1
|
Cao X, Wei X, Shao Y, Li D, Zhu J. Jatropha curcas seed oil for possible human consumption: A toxicological assessment of its phorbol esters. Toxicol Rep 2025; 14:101870. [PMID: 39802603 PMCID: PMC11721233 DOI: 10.1016/j.toxrep.2024.101870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
Jatropha curcas seeds are known for their high oil content, and the oil extracted from these seeds has been traditionally utilized in biodiesel production. The presence of toxic compounds, specifically phorbol esters (PEs), in Jatropha curcas seed oil (JCSO) has blocked its use for human consumption. This article presents a thorough literature review that summarizes the latest research on the toxicological effects, including acute toxicity, genotoxicity, carcinogenicity, and chronic toxicity associated with Jatropha curcas phorbol esters (JCPEs). It also provides an overview of current detoxification strategies. A quantitative risk assessment was performed using the Benchmark Dose (BMD) approach, revealing an Acute Reference Dose (ARfD) of 139.64 μg kg-1 body weight for JCPEs (expressed as 12-O-tetradecanoyl-phorbol-13-acetate equivalent). Moreover, a Health-Based Guidance Value (HBGV) for JCPEs in a sub-chronic exposure context was established at 0.0105 mg kg-1 body weight per day. These results have guided the formulation of detoxification goals for JCSO, targeting a detoxification rate of 99.5 %, along with recommendations for future research to investigate the feasibility of using JCSO in food products.
Collapse
Affiliation(s)
- Xinyuan Cao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, China
| | - Xiaoyang Wei
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, China
| | - Yuanyuan Shao
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, China
| | - Dongbing Li
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, China
| | - Jesse Zhu
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute, University of Nottingham Ningbo China, Ningbo 315048, China
- Department of Chemical and Biochemical Engineering, Western University, London, Ontario N6A 3K7, Canada
| |
Collapse
|
2
|
Chen Y, Chen H, Han Z, Cui Y, Situ C, Qi Y, Cheng Q, Li Y. PRKCQ Is Dispensable for Spermatogenesis in Mice. Cell Biol Int 2025; 49:522-533. [PMID: 40051302 DOI: 10.1002/cbin.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/20/2025] [Accepted: 02/11/2025] [Indexed: 04/15/2025]
Abstract
Protein kinase C (PKC) family is evolutionally conserved and involved in various signaling cascades in all cells. Of the family, PRKCQ is dominatingly expressed in testis, however, its molecular functionality in spermatogenesis and male fertility remains unclear. To evaluate the role of PRKCQ in spermatogenesis, Prkcq knockout mice were generated using CRISPR/Cas9 system. Histological and immunofluorescence assays by different markers were employed to assess the testicular cells variation. Sperm parameters were analyzed by computer-assisted sperm analyzer. qPCR assay was used to examine the expression levels of other PKC family genes. We found that PRKCQ was conserved throughout evolution and highly expressed in testis. Prkcq-/- mice were successfully generated and developed viably. Normal fertility was observed in Prkcq-/- males. Prkcq-/- mice exhibited no defects in spermatogenic cells and mature sperm were full in epididymis. Furthermore, there were no differences in sperm motility and progressive motility between Prkcq-/- males and controls. Our findings report a detailed phenotypic analysis of Prkcq-/- males and indicate that PRKCQ is not required for spermatogenesis in male mice, which can provide basic information for other researchers.
