1
|
Tang Y, Ma H, Shen H. Self-cascade and self-activated nanozyme based on Au quantum dot modified covalent organic framework for rapid and sensitive detection of live bacteria. Mikrochim Acta 2025; 192:362. [PMID: 40383856 DOI: 10.1007/s00604-025-07182-1] [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: 02/06/2025] [Accepted: 04/17/2025] [Indexed: 05/20/2025]
Abstract
Multi-enzymes-guided cascade biocatalysis plays an important role in both nature and industry. Nevertheless, the inherent defects of natural enzymes (e.g., unattractive robustness, sensitivity, and reproducibility under severe catalytic environments) have limited their wider employment. Here, a self-cascade nanozyme was synthesized via depositing Au quantum dots (Au QDs) on iron ions and cysteine-doped porphyrin covalent organic framework (Fe@cpCOF). The in situ introduction of cysteine created a beneficial microenvironment around the iron-porphyrin catalytic center, facilitating the activity of the nanozyme. Through the regulation of Au QDs deposition amount on the surface of Fe@cpCOF, the synthetic nanozyme not only possessed robust glucose oxidase (GOx) mimicking activity but also demonstrated promoted peroxidase (POD) mimicking activity. In the self-cascade system, the innocuous glucose could be constantly transformed to sufficient gluconic acid and H2O2 by Au QDs, preventing the direct application of noxious H2O2 and reducing the detrimental by-effects. In addition, the product gluconic acid decreases the pH of the microenvironment, significantly activating the POD-like bioactivity of Fe@cpCOF. The obtained Au-Fe@cpCOF nanozyme was utilized to simulate the multi-step biocatalytic process in nature, thus constructing an enzyme-free self-cascade biocatalytic sensing platform for specific and wide-spectrum analysis of live bacteria. This study provides a facile assay for pathogen detection in both clinical and daily life.
Collapse
Affiliation(s)
- Yan Tang
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, Zhejiang, China
- The Research Institute of Advanced Technology, Ningbo University, Ningbo, 315211, Zhejiang, China
| | - Hongmei Ma
- Office of Research Affairs, Zhejiang Wanli University, Ningbo, 315100, Zhejiang, China.
| | - Hao Shen
- Key Laboratory of Advanced Mass Spectrometry and Molecular Analysis of Zhejiang Province, Institute of Mass Spectrometry, School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
2
|
Liu J, Zuo Z, Ewing M, Cao Q, Cao L, Li Q, Finkel T, Leppla SH, Liu S. ERK pathway reactivation prevents anthrax toxin lethality in mice. Nat Microbiol 2025; 10:1145-1155. [PMID: 40155776 DOI: 10.1038/s41564-025-01977-x] [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: 07/19/2024] [Accepted: 03/03/2025] [Indexed: 04/01/2025]
Abstract
Lethal toxin (LT), the major virulence factor of Bacillus anthracis, proteolytically inactivates MEKs and disables downstream ERK, p38 and JNK pathway signalling leading to tissue damage and mortality. Therapies for LT-induced damage after host cell internalization of the toxin are lacking. Here we constructed MEK variants in which the LT proteolytic site was modified: MEK2(P10V/A11D), MEK3(I27D) and MEK6(I15D). These variants were resistant to proteolysis by LT. Expression in cells enabled sustained activation of ERK and p38 pathways and promoted cell survival upon LT treatment. Survival of LT- or B. anthracis-challenged MEK variant transgenic mice also increased compared with controls. We found that LT-mediated disruption of both ERK and p38 pathway is essential for anthrax pathogenesis. We show that engagement of upstream receptor tyrosine kinases reactivated the LT-disrupted ERK pathway, as did administering a cocktail of EGF, GM-CSF and FGF2 growth factors, which significantly increased survival of LT- or B. anthracis-challenged mice. These findings offer potential towards developing damage-limiting therapeutic strategies for anthrax.
Collapse
Affiliation(s)
- Jie Liu
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Zehua Zuo
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Michael Ewing
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Qing Cao
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Department of Environmental and Occupational Health, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Liu Cao
- Health Sciences Institute, China Medical University, Shenyang, China
| | - Qi Li
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Toren Finkel
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Stephen H Leppla
- Microbial Pathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Shihui Liu
- Aging Institute of University of Pittsburgh and University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
3
|
Ullah S, Khan SA, Jan S, Din SU, Muhammad N, Rehman ZU, Jan A, Tariq M, Muhammad N, Ghani A, Wasif N, Khan S. Truncated Variants in FAM20A and WDR72 Genes Underlie Autosomal Recessive Amelogenesis Imperfecta in Four Pakistani Families. Biochem Genet 2025:10.1007/s10528-025-11087-2. [PMID: 40108106 DOI: 10.1007/s10528-025-11087-2] [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: 01/13/2025] [Accepted: 03/13/2025] [Indexed: 03/22/2025]
Abstract
Amelogenesis Imperfecta (AI) is a set of hereditary diseases affecting enamel development, leading to various types of enamel defects, potentially impacting oral health unassociated with other generalized defects. AI manifests in syndromic and non-syndromic forms and can be inherited through autosomal recessive, autosomal dominant, or X-linked inheritance patterns. Genetic studies have identified sequence variants in a number of genes (≥ 70) linked to both syndromic and non-syndromic AI, highlighting the genetic diversity underlying the condition. The current study involved clinical evaluation and exome sequencing, aimed at identifying the causative variants in four unrelated consanguineous Pakistani families presenting AI phenotypes. The exome sequencing results revealed a novel homozygous frameshift variant FAM20A: NM_017565.4, c.188dupA; p.(Asp63Glufs*17) in families A, B, and C while a nonsense homozygous variant WDR72: NM_182758.4, c.2686C > T; p. (Arg896*) in family D. The segregation of both variants was confirmed by Sanger sequencing. Bioinformatics analysis predicted the pathogenicity of these genetic variants. These alterations suggest functional consequences, potentially impairing the FAM20A and WDR72 proteins and causing dental anomalies. This investigation significantly broadens our understanding of FAM20A and WDR72's involvement in AI. Furthermore, this study highlights the genetic heterogeneity of AI (involving FAM20A and WDR72 in this study) within the Pakistani population.
Collapse
Affiliation(s)
- Sadaqat Ullah
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Sher Alam Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
- Department of Computer Science and Bioinformatics, Khushal Khan Khatak University, Karak, Pakistan
| | - Samin Jan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Salah Ud Din
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Nazif Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Zia Ur Rehman
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Abid Jan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Muhammad Tariq
- Department of Medical Laboratory Technology, University College of Duba, University of Tabuk, Tabuk, Kingdom of Saudi Arabia
| | - Noor Muhammad
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Abdul Ghani
- Department of Chemistry, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan
| | - Naveed Wasif
- Institute of Human Genetics, Ulm University and Ulm University Medical Center, 89081, Ulm, Germany.
- Institute of Human Genetics, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany.
| | - Saadullah Khan
- Department of Biotechnology and Genetic Engineering, Kohat University of Science & Technology (KUST), Khyber Pakhtunkhwa, Kohat, Pakistan.
| |
Collapse
|
4
|
Tindell SJ, Boeving AG, Aebersold J, Arkov AL. Multiple domains of scaffold Tudor protein play non-redundant roles in Drosophila germline. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.13.643173. [PMID: 40166263 PMCID: PMC11956945 DOI: 10.1101/2025.03.13.643173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Scaffold proteins play crucial roles in subcellular organization and function. In many organisms, proteins with multiple Tudor domains are required for the assembly of membraneless RNA-protein organelles (germ granules) in germ cells. Tudor domains are protein-protein interaction modules which bind to methylated polypeptides. Drosophila Tudor protein contains eleven Tudor domains, which is the highest number known in a single protein. The role of each of these domains in germ cell formation has not been systematically tested and it is not clear if some domains are functionally redundant. Using CRISPR methodology, we generated mutations in several uncharacterized Tudor domains and showed that they all caused defects in germ cell formation. Mutations in individual domains affected Tudor protein differently causing reduction in protein levels, defects in subcellular localization and in the assembly of germ granules. Our data suggest that multiple domains of Tudor protein are all needed for efficient germ cell formation highlighting the rational for keeping many Tudor domains in protein scaffolds of biomolecular condensates in Drosophila and other organisms.
Collapse
Affiliation(s)
- Samuel J. Tindell
- Department of Biological Sciences, Murray State University, Murray, KY, USA
| | - Alyssa G. Boeving
- Department of Biological Sciences, Murray State University, Murray, KY, USA
| | - Julia Aebersold
- Micro/Nano Technology Center, University of Louisville, Louisville, KY, USA
| | - Alexey L. Arkov
- Department of Biological Sciences, Murray State University, Murray, KY, USA
| |
Collapse
|
5
|
Orand T, Delaforge E, Lee A, Kragelj J, Tengo M, Tengo L, Blackledge M, Boeri Erba E, Davis RJ, Palencia A, Jensen MR. Bipartite binding of the intrinsically disordered scaffold protein JIP1 to the kinase JNK1. Proc Natl Acad Sci U S A 2025; 122:e2419915122. [PMID: 39999166 PMCID: PMC11892650 DOI: 10.1073/pnas.2419915122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 01/18/2025] [Indexed: 02/27/2025] Open
Abstract
Scaffold proteins are key players in many signaling pathways where they ensure spatial and temporal control of molecular interactions by simultaneous tethering of multiple signaling components. The protein JIP1 acts as a scaffold within the c-Jun N-terminal kinase (JNK) signaling pathway by assembling three kinases, MLK3, MKK7, and JNK, into a macromolecular complex that enables their specific activation. The recruitment of these kinases depends on the 450-amino acid intrinsically disordered tail of JIP1, however, the structural details of this tail and the molecular mechanisms by which it binds kinases have remained elusive. Here, we provide an atomic resolution structural description of the JIP1 tail, and we study its interaction with the kinase JNK1. Using NMR spectroscopy, we show that JNK1 not only engages with the well-known docking site motif (D-motif) of JIP1, but also interacts with a noncanonical F-motif. We determine the crystal structure of the JIP1-JNK1 complex at 2.35 Å resolution revealing a bipartite binding mode of JIP1. Our work provides insights into the sequence determinants of F-motifs suggesting that these motifs may be more prevalent in JNK substrates than previously recognized. More broadly, our study highlights the power of NMR spectroscopy in uncovering kinase interaction motifs within disordered scaffold proteins, and it paves the way for atomic-resolution interaction studies of JIP1 with its multitude of interaction partners.
Collapse
Affiliation(s)
- Thibault Orand
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Elise Delaforge
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA01655
| | - Jaka Kragelj
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Maud Tengo
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Laura Tengo
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Martin Blackledge
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Elisabetta Boeri Erba
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| | - Roger J. Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA01655
| | - Andrés Palencia
- Institute for Advanced Biosciences, Structural Biology of Novel Targets in Human Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble38000, France
| | - Malene Ringkjøbing Jensen
- Université Grenoble Alpes, Commissariat à l’Énergie Atomique et aux Énergies Alternatives, CNRS, Institut de Biologie Structurale, Grenoble38044, France
| |
Collapse
|
6
|
Pardon E, Wohlkönig A, Steyaert J. Allosteric modulation of protein-protein interactions in signal transduction with Nanobodies. Curr Opin Struct Biol 2025; 92:103022. [PMID: 40024205 DOI: 10.1016/j.sbi.2025.103022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 03/04/2025]
Abstract
Nanobodies (Nbs), the variable domains of heavy-chain only antibodies that naturally occur in camelids, are exquisite molecular tools to stabilize dynamic proteins in unique functional conformations. Recent developments in Nb discovery allow to select allosteric Nbs that perturb the distribution of conformational ensembles of protein complexes that mediate signaling, leading to the allosteric modulation of the signals they transmit. Evidence is also accumulating that such conformational specific Nbs do not stabilize new conformational states but rather change the distribution of existing states to allosterically induce transitions, imprinted by the natural ligands of the system.
Collapse
Affiliation(s)
- Els Pardon
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium
| | - Alex Wohlkönig
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium
| | - Jan Steyaert
- VIB-VUB Center for Structural Biology, VIB, Pleinlaan 2, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, Pleinlaan 2, Brussels, Belgium.
| |
Collapse
|
7
|
van Veldhuisen T, Dijkstra RMJ, Koops AA, Cossar PJ, van Hest JCM, Brunsveld L. Modulation of Protein-Protein Interactions with Molecular Glues in a Synthetic Condensate Platform. J Am Chem Soc 2025; 147:5386-5397. [PMID: 39874979 PMCID: PMC11826995 DOI: 10.1021/jacs.4c17567] [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: 12/09/2024] [Revised: 01/09/2025] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Misregulation of protein-protein interactions (PPIs) underlies many diseases; hence, molecules that stabilize PPIs, known as molecular glues, are promising drug candidates. Identification of novel molecular glues is highly challenging among others because classical biochemical assays in dilute aqueous conditions have limitations for evaluating weak PPIs and their stabilization by molecular glues. This hampers the systematic discovery and evaluation of molecular glues. Here, we present a synthetic condensate platform for the study of PPIs and molecular glues in a crowded macromolecular environment that more closely resembles the dense cellular milieu. With this platform, weak PPIs can be enhanced by sequestration. The condensates, based on amylose derivatives, recruit the hub protein 14-3-3 via affinity-based uptake, which results in high local protein concentrations ideal for the efficient screening of molecular glues. Clients of 14-3-3 are sequestered in the condensates based on their enhanced affinity upon treatment with molecular glues. Fine control over the condensate environment is illustrated by modulating the reactivity of dynamic covalent molecular glues by the adjustment of pH and the redox environment. General applicability of the system for screening of molecular glues is highlighted by using the nuclear receptor PPARγ, which recruits coregulators via an allosteric PPI stabilization mechanism. The condensate environment thus provides a unique dense molecular environment to enhance weak PPIs and enable subsequent evaluation of small-molecule stabilization in a molecular setting chemically en route to the cellular interior.
