1
|
Howe J, Barbar EJ. Dynamic interactions of dimeric hub proteins underlie their diverse functions and structures: A comparative analysis of 14-3-3 and LC8. J Biol Chem 2025; 301:108416. [PMID: 40107617 PMCID: PMC12017986 DOI: 10.1016/j.jbc.2025.108416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 03/22/2025] Open
Abstract
Hub proteins interact with a host of client proteins and regulate multiple cellular functions. Dynamic hubs have a single binding interface for one client at a time resulting in competition among clients with the highest affinity. Dynamic dimeric hubs with two identical sites bind either two different client proteins or two chains of the same client to form homogenous complexes and could also form heterogeneous mixtures of interconverting complexes. Here, we review the interactions of the dimeric hubs 14-3-3 and LC8. 14-3-3 is a phosphoserine/threonine binding protein involved in structuring client proteins and regulating their phosphorylation. LC8 is involved in promoting the dimerization of client peptides and the rigidification of their disordered regions. Both 14-3-3 and LC8 are essential genes, with 14-3-3 playing a crucial role in apoptosis and cell cycle regulation, while LC8 is critical for the assembly of proteins involved in transport, DNA repair, and transcription. Interestingly, both protein dimers can dissociate by phosphorylation, which results in their interactome-wide changes. Their interactions are also regulated by the phosphorylation of their clients. Both form heterogeneous complexes with various functions including phase separation, signaling, and viral hijacking where they restrict the conformational heterogeneity of their dimeric clients that bind nucleic acids. This comparative analysis highlights the importance of dynamic protein-protein interactions in the diversity of functions of 14-3-3 and LC8 and how small differences in structures of interfaces explain why 14-3-3 is primarily involved in the regulation of phosphorylation states while LC8 is primarily involved in the regulation of assembly of large dynamic complexes.
Collapse
Affiliation(s)
- Jesse Howe
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA
| | - Elisar J Barbar
- Oregon State University, Department of Biochemistry and Biophysics, Corvallis, Oregon, USA.
| |
Collapse
|
2
|
Lin Q, Colussi DM, Stathopulos PB. The apo LETM1 F-EF-hand adopts a closed conformation that underlies a multi-modal sensory role in mitochondria. FEBS Lett 2025; 599:971-988. [PMID: 39927520 PMCID: PMC11995678 DOI: 10.1002/1873-3468.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 02/11/2025]
Abstract
Leucine zipper EF-hand containing transmembrane protein-1 (LETM1) plays a critical role in mitochondrial function, with haploinsufficiency linked to Wolf-Hirschhorn syndrome. Here, we present the solution NMR structure of the calcium (Ca2+)-depleted LETM1 EF-hand domain, revealing a closed conformation facilitated by a distinct F1-helix pivot rather than decreased interhelical angle. Further, we observe regiospecific unfolding in response to hot and cold denaturation and show H662 has a pKa in-line with physiological pH fluctuations. Finally, we demonstrate Ca2+-dependent transient interactions between the EF-hand and other LETM1 or GHITM protein domains. Collectively, our data reveal the apo-to-holo structural dynamics and mechanisms underlying the multi-modal sensing by the LETM1 EF-hand domain, highlighting its role as an adaptable regulatory element within the mitochondrial matrix.
Collapse
Affiliation(s)
- Qi‐Tong Lin
- Department of Physiology and Pharmacology, Schulich School of Medicine and DentistryUniversity of Western OntarioLondonCanada
| | - Danielle M. Colussi
- Department of Physiology and Pharmacology, Schulich School of Medicine and DentistryUniversity of Western OntarioLondonCanada
| | - Peter B. Stathopulos
- Department of Physiology and Pharmacology, Schulich School of Medicine and DentistryUniversity of Western OntarioLondonCanada
| |
Collapse
|
3
|
Turina P, Dal Cortivo G, Enriquez Sandoval CA, Alexov E, Ascher DB, Babbi G, Bakolitsa C, Casadio R, Fariselli P, Folkman L, Kamandula A, Katsonis P, Li D, Lichtarge O, Martelli PL, Panday SK, Pires DEV, Portelli S, Pucci F, Rodrigues CHM, Rooman M, Savojardo C, Schwersensky M, Shen Y, Strokach AV, Sun Y, Woo J, Radivojac P, Brenner SE, Dell'Orco D, Capriotti E. Assessing the predicted impact of single amino acid substitutions in calmodulin for CAGI6 challenges. Hum Genet 2025; 144:113-125. [PMID: 39714488 PMCID: PMC11975486 DOI: 10.1007/s00439-024-02720-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Accepted: 12/02/2024] [Indexed: 12/24/2024]
Abstract
Recent thermodynamic and functional studies have been conducted to evaluate the impact of amino acid substitutions on Calmodulin (CaM). The Critical Assessment of Genome Interpretation (CAGI) data provider at University of Verona (Italy) measured the melting temperature (Tm) and the percentage of unfolding (%unfold) of a set of CaM variants (CaM challenge dataset). Thermodynamic measurements for the equilibrium unfolding of CaM were obtained by monitoring far-UV Circular Dichroism as a function of temperature. These measurements were used to determine the Tm and the percentage of protein remaining unfolded at the highest temperature. The CaM challenge dataset, comprising a total of 15 single amino acid substitutions, was used to evaluate the effectiveness of computational methods in predicting the Tm and unfolding percentages associated with the variants, and categorizing them as destabilizing or not. For the sixth edition of CAGI, nine independent research groups from four continents (Asia, Australia, Europe, and North America) submitted over 52 sets of predictions, derived from various approaches. In this manuscript, we summarize the results of our assessment to highlight the potential limitations of current algorithms and provide insights into the future development of more accurate prediction tools. By evaluating the thermodynamic stability of CaM variants, this study aims to enhance our understanding of the relationship between amino acid substitutions and protein stability, ultimately contributing to more accurate predictions of the effects of genetic variants.
Collapse
Affiliation(s)
- Paola Turina
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Giuditta Dal Cortivo
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy
| | | | - Emil Alexov
- Department of Physics and Astronomy, Clemson University, Clemson, SC, 29634, USA
| | - David B Ascher
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Giulia Babbi
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Constantina Bakolitsa
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
| | - Rita Casadio
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, Turin, Italy
| | - Lukas Folkman
- Institute for Integrated and Intelligent Systems, Griffith University, Southport, QLD, Australia
| | - Akash Kamandula
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Panagiotis Katsonis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Dong Li
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Olivier Lichtarge
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Pier Luigi Martelli
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | | | - Douglas E V Pires
- School of Computing and Information Systems, The University of Melbourne, Melbourne, VIC, 3053, Australia
| | - Stephanie Portelli
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
- School of Chemistry and Molecular Biosciences, Australian Centre for Ecogenomics, University of Queensland, St Lucia, QLD, 4072, Australia
| | - Fabrizio Pucci
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Carlos H M Rodrigues
- Computational Biology and Clinical Informatics, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Marianne Rooman
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Castrense Savojardo
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy
| | - Martin Schwersensky
- Computational Biology and Bioinformatics, Université Libre de Bruxelles, 50 Roosevelt Ave, 1050, Brussels, Belgium
| | - Yang Shen
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | - Alexey V Strokach
- Department of Computer Science, University of Toronto, Toronto, ON, Canada
| | - Yuanfei Sun
- Department of Electrical and Computer Engineering Texas, A&M University, College Station, TX, USA
| | | | - Predrag Radivojac
- Khoury College of Computer Sciences, Northeastern University, Boston, MA, 02115, USA
| | - Steven E Brenner
- Department of Plant and Microbial Biology and Center for Computational Biology, University of California, Berkeley, CA, USA
- Biophysics Graduate Group, University of California, Berkeley, CA, 94720, USA
- Center for Computational Biology, University of California, Berkeley, CA, 94720, USA
| | - Daniele Dell'Orco
- Department of Neurosciences, Biomedicine, and Movement Sciences, Section of Biological Chemistry, University of Verona, 37134, Verona, Italy.
| | - Emidio Capriotti
- Department of Pharmacy and Biotechnology, University of Bologna, 40126, Bologna, Italy.
- Computational Genomics Platform, IRCCS University Hospital of Bologna, 40138, Bologna, Italy.
| |
Collapse
|
4
|
Wang L, Fan Z, Gao K, Qin Y. Integration of transcriptomics and metabolomics of 2-furoic acid-treated Meloidogyne incognita reveals a calmodulin gene associated with motility and pathogenicity. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 220:109480. [PMID: 39827706 DOI: 10.1016/j.plaphy.2025.109480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/30/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
Infections caused by root-knot nematodes (RKNs) significantly impair vegetable growth and crop yield, posing a severe threat to global food security. Our previous study indicated that fungal-derived 2-furoic acid was a promising lead compound for the exploitation of eco-friendly nematicides. However, the exact molecular mechanism remains poorly understood. In this study, we observed behavioral changes in the nematodes following treatment with 100 μg/mL of 2-furoic acid. Subsequently, transcriptomics and metabolomics were combined to identify changes in differentially expressed genes (DEGs) and differentially expressed metabolites (DEMs). Transcriptome analysis revealed 199 up-regulated and 109 down-regulated DEGs. Metabolomics analysis indicated that 140 metabolites (72 up-regulated and 68 down-regulated) exhibited significant differences. Notably, the integrated transcriptomics and metabolomics analysis identified a total of 72 DEGs and 54 DEMs annotated across 51 pathways, including lipid metabolism and amino acid metabolism. Intriguingly, 14 out of the 51 pathways were simultaneously annotated to the downregulated calmodulin gene (cam). RNA interference (RNAi) results demonstrated that the down-regulated cam, a potential drug target for controlling RKNs, significantly reduced the motility, infectivity, and pathogenicity of M. incognita. Our results provide insights into the molecular basis of 2-furoic acid against M. incognita and offer a valuable theoretical foundation for the design of eco-friendly nematicides.
Collapse
Affiliation(s)
- Linsong Wang
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao, 266237, China
| | - Zhaoqian Fan
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao, 266237, China
| | - Kun Gao
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao, 266237, China
| | - Yukun Qin
- CAS and Shandong Province Key Laboratory of Experimental Marine Biology, Center for Ocean Mega-Science, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, 266071, China; Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), No. 1 Wenhai Road, Qingdao, 266237, China.
| |
Collapse
|
5
|
Sabek Y, Zhang Z, Nishibe N, Maruta S. Ionic control of small GTPase HRas using calmodulin. J Biochem 2025; 177:153-161. [PMID: 39696662 DOI: 10.1093/jb/mvae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/20/2024] Open
Abstract
HRas is a small GTPase that plays physiologically important roles in various intracellular signal transduction processes, such as cell growth and proliferation. The structure and action mechanisms of HRas have been well characterized, leading to its widespread use as a molecular switch in bionanomachines. Calmodulin (CaM), a calcium ion-binding protein, acts as an ion-binding molecular switch and activates the target enzymes. We previously demonstrated that the fusion protein of HRas (M13-HRas) with the CaM target peptide M13 at the N-terminus of HRas exhibits reversible regulation of GTPase activity and the interaction between M13-HRas and the downstream signalling factor Raf by calcium ions with CaM. In this study, we prepared two new HRas fusion proteins with the M13 peptide at the C-terminus (HRas-M13) and both termini (M13-HRas-M13) of HRas and analysed the calcium-dependent regulation of HRas function. M13-HRas-M13 more efficiently controlled GTPase, interaction with Raf and the HRas regulator GEF by calcium ions with CaM.
Collapse
Affiliation(s)
- Yassine Sabek
- Department of Biosciences, Graduate School of Science and Engineering, Soka University, 1-236 Tangi-cho, Hachioji, Tokyo 192-8577, Japan
| | - Ziyun Zhang
- Department of Biosciences, Graduate School of Science and Engineering, Soka University, 1-236 Tangi-cho, Hachioji, Tokyo 192-8577, Japan
| | - Nobuyuki Nishibe
- Department of Biosciences, Graduate School of Science and Engineering, Soka University, 1-236 Tangi-cho, Hachioji, Tokyo 192-8577, Japan
| | - Shinsaku Maruta
- Department of Biosciences, Graduate School of Science and Engineering, Soka University, 1-236 Tangi-cho, Hachioji, Tokyo 192-8577, Japan
| |
Collapse
|
6
|
Amemiya Y, Ioi Y, Araki M, Kontani K, Maki M, Shibata H, Takahara T. Calmodulin enhances mTORC1 signaling by preventing TSC2-Rheb binding. J Biol Chem 2025; 301:108122. [PMID: 39716490 PMCID: PMC11787510 DOI: 10.1016/j.jbc.2024.108122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/19/2024] [Accepted: 12/15/2024] [Indexed: 12/25/2024] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) functions as a master regulator of cell growth and proliferation. We previously demonstrated that intracellular calcium ion (Ca2+) concentration modulates the mTORC1 pathway via binding of the Ca2+ sensor protein calmodulin (CaM) to tuberous sclerosis complex 2 (TSC2), a critical negative regulator of mTORC1. However, the precise molecular mechanism by which Ca2+/CaM modulates mTORC1 activity remains unclear. Here, we performed a binding assay based on nano-luciferase reconstitution, a method for detecting weak interactions between TSC2 and its target, Ras homolog enriched in the brain (Rheb), an activator of mTORC1. CaM inhibited the binding of TSC2 to Rheb in a Ca2+-dependent manner. Live-cell imaging analysis indicated increased interaction between the CaM-binding region of TSC2 and CaM in response to elevated intracellular Ca2+ levels. Furthermore, treatment with carbachol, an acetylcholine analog, elevated intracellular Ca2+ levels and activated mTORC1. Notably, carbachol-induced activation of mTORC1 was inhibited by CaM inhibitors, corroborating the role of Ca2+/CaM in promoting the mTORC1 pathway. Consistent with the effect of Ca2+/CaM on the TSC2-Rheb interaction, increased intracellular Ca2+ concentration promoted the dissociation of TSC2 from lysosomes without affecting Akt-dependent phosphorylation of TSC2, suggesting that the regulatory mechanism of TSC2 by Ca2+/CaM is distinct from the previously established action mechanism of TSC2. Collectively, our findings offer mechanistic insights into TSC2-Rheb regulation mediated by Ca2+/CaM, which links Ca2+ signaling to mTORC1 activation.