Collapse
Affiliation(s)
- Yu Chen
- Medical Research Center, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Hong Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Zhongyan Han
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Yiqiang Cui
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Chenghao Situ
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Yaling Qi
- State Key Laboratory of Reproductive Medicine and Offspring Health, Department of Histology and Embryology, Nanjing Medical University, Nanjing, China
| | - Qing Cheng
- Women's Hospital of Nanjing Medical University, Nanjing Women and Children's Healthcare Hospital, Nanjing, China
| | - Yan Li
- Department of Clinical Laboratory, Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
3
|
Morita K, Hatano A, Kokaji T, Sugimoto H, Tsuchiya T, Ozaki H, Egami R, Li D, Terakawa A, Ohno S, Inoue H, Inaba Y, Suzuki Y, Matsumoto M, Takahashi M, Izumi Y, Bamba T, Hirayama A, Soga T, Kuroda S. Structural robustness and temporal vulnerability of the starvation-responsive metabolic network in healthy and obese mouse liver. Sci Signal 2025; 18:eads2547. [PMID: 40261956 DOI: 10.1126/scisignal.ads2547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 11/13/2024] [Accepted: 04/02/2025] [Indexed: 04/24/2025]
Abstract
Adaptation to starvation is a multimolecular and temporally ordered process. We sought to elucidate how the healthy liver regulates various molecules in a temporally ordered manner during starvation and how obesity disrupts this process. We used multiomic data collected from the plasma and livers of wild-type and leptin-deficient obese (ob/ob) mice at multiple time points during starvation to construct a starvation-responsive metabolic network that included responsive molecules and their regulatory relationships. Analysis of the network structure showed that in wild-type mice, the key molecules for energy homeostasis, ATP and AMP, acted as hub molecules to regulate various metabolic reactions in the network. Although neither ATP nor AMP was responsive to starvation in ob/ob mice, the structural properties of the network were maintained. In wild-type mice, the molecules in the network were temporally ordered through metabolic processes coordinated by hub molecules, including ATP and AMP, and were positively or negatively coregulated. By contrast, both temporal order and coregulation were disrupted in ob/ob mice. These results suggest that the metabolic network that responds to starvation was structurally robust but temporally disrupted by the obesity-associated loss of responsiveness of the hub molecules. In addition, we propose how obesity alters the response to intermittent fasting.
Collapse
Affiliation(s)
- Keigo Morita
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Atsushi Hatano
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
- Laboratory for Integrated Cellular Systems, RIKEN Center for Integrative Medical Sciences, Kanagawa 230-0045 Japan
| | - Toshiya Kokaji
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Data Science Center, Nara Institute of Science and Technology, Nara 630-0192, Japan
| | - Hikaru Sugimoto
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Takaho Tsuchiya
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Haruka Ozaki
- Bioinformatics Laboratory, Institute of Medicine, University of Tsukuba, Ibaraki 305-8575, Japan
- Center for Artificial Intelligence Research, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Riku Egami
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba 277-8562, Japan
| | - Dongzi Li
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Akira Terakawa
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
| | - Satoshi Ohno
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Department of AI Systems Medicine, M&D Data Science Center, Institute of Integrated Research, Institute of Science Tokyo, Tokyo 113-8510, Japan
| | - Hiroshi Inoue
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yuka Inaba
- Metabolism and Nutrition Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Ishikawa 920-8641, Japan
| | - Yutaka Suzuki
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba 277-8562, Japan
| | - Masaki Matsumoto
- Department of Omics and Systems Biology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-8510, Japan
| | - Masatomo Takahashi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Takeshi Bamba
- Division of Metabolomics, Medical Research Center for High Depth Omics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0052, Japan
| | - Tomoyoshi Soga
- Institute for Advanced Biosciences, Keio University, Yamagata 997-0052, Japan
- Human Biology-Microbiome-Quantum Research Center (WPI-Bio2Q), Keio University, Tokyo 108-8345, Japan
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, University of Tokyo, Tokyo 113-0033, Japan
- Department of Biochemistry and Molecular Biology, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
- Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Chiba 277-8562, Japan
| |
Collapse
|
4
|
