602,219 results on '"Protein Binding"'
Search Results
2. Tridentate chelating ligand coordinated Co(III) complexes: Synthesis, crystal structure, DFT/TD-DFT calculation, studies of interaction with proteins and molecular docking
- Author
-
Paul, Aparup, Puschmann, Horst, and Manna, Subal Chandra
- Published
- 2025
- Full Text
- View/download PDF
3. High-throughput screening of protein interactions with per- and polyfluoroalkyl substances (PFAS) used in photolithography
- Author
-
Cao, Yuexin and Ng, Carla A.
- Published
- 2025
- Full Text
- View/download PDF
4. Implementation of a novel method for the determination of plasma protein binding of highly bound compounds using the equilibrium dialysis method coupled with extraction to the organic phase and its comparison to known methods
- Author
-
Gogola, Dawid, Novak Ratajczak, Sanja, Gabor-Worwa, Ewelina, Kowal-Chwast, Anna, Gaud, Nilesh, Latacz, Gniewomir, Brzózka, Krzysztof, and Kuś, Kamil
- Published
- 2025
- Full Text
- View/download PDF
5. Population pharmacokinetics and dosing simulations of temocillin in liver-transplanted paediatric patients: a prospective, open-label, non-randomized study
- Author
-
Ngougni Pokem, Perrin, Stéphenne, Xavier, Liu, Xin, Parker, Suzanne L., Van der Linden, Dimitri, Godet, Marie-Laura, Wijnant, Gert-Jan, Chatzis, Olga, Houtekie, Laurent, Haenecour, Astrid, Sokal, Etienne, Roberts, Jason A., Elens, Laure, and Van Bambeke, Françoise
- Published
- 2024
- Full Text
- View/download PDF
6. Unveiling the molecular interactions between alkyl imidazolium ionic liquids and human serum albumin: Implications for toxicological significance
- Author
-
Meng, Shizhen, Yu, Qingqing, Li, Ming, Liu, Xin, Zhao, Xiaole, Wu, Kejia, Wang, Qiao, Liu, Yan, Wu, Yongning, and Gong, Zhiyong
- Published
- 2023
- Full Text
- View/download PDF
7. Profiling in-vitro release of verteporfin from VISUDYNE® liposomal formulation and investigating verteporfin binding to human serum albumin
- Author
-
Siriwardane, Dumindika A., Jiang, Wenlei, and Mudalige, Thilak
- Published
- 2023
- Full Text
- View/download PDF
8. Role of protein–protein interactions on organization and dynamics of a model chromatin.
- Author
-
Swain, Pinaki, Choubey, Sandeep, and Vemparala, Satyavani
- Subjects
- *
CARRIER proteins , *CHROMATIN , *PROTEIN binding , *PROTEIN-protein interactions , *CHROMOSOMES - Abstract
The three-dimensional organization of chromatin is influenced by chromatin-binding proteins through both specific and non-specific interactions. However, the roles of chromatin sequence and the interactions between binding proteins in shaping chromatin structure remain elusive. By employing a simple polymer-based model of chromatin that explicitly considers sequence-dependent protein binding and protein–protein interactions, we elucidate a mechanism for chromatin organization. We find that tuning protein–protein interactions and protein concentration is sufficient to either promote or inhibit chromatin compartmentalization. Moreover, chromatin sequence and protein–protein attraction strongly affect the structural and dynamic exponents that describe the spatiotemporal organization of chromatin. Strikingly, our model's predictions for the exponents governing chromatin structure and dynamics successfully capture experimental observations, in sharp contrast to previous chromatin models. Overall, our findings have the potential to reinterpret data obtained from various chromosome conformation capture technologies, laying the groundwork for advancing our understanding of chromatin organization. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Surprising features of nuclear receptor interaction networks revealed by live-cell single-molecule imaging.
- Author
-
Dahal, Liza, Graham, Thomas, Dailey, Gina, Heckert, Alec, Tjian, Robert, and Darzacq, Xavier
- Subjects
chromosomes ,gene expression ,gene regulatory networks ,human ,molecular biophysics ,nuclear receptors ,protein-protein interactions ,single-molecule tracking ,structural biology ,transcription factors ,Humans ,Single Molecule Imaging ,Retinoid X Receptors ,Chromatin ,Receptors ,Retinoic Acid ,Protein Binding ,Protein Multimerization ,Receptors ,Cytoplasmic and Nuclear ,Cell Line ,Tumor - Abstract
Type II nuclear receptors (T2NRs) require heterodimerization with a common partner, the retinoid X receptor (RXR), to bind cognate DNA recognition sites in chromatin. Based on previous biochemical and overexpression studies, binding of T2NRs to chromatin is proposed to be regulated by competition for a limiting pool of the core RXR subunit. However, this mechanism has not yet been tested for endogenous proteins in live cells. Using single-molecule tracking (SMT) and proximity-assisted photoactivation (PAPA), we monitored interactions between endogenously tagged RXR and retinoic acid receptor (RAR) in live cells. Unexpectedly, we find that higher expression of RAR, but not RXR, increases heterodimerization and chromatin binding in U2OS cells. This surprising finding indicates the limiting factor is not RXR but likely its cadre of obligate dimer binding partners. SMT and PAPA thus provide a direct way to probe which components are functionally limiting within a complex TF interaction network providing new insights into mechanisms of gene regulation in vivo with implications for drug development targeting nuclear receptors.
- Published
- 2025
10. Structural insights into GrpEL1-mediated nucleotide and substrate release of human mitochondrial Hsp70.
- Author
-
Morizono, Marc, McGuire, Kelly, Birouty, Natalie, and Herzik, Mark
- Subjects
HSP70 Heat-Shock Proteins ,Humans ,Cryoelectron Microscopy ,Mitochondria ,Molecular Dynamics Simulation ,Mitochondrial Proteins ,Nucleotides ,Protein Binding - Abstract
Maintenance of protein homeostasis is necessary for cell viability and depends on a complex network of chaperones and co-chaperones, including the heat-shock protein 70 (Hsp70) system. In human mitochondria, mitochondrial Hsp70 (mortalin) and the nucleotide exchange factor (GrpEL1) work synergistically to stabilize proteins, assemble protein complexes, and facilitate protein import. However, our understanding of the molecular mechanisms guiding these processes is hampered by limited structural information. To elucidate these mechanistic details, we used cryoEM to determine structures of full-length human mortalin-GrpEL1 complexes in previously unobserved states. Our structures and molecular dynamics simulations allow us to delineate specific roles for mortalin-GrpEL1 interfaces and to identify steps in GrpEL1-mediated nucleotide and substrate release by mortalin. Subsequent analyses reveal conserved mechanisms across bacteria and mammals and facilitate a complete understanding of sequential nucleotide and substrate release for the Hsp70 chaperone system.
- Published
- 2024
11. Identifying Strong Neoantigen MHC-I/II Binding Candidates for Targeted Immunotherapy with SINE.
- Author
-
Bendik, Joseph, Castro, Andrea, Califano, Joseph, Carter, Hannah, and Guo, Theresa
- Subjects
alternative splicing ,head and neck cancer ,melanoma ,neoantigen ,software ,targeted immunotherapy response ,Humans ,Antigens ,Neoplasm ,Immunotherapy ,Histocompatibility Antigens Class I ,Head and Neck Neoplasms ,Histocompatibility Antigens Class II ,Melanoma ,Protein Binding ,Squamous Cell Carcinoma of Head and Neck ,Peptides - Abstract
The discovery of tumor-derived neoantigens which elicit an immune response through major histocompatibility complex (MHC-I/II) binding has led to significant advancements in immunotherapy. While many neoantigens have been discovered through the identification of non-synonymous mutations, the rate of these is low in some cancers, including head and neck squamous cell carcinoma. Therefore, the identification of neoantigens through additional means, such as aberrant splicing, is necessary. To achieve this, we developed the splice isoform neoantigen evaluator (SINE) pipeline. Our tool documents peptides present on spliced or inserted genomic regions of interest using Patient Harmonic-mean Best Rank scores, calculating the MHC-I/II binding affinity across the complete human leukocyte antigen landscape. Here, we found 125 potentially immunogenic events and 9 principal binders in a cohort of head and neck cancer patients where the corresponding wild-type peptides display no MHC-I/II affinity. Further, in a melanoma cohort of patients treated with anti-PD1 therapy, the expression of immunogenic splicing events identified by SINE predicted response, potentially indicating the existence of immune editing in these tumors. Overall, we demonstrate SINEs ability to identify clinically relevant immunogenic neojunctions, thus acting as a useful tool for researchers seeking to understand the neoantigen landscape from aberrant splicing in cancer.
- Published
- 2024
12. Legionella uses host Rab GTPases and BAP31 to create a unique ER niche.
- Author
-
Chadha, Attinder, Yanai, Yu, Oide, Hiromu, Wakana, Yuichi, Inoue, Hiroki, Saha, Saradindu, Paul, Manish, Tagaya, Mitsuo, Arasaki, Kohei, and Mukherjee, Shaeri
- Subjects
CP: Cell biology ,CP: Microbiology ,Legionella ,Rab GTPase ,bacteria ,endoplasmic reticulum ,pathogen ,rab GTP-Binding Proteins ,Endoplasmic Reticulum ,Humans ,Legionella pneumophila ,Vacuoles ,Host-Pathogen Interactions ,Bacterial Proteins ,HeLa Cells ,Protein Binding ,Animals ,Membrane Proteins - Abstract
The bacterium Legionella pneumophila secretes numerous effector proteins that manipulate endoplasmic reticulum (ER)-derived vesicles to form the Legionella-containing vacuole (LCV). Despite extensive studies, whether the LCV membrane is separate from or connected to the host ER network remains unclear. Here, we show that the smooth ER (sER) is closely associated with the LCV early in infection. Remarkably, Legionella forms a distinct rough ER (rER) niche at later stages, disconnected from the host ER network. We discover that host small GTPases Rab10 and Rab4 and an ER protein, BAP31, play crucial roles in transitioning the LCV from an sER to an rER. Additionally, we have identified a Legionella effector, Lpg1152, that binds to BAP31. Interestingly, the optimal growth of Legionella is dependent on both BAP31 and Lpg1152. These findings detail the complex interplay between host and pathogen in transforming the LCV membrane from a host-associated sER to a distinct rER.