Collapse
Affiliation(s)
- Yuna Amemiya
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Yuichiro Ioi
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Makoto Araki
- Department of Biochemistry, Meiji Pharmaceutical University, Tokyo, Japan
| | - Kenji Kontani
- Department of Biochemistry, Meiji Pharmaceutical University, Tokyo, Japan
| | - Masatoshi Maki
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Hideki Shibata
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan
| | - Terunao Takahara
- Graduate School of Bioagricultural Sciences, Nagoya University, Nagoya, Japan.
| |
Collapse
|
7
|
Thapa G, Bhattacharya A, Bhattacharya S. Dynamics of AKAP/Calmodulin complex is largely driven by ionic occupancy state. J Mol Graph Model 2025; 134:108904. [PMID: 39547129 DOI: 10.1016/j.jmgm.2024.108904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/28/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024]
Abstract
AKAP79/150 is a scaffold protein found in dendritic spines and other neuronal compartments. It localizes and regulates phosphorylation by protein kinase A and C and is, in turn regulated by Ca2+, mediated by Calmodulin (CaM). Thus, the interaction of AKAP79/150 with CaM is of biological interest. A 2017 study used a peptide cross linking coupled to mass spectrometry (XLMS) to identify the CaM binding site on AKAP79/150 and subsequently solved an X-ray crystallography structure of CaM in complex with a short helical AKAP79/150 peptide. The XRD structure revealed an unusual mixed ionic occupancy state of CaM as bound to the AKAP79/150 peptide. In this molecular dynamics-based study, we have explored the motional modes of the CaM-AKAP helix complex under three ionic occupancy conditions. Our results indicate that the dynamics of this CaM backbone is largely dominated by the ionic occupancy state. We find that binding of the AKAP79/150 peptide to CaM is not preferentially stabilized in energetic terms in the Ca2+ state as compared to apo. However, the Mg2+ state is destabilized energetically as compared to the apo state. In addition, in the Ca2+ state, the AKAP79/150 peptide appears to be preferentially stabilized by additional hydrogen bonds. Our simulations suggest that further structural biology studies should be carried out, with a focus on driving the system equilibrium to full Ca2+ occupancy. NMR studies may be able to capture conformational states which are not seen in crystals.
Collapse
Affiliation(s)
- Gauri Thapa
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| | | | - Swati Bhattacharya
- Department of Chemical Engineering, Indian Institute of Technology Bombay, Mumbai, 400076, India.
| |
Collapse
|
8
|
Sanders JH, Taiwo KM, Adekanye GA, Bali A, Zhang Y, Paulsen CE. Calmodulin binding is required for calcium mediated TRPA1 desensitization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627969. [PMID: 39713425 PMCID: PMC11661184 DOI: 10.1101/2024.12.11.627969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Calcium (Ca2+) ions affect nearly all aspects of biology. Excessive Ca2+ entry is cytotoxic and Ca2+-mobilizing receptors have evolved diverse mechanisms for tight regulation that often include Calmodulin (CaM). TRPA1, an essential Ca2+-permeable ion channel involved in pain signaling and inflammation, exhibits complex Ca2+ regulation with initial channel potentiation followed by rapid desensitization. The molecular mechanisms of TRPA1 Ca2+ regulation and whether CaM plays a role remain elusive. We find that TRPA1 binds CaM best at basal Ca2+ concentration, that they co-localize in resting cells, and that CaM suppresses TRPA1 activity. Combining biochemical, biophysical, modeling, NMR spectroscopy, and functional approaches, we identify an evolutionarily conserved, high-affinity CaM binding element in the distal TRPA1 C-terminus (DCTCaMBE). Genetic or biochemical perturbation of Ca2+/CaM binding to the TRPA1 DCTCaMBE yields hyperactive channels that exhibit drastic slowing of desensitization with no effect on potentiation. Ca2+/CaM TRPA1 regulation does not require the N-lobe, raising the possibility that CaM is not the Ca2+ sensor, per se. Higher extracellular Ca2+ can partially rescue slowed desensitization suggesting Ca2+/CaM binding to the TRPA1 DCTCaMBE primes an intrinsic TRPA1 Ca2+ binding site that, upon binding Ca2+, triggers rapid desensitization. Collectively, our results identify a critical regulatory element in an unstructured TRPA1 region highlighting the importance of these domains, they reveal Ca2+/CaM is an essential TRPA1 auxiliary subunit required for rapid desensitization that establishes proper channel function with implications for all future TRPA1 work, and they uncover a mechanism for receptor regulation by Ca2+/CaM that expands the scope of CaM biology.
Collapse
Affiliation(s)
- Justin H. Sanders
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Kehinde M. Taiwo
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Glory A. Adekanye
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Avnika Bali
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Yuekang Zhang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Candice E. Paulsen
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
9
|
Liu X, Guo P, Yu Q, Gao SQ, Yuan H, Tan X, Lin YW. Site-specific incorporation of 19F-nulcei at protein C-terminus to probe allosteric conformational transitions of metalloproteins. Commun Biol 2024; 7:1613. [PMID: 39627324 PMCID: PMC11615248 DOI: 10.1038/s42003-024-07331-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/27/2024] [Indexed: 12/06/2024] Open
Abstract
Allosteric conformational change is an important paradigm in the regulation of protein function, which is typically triggered by the binding of small cofactors, metal ions or protein partners. Here, we found those conformational transitions can be effectively monitored by 19F NMR, facilitated by a site-specific 19F incorporation strategy at the protein C-terminus using asparaginyl endopeptidase (AEP). Three case studies show that C-terminal 19F-nuclei can reveal protein dynamics not only adjacent but also distal to C-terminus, including those occurring in a hemoprotein neuroglobin (Ngb), calmodulin (CaM), and a cobalt metalloregulator (CoaR) responding to both cobalt and tetrapyrrole. In Ngb, the heme orientation disorder is affected by missense mutations that perturb backbone rigidity or surface charges close to the heme axial ligands. In CaM, the C-terminal 19F-nuclei is an ideal probe for detecting the binding states of Ca2+, peptides and inhibitors. Furthermore, multiple 19F-moieties were incorporated into the two domains of CoaR, revealing the intrinsically disordered C-terminal metal binding tail might be an allosteric conformational switch to maintain cobalt homeostasis and balance corrinoid biosynthesis. This study demonstrates that the AEP-based 19F-modification strategy can be applied to various targets to study allosteric regulation, especially for those biological processes modulated by the protein C-terminus.
Collapse
Affiliation(s)
- Xichun Liu
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China.
| | - Pengfei Guo
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China
| | - Qiufan Yu
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China
| | - Shu-Qin Gao
- Key Lab of Protein Structure and Function of Universities in Hunan Province, Hengyang Medical School, University of South China, Hengyang, China
| | - Hong Yuan
- Department of Chemistry & Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Xiangshi Tan
- Department of Chemistry & Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Ying-Wu Lin
- School of Chemistry and Chemical Engineering, University of South China, Hengyang, China.
- Key Lab of Protein Structure and Function of Universities in Hunan Province, Hengyang Medical School, University of South China, Hengyang, China.
| |
Collapse
|
10
|
Dutta S, Basu R, Pal A, Kunalika MH, Chattopadhyay S. The homeostasis of AtMYB4 is maintained by ARA4, HY5, and CAM7 during Arabidopsis seedling development. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 120:2515-2535. [PMID: 39526498 DOI: 10.1111/tpj.17126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 09/24/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Calmodulin7 (CAM7) is a key transcription factor of Arabidopsis seedling development. CAM7 works together with HY5 bZIP protein to promote photomorphogenesis at various wavelengths of light. In this study, we show that AtMYB4, identified from a yeast two-hybrid screen, physically interacts with CAM7 and works as a positive regulator of photomorphogenesis at various wavelengths of light. CAM7 and HY5 directly bind to the promoter of AtMYB4 to promote its expression for photomorphogenic growth. On the other hand, ARA4, identified from the same yeast two-hybrid screen, works as a negative regulator of photomorphogenic growth specifically in white light. The double mutant analysis reveals that the altered hypocotyl elongation of atmyb4 and ara4 is either partly or completely suppressed by additional loss of function of CAM7. Furthermore, ARA4 genetically interacts with AtMYB4 in an antagonistic manner to suppress the elongated hypocotyl phenotype of atmyb4. The transactivation studies reveal that while CAM7 activates the promoter of AtMYB4 in association with HY5, ARA4 negatively regulates AtMYB4 expression. Taken together, these results demonstrate that working as a negative regulator of photomorphogenesis, ARA4 plays a balancing act on CAM7 and HY5-mediated regulation of AtMYB4.
Collapse
Affiliation(s)
- Siddhartha Dutta
- Department of Biotechnology, National Institute of Technology, Durgapur, 713209, India
| | - Riya Basu
- Department of Biotechnology, National Institute of Technology, Durgapur, 713209, India
| | - Abhideep Pal
- Department of Biotechnology, National Institute of Technology, Durgapur, 713209, India
| | - M H Kunalika
- Department of Biotechnology, National Institute of Technology, Durgapur, 713209, India
| | - Sudip Chattopadhyay
- Department of Biotechnology, National Institute of Technology, Durgapur, 713209, India
| |
Collapse
|
11
|
Daniel-Mozo M, Rombolá-Caldentey B, Mendoza I, Ragel P, De Luca A, Carranco R, Alcaide AM, Ausili A, Cubero B, Schumacher K, Quintero FJ, Albert A, Pardo JM. The vacuolar K +/H + exchangers and calmodulin-like CML18 constitute a pH-sensing module that regulates K + status in Arabidopsis. SCIENCE ADVANCES 2024; 10:eadp7658. [PMID: 39536104 PMCID: PMC11559620 DOI: 10.1126/sciadv.adp7658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Shifts in cytosolic pH have been recognized as key signaling events and mounting evidence supports the interdependence between H+ and Ca2+ signaling in eukaryotic cells. Among the cellular pH-stats, K+/H+ exchange at various membranes is paramount in plant cells. Vacuolar K+/H+ exchangers of the NHX (Na+,K+/H+ exchanger) family control luminal pH and, together with K+ and H+ transporters at the plasma membrane, have been suggested to also regulate cytoplasmic pH. We show the regulation of vacuolar K+/H+ exchange by cytoplasmic pH and the calmodulin-like protein CML18 in Arabidopsis. The crystal structure and physicochemical properties of CML18 indicate that this protein senses pH shifts. Interaction of CML18 with tonoplast exchangers NHX1 and NHX2 was favored at acidic pH, a physiological condition elicited by K+ starvation in Arabidopsis roots, whereas excess K+ produced cytoplasmic alkalinization and CML18 dissociation. These results imply that the pH-responsive NHX-CML18 module is an essential component of the cellular K+- and pH-stats.
Collapse
Affiliation(s)
- Miguel Daniel-Mozo
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - Belén Rombolá-Caldentey
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Imelda Mendoza
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Paula Ragel
- Centre for Organismal Studies, University of Heidelberg, Heidelberg 69120, Germany
| | - Anna De Luca
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Raul Carranco
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Ana M. Alcaide
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Alessio Ausili
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Beatriz Cubero
- Instituto de Recursos Naturales y Agrobiología, Consejo Superior de Investigaciones Cientificas, Seville 41012, Spain
| | - Karin Schumacher
- Centre for Organismal Studies, University of Heidelberg, Heidelberg 69120, Germany
| | - Francisco J. Quintero
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| | - Armando Albert
- Instituto de Química Física Blas Cabrera, Consejo Superior de Investigaciones Científicas, Madrid 28006, Spain
| | - José M. Pardo
- Institute of Plant Biochemistry and Photosynthesis, Consejo Superior de Investigaciones Cientificas and University of Seville, Seville 41092, Spain
| |
Collapse
|
12
|
Kumari D, Prasad BD, Dwivedi P. Genome-wide analysis of calmodulin binding Protein60 candidates in the important crop plants. Mol Biol Rep 2024; 51:1105. [PMID: 39476040 DOI: 10.1007/s11033-024-10032-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/15/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND Efficient management of environmental stresses is essential for sustainable crop production. Calcium (Ca²⁺) signaling plays a crucial role in regulating responses to both biotic and abiotic stresses, particularly during host-pathogen interactions. In Arabidopsis thaliana, calmodulin-binding protein 60 (CBP60) family members, such as AtCBP60g, AtCBP60a, and AtSARD1, have been well characterized for their involvement in immune regulation. However, a comprehensive understanding of CBP60 genes in major crops remains limited. METHODS In this study, we utilized the Phytozome v12.1 database to identify and analyze CBP60 genes in agriculturally important crops. Expression patterns of a Oryza sativa (rice) CBP60 gene, OsCBP60bcd-1, were assessed in resistant and susceptible rice genotypes in response to infection by the bacterial pathogen Xanthomonas oryzae. Localization of CBP60 proteins was analyzed to predict their functional roles, and computational promoter analysis was performed to identify stress-responsive cis-regulatory elements. RESULTS Phylogenetic analysis revealed that most CBP60 genes in crops belong to the immune-related clade. Expression analysis showed that OsCBP60bcd-1 was significantly upregulated in the resistant rice genotype upon pathogen infection. Subcellular localization studies suggested that the majority of CBP60 proteins are nuclear-localized, indicating a potential role as transcription factors. Promoter analysis identified diverse stress-responsive cis-regulatory elements in the promoters of CBP60 genes, highlighting their regulatory potential under stress conditions. CONCLUSION The upregulation of OsCBP60bcd-1 in response to Xanthomonas oryzae and the presence of stress-responsive elements in its promoter underscore the importance of CBP60 genes in pathogen defense. These findings provide a basis for further investigation into the functional roles of CBP60 genes in crop disease resistance, with implications for enhancing stress resilience in agricultural species.
