412 results on '"Pseudokinase"'
Search Results
2. Discovery of CZY43 as a new small-molecule degrader of pseudokinase HER3
- Author
-
Chen, Zhiyuan, He, Rui, Huang, Shengjie, Zhou, Yang, Zhang, Zhang, Wang, Zhen, and Ding, Ke
- Published
- 2025
- Full Text
- View/download PDF
3. Protein shapeshifting in necroptotic cell death signaling
- Author
-
Hoblos, Hanadi, Cawthorne, Wayne, Samson, André L., and Murphy, James M.
- Published
- 2024
- Full Text
- View/download PDF
4. Molecular basis of JAK kinase regulation guiding therapeutic approaches: Evaluating the JAK3 pseudokinase domain as a drug target
- Author
-
Virtanen, Anniina, Kettunen, Vivian, Musta, Kirsikka, Räkköläinen, Veera, Knapp, Stefan, Haikarainen, Teemu, and Silvennoinen, Olli
- Published
- 2024
- Full Text
- View/download PDF
5. The role of tribbles homolog 2 in cell proliferation.
- Author
-
Zhang, Wenkang, Li, Mingkang, Zhang, Minhao, Yan, Gaoliang, and Tang, Chengchun
- Subjects
- *
PROTEIN kinase B , *CELL communication , *MITOGEN-activated protein kinases , *VASCULAR smooth muscle , *MEDICAL sciences , *UBIQUITINATION - Abstract
Tribbles homolog 2 (TRIB2), a pseudoserine/threonine kinase, is a member of the TRIB family. TRIB2 primarily regulates cell proliferation through its scaffold or adaptor effect on promoting the degradation of target proteins by E3 ligase-dependent ubiquitination and regulating mitogen-activated protein kinase (MAPK) and protein kinase B (AKT) signaling pathways. TRIB2 is not only involved in the physiological proliferation of cells (granulosa cells, myoblasts, naive T cells, and thymocytes) during normal development but also in the pathological proliferation of vascular smooth muscle cells and a variety of cancer cells (lung cancer cells, liver cancer cells, leukemia cells, pancreatic cancer cells, gastric cancer cells, prostate cancer cells, thyroid cancer cells, cervical cancer cells, melanoma cells, colorectal cancer cells, ovarian cancer cells and osteosarcoma cells) under disease conditions. Its expression level and functional role predominantly hinge on the specific tissue and cell type it targets. This review elucidates the specific mechanisms of TRIB2 in physiological and pathological cell proliferation from the perspective of different kinds of cells. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
6. The expanding landscape of canonical and non-canonical protein phosphorylation.
- Author
-
Houles, Thibault, Yoon, Sang-Oh, and Roux, Philippe P.
- Subjects
- *
PHOSPHOPROTEIN phosphatases , *PROTEIN kinases , *CELL communication , *MOLECULAR switches , *CATALYTIC activity - Abstract
The phosphoproteome is vast and intricate, with many aspects of its extent and site-specific functional roles still largely unexplored. While traditional studies have primarily focused on Ser/Thr and Tyr phosphorylation, new methodologies are emerging to study non-canonical phosphorylation events. Protein phosphatases play critical roles in cellular signaling, with recent studies highlighting their complexity and involvement in various biological processes. A significant portion of the kinome, referred to as the 'dark' kinome, remains understudied and holds great potential for novel discoveries. Pseudokinases and pseudophosphatases, despite lacking catalytic activity, play crucial roles in cell signaling. Recent advances in experimental and computational analyses have improved the understanding of kinase–substrate and phosphatase–substrate interactions. Protein phosphorylation is a crucial regulatory mechanism in cell signaling, acting as a molecular switch that modulates protein function. Catalyzed by protein kinases and reversed by phosphoprotein phosphatases, it is essential in both normal physiological and pathological states. Recent advances have uncovered a vast and intricate landscape of protein phosphorylation that include histidine phosphorylation and more unconventional events, such as pyrophosphorylation and polyphosphorylation. Many questions remain about the true size of the phosphoproteome and, more importantly, its site-specific functional relevance. The involvement of unconventional actors such as pseudokinases and pseudophosphatases adds further complexity to be resolved. This review explores recent discoveries and ongoing challenges, highlighting the need for continued research to fully elucidate the roles and regulation of protein phosphorylation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. NRBP1 promotes malignant phenotypes of glioblastoma by regulating PI3K/Akt activation.
- Author
-
Zhang, Anli, Peng, Shichao, Sun, Sibai, Ye, Shan, Zhao, Ye, and Wu, Qiang
- Subjects
- *
PI3K/AKT pathway , *CELL migration , *TUMOR growth , *GENE expression , *CYTOLOGY - Abstract
Objectives: Glioblastoma (GBM) is the most aggressive of intracranial gliomas. Despite the maximal treatment intervention, the median survival rate is still about 14–16 months. Nuclear receptor‐binding protein 1 (NRBP1) has a potential growth‐promoting role on biology function of cells. In this study, we investigated whether NRBP1 promotes GBM malignant phenotypes and the potential mechanisms. Methods: The correlation between NRBP1 and glioma grade, prognosis in TCGA/CGGA databases and our clinical data were analyzed. Next, we conducted knockout and overexpression of NRBP1 on GBM cells to verify that NRBP1 promoted cell proliferation, invasion, and migration in vitro and in vivo. Finally, we detected the impact of NRBP1 on PI3K/Akt signaling pathway and EMT. Results: There was a correlation between elevated NRBP1 expression and advanced stage glioma, as well as decreased overall and disease‐free survival. The suppression of proliferation, invasion, and migration of tumor cells was observed upon NRBP1 knockout, and in vitro studies also demonstrated the induction of apoptotic cell death. Whereas, its overexpression is associated with high multiplication rate, migration, invasion, and apoptotic escape. GO enrichment and KEGG analysis revealed that NRBP1 regulated differentially expressed gene clusters are involved in PI3K/Akt signaling pathway, as well as EMT mediated by this pathway. Moreover, the effects of NRBP1 knockdown and overexpression on GBM were mitigated by MK‐2206 and SC79, both of which respectively function as an inhibitor and an activator of the PI3K/Akt signaling pathway. Similarly, the suppression of NRBP1 led to a decrease in tumor growth, whereas its overexpression promoted tumor growth in a mouse model. Conclusions: This study shows that NRBP1 promotes malignant phenotypes in GBM by activating PI3K/Akt pathway. Hence, it can function as both a predictive indicator and a new target for therapies in GBM treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Enzyme Is the Name—Adapter Is the Game.
- Author
-
Huber, Michael and Brummer, Tilman
- Subjects
- *
BRUTON tyrosine kinase , *SCAFFOLD proteins , *MITOGEN-activated protein kinases , *PHOSPHATIDYLINOSITOL 3-kinases , *CATALYTIC domains - Abstract
Signaling proteins in eukaryotes usually comprise a catalytic domain coupled to one or several interaction domains, such as SH2 and SH3 domains. An additional class of proteins critically involved in cellular communication are adapter or scaffold proteins, which fulfill their purely non-enzymatic functions by organizing protein–protein interactions. Intriguingly, certain signaling enzymes, e.g., kinases and phosphatases, have been demonstrated to promote particular cellular functions by means of their interaction domains only. In this review, we will refer to such a function as "the adapter function of an enzyme". Though many stories can be told, we will concentrate on several proteins executing critical adapter functions in cells of the immune system, such as Bruton´s tyrosine kinase (BTK), phosphatidylinositol 3-kinase (PI3K), and SH2-containing inositol phosphatase 1 (SHIP1), as well as in cancer cells, such as proteins of the rat sarcoma/extracellular signal-regulated kinase (RAS/ERK) mitogen-activated protein kinase (MAPK) pathway. We will also discuss how these adaptor functions of enzymes determine or even undermine the efficacy of targeted therapy compounds, such as ATP-competitive kinase inhibitors. Thereby, we are highlighting the need to develop pharmacological approaches, such as proteolysis-targeting chimeras (PROTACs), that eliminate the entire protein, and thus both enzymatic and adapter functions of the signaling protein. We also review how genetic knock-out and knock-in approaches can be leveraged to identify adaptor functions of signaling proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. KINtaro: protein kinase-like database
- Author
-
Baranowski, Bartosz, Krysińska, Marianna, and Gradowski, Marcin
- Published
- 2024
- Full Text
- View/download PDF
10. "Oh, Dear We Are in Tribble": An Overview of the Oncogenic Functions of Tribbles 1.
- Author
-
Singh, Karnika, Showalter, Christian A., Manring, Heather R., Haque, Saikh Jaharul, and Chakravarti, Arnab
- Subjects
- *
LIPID metabolism , *GLIOMAS , *DRUG resistance in cancer cells , *CELL proliferation , *BREAST tumors , *ANTINEOPLASTIC agents , *ACUTE promyelocytic leukemia , *TUMOR markers , *CELLULAR signal transduction , *ATHEROSCLEROSIS , *PROSTATE tumors , *COLORECTAL cancer , *SMALL molecules , *GENE expression , *ONCOGENES , *INFLAMMATION , *CELL survival , *LUNG cancer , *CELL differentiation , *IMMUNITY , *HEPATOCELLULAR carcinoma - Abstract
Simple Summary: Pseudokinases have evolved from conventional kinases and differ from them in their inability to phosphorylate their substrates. The Tribbles pseudokinases have evolved from the CAMK family of kinases and include three members: Tribbles 1, 2, and 3. Tribbles 1, which evolved later in metazoan lineage, appears to have a regulatory function. It interacts with a variety of substrates through different domains embedded in its molecular structure. Each interaction regulates cellular processes involved in cell division, survival, and metabolism. These processes also extrapolate to cancer development and other diseases. Based on its involvement in disorders and therapy resistance, it can be considered a potential candidate for drug development. The improved knowledge about its function can be utilized to design small-molecule inhibitors against TRIB1. Pseudokinases are catalytically inactive proteins in the human genome that lack the ability to transfer phosphate from ATP to their substrates. The Tribbles family of pseudokinases contains three members: Tribbles 1, 2, and 3. Tribbles 1 has recently gained importance because of its involvement in various diseases, including cancer. It acts as a scaffolding protein that brings about the degradation of its substrate proteins, such as C/EBPα/β, MLXIPL, and RAR/RXRα, among others, via the ubiquitin proteasome system. It also serves as an adapter protein, which sequesters different protein molecules and activates their downstream signaling, leading to processes, such as cell survival, cell proliferation, and lipid metabolism. It has been implicated in cancers such as AML, prostate cancer, breast cancer, CRC, HCC, and glioma, where it activates oncogenic signaling pathways such as PI3K-AKT and MAPK and inhibits the anti-tumor function of p53. TRIB1 also causes treatment resistance in cancers such as NSCLC, breast cancer, glioma, and promyelocytic leukemia. All these effects make TRIB1 a potential drug target. However, the lack of a catalytic domain renders TRIB1 "undruggable", but knowledge about its structure, conformational changes during substrate binding, and substrate binding sites provides an opportunity to design small-molecule inhibitors against specific TRIB1 interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Deep dental phenotyping and a novel FAM20A variant in patients with amelogenesis imperfecta type IG.
