1
|
Sander M, Wu Z, Strasburger CJ. Short stature explained by dimerization of human growth hormone induced by a p.C53S point mutation. J Biol Chem 2020; 295:4893-4901. [PMID: 32132170 DOI: 10.1074/jbc.ra119.009101] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 02/29/2020] [Indexed: 11/06/2022] Open
Abstract
A homozygous mutation in growth hormone 1 (GH1) was recently identified in an individual with growth failure. This mutation, c.705G>C, causes replacement of cysteine at position 53 of the 191-amino-acid sequence of 22 kDa human GH (hGH) with serine (p.C53S). This hGH molecule (hereafter referred to as GH-C53S) lacks the disulfide bond between p.Cys-53 and p.Cys-165, which is highly conserved among species. It has been reported previously that monomeric GH-C53S has reduced bioactivity compared with WT GH (GH-WT) because of its decreased ability to bind and activate the GH receptor in vitro In this study, we discovered that substitution of p.Cys-53 in hGH significantly increased formation of hGH dimers in pituitary cells. We expressed His-tagged hGH variants in the cytoplasm of genetically modified Rosetta-gami B DE3 Escherichia coli cells, facilitating high-yield production. We observed that the bioactivity of monomeric GH-C53S is 25.2% of that of GH-WT and that dimeric GH-C53S-His has no significant bioactivity in cell proliferation assays. We also found that the expression of GH-C53S in pituitary cells deviates from that of GH-WT. GH-C53S was exclusively stained in the Golgi apparatus, and no secretory granules formed for this variant, impairing its stimulated release. In summary, the unpaired Cys-165 in GH-C53S forms a disulfide bond linking two hGH molecules in pituitary cells. We conclude that the GH-C53S dimer is inactive and responsible for the growth failure in the affected individual.
Collapse
Affiliation(s)
- Max Sander
- Department of Endocrinology, Diabetes, and Nutritional Medicine, Campus Charité Mitte, Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Zida Wu
- Department of Endocrinology, Diabetes, and Nutritional Medicine, Campus Charité Mitte, Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| | - Christian J Strasburger
- Department of Endocrinology, Diabetes, and Nutritional Medicine, Campus Charité Mitte, Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
2
|
Petersen M, Gandhi PS, Buchardt J, Alanentalo T, Fels JJ, Johansen NL, Helding-Kvist P, Vad K, Thygesen P. Tissue Distribution and Receptor Activation by Somapacitan, a Long Acting Growth Hormone Derivative. Int J Mol Sci 2020; 21:ijms21041181. [PMID: 32053994 PMCID: PMC7072805 DOI: 10.3390/ijms21041181] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/07/2020] [Accepted: 02/08/2020] [Indexed: 11/16/2022] Open
Abstract
Somapacitan is a long-acting, once-weekly, albumin-binding growth hormone (GH) derivative. The reversible albumin-binding properties leads to prolonged circulation half-life. Here, we investigated and compared somapacitan with human GH on downstream receptor signaling in primary hepatocytes and hepatocellular models and using isothermal titration calorimetry to characterize receptor binding of somapacitan in the presence or absence of human serum albumin (HSA). With non-invasive fluorescence imaging we quantitatively visualize and compare the temporal distribution and examine the tissue-specific growth hormone receptor (GHR) activation at distribution sites. We found that signaling kinetics were slightly more rapid and intense for GH compared with somapacitan. Receptor binding isotherms were characterized by a high and a low affinity interaction site with or without HSA. Using in vivo optical imaging we found prolonged systemically biodistribution of somapacitan compared with GH, which correlated with plasma pharmacokinetics. Ex vivo mouse organ analysis revealed that the temporal fluorescent intensity in livers dosed with somapacitan was significantly increased compared with GH-dosed livers and correlated with the degree of downstream GHR activation. Finally, we show that fluorescent-labeled analogs distributed to the hypertrophic zone in the epiphysis of proximal tibia of hypophysectomized rats and that somapacitan and GH activate the GHR signaling in epiphyseal tissues.
Collapse
Affiliation(s)
- Maj Petersen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | | - Jens Buchardt
- Global Research Technologies, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Tomas Alanentalo
- Umeå Centre for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
| | | | | | | | - Knud Vad
- Global Development, Novo Nordisk A/S, 2860 Søborg, Denmark
| | - Peter Thygesen
- Global Drug Discovery, Novo Nordisk A/S, 2760 Måløv, Denmark
- Correspondence: ; Tel.: +45-3075-4617
| |
Collapse
|
3
|
Gertler A, Solomon G. Pegylated Human Leptin D23L Mutant-Preparation and Biological Activity In Vitro and In Vivo in Male ob/ob Mice. Endocrinology 2019; 160:891-898. [PMID: 30802285 DOI: 10.1210/en.2018-00643] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 02/20/2019] [Indexed: 02/06/2023]
Abstract
Recombinant monomeric human leptin (hLEP) and its D23L mutant were prepared in Escherichia coli and pegylated at their N-terminus using 20-kDa methoxy pegylated (PEG)-propionylaldehyde. As determined by both SDS-PAGE and size-exclusion chromatography, the pegylated proteins consisted of >90% monopegylated and <10% double-pegylated species. Circular dichroism spectra showed that their secondary structure, characteristic of all four α-helix bundle cytokines, was not affected by either the D23L mutation or pegylation. Because of the D23L mutation, affinity for hLEP receptor increased 25- and 40-fold for the pegylated and nonpegylated mutant, respectively. However, whereas the proliferation-promoting activity in vitro of nonmutated and mutated nonpegylated hLEP was identical, that of the respective pegylated mutant was approximately sixfold higher compared with the pegylated nonmutated hLEP. This difference was also seen in vivo. Both pegylated hLEPs at all doses significantly decreased body weight and food consumption, as compared with the vehicle-treated control. Once-daily administration of pegylated hLEP D23L at doses of 0.1, 0.3, and 1 mg/kg for 14 consecutive days in ob/ob mice resulted in significantly decreased body weight and food consumption as compared with respective pegylated hLEP-treated animals, with the biggest difference observed at 0.1 mg/kg. Repeated administration of either pegylated hLEP D23L or pegylated hLEP significantly decreased blood glucose levels compared with the control before glucose challenge and after oral glucose tolerance test, but with no difference between the two treatments. The pegylated hLEP D23L mutant seems to be a more potent reagent suitable for in vivo studies than the pegylated nonmutated hLEP.
Collapse
Affiliation(s)
- Arieh Gertler
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Gili Solomon
- Institute of Biochemistry, Food Science and Nutrition, The Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| |
Collapse
|
4
|
Bourg S, Jacob L, Menu F, Rajon E. Hormonal pleiotropy and the evolution of allocation trade-offs. Evolution 2019; 73:661-674. [PMID: 30734273 DOI: 10.1111/evo.13693] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 01/09/2019] [Indexed: 12/15/2022]
Abstract
Recent empirical evidence suggests that trade-off relationships can evolve, challenging the classical image of their high entrenchment. For energy reliant traits, this relationship should depend on the endocrine system that regulates resource allocation. Here, we model changes in this system by mutating the expression and conformation of its constitutive hormones and receptors. We show that the shape of trade-offs can indeed evolve in this model through the combined action of genetic drift and selection, such that their evolutionarily expected curvature and length depend on context. In particular, the shape of a trade-off should depend on the cost associated with resource storage, itself depending on the traded resource and on the ecological context. Despite this convergence at the phenotypic level, we show that a variety of physiological mechanisms may evolve in similar simulations, suggesting redundancy at the genetic level. This model should provide a useful framework to interpret and unify the overly complex observations of evolutionary endocrinology and evolutionary ecology.
Collapse
Affiliation(s)
- Salomé Bourg
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622 Villeurbanne, France
| | - Laurent Jacob
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622 Villeurbanne, France
| | - Frédéric Menu
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622 Villeurbanne, France
| | - Etienne Rajon
- Univ Lyon, Université Lyon 1, CNRS, Laboratoire de Biométrie et Biologie Évolutive UMR5558, F-69622 Villeurbanne, France
| |
Collapse
|
5
|
Spangler JB, Trotta E, Tomala J, Peck A, Young TA, Savvides CS, Silveria S, Votavova P, Salafsky J, Pande VS, Kovar M, Bluestone JA, Garcia KC. Engineering a Single-Agent Cytokine/Antibody Fusion That Selectively Expands Regulatory T Cells for Autoimmune Disease Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 201:2094-2106. [PMID: 30104245 DOI: 10.4049/jimmunol.1800578] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 07/19/2018] [Indexed: 12/18/2022]
Abstract
IL-2 has been used to treat diseases ranging from cancer to autoimmune disorders, but its concurrent immunostimulatory and immunosuppressive effects hinder efficacy. IL-2 orchestrates immune cell function through activation of a high-affinity heterotrimeric receptor (composed of IL-2Rα, IL-2Rβ, and common γ [γc]). IL-2Rα, which is highly expressed on regulatory T (TReg) cells, regulates IL-2 sensitivity. Previous studies have shown that complexation of IL-2 with the JES6-1 Ab preferentially biases cytokine activity toward TReg cells through a unique mechanism whereby IL-2 is exchanged from the Ab to IL-2Rα. However, clinical adoption of a mixed Ab/cytokine complex regimen is limited by stoichiometry and stability concerns. In this study, through structure-guided design, we engineered a single agent fusion of the IL-2 cytokine and JES6-1 Ab that, despite being covalently linked, preserves IL-2 exchange, selectively stimulating TReg expansion and exhibiting superior disease control to the mixed IL-2/JES6-1 complex in a mouse colitis model. These studies provide an engineering blueprint for resolving a major barrier to the implementation of functionally similar IL-2/Ab complexes for treatment of human disease.
