona pellucida, is an oocyte-specific extracellular matrix comprising two to six zona pellucida (ZP) glycoproteins. The egg coat plays important roles in fertilization, especially in species-specific interactions with sperm to induce the sperm acrosome reaction and to form the block to polyspermy. It is suggested that the physiological functions of the egg coat are mediated and/or regulated coordinately by peptide and carbohydrate moieties of the ZP glycoproteins that are spatially arranged in the egg coat, whereas a comprehensive understanding of the architecture of vertebrate egg-coat matrix remains elusive. Here, we deduced the orientations and/or distributions of chicken ZP glycoproteins, ZP1, ZP3 and ZPD, in the egg-coat matrix by confocal immunofluorescent microscopy, and in the ZP1–ZP3 complexes generated in vitro by co-immunoprecipitation assays. We further confirmed interdomain interactions of the ZP glycoproteins by far-Western blot analyses of the egg-coat proteins and pull-down assays of ZP1 in the serum, using recombinant domains of ZP glycoproteins as probes. Our results suggest that the ZP1 and ZP3 bind through their ZP-C domains to form the ZP1–ZP3 complexes and fibrils, which are assembled into bundles through interactions between the repeat domains of ZP1 to form the ZP1–ZP3 matrix, and that the ZPD molecules self-associate and bind to the ZP1–ZP3 matrix through its ZP-N and ZP-C domains to form the egg-coat matrix. Based on these results, we propose a tentative model for the architecture of the chicken egg-coat matrix that might be applicable to other vertebrate ones.
Collapse
Key Words
- CBB, Coomassie Brilliant Blue
- DIC, differential interference contrast
- DTT, dithiothreitol
- EGF, epidermal growth factor
- EHP, external hydrophobic patch
- Egg coat
- Extracellular matrix
- Fertilization
- His6, hexahistidine
- IHP, internal hydrophobic patch
- Interdomain interaction
- MBP, maltose binding protein
- RT, room temperature
- TGFR, transforming growth factor-β receptor
- THP, Tamm–Horsfall protein
- Trx, thioredoxin
- ZP, zona pellucida
- Zona pellucida
Collapse
Affiliation(s)
- Hiroki Okumura
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, Nagoya 468-8502, Japan
- Corresponding author. Tel.: +81 52 838 2451; fax: +81 52 833 5524.
| | - Takahiro Sato
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, Nagoya 468-8502, Japan
| | - Rio Sakuma
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, Nagoya 468-8502, Japan
| | - Hideaki Fukushima
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, Nagoya 468-8502, Japan
| | - Tsukasa Matsuda
- Department of Applied Molecular Biosciences, Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya 464-8601, Japan
| | - Minoru Ujita
- Department of Applied Biological Chemistry, Graduate School of Agriculture, Meijo University, Nagoya 468-8502, Japan
| |
Collapse
|
39
|
Delolme F, Anastasi C, Alcaraz LB, Mendoza V, Vadon-Le Goff S, Talantikite M, Capomaccio R, Mevaere J, Fortin L, Mazzocut D, Damour O, Zanella-Cléon I, Hulmes DJS, Overall CM, Valcourt U, Lopez-Casillas F, Moali C. Proteolytic control of TGF-β co-receptor activity by BMP-1/tolloid-like proteases revealed by quantitative iTRAQ proteomics. Cell Mol Life Sci 2015; 72:1009-27. [PMID: 25260970 PMCID: PMC11113849 DOI: 10.1007/s00018-014-1733-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 08/29/2014] [Accepted: 09/09/2014] [Indexed: 10/24/2022]
Abstract
The metalloproteinase BMP-1 (bone morphogenetic protein-1) plays a major role in the control of extracellular matrix (ECM) assembly and growth factor activation. Most of the growth factors activated by BMP-1 are members of the TGF-β superfamily known to regulate multiple biological processes including embryonic development, wound healing, inflammation and tumor progression. In this study, we used an iTRAQ (isobaric tags for relative and absolute quantification)-based quantitative proteomic approach to reveal the release of proteolytic fragments from the cell surface or the ECM by BMP-1. Thirty-eight extracellular proteins were found in significantly higher or lower amounts in the conditioned medium of HT1080 cells overexpressing BMP-1 and thus, could be considered as candidate substrates. Strikingly, three of these new candidates (betaglycan, CD109 and neuropilin-1) were TGF-β co-receptors, also acting as antagonists when released from the cell surface, and were chosen for further substrate validation. Betaglycan and CD109 proved to be directly cleaved by BMP-1 and the corresponding cleavage sites were extensively characterized using a new mass spectrometry approach. Furthermore, we could show that the ability of betaglycan and CD109 to interact with TGF-β was altered after cleavage by BMP-1, leading to increased and prolonged SMAD2 phosphorylation in BMP-1-overexpressing cells. Betaglycan processing was also observed in primary corneal keratocytes, indicating a general and novel mechanism by which BMP-1 directly affects signaling by controlling TGF-β co-receptor activity. The proteomic data have been submitted to ProteomeXchange with the identifier PXD000786 and doi: 10.6019/PXD000786 .