- Author
-
Sriwattanapong, Kanokwan, Theerapanon, Thanakorn, Khamwachirapitak, Chompak, Sae‐ear, Pannagorn, Srijunbarl, Anucharte, Porntaveetus, Thantrira, and Shotelersuk, Vorasuk
- Subjects
- *
DENTAL pulp diseases , *AMELOGENESIS imperfecta , *GENETIC mutation , *MOLARS , *PERIODONTITIS , *GENETIC variation , *PERIODONTAL disease , *MALOCCLUSION , *GENE expression , *GENOMICS , *GINGIVAL hyperplasia , *RESEARCH funding , *DENTAL enamel , *CELL separation , *KIDNEY calcification , *PHENOTYPES - Abstract
Objectives: To identify etiologic variants and perform deep dental phenotyping in patients with amelogenesis imperfecta (AI). Methods: Three patients of two unrelated families were evaluated. Genetic variants were investigated by exome and Sanger sequencing. An unerupted permanent third molar (AI1) from Patient1 and a deciduous first molar (AI2) from Patient2, along with three tooth‐type matched controls for each were characterized. Results: All three patients harbored biallelic pathogenic variants in FAM20A, indicating AI1G. Of the four identified variants, one, c.1231C > T p.(Arg411Trp), was novel. Patient1 possessed the largest deletion, 7531 bp, ever identified in FAM20A. In addition to hypoplastic enamel, multiple impacted teeth, intrapulpal calcification, pericoronal radiolucencies, malocclusion, and periodontal infections were found in all three patients, gingival hyperplasia in Patient1 and Patient2, and alveolar bone exostosis in Patient3. Surface roughness was increased in AI1 but decreased in AI2. Decreased enamel mineral density, hardness, and elastic modulus were observed in AI1 enamel and dentin and AI2 dentin, along with decreased phosphorus, increased carbon, and increased calcium/phosphorus and carbon/oxygen ratios. Severely collapsed enamel rods and disorganized dentin–enamel junction were observed. Conclusions: We report a novel FAM20A variant and, for the first time, the defective mineral composition and physical/mechanical properties of AI1G teeth. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. PD-L1 expression is regulated by ATP-binding of the ERBB3 pseudokinase domain
- Author
-
Yamu Li, Zhonghua Liu, Yiqing Zhao, Jie Yang, Tsan Sam Xiao, Ronald A. Conlon, and Zhenghe Wang
- Subjects
Colon cancer ,ERBB3 ,Immunotherapy ,PD-L1 ,Pseudokinase ,Medicine (General) ,R5-920 ,Genetics ,QH426-470 - Abstract
How PD-L1 expression is regulated in cancer is poorly understood. Here, we report that the ATP-binding activity of ERBB3 pseudokinase regulates PD-L1 gene expression in colorectal cancers (CRCs). ERBB3 is one of the four members of the EGF receptor family, all with protein tyrosine kinase domains. ERBB3 is a pseudokinase with a high binding affinity to ATP. We showed that ERBB3 ATP-binding inactivation mutant reduces tumorigenicity in genetically engineered mouse models and impairs xenograft tumor growth of CRC cell lines. The ERBB3 ATP-binding mutant cells dramatically reduce IFN-γ-induced PD-L1 expression. Mechanistically, ERBB3 regulates IFN-γ-induced PD-L1 expression through the IRS1-PI3K-PDK1-RSK-CREB signaling axis. CREB is the transcription factor that regulates PD-L1 gene expression in CRC cells. Knockin of a tumor-derived ERBB3 mutation located in the kinase domain sensitizes mouse colon cancers to anti-PD1 antibody therapy, suggesting that ERBB3 mutations could be predictive biomarkers for tumors amenable to immune checkpoint therapy.
- Published
- 2023
- Full Text
- View/download PDF
13. Ulk4 promotes Shh signaling by regulating Stk36 ciliary localization and Gli2 phosphorylation
- Author
-
Mengmeng Zhou, Yuhong Han, and Jin Jiang
- Subjects
Hedgehog ,Ulk4 ,pseudokinase ,Gli2 ,primary cilium ,Stk36 ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis through the Gli family of transcription factors. Gli is thought to be activated at the tip of primary cilium, but the underlying mechanism has remained poorly understood. Here, we show that Unc-51-like kinase 4 (Ulk4), a pseudokinase and a member of the Ulk kinase family, acts in conjunction with another Ulk family member Stk36 to promote Gli2 phosphorylation and Hh pathway activation. Ulk4 interacts with Stk36 through its N-terminal region containing the pseudokinase domain and with Gli2 via its regulatory domain to bridge the kinase and substrate. Although dispensable for Hh-induced Stk36 kinase activation, Ulk4 is essential for Stk36 ciliary tip localization, Gli2 phosphorylation, and activation. In response to Hh, both Ulk4 and Stk36 colocalize with Gli2 at ciliary tip, and Ulk4 and Stk36 depend on each other for their ciliary tip accumulation. We further show that ciliary localization of Ulk4 depends on Stk36 kinase activity and phosphorylation of Ulk4 on Thr1023, and that ciliary tip accumulation of Ulk4 is essential for its function in the Hh pathway. Taken together, our results suggest that Ulk4 regulates Hh signaling by promoting Stk36-mediated Gli2 phosphorylation and activation at ciliary tip.
- Published
- 2023
- Full Text
- View/download PDF
14. 假激酶在细胞信号转导中的功能.
- Author
-
伍 铤 and 王 韵
- Abstract
Protein kinases regulate signaling pathways in eukaryotic cells. The phosphorylation function of kinases has attracted the attention of scientists and been deeply studied. However, some kinases are called “pseudokinases” due to missing of the key amino acids in the key motif during evolution, which leads to disruption of their phosphoryl-transfer activity. The fact that these pseudokinases are conserved across species argues that although pseudokinases have lost their phosphoryl-transfer activity, they may acquire the phosphorylation-independent functions and participate in the cellular signal transduction as well. The phosphorylation-independent functions of pseudokinases are listed as follows: 1. Directly regulating the phosphorylation activity of active kinases by binding with them; 2. Regulating the phosphorylation activity of active kinases by competitive inhibition; 3. Acting as molecular switches to perform key non-catalytic functions; 4. Acting as signaling scaffolds to assist in cell signaling; 5. Exerting catalytic functions other than phosphorylation activity. These functions have been shown to be involved in important physiological processes such as development, immunity and metabolism. Mutations or abnormal expression of pseudokinases are associated with the occurrence of various diseases such as tumors, neurological diseases, metabolic diseases and autoimmune diseases. In this article, we summarize the functions of pseudokinases in cellular signal transduction and provide a perspective for the future directions in the study of pseudokinases. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
15. A novel chemical attack on Notch-mediated transcription by targeting the NACK ATPase
- Author
-
Giulia Diluvio, Tanya T. Kelley, Mohini Lahiry, Annamil Alvarez-Trotta, Ellen M. Kolb, Elena Shersher, Luisana Astudillo, Rhett A. Kovall, Stephan C. Schürer, and Anthony J. Capobianco
- Subjects
small-molecule inhibitor ,ATPase ,Notch signaling ,pseudokinase ,SGK223 ,esophageal adenocarcinoma ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Notch activation complex kinase (NACK) is a component of the Notch transcriptional machinery critical for the Notch-mediated tumorigenesis. However, the mechanism through which NACK regulates Notch-mediated transcription is not well understood. Here, we demonstrate that NACK binds and hydrolyzes ATP and that only ATP-bound NACK can bind to the Notch ternary complex (NTC). Considering this, we sought to identify inhibitors of this ATP-dependent function and, using computational pipelines, discovered the first small-molecule inhibitor of NACK, Z271-0326, that directly blocks the activity of Notch-mediated transcription and shows potent antineoplastic activity in PDX mouse models. In conclusion, we have discovered the first inhibitor that holds promise for the efficacious treatment of Notch-driven cancers by blocking the Notch activity downstream of the NTC.