- Published
- 2024
13. Role of the αC-β4 loop in protein kinase structure and dynamics.
- Author
-
Wu, Jian, Jonniya, Nisha, Hirakis, Sophia, Olivieri, Cristina, Veglia, Gianluigi, Kornev, Alexandr, and Taylor, Susan
- Subjects
F100A mutant ,local spatial pattern ,molecular biophysics ,molecular dynamics simulations ,none ,protein kinase A ,structural biology ,αC-β4 loop ,Protein Kinases ,Adenosine Triphosphate ,Protein Structure ,Secondary ,Models ,Molecular ,Protein Conformation ,Protein Binding ,Humans ,Mutation - Abstract
Although the αC-β4 loop is a stable feature of all protein kinases, the importance of this motif as a conserved element of secondary structure, as well as its links to the hydrophobic architecture of the kinase core, has been underappreciated. We first review the motif and then describe how it is linked to the hydrophobic spine architecture of the kinase core, which we first discovered using a computational tool, local spatial Pattern (LSP) alignment. Based on NMR predictions that a mutation in this motif abolishes the synergistic high-affinity binding of ATP and a pseudo substrate inhibitor, we used LSP to interrogate the F100A mutant. This comparison highlights the importance of the αC-β4 loop and key residues at the interface between the N- and C-lobes. In addition, we delved more deeply into the structure of the apo C-subunit, which lacks ATP. While apo C-subunit showed no significant changes in backbone dynamics of the αC-β4 loop, we found significant differences in the side chain dynamics of K105. The LSP analysis suggests disruption of communication between the N- and C-lobes in the F100A mutant, which would be consistent with the structural changes predicted by the NMR spectroscopy.
- Published
- 2024
14. Synthesis and structural characterization of the heavy tricysteinylpnictines, models of protein-bound As( iii ), Sb( iii ), and Bi( iii )
- Author
-
Hollow, Sophia E and Johnstone, Timothy C
- Subjects
Inorganic Chemistry ,Chemical Sciences ,Antimony ,Models ,Molecular ,Bismuth ,Cysteine ,Coordination Complexes ,Proteins ,Molecular Structure ,Crystallography ,X-Ray ,Protein Binding ,Theoretical and Computational Chemistry ,Other Chemical Sciences ,Inorganic & Nuclear Chemistry ,Inorganic chemistry - Abstract
The heavier group 15 elements As, Sb, and Bi are more restricted in their biochemistry than the nearly ubiquitous lighter congeners N and P, but organisms do encounter compounds of these elements as environmental toxins, starting materials for secondary metabolite biosynthesis, substrates for primary metabolism, or exogenously applied medicines. Under many physiological conditions, these compounds are transformed into pnictogen(III) species, the soft Lewis acidic character of which leads them to interact strongly with biologically relevant soft Lewis bases such as small-molecule thiols or cysteine residues of proteins and peptides. The archetypal complexes As(Cys)3, Sb(Cys)3, and Bi(Cys)3 have been studied in the past but a lack of detailed information about their molecular structures has hampered the analysis of protein structures featuring As(III), Sb(III), and Bi(III) bound to cysteine thiolate residues. In many cases, the formation of such protein adducts is proposed to play a key role in the mechanism of action of inorganic drugs that feature these elements. Here, we refine synthetic strategies to access As(Cys)3, Sb(Cys)3, and Bi(Cys)3, describe their crystal structures, analyze structural trends across the series and across Pn(SR)3 compounds deposited in the Cambridge Structural Database, and compare their features to the structures of proteins with these centers bound to Cys3 motifs. Significant differences were noted for many of the protein structures.
- Published
- 2024
15. Mapping protein–DNA interactions with DiMeLo-seq
- Author
-
Maslan, Annie, Altemose, Nicolas, Marcus, Jeremy, Mishra, Reet, Brennan, Lucy D, Sundararajan, Kousik, Karpen, Gary, Straight, Aaron F, and Streets, Aaron
- Subjects
Biological Sciences ,Bioinformatics and Computational Biology ,Genetics ,Biotechnology ,Human Genome ,Nanotechnology ,Bioengineering ,2.1 Biological and endogenous factors ,Generic health relevance ,Humans ,DNA ,DNA Methylation ,High-Throughput Nucleotide Sequencing ,Nanopore Sequencing ,Protein Binding ,Sequence Analysis ,DNA ,Chemical Sciences ,Medical and Health Sciences ,Bioinformatics - Abstract
We recently developed directed methylation with long-read sequencing (DiMeLo-seq) to map protein-DNA interactions genome wide. DiMeLo-seq is capable of mapping multiple interaction sites on single DNA molecules, profiling protein binding in the context of endogenous DNA methylation, identifying haplotype-specific protein-DNA interactions and mapping protein-DNA interactions in repetitive regions of the genome that are difficult to study with short-read methods. With DiMeLo-seq, adenines in the vicinity of a protein of interest are methylated in situ by tethering the Hia5 methyltransferase to an antibody using protein A. Protein-DNA interactions are then detected by direct readout of adenine methylation with long-read, single-molecule DNA sequencing platforms such as Nanopore sequencing. Here we present a detailed protocol and practical guidance for performing DiMeLo-seq. This protocol can be run on nuclei from fresh, lightly fixed or frozen cells. The protocol requires 1-2 d for performing in situ targeted methylation, 1-5 d for library preparation depending on desired fragment length and 1-3 d for Nanopore sequencing depending on desired sequencing depth. The protocol requires basic molecular biology skills and equipment, as well as access to a Nanopore sequencer. We also provide a Python package, dimelo, for analysis of DiMeLo-seq data.
- Published
- 2024
16. Chemical shift assignments of the α-actinin C-terminal EF-hand domain bound to a cytosolic C0 domain of GluN1 (residues 841-865) from the NMDA receptor.
- Author
-
Bej, Aritra, Hell, Johannes, and Ames, James
- Subjects
C0 domain ,Calcium ,GluN1 ,NMDA receptor ,NMR ,a-actinin ,Humans ,Actinin ,Cytosol ,EF Hand Motifs ,Nuclear Magnetic Resonance ,Biomolecular ,Protein Binding ,Protein Domains ,Receptors ,N-Methyl-D-Aspartate - Abstract
N-methyl-D-aspartate receptors (NMDARs) consist of glycine-binding GluN1 and glutamate-binding GluN2 subunits that form tetrameric ion channels. NMDARs in the brain are important for controlling neuronal excitability to promote synaptic plasticity. The cytoskeletal protein, α-actinin-1 (100 kDa, called ACTN1) binds to the cytosolic C0 domain of GluN1 (residues 841-865) that may play a role in the Ca2+-dependent desensitization of NMDAR channels. Mutations that disrupt NMDAR channel function are linked to Alzheimers disease, depression, stroke, epilepsy, and schizophrenia. NMR chemical shift assignments are reported here for the C-terminal EF-hand domain of ACTN1 (residues 824-892, called ACTN_EF34) and ACTN_EF34 bound to the GluN1 C0 domain (BMRB numbers 52385 and 52386, respectively).
- Published
- 2024
17. Ribosome-inactivation by a class of widely distributed C-tail anchored membrane proteins
- Author
-
Njenga, Robert Karari, Boele, Julian, Drepper, Friedel, Sinha, Kasturica, Marouda, Eirini, Huesgen, Pitter F, Blaby-Haas, Crysten, and Koch, Hans-Georg
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Infection ,Generic health relevance ,Ribosomes ,Protein Biosynthesis ,Protein Binding ,Membrane Proteins ,Ribosome Subunits ,Large ,Bacterial ,Escherichia coli ,Models ,Molecular ,Escherichia coli Proteins ,Ribosomal Proteins ,Binding Sites ,Bacterial Proteins ,ElaB ,YgaM ,YqjD ,antimicrobial peptides ,macrolide antibiotics ,ribosomes ,stationary phase ,stress response ,translation ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Ribosome hibernation is a commonly used strategy that protects ribosomes under unfavorable conditions and regulates developmental processes. Multiple ribosome-hibernation factors have been identified in all domains of life, but due to their structural diversity and the lack of a common inactivation mechanism, it is currently unknown how many different hibernation factors exist. Here, we show that the YqjD/ElaB/YgaM paralogs, initially discovered as membrane-bound ribosome binding proteins in E. coli, constitute an abundant class of ribosome-hibernating proteins, which are conserved across all proteobacteria and some other bacterial phyla. Our data demonstrate that they inhibit in vitro protein synthesis by interacting with the 50S ribosomal subunit. In vivo cross-linking combined with mass spectrometry revealed their specific interactions with proteins surrounding the ribosomal tunnel exit and even their penetration into the ribosomal tunnel. Thus, YqjD/ElaB/YgaM inhibit translation by blocking the ribosomal tunnel and thus mimic the activity of antimicrobial peptides and macrolide antibiotics.
- Published
- 2024
18. Effects of Forming Lactoferrin-Milk Protein Complexes on Lactoferrin Functionality and Intestinal Development in Infancy.
- Author
-
Jiang, Rulan, Du, Xiaogu, and Lönnerdal, Bo
- Subjects
functionality of lactoferrin ,lactoferrin ,protein complex ,α-lactalbumin ,Lactoferrin ,Humans ,Caco-2 Cells ,Lactalbumin ,Whey Proteins ,Infant ,Intestinal Mucosa ,Animals ,Intestines ,Milk Proteins ,Epithelial Cells ,Cattle ,Digestion ,Milk ,Cell Proliferation ,Protein Binding - Abstract
BACKGROUND/OBJECTIVES: Lactoferrin (Lf) is an iron-binding glycoprotein with multiple bioactivities, including promotion of cell proliferation and differentiation, immunomodulation, and antimicrobial activity. Lf, a basic glycoprotein, can bind to α-lactalbumin (α-Lac), an acidic whey protein. The current study aimed to evaluate whether Lf forms protein complexes with α-Lac and proteins/peptides from whey protein hydrolysate (WPH) and nonfat bovine milk powder (MP) and whether forming protein complexes influences resistance to gastrointestinal digestion and affects the bioactivities of Lf in human intestinal epithelial cells (HIECs and differentiated Caco-2 cells). METHODS: Lf was blended with α-Lac, WPH, or MP. Assays were conducted to evaluate the bioactivities of proteins (Lf, α-Lac, WPH, or MP) and Lf-protein blends on HIECs and Caco-2 cells. RESULTS: (1) Lf forms complexes with α-Lac and proteins/peptides from WPH and MP; (2) compared with Lf alone, complexed Lf shows greater resistance to in vitro digestion; (3) forming protein complexes does not affect Lfs binding to the Lf receptor or its uptake by HIECs; and (4) forming protein complexes does not impact Lfs bioactivities, including the promotion of cell proliferation and differentiation, reduction of cell permeability by upregulating tight-junction proteins, immune modulation through the regulation of IL-18, inhibition of enteropathogenic Escherichia coli growth, and modulation of immune responses to EPEC infection. CONCLUSIONS: Lf forms complexes with α-Lac and other milk proteins/peptides from WPH and MP in protein blends, and forming complexes does not affect the functionalities of Lf.