Collapse
Affiliation(s)
- Thijs
W. van Veldhuisen
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Renske M. J. Dijkstra
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Auke A. Koops
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Peter J. Cossar
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Jan C. M. van Hest
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| | - Luc Brunsveld
- Laboratory of Chemical Biology, Department
of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, The Netherlands
| |
Collapse
|
8
|
Clark LK, Cullati SN. Activation is only the beginning: mechanisms that tune kinase substrate specificity. Biochem Soc Trans 2025:BST20241420. [PMID: 39907081 DOI: 10.1042/bst20241420] [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: 09/10/2024] [Revised: 10/22/2024] [Accepted: 10/29/2024] [Indexed: 02/06/2025]
Abstract
Kinases are master coordinators of cellular processes, but to appropriately respond to the changing cellular environment, each kinase must recognize its substrates, target only those proteins on the correct amino acids, and in many cases, only phosphorylate a subset of potential substrates at any given time. Therefore, regulation of kinase substrate specificity is paramount to proper cellular function, and multiple mechanisms can be employed to achieve specificity. At the smallest scale, characteristics of the substrate such as its linear peptide motif and three-dimensional structure must be complementary to the substrate binding surface of the kinase. This surface is dynamically shaped by the activation loop and surrounding region of the substrate binding groove, which can adopt multiple conformations, often influenced by post-translational modifications. Domain-scale conformational changes can also occur, such as the interaction with pseudosubstrate domains or other regulatory domains in the kinase. Kinases may multimerize or form complexes with other proteins that influence their structure, function, and/or subcellular localization at different times and in response to different signals. This review will illustrate these mechanisms by examining recent work on four serine/threonine kinases: Aurora B, CaMKII, GSK3β, and CK1δ. We find that these mechanisms are often shared by this diverse set of kinases in diverse cellular contexts, so they may represent common strategies that cells use to regulate cell signaling, and it will be enlightening to continue to learn about the depth and robustness of kinase substrate specificity in additional systems.
Collapse
Affiliation(s)
- Landon K Clark
- Department of Chemistry, Western Washington University, Bellingham, WA, U.S.A
| | - Sierra N Cullati
- Department of Chemistry, Western Washington University, Bellingham, WA, U.S.A
| |
Collapse
|
9
|
Currin-Ross D, Al-Izzi SC, Noordstra I, Yap AS, Morris RG. Advecting scaffolds: Controlling the remodeling of actomyosin with anillin. Phys Rev E 2025; 111:024403. [PMID: 40103056 DOI: 10.1103/physreve.111.024403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 11/13/2024] [Indexed: 03/20/2025]
Abstract
We propose and analyze an active hydrodynamic theory that characterizes the effects of the scaffold protein anillin. Anillin is found at major sites of cortical activity, such as adherens junctions and the cytokinetic furrow, where the canonical regulator of actomyosin remodeling is the small GTPase, RhoA. RhoA acts via intermediary "effectors" to increase both the rates of activation of myosin motors and the polymerization of actin filaments. Anillin has been shown to scaffold this action of RhoA-improving critical rates in the signaling pathway without altering the essential biochemistry-but its contribution to the wider spatiotemporal organization of the cortical cytoskeleton remains poorly understood. Here we combine analytics and numerics to show how anillin can nontrivially regulate the cytoskeleton at hydrodynamic scales. At short times, anillin can amplify or dampen existing contractile instabilities, as well as alter the parameter ranges over which they occur. At long times, it can change both the size and speed of steady-state traveling pulses. The primary mechanism that underpins these behaviors is established to be the advection of anillin by myosin II motors, with the specifics relying on the values of two coupling parameters. These codify anillin's effect on local signaling kinetics and can be traced back to its interaction with the acidic phospholipid phosphatidylinositol 4,5-bisphosphate (PIP_{2}), thereby establishing a putative connection between actomyosin remodeling and membrane composition.
Collapse
Affiliation(s)
- Denni Currin-Ross
- The University of Queensland, Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, Brisbane 4000, Australia
- UNSW, School of Physics, Sydney, NSW 2052, Australia
- UNSW, EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, Sydney 2052, Australia
| | - Sami C Al-Izzi
- UNSW, School of Physics, Sydney, NSW 2052, Australia
- UNSW, ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Node, Sydney, NSW 2052, Australia
| | - Ivar Noordstra
- The University of Queensland, Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, Brisbane 4000, Australia
| | - Alpha S Yap
- The University of Queensland, Centre for Cell Biology of Chronic Disease, Institute for Molecular Bioscience, Brisbane 4000, Australia
| | - Richard G Morris
- UNSW, School of Physics, Sydney, NSW 2052, Australia
- UNSW, EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, Sydney 2052, Australia
- UNSW, ARC Centre of Excellence for the Mathematical Analysis of Cellular Systems, Node, Sydney, NSW 2052, Australia
| |
Collapse
|
10
|
Cheng L, Meliala I, Kong Y, Chen J, Proud CG, Björklund M. PEBP1 amplifies mitochondrial dysfunction-induced integrated stress response. eLife 2025; 13:RP102852. [PMID: 39878441 PMCID: PMC11778924 DOI: 10.7554/elife.102852] [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] [Indexed: 01/31/2025] Open
Abstract
Mitochondrial dysfunction is involved in numerous diseases and the aging process. The integrated stress response (ISR) serves as a critical adaptation mechanism to a variety of stresses, including those originating from mitochondria. By utilizing mass spectrometry-based cellular thermal shift assay (MS-CETSA), we uncovered that phosphatidylethanolamine-binding protein 1 (PEBP1), also known as Raf kinase inhibitory protein (RKIP), is thermally stabilized by stresses which induce mitochondrial ISR. Depletion of PEBP1 impaired mitochondrial ISR activation by reducing eukaryotic translation initiation factor 2α (eIF2α) phosphorylation and subsequent ISR gene expression, which was independent of PEBP1's role in inhibiting the RAF/MEK/ERK pathway. Consistently, overexpression of PEBP1 potentiated ISR activation by heme-regulated inhibitor (HRI) kinase, the principal eIF2α kinase in the mitochondrial ISR pathway. Real-time interaction analysis using luminescence complementation in live cells revealed an interaction between PEBP1 and eIF2α, which was disrupted by eIF2α S51 phosphorylation. These findings suggest a role for PEBP1 in amplifying mitochondrial stress signals, thereby facilitating an effective cellular response to mitochondrial dysfunction. Therefore, PEBP1 may be a potential therapeutic target for diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Ling Cheng
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) InstituteHainingChina
| | - Ian Meliala
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) InstituteHainingChina
| | - Yidi Kong
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) InstituteHainingChina
| | - Jingyuan Chen
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) InstituteHainingChina
| | - Christopher G Proud
- Lifelong Health, South Australian Health & Medical Research InstituteAdelaideAustralia
| | - Mikael Björklund
- Centre for Cellular Biology and Signalling, Zhejiang University-University of Edinburgh (ZJU-UoE) InstituteHainingChina
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
11
|
Jia B, Ge J, Ma Y, Sun X, Li Z, Jiang S, Yu H. Spatially Preorganized Hybridization Chain Reaction for the Prompt Diagnosis of Inflammation. Angew Chem Int Ed Engl 2024:e202421022. [PMID: 39716958 DOI: 10.1002/anie.202421022] [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: 11/05/2024] [Revised: 12/17/2024] [Accepted: 12/23/2024] [Indexed: 12/25/2024]
Abstract
Biological systems utilize precise spatial organization to facilitate and regulate information transmission within signaling networks. Inspired by this, artificial scaffolds that enable delicate spatial arrangements are desirable to increase the local concentration of reactants, expedite specific interactions, and minimize undesired interference. In this study, we presented an integrated biosensing nanodevice, termed TRI-HCR, in which hybridization chain reaction (HCR) probes were precisely organized on a triangular DNA origami nanostructure (TRI) with finely-tuned distance, quantity, and pattern. Compared to traditional HCR in the free form, this nanodevice demonstrated increased reaction rate and signal level. We further employed the optimized TRI-HCR for in vivo imaging of a nucleic acid biomarker of inflammatory diseases. In both acute gouty arthritis (AGA) and sepsis-associated acute kidney injury (SA-AKI) model mice, TRI-HCR was capable of diagnosing inflammation in the early stages, significantly earlier than histological examination. We anticipate that this precise spatial preorganization strategy for HCR holds promise for broader applications in early disease detection and monitoring.
Collapse
Affiliation(s)
- Bin Jia
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Jingru Ge
- State Key Laboratory of Analytical Chemistry for Life Science, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Yuxuan Ma
- State Key Laboratory of Analytical Chemistry for Life Science, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Xiaolei Sun
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Zhe Li
- State Key Laboratory of Analytical Chemistry for Life Science, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Shuoxing Jiang
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| | - Hanyang Yu
- State Key Laboratory of Coordination Chemistry, Department of Biomedical Engineering, College of Engineering and Applied Sciences, Chemistry and Biomedicine Innovation Center (ChemBIC), ChemBioMed Interdisciplinary Research Center, Nanjing University, Nanjing, Jiangsu, 210023, P. R. China
| |
Collapse
|
12
|
Jozwiak A, Panda S, Akiyama R, Yoneda A, Umemoto N, Saito K, Yasumoto S, Muranaka T, Gharat SA, Kazachkova Y, Dong Y, Arava S, Goliand I, Nevo R, Rogachev I, Meir S, Mizutani M, Aharoni A. A cellulose synthase-like protein governs the biosynthesis of Solanum alkaloids. Science 2024; 386:eadq5721. [PMID: 39700293 DOI: 10.1126/science.adq5721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 11/01/2024] [Indexed: 12/21/2024]
Abstract
Decades of research on the infamous antinutritional steroidal glycoalkaloids (SGAs) in Solanaceae plants have provided deep insights into their metabolism and roles. However, engineering SGAs in heterologous hosts has remained a challenge. We discovered that a protein evolved from the machinery involved in building plant cell walls is the crucial link in the biosynthesis of SGAs. We show that cellulose synthase-like M [GLYCOALKALOID METABOLISM15 (GAME15)] functions both as a cholesterol glucuronosyltransferase and a scaffold protein. Silencing GAME15 depletes SGAs, which makes plants more vulnerable to pests. Our findings illuminate plant evolutionary adaptations that balance chemical defense and self-toxicity and open possibilities for producing steroidal compounds in heterologous systems for food, cosmetics, and pharmaceuticals.
Collapse
Affiliation(s)
- Adam Jozwiak
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
- Department of Botany and Plant Sciences, University of California, Riverside, CA, USA
| | - Sayantan Panda
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
- Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Ryota Akiyama
- Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Ayano Yoneda
- Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Naoyuki Umemoto
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa, Japan
| | - Kazuki Saito
- RIKEN Center for Sustainable Resource Science, Yokohama, Kanagawa, Japan
| | - Shuhei Yasumoto
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Toshiya Muranaka
- Department of Biotechnology, Graduate School of Engineering, Osaka University, Osaka, Japan
| | - Sachin A Gharat
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yana Kazachkova
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yonghui Dong
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Shlomy Arava
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Inna Goliand
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Reinat Nevo
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Ilana Rogachev
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Sagit Meir
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Masaharu Mizutani
- Graduate School of Agricultural Science, Kobe University, Hyogo, Japan
| | - Asaph Aharoni
- Department of Plant and Environmental Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
13
|
Zhao C, Jiang X, Wang M, Gui S, Yan X, Dong Y, Liu D. Constructing protein-functionalized DNA origami nanodevices for biological applications. NANOSCALE 2024; 17:142-157. [PMID: 39564893 DOI: 10.1039/d4nr03599b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2024]
Abstract
In living systems, proteins participate in various physiological processes and the clustering of multiple proteins is essential for efficient signaling. Therefore, understanding the effects of the number, distance and orientation of proteins is of great significance. With programmability and addressability, DNA origami technology has enabled fabrication of sophisticated nanostructures with precise arrangement and orientation control of proteins to investigate the effects of these parameters on protein-involved cellular processes. Herein, we highlight the construction and applications of protein-functionalized DNA origami nanodevices. After the introduction of the structural design principles of DNA origami and the strategies of protein-DNA conjugation, the emerging applications of protein-functionalized DNA origami nanodevices with controlled key parameters are mainly discussed, including the regulation of enzyme cascade reactions, modulation of cellular behaviours, drug delivery therapy and protein structural analysis. Finally, the current challenges and potential directions of protein-functionalized DNA origami nanodevices are also presented, advancing their applications in biomedicine, cell biology and structural biology.