- Published
- 2023
- Full Text
- View/download PDF
16. Structure and function of the pseudokinases IRAK2 and IRAK3 and their potential as drug targets
- Author
-
Lange, Sven Manfred, Cohen, Philip, Kulathu, Yogesh, and Nelen, Marina
- Subjects
572 ,pseudokinase ,IRAK ,TRAF6 ,Drug discovery ,crystallography ,Structural biology ,Immunity - Abstract
The Toll-like receptors (TLRs) and interleukin-1 receptors (IL-1R) are critical components of the innate immune system that recruit the adapter protein MyD88 (myeloid differentiation primary response gene 88), which serves as an assembly platform for members of the interleukin-1 receptor associated kinase (IRAK) family to form an oligomeric signalling complex called the Myddosome. Subsequently, IRAK kinases and pseudokinases recruit and activate a number of E3 ubiquitin ligases that trigger downstream signalling events to induce the production of pro- and anti-inflammatory mediators. The emergence of crystal structures and development of specific inhibitors of the catalytically active IRAK family members, IRAK1 and IRAK4, have advanced our understanding of their roles in the TLR/IL-1R signalling pathway. However, there is no structural or mechanistic information about the two IRAK pseudokinases, IRAK2 and IRAK3. For this reason, I focused my research on the structural characterisation of the IRAK pseudokinases, to decode their roles within MyD88-dependent signalling. Chapter 3 is focused on the pseudokinase domains of IRAK2 and IRAK3. Chapter 4 describes the identification and characterisation of small molecules that bind to IRAK2 and IRAK3, to evaluate their potential as drug targets. Chapter 5 centres on the interaction of the E3 ubiquitin ligase TRAF6 with IRAK2 and IRAK3. I discovered that the kinase domain of IRAK2 is monomeric in solution, while IRAK3 is in a concentration-dependent equilibrium of monomeric and dimeric states. ATP-binding assays revealed that IRAK2 binds ATP in a Mg-dependent fashion, while IRAK3 does not bind ATP, but binds to the ATP-competitive pankinase inhibitor staurosporine. I present the first crystal structure of the pseudokinase domain of human IRAK3, which reveals a pseudoactive conformation and provides support for IRAK3's inability to bind ATP. IRAK3 forms a hexamer in the crystal via two alternating pseudosymmetric homo-dimer interfaces: The αC-to-αC interface, which centres on the αC-helices of IRAK3, and the αG-to-αG interface with central αG-helices. Structure-guided point-mutations in the αC-to-αC interface identified this interface to form the in-solution dimer of IRAK3. Interestingly, I found that a higher-order assembly of IRAK3 pseudokinase domains with alternating αC-to-αC and αG-to-αG interfaces would create a helical IRAK3 filament. Strikingly, I identified a third putative interface on the IRAK3 surface in a conservation analysis of vertebrate IRAK3 sequences. The third interface is centred around the αEF-helix of IRAK3 and closely resembles the αEF-to-αEF interface of the previously reported IRAK4 homo-dimer. Mutations in IRAK3 that are linked to the pathogenesis of early-onset persistent asthma lie in the putative αEF-interface of IRAK3. Intriguingly, the three distinct dimerisation interfaces on the surface of the pseudokinase domain of IRAK3 do not overlap and allowed for the creation of a higher-order assembly model of IRAK3 and IRAK4 molecules. The model resembles a potential IRAK3-mediated inhibited state of the IRAK4 kinase domain, which would be a novel mechanism of kinase inhibition by a pseudokinase oligomer. Furthermore, these findings demonstrate for the first time that the ability of the Myddosome to signal may be tightly regulated by oligomerisation of the kinase and pseudokinase domains of the IRAKs. In collaboration with Janssen Pharmaceutica, I performed a high-throughput assay to screen for small molecule binders of the IRAK pseudokinases. I identified and started to characterise twelve small molecules that bind to the IRAK3 pseudokinase domain with nanomolar affinities. Lastly, I detected a highly conserved helical region in the C-terminal domains of IRAK1 and IRAK2, which might be the first clue for a function of the C-terminal domains of the IRAKs in addition to their role in TRAF6-binding. I investigated the interaction between TRAF6 and the C-terminal domains of IRAKs and developed a high-throughput HTRF assay for the identification of protein-protein interaction inhibitors to disrupt this interaction. Together, this work lays the foundation to explore the IRAK pseudokinases as potential drug targets for the treatment of innate immune diseases in the future.
- Published
- 2020
17. ALPK3 Functions as a Pseudokinase.
- Author
-
Feng, Wei, Bogomolovas, Julius, Wang, Li, Li, Mengchen, and Chen, Ju
- Subjects
- *
CARDIOMYOPATHIES , *PHOSPHORYLATION , *CRISPRS , *GENES , *MICE - Abstract
The article discusses the ALPK3 gene and its potential role in cardiomyopathies. While there are conflicting findings regarding ALPK3's kinase activity, recent studies suggest that ALPK3 functions as a pseudokinase rather than a true kinase. The article presents evidence from in vitro and in vivo experiments, including an in vitro phosphorylation assay and the generation of knock-in mice with a mutated ALPK3 gene. These findings suggest that ALPK3 variants associated with cardiomyopathy may affect other aspects of the protein rather than its kinase activity. [Extracted from the article]
- Published
- 2023
- Full Text
- View/download PDF
18. PEAK3/C19orf35 pseudokinase, a new NFK3 kinase family member, inhibits CrkII through dimerization
- Author
-
Lopez, Mitchell L, Lo, Megan, Kung, Jennifer E, Dudkiewicz, Małgorzata, Jang, Gwendolyn M, Von Dollen, John, Johnson, Jeffrey R, Krogan, Nevan J, Pawłowski, Krzysztof, and Jura, Natalia
- Subjects
Aetiology ,1.1 Normal biological development and functioning ,2.1 Biological and endogenous factors ,Underpinning research ,Generic health relevance ,Cancer ,Amino Acid Motifs ,Amino Acid Sequence ,Animals ,COS Cells ,Cell Membrane ,Cell Shape ,Chlorocebus aethiops ,Conserved Sequence ,Cytoskeletal Proteins ,Evolution ,Molecular ,HEK293 Cells ,Humans ,Phylogeny ,Protein Binding ,Protein Domains ,Protein Interaction Mapping ,Protein Multimerization ,Protein-Tyrosine Kinases ,Proto-Oncogene Proteins c-crk ,protein kinase ,pseudokinase ,NKF3 family ,CrkII ,motility - Abstract
Members of the New Kinase Family 3 (NKF3), PEAK1/SgK269 and Pragmin/SgK223 pseudokinases, have emerged as important regulators of cell motility and cancer progression. Here, we demonstrate that C19orf35 (PEAK3), a newly identified member of the NKF3 family, is a kinase-like protein evolutionarily conserved across mammals and birds and a regulator of cell motility. In contrast to its family members, which promote cell elongation when overexpressed in cells, PEAK3 overexpression does not have an elongating effect on cell shape but instead is associated with loss of actin filaments. Through an unbiased search for PEAK3 binding partners, we identified several regulators of cell motility, including the adaptor protein CrkII. We show that by binding to CrkII, PEAK3 prevents the formation of CrkII-dependent membrane ruffling. This function of PEAK3 is reliant upon its dimerization, which is mediated through a split helical dimerization domain conserved among all NKF3 family members. Disruption of the conserved DFG motif in the PEAK3 pseudokinase domain also interferes with its ability to dimerize and subsequently bind CrkII, suggesting that the conformation of the pseudokinase domain might play an important role in PEAK3 signaling. Hence, our data identify PEAK3 as an NKF3 family member with a unique role in cell motility driven by dimerization of its pseudokinase domain.
- Published
- 2019
19. JAK1 Pseudokinase V666G Mutant Dominantly Impairs JAK3 Phosphorylation and IL-2 Signaling.
- Author
-
Grant, Alice H., Rodriguez, Alejandro C., Rodriguez Moncivais, Omar J., Sun, Shengjie, Li, Lin, Mohl, Jonathon E., Leung, Ming-Ying, Kirken, Robert A., and Rodriguez, Georgialina
- Subjects
- *
JANUS kinases , *PHOSPHORYLATION , *INTERLEUKIN-2 - Abstract
Overactive Janus kinases (JAKs) are known to drive leukemia, making them well-suited targets for treatment. We sought to identify new JAK-activating mutations and instead found a JAK1-inactivating pseudokinase mutation, V666G. In contrast to other pseudokinase mutations that canonically lead to an active kinase, the JAK1 V666G mutation led to under-activation seen by reduced phosphorylation. To understand the functional role of JAK1 V666G in modifying kinase activity we investigated its influence on other JAK kinases and within the Interleukin-2 pathway. JAK1 V666G not only inhibited its own activity, but its presence could inhibit other JAK kinases. These findings provide new insights into the potential of JAK1 pseudokinase to modulate its own activity, as well as of other JAK kinases. Thus, the features of the JAK1 V666 region in modifying JAK kinases can be exploited to allosterically inhibit overactive JAKs. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
20. Discovery of the first selective and potent PROTAC degrader for the pseudokinase TRIB2.
- Author
-
Wen, Chaowei, Gajjala, Prathibha R., Liu, Yihan, Chen, Bingzhong, Bal, Mehtab S., Sutaria, Payal, Yuanyuan, Qiao, Zheng, Yang, Zhou, Yang, Zhang, Jinwei, Huang, Weixue, Ren, Xiaomei, Wang, Zhen, Ding, Ke, Chinnaiyan, Arul M., and Zhou, Fengtao
- Subjects
- *
PROTEOLYSIS , *INHIBITION of cellular proliferation , *CANCER cells , *PROTEIN kinases , *PROSTATE cancer - Abstract
Pseudokinase TRIB2, a member of the CAMK Ser/Thr protein kinase family, regulates various cellular processes through phosphorylation-independent mechanisms. Dysregulation of TRIB2 has been implicated in promoting tumor growth, metastasis, and therapy resistance, making it a promising target for cancer treatment. In this study, we designed and synthesized a series of TRIB2 PROTAC degraders by conjugating a TRIB2 binder 1 with VHL or CRBN ligands via linkers of varying lengths and compositions. Among these compounds, 5k demonstrated potent TRIB2 degradation with a DC 50 value of 16.84 nM (95 % CI: 13.66–20.64 nM) in prostate cancer PC3 cells. Mechanistic studies revealed that 5k directly interacted with TRIB2, selectively inducing its degradation through a CRBN-dependent ubiquitin-proteasomal pathway. Moreover, 5k outperformed the TRIB2 binder alone in inhibiting cell proliferation and inducing apoptosis, confirming that TRIB2 protein degradation could be a promising therapeutic strategy for TRIB2-associated cancers. Additionally, compound 5k also serves as an effective tool for probing TRIB2 biology. [Display omitted] • A potent TRIB2 degrader 5k was designed, synthesized and evaluated. • Compound 5k potently degrades TRIB2 with a DC 50 of 16.84 nM in PC3 cell lines. • Compound 5k induces TRIB2 degradation through ubiquitin-proteasomal pathways. • Compound 5k exhibited good antiproliferative activities against prostate cancer cells. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
21. The pseudokinase TRIB1 toggles an intramolecular switch to regulate COP1 nuclear export
- Author
-
Kung, Jennifer E and Jura, Natalia
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Active Transport ,Cell Nucleus ,Amino Acid Sequence ,Cell Nucleus ,HEK293 Cells ,Humans ,Intracellular Signaling Peptides and Proteins ,Karyopherins ,Protein Binding ,Protein Serine-Threonine Kinases ,Receptors ,Cytoplasmic and Nuclear ,Sequence Homology ,Ubiquitin ,Ubiquitin-Protein Ligases ,Ubiquitination ,WD40 Repeats ,COP1 ,E3 ubiquitin ligase ,nuclear export ,pseudokinase ,Tribbles ,Information and Computing Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
COP1 is a highly conserved ubiquitin ligase that regulates diverse cellular processes in plants and metazoans. Tribbles pseudokinases, which only exist in metazoans, act as scaffolds that interact with COP1 and its substrates to facilitate ubiquitination. Here, we report that, in addition to this scaffolding role, TRIB1 promotes nuclear localization of COP1 by disrupting an intramolecular interaction between the WD40 domain and a previously uncharacterized regulatory site within COP1. This site, which we have termed the pseudosubstrate latch (PSL), resembles the consensus COP1-binding motif present in known COP1 substrates. Our findings support a model in which binding of the PSL to the WD40 domain stabilizes a conformation of COP1 that is conducive to CRM1-mediated nuclear export, and TRIB1 displaces this intramolecular interaction to induce nuclear retention of COP1. Coevolution of Tribbles and the PSL in metazoans further underscores the importance of this role of Tribbles in regulating COP1 function.