- Published
- 2024
19. Nonspecific Yet Selective Interactions Contribute to Small Molecule Condensate Binding.
- Author
-
Wang, Cong, Kilgore, Henry, Latham, Andrew, and Zhang, Bin
- Subjects
Ligands ,Small Molecule Libraries ,Molecular Dynamics Simulation ,Hydrophobic and Hydrophilic Interactions ,Binding Sites ,Thermodynamics ,Protein Binding - Abstract
Biomolecular condensates are essential in various cellular processes, and their misregulation has been demonstrated to underlie disease. Small molecules that modulate condensate stability and material properties offer promising therapeutic approaches, but mechanistic insights into their interactions with condensates remain largely lacking. We employ a multiscale approach to enable long-time, equilibrated all-atom simulations of various condensate-ligand systems. Systematic characterization of the ligand binding poses reveals that condensates can form diverse and heterogeneous chemical environments with one or multiple chains to bind small molecules. Unlike traditional protein-ligand interactions, these chemical environments are dominated by nonspecific hydrophobic interactions. Nevertheless, the chemical environments feature unique amino acid compositions and physicochemical properties that favor certain small molecules over others, resulting in varied ligand partitioning coefficients within condensates. Notably, different condensates share similar sets of chemical environments but at different populations. This population shift drives ligand selectivity toward specific condensates. Our approach can enhance the interpretation of experimental screening data and may assist in the rational design of small molecules targeting specific condensates.
- Published
- 2024
20. SPOP-mediated RIPK3 destabilization desensitizes LPS/sMAC/zVAD-induced necroptotic cell death.
- Author
-
Lee, Ga-Eun, Bang, Geul, Byun, Jiin, Chen, Weidong, Jeung, Dohyun, Cho, Hana, Lee, Joo, Kang, Han, Lee, Hye, Kim, Jin, Kim, Kwang, Wu, Juan, Nam, Soo-Bin, Kwon, Young, Lee, Cheol-Jung, and Cho, Yong-Yeon
- Subjects
Kinases ,Necroptosis ,RIPK3 ,SPOP ,Ubiquitination ,Receptor-Interacting Protein Serine-Threonine Kinases ,Humans ,Nuclear Proteins ,Repressor Proteins ,Ubiquitination ,Phosphorylation ,Lipopolysaccharides ,Colonic Neoplasms ,Necroptosis ,HEK293 Cells ,Cell Line ,Tumor ,Cell Death ,Protein Stability ,Protein Binding - Abstract
RIPK1/RIPK3-MLKL signaling molecules are fundamental in initiating necroptotic cell death, but their roles in the development of colon cancer are unclear. This study reports that RIPK3 interacted with SPOP, a component of the E3 ligase within the Cul3 complex. This interaction leads to K48-linked ubiquitination and subsequent proteasomal degradation of RIPK3. Two distinct degron motifs, PETST and SPTST, were identified within the linker domain of RIPK3 for SPOP. RIPK3 phosphorylations at Thr403 by PIM2 and at Thr412/Ser413 by ERK2 are essential to facilitate its interaction with SPOP. Computational docking studies and immunoprecipitation analyses showed that these PIM2 and ERK2 phosphorylations bolster the stability of the RIPK3-SPOP interaction. In particular, mutations of RIPK3 at the degron motifs extended the half-life of RIPK3 by preventing its phosphorylation and subsequent ubiquitination. The deletion of SPOP, which led to increased stability of the RIPK3 protein, intensified LPS/sMAC/zVAD-induced necroptotic cell death in colon cancer cells. These findings underscore the critical role of the SPOP-mediated RIPK3 stability regulation pathway in controlling necroptotic cell death.
- Published
- 2024
21. HURP regulates Kif18A recruitment and activity to synergistically control microtubule dynamics.
- Author
-
Perez-Bertoldi, Juan, Zhao, Yuanchang, Thawani, Akanksha, Yildiz, Ahmet, and Nogales, Eva
- Subjects
Kinesins ,Microtubules ,Humans ,Protein Binding ,Cryoelectron Microscopy ,Mitosis ,Binding Sites ,Single Molecule Imaging ,Kinetochores - Abstract
During mitosis, microtubule dynamics are regulated to ensure proper alignment and segregation of chromosomes. The dynamics of kinetochore-attached microtubules are regulated by hepatoma-upregulated protein (HURP) and the mitotic kinesin-8 Kif18A, but the underlying mechanism remains elusive. Using single-molecule imaging in vitro, we demonstrate that Kif18A motility is regulated by HURP. While sparse decoration of HURP activates the motor, higher concentrations hinder processive motility. To shed light on this behavior, we determine the binding mode of HURP to microtubules using cryo-EM. The structure helps rationalize why HURP functions as a microtubule stabilizer. Additionally, HURP partially overlaps with the microtubule-binding site of the Kif18A motor domain, indicating that excess HURP inhibits Kif18A motility by steric exclusion. We also observe that HURP and Kif18A function together to suppress dynamics of the microtubule plus-end, providing a mechanistic basis for how they collectively serve in microtubule length control.
- Published
- 2024
22. Lysosomal enzyme binding to the cation-independent mannose 6-phosphate receptor is regulated allosterically by insulin-like growth factor 2
- Author
-
Bohnsack, Richard N, Misra, Sandeep K, Liu, Jianfang, Ishihara-Aoki, Mayumi, Pereckas, Michaela, Aoki, Kazuhiro, Ren, Gang, Sharp, Joshua S, and Dahms, Nancy M
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Insulin-Like Growth Factor II ,Receptor ,IGF Type 2 ,Lysosomes ,Protein Binding ,Humans ,Binding Sites ,Animals ,Allosteric Regulation ,Cattle ,Glycosylation ,Mannosephosphates ,Insulin-Like Peptides - Abstract
The cation-independent mannose 6-phosphate receptor (CI-MPR) is clinically significant in the treatment of patients with lysosomal storage diseases because it functions in the biogenesis of lysosomes by transporting mannose 6-phosphate (M6P)-containing lysosomal enzymes to endosomal compartments. CI-MPR is multifunctional and modulates embryonic growth and fetal size by downregulating circulating levels of the peptide hormone insulin-like growth factor 2 (IGF2). The extracellular region of CI-MPR comprises 15 homologous domains with binding sites for M6P-containing ligands located in domains 3, 5, 9, and 15, whereas IGF2 interacts with residues in domain 11. How a particular ligand affects the receptor's conformation or its ability to bind other ligands remains poorly understood. To address these questions, we purified a soluble form of the receptor from newborn calf serum, carried out glycoproteomics to define the N-glycans at its 19 potential glycosylation sites, probed its ability to bind lysosomal enzymes in the presence and absence of IGF2 using surface plasmon resonance, and assessed its conformation in the presence and absence of IGF2 by negative-staining electron microscopy and hydroxyl radical protein footprinting studies. Together, our findings support the hypothesis that IGF2 acts as an allosteric inhibitor of lysosomal enzyme binding by inducing global conformational changes of CI-MPR.
- Published
- 2024
23. Microtubule-associated protein, MAP1B, encodes functionally distinct polypeptides
- Author
-
Tan, Tracy C, Shen, Yusheng, Stine, Lily B, Mitchell, Barbara, Okada, Kyoko, McKenney, Richard J, and Ori-McKenney, Kassandra M
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Microtubule-Associated Proteins ,Humans ,Microtubules ,Kinesins ,Actin Cytoskeleton ,Animals ,Protein Binding ,Phosphorylation ,Dyneins ,DYRK1a ,actin ,dynein ,kinesin ,microtubule-associated protein ,microtubules ,phosphorylation ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Microtubule-associated protein, MAP1B, is crucial for neuronal morphogenesis and disruptions in MAP1B function are correlated with neurodevelopmental disorders. MAP1B encodes a single polypeptide that is processed into discrete proteins, a heavy chain (HC) and a light chain (LC); however, it is unclear if these two chains operate individually or as a complex within the cell. In vivo studies have characterized the contribution of MAP1B HC and LC to microtubule and actin-based processes, but their molecular mechanisms of action are unknown. Using in vitro reconstitution with purified proteins, we dissect the biophysical properties of the HC and LC and uncover distinct binding behaviors and functional roles for these MAPs. Our biochemical assays indicate that MAP1B HC and LC do not form a constitutive complex, supporting the hypothesis that these proteins operate independently within cells. Both HC and LC inhibit the microtubule motors, kinesin-3, kinesin-4, and dynein, and differentially affect the severing activity of spastin. Notably, MAP1B LC binds to actin filaments in vitro and can simultaneously bind and cross-link actin filaments and microtubules, a function not observed for MAP1B HC. Phosphorylation of MAP1B HC by dual-specificity, tyrosine phosphorylation-regulated kinase 1a negatively regulates its actin-binding activity without significantly affecting its microtubule-binding capacity, suggesting a dynamic contribution of MAP1B HC in cytoskeletal organization. Overall, our study provides new insights into the distinct functional properties of MAP1B HC and LC, underscoring their roles in coordinating cytoskeletal networks during neuronal development.
- Published
- 2024
24. Bone-marrow macrophage-derived GPNMB protein binds to orphan receptor GPR39 and plays a critical role in cardiac repair
- Author
-
Ramadoss, Sivakumar, Qin, Juan, Tao, Bo, Thomas, Nathan E, Cao, Edward, Wu, Rimao, Sandoval, Daniel R, Piermatteo, Ann, Grunddal, Kaare V, Ma, Feiyang, Li, Shen, Sun, Baiming, Zhou, Yonggang, Wan, Jijun, Pellegrini, Matteo, Holst, Birgitte, Lusis, Aldons J, Gordts, Philip LSM, and Deb, Arjun
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Heart Disease - Coronary Heart Disease ,Cardiovascular ,Heart Disease ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Animals ,Receptors ,G-Protein-Coupled ,Humans ,Macrophages ,Membrane Glycoproteins ,Myocardial Infarction ,Mice ,Knockout ,Disease Models ,Animal ,Myocytes ,Cardiac ,Male ,Mice ,Inbred C57BL ,Signal Transduction ,Ventricular Function ,Left ,Heart Failure ,Female ,Mice ,Cells ,Cultured ,Ventricular Dysfunction ,Left ,Bone Marrow Transplantation ,Protein Binding ,Regeneration ,Eye Proteins - Abstract
Glycoprotein nonmetastatic melanoma protein B (GPNMB) is a type I transmembrane protein initially identified in nonmetastatic melanomas and has been associated with human heart failure; however, its role in cardiac injury and function remains unclear. Here we show that GPNMB expression is elevated in failing human and mouse hearts after myocardial infarction (MI). Lineage tracing and bone-marrow transplantation reveal that bone-marrow-derived macrophages are the main source of GPNMB in injured hearts. Using genetic loss-of-function models, we demonstrate that GPNMB deficiency leads to increased mortality, cardiac rupture and rapid post-MI left ventricular dysfunction. Conversely, increasing circulating GPNMB levels through viral delivery improves heart function after MI. Single-cell transcriptomics show that GPNMB enhances myocyte contraction and reduces fibroblast activation. Additionally, we identified GPR39 as a receptor for circulating GPNMB, with its absence negating the beneficial effects. These findings highlight a pivotal role of macrophage-derived GPNMBs in post-MI cardiac repair through GPR39 signaling.