Collapse
Affiliation(s)
- Frédéric Delolme
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
- Centre Commun de Microanalyse des Protéines, UMS 3444, Institut de Biologie et Chimie des Protéines, 69367 Lyon, France
| | - Cyril Anastasi
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Lindsay B. Alcaraz
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon (CRCL), Université de Lyon, Centre Léon Bérard, 69373 Lyon, France
| | - Valentin Mendoza
- Instituto de Fisiologia Celular, Universidad Nacional Autonoma de Mexico, 04510 Mexico, Mexico
| | - Sandrine Vadon-Le Goff
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Maya Talantikite
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Robin Capomaccio
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Jimmy Mevaere
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Laëtitia Fortin
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | - Dominique Mazzocut
- Centre Commun de Microanalyse des Protéines, UMS 3444, Institut de Biologie et Chimie des Protéines, 69367 Lyon, France
| | - Odile Damour
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
- Banque de Tissus et Cellules, Hospices Civils de Lyon, 69437 Lyon, France
| | - Isabelle Zanella-Cléon
- Centre Commun de Microanalyse des Protéines, UMS 3444, Institut de Biologie et Chimie des Protéines, 69367 Lyon, France
| | - David J. S. Hulmes
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| | | | - Ulrich Valcourt
- INSERM U1052, CNRS UMR 5286, Centre de Recherche en Cancérologie de Lyon (CRCL), Université de Lyon, Centre Léon Bérard, 69373 Lyon, France
| | - Fernando Lopez-Casillas
- Instituto de Fisiologia Celular, Universidad Nacional Autonoma de Mexico, 04510 Mexico, Mexico
| | - Catherine Moali
- UMR 5305, Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique, CNRS/Université de Lyon, 69367 Lyon, France
| |
Collapse
|
40
|
Iozzo RV, Schaefer L. Proteoglycan form and function: A comprehensive nomenclature of proteoglycans. Matrix Biol 2015; 42:11-55. [PMID: 25701227 PMCID: PMC4859157 DOI: 10.1016/j.matbio.2015.02.003] [Citation(s) in RCA: 850] [Impact Index Per Article: 85.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 02/07/2023]
Abstract
We provide a comprehensive classification of the proteoglycan gene families and respective protein cores. This updated nomenclature is based on three criteria: Cellular and subcellular location, overall gene/protein homology, and the utilization of specific protein modules within their respective protein cores. These three signatures were utilized to design four major classes of proteoglycans with distinct forms and functions: the intracellular, cell-surface, pericellular and extracellular proteoglycans. The proposed nomenclature encompasses forty-three distinct proteoglycan-encoding genes and many alternatively-spliced variants. The biological functions of these four proteoglycan families are critically assessed in development, cancer and angiogenesis, and in various acquired and genetic diseases where their expression is aberrant.