Collapse
Affiliation(s)
- Diksha Kumari
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Bishun Deo Prasad
- Department of Agricultural Biotechnology & Molecular Biology, College of Basic Sciences & Humanities, Dr. Rajendra Prasad Central Agricultural University, Pusa, Samastipur, India.
| | - Padmanabh Dwivedi
- Department of Plant Physiology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
13
|
Li J, Meng S, Zhang Z, Wang Y, Li Z, Yan S, Shen J, Liu X, Zhang S. Nanoparticle-mediated calmodulin dsRNA and cyantraniliprole co-delivery system: High-efficient control of two key pear pests while ensuring safety for natural enemy insects. Int J Biol Macromol 2024; 277:134478. [PMID: 39102908 DOI: 10.1016/j.ijbiomac.2024.134478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/26/2024] [Accepted: 08/02/2024] [Indexed: 08/07/2024]
Abstract
Currently, the predominant method for managing pests in orchards is chemical control. However, prolonged use of chemicals leads to resistance issues and raise ecological safety. A promising approach to tackle these challenges involves nanoparticles-mediated delivery system of dsRNA and pesticides. Despite its potential, this strategy has not been widely applied in controlling pests in pear orchards. In this study, we developed a nanoparticle-mediated ternary biopesticide to tackle resistance and safety concerns associated with calmodulin dsRNA and cyantraniliprole. Initially, we assessed the effectiveness of cyantraniliprole against two key pear pests, Grapholita molesta and Cacopsylla chinensis. Subsequently, we observed an upregualtion of genes CaM and CN following cyantraniliprole treatment. Furthermore, inhibiting or silencing GmCaM and CcGaM enhanced the sensitivity to cyantraniliprole more effectively. By introducing hairpin RNA into the pET30a-BL21 RNaseIII- system to silence GmCaM and CcCaM, we developed a nanoparticle-mediated co-delivery system that exhibited improved control over these two pests. Importantly, our research demonstrated that using reduced cyantraniliprole dosages through ternary biopesticides could help mitigate risks to natural enemies. Overall, our research emphasizes the enhanced effectiveness of ternary biopesticides in boosting the performance of dsRNA and pesticide against pear pests, while fostering environmental sustainability-a novel advancement in this field.
Collapse
Affiliation(s)
- Jianying Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shili Meng
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhixian Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Yilin Wang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Zhen Li
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Shuo Yan
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Jie Shen
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China
| | - Xiaoxia Liu
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China.
| | - Songdou Zhang
- Department of Entomology and MOA Key Lab of Pest Monitoring and Green Management, College of Plant Protection, China Agricultural University, Beijing, China; Sanya Institute of China Agricultural University, 572025 Sanya City, Hainan Province, China.
| |
Collapse
|
14
|
Scollo F, Tempra C, Evci H, Riopedre-Fernandez M, Olżyńska A, Javanainen M, Uday A, Cebecauer M, Cwiklik L, Martinez-Seara H, Jungwirth P, Jurkiewicz P, Hof M. Can calmodulin bind to lipids of the cytosolic leaflet of plasma membranes? Open Biol 2024; 14:240067. [PMID: 39288811 PMCID: PMC11500697 DOI: 10.1098/rsob.240067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 09/19/2024] Open
Abstract
Calmodulin (CaM) is a ubiquitous calcium-sensitive messenger in eukaryotic cells. It was previously shown that CaM possesses an affinity for diverse lipid moieties, including those found on CaM-binding proteins. These facts, together with our observation that CaM accumulates in membrane-rich protrusions of HeLa cells upon increased cytosolic calcium, motivated us to perform a systematic search for unmediated CaM interactions with model lipid membranes mimicking the cytosolic leaflet of plasma membranes. A range of experimental techniques and molecular dynamics simulations prove unambiguously that CaM interacts with lipid bilayers in the presence of calcium ions. The lipids phosphatidylserine (PS) and phosphatidylethanolamine (PE) hold the key to CaM-membrane interactions. Calcium induces an essential conformational rearrangement of CaM, but calcium binding to the headgroup of PS also neutralizes the membrane negative surface charge. More intriguingly, PE plays a dual role-it not only forms hydrogen bonds with CaM, but also destabilizes the lipid bilayer increasing the exposure of hydrophobic acyl chains to the interacting proteins. Our findings suggest that upon increased intracellular calcium concentration, CaM and the cytosolic leaflet of cellular membranes can be functionally connected.
Collapse
Affiliation(s)
- Federica Scollo
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Carmelo Tempra
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 166 10 Prague 6, Czech Republic
| | - Hüseyin Evci
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
- Department of Chemistry, Faculty of Science, University of South Bohemia in České Budějovice, 370 05 České Budějovice, Czech Republic
| | - Miguel Riopedre-Fernandez
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 166 10 Prague 6, Czech Republic
| | - Agnieszka Olżyńska
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Matti Javanainen
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 166 10 Prague 6, Czech Republic
- Institute of Biotechnology, University of Helsinki, 00790 Helsinki, Finland
| | - Arunima Uday
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Marek Cebecauer
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Lukasz Cwiklik
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Hector Martinez-Seara
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 166 10 Prague 6, Czech Republic
| | - Pavel Jungwirth
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo nam. 2, 166 10 Prague 6, Czech Republic
| | - Piotr Jurkiewicz
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| | - Martin Hof
- J. Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 2155/3, 182 23 Prague 8, Czech Republic
| |
Collapse
|
15
|
O’Day DH. Calcium and Non-Penetrating Traumatic Brain Injury: A Proposal for the Implementation of an Early Therapeutic Treatment for Initial Head Insults. Biomolecules 2024; 14:853. [PMID: 39062567 PMCID: PMC11274459 DOI: 10.3390/biom14070853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/04/2024] [Accepted: 07/13/2024] [Indexed: 07/28/2024] Open
Abstract
Finding an effective treatment for traumatic brain injury is challenging for multiple reasons. There are innumerable different causes and resulting levels of damage for both penetrating and non-penetrating traumatic brain injury each of which shows diverse pathophysiological progressions. More concerning is that disease progression can take decades before neurological symptoms become obvious. Currently, the primary treatment for non-penetrating mild traumatic brain injury, also called concussion, is bed rest despite the fact the majority of emergency room visits for traumatic brain injury are due to this mild form. Furthermore, one-third of mild traumatic brain injury cases progress to long-term serious symptoms. This argues for the earliest therapeutic intervention for all mild traumatic brain injury cases which is the focus of this review. Calcium levels are greatly increased in damaged brain regions as a result of the initial impact due to tissue damage as well as disrupted ion channels. The dysregulated calcium level feedback is a diversity of ways to further augment calcium neurotoxicity. This suggests that targeting calcium levels and function would be a strong therapeutic approach. An effective calcium-based traumatic brain injury therapy could best be developed through therapeutic programs organized in professional team sports where mild traumatic brain injury events are common, large numbers of subjects are involved and professional personnel are available to oversee treatment and documentation. This review concludes with a proposal with that focus.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
16
|
Hong L, Kortemme T. An integrative approach to protein sequence design through multiobjective optimization. PLoS Comput Biol 2024; 20:e1011953. [PMID: 38991035 PMCID: PMC11265717 DOI: 10.1371/journal.pcbi.1011953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/23/2024] [Accepted: 06/25/2024] [Indexed: 07/13/2024] Open
Abstract
With recent methodological advances in the field of computational protein design, in particular those based on deep learning, there is an increasing need for frameworks that allow for coherent, direct integration of different models and objective functions into the generative design process. Here we demonstrate how evolutionary multiobjective optimization techniques can be adapted to provide such an approach. With the established Non-dominated Sorting Genetic Algorithm II (NSGA-II) as the optimization framework, we use AlphaFold2 and ProteinMPNN confidence metrics to define the objective space, and a mutation operator composed of ESM-1v and ProteinMPNN to rank and then redesign the least favorable positions. Using the two-state design problem of the foldswitching protein RfaH as an in-depth case study, and PapD and calmodulin as examples of higher-dimensional design problems, we show that the evolutionary multiobjective optimization approach leads to significant reduction in the bias and variance in RfaH native sequence recovery, compared to a direct application of ProteinMPNN. We suggest that this improvement is due to three factors: (i) the use of an informative mutation operator that accelerates the sequence space exploration, (ii) the parallel, iterative design process inherent to the genetic algorithm that improves upon the ProteinMPNN autoregressive sequence decoding scheme, and (iii) the explicit approximation of the Pareto front that leads to optimal design candidates representing diverse tradeoff conditions. We anticipate this approach to be readily adaptable to different models and broadly relevant for protein design tasks with complex specifications.
Collapse
Affiliation(s)
- Lu Hong
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
| | - Tanja Kortemme
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, United States of America
- Quantitative Biosciences Institute, University of California, San Francisco, California, United States of America
- Chan Zuckerberg Biohub, San Francisco, California, United States of America
| |
Collapse
|
17
|
Greene D, Shiferaw Y. Identifying Key Binding Interactions Between the Cardiac L-Type Calcium Channel and Calmodulin Using Molecular Dynamics Simulations. J Phys Chem B 2024; 128:6097-6111. [PMID: 38870543 PMCID: PMC11215769 DOI: 10.1021/acs.jpcb.4c02251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/15/2024]
Abstract
Defects in the binding of the calcium sensing protein calmodulin (CaM) to the L-type calcium channel (CaV1.2) or to the ryanodine receptor type 2 (RyR2) can lead to dangerous cardiac arrhythmias with distinct phenotypes, such as long-QT syndrome (LQTS) and catecholaminergic ventricular tachycardia (CPVT). Certain CaM mutations lead to LQTS while other mutations lead to CPVT, but the mechanisms by which a specific mutation can lead to each disease phenotype are not well-understood. In this study, we use long, 2 μs molecular dynamics simulations and a multitrajectory approach to identify the key binding interactions between the IQ domain of CaV1.2 and CaM. Five key interactions are found between CaV1.2 and CaM in the C-lobe, 1 in the central linker, and 2 in the N-lobe. In addition, while 5 key interactions appear between residues 120-149 in the C-lobe of CaM when it interacts with CaV1.2, only 1 key interaction is found within this region of CaM when it interacts with the RyR2. We show that this difference in the distribution of key interactions correlates with the known distribution of CaM mutations that lead to LQTS or CPVT. This correlation suggests that a disruption of key binding interactions is a plausible mechanism that can lead to these two different disease phenotypes.
Collapse
Affiliation(s)
- D’Artagnan Greene
- Department of Physics and
Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, California 91330-8268, United States of
America
| | - Yohannes Shiferaw
- Department of Physics and
Astronomy, California State University Northridge, 18111 Nordhoff Street, Northridge, California 91330-8268, United States of
America
| |
Collapse
|
18
|
Thines L, Jang H, Li Z, Sayedyahossein S, Maloney R, Nussinov R, Sacks DB. Disruption of Ca 2+/calmodulin:KSR1 interaction lowers ERK activation. Protein Sci 2024; 33:e4982. [PMID: 38591710 PMCID: PMC11002989 DOI: 10.1002/pro.4982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/22/2024] [Accepted: 03/16/2024] [Indexed: 04/10/2024]
Abstract
KSR1, a key scaffold protein for the MAPK pathway, facilitates ERK activation upon growth factor stimulation. We recently demonstrated that KSR1 binds the Ca2+-binding protein calmodulin (CaM), thereby providing an intersection between KSR1-mediated and Ca2+ signaling. In this study, we set out to generate a KSR1 point mutant with reduced Ca2+/CaM binding in order to unravel the functional implications of their interaction. To do so, we solved the structural determinants of complex formation. Using purified fragments of KSR1, we showed that Ca2+/CaM binds to the CA3 domain of KSR1. We then used in silico molecular modeling to predict contact residues for binding. This approach identified two possible modes of interaction: (1) binding of extended Ca2+/CaM to a globular conformation of KSR1-CA3 via electrostatic interactions or (2) binding of collapsed Ca2+/CaM to α-helical KSR1-CA3 via hydrophobic interactions. Experimentally, site-directed mutagenesis of the predicted contact residues for the two binding models favored that where collapsed Ca2+/CaM binds to the α-helical conformation of KSR1-CA3. Importantly, replacing KSR1-Phe355 with Asp reduces Ca2+/CaM binding by 76%. The KSR1-F355D mutation also significantly impairs the ability of EGF to activate ERK, which reveals that Ca2+/CaM binding promotes KSR1-mediated MAPK signaling. This work, by uncovering structural insight into the binding of KSR1 to Ca2+/CaM, identifies a KSR1 single-point mutant as a bioreagent to selectively study the crosstalk between Ca2+ and KSR1-mediated signaling.