- Published
- 2024
25. Prediction of Threonine-Tyrosine Kinase Receptor-Ligand Unbinding Kinetics with Multiscale Milestoning and Metadynamics.
- Author
-
Votapka, Lane, Ojha, Anupam, Asada, Naoya, and Amaro, Rommie
- Subjects
Ligands ,Kinetics ,Molecular Dynamics Simulation ,Thermodynamics ,Protein Kinase Inhibitors ,Protein Binding ,Protein Serine-Threonine Kinases - Abstract
Accurately describing protein-ligand binding and unbinding kinetics remains challenging. Computational calculations are difficult and costly, while experimental measurements often lack molecular detail and can be unobtainable. Here, we extend our multiscale milestoning method, Simulation-Enabled Estimation of Kinetics Rates (SEEKR), with metadynamics molecular dynamics simulations to yield accurate small molecule drug residence times. Using the pharmaceutically relevant threonine-tyrosine kinase (TTK) and eight long-residence-time (tens of seconds to hours) inhibitors, we demonstrate accurate prediction of absolute and rank-ordered ligand residence times and free energies of binding.
- Published
- 2024
26. Structural basis of μ-opioid receptor targeting by a nanobody antagonist.
- Author
-
Yu, Jun, Kumar, Amit, Zhang, Xuefeng, Martin, Charlotte, Van Holsbeeck, Kevin, Raia, Pierre, Koehl, Antoine, Laeremans, Toon, Steyaert, Jan, Manglik, Aashish, Ballet, Steven, Boland, Andreas, and Stoeber, Miriam
- Subjects
Receptors ,Opioid ,mu ,Single-Domain Antibodies ,Humans ,Cryoelectron Microscopy ,Ligands ,HEK293 Cells ,Animals ,Protein Binding ,Binding Sites ,Models ,Molecular ,Analgesics ,Opioid ,Peptides ,Cyclic - Abstract
The μ-opioid receptor (μOR), a prototypical G protein-coupled receptor (GPCR), is the target of opioid analgesics such as morphine and fentanyl. Due to the severe side effects of current opioid drugs, there is considerable interest in developing novel modulators of μOR function. Most GPCR ligands today are small molecules, however biologics, including antibodies and nanobodies, represent alternative therapeutics with clear advantages such as affinity and target selectivity. Here, we describe the nanobody NbE, which selectively binds to the μOR and acts as an antagonist. We functionally characterize NbE as an extracellular and genetically encoded μOR ligand and uncover the molecular basis for μOR antagonism by determining the cryo-EM structure of the NbE-μOR complex. NbE displays a unique ligand binding mode and achieves μOR selectivity by interactions with the orthosteric pocket and extracellular receptor loops. Based on a β-hairpin loop formed by NbE that deeply protrudes into the μOR, we design linear and cyclic peptide analogs that recapitulate NbEs antagonism. The work illustrates the potential of nanobodies to uniquely engage with GPCRs and describes lower molecular weight μOR ligands that can serve as a basis for therapeutic developments.
- Published
- 2024
27. Combinatorial transcription factor binding encodes cis-regulatory wiring of mouse forebrain GABAergic neurogenesis
- Author
-
Catta-Preta, Rinaldo, Lindtner, Susan, Ypsilanti, Athena, Seban, Nicolas, Price, James D, Abnousi, Armen, Su-Feher, Linda, Wang, Yurong, Cichewicz, Karol, Boerma, Sally A, Juric, Ivan, Jones, Ian R, Akiyama, Jennifer A, Hu, Ming, Shen, Yin, Visel, Axel, Pennacchio, Len A, Dickel, Diane E, Rubenstein, John LR, and Nord, Alex S
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Biological Sciences ,Stem Cell Research ,Human Genome ,Genetics ,Prosencephalon ,Chromatin ,Animals ,Mice ,Histones ,Transcription Factors ,Gene Expression Regulation ,Developmental ,Protein Binding ,Enhancer Elements ,Genetic ,Promoter Regions ,Genetic ,Neurogenesis ,GABAergic Neurons ,Chromatin Immunoprecipitation Sequencing ,GABAergic cortical interneurons ,chromatin conformation ,combinatorial TF binding ,evolutionary conservation ,gene regulatory network ,neurogenesis ,transcription factors ,Medical and Health Sciences ,Developmental Biology ,Biochemistry and cell biology - Abstract
Transcription factors (TFs) bind combinatorially to cis-regulatory elements, orchestrating transcriptional programs. Although studies of chromatin state and chromosomal interactions have demonstrated dynamic neurodevelopmental cis-regulatory landscapes, parallel understanding of TF interactions lags. To elucidate combinatorial TF binding driving mouse basal ganglia development, we integrated chromatin immunoprecipitation sequencing (ChIP-seq) for twelve TFs, H3K4me3-associated enhancer-promoter interactions, chromatin and gene expression data, and functional enhancer assays. We identified sets of putative regulatory elements with shared TF binding (TF-pRE modules) that orchestrate distinct processes of GABAergic neurogenesis and suppress other cell fates. The majority of pREs were bound by one or two TFs; however, a small proportion were extensively bound. These sequences had exceptional evolutionary conservation and motif density, complex chromosomal interactions, and activity as in vivo enhancers. Our results provide insights into the combinatorial TF-pRE interactions that activate and repress expression programs during telencephalon neurogenesis and demonstrate the value of TF binding toward modeling developmental transcriptional wiring.
- Published
- 2024
28. Large-scale map of RNA-binding protein interactomes across the mRNA life cycle
- Author
-
Street, Lena A, Rothamel, Katherine L, Brannan, Kristopher W, Jin, Wenhao, Bokor, Benjamin J, Dong, Kevin, Rhine, Kevin, Madrigal, Assael, Al-Azzam, Norah, Kim, Jenny Kim, Ma, Yanzhe, Gorhe, Darvesh, Abdou, Ahmed, Wolin, Erica, Mizrahi, Orel, Ahdout, Joshua, Mujumdar, Mayuresh, Doron-Mandel, Ella, Jovanovic, Marko, and Yeo, Gene W
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Genetics ,1.1 Normal biological development and functioning ,Generic health relevance ,Neurological ,Humans ,RNA ,Messenger ,RNA-Binding Proteins ,Protein Interaction Maps ,Protein Binding ,HeLa Cells ,Protein Interaction Mapping ,Ribonucleoproteins ,Small Nuclear ,HEK293 Cells ,Mass Spectrometry ,RNA Splicing ,Hela Cells ,ERH ,IP-MS ,RBP ,RNA-binding proteins ,RNA-dependent protein interactions ,SEC-MS ,SNRNP200 ,interactome ,mRNA life-cycle ,protein-protein interactions ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
mRNAs interact with RNA-binding proteins (RBPs) throughout their processing and maturation. While efforts have assigned RBPs to RNA substrates, less exploration has leveraged protein-protein interactions (PPIs) to study proteins in mRNA life-cycle stages. We generated an RNA-aware, RBP-centric PPI map across the mRNA life cycle in human cells by immunopurification-mass spectrometry (IP-MS) of ∼100 endogenous RBPs with and without RNase, augmented by size exclusion chromatography-mass spectrometry (SEC-MS). We identify 8,742 known and 20,802 unreported interactions between 1,125 proteins and determine that 73% of the IP-MS-identified interactions are RNA regulated. Our interactome links many proteins, some with unknown functions, to specific mRNA life-cycle stages, with nearly half associated with multiple stages. We demonstrate the value of this resource by characterizing the splicing and export functions of enhancer of rudimentary homolog (ERH), and by showing that small nuclear ribonucleoprotein U5 subunit 200 (SNRNP200) interacts with stress granule proteins and binds cytoplasmic RNA differently during stress.
- Published
- 2024
29. Molecular mechanism of contactin 2 homophilic interaction
- Author
-
Fan, Shanghua, Liu, Jianfang, Chofflet, Nicolas, Bailey, Aaron O, Russell, William K, Zhang, Ziqi, Takahashi, Hideto, Ren, Gang, and Rudenko, Gabby
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Neurological ,Humans ,Protein Binding ,Protein Multimerization ,Cryoelectron Microscopy ,Contactin 2 ,Models ,Molecular ,Binding Sites ,HEK293 Cells ,adhesion molecule ,contactins ,cross-linking mass spectrometry ,cryo-EM ,molecular recognition mechanism ,molecular specificity ,neuronal guidance molecule ,protein structure ,protein:protein interaction ,single-particle analysis ,Chemical Sciences ,Information and Computing Sciences ,Biophysics ,Biological sciences ,Chemical sciences - Abstract
Contactin 2 (CNTN2) is a cell adhesion molecule involved in axon guidance, neuronal migration, and fasciculation. The ectodomains of CNTN1-CNTN6 are composed of six Ig domains (Ig1-Ig6) and four FN domains. Here, we show that CNTN2 forms transient homophilic interactions (KD ∼200 nM). Cryo-EM structures of full-length CNTN2 and CNTN2_Ig1-Ig6 reveal a T-shaped homodimer formed by intertwined, parallel monomers. Unexpectedly, the horseshoe-shaped Ig1-Ig4 headpieces extend their Ig2-Ig3 tips outwards on either side of the homodimer, while Ig4, Ig5, Ig6, and the FN domains form a central stalk. Cross-linking mass spectrometry and cell-based binding assays confirm the 3D assembly of the CNTN2 homodimer. The interface mediating homodimer formation differs between CNTNs, as do the homophilic versus heterophilic interaction mechanisms. The CNTN family thus encodes a versatile molecular platform that supports a very diverse portfolio of protein interactions and that can be leveraged to strategically guide neural circuit development.