Collapse
Affiliation(s)
- Chuangyuan Zhao
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Xinran Jiang
- School of Life Sciences Fudan University, Shanghai, 200433, China
| | - Miao Wang
- Chemistry and chemical biology, Cornell university, 122 Baker Laboratory, Ithaca, NY 14853, USA
| | - Songbai Gui
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, 100071, Beijing, China.
| | - Xin Yan
- Department of Sports Medicine, Beijing Key Laboratory of Sports Injuries, Peking University Third Hospital, Beijing, 100191, China.
| | - Yuanchen Dong
- CAS Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, 100190, China
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Dongsheng Liu
- Engineering Research Center of Advanced Rare Earth Materials, (Ministry of Education), Department of Chemistry, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
14
|
Leopold AV, Verkhusha VV. Engineering signalling pathways in mammalian cells. Nat Biomed Eng 2024; 8:1523-1539. [PMID: 39237709 PMCID: PMC11852397 DOI: 10.1038/s41551-024-01237-z] [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: 10/04/2023] [Accepted: 06/14/2024] [Indexed: 09/07/2024]
Abstract
In mammalian cells, signalling pathways orchestrate cellular growth, differentiation and survival, as well as many other processes that are essential for the proper functioning of cells. Here we describe cutting-edge genetic-engineering technologies for the rewiring of signalling networks in mammalian cells. Specifically, we describe the recombination of native pathway components, cross-kingdom pathway transplantation, and the development of de novo signalling within cells and organelles. We also discuss how, by designing signalling pathways, mammalian cells can acquire new properties, such as the capacity for photosynthesis, the ability to detect cancer and senescent cell markers or to synthesize hormones or metabolites in response to chemical or physical stimuli. We also review the applications of mammalian cells in biocomputing. Technologies for engineering signalling pathways in mammalian cells are advancing basic cellular biology, biomedical research and drug discovery.
Collapse
Affiliation(s)
- Anna V Leopold
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Vladislav V Verkhusha
- Medicum, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
- Department of Genetics and Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
15
|
Guo B, Kim EJ, Zhu Y, Wang K, Russinova E. Shaping Brassinosteroid Signaling through Scaffold Proteins. PLANT & CELL PHYSIOLOGY 2024; 65:1608-1617. [PMID: 38590034 DOI: 10.1093/pcp/pcae040] [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: 01/02/2024] [Revised: 03/06/2024] [Accepted: 04/04/2024] [Indexed: 04/10/2024]
Abstract
Cellular responses to internal and external stimuli are orchestrated by intricate intracellular signaling pathways. To ensure an efficient and specific information flow, cells employ scaffold proteins as critical signaling organizers. With the ability to bind multiple signaling molecules, scaffold proteins can sequester signaling components within specific subcellular domains or modulate the efficiency of signal transduction. Scaffolds can also tune the output of signaling pathways by serving as regulatory targets. This review focuses on scaffold proteins associated with the plant GLYCOGEN SYNTHASE KINASE3-like kinase, BRASSINOSTEROID-INSENSITIVE2 (BIN2), that serves as a key negative regulator of brassinosteroid (BR) signaling. Here, we summarize current understanding of how scaffold proteins actively shape BR signaling outputs and cross-talk in plant cells via interactions with BIN2.
Collapse
Affiliation(s)
- Boyu Guo
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technolgiepark 71, Ghent 9052, Belgium
- Center for Plant Systems Biology, VIB, Technolgiepark 71, Ghent 9052, Belgium
- College of Life Sciences, Wuhan University, 299 Bayi Road, Wuchang District, Wuhan 430072, China
| | - Eun-Ji Kim
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technolgiepark 71, Ghent 9052, Belgium
- Center for Plant Systems Biology, VIB, Technolgiepark 71, Ghent 9052, Belgium
| | - Yuxian Zhu
- College of Life Sciences, Wuhan University, 299 Bayi Road, Wuchang District, Wuhan 430072, China
| | - Kun Wang
- College of Life Sciences, Wuhan University, 299 Bayi Road, Wuchang District, Wuhan 430072, China
| | - Eugenia Russinova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, Technolgiepark 71, Ghent 9052, Belgium
- Center for Plant Systems Biology, VIB, Technolgiepark 71, Ghent 9052, Belgium
| |
Collapse
|
16
|
Kang J, Schroeder ME, Lee Y, Kapoor C, Yu E, Tarr TB, Titterton K, Zeng M, Park D, Niederst E, Wei D, Feng G, Boyden ES. Multiplexed expansion revealing for imaging multiprotein nanostructures in healthy and diseased brain. Nat Commun 2024; 15:9722. [PMID: 39521775 PMCID: PMC11550395 DOI: 10.1038/s41467-024-53729-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Proteins work together in nanostructures in many physiological contexts and disease states. We recently developed expansion revealing (ExR), which expands proteins away from each other, in order to support better labeling with antibody tags and nanoscale imaging on conventional microscopes. Here, we report multiplexed expansion revealing (multiExR), which enables high-fidelity antibody visualization of >20 proteins in the same specimen, over serial rounds of staining and imaging. Across all datasets examined, multiExR exhibits a median round-to-round registration error of 39 nm, with a median registration error of 25 nm when the most stringent form of the protocol is used. We precisely map 23 proteins in the brain of 5xFAD Alzheimer's model mice, and find reductions in synaptic protein cluster volume, and co-localization of specific AMPA receptor subunits with amyloid-beta nanoclusters. We visualize 20 synaptic proteins in specimens of mouse primary somatosensory cortex. multiExR may be of broad use in analyzing how different kinds of protein are organized amidst normal and pathological processes in biology.
Collapse
Affiliation(s)
- Jinyoung Kang
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
| | - Margaret E Schroeder
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
| | - Youngmi Lee
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Chaitanya Kapoor
- Department of Electrical and Electronics Engineering, BITS Pilani, Rajasthan, India
| | - Eunah Yu
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Tyler B Tarr
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kat Titterton
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Menglong Zeng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Demian Park
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
| | - Emily Niederst
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA
| | - Donglai Wei
- Department of Computer Science, Boston College, Chestnut Hill, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA
- Yang Tan Collective, MIT, Cambridge, MA, USA
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Edward S Boyden
- McGovern Institute for Brain Research, MIT, Cambridge, MA, USA.
- Yang Tan Collective, MIT, Cambridge, MA, USA.
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA.
- Center for Neurobiological Engineering and K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, USA.
- Department of Biological Engineering, MIT, Cambridge, MA, USA.
- Koch Institute, MIT, Cambridge, MA, USA.
- Howard Hughes Medical Institute, Cambridge, MA, USA.
- Media Arts and Sciences, MIT, Cambridge, MA, USA.
| |
Collapse
|
17
|
Zhu W, Fu Y, Zhou H, Zhou Y, Zhang D, Wang Y, Su Y, Li Z, Liang J. RACK1 links phyB and BES1 to coordinate brassinosteroid-dependent root meristem development. THE NEW PHYTOLOGIST 2024; 244:883-899. [PMID: 39149918 DOI: 10.1111/nph.20055] [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: 06/08/2024] [Accepted: 07/29/2024] [Indexed: 08/17/2024]
Abstract
Light and brassinosteroids (BR) are indispensable for plant growth and control cell division in the apical meristem. However, how external light signals cooperate with internal brassinosteroids to program root meristem development remains elusive. We reveal that the photoreceptor phytochrome B (phyB) guides the scaffold protein RACK1 to coordinate BR signaling for maintaining root meristematic activity. phyB and RACK1 promote early root meristem development. Mechanistically, RACK1 could reinforce the phyB-SPA1 association by interacting with both phyB and SPA1, which indirectly affects COP1-dependent RACK1 degradation, resulting in the accumulation of RACK1 in roots. Subsequently, RACK1 interacts with BES1 to repress its DNA-binding activity toward the target gene CYCD3;1, leading to the release of BES1-mediated inhibition of CYCD3;1 transcription, and hence the promotion of root meristem development. Our study provides mechanistic insights into the regulation of root meristem development by combination of light and phytohormones signals through the photoreceptors and scaffold proteins.
Collapse
Affiliation(s)
- Wei Zhu
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yajuan Fu
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Hua Zhou
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yeling Zhou
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dayan Zhang
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yuzhu Wang
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yujing Su
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiyong Li
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiansheng Liang
- Shenzhen Key Laboratory of Plant Genetic Engineering and Molecular Design, Department of Biology, School of Life Sciences, Institute of Plant and Food Science, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
18
|
Philbrook A, O’Donnell MP, Grunenkovaite L, Sengupta P. Cilia structure and intraflagellar transport differentially regulate sensory response dynamics within and between C. elegans chemosensory neurons. PLoS Biol 2024; 22:e3002892. [PMID: 39591402 PMCID: PMC11593760 DOI: 10.1371/journal.pbio.3002892] [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: 05/10/2024] [Accepted: 10/10/2024] [Indexed: 11/28/2024] Open
Abstract
Sensory neurons contain morphologically diverse primary cilia that are built by intraflagellar transport (IFT) and house sensory signaling molecules. Since both ciliary structural and signaling proteins are trafficked via IFT, it has been challenging to decouple the contributions of IFT and cilia structure to neuronal responses. By acutely inhibiting IFT without altering cilia structure and vice versa, here we describe the differential roles of ciliary trafficking and sensory ending morphology in shaping chemosensory responses in Caenorhabditis elegans. We show that a minimum cilium length but not continuous IFT is necessary for a subset of responses in the ASH nociceptive neurons. In contrast, neither cilia nor continuous IFT are necessary for odorant responses in the AWA olfactory neurons. Instead, continuous IFT differentially modulates response dynamics in AWA. Upon acute inhibition of IFT, cilia-destined odorant receptors are shunted to ectopic branches emanating from the AWA cilia base. Spatial segregation of receptors in these branches from a cilia-restricted regulatory kinase results in odorant desensitization defects, highlighting the importance of precise organization of signaling molecules at sensory endings in regulating response dynamics. We also find that adaptation of AWA responses upon repeated exposure to an odorant is mediated by IFT-driven removal of its cognate receptor, whereas adaptation to a second odorant is regulated via IFT-independent mechanisms. Our results reveal unexpected complexity in the contribution of IFT and cilia organization to the regulation of responses even within a single chemosensory neuron type and establish a critical role for these processes in the precise modulation of olfactory behaviors.
Collapse
Affiliation(s)
- Alison Philbrook
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Michael P. O’Donnell
- Department of Molecular, Cellular, and Developmental Biology, Yale University, Connecticut, United States of America
| | - Laura Grunenkovaite
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| | - Piali Sengupta
- Department of Biology, Brandeis University, Waltham, Massachusetts, United States of America
| |
Collapse
|
19
|
Bova V, Mannino D, Capra AP, Lanza M, Palermo N, Filippone A, Esposito E. CK and LRRK2 Involvement in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:11661. [PMID: 39519213 PMCID: PMC11546471 DOI: 10.3390/ijms252111661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/21/2024] [Accepted: 10/27/2024] [Indexed: 11/16/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are currently the most widespread neuronal pathologies in the world. Among these, the most widespread are Alzheimer's disease (AD), dementia, Parkinson's disease (PD), amyotrophic lateral sclerosis (ALS), and Huntington's disease (HD)-all characterized by a progressive loss of neurons in specific regions of the brain leading to varied clinical symptoms. At the basis of neurodegenerative diseases, an emerging role is played by genetic mutations in the leucine-rich repeat kinase 2 (LRRK2) gene that cause increased LRRK2 activity with consequent alteration of neuronal autophagy pathways. LRRK2 kinase activity requires GTPase activity which functions independently of kinase activity and is required for neurotoxicity and to potentiate neuronal death. Important in the neurodegeneration process is the upregulation of casein kinase (CK), which causes the alteration of the AMPK pathway by enhancing the phosphorylation of α-synuclein and huntingtin proteins, known to be involved in PD and HD, and increasing the accumulation of the amyloid-β protein (Aβ) for AD. Recent research has identified CK of the kinases upstream of LRRK2 as a regulator of the stability of the LRRK2 protein. Based on this evidence, this review aims to understand the direct involvement of individual kinases in NDDs and how their crosstalk may impact the pathogenesis and early onset of neurodegenerative diseases.