Collapse
Affiliation(s)
- Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology and the Cancer Cell Biology and Signaling Program, Kimmel Cancer Center, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, USA.
| | - Liliana Schaefer
- Pharmazentrum Frankfurt/ZAFES, Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany.
| |
Collapse
|
41
|
Makanji Y, Zhu J, Mishra R, Holmquist C, Wong WPS, Schwartz NB, Mayo KE, Woodruff TK. Inhibin at 90: from discovery to clinical application, a historical review. Endocr Rev 2014; 35:747-94. [PMID: 25051334 PMCID: PMC4167436 DOI: 10.1210/er.2014-1003] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
When it was initially discovered in 1923, inhibin was characterized as a hypophysiotropic hormone that acts on pituitary cells to regulate pituitary hormone secretion. Ninety years later, what we know about inhibin stretches far beyond its well-established capacity to inhibit activin signaling and suppress pituitary FSH production. Inhibin is one of the major reproductive hormones involved in the regulation of folliculogenesis and steroidogenesis. Although the physiological role of inhibin as an activin antagonist in other organ systems is not as well defined as it is in the pituitary-gonadal axis, inhibin also modulates biological processes in other organs through paracrine, autocrine, and/or endocrine mechanisms. Inhibin and components of its signaling pathway are expressed in many organs. Diagnostically, inhibin is used for prenatal screening of Down syndrome as part of the quadruple test and as a biochemical marker in the assessment of ovarian reserve. In this review, we provide a comprehensive summary of our current understanding of the biological role of inhibin, its relationship with activin, its signaling mechanisms, and its potential value as a diagnostic marker for reproductive function and pregnancy-associated conditions.
Collapse
Affiliation(s)
- Yogeshwar Makanji
- Department of Obstetrics and Gynecology (Y.M., J.Z., C.H., W.P.S.W., T.K.W.), Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60610; Center for Molecular Innovation and Drug Discovery (R.M., C.H.), Chemistry of Life Processes Institute, Northwestern University, Evanston, Illinois 60208; and Department of Molecular Biosciences (N.B.S., K.E.M., T.K.W.), Center for Reproductive Science, Northwestern University, Evanston, Illinois 60208
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Posttranslational modifications of zona pellucida proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 759:111-40. [PMID: 25030762 DOI: 10.1007/978-1-4939-0817-2_6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The zona pellucida (ZP), which surrounds the mammalian oocyte, functions in various aspects of fertilization. The ZP consists of three or four glycoproteins, which are derived from transmembrane proteins that lack the ability to self-assemble. Following posttranslational processing at specific sites, ectodomains of ZP precursor proteins are released from the membrane and begin to form a matrix. Glycosylational modification is thought to be involved in species-selective sperm recognition by ZP proteins. However, in mice, the supramolecular structure of the zona matrix is also important in sperm recognition. One ZP protein, ZP2, is processed at a specific site upon fertilization by ovastacin, which is released from cortical granules inside the oocyte. This phenomenon is involved in the block to polyspermy. The proteolysis of ubiquitinated ZP proteins by a sperm-associated proteasome is involved in penetration of the zona matrix by sperm, at least in the pigs. Thus, the posttranslational modification of ZP proteins is closely tied to ZP formation and the regulation of sperm-oocyte interactions.
Collapse
|
43
|
Plum, an immunoglobulin superfamily protein, regulates axon pruning by facilitating TGF-β signaling. Neuron 2013; 78:456-68. [PMID: 23664613 DOI: 10.1016/j.neuron.2013.03.004] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/26/2013] [Indexed: 11/22/2022]
Abstract
Axon pruning during development is essential for proper wiring of the mature nervous system, but its regulation remains poorly understood. We have identified an immunoglobulin superfamily (IgSF) transmembrane protein, Plum, that is cell autonomously required for axon pruning of mushroom body (MB) γ neurons and for ectopic synapse refinement at the developing neuromuscular junction in Drosophila. Plum promotes MB γ neuron axon pruning by regulating the expression of Ecdysone Receptor-B1, a key initiator of axon pruning. Genetic analyses indicate that Plum acts to facilitate signaling of Myoglianin, a glial-derived TGF-β, on MB γ neurons upstream of the type-I TGF-β receptor Baboon. Myoglianin, Baboon, and Ecdysone Receptor-B1 are also required for neuromuscular junction ectopic synapse refinement. Our study highlights both IgSF proteins and TGF-β facilitation as key promoters of developmental axon elimination and demonstrates a mechanistic conservation between MB axon pruning during metamorphosis and the refinement of ectopic larval neuromuscular connections.