Collapse
Affiliation(s)
- Jamie B Spangler
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305.,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| | - Eleonora Trotta
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143
| | - Jakub Tomala
- Laboratory of Tumor Immunology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, 14220 Prague 4-Krc, Czech Republic
| | - Ariana Peck
- Department of Biochemistry, Stanford University, Stanford, CA 94305
| | | | | | - Stephanie Silveria
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143
| | - Petra Votavova
- Laboratory of Tumor Immunology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, 14220 Prague 4-Krc, Czech Republic
| | | | - Vijay S Pande
- Department of Bioengineering, Stanford University, Stanford, CA 94305; and
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Academy of Sciences of the Czech Republic, 14220 Prague 4-Krc, Czech Republic
| | - Jeffrey A Bluestone
- Diabetes Center, University of California San Francisco, San Francisco, CA 94143.,Sean N. Parker Autoimmune Research Laboratory, University of California San Francisco, San Francisco, CA 94143
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305; .,Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305.,Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
6
|
Hong P, Lan H, Li Y, Fu Z, Zheng X. Different intracellular signalling properties induced by human and porcine growth hormone. Gen Comp Endocrinol 2016; 229:67-73. [PMID: 26944485 DOI: 10.1016/j.ygcen.2016.02.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/25/2016] [Accepted: 02/29/2016] [Indexed: 11/16/2022]
Abstract
Growth hormone (GH) is reportedly species-specific. Primate growth hormone can trigger non-primate growth hormone receptor (GHR), but primates GHR cannot be activated by non-primate GH. However, it is also unclear that why primate GH and non-primate GH have different biological activities. Thus, we analysed primate growth hormone (human growth hormone (hGH)) or non-primate GH (porcine growth hormone (pGH))-induced intracellular signalling in 3T3-F442A cells and rat hepatocytes in a dose- and time-dependent manner to explore the different biological activities between them. The results revealed that both hGH and pGH can activate Janus kinase 2 (JAK2), Signal transducers and activators of transcription 1, 3 and 5 (STATs 1, 3 and 5) and extracellular signal-regulated kinase 1/2 (ERK1/2). There were no significant differences in JAK2 or ERK1/2 tyrosine phosphorylation after hGH and pGH treatment, but there were different between hGH and pGH in STAT/1/3/5 tyrosine phosphorylation, and JAK2, STAT/1/3/5 tyrosine phosphorylation was time-dependent and dose-dependent, whereas ERK1/2 was not. Both hGH and pGH demonstrated similar kinetics for STATs 1, 3 and 5 phosphorylation, but the pGH-mediated tyrosine phosphorylation was weaker than that mediated by hGH. Our observations indicated that the levels of main signalling proteins phosphorylation triggered by hGH or pGH were not exactly the same, which may explain the different biological activities showed by primate GH and non-primate GH.
Collapse
Affiliation(s)
- Pan Hong
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun 130118, China
| | - Hainan Lan
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun 130118, China
| | - Yumeng Li
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun 130118, China
| | - Zhiling Fu
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun 130118, China
| | - Xin Zheng
- College of Animal Science and Technology, Jilin Agricultural University, Xincheng Street 2888, Changchun 130118, China.
| |
Collapse
|
7
|
Mitchell AC, Briquez PS, Hubbell JA, Cochran JR. Engineering growth factors for regenerative medicine applications. Acta Biomater 2016; 30:1-12. [PMID: 26555377 PMCID: PMC6067679 DOI: 10.1016/j.actbio.2015.11.007] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Revised: 10/27/2015] [Accepted: 11/06/2015] [Indexed: 01/10/2023]
Abstract
Growth factors are important morphogenetic proteins that instruct cell behavior and guide tissue repair and renewal. Although their therapeutic potential holds great promise in regenerative medicine applications, translation of growth factors into clinical treatments has been hindered by limitations including poor protein stability, low recombinant expression yield, and suboptimal efficacy. This review highlights current tools, technologies, and approaches to design integrated and effective growth factor-based therapies for regenerative medicine applications. The first section describes rational and combinatorial protein engineering approaches that have been utilized to improve growth factor stability, expression yield, biodistribution, and serum half-life, or alter their cell trafficking behavior or receptor binding affinity. The second section highlights elegant biomaterial-based systems, inspired by the natural extracellular matrix milieu, that have been developed for effective spatial and temporal delivery of growth factors to cell surface receptors. Although appearing distinct, these two approaches are highly complementary and involve principles of molecular design and engineering to be considered in parallel when developing optimal materials for clinical applications. STATEMENT OF SIGNIFICANCE Growth factors are promising therapeutic proteins that have the ability to modulate morphogenetic behaviors, including cell survival, proliferation, migration and differentiation. However, the translation of growth factors into clinical therapies has been hindered by properties such as poor protein stability, low recombinant expression yield, and non-physiological delivery, which lead to suboptimal efficacy and adverse side effects. To address these needs, researchers are employing clever molecular and material engineering and design strategies to both improve the intrinsic properties of growth factors and effectively control their delivery into tissue. This review highlights examples of interdisciplinary tools and technologies used to augment the therapeutic potential of growth factors for clinical applications in regenerative medicine.
Collapse
Affiliation(s)
- Aaron C Mitchell
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Priscilla S Briquez
- Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jeffrey A Hubbell
- Institute for Bioengineering, School of Life Sciences and School of Engineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland; Institute for Molecular Engineering, University of Chicago, Chicago, IL, USA; Materials Science Division, Argonne National Laboratory, Argonne, IL, USA.
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA, USA; Department of Chemical Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Moraga I, Richter D, Wilmes S, Winkelmann H, Jude K, Thomas C, Suhoski MM, Engleman EG, Piehler J, Garcia KC. Instructive roles for cytokine-receptor binding parameters in determining signaling and functional potency. Sci Signal 2015; 8:ra114. [PMID: 26554818 DOI: 10.1126/scisignal.aab2677] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cytokines dimerize cell surface receptors to activate signaling and regulate many facets of the immune response. Many cytokines have pleiotropic effects, inducing a spectrum of redundant and distinct effects on different cell types. This pleiotropy has hampered cytokine-based therapies, and the high doses required for treatment often lead to off-target effects, highlighting the need for a more detailed understanding of the parameters controlling cytokine-induced signaling and bioactivities. Using the prototypical cytokine interleukin-13 (IL-13), we explored the interrelationships between receptor binding and a wide range of downstream cellular responses. We applied structure-based engineering to generate IL-13 variants that covered a spectrum of binding strengths for the receptor subunit IL-13Rα1. Engineered IL-13 variants representing a broad range of affinities for the receptor exhibited similar potencies in stimulating the phosphorylation of STAT6 (signal transducer and activator of transcription 6). Delays in the phosphorylation and nuclear translocation of STAT6 were only apparent for those IL-13 variants with markedly reduced affinities for the receptor. From these data, we developed a mechanistic model that quantitatively reproduced the kinetics of STAT6 phosphorylation for the entire spectrum of binding affinities. Receptor endocytosis played a key role in modulating STAT6 activation, whereas the lifetime of receptor-ligand complexes at the plasma membrane determined the potency of the variant for inducing more distal responses. This complex interrelationship between extracellular ligand binding and receptor function provides the foundation for new mechanism-based strategies that determine the optimal cytokine dose to enhance therapeutic efficacy.
Collapse
Affiliation(s)
- Ignacio Moraga
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - David Richter
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Stephan Wilmes
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Hauke Winkelmann
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany
| | - Kevin Jude
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Christoph Thomas
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Megan M Suhoski
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Edgar G Engleman
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA
| | - Jacob Piehler
- Department of Biology, University of Osnabrück, 49076 Osnabrück, Germany.
| | - K Christopher Garcia
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305-5345, USA. Department of Molecular and Cellular Physiology and Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345, USA.
| |
Collapse
|
9
|
High efficiency cell-specific targeting of cytokine activity. Nat Commun 2015; 5:3016. [PMID: 24398568 DOI: 10.1038/ncomms4016] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 11/26/2013] [Indexed: 11/09/2022] Open
Abstract
Systemic toxicity currently prevents exploiting the huge potential of many cytokines for medical applications. Here we present a novel strategy to engineer immunocytokines with very high targeting efficacies. The method lies in the use of mutants of toxic cytokines that markedly reduce their receptor-binding affinities, and that are thus rendered essentially inactive. Upon fusion to nanobodies specifically binding to marker proteins, activity of these cytokines is selectively restored for cell populations expressing this marker. This 'activity-by-targeting' concept was validated for type I interferons and leptin. In the case of interferon, activity can be directed to target cells in vitro and to selected cell populations in mice, with up to 1,000-fold increased specific activity. This targeting strategy holds promise to revitalize the clinical potential of many cytokines.
Collapse
|
10
|
Abstract
Bone morphogenetic proteins (BMPs), together with the eponymous transforming growth factor (TGF) β and the Activins form the TGFβ superfamily of ligands. This protein family comprises more than 30 structurally highly related proteins, which determine formation, maintenance, and regeneration of tissues and organs. Their importance for the development of multicellular organisms is evident from their existence in all vertebrates as well as nonvertebrate animals. From their highly specific functions in vivo either a strict relation between a particular ligand and its cognate cellular receptor and/or a stringent regulation to define a distinct temperospatial expression pattern for the various ligands and receptor is expected. However, only a limited number of receptors are found to serve a large number of ligands thus implicating highly promiscuous ligand-receptor interactions instead. Since in tissues a multitude of ligands are often found, which signal via a highly overlapping set of receptors, this raises the question how such promiscuous interactions between different ligands and their receptors can generate concerted and highly specific cellular signals required during embryonic development and tissue homeostasis.