Collapse
Affiliation(s)
- Valentina Bova
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Anna Paola Capra
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Marika Lanza
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Nicoletta Palermo
- Department of Biochemical, Dental, Morphological and Functional Imaging, University of Messina, Via Consolare Valeria, 98125 Messina, Italy;
| | - Alessia Filippone
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceuticals and Environmental Sciences, University of Messina, Viale Stagno d’Alcontres, 98166 Messina, Italy; (V.B.); (D.M.); (A.P.C.); (M.L.); (E.E.)
| |
Collapse
|
20
|
Leslie K, Berry SS, Miller GJ, Mahon CS. Sugar-Coated: Can Multivalent Glycoconjugates Improve upon Nature's Design? J Am Chem Soc 2024; 146:27215-27232. [PMID: 39340450 PMCID: PMC11467903 DOI: 10.1021/jacs.4c08818] [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] [Revised: 09/16/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Multivalent interactions between receptors and glycans play an important role in many different biological processes, including pathogen infection, self-recognition, and the immune response. The growth in the number of tools and techniques toward the assembly of multivalent glycoconjugates means it is possible to create synthetic systems that more and more closely resemble the diversity and complexity we observe in nature. In this Perspective we present the background to the recognition and binding enabled by multivalent interactions in nature, and discuss the strategies used to construct synthetic glycoconjugate equivalents. We highlight key discoveries and the current state of the art in their applications to glycan arrays, vaccines, and other therapeutic and diagnostic tools, with an outlook toward some areas we believe are of most interest for future work in this area.
Collapse
Affiliation(s)
- Kathryn
G. Leslie
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| | - Sian S. Berry
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Gavin J. Miller
- Centre
for Glycoscience and School of Chemical and Physical Sciences, Keele University, Keele, Staffordshire ST5 5BG, United Kingdom
| | - Clare S. Mahon
- Department
of Chemistry, Durham University, Durham DH1 3LE, United Kingdom
| |
Collapse
|
21
|
Bradley D, Hogrebe A, Dandage R, Dubé AK, Leutert M, Dionne U, Chang A, Villén J, Landry CR. The fitness cost of spurious phosphorylation. EMBO J 2024; 43:4720-4751. [PMID: 39256561 PMCID: PMC11480408 DOI: 10.1038/s44318-024-00200-7] [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: 10/25/2023] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 09/12/2024] Open
Abstract
The fidelity of signal transduction requires the binding of regulatory molecules to their cognate targets. However, the crowded cell interior risks off-target interactions between proteins that are functionally unrelated. How such off-target interactions impact fitness is not generally known. Here, we use Saccharomyces cerevisiae to inducibly express tyrosine kinases. Because yeast lacks bona fide tyrosine kinases, the resulting tyrosine phosphorylation is biologically spurious. We engineered 44 yeast strains each expressing a tyrosine kinase, and quantitatively analysed their phosphoproteomes. This analysis resulted in ~30,000 phosphosites mapping to ~3500 proteins. The number of spurious pY sites generated correlates strongly with decreased growth, and we predict over 1000 pY events to be deleterious. However, we also find that many of the spurious pY sites have a negligible effect on fitness, possibly because of their low stoichiometry. This result is consistent with our evolutionary analyses demonstrating a lack of phosphotyrosine counter-selection in species with tyrosine kinases. Our results suggest that, alongside the risk for toxicity, the cell can tolerate a large degree of non-functional crosstalk as interaction networks evolve.
Collapse
Affiliation(s)
- David Bradley
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, QC, Canada
- Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Université du Québec à Montréal, Montréal, QC, Canada
- Université Laval Big Data Research Center (BDRC_UL), Québec, QC, Canada
- Department of Biology, Université Laval, Québec, QC, Canada
| | - Alexander Hogrebe
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Rohan Dandage
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, QC, Canada
- Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Université du Québec à Montréal, Montréal, QC, Canada
- Université Laval Big Data Research Center (BDRC_UL), Québec, QC, Canada
- Department of Biology, Université Laval, Québec, QC, Canada
| | - Alexandre K Dubé
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, QC, Canada
- Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Université du Québec à Montréal, Montréal, QC, Canada
- Université Laval Big Data Research Center (BDRC_UL), Québec, QC, Canada
- Department of Biology, Université Laval, Québec, QC, Canada
| | - Mario Leutert
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Institute of Molecular Systems Biology, ETH Zürich, Zürich, Switzerland
| | - Ugo Dionne
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, QC, Canada
- Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Université du Québec à Montréal, Montréal, QC, Canada
- Université Laval Big Data Research Center (BDRC_UL), Québec, QC, Canada
- Department of Biology, Université Laval, Québec, QC, Canada
| | - Alexis Chang
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
| | - Judit Villén
- Department of Genome Sciences, University of Washington, Seattle, WA, USA.
| | - Christian R Landry
- Institut de Biologie Intégrative et des Systèmes (IBIS), Université Laval, Québec, QC, Canada.
- Department of Biochemistry, Microbiology and Bioinformatics, Université Laval, Québec, QC, Canada.
- Quebec Network for Research on Protein Function, Engineering, and Applications (PROTEO), Université du Québec à Montréal, Montréal, QC, Canada.
- Université Laval Big Data Research Center (BDRC_UL), Québec, QC, Canada.
- Department of Biology, Université Laval, Québec, QC, Canada.
| |
Collapse
|
22
|
Taghvimi S, Soltani Fard E, Khatami SH, Zafaranchi Z M S, Taheri-Anganeh M, Movahedpour A, Ghasemi H. lncRNA HOTAIR and Cardiovascular diseases. Funct Integr Genomics 2024; 24:165. [PMID: 39294422 DOI: 10.1007/s10142-024-01444-6] [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: 07/09/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/20/2024]
Abstract
Cardiovascular diseases (CVDs) a major contributor to global mortality rates, with a steadily rising prevalence observed across the world. Understanding the molecular mechanisms that underlie the signaling pathways implicated in the pathogenesis of CVDs represents a salient and advantageous avenue toward the development of precision and targeted therapeutics. A recent development in CVDs research is the discovery of long non-coding RNAs (lncRNAs), which are now understood to have crucial roles in the onset and development of several pathophysiological processes. The distinct expression patterns exhibited by lncRNAs in various CVDs contexts, present a significant opportunity for their utilization as both biomarkers and targets for therapeutic intervention. Among the various identified lncRNAs, HOX antisense intergenic RNA (HOTAIR) functions as signaling molecules that are significantly implicated in the pathogenesis of cardiovascular disorders in response to risk factors. HOTAIR has been observed to circulate within the bloodstream and possesses an integral epigenetic regulatory function in the transcriptional pathways of many diseases. Recent studies have suggested that HOTAIR offers promise as a biomarker for the detection and treatment of CVDs. The investigation on HOTAIR's role in CVDs, however, is still in its early phases. The goal of the current study is to give a thorough overview of recent developments in the field of analyzing the molecular mechanism of HOTAIR in controlling the pathophysiological processes of CVDs as well as its possible therapeutic uses.
Collapse
Affiliation(s)
- Sina Taghvimi
- Department of Biology, Faculty of Sciences, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Elahe Soltani Fard
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
- Department of Molecular Medicine, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Seyyed Hossein Khatami
- Student Research Committee, Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sara Zafaranchi Z M
- Department of Molecular Medicine, School of Advanced Technologies in Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mortaza Taheri-Anganeh
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Ahmad Movahedpour
- Cellular and Molecular Research Center, Yasuj University of Medical Sciences, Yasuj, Iran.
| | - Hassan Ghasemi
- Research Center for Environmental Contaminants (RCEC), Abadan University of Medical Sciences, Abadan, Iran.
| |
Collapse
|
23
|
Meziadi A, Bloquert V, Greschner AA, de Haan HW, Gauthier MA. Harnessing Water Competition to Drive Enzyme Crosstalk. Biomacromolecules 2024; 25:6072-6081. [PMID: 39150387 DOI: 10.1021/acs.biomac.4c00727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
In nature, enzymatic pathways often involve compartmentalization effects that can modify the intrinsic activity and specificity of the different enzymes involved. Consequently, extensive research has focused on replicating and studying the compartmentalization effects on individual enzymes and on multistep enzyme "cascade" reactions. This study explores the influence of compartmentalization achieved using molecular crowding on the glucose oxidase/horseradish peroxidase (GOx/HRP) cascade reaction. The crowder tested is methoxy poly(ethylene glycol) (mPEG) that can, depending on conditions, promote GOx and HRP coassociation at the nanoscale and extend their contact time. Low-molecular-weight mPEG (0.35 kDa), but not mPEG of higher molecular weights (5 or 20 kDa), significantly enhanced the cascade reaction where up to a 20-fold increase in the rate of the cascade reaction was observed under some conditions. The combined analyses emphasize the particularity of low-molecular-weight mPEG and point toward mPEG-induced coassociation of HRP and GOx, producing nearest crowded neighbor effects of HRP on GOx, and vice versa. These altered the nanoscale environments of these enzymes, which influenced substrate affinity. Using mPEG to promote protein coassociation is simple and does not chemically modify the proteins studied. This approach could be of interest for more broadly characterizing nearest crowded neighbor effects (i.e., protein-protein interactions) for multiprotein systems (i.e., more than just two), thus making it an interesting tool for studying very complex systems, such as those found in nature.
Collapse
Affiliation(s)
- Ahlem Meziadi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Victoria Bloquert
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Andrea A Greschner
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| | - Hendrick W de Haan
- Faculty of Science, University of Ontario Institute of Technology, Oshawa, Ontario L1H 7K4, Canada
| | - Marc A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, Quebec J3X 1P7, Canada
| |
Collapse
|
24
|
Mariño Pérez L, Ielasi FS, Lee A, Delaforge E, Juyoux P, Tengo M, Davis RJ, Palencia A, Jensen MR. Structural basis of homodimerization of the JNK scaffold protein JIP2 and its heterodimerization with JIP1. Structure 2024; 32:1394-1403.e5. [PMID: 39013462 DOI: 10.1016/j.str.2024.06.010] [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: 01/22/2024] [Revised: 03/18/2024] [Accepted: 06/19/2024] [Indexed: 07/18/2024]
Abstract
The scaffold proteins JIP1 and JIP2 intervene in the c-Jun N-terminal kinase (JNK) pathway to mediate signaling specificity by coordinating the simultaneous assembly of multiple kinases. Using NMR, we demonstrate that JIP1 and JIP2 heterodimerize via their SH3 domains with the affinity of heterodimerization being comparable to homodimerization. We present the high-resolution crystal structure of the JIP2-SH3 homodimer and the JIP1-JIP2-SH3 heterodimeric complex. The JIP2-SH3 structure reveals how charge differences in residues at its dimer interface lead to formation of compensatory hydrogen bonds and salt bridges, distinguishing it from JIP1-SH3. In the JIP1-JIP2-SH3 complex, structural features of each homodimer are employed to stabilize the heterodimer. Building on these insights, we identify key residues crucial for stabilizing the dimer of both JIP1 and JIP2. Through targeted mutations in cellulo, we demonstrate a functional role for the dimerization of the JIP1 and JIP2 scaffold proteins in activation of the JNK signaling pathway.
Collapse
Affiliation(s)
- Laura Mariño Pérez
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France; Departament de Química, Universitat de les Illes Balears, Institut Universitari d'Investigació en Ciències de la Salut (IUNICS), Institut de Recerca en Ciències de la Salut (IdISBa), Palma, Spain
| | - Francesco S Ielasi
- Institute for Advanced Biosciences (IAB), Structural Biology of Novel Targets in Human Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France
| | - Alexandra Lee
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | | | - Pauline Juyoux
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Maud Tengo
- University Grenoble Alpes, CEA, CNRS, IBS, Grenoble, France
| | - Roger J Davis
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Andrés Palencia
- Institute for Advanced Biosciences (IAB), Structural Biology of Novel Targets in Human Diseases, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, Grenoble, France.
| | | |
Collapse
|
25
|
Enos MD, Gavagan M, Jameson N, Zalatan JG, Weis WI. Structural and functional effects of phosphopriming and scaffolding in the kinase GSK-3β. Sci Signal 2024; 17:eado0881. [PMID: 39226374 PMCID: PMC11461088 DOI: 10.1126/scisignal.ado0881] [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: 01/18/2024] [Accepted: 08/02/2024] [Indexed: 09/05/2024]
Abstract
Glycogen synthase kinase 3β (GSK-3β) targets specific signaling pathways in response to distinct upstream signals. We used structural and functional studies to dissect how an upstream phosphorylation step primes the Wnt signaling component β-catenin for phosphorylation by GSK-3β and how scaffolding interactions contribute to this reaction. Our crystal structure of GSK-3β bound to a phosphoprimed β-catenin peptide confirmed the expected binding mode of the phosphoprimed residue adjacent to the catalytic site. An aspartate phosphomimic in the priming site of β-catenin adopted an indistinguishable structure but reacted approximately 1000-fold slower than the native phosphoprimed substrate. This result suggests that substrate positioning alone is not sufficient for catalysis and that native phosphopriming interactions are necessary. We also obtained a structure of GSK-3β with an extended peptide from the scaffold protein Axin that bound with greater affinity than that of previously crystallized Axin fragments. This structure neither revealed additional contacts that produce the higher affinity nor explained how substrate interactions in the GSK-3β active site are modulated by remote Axin binding. Together, our findings suggest that phosphopriming and scaffolding produce small conformational changes or allosteric effects, not captured in the crystal structures, that activate GSK-3β and facilitate β-catenin phosphorylation. These results highlight limitations in our ability to predict catalytic activity from structure and have potential implications for the role of natural phosphomimic mutations in kinase regulation and phosphosite evolution.