Collapse
|
44
|
Diestel U, Resch M, Meinhardt K, Weiler S, Hellmann TV, Mueller TD, Nickel J, Eichler J, Muller YA. Identification of a Novel TGF-β-Binding Site in the Zona Pellucida C-terminal (ZP-C) Domain of TGF-β-Receptor-3 (TGFR-3). PLoS One 2013; 8:e67214. [PMID: 23826237 PMCID: PMC3695229 DOI: 10.1371/journal.pone.0067214] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 05/16/2013] [Indexed: 12/30/2022] Open
Abstract
The zona pellucida (ZP) domain is present in extracellular proteins such as the zona pellucida proteins and tectorins and participates in the formation of polymeric protein networks. However, the ZP domain also occurs in the cytokine signaling co-receptor transforming growth factor β (TGF-β) receptor type 3 (TGFR-3, also known as betaglycan) where it contributes to cytokine ligand recognition. Currently it is unclear how the ZP domain architecture enables this dual functionality. Here, we identify a novel major TGF-β-binding site in the FG loop of the C-terminal subdomain of the murine TGFR-3 ZP domain (ZP-C) using protein crystallography, limited proteolysis experiments, surface plasmon resonance measurements and synthetic peptides. In the murine 2.7 Å crystal structure that we are presenting here, the FG-loop is disordered, however, well-ordered in a recently reported homologous rat ZP-C structure. Surprisingly, the adjacent external hydrophobic patch (EHP) segment is registered differently in the rat and murine structures suggesting that this segment only loosely associates with the remaining ZP-C fold. Such a flexible and temporarily-modulated association of the EHP segment with the ZP domain has been proposed to control the polymerization of ZP domain-containing proteins. Our findings suggest that this flexibility also extends to the ZP domain of TGFR-3 and might facilitate co-receptor ligand interaction and presentation via the adjacent FG-loop. This hints that a similar C-terminal region of the ZP domain architecture possibly regulates both the polymerization of extracellular matrix proteins and cytokine ligand recognition of TGFR-3.
Collapse
Affiliation(s)
- Uschi Diestel
- Lehrstuhl fuer Biotechnik, Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Marcus Resch
- Lehrstuhl fuer Biotechnik, Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Kathrin Meinhardt
- Lehrstuhl fuer Biotechnik, Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Sigrid Weiler
- Lehrstuhl fuer Biotechnik, Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Tina V. Hellmann
- Julius-von-Sachs-Institut fuer Biowissenschaften, Lehrstuhl fuer Botanik I, University of Wuerzburg, Wuerzburg, Germany
| | - Thomas D. Mueller
- Julius-von-Sachs-Institut fuer Biowissenschaften, Lehrstuhl fuer Botanik I, University of Wuerzburg, Wuerzburg, Germany
| | - Joachim Nickel
- Lehrstuhl fuer Tissue Engineering und Regenerative Medizin, University of Wuerzburg, Wuerzburg, Germany
| | - Jutta Eichler
- Department of Chemistry and Pharmacy, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Yves A. Muller
- Lehrstuhl fuer Biotechnik, Department of Biology, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
- * E-mail:
| |
Collapse
|
45
|
Pimenta J, Sardinha J, Marques CC, Domingos A, Baptista MC, Barbas JP, Martins IC, Mesquita P, Pessa P, Soares R, Viegas A, Cabrita E, Horta EMA, Fontes CA, Prates AMJ, Pereira MLNR. Inhibition of ovine in vitro fertilization by anti-Prt antibody: hypothetical model for Prt/ZP interaction. Reprod Biol Endocrinol 2013; 11:25. [PMID: 23531155 PMCID: PMC3617107 DOI: 10.1186/1477-7827-11-25] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/18/2013] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The impact of prion proteins in the rules that dictate biological reproduction is still poorly understood. Likewise, the role of prnt gene, encoding the prion-like protein testis specific (Prt), in ram reproductive physiology remains largely unknown. In this study, we assessed the effect of Prt in ovine fertilization by using an anti-Prt antibody (APPA) in fertilization medium incubated with spermatozoa and oocytes. Moreover, a computational model was constructed to infer how the results obtained could be related to a hypothetical role for Prt in sperm-zona pellucida (ZP) binding. METHODS Mature ovine oocytes were transferred to fertilization