- Published
- 2019
22. PK1 from Drosophila obscurin is an inactive pseudokinase with scaffolding properties
- Author
-
Thomas Zacharchenko, Till Dorendorf, Nicolas Locker, Evert Van Dijk, Anja Katzemich, Kay Diederichs, Belinda Bullard, and Olga Mayans
- Subjects
pseudokinase ,X-ray crystallography ,muscle sarcomere ,Biology (General) ,QH301-705.5 - Abstract
Obscurins are large filamentous proteins with crucial roles in the assembly, stability and regulation of muscle. Characteristic of these proteins is a tandem of two C-terminal kinase domains, PK1 and PK2, that are separated by a long intrinsically disordered sequence. The significance of this conserved domain arrangement is unknown. Our study of PK1 from Drosophila obscurin shows that this is a pseudokinase with features typical of the CAM-kinase family, but which carries a minimalistic regulatory tail that no longer binds calmodulin or has mechanosensory properties typical of other sarcomeric kinases. PK1 binds ATP with high affinity, but in the absence of magnesium and lacks detectable phosphotransfer activity. It also has a highly diverged active site, strictly conserved across arthropods, that might have evolved to accommodate an unconventional binder. We find that PK1 interacts with PK2, suggesting a functional relation to the latter. These findings lead us to speculate that PK1/PK2 form a pseudokinase/kinase dual system, where PK1 might act as an allosteric regulator of PK2 and where mechanosensing properties, akin to those described for regulatory tails in titin-like kinases, might now reside on the unstructured interkinase segment. We propose that the PK1-interkinase-PK2 region constitutes an integrated functional unit in obscurin proteins.
- Published
- 2023
- Full Text
- View/download PDF
23. Deucravacitinib is an allosteric TYK2 protein kinase inhibitor FDA-approved for the treatment of psoriasis
- Author
-
Robert Roskoski, Jr.
- Subjects
Cytokines ,JAK-STAT signaling ,Immune response ,Inflammation ,Protein kinase inhibitor classification ,Pseudokinase ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Psoriasis is a heterogeneous, inflammatory, autoimmune skin disease that affects up to 2% of the world’s population. There are many treatment modalities including topical medicines, ultraviolet light therapy, monoclonal antibodies, and several oral medications. Cytokines play a central role in the pathogenesis of this disorder including TNF-α, (tumor necrosis factor-α) IL-17A (interleukin-17A), IL-17F, IL-22, and IL-23. Cytokine signaling involves transduction mediated by the JAK-STAT pathway. There are four JAKS (JAK1/2/3 and TYK2) and six STATS (signal transducer and activators of transcription). Janus kinases contain an inactive JH2 domain that is aminoterminal to the active JH1 domain. Under basal conditions, the JH2 domain inhibits the activity of the JH1 domain. Deucravacitinib is an orally effective N-trideuteromethyl-pyridazine derivative that targets and stabilizes the TYK2 JH2 domain and thereby blocks TYK2 JH1 activity. Seven other JAK inhibitors, which target the JAK family JH1 domain, are prescribed for the treatment of neoplastic and other inflammatory diseases. The use of deuterium in the trimethylamide decreases the rate of demethylation and slows the production of a metabolite that is active against a variety of targets in addition to TYK2. A second unique aspect in the development of deucravacitinib is the targeting of a pseudokinase domain. Deucravacitinib is rather specific for TYK2 and its toxic effects are much less than those of the other FDA-approved JAK inhibitors. The successful development of deucravacitinib may stimulate the development of additional pseudokinase ligands for the JAK family and for other kinase families as well.
- Published
- 2023
- Full Text
- View/download PDF
24. The web of death: the expanding complexity of necroptotic signaling.
- Author
-
Horne, Christopher R., Samson, André L., and Murphy, James M.
- Subjects
- *
APOPTOSIS , *DEATH receptors , *PROTEIN kinases , *PROTEIN-protein interactions , *CELL death , *PROGRAMMED cell death 1 receptors - Abstract
The past decade has seen the emergence of the necroptosis programmed cell death pathway as an important contributor to the pathophysiology of myriad diseases. The receptor interacting protein kinase (RIPK)1 and RIPK3, and the pseudokinase executioner protein, mixed lineage kinase domain-like (MLKL), have grown to prominence as the core pathway components. Depending on cellular context, these proteins also serve as integrators of signals, such as post-translational modifications and protein or metabolite interactions, adding layers of complexity to pathway regulation. Here, we describe the emerging picture of the web of proteins that tune necroptotic signal transduction and how these events have diverged across species, presumably owing to selective pressures of pathogens upon the RIPK3–MLKL protein pair. Necroptosis is a lytic programmed cell death pathway initiated by death receptors and pathogen receptors, which is believed to have ancestrally evolved for host defense, but is frequently dysregulated in human disease. The core components of the pathway are the protein kinases, receptor interacting protein kinase (RIPK)1 and RIPK3, and the pseudokinase mixed lineage kinase domain-like (MLKL), which are responsible for membrane perturbation and cell death. The past decade has seen increasing complexity and implication of additional layers of regulation, through protein and metabolite interactions and post-translational modifications, to tune the activities and necroptotic functions of RIPK1, RIPK3, and MLKL. Many RIPK1, RIPK3, and MLKL modifications and interactions appear to be dependent on the cellular context, the necroptotic stimulus, and the species in question, which limits reductionist approaches to understanding the pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
25. Identification of Novel Small Molecule Ligands for JAK2 Pseudokinase Domain.
- Author
-
Virtanen, Anniina T., Haikarainen, Teemu, Sampathkumar, Parthasarathy, Palmroth, Maaria, Liukkonen, Sanna, Liu, Jianping, Nekhotiaeva, Natalia, Hubbard, Stevan R., and Silvennoinen, Olli
- Subjects
- *
SMALL molecules , *LIGANDS (Biochemistry) , *MYELOPROLIFERATIVE neoplasms , *STRUCTURAL optimization , *CELL survival , *THROMBOPOIETIN receptors - Abstract
Hyperactive mutation V617F in the JAK2 regulatory pseudokinase domain (JH2) is prevalent in patients with myeloproliferative neoplasms. Here, we identified novel small molecules that target JH2 of JAK2 V617F and characterized binding via biochemical and structural approaches. Screening of 107,600 small molecules resulted in identification of 55 binders to the ATP-binding pocket of recombinant JAK2 JH2 V617F protein at a low hit rate of 0.05%, which indicates unique structural characteristics of the JAK2 JH2 ATP-binding pocket. Selected hits and structural analogs were further assessed for binding to JH2 and JH1 (kinase) domains of JAK family members (JAK1-3, TYK2) and for effects on MPN model cell viability. Crystal structures were determined with JAK2 JH2 wild-type and V617F. The JH2-selective binders were identified in diaminotriazole, diaminotriazine, and phenylpyrazolo-pyrimidone chemical entities, but they showed low-affinity, and no inhibition of MPN cells was detected, while compounds binding to both JAK2 JH1 and JH2 domains inhibited MPN cell viability. X-ray crystal structures of protein-ligand complexes indicated generally similar binding modes between the ligands and V617F or wild-type JAK2. Ligands of JAK2 JH2 V617F are applicable as probes in JAK-STAT research, and SAR optimization combined with structural insights may yield higher-affinity inhibitors with biological activity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. Looking lively: emerging principles of pseudokinase signaling.
- Author
-
Sheetz, Joshua B. and Lemmon, Mark A.
- Subjects
- *
PROTEIN kinases , *SMALL molecules , *PROTEIN folding , *DRUG target , *G proteins , *G protein coupled receptors - Abstract
Progress towards understanding catalytically 'dead' protein kinases – pseudokinases – in biology and disease has hastened over the past decade. An especially lively area for structural biology, pseudokinases appear to be strikingly similar to their kinase relatives, despite lacking key catalytic residues. Distinct active- and inactive-like conformation states, which are crucial for regulating bona fide protein kinases, are conserved in pseudokinases and appear to be essential for function. We discuss recent structural data on conformational transitions and nucleotide binding by pseudokinases, from which some common principles emerge. In both pseudokinases and bona fide kinases, a conformational toggle appears to control the ability to interact with signaling effectors. We also discuss how biasing this conformational toggle may provide opportunities to target pseudokinases pharmacologically in disease. Approximately 10% of proteins (58) in the human kinome are pseudokinases: they display the protein kinase fold but lack key conserved residues or have been observed experimentally to lack protein kinase activity. Several pseudokinases are implicated in human diseases, making them potential drug targets. A few domains first thought to qualify as pseudokinases retain kinase activity through compensatory changes, or have alternative catalytic activities. Many pseudokinases have compensatory mutations that allow retained ATP binding, despite being inactive kinases. Like their bona fide kinase relatives, several pseudokinases toggle conformationally between active-like and inactive-like states, and this controls their ability to interact with signaling effectors – suggesting analogies to regulation by small G proteins. Insights into conformation-dependent signaling by pseudokinases set the stage for their pharmacological targeting with conformational disruptors, and one pseudokinase-binding small molecule (targeting TYK2) is now in clinical development. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