Collapse
Affiliation(s)
- Michael D. Enos
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Maire Gavagan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Noel Jameson
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - Jesse G. Zalatan
- Department of Chemistry, University of Washington, Seattle, WA 98195, USA
| | - William I. Weis
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94035, USA
- Department Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94035, USA
| |
Collapse
|
26
|
Bremer HJ, Herppich AA, Pflum MKH. Kinase-catalyzed crosslinking: A comparison of ATP-crosslinker analogs. Bioorg Med Chem Lett 2024; 109:129841. [PMID: 38838920 PMCID: PMC11305616 DOI: 10.1016/j.bmcl.2024.129841] [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: 04/04/2024] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 06/07/2024]
Abstract
Protein phosphorylation is catalyzed by kinases to regulate cellular events and disease states. Identifying kinase-substrate relationships represents a powerful strategy to understand cell biology and disease yet remains challenging due to the rapid dynamics of phosphorylation. Over the last decade, several γ-phosphoryl modified ATP analogs containing crosslinkers were developed to covalently conjugate kinases, their substrates, and their associated proteins for subsequent characterization. Here, kinetics and crosslinking experiments demonstrated that the UV-activated analogs, ATP-aryl azide and ATP-benzophenone, offered the most robust crosslinking, whereas electrophilic ATP-aryl fluorosulfate promoted the most effective proximity-enabled crosslinking. The data will guide future applications of kinase-catalyzed crosslinking to study normal and disease biology.
Collapse
Affiliation(s)
- Hannah J Bremer
- Wayne State University, Department of Chemistry, 5101 Cass Ave., Detroit, MI 48202, United States
| | - Andrew A Herppich
- Wayne State University, Department of Chemistry, 5101 Cass Ave., Detroit, MI 48202, United States
| | - Mary Kay H Pflum
- Wayne State University, Department of Chemistry, 5101 Cass Ave., Detroit, MI 48202, United States.
| |
Collapse
|
27
|
Cabral G, Moss WJ, Brown KM. Proteomic approaches for protein kinase substrate identification in Apicomplexa. Mol Biochem Parasitol 2024; 259:111633. [PMID: 38821187 PMCID: PMC11194964 DOI: 10.1016/j.molbiopara.2024.111633] [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: 03/30/2024] [Revised: 05/10/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024]
Abstract
Apicomplexa is a phylum of protist parasites, notable for causing life-threatening diseases including malaria, toxoplasmosis, cryptosporidiosis, and babesiosis. Apicomplexan pathogenesis is generally a function of lytic replication, dissemination, persistence, host cell modification, and immune subversion. Decades of research have revealed essential roles for apicomplexan protein kinases in establishing infections and promoting pathogenesis. Protein kinases modify their substrates by phosphorylating serine, threonine, tyrosine, or other residues, resulting in rapid functional changes in the target protein. Post-translational modification by phosphorylation can activate or inhibit a substrate, alter its localization, or promote interactions with other proteins or ligands. Deciphering direct kinase substrates is crucial to understand mechanisms of kinase signaling, yet can be challenging due to the transient nature of kinase phosphorylation and potential for downstream indirect phosphorylation events. However, with recent advances in proteomic approaches, our understanding of kinase function in Apicomplexa has improved dramatically. Here, we discuss methods that have been used to identify kinase substrates in apicomplexan parasites, classifying them into three main categories: i) kinase interactome, ii) indirect phosphoproteomics and iii) direct labeling. We briefly discuss each approach, including their advantages and limitations, and highlight representative examples from the Apicomplexa literature. Finally, we conclude each main category by introducing prospective approaches from other fields that would benefit kinase substrate identification in Apicomplexa.
Collapse
Affiliation(s)
- Gabriel Cabral
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - William J Moss
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kevin M Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
28
|
Mashima T, Yamanaka M, Yoshida A, Kobayashi N, Kanaoka Y, Uchihashi T, Hirota S. Construction of ligand-binding controlled hemoprotein assemblies utilizing 3D domain swapping. Chem Commun (Camb) 2024; 60:9440-9443. [PMID: 39139060 DOI: 10.1039/d4cc03129f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
Association-controllable hemoprotein assemblies were constructed from a fusion protein containing two c-type cytochrome units using 3D domain swapping. The hemoprotein assembly exhibited a dynamic exchange between cyclic and linear structures and could be regulated by carbon monoxide (CO) and imidazole binding.
Collapse
Affiliation(s)
- Tsuyoshi Mashima
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan.
- Medilux Research Center, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan
| | - Masaru Yamanaka
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan.
| | - Atsuki Yoshida
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan.
| | - Naoya Kobayashi
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan.
| | - Yui Kanaoka
- Department of Physics, Nagoya University, Nagoya, 464-8602, Japan
| | - Takayuki Uchihashi
- Department of Physics, Nagoya University, Nagoya, 464-8602, Japan
- Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Higashiyama 5-1, Myodaiji, Okazaki, 444-0864, Japan
| | - Shun Hirota
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, 8916-5 Takayama, Ikoma, Nara 630-0192, Japan.
| |
Collapse
|
29
|
Helenek C, Krzysztoń R, Petreczky J, Wan Y, Cabral M, Coraci D, Balázsi G. Synthetic gene circuit evolution: Insights and opportunities at the mid-scale. Cell Chem Biol 2024; 31:1447-1459. [PMID: 38925113 PMCID: PMC11330362 DOI: 10.1016/j.chembiol.2024.05.018] [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: 02/12/2024] [Revised: 05/07/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Directed evolution focuses on optimizing single genetic components for predefined engineering goals by artificial mutagenesis and selection. In contrast, experimental evolution studies the adaptation of entire genomes in serially propagated cell populations, to provide an experimental basis for evolutionary theory. There is a relatively unexplored gap at the middle ground between these two techniques, to evolve in vivo entire synthetic gene circuits with nontrivial dynamic function instead of single parts or whole genomes. We discuss the requirements for such mid-scale evolution, with hypothetical examples for evolving synthetic gene circuits by appropriate selection and targeted shuffling of a seed set of genetic components in vivo. Implementing similar methods should aid the rapid generation, functionalization, and optimization of synthetic gene circuits in various organisms and environments, accelerating both the development of biomedical and technological applications and the understanding of principles guiding regulatory network evolution.
Collapse
Affiliation(s)
- Christopher Helenek
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rafał Krzysztoń
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Julia Petreczky
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Chemistry, Stony Brook University, Stony Brook, NY 11794, USA
| | - Yiming Wan
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Mariana Cabral
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA
| | - Damiano Coraci
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | - Gábor Balázsi
- The Louis and Beatrice Laufer Center for Physical and Quantitative Biology, Stony Brook University, Stony Brook, NY 11794, USA; Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY 11794, USA; Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
30
|
Trautmann G, Block K, Gutsmann M, Besnard S, Furlan S, Denise P, Volpe P, Blottner D, Salanova M. Increased Homer Activity and NMJ Localization in the Vestibular Lesion het-/- Mouse soleus Muscle. Int J Mol Sci 2024; 25:8577. [PMID: 39201265 PMCID: PMC11354602 DOI: 10.3390/ijms25168577] [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: 06/28/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
We investigated the shuttling of Homer protein isoforms identified in soluble (cytosolic) vs. insoluble (membrane-cytoskeletal) fraction and Homer protein-protein interaction/activation in the deep postural calf soleus (SOL) and non-postural gastrocnemius (GAS) muscles of het-/- mice, i.e., mice with an autosomal recessive variant responsible for a vestibular disorder, in order to further elucidate a) the underlying mechanisms of disrupted vestibular system-derived modulation on skeletal muscle, and b) molecular signaling at respective neuromuscular synapses. Heterozygote mice muscles served as the control (CTR). An increase in Homer cross-linking capacity was present in the SOL muscle of het-/- mice as a compensatory mechanism for the altered vestibule system function. Indeed, in both fractions, different Homer immunoreactive bands were detectable, as were Homer monomers (~43-48 kDa), Homer dimers (~100 kDa), and several other Homer multimer bands (>150 kDA). The het-/- GAS particulate fraction showed no Homer dimers vs. SOL. The het-/- SOL soluble fraction showed a twofold increase (+117%, p ≤ 0.0004) in Homer dimers and multimers. Homer monomers were completely absent from the SOL independent of the animals studied, suggesting muscle-specific changes in Homer monomer vs. dimer expression in the postural SOL vs. the non-postural GAS muscles. A morphological assessment showed an increase (+14%, p ≤ 0.0001) in slow/type-I myofiber cross-sectional area in the SOL of het-/- vs. CTR mice. Homer subcellular immuno-localization at the neuromuscular junction (NMJ) showed an altered expression in the SOL of het-/-mice, whereas only not-significant changes were found for all Homer isoforms, as judged by RT-qPCR analysis. Thus, muscle-specific changes, myofiber properties, and neuromuscular signaling mechanisms share causal relationships, as highlighted by the variable subcellular Homer isoform expression at the instable NMJs of vestibular lesioned het-/- mice.
Collapse
Affiliation(s)
- Gabor Trautmann
- Institute of Integrative Neuroanatomy, Neuromuscular Signaling and System, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (D.B.)
- Center of Space Medicine Berlin, 10115 Berlin, Germany
| | - Katharina Block
- Institute of Integrative Neuroanatomy, Neuromuscular Signaling and System, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (D.B.)
- Center of Space Medicine Berlin, 10115 Berlin, Germany
| | - Martina Gutsmann
- Institute of Integrative Neuroanatomy, Neuromuscular Signaling and System, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (D.B.)
| | - Stéphane Besnard
- UR VERTEX 7480, CHU de Caen, Université de Caen Normandie, 10115 Caen, France;
| | - Sandra Furlan
- C.N.R. Institute of Neuroscience, 14000 Padova, Italy;
| | - Pierre Denise
- COMETE U1075, INSERM, CYCERON, CHU de Caen, Normandie Université, Université de Caen Normandie, 10115 Caen, France;
| | - Pompeo Volpe
- Department of Biomedical Sciences, University of Padova, 14000 Padova, Italy;
| | - Dieter Blottner
- Institute of Integrative Neuroanatomy, Neuromuscular Signaling and System, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (D.B.)
- Center of Space Medicine Berlin, 10115 Berlin, Germany
| | - Michele Salanova
- Institute of Integrative Neuroanatomy, Neuromuscular Signaling and System, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Philippstrasse 12, 10115 Berlin, Germany; (G.T.); (K.B.); (M.G.); (D.B.)
- Center of Space Medicine Berlin, 10115 Berlin, Germany
| |
Collapse
|
31
|
Oskoei V, Mathesh M, Yang W. Enhancing Substrate Channeling with Multi-Enzyme Architectures in Hydrogen-Bonded Organic Frameworks. Chemistry 2024; 30:e202401256. [PMID: 38719746 DOI: 10.1002/chem.202401256] [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: 03/28/2024] [Indexed: 07/03/2024]
Abstract
Hydrogen-bonded organic frameworks (HOF) represent an emerging category of organic structures with high crystallinity and metal-free, which are not commonly observed in alternative porous organic frameworks. These needle-like porous structure can help in stabilizing enzymes and allow transfer of molecules between enzymes participating in cascade reactions for enhanced substrate channelling. Herein, we systematically synthesized and investigated the stability of HOF at extreme conditions followed by one-pot encapsulation of single and bi-enzyme systems. Firstly, we observed HOF to be stable at pH 1 to 14 and at high temperatures (up to 115 °C). Secondly, the encapsulated glucose oxidase enzyme (GOX) showed 80 % and 90 % of its original activity at 70 °C and pH 11, respectively. Thirdly, transient time close to 0 seconds was observed for HOF encapsulated bi-enzyme cascade reaction system demonstrating a 4.25-fold improvement in catalytic activity when compared to free enzymes with enhanced substrate channelling. Our findings showcase a facile system synthesized under ambient conditions to encapsulate and stabilize enzymes at extreme conditions.
Collapse
Affiliation(s)
- Vahide Oskoei
- Centre for Sustainable Products School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, 3216, Australia
| | - Motilal Mathesh
- Centre for Sustainable Products School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, 3216, Australia
| | - Wenrong Yang
- Centre for Sustainable Products School of Life and Environmental Sciences, Deakin University, Geelong, Victoria, 3216, Australia
| |
Collapse
|
32
|
Chen L, Hao M, Huang W, Yu S, Shen H, Yang F, Wang L, Tu H. Integration of multienzyme co-immobilization and biomimetic catalysis in magnetic metal-organic framework nanoflowers for α-amylase detection in fermentation samples. Food Chem 2024; 446:138773. [PMID: 38402762 DOI: 10.1016/j.foodchem.2024.138773] [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: 11/21/2023] [Revised: 01/15/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
Multiple enzymes induce biological cascade catalysis is essential in nature and industrial production. However, the shortcomings of enzymes, including unsatisfactory stability, reusability, and sensitivity in harsh microenvironment, have restricted their broader use. Here, we report a facile method for fabricating a cascade system by combining the benefits of immobilized enzymes and biomimetic catalysis based on magnetic metal-organic framework nanoflowers (mMOFNFs). mMOFNFs prepared through the layered double hydroxide-derived strategy exhibited remarkable peroxidase-like activity and accessible amino interface, enabling it to serve not only as a reliable carrier for α-glucosidase and glucose oxidase fixation, but also as a nanozyme participating in cascade. On this basis, a colorimetric biosensor of excellent sensitivity and selectivity for α-amylase detection was constructed with a wide range (2-225 U L-1), low detection limit (2.48 U L-1), and rapid operation (30 min). This work provides a versatile strategy for establishing multi-enzyme cascade systems and rapid analysis of α-amylase.