Collapse
Affiliation(s)
- Thomas D Mueller
- Department Plant Physiology and Biophysics, Julius-von-Sachs Institute of the University Wuerzburg, Wuerzburg, Germany.
| |
Collapse
|
11
|
Zhang C, Nygaard M, Haxholm GW, Boutillon F, Bernadet M, Hoos S, England P, Broutin I, Kragelund BB, Goffin V. A Residue Quartet in the Extracellular Domain of the Prolactin Receptor Selectively Controls Mitogen-activated Protein Kinase Signaling. J Biol Chem 2015; 290:11890-904. [PMID: 25784554 DOI: 10.1074/jbc.m115.639096] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Indexed: 11/06/2022] Open
Abstract
Cytokine receptors elicit several signaling pathways, but it is poorly understood how they select and discriminate between them. We have scrutinized the prolactin receptor as an archetype model of homodimeric cytokine receptors to address the role of the extracellular membrane proximal domain in signal transfer and pathway selection. Structure-guided manipulation of residues involved in the receptor dimerization interface identified one residue (position 170) that in cell-based assays profoundly altered pathway selectivity and species-specific bio-characteristics. Subsequent in vitro spectroscopic and nuclear magnetic resonance analyses revealed that this residue was part of a residue quartet responsible for specific local structural changes underlying these effects. This included alteration of a novel aromatic T-stack within the membrane proximal domain, which promoted selective signaling affecting primarily the MAPK (ERK1/2) pathway. Importantly, activation of the MAPK pathway correlated with in vitro stabilities of ternary ligand·receptor complexes, suggesting a threshold mean lifetime of the complex necessary to achieve maximal activation. No such dependence was observed for STAT5 signaling. Thus, this study establishes a residue quartet in the extracellular membrane proximal domain of homodimeric cytokine receptors as a key regulator of intracellular signaling discrimination.
Collapse
Affiliation(s)
- Chi Zhang
- From the Inserm, U1151, Institut Necker Enfants Malades, Equipe Physiopathologie des Hormones PRL/GH, Paris 75014, France, the Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Mads Nygaard
- the Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Gitte W Haxholm
- the Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Florence Boutillon
- From the Inserm, U1151, Institut Necker Enfants Malades, Equipe Physiopathologie des Hormones PRL/GH, Paris 75014, France, the Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Marie Bernadet
- From the Inserm, U1151, Institut Necker Enfants Malades, Equipe Physiopathologie des Hormones PRL/GH, Paris 75014, France, the Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France
| | - Sylviane Hoos
- the Institut Pasteur, Plateforme de Biophysique des Macromolécules et de leurs Interactions, Département de Biologie Structurale et Chimie, Paris 75015, France, and
| | - Patrick England
- the Institut Pasteur, Plateforme de Biophysique des Macromolécules et de leurs Interactions, Département de Biologie Structurale et Chimie, Paris 75015, France, and
| | - Isabelle Broutin
- the Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France, Laboratoire de Cristallographie et RMN Biologiques CNRS, UMR 8015, Paris 75006, France
| | - Birthe B Kragelund
- the Structural Biology and NMR Laboratory, Department of Biology, University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Vincent Goffin
- From the Inserm, U1151, Institut Necker Enfants Malades, Equipe Physiopathologie des Hormones PRL/GH, Paris 75014, France, the Université Paris Descartes, Sorbonne Paris Cité, Paris 75006, France,
| |
Collapse
|
12
|
Petridou B. Differences in affinities between the homologous and the heterologous rabbit prolactin-receptor interaction with respect to proliferation and differentiation activities. Gen Comp Endocrinol 2015; 213:118-29. [PMID: 25449135 DOI: 10.1016/j.ygcen.2014.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 09/08/2014] [Accepted: 10/23/2014] [Indexed: 11/23/2022]
Abstract
Interspecies differences in PRL-receptor binding and their relationship with bioactivity deserve investigation since cross-reactivity is relevant to the design of many experiments. We have previously shown that the lower affinity of rabbit prolactin (rbPRL) binding to its homologous receptor is due to its faster and more complete dissociation compared with that of ovine PRL (oPRL). In order to obtain sufficient amounts of rbPRL to study the functional consequences of its low affinity homologous interaction, rbPRL was expressed recombinantly in Escherichia coli (rec rbPRL) as insoluble inclusion bodies, refolded and purified to homogeneity, yielding electrophoretically pure, over 98% monomeric rec rbPRL. Proper renaturation of rec rbPRL was evidenced by comparison of its CD spectra, binding parameters and bioactivity with those determined for the rbPRL. The binding potency of rec rbPRL to its receptor, expressed either endogenously in the mammary gland or recombinantly in mammalian cells is one log unit lower than that to the receptor expressed recombinantly in insect cells. This difference is probably related to differences in cell-dependent receptor densities. The proliferation potency of rbPRL or rec rbPRL was one log unit lower than that of oPRL, consistent with its lower binding affinity, but the differentiation potencies of these PRLs were similar. Thus, the proliferation activity is sensitive to PRL-receptor affinity and dissociation kinetics, whereas the differentiation response is marginally modulated.
Collapse
Affiliation(s)
- Barbara Petridou
- UMR 1313 Génétique Animale et Biologie Intégrative, INRA Institut National de la Recherche Agronomique, F-78352 Jouy-en-Josas, France
| |
Collapse
|
13
|
Uzé G, Tavernier J. High efficiency targeting of IFN-α activity: possible applications in fighting tumours and infections. Cytokine Growth Factor Rev 2014; 26:179-82. [PMID: 25466630 DOI: 10.1016/j.cytogfr.2014.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 10/22/2014] [Indexed: 10/24/2022]
Abstract
In this short review, we summarize how insights into the structure and dynamics of interferon-receptor complex assembly and activation guided the design of a novel class of engineered type I interferons that combine a largely lost potency on non-targeted cells with high activity on targeted cells. These novel interferons are expected to exhibit lower systemic toxicities compared to other interferon therapy modalities and could open avenues to revive these cytokines for the treatment of patients suffering of cancer and viral infections.
Collapse
Affiliation(s)
- Gilles Uzé
- CNRS UMR 5235, University Montpellier, Place Eugène Bataillon, 34095 Montpellier Cedex 5, France.
| | - Jan Tavernier
- Department of Medical Protein Research, Flanders Institute for Biotechnology, Ghent University, Faculty of Medicine and Health Sciences, Ghent, BE 9000, Belgium.
| |
Collapse
|
14
|
Kepsutlu B, Kizilel R, Kizilel S. Quantification of interactions among circadian clock proteins via surface plasmon resonance. J Mol Recognit 2014; 27:458-69. [PMID: 24895278 DOI: 10.1002/jmr.2367] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/01/2013] [Accepted: 01/29/2014] [Indexed: 11/11/2022]
Abstract
Circadian clock is an internal time keeping system recurring 24 h daily rhythm in physiology and behavior of organisms. Circadian clock contains transcription and translation feedback loop involving CLOCK/NPAS2, BMAL1, Cry1/2, and Per1/2. In common, heterodimer of CLOCK/NPAS2 and BMAL1 binds to EBOX element in the promoter of Per and Cry genes in order to activate their transcription. CRY and PER making heterodimeric complexes enter the nucleus in order to inhibit their own BMAL1-CLOCK-activated transcription. The aim of this study was to investigate and quantify real-time binding affinities of clock proteins among each other on and off DNA modes using surface plasmon resonance. The pairwise interaction coefficients among clock proteins, as well as interaction of PER2, CRY2, and PER2 : CRY2 proteins with BMAL1 : CLOCK complex in the presence and absence of EBOX motif have been investigated via analysis of surface plasmon resonance data with pseudo first-order reaction kinetics approximation and via nonlinear regression curve fitting. The results indicated that CRY2 and PER2, BMAL1, and CLOCK proteins form complexes in vitro and that PER2, CRY2 and PER2 : CRY2 complex have similar affinities toward BMAL1 : CLOCK complex. CRY2 protein had the highest affinity toward EBOX complex, whereas PER2 and CRY2 : PER2 complexes displayed low affinity toward EBOX complex. The quantification of the interaction between clock proteins is critical to understand the operation mechanism of the biological clock and to address the behavioral and physiological disorders, and it will be useful for the design of new drugs toward clock-related diseases.