Li W, Zhu K, Liu Y, Liu M, Chen Q. Recent advances in PKC inhibitor development: Structural design strategies and therapeutic applications. Eur J Med Chem 2025; 287:117290. [PMID: 39904144 DOI: 10.1016/j.ejmech.2025.117290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Protein kinase C (PKC) isozymes play critical roles in diverse cellular processes and are implicated in numerous diseases, including cancer, diabetes, and autoimmune disorders. Despite extensive research efforts spanning four decades, only one PKC inhibitor has received clinical approval, highlighting the challenges in developing selective and efficacious PKC-targeting therapeutics. Here we review recent advances in the development of small-molecule PKC inhibitors, focusing on structural design strategies, pharmacological activities, and structure-activity relationships. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and natural product derivatization that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly for PKCα and PKCβ, which have proven crucial for therapeutic applications. We discuss how integration of computational methods, structural biology insights, and rational design principles has advanced our understanding of PKC inhibition mechanisms. This comprehensive analysis reveals key challenges in PKC drug development, including the need for enhanced selectivity and reduced off-target effects, while highlighting promising directions for future therapeutic development. Our findings provide a framework for designing next-generation PKC inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuyin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Meixi Liu
- Department of Endocrinology, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, 618000, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
5
|
Su Q, Zhang J, Lin W, Zhang JF, Newton AC, Mehta S, Yang J, Zhang J. Sensitive fluorescent biosensor reveals differential subcellular regulation of PKC. Nat Chem Biol 2025; 21:501-511. [PMID: 39394268 DOI: 10.1038/s41589-024-01758-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 09/20/2024] [Indexed: 10/13/2024]
Abstract
The protein kinase C (PKC) family of serine and threonine kinases, consisting of three distinctly regulated subfamilies, has been established as critical for various cellular functions. However, how PKC enzymes are regulated at different subcellular locations, particularly at emerging signaling hubs, is unclear. Here we present a sensitive excitation ratiometric C kinase activity reporter (ExRai-CKAR2) that enables the detection of minute changes (equivalent to 0.2% of maximum stimulation) in subcellular PKC activity. Using ExRai-CKAR2 with an enhanced diacylglycerol (DAG) biosensor, we uncover that G-protein-coupled receptor stimulation triggers sustained PKC activity at the endoplasmic reticulum and lysosomes, differentially mediated by Ca2+-sensitive conventional PKC and DAG-sensitive novel PKC, respectively. The high sensitivity of ExRai-CKAR2, targeted to either the cytosol or partitioning defective complexes, further enabled us to detect previously inaccessible endogenous atypical PKC activity in three-dimensional organoids. Taken together, ExRai-CKAR2 is a powerful tool for interrogating PKC regulation in response to physiological stimuli.
Collapse
Affiliation(s)
- Qi Su
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Wei Lin
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jin-Fan Zhang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Alexandra C Newton
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sohum Mehta
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA.
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA.
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
6
|
Li Y, Wang H, Wang H, Wang T, Wu D, Wei W. Molecular Mechanisms of Pathogenic Fungal Virulence Regulation by Cell Membrane Phospholipids. J Fungi (Basel) 2025; 11:256. [PMID: 40278077 DOI: 10.3390/jof11040256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/17/2025] [Accepted: 03/25/2025] [Indexed: 04/26/2025] Open
Abstract
Pathogenic fungi represent a growing concern for human health, necessitating a deeper understanding of their molecular mechanisms of virulence to formulate effective antifungal strategies. Recent research has increasingly highlighted the role of phospholipid components in fungal cell membranes, which are not only vital for maintaining cellular integrity but also significantly influence fungal pathogenicity. This review focuses on the impact of membrane phospholipid composition on fungal growth, morphogenesis, stress responses, and interactions with host cells. To be specific, membrane phospholipid composition critically influences fungal virulence by modulating growth dynamics and morphogenesis, such as the transition from yeast to hyphal forms, which enhances tissue invasion. Additionally, phospholipids mediate stress adaptation, enabling fungi to withstand host-derived oxidative and osmotic stresses, crucial for survival within hostile host environments. Phospholipid asymmetry also impacts interactions with host cells, including adhesion, phagocytosis evasion, and the secretion of virulence factors like hydrolytic enzymes. These adaptations collectively enhance fungal pathogenicity by promoting colonization, immune evasion, and damage to host tissues, directly linking membrane architecture to infection outcomes. By elucidating the molecular mechanisms involved, we aim to underscore the potential of targeting phospholipid metabolic pathways as a promising avenue for antifungal therapy. A comprehensive understanding of how membrane phospholipid composition regulates the virulence of pathogenic fungi can provide valuable insights for developing novel antifungal strategies.