Collapse
Affiliation(s)
- Liangqiang Chen
- Kweichow Moutai Group, Renhuai, Guizhou 564501, China; Guizhou Key Laboratory of Microbial Resources Exploration in Fermentation Industry, Kweichow Moutai Group, Zunyi 564501, China
| | - Mengdi Hao
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Wanqiu Huang
- Kweichow Moutai Group, Renhuai, Guizhou 564501, China; Guizhou Key Laboratory of Microbial Resources Exploration in Fermentation Industry, Kweichow Moutai Group, Zunyi 564501, China
| | - Shaoning Yu
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| | - Hao Shen
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Fan Yang
- Kweichow Moutai Group, Renhuai, Guizhou 564501, China; Guizhou Key Laboratory of Microbial Resources Exploration in Fermentation Industry, Kweichow Moutai Group, Zunyi 564501, China
| | - Li Wang
- Kweichow Moutai Group, Renhuai, Guizhou 564501, China; Guizhou Key Laboratory of Microbial Resources Exploration in Fermentation Industry, Kweichow Moutai Group, Zunyi 564501, China
| | - Huabin Tu
- Kweichow Moutai Group, Renhuai, Guizhou 564501, China; Guizhou Key Laboratory of Microbial Resources Exploration in Fermentation Industry, Kweichow Moutai Group, Zunyi 564501, China.
| |
Collapse
|
33
|
Welles RM, Sojitra KA, Garabedian MV, Xia B, Wang W, Guan M, Regy RM, Gallagher ER, Hammer DA, Mittal J, Good MC. Determinants that enable disordered protein assembly into discrete condensed phases. Nat Chem 2024; 16:1062-1072. [PMID: 38316988 PMCID: PMC11929961 DOI: 10.1038/s41557-023-01423-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 12/13/2023] [Indexed: 02/07/2024]
Abstract
Cells harbour numerous mesoscale membraneless compartments that house specific biochemical processes and perform distinct cellular functions. These protein- and RNA-rich bodies are thought to form through multivalent interactions among proteins and nucleic acids, resulting in demixing via liquid-liquid phase separation. Proteins harbouring intrinsically disordered regions (IDRs) predominate in membraneless organelles. However, it is not known whether IDR sequence alone can dictate the formation of distinct condensed phases. We identified a pair of IDRs capable of forming spatially distinct condensates when expressed in cells. When reconstituted in vitro, these model proteins do not co-partition, suggesting condensation specificity is encoded directly in the polypeptide sequences. Through computational modelling and mutagenesis, we identified the amino acids and chain properties governing homotypic and heterotypic interactions that direct selective condensation. These results form the basis of physicochemical principles that may direct subcellular organization of IDRs into specific condensates and reveal an IDR code that can guide construction of orthogonal membraneless compartments.
Collapse
Affiliation(s)
- Rachel M Welles
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kandarp A Sojitra
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Mikael V Garabedian
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Boao Xia
- Bioengineering Graduate Program, Rice University, Houston, TX, USA
| | - Wentao Wang
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
| | - Muyang Guan
- Chemical and Biomolecular Engineering Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
| | - Roshan M Regy
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Elizabeth R Gallagher
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel A Hammer
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
- Chemical and Biomolecular Engineering Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
- Chemical and Biomolecular Engineering Department, University of Pennsylvania, Philadelphia, PA, USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Chemistry, Texas A&M University, College Station, TX, USA.
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
| | - Matthew C Good
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA, USA.
- Cell and Molecular Biology Graduate Program, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
34
|
Rekhi S, Garcia CG, Barai M, Rizuan A, Schuster BS, Kiick KL, Mittal J. Expanding the molecular language of protein liquid-liquid phase separation. Nat Chem 2024; 16:1113-1124. [PMID: 38553587 PMCID: PMC11230844 DOI: 10.1038/s41557-024-01489-x] [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: 03/20/2023] [Accepted: 02/27/2024] [Indexed: 04/07/2024]
Abstract
Understanding the relationship between a polypeptide sequence and its phase separation has important implications for analysing cellular function, treating disease and designing novel biomaterials. Several sequence features have been identified as drivers for protein liquid-liquid phase separation (LLPS), schematized as a 'molecular grammar' for LLPS. Here we further probe how sequence modulates phase separation and the material properties of the resulting condensates, targeting sequence features previously overlooked in the literature. We generate sequence variants of a repeat polypeptide with either no charged residues, high net charge, no glycine residues or devoid of aromatic or arginine residues. All but one of 12 variants exhibited LLPS, albeit to different extents, despite substantial differences in composition. Furthermore, we find that all the condensates formed behaved like viscous fluids, despite large differences in their viscosities. Our results support the model of multiple interactions between diverse residue pairs-not just a handful of residues-working in tandem to drive the phase separation and dynamics of condensates.
Collapse
Affiliation(s)
- Shiv Rekhi
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | | | - Mayur Barai
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Azamat Rizuan
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA
| | - Benjamin S Schuster
- Department of Chemical and Biochemical Engineering, Rutgers, the State University of New Jersey, Piscataway, NJ, USA.
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, Newark, DE, USA.
- Department of Biomedical Engineering, University of Delaware, Newark, DE, USA.
| | - Jeetain Mittal
- Artie McFerrin Department of Chemical Engineering, Texas A&M University, College Station, TX, USA.
- Department of Chemistry, Texas A&M University, College Station, TX, USA.
- Interdisciplinary Graduate Program in Genetics and Genomics, Texas A&M University, College Station, TX, USA.
| |
Collapse
|
35
|
Philbrook A, O'Donnell MP, Grunenkovaite L, Sengupta P. Differential modulation of sensory response dynamics by cilia structure and intraflagellar transport within and across chemosensory neurons. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.16.594529. [PMID: 38798636 PMCID: PMC11118401 DOI: 10.1101/2024.05.16.594529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Sensory neurons contain morphologically diverse primary cilia that are built by intraflagellar transport (IFT) and house sensory signaling molecules. Since both ciliary structural and signaling proteins are trafficked via IFT, it has been challenging to decouple the contributions of IFT and cilia structure to neuronal responses. By acutely inhibiting IFT without altering cilia structure and vice versa , here we describe the differential roles of ciliary trafficking and sensory ending morphology in shaping chemosensory responses in C. elegans. We show that a minimum cilium length but not continuous IFT is necessary for a subset of responses in the ASH nociceptive neurons. In contrast, neither cilia nor continuous IFT are necessary for odorant responses in the AWA olfactory neurons. Instead, continuous IFT differentially modulates response dynamics in AWA. Upon acute inhibition of IFT, cilia-destined odorant receptors are shunted to ectopic branches emanating from the cilia base. Spatial segregation of receptors in these branches from a cilia-restricted regulatory kinase results in odorant desensitization defects, highlighting the importance of precise organization of signaling molecules at sensory endings in regulating response dynamics. We also find that adaptation of AWA responses upon repeated exposure to an odorant is mediated by IFT-driven removal of its cognate receptor, whereas adaptation to a second odorant is regulated via IFT-independent mechanisms. Our results reveal unexpected complexity in the contribution of IFT and cilia organization to the regulation of responses even within a single chemosensory neuron type, and establish a critical role for these processes in the precise modulation of olfactory behaviors.
Collapse
|
36
|
Fu Y, Zhu W, Zhou Y, Su Y, Li Z, Zhang D, Zhang D, Shen J, Liang J. RACK1A promotes hypocotyl elongation by scaffolding light signaling components in Arabidopsis. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2024; 66:956-972. [PMID: 38558526 DOI: 10.1111/jipb.13651] [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: 12/29/2023] [Accepted: 03/07/2024] [Indexed: 04/04/2024]
Abstract
Plants deploy versatile scaffold proteins to intricately modulate complex cell signaling. Among these, RACK1A (Receptors for Activated C Kinase 1A) stands out as a multifaceted scaffold protein functioning as a central integrative hub for diverse signaling pathways. However, the precise mechanisms by which RACK1A orchestrates signal transduction to optimize seedling development remain largely unclear. Here, we demonstrate that RACK1A facilitates hypocotyl elongation by functioning as a flexible platform that connects multiple key components of light signaling pathways. RACK1A interacts with PHYTOCHROME INTERACTING FACTOR (PIF)3, enhances PIF3 binding to the promoter of BBX11 and down-regulates its transcription. Furthermore, RACK1A associates with ELONGATED HYPOCOTYL 5 (HY5) to repress HY5 biochemical activity toward target genes, ultimately contributing to hypocotyl elongation. In darkness, RACK1A is targeted by CONSTITUTIVELY PHOTOMORPHOGENIC (COP)1 upon phosphorylation and subjected to COP1-mediated degradation via the 26 S proteasome system. Our findings provide new insights into how plants utilize scaffold proteins to regulate hypocotyl elongation, ensuring proper skoto- and photo-morphogenic development.
Collapse
Affiliation(s)
- Yajuan Fu
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Wei Zhu
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yeling Zhou
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Yujing Su
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Zhiyong Li
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dayan Zhang
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Dong Zhang
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jinyu Shen
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Jiansheng Liang
- Key Laboratory of Molecular Design for Plant Cell Factory of Guangdong Higher Education Institutes, Department of Biology, School of Life Sciences, Institute of Plant and Food Sciences, Southern University of Science and Technology, Shenzhen, 518055, China
| |
Collapse
|
37
|
Ball NJ, Barnett SFH, Goult BT. Mechanically operated signalling scaffolds. Biochem Soc Trans 2024; 52:517-527. [PMID: 38572868 PMCID: PMC11088903 DOI: 10.1042/bst20221194] [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: 01/03/2024] [Revised: 03/20/2024] [Accepted: 03/25/2024] [Indexed: 04/05/2024]
Abstract
Cellular signalling is a complex process and involves cascades of enzymes that, in response to a specific signal, give rise to exact cellular responses. Signalling scaffold proteins organise components of these signalling pathways in space and time to co-ordinate signalling outputs. In this review we introduce a new class of mechanically operated signalling scaffolds that are built into the cytoskeletal architecture of the cell. These proteins contain force-dependent binary switch domains that integrate chemical and mechanical signals to introduce quantised positional changes to ligands and persistent alterations in cytoskeletal architecture providing mechanomemory capabilities. We focus on the concept of spatial organisation, and how the cell organises signalling molecules at the plasma membrane in response to specific signals to create order and distinct signalling outputs. The dynamic positioning of molecules using binary switches adds an additional layer of complexity to the idea of scaffolding. The switches can spatiotemporally organise enzymes and substrates dynamically, with the introduction of ∼50 nm quantised steps in distance between them as the switch patterns change. Together these different types of signalling scaffolds and the proteins engaging them, provide a way for an ordering of molecules that extends beyond current views of the cell.
Collapse
Affiliation(s)
- Neil J. Ball
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, U.K
| | | | - Benjamin T. Goult
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool L69 7ZB, U.K
| |
Collapse
|
38
|
Shanmugam R, Anderson R, Schiemann AH, Sattlegger E. Evidence that Xrn1 is in complex with Gcn1, and is required for full levels of eIF2α phosphorylation. Biochem J 2024; 481:481-498. [PMID: 38440860 PMCID: PMC11088878 DOI: 10.1042/bcj20220531] [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: 01/19/2023] [Revised: 02/04/2024] [Accepted: 03/05/2024] [Indexed: 03/06/2024]
Abstract
The protein kinase Gcn2 and its effector protein Gcn1 are part of the general amino acid control signalling (GAAC) pathway best known in yeast for its function in maintaining amino acid homeostasis. Under amino acid limitation, Gcn2 becomes activated, subsequently increasing the levels of phosphorylated eIF2α (eIF2α-P). This leads to the increased translation of transcriptional regulators, such as Gcn4 in yeast and ATF4 in mammals, and subsequent re-programming of the cell's gene transcription profile, thereby allowing cells to cope with starvation. Xrn1 is involved in RNA decay, quality control and processing. We found that Xrn1 co-precipitates Gcn1 and Gcn2, suggesting that these three proteins are in the same complex. Growth under starvation conditions was dependent on Xrn1 but not on Xrn1-ribosome association, and this correlated with reduced eIF2α-P levels. Constitutively active Gcn2 leads to a growth defect due to eIF2α-hyperphosphorylation, and we found that this phenotype was independent of Xrn1, suggesting that xrn1 deletion does not enhance eIF2α de-phosphorylation. Our study provides evidence that Xrn1 is required for efficient Gcn2 activation, directly or indirectly. Thus, we have uncovered a potential new link between RNA metabolism and the GAAC.