medium alone (control) or supplemented with APPA, or pre-immune serum (CSerum). Oocytes were inseminated with ovine spermatozoa and after 18 h, presumptive zygotes (n=142) were fixed to evaluate fertilization rates or transferred (n=374) for embryo culture until D6-7. Predicted ovine Prt tertiary structure was compared with data obtained by circular dichroism spectroscopy (CD) and a protein-protein computational docking model was estimated for a hypothetical Prt/ZP interaction. RESULTS The fertilizing rate was lower (P=0.006) in APPA group (46.0+/-6.79%) when compared to control (78.5+/-7.47%) and CSerum (64.5+/-6.65%) groups. In addition, the cleavage rate was higher (P<0.0001) in control (44.1+/-4.15%) than in APPA group (19.7+/-4.22%). Prt CD spectroscopy showed a 22% alpha-helical structure in 30% (m/v) aqueous trifluoroethanol (TFE) and 17% alpha in 0.6% (m/v) TFE. The predominant alpha-helical secondary structure detected correlates with the predicted three dimensional structure for ovine Prt, which was subsequently used to test Prt/ZP docking. Computational analyses predicted a favorable Prt-binding activity towards ZP domains. CONCLUSIONS Our data indicates that the presence of APPA reduces the number of fertilized oocytes and of cleaved embryos. Moreover, the CD analysis data reinforces the predicted ovine Prt trend towards an alpha-helical structure. Predicted protein-protein docking suggests a possible interaction between Prt and ZP, thus supporting an important role for Prt in ovine fertilization.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Circular Dichroism
- Egg Proteins/chemistry
- Egg Proteins/genetics
- Egg Proteins/metabolism
- Embryo, Mammalian/drug effects
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Female
- Fertilization in Vitro/drug effects
- Male
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
- Models, Molecular
- Molecular Sequence Data
- Prions/chemistry
- Prions/immunology
- Prions/metabolism
- Protein Binding
- Protein Conformation/drug effects
- Protein Structure, Secondary/drug effects
- Protein Structure, Tertiary
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Sequence Homology, Amino Acid
- Sheep
- Sperm-Ovum Interactions/drug effects
- Time Factors
- Trifluoroethanol/chemistry
- Trifluoroethanol/pharmacology
- Zona Pellucida/metabolism
- Zona Pellucida Glycoproteins
Collapse
Affiliation(s)
- Jorge Pimenta
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
- CIISA, Faculdade de Medicina Veterinária (FMV), Universidade Técnica de Lisboa, Lisbon, Portugal
| | - João Sardinha
- REQUIMTE/CQFB, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Carla C Marques
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
| | - Ana Domingos
- IHMT-CMDT – Instituto de Higiene e Medicina Tropical, Centro de Malária e Doenças Tropicais, Lisbon, Portugal
| | - Maria C Baptista
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
| | - João P Barbas
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
| | - Ivo C Martins
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Av. Prof. Egas Moniz, Lisbon, 1649-028, Portugal
| | - Patrícia Mesquita
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
| | - Pedro Pessa
- Hospital Universitário de Coimbra, Coimbra, Portugal
| | - Rui Soares
- REQUIMTE/CQFB, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Aldino Viegas
- REQUIMTE/CQFB, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - Eurico Cabrita
- REQUIMTE/CQFB, Departamento de Química, Faculdade de Ciências e Tecnologia, Universidade Nova de Lisboa, Caparica, Portugal
| | - EM António Horta
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
| | - Carlos A Fontes
- CIISA, Faculdade de Medicina Veterinária (FMV), Universidade Técnica de Lisboa, Lisbon, Portugal
| | - AM José Prates
- CIISA, Faculdade de Medicina Veterinária (FMV), Universidade Técnica de Lisboa, Lisbon, Portugal
| | - MLN Rosa Pereira
- Unidade de Biotecnologia e Recursos Genéticos, Instituto Nacional de Investigação Agrária e Veterinária Santarém, Quinta da Fonte Boa, Vale de Santarém, 2005-048, Portugal
- Escola Universitária Vasco da Gama, Coimbra, Portugal
| |
Collapse
|
46
|