27. A pseudokinase version of the histidine kinase ChrS promotes high heme tolerance of Corynebacterium glutamicum.
- Author
-
Krüger, Aileen and Frunzke, Julia
- Subjects
CORYNEBACTERIUM glutamicum ,HEME ,HISTIDINE ,FRAMESHIFT mutation ,IRON ,HISTIDINE kinases - Abstract
Heme is an essential cofactor for almost all living cells by acting as prosthetic group for various proteins or serving as alternative iron source. However, elevated levels are highly toxic for cells. Several corynebacterial species employ two paralogous, heme-responsive two-component systems (TCS), ChrSA and HrrSA, to cope with heme stress and to maintain intracellular heme homeostasis. Significant cross-talk at the level of phosphorylation between these systems was previously demonstrated. In this study, we have performed a laboratory evolution experiment to adapt Corynebacterium glutamicum to increasing heme levels. Isolated strains showed a highly increased tolerance to heme growing at concentrations of up to 100 µM. The strain featuring the highest heme tolerance harbored a frameshift mutation in the catalytical and ATPase-domain (CA-domain) of the chrS gene, converting it into a catalytically-inactive pseudokinase (ChrS_CA-fs). Reintroduction of the respective mutation in the parental C. glutamicum strain confirmed high heme tolerance and showed a drastic upregulation of hrtBA encoding a heme export system, conserved in Firmicutes and Actinobacteria. The strain encoding the ChrS pseudokinase variant showed significantly higher heme tolerance than a strain lacking chrS. Mutational analysis revealed that induction of hrtBA in the evolved strain is solely mediated via the cross-phosphorylation of the response regulator (RR) ChrA by the kinase HrrS and BACTH assays revealed the formation of heterodimers between HrrS and ChrS. Overall, our results emphasize an important role of the ChrS pseudokinase in high heme tolerance of the evolved C. glutamicum and demonstrate the promiscuity in heme-dependent signaling of the paralogous two-component systems facilitating fast adaptation to changing environmental conditions. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
28. Vps34 Kinase Domain Dynamics Regulate the Autophagic PI 3-Kinase Complex
- Author
-
Stjepanovic, Goran, Baskaran, Sulochanadevi, Lin, Mary G, and Hurley, James H
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Generic health relevance ,Adaptor Proteins ,Vesicular Transport ,Allosteric Regulation ,Autophagy ,Autophagy-Related Proteins ,Beclin-1 ,Class III Phosphatidylinositol 3-Kinases ,HEK293 Cells ,Humans ,Mass Spectrometry ,Mutation ,Protein Interaction Domains and Motifs ,Saccharomyces cerevisiae ,Saccharomyces cerevisiae Proteins ,Signal Transduction ,Structure-Activity Relationship ,Vacuolar Sorting Protein VPS15 ,allostery ,autophagy ,crosslinking mass spectrometry ,electron microscopy ,lipid kinase ,phosphoinositide ,protein dynamics ,protein kinase ,protein structure ,pseudokinase ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The class III phosphatidylinositol 3-kinase complex I (PI3KC3-C1) is required for the initiation of essentially all macroautophagic processes. PI3KC3-C1 consists of the lipid kinase catalytic subunit VPS34, the VPS15 scaffold, and the regulatory BECN1 and ATG14 subunits. The VPS34 catalytic domain and BECN1:ATG14 subcomplex do not touch, and it is unclear how allosteric signals are transmitted to VPS34. We used EM and crosslinking mass spectrometry to dissect five conformational substates of the complex, including one in which the VPS34 catalytic domain is dislodged from the complex but remains tethered by an intrinsically disordered linker. A "leashed" construct prevented dislodging without interfering with the other conformations, blocked enzyme activity in vitro, and blocked autophagy induction in yeast cells. This pinpoints the dislodging and tethering of the VPS34 catalytic domain, and its regulation by VPS15, as a master allosteric switch in autophagy induction.
- Published
- 2017
29. The Antibacterial Type VII Secretion System of Bacillus subtilis: Structure and Interactions of the Pseudokinase YukC/EssB
- Author
-
Matteo Tassinari, Thierry Doan, Marco Bellinzoni, Maïalene Chabalier, Mathilde Ben-Assaya, Mariano Martinez, Quentin Gaday, Pedro M. Alzari, Eric Cascales, Rémi Fronzes, and Francesca Gubellini
- Subjects
type VIIb secretion system ,Bacillus subtilis ,bacterial competition ,pseudokinase ,crystallographic structure ,bacterial two-hybrid assay ,Microbiology ,QR1-502 - Abstract
ABSTRACT Type VIIb secretion systems (T7SSb) were recently proposed to mediate different aspects of Firmicutes physiology, including bacterial pathogenicity and competition. However, their architecture and mechanism of action remain largely obscure. Here, we present a detailed analysis of the T7SSb-mediated bacterial competition in Bacillus subtilis, using the effector YxiD as a model for the LXG secreted toxins. By systematically investigating protein-protein interactions, we reveal that the membrane subunit YukC contacts all T7SSb components, including the WXG100 substrate YukE and the LXG effector YxiD. YukC’s crystal structure shows unique features, suggesting an intrinsic flexibility that is required for T7SSb antibacterial activity. Overall, our results shed light on the role and molecular organization of the T7SSb and demonstrate the potential of B. subtilis as a model system for extensive structure-function studies of these secretion machineries. IMPORTANCE Type VII secretion systems mediate protein extrusion from Gram-positive bacteria and are classified as T7SSa and T7SSb in Actinobacteria and in Firmicutes, respectively. Despite the genetic divergence of T7SSa and T7SSb, the high degree of structural similarity of their WXG100 substrates suggests similar secretion mechanisms. Recent advances revealed the structures of several T7SSa cytoplasmic membrane complexes, but the molecular mechanism of secretion and the T7SSb architecture remain obscure. Here, we provide hints on the organization of T7SSb in B. subtilis and a high-resolution structure of its central pseudokinase subunit, opening new perspectives for the understanding of the T7SSb secretion mechanism by using B. subtilis as an amenable bacterial model.
- Published
- 2022
- Full Text
- View/download PDF
30. A pseudokinase version of the histidine kinase ChrS promotes high heme tolerance of Corynebacterium glutamicum
- Author
-
Aileen Krüger and Julia Frunzke
- Subjects
adaptive laboratory evolution (ALE) ,heme ,pseudokinase ,two-component system (TCS) ,histidine kinase ,CA-domain ,Microbiology ,QR1-502 - Abstract
Heme is an essential cofactor for almost all living cells by acting as prosthetic group for various proteins or serving as alternative iron source. However, elevated levels are highly toxic for cells. Several corynebacterial species employ two paralogous, heme-responsive two-component systems (TCS), ChrSA and HrrSA, to cope with heme stress and to maintain intracellular heme homeostasis. Significant cross-talk at the level of phosphorylation between these systems was previously demonstrated. In this study, we have performed a laboratory evolution experiment to adapt Corynebacterium glutamicum to increasing heme levels. Isolated strains showed a highly increased tolerance to heme growing at concentrations of up to 100 μM. The strain featuring the highest heme tolerance harbored a frameshift mutation in the catalytical and ATPase-domain (CA-domain) of the chrS gene, converting it into a catalytically-inactive pseudokinase (ChrS_CA-fs). Reintroduction of the respective mutation in the parental C. glutamicum strain confirmed high heme tolerance and showed a drastic upregulation of hrtBA encoding a heme export system, conserved in Firmicutes and Actinobacteria. The strain encoding the ChrS pseudokinase variant showed significantly higher heme tolerance than a strain lacking chrS. Mutational analysis revealed that induction of hrtBA in the evolved strain is solely mediated via the cross-phosphorylation of the response regulator (RR) ChrA by the kinase HrrS and BACTH assays revealed the formation of heterodimers between HrrS and ChrS. Overall, our results emphasize an important role of the ChrS pseudokinase in high heme tolerance of the evolved C. glutamicum and demonstrate the promiscuity in heme-dependent signaling of the paralogous two-component systems facilitating fast adaptation to changing environmental conditions.
- Published
- 2022
- Full Text
- View/download PDF
31. Back From the Dead: The Atypical Kinase Activity of a Pseudokinase Regulator of Cation Fluxes During Inducible Immunity.
- Author
-
Brauer, Elizabeth K., Ahsan, Nagib, Popescu, George V., Thelen, Jay J., and Popescu, Sorina C.
- Subjects
CARRIER proteins ,PEPTIDES ,PROTEIN kinases ,CYTOKININS ,NATURAL immunity ,CALMODULIN ,ROOT growth ,PROTEIN kinase C - Abstract
Pseudokinases are thought to lack phosphotransfer activity due to altered canonical catalytic residues within their kinase domain. However, a subset of pseudokinases maintain activity through atypical phosphotransfer mechanisms. The Arabidopsis ILK1 is a pseudokinase from the Raf-like MAP3K family and is the only known plant pseudokinase with confirmed protein kinase activity. ILK1 activity promotes disease resistance and molecular pattern-induced root growth inhibition through its stabilization of the HAK5 potassium transporter with the calmodulin-like protein CML9. ILK1 also has a kinase-independent function in salt stress suggesting that it interacts with additional proteins. We determined that members of the ILK subfamily are the sole pseudokinases within the Raf-like MAP3K family and identified 179 novel putative ILK1 protein interactors. We also identified 70 novel peptide targets for ILK1, the majority of which were phosphorylated in the presence of Mn2C instead of Mg2C in line with modifications in ILK1's DFG cofactor binding domain. Overall, the ILK1-targeted or interacting proteins included diverse protein types including transporters (HAK5, STP1), protein kinases (MEKK1, MEKK3), and a cytokinin receptor (AHK2). The expression of 31 genes encoding putative ILK1-interacting or phosphorylated proteins, including AHK2, were altered in the root and shoot in response to molecular patterns suggesting a role for these genes in immunity. We describe a potential role for ILK1 interactors in the context of cation-dependent immune signaling, highlighting the importance of KC in MAMP responses. This work further supports the notion that ILK1 is an atypical kinase with an unusual cofactor dependence that may interact with multiple proteins in the cell. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
32. Insight into the mechanism of molecular recognition between human Integrin-Linked Kinase and Cpd22 and its implication at atomic level.
- Author
-
García-Marín, Javier, Rodríguez-Puyol, Diego, and Vaquero, Juan J.
- Subjects
- *
SURFACE plasmon resonance , *MOLECULAR recognition , *MOLECULAR dynamics , *COMPUTATIONAL chemistry , *CHEMICAL potential , *BINDING sites - Abstract
Absract: Pseudokinases have received increasing attention over the past decade because of their role in different physiological phenomena. Although pseudokinases lack several active-site residues, thereby hindering their catalytic activity, recent discoveries have shown that these proteins can play a role in intracellular signaling thanks to their non-catalytic functions. Integrin-linked kinase (ILK) was discovered more than two decades ago and was subsequently validated as a promising target for neoplastic diseases. Since then, only a few small-molecule inhibitors have been described, with the V-shaped pyrazole Cpd22 being the most interesting and characterized. However, little is known about its detailed mechanism of action at atomic level. In this study, using a combination of computational chemistry methods including PELE calculations, docking, molecular dynamics and experimental surface plasmon resonance, we were able to prove the direct binding of this molecule to ILK, thus providing the basis of its molecular recognition by the protein and the effect over its architecture. Our breakthroughs show that Cpd22 binding stabilizes the ILK domain by binding to the pseudo-active site in a similar way to the ATP, possibly modulating its scaffolding properties as pseudokinase. Moreover, our results explain the experimental observations obtained during Cpd22 development, thus paving the way to the development of new chemical probes and potential drugs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