- Published
- 2024
30. Atomistic mechanisms of the regulation of small-conductance Ca2+-activated K+ channel (SK2) by PIP2
- Author
-
Woltz, Ryan L, Zheng, Yang, Choi, Woori, Ngo, Khoa, Trinh, Pauline, Ren, Lu, Thai, Phung N, Harris, Brandon J, Han, Yanxiao, Rouen, Kyle C, Mateos, Diego Lopez, Jian, Zhong, Chen-Izu, Ye, Dickson, Eamonn J, Yamoah, Ebenezer N, Yarov-Yarovoy, Vladimir, Vorobyov, Igor, Zhang, Xiao-Dong, and Chiamvimonvat, Nipavan
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Cardiovascular ,1.1 Normal biological development and functioning ,5.1 Pharmaceuticals ,Phosphatidylinositol 4 ,5-Diphosphate ,Small-Conductance Calcium-Activated Potassium Channels ,Molecular Dynamics Simulation ,Animals ,Calmodulin ,Humans ,Ion Channel Gating ,Calcium ,Protein Binding ,Myocytes ,Cardiac ,small conductance Ca2+- activated K plus channel ,phosphatidylinositol 4 ,5-bisphosphate ,optogenetics ,calmodulin ,atrial arrhythmias ,phosphatidylinositol 4 ,5-bisphosphate ,small conductance Ca2+-activated K+ channel - Abstract
Small-conductance Ca2+-activated K+ channels (SK, KCa2) are gated solely by intracellular microdomain Ca2+. The channel has emerged as a therapeutic target for cardiac arrhythmias. Calmodulin (CaM) interacts with the CaM binding domain (CaMBD) of the SK channels, serving as the obligatory Ca2+ sensor to gate the channels. In heterologous expression systems, phosphatidylinositol 4,5-bisphosphate (PIP2) coordinates with CaM in regulating SK channels. However, the roles and mechanisms of PIP2 in regulating SK channels in cardiomyocytes remain unknown. Here, optogenetics, magnetic nanoparticles, combined with Rosetta structural modeling, and molecular dynamics (MD) simulations revealed the atomistic mechanisms of how PIP2 works in concert with Ca2+-CaM in the SK channel activation. Our computational study affords evidence for the critical role of the amino acid residue R395 in the S6 transmembrane segment, which is localized in propinquity to the intracellular hydrophobic gate. This residue forms a salt bridge with residue E398 in the S6 transmembrane segment from the adjacent subunit. Both R395 and E398 are conserved in all known isoforms of SK channels. Our findings suggest that the binding of PIP2 to R395 residue disrupts the R395:E398 salt bridge, increasing the flexibility of the transmembrane segment S6 and the activation of the channel. Importantly, our findings serve as a platform for testing of structural-based drug designs for therapeutic inhibitors and activators of the SK channel family. The study is timely since inhibitors of SK channels are currently in clinical trials to treat atrial arrhythmias.
- Published
- 2024
31. Rational Design of High Affinity Interaction Between CC Chemokine Binding Protein vCCI and CCL17/TARC
- Author
-
Guan, Wenyan, Stark, Lauren E, Zhang, Ning, Bains, Arjan, Martinez, Airam, Dupureur, Cynthia M, Colvin, Michael E, and LiWang, Patricia J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Infectious Diseases ,5.1 Pharmaceuticals ,Chemokine CCL17 ,Humans ,Viral Proteins ,Protein Binding ,Molecular Dynamics Simulation ,Amino Acid Sequence ,Chemokines ,CC ,Mutation ,Medicinal and Biomolecular Chemistry ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics ,Medicinal and biomolecular chemistry - Abstract
The poxvirus-derived protein vCCI (viral CC chemokine inhibitor) binds almost all members of the CC chemokine family with nanomolar affinity, inhibiting their pro-inflammatory actions. Understanding the affinity and specificity of vCCI could lead to new anti-inflammatory therapeutics. CCL17, also known as TARC, is unusual among CC chemokines by having only micromolar binding to vCCI. We have used sequence analysis and molecular simulations to determine the cause of this weak binding, which identified several locations in CCL17 where mutations seemed likely to improve binding to vCCI. Based on the aforementioned analysis, we expressed and tested multiple mutants of CCL17. We found two single point mutants V44K and Q45R that increased binding affinity to vCCI by 2-3-fold and, in combination, further improved affinity by 7-fold. The CCL17 triple mutant G17R/V44K/Q45R yielded a Kd of 0.25 ± 0.13 μM, a 68-fold improvement in affinity compared to the complex with wild-type CCL17. A quadruple mutant G17R/V44K/Q45R/R57W showed high affinity (0.59 ± 0.09 μM) compared to the wild type but lower affinity than the triple mutant. This work demonstrates that sequence comparisons and molecular simulations can predict chemokine mutations that increase the level of binding to vCCI, an important first step in developing engineered chemokine inhibitors useful for anti-inflammatory therapy.
- Published
- 2024
32. Structural basis of TRPV1 modulation by endogenous bioactive lipids
- Author
-
Arnold, William R, Mancino, Adamo, Moss, Frank R, Frost, Adam, Julius, David, and Cheng, Yifan
- Subjects
Biomedical and Clinical Sciences ,Neurosciences ,Pain Research ,Chronic Pain ,5.1 Pharmaceuticals ,TRPV Cation Channels ,Animals ,Rats ,Lysophospholipids ,Binding Sites ,Protein Binding ,Models ,Molecular ,Humans ,Phosphatidylinositols ,Protein Conformation ,HEK293 Cells ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biophysics ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
TRP ion channels are modulated by phosphoinositide lipids, but the underlying structural mechanisms remain unclear. The capsaicin- and heat-activated receptor, TRPV1, has served as a model for deciphering lipid modulation, which is relevant to understanding how pro-algesic agents enhance channel activity in the setting of inflammatory pain. Identification of a pocket within the TRPV1 transmembrane core has provided initial clues as to how phosphoinositide lipids bind to and regulate the channel. Here we show that this regulatory pocket in rat TRPV1 can accommodate diverse lipid species, including the inflammatory lipid lysophosphatidic acid, whose actions are determined by their specific modes of binding. Furthermore, we show that an empty-pocket channel lacking an endogenous phosphoinositide lipid assumes an agonist-like state, even at low temperature, substantiating the concept that phosphoinositide lipids serve as negative TRPV1 modulators whose ejection from the binding pocket is a critical step toward activation by thermal or chemical stimuli.
- Published
- 2024
33. Integrin mechanosensing relies on a pivot-clip mechanism to reinforce cell adhesion.
- Author
-
Montes, Andre, Barroso, Anahi, Wang, Wei, OConnell, Grace, Tepole, Adrian, and Mofrad, Mohammad
- Subjects
Cell Adhesion ,Integrin alpha5beta1 ,Fibronectins ,Molecular Dynamics Simulation ,Mechanotransduction ,Cellular ,Binding Sites ,Humans ,Protein Binding ,Oligopeptides - Abstract
Cells intricately sense mechanical forces from their surroundings, driving biophysical and biochemical activities. This mechanosensing phenomenon occurs at the cell-matrix interface, where mechanical forces resulting from cellular motion, such as migration or matrix stretching, are exchanged through surface receptors, primarily integrins, and their corresponding matrix ligands. A pivotal player in this interaction is the α5β1 integrin and fibronectin (FN) bond, known for its role in establishing cell adhesion sites for migration. However, upregulation of the α5β1-FN bond is associated with uncontrolled cell metastasis. This bond operates through catch bond dynamics, wherein the bond lifetime paradoxically increases with greater force. The mechanism sustaining the characteristic catch bond dynamics of α5β1-FN remains unclear. Leveraging molecular dynamics simulations, our approach unveils a pivot-clip mechanism. Two key binding sites on FN, namely the synergy site and the RGD (Arg-Gly-Asp) motif, act as active points for structural changes in α5β1 integrin. Conformational adaptations at these sites are induced by a series of hydrogen bond formations and breaks at the synergy site. We disrupt these adaptations through a double mutation on FN, known to reduce cell adhesion. A whole-cell finite-element model is employed to elucidate how the synergy site may promote dynamic α5β1-FN binding, resisting cell contraction. In summary, our study integrates molecular- and cellular-level modeling to propose that FNs synergy site reinforces cell adhesion through enhanced binding dynamics and a mechanosensitive pivot-clip mechanism. This work sheds light on the interplay between mechanical forces and cell-matrix interactions, contributing to our understanding of cellular behaviors in physiological and pathological contexts.
- Published
- 2024
34. Alteration of Neuropilin‑1 and Heparan Sulfate Interaction Impairs Murine B16 Tumor Growth
- Author
-
Painter, Chelsea D, Sankaranarayanan, Nehru Viji, Nagarajan, Balaji, Clausen, Thomas Mandel, West, Alan MV, Setiawan, Nicollette J, Park, Jeeyoung, Porell, Ryan N, Bartels, Phillip L, Sandoval, Daniel R, Vasquez, Gabriel J, Chute, John P, Godula, Kamil, Vander Kooi, Craig W, Gordts, Philip LSM, Corbett, Kevin D, Termini, Christina M, Desai, Umesh R, and Esko, Jeffrey D
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Neuropilin-1 ,Animals ,Heparitin Sulfate ,Mice ,Melanoma ,Experimental ,Protein Binding ,Binding Sites ,Mice ,Inbred C57BL ,Heparin ,Molecular Dynamics Simulation ,Mutation ,Chemical Sciences ,Organic Chemistry ,Biological sciences ,Chemical sciences - Abstract
Neuropilin-1 acts as a coreceptor with vascular endothelial growth factor receptors to facilitate binding of its ligand, vascular endothelial growth factor. Neuropilin-1 also binds to heparan sulfate, but the functional significance of this interaction has not been established. A combinatorial library screening using heparin oligosaccharides followed by molecular dynamics simulations of a heparin tetradecasaccharide suggested a highly conserved binding site composed of amino acid residues extending across the b1 and b2 domains of murine neuropilin-1. Mutagenesis studies established the importance of arginine513 and lysine514 for binding of heparin to a recombinant form of Nrp1 composed of the a1, a2, b1, and b2 domains. Recombinant Nrp1 protein bearing R513A,K514A mutations showed a significant loss of heparin-binding, heparin-induced dimerization, and heparin-dependent thermal stabilization. Isothermal calorimetry experiments suggested a 1:2 complex of heparin tetradecasaccharide:Nrp1. To study the impact of altered heparin binding in vivo, a mutant allele of Nrp1 bearing the R513A,K514A mutations was created in mice (Nrp1D) and crossbred to Nrp1+/- mice to examine the impact of altered heparan sulfate binding. Analysis of tumor formation showed variable effects on tumor growth in Nrp1D/D mice, resulting in a frank reduction in tumor growth in Nrp1D/- mice. Expression of mutant Nrp1D protein was normal in tissues, suggesting that the reduction in tumor growth was due to the altered binding of heparin/heparan sulfate to neuropilin-1. These findings suggest that the interaction of neuropilin-1 with heparan sulfate modulates its stability and its role in tumor formation and growth.