Collapse
Affiliation(s)
- Louise Thines
- Department of Laboratory MedicineNational Institutes of HealthBethesdaMarylandUSA
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
| | - Zhigang Li
- Department of Laboratory MedicineNational Institutes of HealthBethesdaMarylandUSA
| | - Samar Sayedyahossein
- Department of Laboratory MedicineNational Institutes of HealthBethesdaMarylandUSA
| | - Ryan Maloney
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation LaboratoryNational Cancer InstituteFrederickMarylandUSA
- Department of Human Molecular Genetics and BiochemistrySackler School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - David B. Sacks
- Department of Laboratory MedicineNational Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
19
|
Yang X, Zhu M, Lu X, Wang Y, Xiao J. Architecture and activation of human muscle phosphorylase kinase. Nat Commun 2024; 15:2719. [PMID: 38548794 PMCID: PMC10978961 DOI: 10.1038/s41467-024-47049-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 03/18/2024] [Indexed: 04/01/2024] Open
Abstract
The study of phosphorylase kinase (PhK)-regulated glycogen metabolism has contributed to the fundamental understanding of protein phosphorylation; however, the molecular mechanism of PhK remains poorly understood. Here we present the high-resolution cryo-electron microscopy structures of human muscle PhK. The 1.3-megadalton PhK α4β4γ4δ4 hexadecamer consists of a tetramer of tetramer, wherein four αβγδ modules are connected by the central β4 scaffold. The α- and β-subunits possess glucoamylase-like domains, but exhibit no detectable enzyme activities. The α-subunit serves as a bridge between the β-subunit and the γδ subcomplex, and facilitates the γ-subunit to adopt an autoinhibited state. Ca2+-free calmodulin (δ-subunit) binds to the γ-subunit in a compact conformation. Upon binding of Ca2+, a conformational change occurs, allowing for the de-inhibition of the γ-subunit through a spring-loaded mechanism. We also reveal an ADP-binding pocket in the β-subunit, which plays a role in allosterically enhancing PhK activity. These results provide molecular insights of this important kinase complex.
Collapse
Affiliation(s)
- Xiaoke Yang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, P.R. China
| | - Mingqi Zhu
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, P.R. China
| | - Xue Lu
- Changping Laboratory, Beijing, P.R. China
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, P.R. China
| | - Yuxin Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, P.R. China
| | - Junyu Xiao
- State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, P.R. China.
- Changping Laboratory, Beijing, P.R. China.
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, P.R. China.
| |
Collapse
|
20
|
Lisek M, Tomczak J, Boczek T, Zylinska L. Calcium-Associated Proteins in Neuroregeneration. Biomolecules 2024; 14:183. [PMID: 38397420 PMCID: PMC10887043 DOI: 10.3390/biom14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/01/2024] [Indexed: 02/25/2024] Open
Abstract
The dysregulation of intracellular calcium levels is a critical factor in neurodegeneration, leading to the aberrant activation of calcium-dependent processes and, ultimately, cell death. Ca2+ signals vary in magnitude, duration, and the type of neuron affected. A moderate Ca2+ concentration can initiate certain cellular repair pathways and promote neuroregeneration. While the peripheral nervous system exhibits an intrinsic regenerative capability, the central nervous system has limited self-repair potential. There is evidence that significant variations exist in evoked calcium responses and axonal regeneration among neurons, and individual differences in regenerative capacity are apparent even within the same type of neurons. Furthermore, some studies have shown that neuronal activity could serve as a potent regulator of this process. The spatio-temporal patterns of calcium dynamics are intricately controlled by a variety of proteins, including channels, ion pumps, enzymes, and various calcium-binding proteins, each of which can exert either positive or negative effects on neural repair, depending on the cellular context. In this concise review, we focus on several calcium-associated proteins such as CaM kinase II, GAP-43, oncomodulin, caldendrin, calneuron, and NCS-1 in order to elaborate on their roles in the intrinsic mechanisms governing neuronal regeneration following traumatic damage processes.
Collapse
Affiliation(s)
| | | | | | - Ludmila Zylinska
- Department of Molecular Neurochemistry, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.); (J.T.); (T.B.)
| |
Collapse
|
21
|
O’Day DH. The Complex Interplay between Toxic Hallmark Proteins, Calmodulin-Binding Proteins, Ion Channels, and Receptors Involved in Calcium Dyshomeostasis in Neurodegeneration. Biomolecules 2024; 14:173. [PMID: 38397410 PMCID: PMC10886625 DOI: 10.3390/biom14020173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 01/23/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Calcium dyshomeostasis is an early critical event in neurodegeneration as exemplified by Alzheimer's (AD), Huntington's (HD) and Parkinson's (PD) diseases. Neuronal calcium homeostasis is maintained by a diversity of ion channels, buffers, calcium-binding protein effectors, and intracellular storage in the endoplasmic reticulum, mitochondria, and lysosomes. The function of these components and compartments is impacted by the toxic hallmark proteins of AD (amyloid beta and Tau), HD (huntingtin) and PD (alpha-synuclein) as well as by interactions with downstream calcium-binding proteins, especially calmodulin. Each of the toxic hallmark proteins (amyloid beta, Tau, huntingtin, and alpha-synuclein) binds to calmodulin. Multiple channels and receptors involved in calcium homeostasis and dysregulation also bind to and are regulated by calmodulin. The primary goal of this review is to show the complexity of these interactions and how they can impact research and the search for therapies. A secondary goal is to suggest that therapeutic targets downstream from calcium dyshomeostasis may offer greater opportunities for success.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
22
|
Holler CV, Petersson NM, Brohus M, Niemelä MA, Iversen ED, Overgaard MT, Iwaï H, Wimmer R. Allosteric changes in protein stability and dynamics as pathogenic mechanism for calmodulin variants not affecting Ca 2+ coordinating residues. Cell Calcium 2024; 117:102831. [PMID: 37995470 DOI: 10.1016/j.ceca.2023.102831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/10/2023] [Accepted: 11/12/2023] [Indexed: 11/25/2023]
Abstract
Mutations in the small, calcium-sensing, protein calmodulin cause cardiac arrhythmia and can ultimately prove lethal. Here, we report the impact of the G113R variant on the structure and dynamics of the calmodulin molecule, both in the presence and in the absence of calcium. We show that the mutation introduces minor changes into the structure of calmodulin and that it changes the thermostability and thus the degree of foldedness at human body temperature. The mutation also severely impacts the intramolecular mobility of calmodulin, especially in the apo form. Glycine 113 acts as an alpha-helical C-capping residue in both apo/ - and Ca2+/calmodulin, but its exchange to arginine has very different effects on the apo and Ca2+ forms. The majority of arrhythmogenic calmodulin variants identified affects residues in the Ca2+ coordinating loops of the two C-domain EF-Hands, causing a 'direct impact on Ca2+ binding'. However, G113R lies outside a Ca2+ coordinating loop and acts differently and more similar to the previously characterized arrhythmogenic N53I. Therefore, we suggest that altered apo/CaM dynamics may be a novel general disease mechanism, defining low-calcium target affinity - or Ca2+ binding kinetics - critical for timely coordination of essential ion-channels in the excitation-contraction cycle.
Collapse
Affiliation(s)
- Christina Vallentin Holler
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Nina Møller Petersson
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Malene Brohus
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | | | - Emil Drivsholm Iversen
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Michael Toft Overgaard
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark
| | - Hideo Iwaï
- Institute of Biotechnology, University of Helsinki, PO Box 65, Helsinki, FIN-00014, Finland
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, Frederik Bajers vej 7H, 9220 Aalborg, Denmark.
| |
Collapse
|
23
|
Saini G, Parasa MK, Clayton KN, Fraseur JG, Bolton SC, Lin KP, Wereley ST, Kinzer-Ursem TL. Immobilization of azide-functionalized proteins to micro- and nanoparticles directly from cell lysate. Mikrochim Acta 2023; 191:46. [PMID: 38129631 PMCID: PMC10739308 DOI: 10.1007/s00604-023-06068-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023]
Abstract
Immobilization of proteins and enzymes on solid supports has been utilized in a variety of applications, from improved protein stability on supported catalysts in industrial processes to fabrication of biosensors, biochips, and microdevices. A critical requirement for these applications is facile yet stable covalent conjugation between the immobilized and fully active protein and the solid support to produce stable, highly bio-active conjugates. Here, we report functionalization of solid surfaces (gold nanoparticles and magnetic beads) with bio-active proteins using site-specific and biorthogonal labeling and azide-alkyne cycloaddition, a click chemistry. Specifically, we recombinantly express and selectively label calcium-dependent proteins, calmodulin and calcineurin, and cAMP-dependent protein kinase A (PKA) with N-terminal azide-tags for efficient conjugation to nanoparticles and magnetic beads. We successfully immobilized the proteins on to the solid supports directly from the cell lysate with click chemistry, forgoing the step of purification. This approach is optimized to yield low particle aggregation and high levels of protein activity post-conjugation. The entire process enables streamlined workflows for bioconjugation and highly active conjugated proteins.
Collapse
Affiliation(s)
- Gunjan Saini
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Mrugesh Krishna Parasa
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Katherine N Clayton
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Julia G Fraseur
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Scott C Bolton
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Kevin P Lin
- Department of Biochemistry, Purdue University, West Lafayette, IN, 47906, USA
| | - Steven T Wereley
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, 47906, USA
| | - Tamara L Kinzer-Ursem
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47906, USA.
| |
Collapse
|
24
|
Liu TT, Xu HH, Liu ZJ, Zhang HP, Zhou HT, Zhu ZX, Wang ZQ, Xue JY, Li Q, Ma Y, You HJ, Luo DL. Downregulated calmodulin expression contributes to endothelial cell impairment in diabetes. Acta Pharmacol Sin 2023; 44:2492-2503. [PMID: 37468692 PMCID: PMC10692162 DOI: 10.1038/s41401-023-01127-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/11/2023] [Indexed: 07/21/2023]
Abstract
Endothelial dysfunction, a central hallmark of cardiovascular pathogenesis in diabetes mellitus, is characterized by impaired endothelial nitric oxide synthase (eNOS) and NO bioavailability. However, the underlying mechanisms remain unclear. Here in this study, we aimed to identify the role of calmodulin (CaM) in diabetic eNOS dysfunction. Human umbilical vein endothelial cells and murine endothelial progenitor cells (EPCs) treated with high glucose (HG) exhibited downregulated CaM mRNA/protein and vascular endothelial growth factor (VEGF) expression with impeded eNOS phosphorylation and cell migration/tube formation. These perturbations were reduplicated in CALM1-knockdown cells but prevented in CALM1-overexpressing cells. EPCs from type 2 diabetes animals behaved similarly to HG-treated normal EPCs, which could be rescued by CALM1-gene transduction. Consistently, diabetic animals displayed impaired eNOS phosphorylation, endothelium-dependent dilation, and CaM expression in the aorta, as well as deficient physical interaction of CaM and eNOS in the gastrocnemius. Local CALM1 gene delivery into a diabetic mouse ischemic hindlimb improved the blunted limb blood perfusion and gastrocnemius angiogenesis, and foot injuries. Diabetic patients showed insufficient foot microvascular autoregulation, eNOS phosphorylation, and NO production with downregulated CaM expression in the arterial endothelium, and abnormal CALM1 transcription in genome-wide sequencing analysis. Therefore, our findings demonstrated that downregulated CaM expression is responsible for endothelium dysfunction and angiogenesis impairment in diabetes, and provided a novel mechanism and target to protect against diabetic endothelial injury.
Collapse
Affiliation(s)
- Tian-Tian Liu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Huan-Huan Xu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Ze-Juan Liu
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - He-Ping Zhang
- Beijing Friendship Hospital, The Affiliated Hospital of Capital Medical University, Beijing, 100065, China
| | - Hai-Tao Zhou
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, and Peaking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zhi-Xiang Zhu
- National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, and Peaking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100021, China
| | - Zhi-Qiang Wang
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Jing-Yi Xue
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Qiang Li
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Yi Ma
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Hong-Jie You
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China
| | - Da-Li Luo
- Department of Pharmacology, Beijing Key Laboratory of Cardiovascular Diseases Related to Metabolic Disturbance, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
25
|
Yoshida A, Ito A, Yasui N, Yamashita A. Direct binding of calmodulin to the cytosolic C-terminal regions of sweet/umami taste receptors. J Biochem 2023; 174:451-459. [PMID: 37527916 PMCID: PMC11033526 DOI: 10.1093/jb/mvad060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/15/2023] [Accepted: 07/26/2023] [Indexed: 08/03/2023] Open
Abstract
Sweet and umami taste receptors recognize chemicals such as sugars and amino acids on their extracellular side and transmit signals into the cytosol of the taste cell. In contrast to ligands that act on the extracellular side of these receptors, little is known regarding the molecules that regulate receptor functions within the cytosol. In this study, we analysed the interaction between sweet and umami taste receptors and calmodulin, a representative Ca2+-dependent cytosolic regulatory protein. High prediction scores for calmodulin binding were observed on the C-terminal cytosolic side of mouse taste receptor type 1 subunit 3 (T1r3), a subunit that is common to both sweet and umami taste receptors. Pull-down assay and surface plasmon resonance analyses showed different affinities of calmodulin to the C-terminal tails of distinct T1r subtypes. Furthermore, we found that T1r3 and T1r2 showed the highest and considerable binding to calmodulin, whereas T1r1 showed weaker binding affinity. Finally, the binding of calmodulin to T1rs was consistently higher in the presence of Ca2+ than in its absence. The results suggested a possibility of the Ca2+-dependent feedback regulation process of sweet and umami taste receptor signaling by calmodulin.