Collapse
Affiliation(s)
- Burcu Kepsutlu
- Chemical and Biological Engineering, Koc University, Sariyer, Istanbul, 34450, Turkey
| | | | | |
Collapse
|
15
|
Liebner R, Mathaes R, Meyer M, Hey T, Winter G, Besheer A. Protein HESylation for half-life extension: Synthesis, characterization and pharmacokinetics of HESylated anakinra. Eur J Pharm Biopharm 2014; 87:378-85. [DOI: 10.1016/j.ejpb.2014.03.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2013] [Revised: 03/13/2014] [Accepted: 03/18/2014] [Indexed: 01/07/2023]
|
16
|
Junnila RK, Wu Z, Strasburger CJ. The role of human growth hormone's C-terminal disulfide bridge. Growth Horm IGF Res 2013; 23:62-67. [PMID: 23478141 DOI: 10.1016/j.ghir.2013.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Revised: 02/04/2013] [Accepted: 02/12/2013] [Indexed: 10/27/2022]
Abstract
OBJECTIVE Human growth hormone (hGH), as well as the other members of the same polypeptide hormone family, have a four-helix bundle structure linked by two disulfide bridges, C53-C165 and C182-C189 in hGH. The C-terminal disulfide bridge of growth hormone is evolutionally conserved but its role is unknown. Our aim was to determine its importance for GH structure and/or function. DESIGN We disrupted the highly conserved C-terminal disulfide bridge of hGH by substituting one or both of its cysteines by alanines. Mutant and wild type hGH genes were expressed in human embryonic kidney (HEK)-293 cells and the hGH analogs were characterized in vitro regarding biological activity, stability and binding to GH receptor (GHR) as well as GH binding protein (GHBP). RESULTS Disrupting the hGH C-terminal disulfide bridge significantly reduces binding affinity to GHR and GHBP. If one of the cysteines is removed, the stability of the molecule is reduced but this feature is reversed when both cysteines are absent. However, despite decreased binding affinity and stability, biological activity is only modestly decreased when the disulfide bridge is removed. CONCLUSIONS Our study reveals the importance of the C-terminal disulfide bridge of GH for receptor binding and the detrimental effect of its unpaired cysteines on stability as well as, to a lesser extent, biological activity. This improved knowledge of structure-function relationships helps better understand the biology of GH and related molecules. This could have an impact on diagnosis and treatment of patients with growth disorders.
Collapse
Affiliation(s)
- R K Junnila
- Department of Clinical Endocrinology, Campus Charité Mitte, Charité Universitaetsmedizin, Berlin, Germany.
| | | | | |
Collapse
|
17
|
Development of a long acting human growth hormone analog suitable for once a week dosing. Bioorg Med Chem Lett 2013; 23:402-6. [DOI: 10.1016/j.bmcl.2012.11.104] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 11/21/2012] [Accepted: 11/26/2012] [Indexed: 11/23/2022]
|
18
|
Abstract
Achieving optimal patient benefit from biological therapies can be hindered by drug instability, rapid clearance requiring frequent dosing or potential immune reactions. One strategy for addressing these challenges is drug modification through PEGylation, a well established process by which one or more molecules of polyethylene glycol (PEG) are covalently attached to a biological or small-molecule drug, effectively transforming it into a therapy with improved pharmacokinetic and pharmacodynamic properties. Numerous PEGylated therapeutics are currently available, all of which have at least comparable efficacy, safety and tolerability to their unmodified forms. A PEGylated form of interferon-β-1a (PEG-IFNβ-1a) is being developed to address an unmet medical need for safer, more effective and more convenient therapies for multiple sclerosis (MS). Phase I study data suggest that PEG-IFNβ-1a should provide patients with a first-line therapy with a more convenient dosing regimen while maintaining the established efficacy, safety and tolerability of presently available IFNβ-1a. The ongoing global ADVANCE phase III study will determine the clinical efficacy of PEG-IFNβ-1a in patients with relapsing MS.
Collapse
|
19
|
Cheng TL, Chuang KH, Chen BM, Roffler SR. Analytical Measurement of PEGylated Molecules. Bioconjug Chem 2012; 23:881-99. [DOI: 10.1021/bc200478w] [Citation(s) in RCA: 92] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Tian-Lu Cheng
- Department of Biomedical Science
and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kuo-Hsiang Chuang
- Department of Biomedical Science
and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bing-Mae Chen
- Institute
of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R. Roffler
- Institute
of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
20
|
Brown NG, Chow DC, Sankaran B, Zwart P, Prasad BVV, Palzkill T. Analysis of the binding forces driving the tight interactions between beta-lactamase inhibitory protein-II (BLIP-II) and class A beta-lactamases. J Biol Chem 2011; 286:32723-35. [PMID: 21775426 PMCID: PMC3173220 DOI: 10.1074/jbc.m111.265058] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2011] [Revised: 06/25/2011] [Indexed: 12/19/2022] Open
Abstract
β-Lactamases hydrolyze β-lactam antibiotics to provide drug resistance to bacteria. β-Lactamase inhibitory protein-II (BLIP-II) is a potent proteinaceous inhibitor that exhibits low picomolar affinity for class A β-lactamases. This study examines the driving forces for binding between BLIP-II and β-lactamases using a combination of presteady state kinetics, isothermal titration calorimetry, and x-ray crystallography. The measured dissociation rate constants for BLIP-II and various β-lactamases ranged from 10(-4) to 10(-7) s(-1) and are comparable with those found in some of the tightest known protein-protein interactions. The crystal structures of BLIP-II alone and in complex with Bacillus anthracis Bla1 β-lactamase revealed no significant side-chain movement in BLIP-II in the complex versus the monomer. The structural rigidity of BLIP-II minimizes the loss of the entropy upon complex formation and, as indicated by thermodynamics experiments, may be a key determinant of the observed potent inhibition of β-lactamases.
Collapse
Affiliation(s)
- Nicholas G. Brown
- From the Departments of Pharmacology
- Biochemistry and Molecular Biology, and
| | | | - Banumathi Sankaran
- The Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - Peter Zwart
- The Berkeley Center for Structural Biology, Lawrence Berkeley National Laboratory, Berkeley, California 94720
| | - B. V. Venkataram Prasad
- Biochemistry and Molecular Biology, and
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030 and
| | - Timothy Palzkill
- From the Departments of Pharmacology
- Biochemistry and Molecular Biology, and
- Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas 77030 and
| |
Collapse
|
21
|
Stochastic receptor expression determines cell fate upon interferon treatment. Mol Cell Biol 2011; 31:3252-66. [PMID: 21690295 DOI: 10.1128/mcb.05251-11] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Type I interferons trigger diverse biological effects by binding a common receptor, composed of IFNAR1 and IFNAR2. Intriguingly, while the activation of an antiviral state is common to all cells, antiproliferative activity and apoptosis affect only part of the population, even when cells are stimulated with saturating interferon concentrations. Manipulating receptor expression by different small interfering RNA (siRNA) concentrations reduced the fraction of responsive cells independent of the interferon used, including a newly generated, extremely tight-binding variant. Reduced receptor numbers increased 50% effective concentrations (EC(50)s) for alpha interferon 2 (IFN-α2) but not for the tight-binding variant. A correlation between receptor numbers, STAT activation, and gene induction is observed. Our data suggest that for a given cell, the response is binary (+/-) and dependent on the stochastic expression levels of the receptors on an individual cell. A low number of receptors suffices for antiviral response and is thus a robust feature common to all cells. Conversely, a high number of receptors is required for antiproliferative activity, which allows for fine-tuning on a single-cell level.
Collapse
|
22
|
Bai H, Yang K, Yu D, Zhang C, Chen F, Lai L. Predicting kinetic constants of protein-protein interactions based on structural properties. Proteins 2010; 79:720-34. [PMID: 21287608 DOI: 10.1002/prot.22904] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2010] [Revised: 07/24/2010] [Accepted: 08/23/2010] [Indexed: 02/01/2023]
Abstract
Elucidating kinetic processes of protein-protein interactions (PPI) helps to understand how basic building blocks affect overall behavior of living systems. In this study, we used structure-based properties to build predictive models for kinetic constants of PPI. A highly diverse PPI dataset, protein-protein kinetic interaction data and structures (PPKIDS), was built. PPKIDS contains 62 PPI with complex structures and kinetic constants measured experimentally. The influence of structural properties on kinetics of PPI was studied using 35 structure-based features, describing different aspects of complex structures. Linear models for the prediction of kinetic constants were built by fitting with selected subsets of structure-based features. The models gave correlation coefficients of 0.801, 0.732, and 0.770 for k(off), k(on), and K(d), respectively, in leave-one-out cross validations. The predictive models reported here use only protein complex structures as input and can be generally applied in PPI studies as well as systems biology modeling. Our study confirmed that different properties play different roles in the kinetic process of PPI. For example, k(on) was affected by overall structural features of complexes, such as the composition of secondary structures, the change of translational and rotational entropy, and the electrostatic interaction; while k(off) was determined by interfacial properties, such as number of contacted atom pairs per 100 Ų. This information provides useful hints for PPI design.
Collapse
Affiliation(s)
- Hongjun Bai
- Beijing National Laboratory for Molecular Sciences, State Key Laboratory of Structural Chemistry for Stable and Unstable Species, College of Chemistry and Molecular Engineering, Peking University, Beijing 100871, China
| | | | | | | | | | | |
Collapse
|
23
|
Shpilman M, Niv-Spector L, Katz M, Varol C, Solomon G, Ayalon-Soffer M, Boder E, Halpern Z, Elinav E, Gertler A. Development and characterization of high affinity leptins and leptin antagonists. J Biol Chem 2010; 286:4429-42. [PMID: 21119198 DOI: 10.1074/jbc.m110.196402] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Leptin is a pleiotropic hormone acting both centrally and peripherally. It participates in a variety of biological processes, including energy metabolism, reproduction, and modulation of the immune response. So far, structural elements affecting leptin binding to its receptor remain unknown. We employed random mutagenesis of leptin, followed by selection of high affinity mutants by yeast surface display and discovered that replacing residue Asp-23 with a non-negatively charged amino acid leads to dramatically enhanced affinity of leptin for its soluble receptor. Rational mutagenesis of Asp-23 revealed the D23L substitution to be most effective. Coupling the Asp-23 mutation with alanine mutagenesis of three amino acids (L39A/D40A/F41A) previously reported to convert leptin into antagonist resulted in potent antagonistic activity. These novel superactive mouse and human leptin antagonists (D23L/L39A/D40A/F41A), termed SMLA and SHLA, respectively, exhibited over 60-fold increased binding to leptin receptor and 14-fold higher antagonistic activity in vitro relative to the L39A/D40A/F41A mutants. To prolong and enhance in vivo activity, SMLA and SHLA were monopegylated mainly at the N terminus. Administration of the pegylated SMLA to mice resulted in a remarkably rapid, significant, and reversible 27-fold more potent increase in body weight (as compared with pegylated mouse leptin antagonist), because of increased food consumption. Thus, recognition and mutagenesis of Asp-23 enabled construction of novel compounds that induce potent and reversible central and peripheral leptin deficiency. In addition to enhancing our understanding of leptin interactions with its receptor, these antagonists enable in vivo study of the role of leptin in metabolic and immune processes and hold potential for future therapeutic use in disease pathologies involving leptin.