Collapse
Affiliation(s)
- Yitong Li
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Hongchen Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Hengxiu Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Tianming Wang
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Daqiang Wu
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| | - Wenfan Wei
- Department of Pathogenic Biology and Immunology, College of Integrated Chinese and Western Medicine (College of Life Science), Anhui University of Chinese Medicine, Hefei 230038, China
- Institute of Integrated Traditional Chinese and Western Medicine, Anhui University of Chinese Medicine, Hefei 230038, China
| |
Collapse
|
7
|
Guo Y, Shen K, Ge S, Zheng Q, Gao J, He X. Exploring the potential causal relationship between fatty acid metabolism ratios and major salivary gland carcinomas: A two-sample Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41802. [PMID: 40101084 PMCID: PMC11922437 DOI: 10.1097/md.0000000000041802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025] Open
Abstract
Major salivary gland carcinomas (MSGCs) is a rare but aggressive cancer, with limited understanding of its metabolic underpinnings. Lipid metabolism, particularly fatty acid metabolism ratios (FAMRs), has been implicated in various cancers, but its role in MSGCs remains unclear. This study aims to explore the potential causal relationships between specific FAMRs and MSGCs using Mendelian randomization (MR) analysis. A 2-sample MR analysis was conducted using summary data from genome-wide association studies. Three FAMRs, including the ratio of diacylglycerol to triglycerides (DAG/TG), total cholesterol (TC) to total lipids (TL) ratio in large very low-density lipoprotein (VLDL; TC/TL in large VLDL), and triglycerides to total lipids ratio in medium VLDL (TG/TL in medium VLDL), were investigated for their potential causal relationships with MSGCs. Sensitivity analyses, including MR-Egger and leave-one-out tests, were performed to assess pleiotropy and the robustness of the results. The DAG/TG and TC/TL ratios in large VLDL were significantly positively associated with an increased risk of MSGCs (OR = 10.921, P = .004 and OR = 2.651, P = .047, respectively). In contrast, the TG/TL ratio in medium VLDL showed a significant negative association with MSGCs risk (OR = 0.460, P = .041). Sensitivity analyses confirmed the robustness of these associations, with no evidence of significant pleiotropy in 2 of the ratios. This study reveals novel insights into the metabolic basis of MSGCs, demonstrating significant associations between specific FAMRs and MSGCs risk. These findings highlight the potential clinical relevance of FAMRs as biomarkers or therapeutic targets in MSGCs. Future studies should focus on diverse populations and mechanistic research to validate these associations and explore their clinical implications.