- Published
- 2024
35. Multi-layered heterochromatin interaction as a switch for DIM2-mediated DNA methylation.
- Author
-
Shao, Zengyu, Lu, Jiuwei, Khudaverdyan, Nelli, and Song, Jikui
- Subjects
Heterochromatin ,DNA Methylation ,Chromosomal Proteins ,Non-Histone ,Histones ,Chromobox Protein Homolog 5 ,Fungal Proteins ,Protein Binding ,Neurospora crassa - Abstract
Functional crosstalk between DNA methylation, histone H3 lysine-9 trimethylation (H3K9me3) and heterochromatin protein 1 (HP1) is essential for proper heterochromatin assembly and genome stability. However, how repressive chromatin cues guide DNA methyltransferases for region-specific DNA methylation remains largely unknown. Here, we report structure-function characterizations of DNA methyltransferase Defective-In-Methylation-2 (DIM2) in Neurospora. The DNA methylation activity of DIM2 requires the presence of both H3K9me3 and HP1. Our structural study reveals a bipartite DIM2-HP1 interaction, leading to a disorder-to-order transition of the DIM2 target-recognition domain that is essential for substrate binding. Furthermore, the structure of DIM2-HP1-H3K9me3-DNA complex reveals a substrate-binding mechanism distinct from that for its mammalian orthologue DNMT1. In addition, the dual recognition of H3K9me3 peptide by the DIM2 RFTS and BAH1 domains allosterically impacts the DIM2-substrate binding, thereby controlling DIM2-mediated DNA methylation. Together, this study uncovers how multiple heterochromatin factors coordinately orchestrate an activity-switching mechanism for region-specific DNA methylation.
- Published
- 2024
36. Huntingtin contains an ubiquitin-binding domain and regulates lysosomal targeting of mitochondrial and RNA-binding proteins
- Author
-
Fote, Gianna M, Eapen, Vinay V, Lim, Ryan G, Yu, Clinton, Salazar, Lisa, McClure, Nicolette R, McKnight, Jharrayne, Nguyen, Thai B, Heath, Marie C, Lau, Alice L, Villamil, Mark A, Miramontes, Ricardo, Kratter, Ian H, Finkbeiner, Steven, Reidling, Jack C, Paulo, Joao A, Kaiser, Peter, Huang, Lan, Housman, David E, Thompson, Leslie M, and Steffan, Joan S
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Huntington's Disease ,Neurodegenerative ,Genetics ,Rare Diseases ,Orphan Drug ,Neurosciences ,Brain Disorders ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Neurological ,Huntingtin Protein ,Lysosomes ,RNA-Binding Proteins ,Humans ,Ubiquitin ,Mitochondria ,Autophagy ,Animals ,Mitochondrial Proteins ,Mice ,Protein Binding ,Huntington Disease ,Peptides ,Huntingtin ,ubiquitin- binding domain ,ubiquitin ,autophagy ,RNA- binding proteins ,RNA-binding proteins ,ubiquitin-binding domain - Abstract
Understanding the normal function of the Huntingtin (HTT) protein is of significance in the design and implementation of therapeutic strategies for Huntington's disease (HD). Expansion of the CAG repeat in the HTT gene, encoding an expanded polyglutamine (polyQ) repeat within the HTT protein, causes HD and may compromise HTT's normal activity contributing to HD pathology. Here, we investigated the previously defined role of HTT in autophagy specifically through studying HTT's association with ubiquitin. We find that HTT interacts directly with ubiquitin in vitro. Tandem affinity purification was used to identify ubiquitinated and ubiquitin-associated proteins that copurify with a HTT N-terminal fragment under basal conditions. Copurification is enhanced by HTT polyQ expansion and reduced by mimicking HTT serine 421 phosphorylation. The identified HTT-interacting proteins include RNA-binding proteins (RBPs) involved in mRNA translation, proteins enriched in stress granules, the nuclear proteome, the defective ribosomal products (DRiPs) proteome and the brain-derived autophagosomal proteome. To determine whether the proteins interacting with HTT are autophagic targets, HTT knockout (KO) cells and immunoprecipitation of lysosomes were used to investigate autophagy in the absence of HTT. HTT KO was associated with reduced abundance of mitochondrial proteins in the lysosome, indicating a potential compromise in basal mitophagy, and increased lysosomal abundance of RBPs which may result from compensatory up-regulation of starvation-induced macroautophagy. We suggest HTT is critical for appropriate basal clearance of mitochondrial proteins and RBPs, hence reduced HTT proteostatic function with mutation may contribute to the neuropathology of HD.
- Published
- 2024
37. Factors affecting protein recovery during Hsp40 affinity profiling.
- Author
-
Montoya, Maureen, Quanrud, Guy, Mei, Liangyong, Moñtano, José, Hong, Caleb, and Genereux, Joseph
- Subjects
AP-MS ,DNAJB1 ,DNAJB8 ,Hsp40 ,Misfolded proteins ,Proteomics ,HSP40 Heat-Shock Proteins ,Humans ,HEK293 Cells ,Proteomics ,Protein Binding ,HSP70 Heat-Shock Proteins ,Protein Stability ,Protein Folding - Abstract
The identification and quantification of misfolded proteins from complex mixtures is important for biological characterization and disease diagnosis, but remains a major bioanalytical challenge. We have developed Hsp40 Affinity Profiling as a bioanalytical approach to profile protein stability in response to cellular stress. In this assay, we ectopically introduce the Hsp40 FlagDNAJB8H31Q into cells and use quantitative proteomics to determine how protein affinity for DNAJB8 changes in the presence of cellular stress, without regard for native clients. Herein, we evaluate potential approaches to improve the performance of this bioanalytical assay. We find that although intracellular crosslinking increases recovery of protein interactors, this is not enough to overcome the relative drop in DNAJB8 recovery. While the J-domain promotes Hsp70 association, it does not affect the yield of protein association with DNAJB8 under basal conditions. By contrast, crosslinking and J-domain ablation both substantially increase relative protein interactor recovery with the structurally distinct Class B Hsp40 DNAJB1 but are completely compensated by poorer yield of DNAJB1 itself. Cellular thermal stress promotes increased affinity between DNAJB8H31Q and interacting proteins, as expected for interactions driven by recognition of misfolded proteins. DNAJB8WT does not demonstrate such a property, suggesting that under stress misfolded proteins are handed off to Hsp70. Hence, we find that DNAJB8H31Q is still our most effective recognition element for the recovery of destabilized client proteins following cellular stress.
- Published
- 2024
38. Novel Z-DNA binding domains in giant viruses
- Author
-
Romero, Miguel F, Krall, Jeffrey B, Nichols, Parker J, Vantreeck, Jillian, Henen, Morkos A, Dejardin, Emmanuel, Schulz, Frederik, Vicens, Quentin, Vögeli, Beat, and Diallo, Mamadou Amadou
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biotechnology ,Genetics ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Generic health relevance ,Infection ,DNA ,Z-Form ,RNA-Binding Proteins ,Giant Viruses ,Protein Domains ,Viral Proteins ,DNA-Binding Proteins ,Immunity ,Innate ,Humans ,Protein Binding ,ADAR1 ,B-Z conversion ,B-to-Z conversion ,ZBP1 ,Zα domain ,innate immunity ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
Z-nucleic acid structures play vital roles in cellular processes and have implications in innate immunity due to their recognition by Zα domains containing proteins (Z-DNA/Z-RNA binding proteins, ZBPs). Although Zα domains have been identified in six proteins, including viral E3L, ORF112, and I73R, as well as, cellular ADAR1, ZBP1, and PKZ, their prevalence across living organisms remains largely unexplored. In this study, we introduce a computational approach to predict Zα domains, leading to the revelation of previously unidentified Zα domain-containing proteins in eukaryotic organisms, including non-metazoan species. Our findings encompass the discovery of new ZBPs in previously unexplored giant viruses, members of the Nucleocytoviricota phylum. Through experimental validation, we confirm the Zα functionality of select proteins, establishing their capability to induce the B-to-Z conversion. Additionally, we identify Zα-like domains within bacterial proteins. While these domains share certain features with Zα domains, they lack the ability to bind to Z-nucleic acids or facilitate the B-to-Z DNA conversion. Our findings significantly expand the ZBP family across a wide spectrum of organisms and raise intriguing questions about the evolutionary origins of Zα-containing proteins. Moreover, our study offers fresh perspectives on the functional significance of Zα domains in virus sensing and innate immunity and opens avenues for exploring hitherto undiscovered functions of ZBPs.
- Published
- 2024
39. A novel reporter for helicase activity in translation uncovers DDX3X interactions
- Author
-
Wilkins, Kevin, Schroeder, Till, Gu, Sohyun, Revalde, Jezrael Lafuente, and Floor, Stephen N
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,DEAD-box RNA Helicases ,Humans ,5' Untranslated Regions ,Protein Biosynthesis ,Genes ,Reporter ,Nucleic Acid Conformation ,RNA ,Messenger ,HEK293 Cells ,Protein Binding ,RNA helicases ,RNA structure ,reporter genes ,translational control ,Developmental Biology ,Biochemistry and cell biology - Abstract
DDX3X regulates the translation of a subset of human transcripts containing complex 5' untranslated regions (5' UTRs). In this study, we developed the helicase activity reporter for translation (HART), which uses DDX3X-sensitive 5' UTRs to measure DDX3X-mediated translational activity in cells. To directly measure RNA structure in DDX3X-dependent mRNAs, we used SHAPE-MaP to determine the secondary structures present in DDX3X-sensitive 5' UTRs and then used HART to investigate how sequence alterations influence DDX3X sensitivity. Additionally, we identified residues 38-44 as potential mediators of DDX3X's interaction with the translational machinery. HART revealed that both DDX3X's association with the translational machinery and its helicase activity are required for its function in promoting the translation of DDX3X-sensitive 5' UTRs. These findings suggest DDX3X plays a crucial role in regulating translation through its interaction with the translational machinery during ribosome scanning and establish the HART reporter as a robust, lentivirally encoded, colorimetric measurement of DDX3X-dependent translation in cells.