Collapse
Affiliation(s)
- Atsuki Yoshida
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ayumi Ito
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Norihisa Yasui
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Atsuko Yamashita
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
26
|
Pedretti M, Favretto F, Troilo F, Giovannoni M, Conter C, Mattei B, Dominici P, Travaglini-Allocatelli C, Di Matteo A, Astegno A. Role of myristoylation in modulating PCaP1 interaction with calmodulin. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2023; 203:108003. [PMID: 37717348 DOI: 10.1016/j.plaphy.2023.108003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 08/09/2023] [Accepted: 09/04/2023] [Indexed: 09/19/2023]
Abstract
Plasma membrane-associated Cation-binding Protein 1 (PCaP1) belongs to the plant-unique DREPP protein family with largely unknown biological functions but ascertained roles in plant development and calcium (Ca2+) signaling. PCaP1 is anchored to the plasma membrane via N-myristoylation and a polybasic cluster, and its N-terminal region can bind Ca2+/calmodulin (CaM). However, the molecular determinants of PCaP1-Ca2+-CaM interaction and the functional impact of myristoylation in the complex formation and Ca2+ sensitivity of CaM remained to be elucidated. Herein, we investigated the direct interaction between Arabidopsis PCaP1 (AtPCaP1) and CaM1 (AtCaM1) using both myristoylated and non-myristoylated peptides corresponding to the N-terminal region of AtPCaP1. ITC analysis showed that AtCaM1 forms a high affinity 1:1 complex with AtPCaP1 peptides and the interaction is strictly Ca2+-dependent. Spectroscopic and kinetic Ca2+ binding studies showed that the myristoylated peptide dramatically increased the Ca2+-binding affinity of AtCaM1 and slowed the Ca2+ dissociation rates from both the C- and N-lobes, thus suggesting that the myristoylation modulates the mechanism of AtPCaP1 recognition by AtCaM1. Furthermore, NMR and CD spectroscopy revealed that the structure of both the N- and C-lobes of Ca2+-AtCaM1 changes markedly in the presence of the myristoylated AtPCaP1 peptide, which assumes a helical structure in the final complex. Overall, our results indicate that AtPCaP1 biological function is strictly related to the presence of multiple ligands, i.e., the myristoyl moiety, Ca2+ ions and AtCaM1 and only a full characterization of their equilibria will allow for a complete molecular understanding of the putative role of PCaP1 as signal protein.
Collapse
Affiliation(s)
- Marco Pedretti
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | - Filippo Favretto
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | - Francesca Troilo
- CNR Institute of Molecular Biology and Pathology, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Moira Giovannoni
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Carolina Conter
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | - Benedetta Mattei
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100, L'Aquila, Italy
| | - Paola Dominici
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| | | | - Adele Di Matteo
- CNR Institute of Molecular Biology and Pathology, P.le Aldo Moro 5, 00185, Rome, Italy.
| | - Alessandra Astegno
- Department of Biotechnology, University of Verona, Strada Le Grazie 15, 37134, Verona, Italy
| |
Collapse
|
27
|
Denesyuk AI, Permyakov SE, Permyakov EA, Johnson MS, Denessiouk K, Uversky VN. Canonical structural-binding modes in the calmodulin-target protein complexes. J Biomol Struct Dyn 2023; 41:7582-7594. [PMID: 36106955 DOI: 10.1080/07391102.2022.2123391] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 09/04/2022] [Indexed: 10/14/2022]
Abstract
Intracellular calcium sensor protein calmodulin (CaM) belongs to the large EF-hand protein superfamily. CaM shows a unique and not fully understood ability to bind to multiple targets, allows them to participate in a variety of regulatory processes. The protein has two approximately symmetrical globular domains (the N- and C-lobes). Analysis of the CaM-binding sites of target proteins showed that they have two hydrophobic 'anchor' amino acids separated by 10 to 17 residues. Consequently, several CaM-binding motifs: {1-10}, {1-11}, {1-13}, {1-14}, {1-16}, {1-17}, differing by the distance between the two anchor residues along the amino acid sequence, have been identified. Despite extensive structural information on the role of target-protein amino acid residues in the formation of complexes with CaM, much less is known about the role of amino acids from CaM contributing to these interactions. In this work, a quantitative analysis of the contact surfaces of CaM and target proteins has been carried out for 35 representative three-dimensional structures. It has been shown that, in addition to the two hydrophobic terminal residues of the target fragment, the interaction also involves residues that are 4 residues earlier in the sequence (binding mode {1-5}). It has also been found that the N- and C-lobes of CaM bind the {1-5} motif located at the ends of the target in a structurally identical manner. Methionine residues at positions 51 (corresponding to 124 in the C-lobe), 71 (144), and 72 (145) of the CaM amino acid sequence are key hydrophobic residues for this interaction. They are located at the N- and C-boundaries of the even EF-hand motifs. The hydrophobic core of CaM ('Ф-quatrefoil') consists of 10 amino acids in the N-lobe (and in the C-lobe): Phe16 (Phe89), Phe19 (Phe92), Ile27 (Ile100), Thr29 (Ala102), Leu32 (Leu105), Ile52 (Ile125), Val55 (Ala128), Ile63 (Val136), Phe65 (Tyr138), and Phe68 (Phe141) and do not intersect with the target-binding methionine residues. CaM belongs to the 'dynamic' group of EF-hand proteins, in which calcium and protein ligand binding causes only global conformational changes but does not alter the conservative 'black' and 'grey' clusters described in our earlier works (PLoS One. 2014; 9(10):e109287). The membership of CaM in the 'dynamic' group is determined by the triggering and protective methionine layer: Met51 (Met124), Met71 (Met144) and Met72 (Met145). HIGHLIGHTSInterchain interactions in the unique 35 CaM complex structures were analyzed.Methionine amino acids of the N- and C-lobes of CaM form triggering and protective layers.Interactions of the target terminal residues with these methionine layers are structurally identical.CaM belonging to the 'dynamic' group is determined by the triggering and protective methionine layer.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alexander I Denesyuk
- Institute for Biological Instrumentation of the, Russian Academy of Sciences, Federal Research Center, "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino Moscow Region, Russia
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Sergei E Permyakov
- Institute for Biological Instrumentation of the, Russian Academy of Sciences, Federal Research Center, "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino Moscow Region, Russia
| | - Eugene A Permyakov
- Institute for Biological Instrumentation of the, Russian Academy of Sciences, Federal Research Center, "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino Moscow Region, Russia
| | - Mark S Johnson
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Konstantin Denessiouk
- Structural Bioinformatics Laboratory, Biochemistry, InFLAMES Research Flagship Center, Faculty of Science and Engineering, Åbo Akademi University, Turku, Finland
| | - Vladimir N Uversky
- Institute for Biological Instrumentation of the, Russian Academy of Sciences, Federal Research Center, "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino Moscow Region, Russia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
28
|
Brohus M, Busuioc AO, Wimmer R, Nyegaard M, Overgaard MT. Calmodulin mutations affecting Gly114 impair binding to the Na V1.5 IQ-domain. Front Pharmacol 2023; 14:1210140. [PMID: 37663247 PMCID: PMC10469309 DOI: 10.3389/fphar.2023.1210140] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 07/25/2023] [Indexed: 09/05/2023] Open
Abstract
Missense variants in CALM genes encoding the Ca2+-binding protein calmodulin (CaM) cause severe cardiac arrhythmias. The disease mechanisms have been attributed to dysregulation of RyR2, for Catecholaminergic Polymorphic Ventricular Tachycardia (CPVT) and/or CaV1.2, for Long-QT Syndrome (LQTS). Recently, a novel CALM2 variant, G114R, was identified in a mother and two of her four children, all of whom died suddenly while asleep at a young age. The G114R variant impairs closure of CaV1.2 and RyR2, consistent with a CPVT and/or mild LQTS phenotype. However, the children carrying the CALM2 G114R variant displayed a phenotype commonly observed with variants in NaV1.5, i.e., Brugada Syndrome (BrS) or LQT3, where death while asleep is a common feature. We therefore hypothesized that the G114R variant specifically would interfere with NaV1.5 binding. Here, we demonstrate that CaM binding to the NaV1.5 IQ-domain is severely impaired for two CaM variants G114R and G114W. The impact was most severe at low and intermediate Ca2+ concentrations (up to 4 µM) resulting in more than a 50-fold reduction in NaV1.5 binding affinity, and a smaller 1.5 to 11-fold reduction at high Ca2+ concentrations (25-400 µM). In contrast, the arrhythmogenic CaM-N98S variant only induced a 1.5-fold reduction in NaV1.5 binding and only at 4 µM Ca2+. A non-arrhythmogenic I10T variant in CaM did not impair NaV1.5 IQ binding. These data suggest that the interaction between NaV1.5 and CaM is decreased with certain CaM variants, which may alter the cardiac sodium current, INa. Overall, these results suggest that the phenotypic spectrum of calmodulinopathies may likely expand to include BrS- and/or LQT3-like traits.
Collapse
Affiliation(s)
- Malene Brohus
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Ana-Octavia Busuioc
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Reinhard Wimmer
- Department of Chemistry and Bioscience, Aalborg University, Aalborg, Denmark
| | - Mette Nyegaard
- Department of Health Science and Technology, Aalborg University, Gistrup, Denmark
| | | |
Collapse
|
29
|
O’Day DH. Alzheimer's Disease beyond Calcium Dysregulation: The Complex Interplay between Calmodulin, Calmodulin-Binding Proteins and Amyloid Beta from Disease Onset through Progression. Curr Issues Mol Biol 2023; 45:6246-6261. [PMID: 37623212 PMCID: PMC10453589 DOI: 10.3390/cimb45080393] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/12/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
A multifactorial syndrome, Alzheimer's disease is the main cause of dementia, but there is no existing therapy to prevent it or stop its progression. One of the earliest events of Alzheimer's disease is the disruption of calcium homeostasis but that is just a prelude to the disease's devastating impact. Calcium does not work alone but must interact with downstream cellular components of which the small regulatory protein calmodulin is central, if not primary. This review supports the idea that, due to calcium dyshomeostasis, calmodulin is a dominant regulatory protein that functions in all stages of Alzheimer's disease, and these regulatory events are impacted by amyloid beta. Amyloid beta not only binds to and regulates calmodulin but also multiple calmodulin-binding proteins involved in Alzheimer's. Together, they act on the regulation of calcium dyshomeostasis, neuroinflammation, amyloidogenesis, memory formation, neuronal plasticity and more. The complex interactions between calmodulin, its binding proteins and amyloid beta may explain why many therapies have failed or are doomed to failure unless they are considered.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
30
|
Manoharan GB, Laurini C, Bottone S, Ben Fredj N, Abankwa DK. K-Ras Binds Calmodulin-Related Centrin1 with Potential Implications for K-Ras Driven Cancer Cell Stemness. Cancers (Basel) 2023; 15:3087. [PMID: 37370697 DOI: 10.3390/cancers15123087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/30/2023] [Accepted: 06/05/2023] [Indexed: 06/29/2023] Open
Abstract
Recent data suggest that K-Ras4B (hereafter K-Ras) can drive cancer cell stemness via calmodulin (CaM)-dependent, non-canonical Wnt-signalling. Here we examined whether another Ca2+-binding protein, the CaM-related centrin1, binds to K-Ras and could mediate some K-Ras functions that were previously ascribed to CaM. While CaM and centrin1 appear to distinguish between peptides that were derived from their classical targets, they both bind to K-Ras in cells. Cellular BRET- and immunoprecipitation data suggest that CaM engages more with K-Ras than centrin1 and that the interaction with the C-terminal membrane anchor of K-Ras is sufficient for this. Surprisingly, binding of neither K-Ras nor its membrane anchor alone to CaM or centrin1 is sensitive to inhibition of prenylation. In support of an involvement of the G-domain of K-Ras in cellular complexes with these Ca2+-binding proteins, we find that oncogenic K-RasG12V displays increased engagement with both CaM and centrin1. This is abrogated by addition of the D38A effector-site mutation, suggesting that K-RasG12V is held together with CaM or centrin1 in complexes with effectors. When treated with CaM inhibitors, the BRET-interaction of K-RasG12V with centrin1 was also disrupted in the low micromolar range, comparable to that with CaM. While CaM predominates in regulating functional membrane anchorage of K-Ras, it has a very similar co-distribution with centrin1 on mitotic organelles. Given these results, a significant overlap of the CaM- and centrin1-dependent functions of K-Ras is suggested.
Collapse
Affiliation(s)
- Ganesh Babu Manoharan
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Christina Laurini
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Sara Bottone
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Nesrine Ben Fredj
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Daniel Kwaku Abankwa
- Cancer Cell Biology and Drug Discovery Group, Department of Life Sciences and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
31
|
Kim C, Kim Y, Lee SJ, Yun SR, Choi J, Kim SO, Yang Y, Ihee H. Visualizing Heterogeneous Protein Conformations with Multi-Tilt Nanoparticle-Aided Cryo-Electron Microscopy Sampling. NANO LETTERS 2023; 23:3334-3343. [PMID: 37068052 PMCID: PMC10141564 DOI: 10.1021/acs.nanolett.3c00313] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Obtaining the heterogeneous conformation of small proteins is important for understanding their biological role, but it is still challenging. Here, we developed a multi-tilt nanoparticle-aided cryo-electron microscopy sampling (MT-NACS) technique that enables the observation of heterogeneous conformations of small proteins and applied it to calmodulin. By imaging the proteins labeled by two gold nanoparticles at multiple tilt angles and analyzing the projected positions of the nanoparticles, the distributions of 3D interparticle distances were obtained. From the measured distance distributions, the conformational changes associated with Ca2+ binding and salt concentration were determined. MT-NACS was also used to track the structural change accompanied by the interaction between amyloid-beta and calmodulin, which has never been observed experimentally. This work offers an alternative platform for studying the functional flexibility of small proteins.