Collapse
Affiliation(s)
- Michal Shpilman
- Robert H Smith Faculty of Agriculture, Food and Environment, Hebrew University, Rehovot 76100, Israel
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Kang L, Bondensgaard K, Li T, Hartmann R, Hjorth SA. Rational design of interleukin-21 antagonist through selective elimination of the gammaC binding epitope. J Biol Chem 2010; 285:12223-31. [PMID: 20167599 DOI: 10.1074/jbc.m110.101444] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The cytokine interleukin (IL)-21 exerts pleiotropic effects acting through innate as well as adaptive immune responses. The activities of IL-21 are mediated through binding to its cognate receptor complex composed of the IL-21 receptor private chain (IL-21Ralpha) and the common gamma-chain (gammaC), the latter being shared by IL-2, IL-4, IL-7, IL-9, and IL-15. The binding energy of the IL-21 ternary complex is predominantly provided by the high affinity interaction between IL-21 and IL-21Ralpha, whereas the interaction between IL-21 and gammaC, albeit essential for signaling, is rather weak. The design of IL-21 analogues, which have lost most or all affinity toward the signaling gammaC chain, while simultaneously maintaining a tight interaction with the private chain, would in theory represent candidates for IL-21 antagonists. We predicted the IL-21 residues, which compose the gammaC binding epitope using homology modeling and alignment with the related cytokines, IL-2 and IL-4. Next we systematically analyzed the predicted binding epitope by a mutagenesis study. Indeed two mutants, which have significantly impaired gammaC affinity with undiminished IL-21Ralpha affinity, were successfully identified. Functional studies confirmed that these two novel hIL-21 double mutants do act as hIL-21 antagonists.
Collapse
Affiliation(s)
- Lishan Kang
- Novo Nordisk China R&D, Beijing 102206, China.
| | | | | | | | | |
Collapse
|
25
|
Rational design and protein engineering of growth factors for regenerative medicine and tissue engineering. Biochem Soc Trans 2009; 37:717-21. [DOI: 10.1042/bst0370717] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Growth factors provide key instructive cues for tissue formation and repair. However, many natural growth factors are limited in their usefulness for tissue engineering and regenerative applications by their poor retention at desired sites of action, short half-lives in vivo, pleiotropic actions and other features. In the present article, we review approaches to rational design of synthetic growth factors based on mechanisms of receptor activation. Such synthetic molecules can function as simplified ligands with potentially tunable specificity and action. Rational and combinatorial protein engineering techniques allow introduction of additional features into these synthetic growth molecules, as well as natural growth factors, which significantly enhance their therapeutic utility.
Collapse
|
26
|
Abstract
Growth hormone (GH) regulates somatic growth, substrate metabolism and body composition. Its actions are elaborated through the GH receptor (GHR). GHR signalling involves the role of at least three major pathways, STATs, MAPK, and PI3-kinase/Akt. GH receptor function can be modulated by changes to the ligand, to the receptor or by factors regulating signal transduction. Insights on the physico-chemical basis of the binding of GH to its receptor and the stoichiometry required for activation of the GH receptor-dimer has led to the development of novel GH agonists and antagonists. Owing to the fact that GH has short half-life, several approaches have been taken to create long-acting GHR agonists. This includes the pegylation, sustained release formulations, and ligand-receptor fusion proteins. Pegylation of a GH analogue (pegvisomant) which binds but not activate signal transduction forms the basis of a new successful approach to the treatment of acromegaly. GH receptors can be regulated at a number of levels, by modifying receptor expression, surface availability and signalling. Insulin, thyroid hormones and sex hormones are among hormones that modulate GHR through some of these mechanisms. Estrogens inhibit GH signalling by stimulating the expression of SOCS proteins which are negative regulators of cytokine receptor signalling. This review of GHR modulators will cover the effects of ligand modification, and of factors regulating receptor expression and signalling.
Collapse
Affiliation(s)
- Vita Birzniece
- Pituitary Research Unit, Garvan Institute of Medical Research and Department of Endocrinology, St. Vincent's Hospital, NSW, Australia
| | | | | |
Collapse
|
27
|
Kotzsch A, Nickel J, Seher A, Sebald W, Müller TD. Crystal structure analysis reveals a spring-loaded latch as molecular mechanism for GDF-5-type I receptor specificity. EMBO J 2009; 28:937-47. [PMID: 19229295 DOI: 10.1038/emboj.2009.37] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2008] [Accepted: 01/23/2009] [Indexed: 01/08/2023] Open
Abstract
Dysregulation of growth and differentiation factor 5 (GDF-5) signalling, a member of the TGF-beta superfamily, is strongly linked to skeletal malformation. GDF-5-mediated signal transduction involves both BMP type I receptors, BMPR-IA and BMPR-IB. However, mutations in either GDF-5 or BMPR-IB lead to similar phenotypes, indicating that in chondrogenesis GDF-5 signalling seems to be exclusively mediated through BMPR-IB. Here, we present structural insights into the GDF-5:BMPR-IB complex revealing how binding specificity for BMPR-IB is generated on a molecular level. In BMPR-IB, a loop within the ligand-binding epitope functions similar to a latch allowing high-affinity binding of GDF-5. In BMPR-IA, this latch is in a closed conformation leading to steric repulsion. The new structural data now provide also a molecular basis of how phenotypically relevant missense mutations in GDF-5 might impair receptor binding and activation.
Collapse
Affiliation(s)
- Alexander Kotzsch
- Lehrstuhl für Botanik I-Molekulare Pflanzenphysiologie und Biophysik, Julius-von-Sachs-Institut für Biowissenschaften (Biozentrum) der Universität Würzburg, Würzburg, Germany
| | | | | | | | | |
Collapse
|
28
|
|
29
|
Kiel C, Aydin D, Serrano L. Association rate constants of ras-effector interactions are evolutionarily conserved. PLoS Comput Biol 2008; 4:e1000245. [PMID: 19096503 PMCID: PMC2588540 DOI: 10.1371/journal.pcbi.1000245] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Accepted: 11/06/2008] [Indexed: 12/31/2022] Open
Abstract
Evolutionary conservation of protein interaction properties has been shown to be a valuable indication for functional importance. Here we use homology interface modeling of 10 Ras-effector complexes by selecting ortholog proteins from 12 organisms representing the major eukaryotic branches, except plants. We find that with increasing divergence time the sequence similarity decreases with respect to the human protein, but the affinities and association rate constants are conserved as predicted by the protein design algorithm, FoldX. In parallel we have done computer simulations on a minimal network based on Ras-effector interactions, and our results indicate that in the absence of negative feedback, changes in kinetics that result in similar binding constants have strong consequences on network behavior. This, together with the previous results, suggests an important biological role, not only for equilibrium binding constants but also for kinetics in signaling processes involving Ras-effector interactions. Our findings are important to take into consideration in system biology approaches and simulations of biological networks. Cellular signal transductions processes are based on protein interactions. Proteins can either associate transiently with each other or form stable complexes, and the strength of the interaction is described by the affinity (the affinity is the ratio between the rate of dissociation and association). Protein complexes with similar affinities can bind and dissociate with different rates, and these rates describe the kinetic properties of protein binding. These kinetic rates are important for signaling; however, to what extent individual changes in such rate constants are biologically important or whether the affinity is more crucial might be different in different signaling processes. In this study we analyze whether association rates are conserved during evolution, because evolutionary conservation of protein biochemical properties is usually a valuable indication of its importance. We analyzed the binding of Ras proteins to effector domains, which are central proteins in many signal transduction pathways, in different organisms. On the basis of homology modeling and energy calculations we find that association rates are conserved, although the sequence similarity decreases compared to the human protein. Our finding should encourage further analysis of the importance of kinetics for cellular signal transduction.
Collapse
Affiliation(s)
- Christina Kiel
- EMBL-CRG Systems Biology Unit, Centre de Regulacio Genomica, Barcelona, Spain.
| | | | | |
Collapse
|
30
|
Kalie E, Jaitin DA, Podoplelova Y, Piehler J, Schreiber G. The Stability of the Ternary Interferon-Receptor Complex Rather than the Affinity to the Individual Subunits Dictates Differential Biological Activities. J Biol Chem 2008; 283:32925-36. [DOI: 10.1074/jbc.m806019200] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
31
|
Jones DS, Silverman AP, Cochran JR. Developing therapeutic proteins by engineering ligand–receptor interactions. Trends Biotechnol 2008; 26:498-505. [DOI: 10.1016/j.tibtech.2008.05.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2008] [Revised: 05/24/2008] [Accepted: 05/27/2008] [Indexed: 12/27/2022]
|
32
|
Higham CE, Trainer PJ. Growth hormone excess and the development of growth hormone receptor antagonists. Exp Physiol 2008; 93:1157-69. [PMID: 18617577 DOI: 10.1113/expphysiol.2008.042515] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In 1990, a single amino acid substitution in the growth hormone (GH) gene at position 119 was found to transform the consequent protein from an agonist to an antagonist at the growth hormone receptor (GHR). Further amino acid substitutions plus prolongation of the half-life of the protein by pegylation resulted in the first clinically effective GHR antagonist, pegvisomant. Following extensive clinical trials, this medication has emerged as the most efficacious therapy for treatment-resistant acromegaly. Subsequent advances in our understanding of GH-GHR interactions and downstream GH signalling pathways suggest that pegvisomant binds to preformed GHR dimers and prevents rotational changes within the receptor-GH complex necessary for intracellular signalling to occur. This article reviews the discovery of pegvisomant, from initial experimental data to successful licensing of the drug for treatment-resistant acromegaly, and discusses its other potential therapeutic uses in diseases with abnormalities in the GH-IGF-I axis.