- Published
- 2024
40. Position-dependent function of human sequence-specific transcription factors
- Author
-
Duttke, Sascha H, Guzman, Carlos, Chang, Max, Delos Santos, Nathaniel P, McDonald, Bayley R, Xie, Jialei, Carlin, Aaron F, Heinz, Sven, and Benner, Christopher
- Subjects
Biochemistry and Cell Biology ,Bioinformatics and Computational Biology ,Genetics ,Biological Sciences ,Human Genome ,1.1 Normal biological development and functioning ,Humans ,Binding Sites ,Gene Expression Regulation ,Genome ,Human ,Nucleotide Motifs ,Promoter Regions ,Genetic ,Protein Binding ,Transcription Factors ,Transcription Initiation Site ,Transcription Initiation ,Genetic ,Genetic Variation ,General Science & Technology - Abstract
Patterns of transcriptional activity are encoded in our genome through regulatory elements such as promoters or enhancers that, paradoxically, contain similar assortments of sequence-specific transcription factor (TF) binding sites1-3. Knowledge of how these sequence motifs encode multiple, often overlapping, gene expression programs is central to understanding gene regulation and how mutations in non-coding DNA manifest in disease4,5. Here, by studying gene regulation from the perspective of individual transcription start sites (TSSs), using natural genetic variation, perturbation of endogenous TF protein levels and massively parallel analysis of natural and synthetic regulatory elements, we show that the effect of TF binding on transcription initiation is position dependent. Analysing TF-binding-site occurrences relative to the TSS, we identified several motifs with highly preferential positioning. We show that these patterns are a combination of a TF's distinct functional profiles-many TFs, including canonical activators such as NRF1, NFY and Sp1, activate or repress transcription initiation depending on their precise position relative to the TSS. As such, TFs and their spacing collectively guide the site and frequency of transcription initiation. More broadly, these findings reveal how similar assortments of TF binding sites can generate distinct gene regulatory outcomes depending on their spatial configuration and how DNA sequence polymorphisms may contribute to transcription variation and disease and underscore a critical role for TSS data in decoding the regulatory information of our genome.
- Published
- 2024
41. Elf1 promotes transcription-coupled repair in yeast by using its C-terminal domain to bind TFIIH.
- Author
-
Selvam, Kathiresan, Xu, Jun, Wilson, Hannah, Oh, Juntaek, Li, Qingrong, Wang, Dong, and Wyrick, John
- Subjects
Transcription Factor TFIIH ,Saccharomyces cerevisiae Proteins ,DNA Repair ,Saccharomyces cerevisiae ,Protein Binding ,Transcription ,Genetic ,Ultraviolet Rays ,Protein Domains ,RNA Polymerase II ,DNA Damage ,Pyrimidine Dimers ,Excision Repair - Abstract
Transcription coupled-nucleotide excision repair (TC-NER) removes DNA lesions that block RNA polymerase II (Pol II) transcription. A key step in TC-NER is the recruitment of the TFIIH complex, which initiates DNA unwinding and damage verification; however, the mechanism by which TFIIH is recruited during TC-NER, particularly in yeast, remains unclear. Here, we show that the C-terminal domain (CTD) of elongation factor-1 (Elf1) plays a critical role in TC-NER in yeast by binding TFIIH. Analysis of genome-wide repair of UV-induced cyclobutane pyrimidine dimers (CPDs) using CPD-seq indicates that the Elf1 CTD in yeast is required for efficient TC-NER. We show that the Elf1 CTD binds to the pleckstrin homology (PH) domain of the p62 subunit of TFIIH in vitro, and identify a putative TFIIH-interaction region (TIR) in the Elf1 CTD that is important for PH binding and TC-NER. The Elf1 TIR shows functional, structural, and sequence similarities to a conserved TIR in the mammalian UV sensitivity syndrome A (UVSSA) protein, which recruits TFIIH during TC-NER in mammalian cells. These findings suggest that the Elf1 CTD acts as a functional counterpart to mammalian UVSSA in TC-NER by recruiting TFIIH in response to Pol II stalling at DNA lesions.
- Published
- 2024
42. Transcription factor binding specificities of the oomycete Phytophthora infestans reflect conserved and divergent evolutionary patterns and predict function.
- Author
-
Vo, Nguyen, Yang, Ally, Leesutthiphonchai, Wiphawee, Liu, Yulong, Hughes, Timothy, and Judelson, Howard
- Subjects
DNA-binding protein ,Gene regulation ,Oomycete ,Phytophthora infestans ,Promoter ,Protein-binding oligonucleotide microarray ,Transcription factor binding site ,Phytophthora infestans ,Transcription Factors ,Evolution ,Molecular ,Phylogeny ,Binding Sites ,Protein Binding - Abstract
BACKGROUND: Identifying the DNA-binding specificities of transcription factors (TF) is central to understanding gene networks that regulate growth and development. Such knowledge is lacking in oomycetes, a microbial eukaryotic lineage within the stramenopile group. Oomycetes include many important plant and animal pathogens such as the potato and tomato blight agent Phytophthora infestans, which is a tractable model for studying life-stage differentiation within the group. RESULTS: Mining of the P. infestans genome identified 197 genes encoding proteins belonging to 22 TF families. Their chromosomal distribution was consistent with family expansions through unequal crossing-over, which were likely ancient since each family had similar sizes in most oomycetes. Most TFs exhibited dynamic changes in RNA levels through the P. infestans life cycle. The DNA-binding preferences of 123 proteins were assayed using protein-binding oligonucleotide microarrays, which succeeded with 73 proteins from 14 families. Binding sites predicted for representatives of the families were validated by electrophoretic mobility shift or chromatin immunoprecipitation assays. Consistent with the substantial evolutionary distance of oomycetes from traditional model organisms, only a subset of the DNA-binding preferences resembled those of human or plant orthologs. Phylogenetic analyses of the TF families within P. infestans often discriminated clades with canonical and novel DNA targets. Paralogs with similar binding preferences frequently had distinct patterns of expression suggestive of functional divergence. TFs were predicted to either drive life stage-specific expression or serve as general activators based on the representation of their binding sites within total or developmentally-regulated promoters. This projection was confirmed for one TF using synthetic and mutated promoters fused to reporter genes in vivo. CONCLUSIONS: We established a large dataset of binding specificities for P. infestans TFs, representing the first in the stramenopile group. This resource provides a basis for understanding transcriptional regulation by linking TFs with their targets, which should help delineate the molecular components of processes such as sporulation and host infection. Our work also yielded insight into TF evolution during the eukaryotic radiation, revealing both functional conservation as well as diversification across kingdoms.
- Published
- 2024
43. Structural basis for the H2AK119ub1-specific DNMT3A-nucleosome interaction.
- Author
-
Chen, Xinyi, Guo, Yiran, Zhao, Ting, Lu, Jiuwei, Fang, Jian, Wang, Yinsheng, Wang, Gang, and Song, Jikui
- Subjects
Nucleosomes ,DNA Methyltransferase 3A ,DNA (Cytosine-5-)-Methyltransferases ,Histones ,Humans ,DNA Methylation ,Protein Binding ,Cryoelectron Microscopy ,Animals ,Mice ,Ubiquitination ,Polycomb Repressive Complex 2 ,HEK293 Cells ,Models ,Molecular - Abstract
Isoform 1 of DNA methyltransferase DNMT3A (DNMT3A1) specifically recognizes nucleosome monoubiquitylated at histone H2A lysine-119 (H2AK119ub1) for establishment of DNA methylation. Mis-regulation of this process may cause aberrant DNA methylation and pathogenesis. However, the molecular basis underlying DNMT3A1-nucleosome interaction remains elusive. Here we report the cryo-EM structure of DNMT3A1s ubiquitin-dependent recruitment (UDR) fragment complexed with H2AK119ub1-modified nucleosome. DNMT3A1 UDR occupies an extensive nucleosome surface, involving the H2A-H2B acidic patch, a surface groove formed by H2A and H3, nucleosomal DNA, and H2AK119ub1. The DNMT3A1 UDRs interaction with H2AK119ub1 affects the functionality of DNMT3A1 in cells in a context-dependent manner. Our structural and biochemical analysis also reveals competition between DNMT3A1 and JARID2, a cofactor of polycomb repression complex 2 (PRC2), for nucleosome binding, suggesting the interplay between different epigenetic pathways. Together, this study reports a molecular basis for H2AK119ub1-dependent DNMT3A1-nucleosome association, with important implications in DNMT3A1-mediated DNA methylation in development.
- Published
- 2024
44. Proximity Graph Networks: Predicting Ligand Affinity with Message Passing Neural Networks.
- Author
-
Gale-Day, Zachary, Shub, Laura, Chuang, Kangway, and Keiser, Michael
- Subjects
Ligands ,Neural Networks ,Computer ,Proteins ,Molecular Docking Simulation ,Protein Binding - Abstract
Message passing neural networks (MPNNs) on molecular graphs generate continuous and differentiable encodings of small molecules with state-of-the-art performance on protein-ligand complex scoring tasks. Here, we describe the proximity graph network (PGN) package, an open-source toolkit that constructs ligand-receptor graphs based on atom proximity and allows users to rapidly apply and evaluate MPNN architectures for a broad range of tasks. We demonstrate the utility of PGN by introducing benchmarks for affinity and docking score prediction tasks. Graph networks generalize better than fingerprint-based models and perform strongly for the docking score prediction task. Overall, MPNNs with proximity graph data structures augment the prediction of ligand-receptor complex properties when ligand-receptor data are available.
- Published
- 2024
45. Selective regulation of a defined subset of inflammatory and immunoregulatory genes by an NF-κB p50-IκBζ pathway.
- Author
-
Daly, Allison, Yeh, George, Soltero, Sofia, and Smale, Stephen
- Subjects
IκBζ ,NF-κB ,inflammation ,innate immunity ,macrophages ,transcription ,Animals ,Mice ,Adaptor Proteins ,Signal Transducing ,Gene Expression Regulation ,I-kappa B Proteins ,Inflammation ,Macrophages ,NF-kappa B p50 Subunit ,Protein Binding ,Signal Transduction ,Toll-Like Receptor 4 ,Transcription Factor RelA ,Male - Abstract
The five NF-κB family members and three nuclear IκB proteins play important biological roles, but the mechanisms by which distinct members of these protein families contribute to selective gene transcription remain poorly understood, especially at a genome-wide scale. Using nascent transcript RNA-seq, we observed considerable overlap between p50-dependent and IκBζ-dependent genes in Toll-like receptor 4 (TLR4)-activated macrophages. Key immunoregulatory genes, including Il6, Il1b, Nos2, Lcn2, and Batf, are among the p50-IκBζ-codependent genes. IκBζ-bound genomic sites are occupied at earlier time points by NF-κB dimers. However, p50-IκBζ codependence does not coincide with preferential binding of either p50 or IκBζ, as RelA co-occupies hundreds of genomic sites with the two proteins. A common feature of p50-IκBζ-codependent genes is a nearby p50/RelA/IκBζ-cobound site exhibiting p50-dependent binding of both RelA and IκBζ. This and other results suggest that IκBζ acts in concert with RelA:p50 heterodimers. Notably, p50-IκBζ-codependent genes comprise a high percentage of genes exhibiting the greatest differential expression between TLR4-stimulated and tumor necrosis factor receptor (TNFR)-stimulated macrophages. Thus, our genome-centric analysis reveals a defined p50-IκBζ pathway that selectively activates a set of key immunoregulatory genes and serves as an important contributor to differential TNFR and TLR4 responses.