Collapse
Affiliation(s)
- Changin Kim
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Yeeun Kim
- Department
of Physics, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
| | - Sang Jin Lee
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - So Ri Yun
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Jungkweon Choi
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Seong Ok Kim
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
| | - Yongsoo Yang
- Department
of Physics, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Y.Y.:
email, ; tel, +82-42-350-7303
| | - Hyotcherl Ihee
- Department
of Chemistry, Korea Advanced Institute of
Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KI
for the BioCentury, Korea Advanced Institute
of Science and Technology (KAIST), Daejeon 34141, Republic
of Korea
- Center
for Advanced Reaction Dynamics, Institute
for Basic Science (IBS), Daejeon 34141, Republic of Korea
- H.I.: email, ; tel, +82-42-350-2844
| |
Collapse
|
32
|
O'Day DH. Phytochemical Interactions with Calmodulin and Critical Calmodulin Binding Proteins Involved in Amyloidogenesis in Alzheimer's Disease. Biomolecules 2023; 13:biom13040678. [PMID: 37189425 DOI: 10.3390/biom13040678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/03/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
An increasing number of plant-based herbal treatments, dietary supplements, medical foods and nutraceuticals and their component phytochemicals are used as alternative treatments to prevent or slow the onset and progression of Alzheimer's disease. Their appeal stems from the fact that no current pharmaceutical or medical treatment can accomplish this. While a handful of pharmaceuticals are approved to treat Alzheimer's, none has been shown to prevent, significantly slow or stop the disease. As a result, many see the appeal of alternative plant-based treatments as an option. Here, we show that many phytochemicals proposed or used as Alzheimer's treatments share a common theme: they work via a calmodulin-mediated mode of action. Some phytochemicals bind to and inhibit calmodulin directly while others bind to and regulate calmodulin-binding proteins, including Aβ monomers and BACE1. Phytochemical binding to Aβ monomers can prevent the formation of Aβ oligomers. A limited number of phytochemicals are also known to stimulate calmodulin gene expression. The significance of these interactions to amyloidogenesis in Alzheimer's disease is reviewed.
Collapse
Affiliation(s)
- Danton H O'Day
- Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada
| |
Collapse
|
33
|
Lai P, Yang X, Li YH, Yin YL, Yao Q, Huang S, Fan YY, Song JK, Zhao GH. Characterization of CpCaM, a protein potentially involved in the growth of Cryptosporidium parvum. Parasitol Res 2023; 122:989-996. [PMID: 36879147 DOI: 10.1007/s00436-023-07803-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/24/2023] [Indexed: 03/08/2023]
Abstract
Cryptosporidium parvum is an important apicomplexan parasite causing severe diarrhea in both humans and animals. Calmodulin (CaM), a multifunctional and universal calcium-binding protein, contributes to the growth and development of apicomplexan parasites, but the role of CaM in C. parvum remains unknown. In this study, the CaM of C. parvum encoded by the cgd2_810 gene was expressed in Escherichia coli, and the biological functions of CpCaM were preliminarily investigated. The transcriptional level of the cgd2_810 gene peaked at 36 h post infection (pi), and the CpCaM protein was mainly located around the nucleus of the whole oocysts, in the middle of sporozoites and around the nucleus of merozoites. Anti-CpCaM antibody reduced the invasion of C. parvum sporozoites by 30.69%. The present study indicates that CpCaM is potentially involved in the growth of C. parvum. Results of the study expand our knowledge on the interaction between host and Cryptosporidium.
Collapse
Affiliation(s)
- Peng Lai
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Xin Yang
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yun-Hui Li
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Yan-Ling Yin
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Qian Yao
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Shuang Huang
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Ying-Ying Fan
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Jun-Ke Song
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| | - Guang-Hui Zhao
- Key Laboratory of Ruminant Disease Prevention and Control (West), College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
34
|
Pollard LW, Coscia SM, Rebowski G, Palmer NJ, Holzbaur ELF, Dominguez R, Ostap EM. Ensembles of human myosin-19 bound to calmodulin and regulatory light chain RLC12B drive multimicron transport. J Biol Chem 2023; 299:102906. [PMID: 36642185 PMCID: PMC9929473 DOI: 10.1016/j.jbc.2023.102906] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/15/2023] Open
Abstract
Myosin-19 (Myo19) controls the size, morphology, and distribution of mitochondria, but the underlying role of Myo19 motor activity is unknown. Complicating mechanistic in vitro studies, the identity of the light chains (LCs) of Myo19 remains unsettled. Here, we show by coimmunoprecipitation, reconstitution, and proteomics that the three IQ motifs of human Myo19 expressed in Expi293 human cells bind regulatory light chain (RLC12B) and calmodulin (CaM). We demonstrate that overexpression of Myo19 in HeLa cells enhances the recruitment of both Myo19 and RLC12B to mitochondria, suggesting cellular association of RLC12B with the motor. Further experiments revealed that RLC12B binds IQ2 and is flanked by two CaM molecules. In vitro, we observed that the maximal speed (∼350 nm/s) occurs when Myo19 is supplemented with CaM, but not RLC12B, suggesting maximal motility requires binding of CaM to IQ-1 and IQ-3. The addition of calcium slowed actin gliding (∼200 nm/s) without an apparent effect on CaM affinity. Furthermore, we show that small ensembles of Myo19 motors attached to quantum dots can undergo processive runs over several microns, and that calcium reduces the attachment frequency and run length of Myo19. Together, our data are consistent with a model where a few single-headed Myo19 molecules attached to a mitochondrion can sustain prolonged motile associations with actin in a CaM- and calcium-dependent manner. Based on these properties, we propose that Myo19 can function in mitochondria transport along actin filaments, tension generation on multiple randomly oriented filaments, and/or pushing against branched actin networks assembled near the membrane surface.
Collapse
Affiliation(s)
- Luther W Pollard
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Stephen M Coscia
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Cell and Molecular Biology Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Grzegorz Rebowski
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nicholas J Palmer
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Biochemistry and Molecular Biophysics Graduate Group, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Erika L F Holzbaur
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Roberto Dominguez
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| | - E Michael Ostap
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA; Pennsylvania Muscle Institute, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
35
|
Wang H, Feng M, Zhong X, Yu Q, Que Y, Xu L, Guo J. Identification of Saccharum CaM gene family and function characterization of ScCaM1 during cold and oxidant exposure in Pichia pastoris. Genes Genomics 2023; 45:103-122. [PMID: 35608775 DOI: 10.1007/s13258-022-01263-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 05/03/2022] [Indexed: 01/11/2023]
Abstract
BACKGROUND Calmodulin (CaM) plays an essential role in binding calcium ions and mediating the interpretation of Ca2+ signals in plants under various stresses. However, the evolutionary relationship of CaM family proteins in Saccharum has not been elucidated. OBJECTIVE To deduce and explore the evolution and function of Saccharum CaM family. METHODS A total of 104 typical CaMs were obtained from Saccharum spontaneum and other 18 plant species. The molecular characteristics and evolution of those CaM proteins were analyzed. A typical CaM gene, ScCaM1, was subsequently cloned from sugarcane (Saccharum spp. hybrid). Its expression patterns in different tissues and under various abiotic stresses were assessed by quantitative real-time PCR. Then the green fluorescent protein was used to determine the subcellular localization of ScCaM1. Finally, the function of ScCaM1 was evaluated via heterologous yeast expression systems. RESULTS Three typical CaM members (SsCaM1, SsCaM2, and SsCaM3) were identified from the S. spontaneum genome database. CaMs were originated from the two last common ancestors before the origin of angiosperms. The number of CaM family members did not correlate to the genome size but correlated with allopolyploidization events. The ScCaM1 was more highly expressed in buds and roots than in other tissues. The expression patterns of ScCaM1 suggested that it was involved in responses to various abiotic stresses in sugarcane via different hormonal signaling pathways. Noteworthily, its expression levels appeared relatively stable during the cold exposure in the cold-tolerant variety but significantly suppressed in the cold-susceptible variety. Moreover, the recombinant yeast (Pichia pastoris) overexpressing ScCaM1 grew better than the wild-type yeast strain under cold and oxidative stresses. It was revealed that the ScCaM1 played a positive role in reactive oxygen species scavenging and conferred enhanced cold and oxidative stress tolerance to cells. CONCLUSION This study provided comprehensive information on the CaM gene family in Saccharum and would facilitate further investigation of their functional characterization.
Collapse
Affiliation(s)
- Hengbo Wang
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Meichang Feng
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Xiaoqiang Zhong
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Qing Yu
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Youxiong Que
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Liping Xu
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
| | - Jinlong Guo
- Key Laboratory of Sugarcane Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, College of Agriculture, Fujian Agriculture and Forestry University, Fuzhou, 350002, China.
| |
Collapse
|
36
|
Young BD, Cook ME, Costabile BK, Samanta R, Zhuang X, Sevdalis SE, Varney KM, Mancia F, Matysiak S, Lattman E, Weber DJ. Binding and Functional Folding (BFF): A Physiological Framework for Studying Biomolecular Interactions and Allostery. J Mol Biol 2022; 434:167872. [PMID: 36354074 PMCID: PMC10871162 DOI: 10.1016/j.jmb.2022.167872] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 09/20/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
EF-hand Ca2+-binding proteins (CBPs), such as S100 proteins (S100s) and calmodulin (CaM), are signaling proteins that undergo conformational changes upon increasing intracellular Ca2+. Upon binding Ca2+, S100 proteins and CaM interact with protein targets and induce important biological responses. The Ca2+-binding affinity of CaM and most S100s in the absence of target is weak (CaKD > 1 μM). However, upon effector protein binding, the Ca2+ affinity of these proteins increases via heterotropic allostery (CaKD < 1 μM). Because of the high number and micromolar concentrations of EF-hand CBPs in a cell, at any given time, allostery is required physiologically, allowing for (i) proper Ca2+ homeostasis and (ii) strict maintenance of Ca2+-signaling within a narrow dynamic range of free Ca2+ ion concentrations, [Ca2+]free. In this review, mechanisms of allostery are coalesced into an empirical "binding and functional folding (BFF)" physiological framework. At the molecular level, folding (F), binding and folding (BF), and BFF events include all atoms in the biomolecular complex under study. The BFF framework is introduced with two straightforward BFF types for proteins (type 1, concerted; type 2, stepwise) and considers how homologous and nonhomologous amino acid residues of CBPs and their effector protein(s) evolved to provide allosteric tightening of Ca2+ and simultaneously determine how specific and relatively promiscuous CBP-target complexes form as both are needed for proper cellular function.
Collapse
Affiliation(s)
- Brianna D Young
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Mary E Cook
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Brianna K Costabile
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Riya Samanta
- Biophysics Graduate Program, University of Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Xinhao Zhuang
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Spiridon E Sevdalis
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Kristen M Varney
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University, New York, NY 10032, USA
| | - Silvina Matysiak
- Biophysics Graduate Program, University of Maryland, College Park, MD 20742, USA; Fischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
| | - Eaton Lattman
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; Department of Physics, Arizona State University, Tempe, AZ 85287, USA
| | - David J Weber
- The Center for Biomolecular Therapeutics (CBT), Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; The Institute of Bioscience and Biotechnology Research (IBBR), Rockville, MD 20850, USA.
| |
Collapse
|
37
|
Mantilla G, Peréz-Gordones MC, Cisneros-Montufar S, Benaim G, Navarro JC, Mendoza M, Ramírez-Iglesias JR. Structural Analysis and Diversity of Calmodulin-Binding Domains in Membrane and Intracellular Ca2+-ATPases. J Membr Biol 2022; 256:159-174. [PMID: 36454258 DOI: 10.1007/s00232-022-00275-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 11/11/2022] [Indexed: 12/03/2022]
Abstract
The plasma membrane and autoinhibited Ca2+-ATPases contribute to the Ca2+ homeostasis in a wide variety of organisms. The enzymatic activity of these pumps is stimulated by calmodulin, which interacts with the target protein through the calmodulin-binding domain (CaMBD). Most information about this region is related to all calmodulin modulated proteins, which indicates general chemical properties and there is no established relation between Ca2+ pump sequences and taxonomic classification. Thus, the aim of this study was to perform an in silico analysis of the CaMBD from several Ca2+-ATPases, in order to determine their diversity and to detect specific patterns and amino acid selection in different species. Patterns related to potential and confirmed CaMBD were detected using sequences retrieved from the literature. The occurrence of these patterns was determined across 120 sequences from 17 taxonomical classes, which were analyzed by a phylogenetic tree to establish phylogenetic groups. Predicted physicochemical characteristics including hydropathy and net charge were calculated for each group of sequences. 22 Ca2+-ATPases sequences from animals, unicellular eukaryotes, and plants were retrieved from bioinformatic databases. These sequences allow us to establish the Patterns 1(GQILWVRGLTRLQTQ), 3(KNPSLEALQRW), and 4(SRWRRLQAEHVKK), which are present at the beginning of putative CaMBD of metazoan, parasites, and land plants. A pattern 2 (IRVVNAFR) was consistently found at the end of most analyzed sequences. The amino acid preference in the CaMBDs changed depending on the phylogenetic groups, with predominance of several aliphatic and charged residues, to confer amphiphilic properties. The results here displayed show a conserved mechanism to contribute to the Ca2+ homeostasis across evolution and may help to detect putative CaMBDs.
Collapse
Affiliation(s)
- Génesis Mantilla
- Research Group of Emerging and Neglected Diseases, Ecoepidemiology and Biodiversity. Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador
- Faculty of Engineering and Applied Sciences, Universidad Internacional SEK (UISEK), Quito, Ecuador
| | - María C Peréz-Gordones
- Instituto de Biología Experimental (IBE), Universidad Central de Venezuela (UCV), Caracas, Venezuela
| | - Soledad Cisneros-Montufar
- Research Group of Emerging and Neglected Diseases, Ecoepidemiology and Biodiversity. Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador
- Faculty of Engineering and Applied Sciences, Universidad Internacional SEK (UISEK), Quito, Ecuador
| | - Gustavo Benaim
- Instituto de Biología Experimental (IBE), Universidad Central de Venezuela (UCV), Caracas, Venezuela
- Instituto de Estudios Avanzados (IDEA), Caracas, Venezuela
| | - Juan-Carlos Navarro
- Research Group of Emerging and Neglected Diseases, Ecoepidemiology and Biodiversity. Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador
- Program of Master in Biomedicine, Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador
| | - Marta Mendoza
- Centro de Estudios Biomédicos y Veterinarios, Instituto de Estudios Científicos y Tecnológicos (IDECYT), Universidad Nacional Experimental Simón Rodríguez, Caracas, Venezuela
| | - José R Ramírez-Iglesias
- Research Group of Emerging and Neglected Diseases, Ecoepidemiology and Biodiversity. Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador.