33. Oncogenic Signalling of PEAK2 Pseudokinase in Colon Cancer.
- Author
-
Lecointre, Céline, Fourgous, Elise, Montarras, Ingrid, Kerneur, Clément, Simon, Valérie, Boublik, Yvan, Bonenfant, Débora, Robert, Bruno, Martineau, Pierre, and Roche, Serge
- Subjects
- *
COLON tumors , *ONCOGENES , *CELL lines - Abstract
Simple Summary: Catalytically inactive kinases, also named pseudokinases, play important roles in the regulation of cell growth and adhesion. While frequently deregulated in human cancer, their role in tumour development is partially elucidated. Here, we report an important tumour function for the pseudokinase PEAK2 in colorectal cancer (CRC) and propose that PEAK2 upregulation can affect cancer cell adhesive properties through an ABL-dependent mechanism to enable cancer progression. Therefore, targeting PEAK2 oncogenic activity with small tyrosine kinases (TK) inhibitors may be of therapeutic interest in colorectal cancer (CRC). The PEAK family pseudokinases are essential components of tyrosine kinase (TK) pathways that regulate cell growth and adhesion; however, their role in human cancer remains unclear. Here, we report an oncogenic activity of the pseudokinase PEAK2 in colorectal cancer (CRC). Notably, high PRAG1 expression, which encodes PEAK2, was associated with a bad prognosis in CRC patients. Functionally, PEAK2 depletion reduced CRC cell growth and invasion in vitro, while its overexpression increased these transforming effects. PEAK2 depletion also reduced CRC development in nude mice. Mechanistically, PEAK2 expression induced cellular protein tyrosine phosphorylation, despite its catalytic inactivity. Phosphoproteomic analysis identified regulators of cell adhesion and F-actin dynamics as PEAK2 targets. Additionally, PEAK2 was identified as a novel ABL TK activator. In line with this, PEAK2 expression localized at focal adhesions of CRC cells and induced ABL-dependent formation of actin-rich plasma membrane protrusions filopodia that function to drive cell invasion. Interestingly, all these PEAK2 transforming activities were regulated by its main phosphorylation site, Tyr413, which implicates the SRC oncogene. Thus, our results uncover a protumoural function of PEAK2 in CRC and suggest that its deregulation affects adhesive properties of CRC cells to enable cancer progression. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
34. Back From the Dead: The Atypical Kinase Activity of a Pseudokinase Regulator of Cation Fluxes During Inducible Immunity
- Author
-
Elizabeth K. Brauer, Nagib Ahsan, George V. Popescu, Jay J. Thelen, and Sorina C. Popescu
- Subjects
pseudokinase ,Raf-like MAP3K ,integrin-linked kinase ,cation signaling ,innate immunity ,microbe-associated molecular pattern ,Plant culture ,SB1-1110 - Abstract
Pseudokinases are thought to lack phosphotransfer activity due to altered canonical catalytic residues within their kinase domain. However, a subset of pseudokinases maintain activity through atypical phosphotransfer mechanisms. The Arabidopsis ILK1 is a pseudokinase from the Raf-like MAP3K family and is the only known plant pseudokinase with confirmed protein kinase activity. ILK1 activity promotes disease resistance and molecular pattern-induced root growth inhibition through its stabilization of the HAK5 potassium transporter with the calmodulin-like protein CML9. ILK1 also has a kinase-independent function in salt stress suggesting that it interacts with additional proteins. We determined that members of the ILK subfamily are the sole pseudokinases within the Raf-like MAP3K family and identified 179 novel putative ILK1 protein interactors. We also identified 70 novel peptide targets for ILK1, the majority of which were phosphorylated in the presence of Mn2+ instead of Mg2+ in line with modifications in ILK1’s DFG cofactor binding domain. Overall, the ILK1-targeted or interacting proteins included diverse protein types including transporters (HAK5, STP1), protein kinases (MEKK1, MEKK3), and a cytokinin receptor (AHK2). The expression of 31 genes encoding putative ILK1-interacting or phosphorylated proteins, including AHK2, were altered in the root and shoot in response to molecular patterns suggesting a role for these genes in immunity. We describe a potential role for ILK1 interactors in the context of cation-dependent immune signaling, highlighting the importance of K+ in MAMP responses. This work further supports the notion that ILK1 is an atypical kinase with an unusual cofactor dependence that may interact with multiple proteins in the cell.
- Published
- 2022
- Full Text
- View/download PDF
35. Palmitoylation-regulated interactions of the pseudokinase calmodulin kinase-like vesicle-associated with membranes and Arc/Arg3.1
- Author
-
Barbara Barylko, Per Niklas Hedde, Clinton A. Taylor, Derk D. Binns, Yu-Kai Huang, Gemma Molinaro, Kimberly M. Huber, David M. Jameson, and Joseph P. Albanesi
- Subjects
CaMKv ,palmitoylation ,pseudokinase ,plasma membrane ,Arc/Arg3.1 ,CaMKv-EGFP ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Calmodulin kinase-like vesicle-associated (CaMKv), a pseudokinase belonging to the Ca2+/calmodulin-dependent kinase family, is expressed predominantly in brain and neural tissue. It may function in synaptic strengthening during spatial learning by promoting the stabilization and enrichment of dendritic spines. At present, almost nothing is known regarding CaMKv structure and regulation. In this study we confirm prior proteomic analyses demonstrating that CaMKv is palmitoylated on Cys5. Wild-type CaMKv is enriched on the plasma membrane, but this enrichment is lost upon mutation of Cys5 to Ser. We further show that CaMKv interacts with another regulator of synaptic plasticity, Arc/Arg3.1, and that the interaction between these two proteins is weakened by mutation of the palmitoylated cysteine in CamKv.
- Published
- 2022
- Full Text
- View/download PDF
36. The Rhoptry Pseudokinase ROP54 Modulates Toxoplasma gondii Virulence and Host GBP2 Loading
- Author
-
Kim, Elliot W, Nadipuram, Santhosh M, Tetlow, Ashley L, Barshop, William D, Liu, Philip T, Wohlschlegel, James A, and Bradley, Peter J
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biodefense ,Emerging Infectious Diseases ,Infectious Diseases ,Foodborne Illness ,2.1 Biological and endogenous factors ,2.2 Factors relating to the physical environment ,Infection ,Toxoplasma gondii ,guanylate binding proteins ,immunity-related GTPases ,pseudokinase ,rhoptry ,virulence ,Immunology ,Microbiology - Abstract
Toxoplasma gondii uses unique secretory organelles called rhoptries to inject an array of effector proteins into the host cytoplasm that hijack host cell functions. We have discovered a novel rhoptry pseudokinase effector, ROP54, which is injected into the host cell upon invasion and traffics to the cytoplasmic face of the parasitophorous vacuole membrane (PVM). Disruption of ROP54 in a type II strain of T. gondii does not affect growth in vitro but results in a 100-fold decrease in virulence in vivo, suggesting that ROP54 modulates some aspect of the host immune response. We show that parasites lacking ROP54 are more susceptible to macrophage-dependent clearance, further suggesting that ROP54 is involved in evasion of innate immunity. To determine how ROP54 modulates parasite virulence, we examined the loading of two known innate immune effectors, immunity-related GTPase b6 (IRGb6) and guanylate binding protein 2 (GBP2), in wild-type and ∆rop54II mutant parasites. While no difference in IRGb6 loading was seen, we observed a substantial increase in GBP2 loading on the parasitophorous vacuole (PV) of ROP54-disrupted parasites. These results demonstrate that ROP54 is a novel rhoptry effector protein that promotes Toxoplasma infections by modulating GBP2 loading onto parasite-containing vacuoles. IMPORTANCE The interactions between intracellular microbes and their host cells can lead to the discovery of novel drug targets. During Toxoplasma infections, host cells express an array of immunity-related GTPases (IRGs) and guanylate binding proteins (GBPs) that load onto the parasite-containing vacuole to clear the parasite. To counter this mechanism, the parasite secretes effector proteins that traffic to the vacuole to disarm the immunity-related loading proteins and evade the immune response. While the interplay between host IRGs and Toxoplasma effector proteins is well understood, little is known about how Toxoplasma neutralizes the GBP response. We describe here a T. gondii pseudokinase effector, ROP54, that localizes to the vacuole upon invasion and is critical for parasite virulence. Toxoplasma vacuoles lacking ROP54 display an increased loading of the host immune factor GBP2, but not IRGb6, indicating that ROP54 plays a distinct role in immune evasion.
- Published
- 2016
37. Pseudokinases repurpose flexibility signatures associated with the protein kinase fold for noncatalytic roles.
- Author
-
Paul, Anindita, Subhadarshini, Seemadri, and Srinivasan, Narayanaswamy
- Abstract
The bilobal protein kinase‐like fold in pseudokinases lack one or more catalytic residues, conserved in canonical protein kinases, and are considered enzymatically deficient. Tertiary structures of pseudokinases reveal that their loops topologically equivalent to activation segments of kinases adopt contracted configurations, which is typically extended in active conformation of kinases. Herein, anisotropic network model based normal mode analysis (NMA) was conducted on 51 active conformation structures of protein kinases and 26 crystal structures of pseudokinases. Our observations indicate that although backbone fluctuation profiles are similar for individual kinase‐pseudokinase families, low intensity mean square fluctuations in pseudo‐activation segment and other sub‐structures impart rigidity to pseudokinases. Analyses of collective motions from functional modes reveal that pseudokinases, compared to active kinases, undergo distinct conformational transitions using the same structural fold. All‐atom NMA of protein kinase‐pseudokinase pairs from each family, sharing high amino acid sequence identities, yielded distinct community clusters, partitioned by residues exhibiting highly correlated fluctuations. It appears that atomic fluctuations from equivalent activation segments guide community membership and network topologies for respective kinase and pseudokinase. Our findings indicate that such adaptations in backbone and side‐chain fluctuations render pseudokinases competent for catalysis‐independent roles. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. Crystal structure of DRIK1, a stress-responsive receptor-like pseudokinase, reveals the molecular basis for the absence of ATP binding
- Author
-
Bruno Aquino, Viviane C. H. da Silva, Katlin B. Massirer, and Paulo Arruda
- Subjects
Protein kinase ,Pseudokinase ,Drought stress ,Biotic stress ,Abiotic stress ,Botany ,QK1-989 - Abstract
Abstract Background Plants reprogram metabolism and development to rapidly adapt to biotic and abiotic stress. Protein kinases play a significant role in this process by phosphorylating protein substrates that activate or inactivate signaling cascades that regulate cellular and metabolic adaptations. Despite their importance in plant biology, a notably small fraction of the plant kinomes has been studied to date. Results In this report, we describe ZmDRIK1, a stress-responsive receptor-like pseudokinase whose expression is downregulated under water restriction. We show the structural features and molecular basis of the absence of ATP binding exhibited by ZmDRIK1. The ZmDRIK1 kinase domain lacks conserved amino acids that are essential for phosphorylation activity. The crystal structure of the ZmDRIK1 kinase domain revealed the presence of a spine formed by the side chain of the triad Leu240, Tyr363, and Leu375 that occludes the ATP binding pocket. Although ZmDRIK1 is unable to bind nucleotides, it does bind the small molecule ENMD-2076 which, in a cocrystal structure, revealed the potential to serve as a ZmDRIK1 inhibitor. Conclusion ZmDRIK1 is a novel receptor-like pseudokinase responsive to biotic and abiotic stress. The absence of ATP binding and consequently, the absence of phosphorylation activity, was proven by the crystal structure of the apo form of the protein kinase domain. The expression profiling of the gene encoding ZmDRIK1 suggests this kinase may play a role in downregulating the expression of stress responsive genes that are not necessary under normal conditions. Under biotic and abiotic stress, ZmDRIK1 is down-regulated to release the expression of these stress-responsive genes.