- Published
- 2024
46. How much does TRPV1 deviate from an ideal MWC-type protein?
- Author
-
Li, Shisheng and Zheng, Jie
- Subjects
TRPV Cation Channels ,Ion Channel Gating ,Models ,Molecular ,Ligands ,Protein Subunits ,Animals ,Protein Binding ,Models ,Biological ,Capsaicin - Abstract
Many ion channels are known to behave as an allosteric protein, coupling environmental stimuli captured by specialized sensing domains to the opening of a central pore. The classic Monod-Wyman-Changeux (MWC) model, originally proposed to describe binding of gas molecules to hemoglobin, has been widely used as a framework for analyzing ion channel gating. Here, we address the issue of how accurately the MWC model predicts activation of the capsaicin receptor TRPV1 by vanilloids. Taking advantage of a concatemeric design that makes it possible to lock TRPV1 in states with zero to four bound vanilloid molecules, we showed quantitatively that the overall gating behavior is satisfactorily predicted by the MWC model. There is, however, a small yet detectable subunit position effect: ligand binding to two kitty-corner subunits is 0.3-0.4 kcal/mol more effective in inducing opening than binding to two neighbor subunits. This difference-less than 10% of the overall energetic contribution from ligand binding-might be due to the restriction on subunit arrangement imposed by the planar membrane; if this is the case, then the position effect is not expected in hemoglobin, in which each subunit is related equivalently to all the other subunits.
- Published
- 2024
47. Current State of Open Source Force Fields in Protein-Ligand Binding Affinity Predictions.
- Author
-
Hahn, David, Gapsys, Vytautas, de Groot, Bert, Mobley, David, and Tresadern, Gary
- Subjects
Ligands ,Proteins ,Molecular Dynamics Simulation ,Protein Binding ,Thermodynamics ,Drug Discovery ,Protein Conformation - Abstract
In drug discovery, the in silico prediction of binding affinity is one of the major means to prioritize compounds for synthesis. Alchemical relative binding free energy (RBFE) calculations based on molecular dynamics (MD) simulations are nowadays a popular approach for the accurate affinity ranking of compounds. MD simulations rely on empirical force field parameters, which strongly influence the accuracy of the predicted affinities. Here, we evaluate the ability of six different small-molecule force fields to predict experimental protein-ligand binding affinities in RBFE calculations on a set of 598 ligands and 22 protein targets. The public force fields OpenFF Parsley and Sage, GAFF, and CGenFF show comparable accuracy, while OPLS3e is significantly more accurate. However, a consensus approach using Sage, GAFF, and CGenFF leads to accuracy comparable to OPLS3e. While Parsley and Sage are performing comparably based on aggregated statistics across the whole dataset, there are differences in terms of outliers. Analysis of the force field reveals that improved parameters lead to significant improvement in the accuracy of affinity predictions on subsets of the dataset involving those parameters. Lower accuracy can not only be attributed to the force field parameters but is also dependent on input preparation and sampling convergence of the calculations. Especially large perturbations and nonconverged simulations lead to less accurate predictions. The input structures, Gromacs force field files, as well as the analysis Python notebooks are available on GitHub.
- Published
- 2024
48. Modeling reveals the strength of weak interactions in stacked-ring assembly.
- Author
-
Lagunes, Leonila, Briggs, Koan, Martin-Holder, Paige, Xu, Zaikun, Maurer, Dustin, Ghabra, Karim, and Deeds, Eric
- Subjects
Models ,Molecular ,Protein Multimerization ,Kinetics ,Protein Binding ,Protein Structure ,Quaternary - Abstract
Cells employ many large macromolecular machines for the execution and regulation of processes that are vital for cell and organismal viability. Interestingly, cells cannot synthesize these machines as functioning units. Instead, cells synthesize the molecular parts that must then assemble into the functional complex. Many important machines, including chaperones such as GroEL and proteases such as the proteasome, comprise protein rings that are stacked on top of one another. While there is some experimental data regarding how stacked-ring complexes such as the proteasome self-assemble, a comprehensive understanding of the dynamics of stacked-ring assembly is currently lacking. Here, we developed a mathematical model of stacked-trimer assembly and performed an analysis of the assembly of the stacked homomeric trimer, which is the simplest stacked-ring architecture. We found that stacked rings are particularly susceptible to a form of kinetic trapping that we term deadlock, in which the system gets stuck in a state where there are many large intermediates that are not the fully assembled structure but that cannot productively react. When interaction affinities are uniformly strong, deadlock severely limits assembly yield. We thus predicted that stacked rings would avoid situations where all interfaces in the structure have high affinity. Analysis of available crystal structures indicated that indeed the majority-if not all-of stacked trimers do not contain uniformly strong interactions. Finally, to better understand the origins of deadlock, we developed a formal pathway analysis and showed that, when all the binding affinities are strong, many of the possible pathways are utilized. In contrast, optimal assembly strategies utilize only a small number of pathways. Our work suggests that deadlock is a critical factor influencing the evolution of macromolecular machines and provides general principles for understanding the self-assembly efficiency of existing machines.
- Published
- 2024
49. The Chlamydia trachomatis Inc Tri1 interacts with TRAF7 to displace native TRAF7 interacting partners.
- Author
-
Herrera, Clara, McMahon, Eleanor, Swaney, Danielle, Sherry, Jessica, Pha, Khavong, Adams-Boone, Kathleen, Johnson, Jeffrey, Krogan, Nevan, Stevers, Meredith, Solomon, David, Elwell, Cherilyn, and Engel, Joanne
- Subjects
Chlamydia trachomatis ,MEKK2 ,MEKK3 ,TRAF7 ,WD40 ,host-pathogen interaction ,inclusion membrane protein ,mass spectrometry ,Humans ,Chlamydia trachomatis ,Host-Pathogen Interactions ,HeLa Cells ,Bacterial Proteins ,Chlamydia Infections ,Signal Transduction ,Tumor Necrosis Factor Receptor-Associated Peptides and Proteins ,Immunity ,Innate ,Protein Binding ,Membrane Proteins ,HEK293 Cells - Abstract
Chlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and of preventable blindness worldwide. This obligate intracellular pathogen replicates within a membrane-bound inclusion, but how it acquires nutrients from the host while avoiding detection by the innate immune system is incompletely understood. C. trachomatis accomplishes this in part through the translocation of a unique set of effectors into the inclusion membrane, the inclusion membrane proteins (Incs). Incs are ideally positioned at the host-pathogen interface to reprogram host signaling by redirecting proteins or organelles to the inclusion. Using a combination of co-affinity purification, immunofluorescence confocal imaging, and proteomics, we characterize the interaction between an early-expressed Inc of unknown function, Tri1, and tumor necrosis factor receptor-associated factor 7 (TRAF7). TRAF7 is a multi-domain protein with a RING finger ubiquitin ligase domain and a C-terminal WD40 domain. TRAF7 regulates several innate immune signaling pathways associated with C. trachomatis infection and is mutated in a subset of tumors. We demonstrate that Tri1 and TRAF7 specifically interact during infection and that TRAF7 is recruited to the inclusion. We further show that the predicted coiled-coil domain of Tri1 is necessary to interact with the TRAF7 WD40 domain. Finally, we demonstrate that Tri1 displaces the native TRAF7 binding partners, mitogen-activated protein kinase kinase kinase 2 (MEKK2), and MEKK3. Together, our results suggest that by displacing TRAF7 native binding partners, Tri1 has the capacity to alter TRAF7 signaling during C. trachomatis infection.IMPORTANCEChlamydia trachomatis is the leading cause of bacterial sexually transmitted infections in the USA and preventable blindness worldwide. Although easily treated with antibiotics, the vast majority of infections are asymptomatic and therefore go untreated, leading to infertility and blindness. This obligate intracellular pathogen evades the immune response, which contributes to these outcomes. Here, we characterize the interaction between a C. trachomatis-secreted effector, Tri1, and a host protein involved in innate immune signaling, TRAF7. We identified host proteins that bind to TRAF7 and demonstrated that Tri1 can displace these proteins upon binding to TRAF7. Remarkably, the region of TRAF7 to which these host proteins bind is often mutated in a subset of human tumors. Our work suggests a mechanism by which Tri1 may alter TRAF7 signaling and has implications not only in the pathogenesis of C. trachomatis infections but also in understanding the role of TRAF7 in cancer.
- Published
- 2024
50. Tandem LIM domain-containing proteins, LIMK1 and LMO1, directly bind to force-bearing keratin intermediate filaments
- Author
-
Kim, Dah Som, Cheah, Joleen S, Lai, Tzu Wei, Zhao, Karen X, Foust, Skylar R, Lee, Yuh-Ru Julie, Lo, Su Hao, Heinrich, Volkmar, and Yamada, Soichiro
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,LIM Domain Proteins ,Humans ,Lim Kinases ,Keratins ,Intermediate Filaments ,Protein Binding ,Animals ,Transcription Factors ,CP: Molecular biology ,FRAP ,LIM domain ,LIMK1 ,LMO1 ,actin ,cell stretch ,cytoskeleton ,fluorescence recovery after photobleaching ,intermediate filament ,keratin ,mechanotransduction ,Medical Physiology ,Biological sciences - Abstract
The cytoskeleton of the cell is constantly exposed to physical forces that regulate cellular functions. Selected members of the LIM (Lin-11, Isl-1, and Mec-3) domain-containing protein family accumulate along force-bearing actin fibers, with evidence supporting that the LIM domain is solely responsible for this force-induced interaction. However, LIM domain's force-induced interactions are not limited to actin. LIMK1 and LMO1, both containing only two tandem LIM domains, are recruited to force-bearing keratin fibers in epithelial cells. This unique recruitment is mediated by their LIM domains and regulated by the sequences outside the LIM domains. Based on in vitro reconstitution of this interaction, LIMK1 and LMO1 directly interact with stretched keratin 8/18 fibers. These results show that LIM domain's mechano-sensing abilities extend to the keratin cytoskeleton, highlighting the diverse role of LIM proteins in force-regulated signaling.
- Published
- 2024
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.