- Program of Master in Biomedicine, Health Sciences Faculty, Universidad Internacional SEK (UISEK), Quito, Ecuador.
| |
Collapse
|
38
|
Erkoc P, von Reumont BM, Lüddecke T, Henke M, Ulshöfer T, Vilcinskas A, Fürst R, Schiffmann S. The Pharmacological Potential of Novel Melittin Variants from the Honeybee and Solitary Bees against Inflammation and Cancer. Toxins (Basel) 2022; 14:toxins14120818. [PMID: 36548715 PMCID: PMC9786170 DOI: 10.3390/toxins14120818] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/24/2022] Open
Abstract
The venom of honeybees is composed of numerous peptides and proteins and has been used for decades as an anti-inflammatory and anti-cancer agent in traditional medicine. However, the bioactivity of specific biomolecular components has been evaluated for the predominant constituent, melittin. So far, only a few melittin-like peptides from solitary bee species have been investigated, and the molecular mechanisms of bee venoms as therapeutic agents remain largely unknown. Here, the preclinical pharmacological activities of known and proteo-transcriptomically discovered new melittin variants from the honeybee and more ancestral variants from phylogenetically older solitary bees were explored in the context of cancer and inflammation. We studied the effects of melittin peptides on cytotoxicity, second messenger release, and inflammatory markers using primary human cells, non-cancer, and cancerous cell lines. Melittin and some of its variants showed cytotoxic effects, induced Ca2+ signaling and inhibited cAMP production, and prevented LPS-induced NO synthesis but did not affect the IP3 signaling and pro-inflammatory activation of endothelial cells. Compared to the originally-described melittin, some phylogenetically more ancestral variants from solitary bees offer potential therapeutic modalities in modulating the in vitro inflammatory processes, and hindering cancer cell viability/proliferation, including aggressive breast cancers, and are worth further investigation.
Collapse
Affiliation(s)
- Pelin Erkoc
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, 60438 Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Correspondence: (P.E.); (B.M.v.R.)
| | - Björn Marcus von Reumont
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Applied Bioinformatics Group, Faculty of Biological Sciences, Goethe University Frankfurt, Max-von-Laue-Str. 13, 60438 Frankfurt, Germany
- Correspondence: (P.E.); (B.M.v.R.)
| | - Tim Lüddecke
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME-BR), 235394 Giessen, Germany
| | - Marina Henke
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), 60596 Frankfurt, Germany
| | - Thomas Ulshöfer
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), 60596 Frankfurt, Germany
| | - Andreas Vilcinskas
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Department of Bioresources, Fraunhofer Institute for Molecular Biology and Applied Ecology (IME-BR), 235394 Giessen, Germany
| | - Robert Fürst
- Institute of Pharmaceutical Biology, Faculty of Biochemistry, Chemistry and Pharmacy, Goethe University Frankfurt, 60438 Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
| | - Susanne Schiffmann
- LOEWE Center for Translational Biodiversity Genomics (LOEWE-TBG), Senckenberganlage 25, 60325 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology (ITMP), 60596 Frankfurt, Germany
| |
Collapse
|
39
|
O’Day DH. Calmodulin Binding Domains in Critical Risk Proteins Involved in Neurodegeneration. Curr Issues Mol Biol 2022; 44:5802-5814. [PMID: 36421678 PMCID: PMC9689381 DOI: 10.3390/cimb44110394] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/15/2022] [Accepted: 11/18/2022] [Indexed: 08/26/2023] Open
Abstract
Neurodegeneration leads to multiple early changes in cognitive, emotional, and social behaviours and ultimately progresses to dementia. The dysregulation of calcium is one of the earliest potentially initiating events in the development of neurodegenerative diseases. A primary neuronal target of calcium is the small sensor and effector protein calmodulin that, in response to calcium levels, binds to and regulates hundreds of calmodulin binding proteins. The intimate and entangled relationship between calmodulin binding proteins and all phases of Alzheimer's disease has been established, but the relationship to other neurodegenerative diseases is just beginning to be evaluated. Risk factors and hallmark proteins from Parkinson's disease (PD; SNCA, Parkin, PINK1, LRRK2, PARK7), Huntington's disease (HD; Htt, TGM1, TGM2), Lewy Body disease (LBD; TMEM175, GBA), and amyotrophic lateral sclerosis/frontotemporal disease (ALS/FTD; VCP, FUS, TDP-43, TBK1, C90rf72, SQSTM1, CHCHD10, SOD1) were scanned for the presence of calmodulin binding domains and, within them, appropriate binding motifs. Binding domains and motifs were identified in multiple risk proteins, some of which are involved in multiple neurodegenerative diseases. The potential calmodulin binding profiles for risk proteins involved in HD, PD, LBD, and ALS/FTD coupled with other studies on proven binding proteins supports the central and potentially critical role for calmodulin in neurodegenerative events.
Collapse
Affiliation(s)
- Danton H. O’Day
- Department of Biology, University of Toronto Mississauga, Mississauga, ON L5L 1C6, Canada;
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON M5S 3G5, Canada
| |
Collapse
|
40
|
Christoffer C, Kihara D. Domain-Based Protein Docking with Extremely Large Conformational Changes. J Mol Biol 2022; 434:167820. [PMID: 36089054 PMCID: PMC9992458 DOI: 10.1016/j.jmb.2022.167820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/31/2022] [Accepted: 09/03/2022] [Indexed: 11/17/2022]
Abstract
Proteins are key components in many processes in living cells, and physical interactions with other proteins and nucleic acids often form key parts of their functions. In many cases, large flexibility of proteins as they interact is key to their function. To understand the mechanisms of these processes, it is necessary to consider the 3D structures of such protein complexes. When such structures are not yet experimentally determined, protein docking has long been present to computationally generate useful structure models. However, protein docking has long had the limitation that the consideration of flexibility is usually limited to very small movements or very small structures. Methods have been developed which handle minor flexibility via normal mode or other structure sampling, but new methods are required to model ordered proteins which undergo large-scale conformational changes to elucidate their function at the molecular level. Here, we present Flex-LZerD, a framework for docking such complexes. Via partial assembly multidomain docking and an iterative normal mode analysis admitting curvilinear motions, we demonstrate the ability to model the assembly of a variety of protein-protein and protein-nucleic acid complexes.
Collapse
Affiliation(s)
- Charles Christoffer
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA
| | - Daisuke Kihara
- Department of Computer Science, Purdue University, West Lafayette, IN 47907, USA; Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue University Center for Cancer Research, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
41
|
Munk M, Villalobo E, Villalobo A, Berchtold MW. Differential expression of the three independent CaM genes coding for an identical protein: Potential relevance of distinct mRNA stability by different codon usage. Cell Calcium 2022; 107:102656. [DOI: 10.1016/j.ceca.2022.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 09/01/2022] [Accepted: 09/25/2022] [Indexed: 11/24/2022]
|
42
|
Chang W, Luo Q, Wu X, Nan Y, Zhao P, Zhang L, Luo A, Jiao W, Zhu Q, Fu Y, Liu Z. OTUB2 exerts tumor-suppressive roles via STAT1-mediated CALML3 activation and increased phosphatidylserine synthesis. Cell Rep 2022; 41:111561. [DOI: 10.1016/j.celrep.2022.111561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/17/2022] [Accepted: 10/04/2022] [Indexed: 12/09/2022] Open
|
43
|
A Modeling and Analysis Study Reveals That CaMKII in Synaptic Plasticity Is a Dominant Affecter in CaM Systems in a T286 Phosphorylation-Dependent Manner. Molecules 2022; 27:molecules27185974. [PMID: 36144710 PMCID: PMC9501549 DOI: 10.3390/molecules27185974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022] Open
Abstract
NMDAR-dependent synaptic plasticity in the hippocampus consists of two opposing forces: long-term potentiation (LTP), which strengthens synapses and long-term depression (LTD), which weakens synapses. LTP and LTD are associated with memory formation and loss, respectively. Synaptic plasticity is controlled at a molecular level by Ca2+-mediated protein signaling. Here, Ca2+ binds the protein, calmodulin (CaM), which modulates synaptic plasticity in both directions. This is because Ca2+-bound CaM activates both LTD-and LTP-inducing proteins. Understanding how CaM responds to Ca2+ signaling and how this translates into synaptic plasticity is therefore important to understanding synaptic plasticity induction. In this paper, CaM activation by Ca2+ and calmodulin binding to downstream proteins was mathematically modeled using differential equations. Simulations were monitored with and without theoretical knockouts and, global sensitivity analyses were performed to determine how Ca2+/CaM signaling occurred at various Ca2+ signals when CaM levels were limiting. At elevated stimulations, the total CaM pool rapidly bound to its protein binding targets which regulate both LTP and LTD. This was followed by CaM becoming redistributed from low-affinity to high-affinity binding targets. Specifically, CaM was redistributed away from LTD-inducing proteins to bind the high-affinity LTP-inducing protein, calmodulin-dependent kinase II (CaMKII). In this way, CaMKII acted as a dominant affecter and repressed activation of opposing CaM-binding protein targets. The model thereby showed a novel form of CaM signaling by which the two opposing pathways crosstalk indirectly. The model also found that CaMKII can repress cAMP production by repressing CaM-regulated proteins, which catalyze cAMP production. The model also found that at low Ca2+ stimulation levels, typical of LTD induction, CaM signaling was unstable and is therefore unlikely to alone be enough to induce synaptic depression. Overall, this paper demonstrates how limiting levels of CaM may be a fundamental aspect of Ca2+ regulated signaling which allows crosstalk among proteins without requiring directly interaction.
Collapse
|
44
|
Sander S, Müller I, Alai MG, Nicke A, Tidow H. New insights into P2X7 receptor regulation: Ca 2+-calmodulin and GDP bind to the soluble P2X7 ballast domain. J Biol Chem 2022; 298:102495. [PMID: 36115462 PMCID: PMC9574498 DOI: 10.1016/j.jbc.2022.102495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 09/09/2022] [Accepted: 09/10/2022] [Indexed: 11/25/2022] Open
Abstract
P2X7 receptors are nonselective cation channels that are activated by extracellular ATP and play important roles in inflammation. They differ from other P2X family members by a large intracellular C-terminus that mediates diverse signaling processes that are little understood. A recent cryo-EM study revealed that the C-terminus of the P2X7 receptor forms a unique cytoplasmic ballast domain that possesses a GDP-binding site as well as a dinuclear Zn2+ site. However, the molecular basis for the regulatory function of the ballast domain as well as the interplay between the various ligands remain unclear. Here, we successfully expressed a soluble trimeric P2X7 ballast domain (P2X7BD) and characterized its ligand binding properties using a biophysical approach. We identified calmodulin (CaM)-binding regions within the ballast domain and found that binding of Ca2+-CaM and GDP to P2X7BD have opposite effects on its stability. Small-angle X-ray scattering experiments indicate that Ca2+-CaM binding disrupts the trimeric state of P2X7BD. Our results provide a possible framework for the intracellular regulation of the P2X7 receptor.
Collapse
Affiliation(s)
- Simon Sander
- The Hamburg Advanced Research Centre for Bioorganic Chemistry (HARBOR) & Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany
| | - Isabel Müller
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Maria Garcia Alai
- European Molecular Biology Laboratory Hamburg, Notkestrasse 85, D-22607 Hamburg, Germany
| | - Annette Nicke
- Walther Straub Institute of Pharmacology and Toxicology, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Henning Tidow
- The Hamburg Advanced Research Centre for Bioorganic Chemistry (HARBOR) & Department of Chemistry, Institute for Biochemistry and Molecular Biology, University of Hamburg, Luruper Chaussee 149, 22761 Hamburg, Germany.
| |
Collapse
|
45
|
Karschin N, Becker S, Griesinger C. Interdomain Dynamics via Paramagnetic NMR on the Highly Flexible Complex Calmodulin/Munc13-1. J Am Chem Soc 2022; 144:17041-17053. [PMID: 36082939 PMCID: PMC9501808 DOI: 10.1021/jacs.2c06611] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Paramagnetic NMR constraints are very useful to study protein interdomain motion, but their interpretation is not always straightforward. On the example of the particularly flexible complex Calmodulin/Munc13-1, we present a new approach to characterize this motion with pseudocontact shifts and residual dipolar couplings. Using molecular mechanics, we sampled the conformational space of the complex and used a genetic algorithm to find ensembles that are in agreement with the data. We used the Bayesian information criterion to determine the ideal ensemble size. This way, we were able to make an accurate, unambiguous, reproducible model of the interdomain motion of Calmodulin/Munc13-1 without prior knowledge about the domain orientation from crystallography.