Alt A, Miguel-Romero L, Donderis J, Aristorena M, Blanco FJ, Round A, Rubio V, Bernabeu C, Marina A. Structural and functional insights into endoglin ligand recognition and binding. PLoS One 2012; 7:e29948. [PMID: 22347366 PMCID: PMC3275592 DOI: 10.1371/journal.pone.0029948] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2011] [Accepted: 12/08/2011] [Indexed: 11/19/2022] Open
Abstract
Endoglin, a type I membrane glycoprotein expressed as a disulfide-linked homodimer on human vascular endothelial cells, is a component of the transforming growth factor (TGF)-β receptor complex and is implicated in a dominant vascular dysplasia known as hereditary hemorrhagic telangiectasia as well as in preeclampsia. It interacts with the type I TGF-β signaling receptor activin receptor-like kinase (ALK)1 and modulates cellular responses to Bone Morphogenetic Protein (BMP)-9 and BMP-10. Structurally, besides carrying a zona pellucida (ZP) domain, endoglin contains at its N-terminal extracellular region a domain of unknown function and without homology to any other known protein, therefore called the orphan domain (OD). In this study, we have determined the recognition and binding ability of full length ALK1, endoglin and constructs encompassing the OD to BMP-9 using combined methods, consisting of surface plasmon resonance and cellular assays. ALK1 and endoglin ectodomains bind, independently of their glycosylation state and without cooperativity, to different sites of BMP-9. The OD comprising residues 22 to 337 was identified among the present constructs as the minimal active endoglin domain needed for partner recognition. These studies also pinpointed to Cys350 as being responsible for the dimerization of endoglin. In contrast to the complete endoglin ectodomain, the OD is a monomer and its small angle X-ray scattering characterization revealed a compact conformation in solution into which a de novo model was fitted.
Collapse
Affiliation(s)
- Aaron Alt
- Instituto de Biomedicina de Valencia, Valencia, Spain
| | | | - Jordi Donderis
- Instituto de Biomedicina de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| | - Mikel Aristorena
- Centro de Investigaciones Biologicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| | - Francisco J. Blanco
- Centro de Investigaciones Biologicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| | - Adam Round
- European Molecular Biology Laboratory, Grenoble, France
| | - Vicente Rubio
- Instituto de Biomedicina de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| | - Carmelo Bernabeu
- Centro de Investigaciones Biologicas, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| | - Alberto Marina
- Instituto de Biomedicina de Valencia, Valencia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras, Instituto de Salud Carlos III, Valencia, Spain
| |
Collapse
|
47
|
Angiogenesis regulation by TGFβ signalling: clues from an inherited vascular disease. Biochem Soc Trans 2011; 39:1659-66. [DOI: 10.1042/bst20110664] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Studies of rare genetic diseases frequently reveal genes that are fundamental to life, and the familial vascular disorder HHT (hereditary haemorrhagic telangiectasia) is no exception. The majority of HHT patients are heterozygous for mutations in either the ENG (endoglin) or the ACVRL1 (activin receptor-like kinase 1) gene. Both genes are essential for angiogenesis during development and mice that are homozygous for mutations in Eng or Acvrl1 die in mid-gestation from vascular defects. Recent development of conditional mouse models in which the Eng or Acvrl1 gene can be depleted in later life have confirmed the importance of both genes in angiogenesis and in the maintenance of a normal vasculature. Endoglin protein is a co-receptor and ACVRL1 is a signalling receptor, both of which are expressed primarily in endothelial cells to regulate TGFβ (transforming growth factor β) signalling in the cardiovasculature. The role of ACVRL1 and endoglin in TGFβ signalling during angiogenesis is now becoming clearer as interactions between these receptors and additional ligands of the TGFβ superfamily, as well as synergistic relationships with other signalling pathways, are being uncovered. The present review aims to place these recent findings into the context of a better understanding of HHT and to summarize recent evidence that confirms the importance of endoglin and ACVRL1 in maintaining normal cardiovascular health.
Collapse
|