Collapse
Affiliation(s)
- C E Higham
- Department of Endocrinology, Christie Hospital, Manchester M20 4BX, UK
| | | |
Collapse
|
33
|
Reichmann D, Rahat O, Cohen M, Neuvirth H, Schreiber G. The molecular architecture of protein-protein binding sites. Curr Opin Struct Biol 2007; 17:67-76. [PMID: 17239579 DOI: 10.1016/j.sbi.2007.01.004] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2006] [Revised: 12/13/2006] [Accepted: 01/10/2007] [Indexed: 11/16/2022]
Abstract
The formation of specific protein interactions plays a crucial role in most, if not all, biological processes, including signal transduction, cell regulation, the immune response and others. Recent advances in our understanding of the molecular architecture of protein-protein binding sites, which facilitates such diversity in binding affinity and specificity, are enabling us to address key questions. What is the amino acid composition of binding sites? What are interface hotspots? How are binding sites organized? What are the differences between tight and weak interacting complexes? How does water contribute to binding? Can the knowledge gained be translated into protein design? And does a universal code for binding exist, or is it the architecture and chemistry of the interface that enable diverse but specific binding solutions?
Collapse
Affiliation(s)
- Dana Reichmann
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
34
|
Jaks E, Gavutis M, Uzé G, Martal J, Piehler J. Differential receptor subunit affinities of type I interferons govern differential signal activation. J Mol Biol 2006; 366:525-39. [PMID: 17174979 DOI: 10.1016/j.jmb.2006.11.053] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2006] [Revised: 11/15/2006] [Accepted: 11/15/2006] [Indexed: 12/17/2022]
Abstract
Type I interferons (IFNs) elicit antiviral, antiproliferative and immunmodulatory responses by binding to a shared cell surface receptor comprising the transmembrane proteins ifnar1 and ifnar2. Activation of differential response patterns by IFNs has been observed, suggesting that members of the family play different roles in innate immunity. The molecular basis for differential signaling has not been identified yet. Here, we have investigated the recognition of various IFNs including several human IFNalpha species, human IFNomega and human IFNbeta as well as ovine IFNtau2 by the receptor subunits in detail. Binding to the extracellular domains of ifnar1 (ifnar1-EC) and ifnar2 (ifnar2-EC) was monitored in real time by reflectance interference and total internal reflection fluorescence spectroscopy. For all IFNs investigated, competitive 1:1 interaction not only with ifnar2-EC but also with ifnar1-EC was shown. Furthermore, ternary complex formation was studied with ifnar1-EC and ifnar2-EC tethered onto solid-supported membranes. These analyses confirmed that the signaling complexes recruited by IFNs have very similar architectures. However, differences in rate and affinity constants over several orders of magnitude were observed for both the interactions with ifnar1-EC and ifnar2-EC. These data were correlated with the potencies of ISGF3 activation, antiviral and anti-proliferative activity on 2fTGH cells. The ISGF3 formation and antiviral activity correlated very well with the binding affinity towards ifnar2. In contrast, the affinity towards ifnar1 played a key role for antiproliferative activity. A striking correlation was observed for relative binding affinities towards ifnar1 and ifnar2 with the differential antiproliferative potency. This correlation was confirmed by systematically engineering IFNalpha2 mutants with very high differential antiproliferative potency.
Collapse
Affiliation(s)
- Eva Jaks
- Institute of Biochemistry, Johann Wolfgang Goethe-University Frankfurt am Main, Germany
| | | | | | | | | |
Collapse
|
35
|
Schlee S, Carmillo P, Whitty A. Quantitative analysis of the activation mechanism of the multicomponent growth-factor receptor Ret. Nat Chem Biol 2006; 2:636-44. [PMID: 17013378 DOI: 10.1038/nchembio823] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2006] [Accepted: 08/16/2006] [Indexed: 12/21/2022]
Abstract
Cytokines and growth factors signal by modulating the interactions between multiple receptor components to form an activated receptor complex. The quantitative details of the activation mechanisms of this important class of receptors are not well understood. Using receptor phosphorylation measurements in live cells, as well as mathematical modeling and data fitting, we have characterized the multistep mechanism by which the GDNF-family neurotrophin artemin (ART), together with its co-receptor GDNF-family receptor alpha3 (GFRalpha3), brings about activation of the Ret receptor tyrosine kinase through formation of a pentameric signaling complex: ART-(GFRalpha3)(2)-(Ret)(2). By systematically varying the concentrations of ART and cell-surface GFRalpha3, we establish both the sequence of steps by which the signaling complex forms and the affinities of all the steps, including the two-dimensional affinities of the steps involving protein-protein interactions between membrane-bound species. Our results reveal the ways in which the individual binary interactions involved in the activation of a multicomponent receptor govern the receptor's functional properties.
Collapse
Affiliation(s)
- Sandra Schlee
- Biogen Idec, Inc., 14 Cambridge Center, Cambridge, Massachusetts 02142, USA
| | | | | |
Collapse
|
36
|
Leung MYK, Ho WKK. Substitution of Serine for Non-disulphide-bond-forming Cysteine in Grass Carp (Ctenopharygodon Idellus) Growth Hormone Improves In Vitro Oxidative Renaturation. BMB Rep 2006; 39:150-7. [PMID: 16584629 DOI: 10.5483/bmbrep.2006.39.2.150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Native grass carp (Ctenopharygodon idellus) growth hormone, has 5 cysteine amino acid residues, forms two disulphide bridges in its mature form. Recombinant grass carp growth hormone, when over-expressed in E. coli, forms inclusion bodies. In vitro oxidative renaturation of guanidine-hydrochloride dissolved recombinant grass carp growth hormone was achieved by sequential dilution and stepwise dialysis at pH 8.5. The redox potential of the refolding cocktail was maintained by glutathione disulphide/glutathione couple. The oxidative refolded protein is heterogeneous, and contains multimers, oligomers and monomers. The presence of non-disulphide-bond-forming cysteine in recombinant grass carp growth hormone enhances intermolecular disulphide bond formation and also nonnative intramolecular disulphide bond formation during protein folding. The non-disulphide-bond-forming cysteine was converted to serine by PCR-mediated site-directed mutagenesis. The resulting 4-cysteine grass carp growth hormone has improved in vitro oxidative refolding properties when studied by gel filtration and reverse phase chromatography. The refolded 4-cysteine form has less hydrophobic aggregate and has only one monomeric isoform. Both refolded 4-cysteine and 5-cysteine forms are active in radioreceptor binding assay.
Collapse
Affiliation(s)
- Michael Yiu-Kwong Leung
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region
| | | |
Collapse
|
37
|
Abstract
Growth hormone (GH) is a major regulator of postnatal growth and metabolism. There are extensive clinical applications for GH or its antagonists, including treatments for dwarfism, cancer and metabolic wasting. Owing to this, there is considerable interest in the mechanisms of GH receptor (GHR) activation. It is conventionally thought that GH induces dimerization of two GHR monomers, which initiates intracellular signaling cascades. However, recent studies have provided evidence for a ligand-induced conformational change within constitutively dimerized GHRs being responsible for activating signaling pathways. This review will relate the new model of GHR activation to the activation of related cytokine receptors and discuss the implication of this new model for the design of small GH mimetics and antagonists for therapeutic use.
Collapse
Affiliation(s)
- Rebecca A Pelekanos
- a University of Queensland, Institute for Molecular Bioscience, Building 80, Services Road, St Lucia, Queensland, 4072, Australia.
| | - Michael J Waters
- b University of Queensland, Institute for Molecular Bioscience, Building 80, Services Road, St Lucia, Queensland, 4072, Australia.
| |
Collapse
|
38
|
Hepner F, Csaszar E, Roitinger E, Pollak A, Lubec G. Massspectrometrical analysis of recombinant human growth hormone Norditropin® reveals amino acid exchange at M14_V14 rhGH. Proteomics 2006; 6:775-84. [PMID: 16372270 DOI: 10.1002/pmic.200500334] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recombinant human growth hormone (rhGH) is used for the treatment of several disorders. Structural integrity of rhGH is of critical importance for its clinical use and modifications thereof may act as markers in situations such as rhGH doping, as illegal rhGH-abuse in sports is of increasing interest. In the current study we investigated homogeneity of Norditropin, a recombinant human growth hormone frequently used in medicine, expressed in E. coli, strain MC1061. The most recent proteomics technologies including 2-DE, MALDI-MS followed by MALDI-MS/MS and LC-MS followed by LC-MS/MS were used for the characterisation of rhGH. MALDI-TOF-TOF and electrospray LC-MS analysis revealed one major protein with an average molecular mass of 22 126.0 Da and some additional minor components. Electrospray LC-MS/MS of the enzymatically digested Norditropin sample showed deamidation of N(12)N(149) and N(159), oxidation of M(14), M(125) and M(170) and one amino acid exchange V(14) for M(14) present in <1% of Norditropin. While deamidation and oxidation may be due to technical reasons, the single amino acid exchange may reflect infidelity of translation rather than codon usage and copy editing by E. coli.