Collapse
Affiliation(s)
- Niels Karschin
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen, Niedersachsen D-37077, Germany
| | - Stefan Becker
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen, Niedersachsen D-37077, Germany
| | - Christian Griesinger
- Max Planck Institute for Multidisciplinary Sciences, Am Fassberg 11, Göttingen, Niedersachsen D-37077, Germany.,Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen D-37075, Germany
| |
Collapse
|
46
|
Sun B, Kekenes-Huskey PM. Calmodulin's Interdomain Linker Is Optimized for Dynamics Signal Transmission and Calcium Binding. J Chem Inf Model 2022; 62:4210-4221. [PMID: 35994621 DOI: 10.1021/acs.jcim.2c00587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Linkers are ubiquitous in multidomain proteins. These linkers are integral to protein functions, and accumulating evidence suggests that the linkers' versatile roles are encoded in their sequences. However, a molecular picture of how amino acid differences in the linker influence protein function is still lacking. By using extensive Gaussian-accelerated MD coupled with dynamic network analysis, we reveal the molecular bases underlying the linker's role in Calmodulin (CaM), a highly conserved Ca2+-signaling hub in eukaryotes. Three CaM constructs comprising a wild-type linker, a flexible linker (four glycines at position D78-S81), and a rigid linker (four prolines at position D78-S81) were simulated. We show that the flexible linker resembles the wild type in allowing CaM to sample a large ensemble of conformations while the rigid linker confines the sampling. Our simulations recapture experimental observations that target binding enhances the Ca2+ affinity to CaM's EF-hand sites at the N-domain. However, only the wild-type linker can both correctly capture the Ca2+ binding order and maintain the α-helical structure of the domain. The other two constructs either bind Ca2+ in an incorrect order or exhibit unfolding of an N-domain helix. We demonstrate that the wild-type linker achieves these outcomes by transmitting interdomain dynamics efficiently. This was evidenced by stronger (anti)correlations among the linker residues, decoupling of the hydrogen bonds between A1-A15 and V35-E45, and structuring of the N-domain for Ca2+ binding. This decoupling was not evident for the other two constructs. Lastly, we show that the wild-type linker's optimal transmission stems from its thermodynamically favorable strain and solvation relative to the other two constructs. Our results show how the linker sequence tunes CaM function, suggesting possible mechanisms for changes in linker properties such as mutations or post-translational modifications to modulate protein/substrate binding.
Collapse
Affiliation(s)
- Bin Sun
- Department of Pharmacology, Harbin Medical University, Harbin 150081, China
| | - Peter M Kekenes-Huskey
- Department of Cell and Molecular Physiology, Loyola University, Chicago, Illinois 60153, United States
| |
Collapse
|
47
|
Stewart AM, Shanmugam M, Kutta RJ, Scrutton NS, Lovett JE, Hay S. Combined Pulsed Electron Double Resonance EPR and Molecular Dynamics Investigations of Calmodulin Suggest Effects of Crowding Agents on Protein Structures. Biochemistry 2022; 61:1735-1742. [PMID: 35979922 PMCID: PMC9454100 DOI: 10.1021/acs.biochem.2c00099] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Calmodulin (CaM) is a highly dynamic Ca2+-binding
protein
that exhibits large conformational changes upon binding Ca2+ and target proteins. Although it is accepted that CaM exists in
an equilibrium of conformational states in the absence of target protein,
the physiological relevance of an elongated helical linker region
in the Ca2+-replete form has been highly debated. In this
study, we use PELDOR (pulsed electron–electron double resonance)
EPR measurements of a doubly spin-labeled CaM variant to assess the
conformational states of CaM in the apo-, Ca2+-bound, and
Ca2+ plus target peptide-bound states. Our findings are
consistent with a three-state conformational model of CaM, showing
a semi-open apo-state, a highly extended Ca2+-replete state,
and a compact target protein-bound state. Molecular dynamics simulations
suggest that the presence of glycerol, and potentially other molecular
crowding agents, has a profound effect on the relative stability of
the different conformational states. Differing experimental conditions
may explain the discrepancies in the literature regarding the observed
conformational state(s) of CaM, and our PELDOR measurements show good
evidence for an extended conformation of Ca2+-replete CaM
similar to the one observed in early X-ray crystal structures.
Collapse
Affiliation(s)
- Andrew M Stewart
- The Roy J. Carver Department of Biochemistry, Biophysics and Molecular Biology, Iowa State University, Ames 50011, Iowa, United States.,Manchester Institute of Biotechnology and Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Muralidharan Shanmugam
- Manchester Institute of Biotechnology and Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Roger J Kutta
- Manchester Institute of Biotechnology and Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K.,Institute of Physical and Theoretical Chemistry, University of Regensburg, Regensburg 93040, Germany
| | - Nigel S Scrutton
- Manchester Institute of Biotechnology and Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| | - Janet E Lovett
- SUPA School of Physics and Astronomy and BSRC, The University of St Andrews, St Andrews KY16 9SS, U.K
| | - Sam Hay
- Manchester Institute of Biotechnology and Department of Chemistry, The University of Manchester, 131 Princess Street, Manchester M1 7DN, U.K
| |
Collapse
|
48
|
Léger C, Pitard I, Sadi M, Carvalho N, Brier S, Mechaly A, Raoux-Barbot D, Davi M, Hoos S, Weber P, Vachette P, Durand D, Haouz A, Guijarro JI, Ladant D, Chenal A. Dynamics and structural changes of calmodulin upon interaction with the antagonist calmidazolium. BMC Biol 2022; 20:176. [PMID: 35945584 PMCID: PMC9361521 DOI: 10.1186/s12915-022-01381-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/29/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Calmodulin (CaM) is an evolutionarily conserved eukaryotic multifunctional protein that functions as the major sensor of intracellular calcium signaling. Its calcium-modulated function regulates the activity of numerous effector proteins involved in a variety of physiological processes in diverse organs, from proliferation and apoptosis, to memory and immune responses. Due to the pleiotropic roles of CaM in normal and pathological cell functions, CaM antagonists are needed for fundamental studies as well as for potential therapeutic applications. Calmidazolium (CDZ) is a potent small molecule antagonist of CaM and one the most widely used inhibitors of CaM in cell biology. Yet, CDZ, as all other CaM antagonists described thus far, also affects additional cellular targets and its lack of selectivity hinders its application for dissecting calcium/CaM signaling. A better understanding of CaM:CDZ interaction is key to design analogs with improved selectivity. Here, we report a molecular characterization of CaM:CDZ complexes using an integrative structural biology approach combining SEC-SAXS, X-ray crystallography, HDX-MS, and NMR. RESULTS We provide evidence that binding of a single molecule of CDZ induces an open-to-closed conformational reorientation of the two domains of CaM and results in a strong stabilization of its structural elements associated with a reduction of protein dynamics over a large time range. These CDZ-triggered CaM changes mimic those induced by CaM-binding peptides derived from physiological protein targets, despite their distinct chemical natures. CaM residues in close contact with CDZ and involved in the stabilization of the CaM:CDZ complex have been identified. CONCLUSION Our results provide molecular insights into CDZ-induced dynamics and structural changes of CaM leading to its inhibition and open the way to the rational design of more selective CaM antagonists. Calmidazolium is a potent and widely used inhibitor of calmodulin, a major mediator of calcium-signaling in eukaryotic cells. Structural characterization of calmidazolium-binding to calmodulin reveals that it triggers open-to-closed conformational changes similar to those induced by calmodulin-binding peptides derived from enzyme targets. These results provide molecular insights into CDZ-induced dynamics and structural changes of CaM leading to its inhibition and open the way to the rational design of more selective CaM antagonists.
Collapse
Affiliation(s)
- Corentin Léger
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France
| | - Irène Pitard
- Biological NMR and HDX-MS Technological Platform, CNRS UMR3528, Université Paris Cité, Institut Pasteur, Paris, 75015, France
| | - Mirko Sadi
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France
- Université Paris Cité, Paris, France
| | - Nicolas Carvalho
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France
- Université Paris Cité, Paris, France
| | - Sébastien Brier
- Biological NMR and HDX-MS Technological Platform, CNRS UMR3528, Université Paris Cité, Institut Pasteur, Paris, 75015, France
| | - Ariel Mechaly
- Plate-forme de Cristallographie-C2RT, Université Paris Cité, CNRS UMR3528, Institut Pasteur, Paris, France
| | - Dorothée Raoux-Barbot
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France
| | - Maryline Davi
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France
| | - Sylviane Hoos
- Plateforme de Biophysique Moléculaire, Université Paris Cité, CNRS UMR3528, Institut Pasteur, Paris, France
| | - Patrick Weber
- Plate-forme de Cristallographie-C2RT, Université Paris Cité, CNRS UMR3528, Institut Pasteur, Paris, France
| | - Patrice Vachette
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Dominique Durand
- Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), 91198, Gif-sur-Yvette, France
| | - Ahmed Haouz
- Plate-forme de Cristallographie-C2RT, Université Paris Cité, CNRS UMR3528, Institut Pasteur, Paris, France
| | - J Iñaki Guijarro
- Biological NMR and HDX-MS Technological Platform, CNRS UMR3528, Université Paris Cité, Institut Pasteur, Paris, 75015, France
| | - Daniel Ladant
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France.
| | - Alexandre Chenal
- Biochemistry of Macromolecular Interactions Unit, Department of Structural Biology and Chemistry, CNRS UMR3528, Institut Pasteur, Paris, 75015, France.
| |
Collapse
|
49
|
Ergun Ayva C, Fiorito MM, Guo Z, Edwardraja S, Kaczmarski JA, Gagoski D, Walden P, Johnston WA, Jackson CJ, Nebl T, Alexandrov K. Exploring Performance Parameters of Artificial Allosteric Protein Switches. J Mol Biol 2022; 434:167678. [PMID: 35709893 DOI: 10.1016/j.jmb.2022.167678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 10/18/2022]
Abstract
Biological information processing networks rely on allosteric protein switches that dynamically interconvert biological signals. Construction of their artificial analogues is a central goal of synthetic biology and bioengineering. Receptor domain insertion is one of the leading methods for constructing chimeric protein switches. Here we present an in vitro expression-based platform for the analysis of chimeric protein libraries for which traditional cell survival or cytometric high throughput assays are not applicable. We utilise this platform to screen a focused library of chimeras between PQQ-glucose dehydrogenase and calmodulin. Using this approach, we identified 50 chimeras (approximately 23% of the library) that were activated by calmodulin-binding peptides. We analysed performance parameters of the active chimeras and demonstrated that their dynamic range and response times are anticorrelated, pointing to the existence of an inherent thermodynamic trade-off. We show that the structure of the ligand peptide affects both the response and activation kinetics of the biosensors suggesting that the structure of a ligand:receptor complex can influence the chimera's activation pathway. In order to understand the extent of structural changes in the reporter protein induced by the receptor domains, we have analysed one of the chimeric molecules by CD spectroscopy and hydrogen-deuterium exchange mass spectrometry. We concluded that subtle ligand-induced changes in the receptor domain propagated into the GDH domain and affected residues important for substrate and cofactor binding. Finally, we used one of the identified chimeras to construct a two-component rapamycin biosensor and demonstrated that core switch optimisation translated into improved biosensor performance.
Collapse
Affiliation(s)
- Cagla Ergun Ayva
- ARC Centre of Excellence in Synthetic Biology, Australia; Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Maria M Fiorito
- ARC Centre of Excellence in Synthetic Biology, Australia; Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Zhong Guo
- ARC Centre of Excellence in Synthetic Biology, Australia; Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Selvakumar Edwardraja
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joe A Kaczmarski
- ARC Centre of Excellence in Synthetic Biology, Australia; Research School of Biology, Australian National University, Canberra, ACT 2601, Australia
| | - Dejan Gagoski
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Patricia Walden
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Wayne A Johnston
- Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia
| | - Colin J Jackson
- ARC Centre of Excellence in Synthetic Biology, Australia; Research School of Biology, Australian National University, Canberra, ACT 2601, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia; Research School of Chemistry, Australian National University, Canberra, ACT 2601, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Australian National University, Canberra, ACT 2601, Australia. https://twitter.com/Jackson_Lab
| | - Tom Nebl
- Biology Group, Biomedical Manufacturing Program, CSIRO, Bayview Ave/Research Way, Clayton, VIC 3168, Australia
| | - Kirill Alexandrov
- ARC Centre of Excellence in Synthetic Biology, Australia; Centre for Agriculture and the Bioeconomy, Queensland University of Technology, Brisbane, QLD 4001, Australia; School of Biology and Environmental Science, Queensland University of Technology, Brisbane, QLD 4001, Australia; CSIRO-QUT Synthetic Biology Alliance, Brisbane, QLD 4001, Australia; Centre for Genomics and Personalised Health, Queensland University of Technology, Brisbane, QLD 4001, Australia.
| |
Collapse
|
50
|
Hidden Multivalency in Phosphatase Recruitment by a Disordered AKAP Scaffold. J Mol Biol 2022; 434:167682. [PMID: 35697294 DOI: 10.1016/j.jmb.2022.167682] [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: 05/08/2022] [Revised: 06/06/2022] [Accepted: 06/08/2022] [Indexed: 11/20/2022]
Abstract
Disordered scaffold proteins provide multivalent landing pads that, via a series of embedded Short Linear Motifs (SLiMs), bring together the components of a complex to orchestrate precise spatial and temporal regulation of cellular processes. One such protein is AKAP5 (previously AKAP79), which contains SLiMs that anchor PKA and Calcineurin, and recruit substrate (the TRPV1 receptor). Calcineurin is anchored to AKAP5 by a well-characterised PxIxIT SLiM. Here we show, using a combination of biochemical and biophysical approaches, that the Calcineurin PxIxIT-binding groove also recognises several hitherto unknown lower-affinity SLiMs in addition to the PxIxIT motif. We demonstrate that the assembly is in reality a complex system with conserved SLiMs spanning a wide affinity range. The capture is analogous to that seen for many DNA-binding proteins that have a weak non-specific affinity for DNA outside the canonical binding site, but different in that it involves (i) two proteins, and (ii) hydrophobic rather than electrostatic interactions. It is also compatible with the requirement for both stable anchoring of the enzyme and responsive downstream signalling. We conclude that the AKAP5 C-terminus is enriched in lower-affinity/mini-SLiMs that, together with the canonical SLiM, maintain a structurally disordered but tightly regulated signalosome.
Collapse
|