- Published
- 2020
- Full Text
- View/download PDF
39. ULK4 in Neurodevelopmental and Neuropsychiatric Disorders
- Author
-
Shilin Luo, Nanxi Zheng, and Bing Lang
- Subjects
ULK4 ,neurodevelopmental disorder ,neuropsychiatric disorder ,pseudokinase ,schizophrenia ,Biology (General) ,QH301-705.5 - Abstract
The gene Unc51-like kinase 4 (ULK4) belongs to the Unc-51-like serine/threonine kinase family and is assumed to encode a pseudokinase with unclear function. Recently, emerging evidence has suggested that ULK4 may be etiologically involved in a spectrum of neuropsychiatric disorders including schizophrenia, but the underlying mechanism remains unaddressed. Here, we summarize the key findings of the structure and function of the ULK4 protein to provide comprehensive insights to better understand ULK4-related neurodevelopmental and neuropsychiatric disorders and to aid in the development of a ULK4-based therapeutic strategy.
- Published
- 2022
- Full Text
- View/download PDF
40. Inhibitors identify an auxiliary role for mTOR signalling in necroptosis execution downstream of MLKL activation.
- Author
-
Garnish SE, Horne CR, Meng Y, Young SN, Jacobsen AV, Hildebrand JM, and Murphy JM
- Subjects
- Humans, Protein Kinase Inhibitors pharmacology, Protein Kinases metabolism, Protein Kinases genetics, Necroptosis drug effects, Necroptosis physiology, TOR Serine-Threonine Kinases metabolism, Signal Transduction
- Abstract
Necroptosis is a lytic and pro-inflammatory form of programmed cell death executed by the terminal effector, the MLKL (mixed lineage kinase domain-like) pseudokinase. Downstream of death and Toll-like receptor stimulation, MLKL is trafficked to the plasma membrane via the Golgi-, actin- and microtubule-machinery, where activated MLKL accumulates until a critical lytic threshold is exceeded and cell death ensues. Mechanistically, MLKL's lytic function relies on disengagement of the N-terminal membrane-permeabilising four-helix bundle domain from the central autoinhibitory brace helix: a process that can be experimentally mimicked by introducing the R30E MLKL mutation to induce stimulus-independent cell death. Here, we screened a library of 429 kinase inhibitors for their capacity to block R30E MLKL-mediated cell death, to identify co-effectors in the terminal steps of necroptotic signalling. We identified 13 compounds - ABT-578, AR-A014418, AZD1480, AZD5363, Idelalisib, Ipatasertib, LJI308, PHA-793887, Rapamycin, Ridaforolimus, SMI-4a, Temsirolimus and Tideglusib - each of which inhibits mammalian target of rapamycin (mTOR) signalling or regulators thereof, and blocked constitutive cell death executed by R30E MLKL. Our study implicates mTOR signalling as an auxiliary factor in promoting the transport of activated MLKL oligomers to the plasma membrane, where they accumulate into hotspots that permeabilise the lipid bilayer to cause cell death., (© 2024 The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
41. A secretory kinase complex regulates extracellular protein phosphorylation.
- Author
-
Cui, Jixin, Xiao, Junyu, Tagliabracci, Vincent S, Wen, Jianzhong, Rahdar, Meghdad, and Dixon, Jack E
- Subjects
Mammary Glands ,Animal ,Cell Line ,Ameloblasts ,Animals ,Mice ,Inbred C57BL ,Humans ,Mice ,Lepidoptera ,Casein Kinase I ,Dental Enamel Proteins ,Recombinant Proteins ,Extracellular Matrix Proteins ,Sequence Alignment ,Signal Transduction ,Gene Expression ,Gene Expression Regulation ,Amino Acid Sequence ,Sequence Homology ,Amino Acid ,Phosphorylation ,Founder Effect ,Molecular Sequence Data ,Female ,biochemistry ,extracellular protein phosphorylation ,human ,kinase complex ,mouse ,pseudokinase ,secretory pathway phosphorylation ,tooth enamel formation ,Mammary Glands ,Animal ,Inbred C57BL ,Sequence Homology ,Amino Acid ,Biochemistry and Cell Biology - Abstract
Although numerous extracellular phosphoproteins have been identified, the protein kinases within the secretory pathway have only recently been discovered, and their regulation is virtually unexplored. Fam20C is the physiological Golgi casein kinase, which phosphorylates many secreted proteins and is critical for proper biomineralization. Fam20A, a Fam20C paralog, is essential for enamel formation, but the biochemical function of Fam20A is unknown. Here we show that Fam20A potentiates Fam20C kinase activity and promotes the phosphorylation of enamel matrix proteins in vitro and in cells. Mechanistically, Fam20A is a pseudokinase that forms a functional complex with Fam20C, and this complex enhances extracellular protein phosphorylation within the secretory pathway. Our findings shed light on the molecular mechanism by which Fam20C and Fam20A collaborate to control enamel formation, and provide the first insight into the regulation of secretory pathway phosphorylation.
- Published
- 2015
42. Exploring nod factor receptors activation process in chickpea by bridging modelling, docking and molecular dynamics simulations.
- Author
-
Palaka, Bhagath Kumar, Vijayakumar, Saravanan, and Roy Choudhury, Swarup
- Subjects
- *
MOLECULAR dynamics , *MOLECULAR docking , *MOLECULAR structure , *CHICKPEA , *LIGAND binding (Biochemistry) , *ROOT-tubercles - Abstract
Plasma membrane-bound receptor proteins play crucial roles in the perception and further transmission of regulatory signals to modulate numerous developmental and metabolic events. Precise functioning and fine-tuning of Nod factor receptor (NFR) mediated signalling is a critical requirement for root nodule symbiosis. Here, we have identified, cloned and phylogenetically characterized chickpea NFR1 and NFR5, which are showing significant homology with other legume NFR receptors. Homology modelling and molecular dynamics simulations highlight the molecular structure of ligand binding ectodomains [EDs] and cytosolic kinase domains [KDs] of NFRs in chickpea. Our detailed structural analysis also revealed that both NFR1 and NFR5 share resemblance as well as dissimilarity in sequence, structure and substrate-binding pocket. Further, molecular docking simulations provide us adequate insights into the active site of receptors where the Nod factor (NF) binds. The outcome of this work sheds light on the binding specificity of NFs towards NFRs and thus may significantly contribute to the design of new strategies in improving root-nodule symbiosis towards meeting the agricultural demands. • Several conserved and variable residues crucial for NF binding and phosphorylation remain in both CaNFR1 and CaNFR5. • CaNFR1 is an active kinase, whereas CaNFR5 is a pseudokinase, as it is not functional for catalysis. • CaNFR1 EDs show high conservation in the domain architecture with other model legumes EDs. • NFRs have strong binding efficacy towards their native NFs compared to other NFs. • CaNFR1-CaNFR5 ED can interact together to form a complex. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
43. tomato receptor-like cytoplasmic kinase, SlZRK1, acts as a negative regulator in wound-induced jasmonic acid accumulation and insect resistance.
- Author
-
Sun, Zongyan, Zang, Yudi, Zhou, Leilei, Song, Yanping, Chen, Di, Zhang, Qiaoli, Liu, Chengxia, Yi, Yuetong, Zhu, Benzhong, Fu, Daqi, Zhu, Hongliang, and Qu, Guiqin
- Subjects
- *
JASMONIC acid , *PLANT resistance to insects , *CELL membranes , *RECEPTOR-like kinases , *CELLULAR signal transduction , *TOMATOES , *WOUND healing - Abstract
Jasmonates accumulate rapidly and act as key regulators in response to mechanical wounding, but few studies have linked receptor-like cytoplasmic kinases (RLCKs) to wound-induced jasmonic acid (JA) signaling cascades. Here, we identified a novel wounding-induced RLCK-XII-2 subfamily member (SlZRK1) in tomato (Solanum lycopersicum) that was closely related to Arabidopsis HOPZ-ETI-DEFICIENT 1 (ZED1)-related kinases 1 based on phylogenetic analysis. SlZRK1 was targeted to the plasma membrane of tobacco mesophyll protoplasts as determined by transient co-expression with the plasma membrane marker mCherry–H+-ATPase. Catalytic residue sequence analysis and an in vitro kinase assay indicated that SlZRK1 may act as a pseudokinase. To further analyse the function of SlZRK1, we developed two stable knock-out mutants by CRISPR/Cas9. Loss of SlZRK1 significantly altered the expression of genes involved in JA biosynthesis, salicylic acid biosynthesis, and ethylene response. Furthermore, after mechanical wounding treatment, slzrk1 mutants increased transcription of early wound-inducible genes involved in JA biosynthesis and signaling. In addition, JA accumulation after wounding and plant resistance to herbivorous insects also were enhanced. Our findings expand plant regulatory networks in the wound-induced JA production by adding RLCKs as a new component in the wound signal transduction pathway. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
44. Investigations into a putative role for the novel BRASSIKIN pseudokinases in compatible pollen-stigma interactions in Arabidopsis thaliana
- Author
-
Jennifer Doucet, Hyun Kyung Lee, Nethangi Udugama, Jianfeng Xu, Baoxiu Qi, and Daphne R. Goring
- Subjects
Compatible pollen ,Stigma ,Signaling ,Pseudokinase ,Receptor-like cytoplasmic kinase ,Brassicaceae ,Botany ,QK1-989 - Abstract
Abstract Background In the Brassicaceae, the early stages of compatible pollen-stigma interactions are tightly controlled with early checkpoints regulating pollen adhesion, hydration and germination, and pollen tube entry into the stigmatic surface. However, the early signalling events in the stigma which trigger these compatible interactions remain unknown. Results A set of stigma-expressed pseudokinase genes, termed BRASSIKINs (BKNs), were identified and found to be present in only core Brassicaceae genomes. In Arabidopsis thaliana Col-0, BKN1 displayed stigma-specific expression while the BKN2 gene was expressed in other tissues as well. CRISPR deletion mutations were generated for the two tandemly linked BKNs, and very mild hydration defects were observed for wild-type Col-0 pollen when placed on the bkn1/2 mutant stigmas. In further analyses, the predominant transcript for the stigma-specific BKN1 was found to have a premature stop codon in the Col-0 ecotype, but a survey of the 1001 Arabidopsis genomes uncovered three ecotypes that encoded a full-length BKN1 protein. Furthermore, phylogenetic analyses identified intact BKN1 orthologues in the closely related outcrossing Arabidopsis species, A. lyrata and A. halleri. Finally, the BKN pseudokinases were found to be plasma-membrane localized through the dual lipid modification of myristoylation and palmitoylation, and this localization would be consistent with a role in signaling complexes. Conclusion In this study, we have characterized the novel Brassicaceae-specific family of BKN pseudokinase genes, and examined the function of BKN1 and BKN2 in the context of pollen-stigma interactions in A. thaliana Col-0. Additionally, premature stop codons were identified in the predicted stigma specific BKN1 gene in a number of the 1001 A. thaliana ecotype genomes, and this was in contrast to the out-crossing Arabidopsis species which carried intact copies of BKN1. Thus, understanding the function of BKN1 in other Brassicaceae species will be a key direction for future studies.