Collapse
Affiliation(s)
- Yongjie Guo
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| | - Kailin Shen
- Department of Intervention, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuqing Ge
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| | - Qi Zheng
- School of Stomatology, Shandong Second Medical University, Wei Fang, China
| | - Jingchao Gao
- School of Stomatology, Shandong Second Medical University, Wei Fang, China
| | - Xiaopo He
- Department of Maxillofacial Surgery, Wei Fang People's Hospital, Wei Fang, China
| |
Collapse
|
8
|
Ruel NM, Hammond JR. Activation of protein kinase C decreases equilibrative nucleobase transporter 1-mediated substrate uptake via phosphorylation of threonine 231. Biochim Biophys Acta Gen Subj 2025; 1869:130765. [PMID: 39837354 DOI: 10.1016/j.bbagen.2025.130765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Protein kinase C (PKC) signalling has been shown to be dysregulated in various cancers including acute lymphoblastic leukemia (ALL). We have previously determined that changes in the expression levels of SLC43A3-encoded equilibrative nucleobase transporter 1 (ENBT1) can significantly alter 6-mercaptopurine (6-MP) toxicity in ALL cells. 6-MP is a common drug used in ALL chemotherapy. Furthermore, it has been reported that activation of PKC by phorbol 12-myristate 13-acetate (PMA) impacts nucleobase uptake via an ENBT1-like transporter in Lilly Laboratories Culture-Porcine Kidney 1 (LLC-PK1) cells. We hypothesized that activation of PKC would also alter ENBT1-mediated uptake of nucleobases in leukemia cell models. Using MOLT-4, SUP-B15, and K562 cells, we incubated the cells with PMA or its inactive isoform 4α-PMA for 30 min and determined changes to ENBT1-mediated substrate uptake. All of the cell lines tested showed decreased ENBT1-mediated substrate uptake when exposed PMA, relative to that observed using 4α-PMA. Pre-incubation with the broad-spectrum PKC inhibitor, Gö6983, reversed the decrease caused by PMA. Finally, to determine the residue responsible for this PKC-mediated effect, we transiently transfected HEK293 cells (which do not express endogenous ENBT1) with wild-type SLC43A3 transcript or constructs mutated to modify the predicted PKC sites in ENBT1. We found that the mutation of threonine 231 to alanine prevents the decrease in ENBT1-mediated uptake following incubation with PMA, suggesting its involvement. This study shows that activation of PKC decreases ENBT1-mediated uptake, suggesting that aberrant activation of PKC in ALL could decrease ENBT1-mediated 6-MP uptake potentially leading to decreased therapeutic efficacy.
Collapse
Affiliation(s)
- Nicholas M Ruel
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | - James R Hammond
- Department of Pharmacology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
9
|
Tioka L, Diez RC, Sönnerborg A, van de Klundert MAA. Latency Reversing Agents and the Road to a HIV Cure. Pathogens 2025; 14:232. [PMID: 40137717 PMCID: PMC11944434 DOI: 10.3390/pathogens14030232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 03/29/2025] Open
Abstract
HIV-1 infection cannot be cured due to the presence of HIV-1 latently infected cells. These cells do not produce the virus, but they can resume virus production at any time in the absence of antiretroviral therapy. Therefore, people living with HIV (PLWH) need to take lifelong therapy. Strategies have been coined to eradicate the viral reservoir by reactivating HIV-1 latently infected cells and subsequently killing them. Various latency reversing agents (LRAs) that can reactivate HIV-1 in vitro and ex vivo have been identified. The most potent LRAs also strongly activate T cells and therefore cannot be applied in vivo. Many LRAs that reactivate HIV in the absence of general T cell activation have been identified and have been tested in clinical trials. Although some LRAs could reduce the reservoir size in clinical trials, so far, they have failed to eradicate the reservoir. More recently, immune modulators have been applied in PLWH, and the first results seem to indicate that these may reduce the reservoir and possibly improve immunological control after therapy interruption. Potentially, combinations of LRAs and immune modulators could reduce the reservoir size, and in the future, immunological control may enable PLWH to live without developing HIV-related disease in the absence of therapy.