Collapse
Affiliation(s)
- Felix Hepner
- Department of Pediatrics, Medical University of Vienna, Vienna, Austria
| | | | | | | | | |
Collapse
|
39
|
Levin AM, Weiss GA. Optimizing the affinity and specificity of proteins with molecular display. MOLECULAR BIOSYSTEMS 2005; 2:49-57. [PMID: 16880922 DOI: 10.1039/b511782h] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Affinity maturation of receptor-ligand interactions represents an important area of academic and pharmaceutical research. Improving affinity and specificity of proteins can tailor potency for both in vivo and in vitro applications. A number of different display platforms including phage display, bacterial and yeast display, ribosome display, and mRNA display can optimize protein affinity and specificity. Here, we will review the advantages and disadvantages of these molecular display methods with a focus on their suitability for protein affinity maturation.
Collapse
Affiliation(s)
- A M Levin
- Department of Chemistry, University of California, Irvine, CA 92697, USA
| | | |
Collapse
|
40
|
Marles JA, Dahesh S, Haynes J, Andrews BJ, Davidson AR. Protein-protein interaction affinity plays a crucial role in controlling the Sho1p-mediated signal transduction pathway in yeast. Mol Cell 2005; 14:813-23. [PMID: 15200958 DOI: 10.1016/j.molcel.2004.05.024] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2004] [Revised: 04/19/2004] [Accepted: 04/28/2004] [Indexed: 01/28/2023]
Abstract
Protein-protein interactions are required for most cellular functions, yet little is known about the relationship between protein-protein interaction affinity and biological activity. To investigate this issue, we engineered a series of mutants that incrementally reduced the affinity of the yeast Sho1p SH3 domain for its in vivo target, the MAP kinase kinase Pbs2p. We demonstrate a strong linear correlation between the binding energy of these mutants and quantitative in vivo outputs from the HOG high-osmolarity response pathway controlled by Sho1p. In addition, we find that reduction in binding affinity for the correct target within this pathway causes a proportional increase in misactivation of the related mating pheromone response pathway and that strong binding affinity alone does not guarantee efficient biological activity. Our experiments also indicate that a second binding surface on the Sho1p SH3 domain is required for its proper in vivo function.
Collapse
Affiliation(s)
- Jennifer A Marles
- Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | |
Collapse
|
41
|
Rao BM, Lauffenburger DA, Wittrup KD. Integrating cell-level kinetic modeling into the design of engineered protein therapeutics. Nat Biotechnol 2005; 23:191-4. [PMID: 15696150 DOI: 10.1038/nbt1064] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Functional genomics and proteomics are identifying many potential drug targets for novel therapeutic proteins, and both rational and combinatorial protein engineering methods are available for creating drug candidates. A central challenge is the definition of the most appropriate design criteria, which will benefit critically from computational kinetic models that incorporate integration from the molecular level to the whole systems level. Interpretation of these processes will require mathematical models that are refined in combination with relevant data derived from quantitative assays, to correctly set biophysical objectives for protein design.
Collapse
Affiliation(s)
- Balaji M Rao
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | |
Collapse
|
42
|
Wan Y, McDevitt A, Shen B, Smythe ML, Waters MJ. Increased Site 1 Affinity Improves Biopotency of Porcine Growth Hormone. J Biol Chem 2004; 279:44775-84. [PMID: 15297460 DOI: 10.1074/jbc.m406092200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Based on phage display optimization studies with human growth hormone (GH), it is thought that the biopotency of GH cannot be increased. This is proposed to be a result of the affinity of the first receptor for hormone far exceeding that which is required to trap the hormone long enough to allow diffusion of the second receptor to form the ternary complex, which initiates signaling. We report here that despite similar site 1 kinetics to the hGH/hGH receptor interaction, the potency of porcine GH for its receptor can be increased up to 5-fold by substituting hGH residues involved in site 1 binding into pGH. Based on extensive mutations and BIAcore studies, we show that the higher potency and site 1 affinity of hGH for the pGHR is primarily a result of a decreased off-rate associated with residues in the extended loop between helices 1 and 2 that interact with the two key tryptophans Trp104 and Trp169 in the receptor binding hot spot. Our mutagenic analysis has also identified a second determinant (Lys165), which in addition to His169, restricts the ability of non-primate hormones to activate hGH receptor. The increased biopotency of GH that we observe can be explained by a model for GH receptor activation where subunit alignment is critical for effective signaling.
Collapse
Affiliation(s)
- Yu Wan
- Institute for Molecular Bioscience and School of Biomedical Sciences, University of Queensland, St. Lucia 4072, Australia
| | | | | | | | | |
Collapse
|
43
|
Lamken P, Lata S, Gavutis M, Piehler J. Ligand-induced Assembling of the Type I Interferon Receptor on Supported Lipid Bilayers. J Mol Biol 2004; 341:303-18. [PMID: 15312780 DOI: 10.1016/j.jmb.2004.05.059] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2004] [Revised: 05/14/2004] [Accepted: 05/17/2004] [Indexed: 11/28/2022]
Abstract
Type I interferons (IFNs) elicit antiviral, antiproliferative and immuno-modulatory responses through binding to a shared receptor consisting of the transmembrane proteins ifnar1 and ifnar2. Differential signaling by different interferons, in particular IFNalphas and IFNbeta, suggests different modes of receptor engagement. Using reflectometric interference spectroscopy (RIfS), we studied kinetics and affinities of the interactions between IFNs and the extracellular receptor domains of ifnar1 (ifnar1-EC) and ifnar2 (ifnar2-EC). For IFNalpha2, we determined a K(D) value of 3 nM and 5 microM for the interaction with ifnar2-EC and ifnar1-EC, respectively. As compared to IFNalpha2, IFNbeta formed complexes with ifnar2-EC as well as ifnar1-EC with substantially higher affinity. For neither IFNalpha2 nor IFNbeta was stabilization of the complex with ifnar1-EC in the presence of soluble ifnar2-EC observed. We investigated ligand-induced complex formation with ifnar1-EC and ifnar2-EC being tethered onto solid-supported, fluid lipid bilayers by RIfS and total internal reflection fluorescence spectroscopy. We observed very stable binding of IFNalpha2 at high receptor surface concentrations with an apparent k(d) value approximately 200 times lower than that for ifnar2-EC alone. The apparent k(d) value was strongly dependent on the surface concentration of the receptor components, suggesting kinetic stabilization. This was corroborated by the fast exchange of labeled IFNalpha2 bound to the receptor by unlabeled IFNalpha2. Taken together, our results indicate that IFN first binds to ifnar2 and subsequently recruits ifnar1 in a transient fashion. In particular, this second step is much more efficient for IFNbeta than for IFNalpha2, which could explain differential activities observed for these IFNs.
Collapse
Affiliation(s)
- Peter Lamken
- Institute of Biochemistry, Johann Wolfgang Goethe-University, Biocenter N210, Marie-Curie-Strasse 9, 60439 Frankfurt am Main, Germany
| | | | | | | |
Collapse
|
44
|
Spies MA, Woodward JJ, Watnik MR, Toney MD. Alanine Racemase Free Energy Profiles from Global Analyses of Progress Curves. J Am Chem Soc 2004; 126:7464-75. [PMID: 15198593 DOI: 10.1021/ja049579h] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Free energy profiles for alanine racemase from Bacillus stearothermophilus have been determined at pH 6.9 and 8.9 from global analysis of racemization progress curves. This required a careful statistical design due to the problems in finding the global minimum in mean square for a system with eight adjustable parameters (i.e., the eight rate constants that describe the stepwise chemical mechanism). The free energy profiles obtained through these procedures are supported by independent experimental evidence: (1). steady-state kinetic constants, (2). solvent viscosity dependence, (3). spectral analysis of reaction intermediates, (4). equilibrium overshoots for progress curves measured in D(2)O, and (5). the magnitudes of calculated intrinsic kinetic isotope effects. The free energy profiles for the enzyme are compared to those of the uncatalyzed and the PLP catalyzed reactions. At pH 6.9, PLP lowers the free energy of activation for deprotonation by 8.4 kcal/mol, while the inclusion of apoenzyme along with PLP additionally lowers it by 11 kcal/mol.
Collapse
Affiliation(s)
- M Ashley Spies
- Department of Chemistry and Statistical Laboratory, University of California, One Shields Avenue, Davis, California 95616, USA
| | | | | | | |
Collapse
|
45
|
Stroud RM, Wells JA. Mechanistic diversity of cytokine receptor signaling across cell membranes. Sci Signal 2004; 2004:re7. [PMID: 15126678 DOI: 10.1126/stke.2312004re7] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Circulating cytokines bind to specific receptors on the cell outer surface to evoke responses inside the cell. Binding of cytokines alters the association between receptor molecules that often cross the membrane only once in a single alpha-helical segment. As a consequence, association of protein domains on the inside of the membrane are also altered. Increasing evidence suggests that an initial "off-state" of associated receptors is perturbed, and brought to an activated state that leads to intracellular signaling and eventually effects a change in DNA transcription. The initial detection event that transduces the change in receptor association is sensitive to both proximity and orientation of the receptors, and probably also to the time that the activated state or receptor association is maintained. Ultimately, a cascade of phosphorylation events is triggered. The initial kinases are sometimes part of the intracellular domains of the receptors. The kinases can also be separate proteins that may be pre-associated with intracellular domains of the receptors, or can be recruited after the intracellular association of the activated receptors. We focus here on each of the cases for which structures of the activated cytokine-receptor complexes are known, in a search for underlying mechanisms. The variations in modes of association, stoichiometries of receptors and cytokines, and orientations before and after activation of these receptors are almost as great as the number of complexes themselves. The principles uncovered nevertheless illustrate the basis for high specificity and fidelity in cytokine-mediated signaling.