Collapse
Affiliation(s)
- Renuka Shanmugam
- School of Natural Sciences, Massey University, Auckland, New Zealand
| | - Reuben Anderson
- School of Natural Sciences, Massey University, Auckland, New Zealand
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Anja H. Schiemann
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Evelyn Sattlegger
- School of Natural Sciences, Massey University, Auckland, New Zealand
- School of Natural Sciences, Massey University, Palmerston North, New Zealand
- Maurice Wilkins Centre for Molecular BioDiscovery, Massey University, Palmerston North, New Zealand
| |
Collapse
|
39
|
Pagano L, Simonetti L, Pennacchietti V, Toto A, Malagrinò F, Ivarsson Y, Gianni S. Exploring the short linear motif-mediated protein-protein interactions of CrkL through ProP-PD. Biochem Biophys Res Commun 2024; 703:149658. [PMID: 38387229 DOI: 10.1016/j.bbrc.2024.149658] [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: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
Adaptor proteins play a pivotal role in cellular signaling mediating a multitude of protein-protein interaction critical for cellular homeostasis. Dysregulation of these interactions has been linked to the onset of various cancer pathologies and exploited by viral pathogens during host cell takeover. CrkL is an adaptor protein composed of an N-terminal SH2 domain followed by two SH3 domains that mediate interactions with diverse partners through the recognition of specific binding motifs. In this study, we employed proteomic peptide-phage display (ProP-PD) to comprehensively explore the short linear motif (SLiM)-based interactions of CrkL. Furthermore, we scrutinized how the binding affinity for selected peptides was influenced in the context of the full-length CrkL versus the isolated N-SH3 domain. Importantly, our results provided insights into SLiM-binding sites within previously reported interactors, as well as revealing novel human and viral ligands, expanding our understanding of the interactions mediated by CrkL and highlighting the significance of SLiM-based interactions in mediating adaptor protein function, with implications for cancer and viral pathologies.
Collapse
Affiliation(s)
- L Pagano
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Universita di Roma, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - L Simonetti
- Department of Chemistry - BMC, Husargatan 3, 751 23, Uppsala, Sweden
| | - V Pennacchietti
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Universita di Roma, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - A Toto
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Universita di Roma, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185, Rome, Italy
| | - F Malagrinò
- Dipartimento di Medicina clinica, sanità pubblica, scienze della vita e dell'ambiente, Università dell'Aquila, Piazzale Salvatore Tommasi 1, L'Aquila, Coppito, 67010, Italy
| | - Y Ivarsson
- Department of Chemistry - BMC, Husargatan 3, 751 23, Uppsala, Sweden.
| | - S Gianni
- Dipartimento di Scienze Biochimiche "A. Rossi Fanelli", Sapienza Universita di Roma, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, 00185, Rome, Italy.
| |
Collapse
|
40
|
Feichtner A, Enzler F, Kugler V, Hoppe K, Mair S, Kremser L, Lindner H, Huber RG, Stelzl U, Stefan E, Torres-Quesada O. Phosphorylation of the compartmentalized PKA substrate TAF15 regulates RNA-protein interactions. Cell Mol Life Sci 2024; 81:162. [PMID: 38568213 PMCID: PMC10991009 DOI: 10.1007/s00018-024-05204-4] [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: 08/18/2023] [Revised: 03/08/2024] [Accepted: 03/09/2024] [Indexed: 04/05/2024]
Abstract
Spatiotemporal-controlled second messengers alter molecular interactions of central signaling nodes for ensuring physiological signal transmission. One prototypical second messenger molecule which modulates kinase signal transmission is the cyclic-adenosine monophosphate (cAMP). The main proteinogenic cellular effectors of cAMP are compartmentalized protein kinase A (PKA) complexes. Their cell-type specific compositions precisely coordinate substrate phosphorylation and proper signal propagation which is indispensable for numerous cell-type specific functions. Here we present evidence that TAF15, which is implicated in the etiology of amyotrophic lateral sclerosis, represents a novel nuclear PKA substrate. In cross-linking and immunoprecipitation experiments (iCLIP) we showed that TAF15 phosphorylation alters the binding to target transcripts related to mRNA maturation, splicing and protein-binding related functions. TAF15 appears to be one of multiple PKA substrates that undergo RNA-binding dynamics upon phosphorylation. We observed that the activation of the cAMP-PKA signaling axis caused a change in the composition of a collection of RNA species that interact with TAF15. This observation appears to be a broader principle in the regulation of molecular interactions, as we identified a significant enrichment of RNA-binding proteins within endogenous PKA complexes. We assume that phosphorylation of RNA-binding domains adds another layer of regulation to binary protein-RNAs interactions with consequences to RNA features including binding specificities, localization, abundance and composition.
Collapse
Affiliation(s)
- Andreas Feichtner
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Florian Enzler
- Daniel Swarovski Research Laboratory, Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Innrain 66/66a, 6020, Innsbruck, Austria
| | - Valentina Kugler
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria
| | - Katharina Hoppe
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Sophia Mair
- Department of Cardiac Surgery, Medical University of Innsbruck, Innrain 66/66a, 6020, Innsbruck, Austria
- Vascage, Center of Clinical Stroke Research, 6020, Innsbruck, Austria
| | - Leopold Kremser
- Division of Clinical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Herbert Lindner
- Division of Clinical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria
| | - Roland G Huber
- Bioinformatics Institute, Agency for Science Technology and Research, Singapore, 138671, Singapore
| | - Ulrich Stelzl
- Institute of Pharmaceutical Sciences, University of Graz, Schubertstrasse 1, 8010, Graz, Austria
| | - Eduard Stefan
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria.
- Institute of Molecular Biology and Center for Molecular Biosciences, University of Innsbruck, Technikerstrasse 25, 6020, Innsbruck, Austria.
| | - Omar Torres-Quesada
- Tyrolean Cancer Research Institute (TKFI), Innrain 66, 6020, Innsbruck, Austria.
- Division of Medical Biochemistry, Biocenter, Medical University of Innsbruck, Innrain 80/82, 6020, Innsbruck, Austria.
| |
Collapse
|
41
|
Du L, Guan Z, Liu Y, Hu D, Gao J, Sun C. Scaffold protein BTB/TAZ domain-containing genes (CmBTs) play a negative role in root development of chrysanthemum. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2024; 341:111997. [PMID: 38280641 DOI: 10.1016/j.plantsci.2024.111997] [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: 10/24/2023] [Revised: 12/05/2023] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Scaffold proteins, which are known as hubs controlling information flow in cells, can function in a diverse array of biological processes in plants. The BTB/TAZ domain-containing scaffold proteins are associated with multiple signaling pathways in plants. However, there have been few studies of the roles of BT scaffold proteins in chrysanthemum to date. In this study, four CmBT genes named as CmBT1, CmBT1-LIKE1 (CmBT1L1), CmBT1-LIKE2 (CmBT1L2), and CmBT5 were cloned based our previous RNA-seq database. The four CmBT genes showed distinctive expression patterns both in different tissues and in response to different stimuli, such as light, sugar, nitrate and auxin. Knockdown of the four CmBTs facilitated the development of adventitious roots and root hair in chrysanthemum. Transcriptome sequencing analysis revealed thousands of differentially expressed genes after knockdown of the four CmBT genes. Moreover, functional annotation suggested that CmBTs play a tethering role as scaffold proteins. Our findings reveal that CmBTs can negatively regulate root development of chrysanthemum by mediating nitrate assimilation, amino acid biosynthesis, and auxin and jasmonic acid (JA) signaling pathways. This study provides new insights into the role of CmBTs in root development of chrysanthemum.
Collapse
Affiliation(s)
- Lianda Du
- College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, Shandong 271018, China
| | - Zhangji Guan
- College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, Shandong 271018, China
| | - Yanhong Liu
- College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, Shandong 271018, China
| | - Dagang Hu
- College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, Shandong 271018, China
| | - Junping Gao
- Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, Department of Ornamental Horticulture, China Agricultural University, Beijing, China
| | - Cuihui Sun
- Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, Department of Ornamental Horticulture, China Agricultural University, Beijing, China; College of Horticulture Science and Engineering, Shandong Agricultural University, Tai-An, Shandong 271018, China.
| |
Collapse
|
42
|
Bock A, Irannejad R, Scott JD. cAMP signaling: a remarkably regional affair. Trends Biochem Sci 2024; 49:305-317. [PMID: 38310024 PMCID: PMC11175624 DOI: 10.1016/j.tibs.2024.01.004] [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: 11/02/2023] [Revised: 12/22/2023] [Accepted: 01/10/2024] [Indexed: 02/05/2024]
Abstract
Louis Pasteur once famously said 'in the fields of observation chance favors only the prepared mind'. Much of chance is being in the right place at the right time. This is particularly true in the crowded molecular environment of the cell where being in the right place is often more important than timing. Although Brownian motion argues that enzymes will eventually bump into substrates, this probability is greatly enhanced if both molecules reside in the same subcellular compartment. However, activation of cell signaling enzymes often requires the transmission of chemical signals from extracellular stimuli to intracellular sites of action. This review highlights new developments in our understanding of cAMP generation and the 3D utilization of this second messenger inside cells.
Collapse
Affiliation(s)
- Andreas Bock
- Rudolf Boehm Institute of Pharmacology and Toxicology, Medical Faculty, Leipzig University, 04107 Leipzig, Germany.
| | - Roshanak Irannejad
- Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA; Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - John D Scott
- Department of Pharmacology, University of Washington Medical Center, Seattle, WA 98195, USA.
| |
Collapse
|
43
|
Herr LA, Fiala GJ, Sagar, Schaffer AM, Hummel JF, Zintchenko M, Raute K, Velasco Cárdenas RMH, Heizmann B, Ebert K, Fehrenbach K, Janowska I, Chan S, Tanriver Y, Minguet S, Schamel WW. Kidins220 and Aiolos promote thymic iNKT cell development by reducing TCR signals. SCIENCE ADVANCES 2024; 10:eadj2802. [PMID: 38489359 PMCID: PMC10942104 DOI: 10.1126/sciadv.adj2802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 02/09/2024] [Indexed: 03/17/2024]
Abstract
Development of T cells is controlled by the signal strength of the TCR. The scaffold protein kinase D-interacting substrate of 220 kilodalton (Kidins220) binds to the TCR; however, its role in T cell development was unknown. Here, we show that T cell-specific Kidins220 knockout (T-KO) mice have strongly reduced invariant natural killer T (iNKT) cell numbers and modest decreases in conventional T cells. Enhanced apoptosis due to increased TCR signaling in T-KO iNKT thymocytes of developmental stages 2 and 3 shows that Kidins220 down-regulates TCR signaling at these stages. scRNA-seq indicated that the transcription factor Aiolos is down-regulated in Kidins220-deficient iNKT cells. Analysis of an Aiolos KO demonstrated that Aiolos is a downstream effector of Kidins220 during iNKT cell development. In the periphery, T-KO iNKT cells show reduced TCR signaling upon stimulation with α-galactosylceramide, suggesting that Kidins220 promotes TCR signaling in peripheral iNKT cells. Thus, Kidins220 reduces or promotes signaling dependent on the iNKT cell developmental stage.
Collapse
Affiliation(s)
- Laurenz A. Herr
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Gina J. Fiala
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Anna-Maria Schaffer
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Jonas F. Hummel
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Marina Zintchenko
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Katrin Raute
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Rubí M.-H. Velasco Cárdenas
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Beate Heizmann
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Karolina Ebert
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
| | - Kerstin Fehrenbach
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Iga Janowska
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
| | - Susan Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Yakup Tanriver
- Institute of Medical Microbiology and Hygiene, Medical Center, University of Freiburg, Germany
- Department of Medicine IV: Nephrology and Primary Care, Medical Center, University of Freiburg, Freiburg, Germany
| | - Susana Minguet
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| | - Wolfgang W. Schamel
- Signaling Research Centers BIOSS and CIBSS; University of Freiburg, Freiburg, Germany
- Department of Immunology, Faculty of Biology, University of Freiburg, Freiburg, Germany
- Centre for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), University of Freiburg, Freiburg, Germany
| |
Collapse
|
44
|
Bacsa B, Hopl V, Derler I. Synthetic Biology Meets Ca 2+ Release-Activated Ca 2+ Channel-Dependent Immunomodulation. Cells 2024; 13:468. [PMID: 38534312 PMCID: PMC10968988 DOI: 10.3390/cells13060468] [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: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/05/2024] [Indexed: 03/28/2024] Open
Abstract
Many essential biological processes are triggered by the proximity of molecules. Meanwhile, diverse approaches in synthetic biology, such as new biological parts or engineered cells, have opened up avenues to precisely control the proximity of molecules and eventually downstream signaling processes. This also applies to a main Ca2+ entry pathway into the cell, the so-called Ca2+ release-activated Ca2+ (CRAC) channel. CRAC channels are among other channels are essential in the immune response and are activated by receptor-ligand binding at the cell membrane. The latter initiates a signaling cascade within the cell, which finally triggers the coupling of the two key molecular components of the CRAC channel, namely the stromal interaction molecule, STIM, in the ER membrane and the plasma membrane Ca2+ ion channel, Orai. Ca2+ entry, established via STIM/Orai coupling, is essential for various immune cell functions, including cytokine release, proliferation, and cytotoxicity. In this review, we summarize the tools of synthetic biology that have been used so far to achieve precise control over the CRAC channel pathway and thus over downstream signaling events related to the immune response.