- Published
- 2019
- Full Text
- View/download PDF
45. Adventures of the undead at kinetochores
- Author
-
Sabine Elowe
- Subjects
mitosis ,kinetochore ,bubr1 ,pseudokinase ,pp2a ,b56 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
At the metaphase-to-anaphase transition, phosphatase activity feeds back to reverse early mitotic phosphorylation events. Our recent work indicates that the pseudokinase domain of the spindle checkpoint protein BUB1 (Budding Uninhibited by Benzimidazoles 1) mitotic checkpoint serine/threonine kinase B (BUB1B, BUBR1) maintains kinase–phosphatase balance at the outer kinetochore during mitotic exit.
- Published
- 2021
- Full Text
- View/download PDF
46. A robust methodology to subclassify pseudokinases based on their nucleotide-binding properties.
- Author
-
Murphy, James M, Zhang, Qingwei, Young, Samuel N, Reese, Michael L, Bailey, Fiona P, Eyers, Patrick A, Ungureanu, Daniela, Hammaren, Henrik, Silvennoinen, Olli, Varghese, Leila N, Chen, Kelan, Tripaydonis, Anne, Jura, Natalia, Fukuda, Koichi, Qin, Jun, Nimchuk, Zachary, Mudgett, Mary Beth, Elowe, Sabine, Gee, Christine L, Liu, Ling, Daly, Roger J, Manning, Gerard, Babon, Jeffrey J, and Lucet, Isabelle S
- Subjects
Cancer ,Underpinning research ,1.1 Normal biological development and functioning ,Generic health relevance ,Amino Acid Sequence ,Animals ,Cell Line ,Humans ,Insecta ,Janus Kinase 2 ,Molecular Sequence Data ,Protein Binding ,Real-Time Polymerase Chain Reaction ,Receptor ,ErbB-3 ,nucleotide binding ,non-catalytic protein-interaction domain ,protein kinase ,pseudoenzyme ,pseudokinase ,Receptor ,erbB-3 ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology - Abstract
Protein kinase-like domains that lack conserved residues known to catalyse phosphoryl transfer, termed pseudokinases, have emerged as important signalling domains across all kingdoms of life. Although predicted to function principally as catalysis-independent protein-interaction modules, several pseudokinase domains have been attributed unexpected catalytic functions, often amid controversy. We established a thermal-shift assay as a benchmark technique to define the nucleotide-binding properties of kinase-like domains. Unlike in vitro kinase assays, this assay is insensitive to the presence of minor quantities of contaminating kinases that may otherwise lead to incorrect attribution of catalytic functions to pseudokinases. We demonstrated the utility of this method by classifying 31 diverse pseudokinase domains into four groups: devoid of detectable nucleotide or cation binding; cation-independent nucleotide binding; cation binding; and nucleotide binding enhanced by cations. Whereas nine pseudokinases bound ATP in a divalent cation-dependent manner, over half of those examined did not detectably bind nucleotides, illustrating that pseudokinase domains predominantly function as non-catalytic protein-interaction modules within signalling networks and that only a small subset is potentially catalytically active. We propose that henceforth the thermal-shift assay be adopted as the standard technique for establishing the nucleotide-binding and catalytic potential of kinase-like domains.
- Published
- 2014
47. BUBR1 Pseudokinase Domain Promotes Kinetochore PP2A-B56 Recruitment, Spindle Checkpoint Silencing, and Chromosome Alignment
- Author
-
Luciano Gama Braga, Angel F. Cisneros, Michelle M. Mathieu, Maxime Clerc, Pauline Garcia, Baptiste Lottin, Chantal Garand, Philippe Thebault, Christian R. Landry, and Sabine Elowe
- Subjects
BUBR1 ,pseudokinase ,PP2A ,B56 ,Kinetochore ,Spindle checkpoint ,Biology (General) ,QH301-705.5 - Abstract
Summary: The balance of phospho-signaling at the outer kinetochore is critical for forming accurate attachments between kinetochores and the mitotic spindle and timely exit from mitosis. A major player in determining this balance is the PP2A-B56 phosphatase, which is recruited to the kinase attachment regulatory domain (KARD) of budding uninhibited by benzimidazole 1-related 1 (BUBR1) in a phospho-dependent manner. This unleashes a rapid, switch-like phosphatase relay that reverses mitotic phosphorylation at the kinetochore, extinguishing the checkpoint and promoting anaphase. Here, we demonstrate that the C-terminal pseudokinase domain of human BUBR1 is required to promote KARD phosphorylation. Mutation or removal of the pseudokinase domain results in decreased PP2A-B56 recruitment to the outer kinetochore attenuated checkpoint silencing and errors in chromosome alignment as a result of imbalance in Aurora B activity. Our data, therefore, elucidate a function for the BUBR1 pseudokinase domain in ensuring accurate and timely exit from mitosis.
- Published
- 2020
- Full Text
- View/download PDF
48. The pseudoGTPase group of pseudoenzymes.
- Author
-
Stiegler, Amy L. and Boggon, Titus J.
- Subjects
- *
DARDARIN , *PROTEIN folding , *ENTAMOEBA histolytica , *CELLULAR signal transduction , *CELL migration , *MITOSIS - Abstract
Pseudoenzymes are emerging as significant mediators and regulators of signal transduction. These proteins maintain enzyme folds and topologies, but are disrupted in the conserved motifs required for enzymatic activity. Among the pseudoenzymes, the pseudoGTPase group of atypical GTPases has recently expanded and includes the Rnd and RGK groups, RhoH and the RhoBTB proteins, mitochondrial RhoGTPase and centaurin‐γ groups, CENP‐M, dynein LIC, Entamoeba histolytica RabX3, leucine‐rich repeat kinase 2, and the p190RhoGAP proteins. The wide range of cellular functions associated with pseudoGTPases includes cell migration and adhesion, membrane trafficking and cargo transport, mitosis, mitochondrial activity, transcriptional control, and autophagy, placing the group in an expanding portfolio of signaling pathways. In this review, we examine how the pseudoGTPases differ from canonical GTPases and consider their mechanistic and functional roles in signal transduction. We review the amino acid differences between the pseudoGTPases and discuss how these proteins can be classified based on their ability to bind nucleotide and their enzymatic activity. We discuss the molecular and structural consequences of amino acid divergence from canonical GTPases and use comparison with the well‐studied pseudokinases to illustrate the classifications. PseudoGTPases are fast becoming recognized as important mechanistic components in a range of cellular roles, and we provide a concise discussion of the currently identified members of this group. Enzymes: small GTPases; EC number: EC 3.6.5.2 [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
49. Cataloguing the dead: breathing new life into pseudokinase research.
- Author
-
Shrestha, Safal, Byrne, Dominic P., Harris, John A., Kannan, Natarajan, and Eyers, Patrick A.
- Subjects
- *
PROTEIN kinases , *PROTEIN-tyrosine kinases , *SMALL molecules , *ERYTHROPOIETIN receptors , *PROTEIN folding , *KINASES - Abstract
Pseudoenzymes are present within many, but not all, known enzyme families and lack one or more conserved canonical amino acids that help define their catalytically active counterparts. Recent findings in the pseudokinase field confirm that evolutionary repurposing of the structurally defined bilobal protein kinase fold permits distinct biological functions to emerge, many of which rely on conformational switching, as opposed to canonical catalysis. In this analysis, we evaluate progress in evaluating several members of the 'dark' pseudokinome that are pertinent to help drive this expanding field. Initially, we discuss how adaptions in erythropoietin‐producing hepatocellular carcinoma (Eph) receptor tyrosine kinase domains resulted in two vertebrate pseudokinases, EphA10 and EphB6, in which co‐evolving sequences generate new motifs that are likely to be important for both nucleotide binding and catalysis‐independent signalling. Secondly, we discuss how conformationally flexible Tribbles pseudokinases, which have radiated in the complex vertebrates, control fundamental aspects of cell signalling that may be targetable with covalent small molecules. Finally, we show how species‐level adaptions in the duplicated canonical kinase protein serine kinase histone (PSKH)1 sequence have led to the appearance of the pseudokinase PSKH2, whose physiological role remains mysterious. In conclusion, we show how the patterns we discover are selectively conserved within specific pseudokinases, and that when they are modelled alongside closely related canonical kinases, many are found to be located in functionally important regions of the conserved kinase fold. Interrogation of these patterns will be useful for future evaluation of these, and other, members of the unstudied human kinome. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
50. Pseudokinases: a tribble‐edged sword.
- Author
-
Richmond, Laura and Keeshan, Karen
- Subjects
- *
SCAFFOLD proteins , *CELL proliferation , *CELL cycle , *CELL death , *SWORDS , *PROTEIN stability - Abstract
Advances in the understanding of the Tribbles family of pseudokinases (TRIB1, TRIB2 and TRIB3) reveal these proteins as potentially valuable biomarkers of disease diagnosis, prognosis, prediction and clinical strategy. In their role as signalling mediators and scaffolding proteins, TRIBs lead to changes in protein stability and activity, which impact on diverse cellular processes such as proliferation, differentiation, cell cycle and cell death. We review the role of TRIB proteins as promising therapeutic targets, with an emphasis on their role in cancer, and as biomarkers, with potential application across diverse pathological processes. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.