Collapse
Affiliation(s)
- Robert M Stroud
- Department of Biochemistry and Biophysics, UCSF Genentech Hall, 600 16th Street, University of California, San Francisco, CA 94143-2240, USA
| | | |
Collapse
|
46
|
Kossiakoff AA. The structural basis for biological signaling, regulation, and specificity in the growth hormone-prolactin system of hormones and receptors. ADVANCES IN PROTEIN CHEMISTRY 2004; 68:147-69. [PMID: 15500861 DOI: 10.1016/s0065-3233(04)68005-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The pituitary hormones growth hormone (GH), prolactin (PRL) and placental lactogen (PL), are members of an extensive cytokine superfamily of hormones and receptors that share many of the same general structure-function relationships in expressing their biological activities. The biology of the pituitary hormones involves a very sophisticated interplay of cross-reactivity and specificity. Biological activity is triggered via a hormone-induced receptor homodimerization process that is regulated by tertiary features of the hormone. These hormones have an asymmetric four alpha-helical bundle structure that gives rise to two receptor binding sites that have distinctly different topographies and electrostatic character. This feature plays an important role in the regulation of these systems by producing binding surfaces with dramatically different binding affinities to the receptor extracellular domains (ECD). As a consequence, the signaling complexes for systems that activate through receptor homodimerization are formed in a controlled sequential step-wise manner. Extensive biochemical and biophysical characterization of the two hormone-receptor interfaces indicate that the energetic properties of the two binding sites are fundamentally different and that the receptor shows extraordinary conformational plasticity to mate with the topographically dissimilar sites on the hormone. An unexpected finding has been that the two hormone binding sites are allosterically coupled; a certain set of mutations in the higher affinity site can produce both conformational and energetic effects in the lower affinity site. These effects are so large that at some level they must have played some role in the evolution of the molecule.
Collapse
Affiliation(s)
- Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, Institute for Biophysical Dynamics, University of Chicago, Cummings Life Sciences Center, Chicago, Illinois 60637, USA
| |
Collapse
|
47
|
Fairlie WD, Uboldi AD, McCoubrie JE, Wang CC, Lee EF, Yao S, De Souza DP, Mifsud S, Metcalf D, Nicola NA, Norton RS, Baca M. Affinity maturation of leukemia inhibitory factor and conversion to potent antagonists of signaling. J Biol Chem 2003; 279:2125-34. [PMID: 14585833 DOI: 10.1074/jbc.m310103200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Leukemia inhibitory factor (LIF)-induced cell signaling occurs following sequential binding to the LIF receptor alpha-chain (LIFR), then to the gp130 co-receptor used by all members of the interleukin-6 family of cytokines. By monovalently displaying human LIF on the surface of M13 phage and randomizing clusters of residues in regions predicted to be important for human LIFR binding, we have identified mutations, which lead to significant increases in affinity for binding to LIFR. Six libraries were constructed in which regions of 4-6 amino acids were randomized then panned against LIFR. Mutations identified in three distinct clusters, residues 53-57, 102-103, and 150-155, gave rise to proteins with significantly increased affinity for binding to both human and mouse LIFR. Combining the mutations for each of these regions further increased the affinity, such that the best mutants bound to human LIFR with >1000-fold higher affinity than wild-type human LIF. NMR analysis indicated that the mutations did not alter the overall structure of the molecule relative to the native protein, although some local changes occurred in the vicinity of the substituted residues. Despite increases in LIFR binding affinity, these mutants did not show any increase in activity as agonists of LIF-induced proliferation of Ba/F3 cells expressing human LIFR and gp130 compared with wild-type LIF. Incorporation of two additional mutations (Q29A and G124R), which were found to abrogate cell signaling, led to the generation of highly potent antagonists of both human and murine LIF-induced bioactivity.
Collapse
Affiliation(s)
- W Douglas Fairlie
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3050, Australia
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Weiss GA, Roth TA, Baldi PF, Sidhu SS. Comprehensive mutagenesis of the C-terminal domain of the M13 gene-3 minor coat protein: the requirements for assembly into the bacteriophage particle. J Mol Biol 2003; 332:777-82. [PMID: 12972250 DOI: 10.1016/s0022-2836(03)00950-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Filamentous bacteriophage assemble at the host membrane in a non-lytic process; the gene-3 minor coat protein (P3) is required for release from the membrane and subsequently, for recognition and infection of a new host. P3 contains at least three distinct domains: two N-terminal domains that mediate host recognition and infection, and a C-terminal domain (P3-C) that is required for release from the host cell following phage assembly and contributes to the structural stability of the phage particle. A comprehensive mutational analysis of the 150 residue P3-C revealed that only 24 side-chains, located within the last 70 residues of sequence, were necessary for efficient incorporation into a wild-type coat. The results reveal that the requirements for the assembly of P3 into the phage particle are quite lax and involve only a few key side-chains. These findings shed light on the functional and structural requirements for filamentous phage assembly, and they may provide guidelines for the engineering of improved coat proteins as scaffolds for phage display technology.
Collapse
Affiliation(s)
- Gregory A Weiss
- Department of Protein Engineering, Genentech Inc., 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | |
Collapse
|
49
|
Duda KM, Brooks CL. Identification of residues outside the two binding sites that are critical for activation of the lactogenic activity of human growth hormone. J Biol Chem 2003; 278:22734-9. [PMID: 12682073 DOI: 10.1074/jbc.m212550200] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Human growth hormone (hGH) binds lactogenic or somatotrophic receptors, creating active heterotrimeric complexes. Comparison of hGH structures, either free or bound to a single lactogenic or somatotrophic receptor, shows binding is associated with structural changes. Changes in hGH structure are unique when binding either lactogenic or somatotrophic receptors and they influence the spatial arrangement of residues constituting the second receptor-binding site. Using site-directed mutagenesis, we identified a contiguous set of largely hydrophobic residues that forms a motif communicating between the two receptor-binding sites of hGH. The residues are external to the receptor-binding epitopes and were identified when their mutation reduced site 2 function without changing site 1 function. The motif includes Phe44, Leu93, Tyr160, Leu163, and Tyr164, located in two hydrophobic clusters between the receptor-binding sites. Their mutation to Glu disrupts hydrophobic interactions and reduces lactogenic activity between 4.7- and 85-fold with little effect on somatotrophic activity or spectroscopic properties. These differential effects indicate that loss of lactogenic activity is not a result of global mis-folding. We propose the loss of lactogenic activity results from disruption of specific hydrophobic clusters that disables the site 1 binding-induced structuring of the second receptor-binding site.
Collapse
Affiliation(s)
- Karen M Duda
- Ohio State Biochemistry Program, Columbus 43221, USA
| | | |
Collapse
|
50
|
Millar DS, Lewis MD, Horan M, Newsway V, Easter TE, Gregory JW, Fryklund L, Norin M, Crowne EC, Davies SJ, Edwards P, Kirk J, Waldron K, Smith PJ, Phillips JA, Scanlon MF, Krawczak M, Cooper DN, Procter AM. Novel mutations of the growth hormone 1 (GH1) gene disclosed by modulation of the clinical selection criteria for individuals with short stature. Hum Mutat 2003; 21:424-40. [PMID: 12655557 DOI: 10.1002/humu.10168] [Citation(s) in RCA: 85] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Subtle mutations in the growth hormone 1 (GH1) gene have been regarded as a comparatively rare cause of short stature. Such lesions were sought in a group of 41 individuals selected for short stature, reduced height velocity, and bone age delay; a group of 11 individuals with short stature and idiopathic growth hormone deficiency (IGHD); and a group of 154 controls. Heterozygous mutations were identified in all three groups but disproportionately in the individuals with short stature, both with (odds ratio 25.2; 95% CI, 5.1-132.2) and without (odds ratio 3.6; 95% CI, 1.0-12.9) IGHD. Twenty-four novel GH1 gene lesions were found. Thirteen novel missense mutations were characterized by assaying the signal transduction activity of in vitro expressed variants; six (T27I, K41R, N47D, S71F, S108R, and T175A) exhibited a reduced ability to activate the JAK/STAT pathway. Molecular modeling suggested that both K41R and T175A might compromise GH receptor binding. Seven GH variants (R16C, K41R, S71F, E74K, Q91L, S108C, and a functional polymorphism, V110I) manifested reduced secretion in rat pituitary cells after allowance had been made for the level of expression attributable to the associated GH1 proximal promoter haplotype. A further leader peptide variant (L-11P) was not secreted. Eleven novel mutations in the GH1 gene promoter were assessed by reporter gene assay but only two, including a GH2 gene-templated gene conversion, were found to be associated with a significantly reduced level of expression. Finally, a novel intron 2 acceptor splice-site mutation, detected in a family with autosomal dominant type II IGHD, was shown to lead to the skipping of exon 3 from the GH1 transcript. A total of 15 novel GH1 gene mutations were thus considered to be of probable phenotypic significance. Such lesions are more prevalent than previously recognized and although most may be insufficient on their own to account for the observed clinical phenotype, they are nevertheless likely to play a contributory role in the etiology of short stature.
Collapse
Affiliation(s)
- David S Millar
- Institute of Medical Genetics, University of Wales College of Medicine, Cardiff, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|