Collapse
Affiliation(s)
- Bernadett Bacsa
- Division of Medical Physics und Biophysics, Medical University of Graz, A-8010 Graz, Austria;
| | - Valentina Hopl
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| | - Isabella Derler
- Institute of Biophysics, JKU Life Science Center, Johannes Kepler University Linz, A-4020 Linz, Austria;
| |
Collapse
|
45
|
Ye C, Zhou T, Deng Y, Wu S, Zeng T, Yang J, Shi YS, Yin Y, Li G. Enhanced performance of enzymes confined in biocatalytic hydrogen-bonded organic frameworks for sensing of glutamate in the central nervous system. Biosens Bioelectron 2024; 247:115963. [PMID: 38147717 DOI: 10.1016/j.bios.2023.115963] [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: 10/22/2023] [Revised: 12/07/2023] [Accepted: 12/20/2023] [Indexed: 12/28/2023]
Abstract
Glutamate (Glu) is a key excitatory neurotransmitter associated with various neurological disorders in the central nervous system, so its measurement is vital to both basic research and biomedical application. In this work, we propose the first example of using biocatalytic hydrogen-bonded organic frameworks (HOFs) as the hosting matrix to encapsulate glutamate oxidase (GLOD) via a de novo approach, fabricating a cascaded-enzyme nanoreactor for Glu biosensing. In this design, the ferriporphyrin ligands can assemble to form Fe-HOFs with high catalase-like activity, while offering a scaffold for the in-situ immobilization of GLOD. Moreover, the formed GLOD@Fe-HOFs are favorable for the efficient diffusion of Glu into the active sites of GLOD via the porous channels, accelerating the cascade reaction with neighboring Fe-HOFs. Consequently, the constructed nanoreactor can offer superior activity and operational stability in the catalytic cascade for Glu biosensing. More importantly, rapid and selective detection can be achieved in the cerebrospinal fluid (CSF) collected from mice in a low sample consumption. Therefore, the successful fabrication of enzyme@HOFs may offer promise to develop high-performance biosensor for further biomedical applications.
Collapse
Affiliation(s)
- Chang Ye
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing, 210032, PR China
| | - Tianci Zhou
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Shuai Wu
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Tianyu Zeng
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China
| | - Jie Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China
| | - Yun Stone Shi
- State Key Laboratory of Pharmaceutical Biotechnology and MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Medical School, Nanjing University, Nanjing, 210032, PR China.
| | - Yongmei Yin
- Women & Children Central Laboratory, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China; Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, PR China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing, 210023, PR China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
46
|
Liu S, He Y, Zhang W, Fu T, Wang L, Zhang Y, Xu Y, Sun H, Zhao H. Self-Cascade Ce-MOF-818 Nanozyme for Sequential Hydrolysis and Oxidation. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306522. [PMID: 37884468 DOI: 10.1002/smll.202306522] [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: 08/01/2023] [Revised: 09/24/2023] [Indexed: 10/28/2023]
Abstract
Mimicking efficient biocatalytic cascades using nanozymes has gained enormous attention in catalytic chemistry, but it remains challenging to develop a nanozyme-based cascade system to sequentially perform the desired reactions. Particularly, the integration of sequential hydrolysis and oxidation reactions into nanozyme-based cascade systems has not yet been achieved, despite their significant roles in various domains. Herein, a self-cascade Ce-MOF-818 nanozyme for sequential hydrolysis and oxidation reactions is developed. Ce-MOF-818 is the first Ce(IV)-based heterometallic metal-organic framework constructed through the coordination of Ce and Cu to distinct groups. It is successfully synthesized using an improved solvothermal method, overcoming the challenge posed by the significant difference in the binding speeds of Ce and Cu to ligands. With excellent organophosphate hydrolase-like (Km = 42.3 µM, Kcat = 0.0208 min-1 ) and catechol oxidase-like (Km = 2589 µM, Kcat = 1.25 s-1 ) activities attributed to its bimetallic active centers, Ce-MOF-818 serves as a promising self-cascade platform for sequential hydrolysis and oxidation. Notably, its catalytic efficiency surpasses that of physically mixed nanozymes by approximately fourfold, owning to the close integration of active sites. The developed hydrolysis-oxidation self-cascade nanozyme has promising potential applications in catalytic chemistry and provides valuable insights into the rational design of nanozyme-based cascade systems.
Collapse
Affiliation(s)
- Sheng Liu
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Yang He
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Weikun Zhang
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Tao Fu
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Liangjie Wang
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Yixin Zhang
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Department of Chemistry, Imperial College London, London, SW7 2AZ, UK
| | - Yi Xu
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
| | - Hao Sun
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Huazhang Zhao
- The Key Laboratory of Water and Sediment Sciences (Ministry of Education), College of Environmental Sciences and Engineering, Peking University, Beijing, 100871, China
- Shanxi Laboratory for Yellow River, College of Environmental & Resource Sciences, Shanxi University, Taiyuan, 030006, China
| |
Collapse
|
47
|
Delesalle C, Vert G, Fujita S. The cell surface is the place to be for brassinosteroid perception and responses. NATURE PLANTS 2024; 10:206-218. [PMID: 38388723 DOI: 10.1038/s41477-024-01621-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/05/2024] [Indexed: 02/24/2024]
Abstract
Adjusting the microenvironment around the cell surface is critical to responding to external cues or endogenous signals and to maintaining cell activities. In plant cells, the plasma membrane is covered by the cell wall and scaffolded with cytoskeletal networks, which altogether compose the cell surface. It has long been known that these structures mutually interact, but the mechanisms that integrate the whole system are still obscure. Here we spotlight the brassinosteroid (BR) plant hormone receptor BRASSINOSTEROID INSENSITIVE1 (BRI1) since it represents an outstanding model for understanding cell surface signalling and regulation. We summarize how BRI1 activity and dynamics are controlled by plasma membrane components and their associated factors to fine-tune signalling. The downstream signals, in turn, manipulate cell surface structures by transcriptional and post-translational mechanisms. Moreover, the changes in these architectures impact BR signalling, resulting in a feedback loop formation. This Review discusses how BRI1 and BR signalling function as central hubs to integrate cell surface regulation.
Collapse
Affiliation(s)
- Charlotte Delesalle
- Plant Science Research Laboratory (LRSV), UMR5546 CNRS/Université Toulouse 3, Auzeville-Tolosane, France
| | - Grégory Vert
- Plant Science Research Laboratory (LRSV), UMR5546 CNRS/Université Toulouse 3, Auzeville-Tolosane, France
| | - Satoshi Fujita
- Plant Science Research Laboratory (LRSV), UMR5546 CNRS/Université Toulouse 3, Auzeville-Tolosane, France.
| |
Collapse
|
48
|
Sun C, Li M, Wang F. Programming and monitoring surface-confined DNA computing. Bioorg Chem 2024; 143:107080. [PMID: 38183684 DOI: 10.1016/j.bioorg.2023.107080] [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: 10/31/2023] [Revised: 12/19/2023] [Accepted: 12/28/2023] [Indexed: 01/08/2024]
Abstract
DNA-based molecular computing has evolved to encompass a diverse range of functions, demonstrating substantial promise for both highly parallel computing and various biomedical applications. Recent advances in DNA computing systems based on surface reactions have demonstrated improved levels of specificity and computational speed compared to their solution-based counterparts that depend on three-dimensional molecular collisions. Herein, computational biomolecular interactions confined by various surfaces such as DNA origamis, nanoparticles, lipid membranes and chips are systematically reviewed, along with their manipulation methodologies. Monitoring techniques and applications for these surface-based computing systems are also described. The advantages and challenges of surface-confined DNA computing are discussed.
Collapse
Affiliation(s)
- Chenyun Sun
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Mingqiang Li
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Fei Wang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules and National Center for Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China.
| |
Collapse
|
49
|
Sieme D, Engelke M, Rezaei-Ghaleh N, Becker S, Wienands J, Griesinger C. Autoinhibition in the Signal Transducer CIN85 Modulates B Cell Activation. J Am Chem Soc 2024; 146:399-409. [PMID: 38111344 PMCID: PMC10786037 DOI: 10.1021/jacs.3c09586] [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: 09/01/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 12/20/2023]
Abstract
Signal transduction by the ligated B cell antigen receptor (BCR) depends on the preorganization of its intracellular components, such as the effector proteins SLP65 and CIN85 within phase-separated condensates. These liquid-like condensates are based on the interaction between three Src homology 3 (SH3) domains and the corresponding proline-rich recognition motifs (PRM) in CIN85 and SLP65, respectively. However, detailed information on the protein conformation and how it impacts the capability of SLP65/CIN85 condensates to orchestrate BCR signal transduction is still lacking. This study identifies a hitherto unknown intramolecular SH3:PRM interaction between the C-terminal SH3 domain (SH3C) of CIN85 and an adjacent PRM. We used high-resolution nuclear magnetic resonance (NMR) experiments to study the flexible linker region containing the PRM and determined the extent of the interaction in multidomain constructs of the protein. Moreover, we observed that the phosphorylation of a serine residue located in the immediate vicinity of the PRM regulates this intramolecular interaction. This allows for a dynamic modulation of CIN85's valency toward SLP65. B cell culture experiments further revealed that the PRM/SH3C interaction is crucial for maintaining the physiological level of SLP65/CIN85 condensate formation, activation-induced membrane recruitment of CIN85, and subsequent mobilization of Ca2+. Our findings therefore suggest that the intramolecular interaction with the adjacent disordered linker is effective in modulating CIN85's valency both in vitro and in vivo. This therefore constitutes a powerful way for the modulation of SLP65/CIN85 condensate formation and subsequent B cell signaling processes within the cell.
Collapse
Affiliation(s)
- Daniel Sieme
- Department
for NMR-based Structural Biology, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Michael Engelke
- Institute
for Cellular and Molecular Immunology, Georg-August
University Göttingen, Humboldtallee 34, 37073 Göttingen, Germany
| | - Nasrollah Rezaei-Ghaleh
- Institute
of Physical Biology, Heinrich Heine University
Düsseldorf, Universitätsstraße
1, 40225 Düsseldorf, Germany
- Institute
of Biological Information Processing, IBI-7: Structural Biochemistry, Forschungszentrum Jülich, Wilhelm-Johnen-Straße, 52428 Jülich, Germany
| | - Stefan Becker
- Department
for NMR-based Structural Biology, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| | - Jürgen Wienands
- Institute
for Cellular and Molecular Immunology, Georg-August
University Göttingen, Humboldtallee 34, 37073 Göttingen, Germany
| | - Christian Griesinger
- Department
for NMR-based Structural Biology, Max Planck
Institute for Multidisciplinary Sciences, Am Fassberg 11, 37077 Göttingen, Germany
| |
Collapse
|
50
|
Kröll S, Burgahn T, Rabe KS, Franzreb M, Niemeyer CM. Nano- and Microscale Confinements in DNA-Scaffolded Enzyme Cascade Reactions. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2304578. [PMID: 37732702 DOI: 10.1002/smll.202304578] [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: 05/31/2023] [Revised: 08/24/2023] [Indexed: 09/22/2023]
Abstract
Artificial reconstruction of naturally evolved principles, such as compartmentalization and cascading of multienzyme complexes, offers enormous potential for the development of biocatalytic materials and processes. Due to their unique addressability at the nanoscale, DNA origami nanostructures (DON) have proven to be an exceptionally powerful tool for studying the fundamental processes in biocatalytic cascades. To systematically investigate the diffusion-reaction network of (co)substrate transfer in enzyme cascades, a model system of stereoselective ketoreductase (KRED) with cofactor regenerating enzyme is assembled in different spatial arrangements on DNA nanostructures and is located in the sphere of microbeads (MB) as a spatially confining nano- and microenvironment, respectively. The results, obtained through the use of highly sensitive analytical methods, Western blot-based quantification of the enzymes, and mass spectrometric (MS) product detection, along with theoretical modeling, provide strong evidence for the presence of two interacting compartments, the diffusion layers around the microbead and the DNA scaffold, which influence the catalytic efficiency of the cascade. It is shown that the microscale compartment exerts a strong influence on the productivity of the cascade, whereas the nanoscale arrangement of enzymes has no influence but can be modulated by the insertion of a diffusion barrier.
Collapse
Affiliation(s)
- Sandra Kröll
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Hermann-von-Helmholtz-Platz 1, D-76344, Eggenstein-Leopoldshafen, Germany
| | - Teresa Burgahn
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Hermann-von-Helmholtz-Platz 1, D-76344, Eggenstein-Leopoldshafen, Germany
| | - Kersten S Rabe
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Hermann-von-Helmholtz-Platz 1, D-76344, Eggenstein-Leopoldshafen, Germany
| | - Matthias Franzreb
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces (IFG), Hermann-von-Helmholtz-Platz 1, D-76344, Eggenstein-Leopoldshafen, Germany
| | - Christof M Niemeyer
- Karlsruhe Institute of Technology (KIT), Institute for Biological Interfaces (IBG 1), Hermann-von-Helmholtz-Platz 1, D-76344, Eggenstein-Leopoldshafen, Germany
| |
Collapse
|