Collapse
Affiliation(s)
- Louis Tioka
- Faculty of Medicine, Erlangen-Nürnberg, Friedrich-Alexander-Universität, 91054 Erlangen, Germany
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Rafael Ceña Diez
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, Department of Medicine Huddinge, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, 17177 Stockholm, Sweden
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, 17177 Stockholm, Sweden
| | | |
Collapse
|
10
|
Montiel I, Bello-Medina PC, Prado-Alcalá RA, Quirarte GL, Verdín-Ruvalcaba LA, Marín-Juárez TA, Medina AC. Involvement of kinases in memory consolidation of inhibitory avoidance training. Rev Neurosci 2025; 36:189-208. [PMID: 39323086 DOI: 10.1515/revneuro-2024-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 09/08/2024] [Indexed: 09/27/2024]
Abstract
The inhibitory avoidance (IA) task is a paradigm widely used to investigate the molecular and cellular mechanisms involved in the formation of long-term memory of aversive experiences. In this review, we discuss studies on different brain structures in rats associated with memory consolidation, such as the hippocampus, striatum, and amygdala, as well as some cortical areas, including the insular, cingulate, entorhinal, parietal and prefrontal cortex. These studies have shown that IA training triggers the release of neurotransmitters, hormones, growth factors, etc., that activate intracellular signaling pathways related to protein kinases, which induce intracellular non-genomic changes or transcriptional mechanisms in the nucleus, leading to the synthesis of proteins. We have summarized the temporal dynamics and crosstalk among protein kinase A, protein kinase C, mitogen activated protein kinase, extracellular-signal-regulated kinase, and Ca2+/calmodulin-dependent protein kinase II described in the hippocampus. Protein kinase activity has been associated with structural changes and synaptic strengthening, resulting in memory storage. However, little is known about the molecular mechanisms involved in intense IA training, which protects memory from typical amnestic treatments, such as protein synthesis inhibitors, and induces increased spinogenesis, suggesting an unexplored mechanism independent of the genomic pathway. This highly emotional experience causes an extinction-resistant memory, as has been observed in some pathological states such as post-traumatic stress disorder. We propose that the changes in spinogenesis observed after intense IA training could be generated by protein kinases via non-genomic pathways.
Collapse
Affiliation(s)
- Ivan Montiel
- Institut Pasteur, Université Paris Cité, Neural Circuits for Spatial Navigation and Memory, Department of Neuroscience, F-75015, Paris, France
- Sorbonne Université, Collège Doctoral, F-75005, Paris, France
| | - Paola C Bello-Medina
- Facultad de Ciencias, Universidad del Tolima, Altos de Santa Helena, Ibagué, Tolima, Colombia
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Gina L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Luis A Verdín-Ruvalcaba
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Tzitzi A Marín-Juárez
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| | - Andrea C Medina
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Campus Juriquilla, Universidad Nacional Autónoma de México, Querétaro, Qro., 76230, Mexico
| |
Collapse
|
11
|
Matsuo M, Liu S, Yamada H, Takemasa E, Suzuki Y, Mogi M. Time-sensitive effects of quercetin on rat basophilic leukemia (RBL-2H3) cell responsiveness and intracellular signaling. PLoS One 2025; 20:e0319103. [PMID: 39992972 PMCID: PMC11849837 DOI: 10.1371/journal.pone.0319103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 02/26/2025] Open
Abstract
Quercetin is known for its ability to inhibit mast cell degranulation and reduce the release of inflammatory mediators. However, it has also been reported to sensitize mast cells, potentially leading to hyperresponsiveness. This necessitates careful optimization of its use in the treatment of chronic inflammatory diseases. To fully harness quercetin's therapeutic potential, this study investigated the effects of quercetin on rat basophilic leukemia (RBL-2H3) cells responsiveness over varying durations of exposure. We employed comprehensive transcriptome analysis and subsequent functional validation of key signaling pathways. Our findings revealed that quercetin initially reduced cell activity with short-term treatment. However, with prolonged exposure, quercetin transiently enhanced both IgE cross-linkage-mediated and non-IgE-mediated responses. Specifically, prolonged quercetin treatment downregulated IgE-mediated degranulation and FcεRI expression, while potentially sensitizing RBL-2H3 cells to other non-IgE secretagogues through enhanced PKC activity. Given quercetin's multifaceted effects on intracellular signaling pathways, it is crucial to further investigate its efficacy and potential risk of adverse effects. Future studies should focus on a deeper understanding of these mechanisms to optimize quercetin's therapeutic applications while mitigating any possible negative outcomes.
Collapse
Affiliation(s)
- Mana Matsuo
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Shuang Liu
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Haruna Yamada
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Erika Takemasa
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| | - Yasuyuki Suzuki
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
- Department of Anesthesiology, Saiseikai Matsuyama Hospital, Matsuyama, Japan
- Research Division, Saiseikai Research Institute of Health Care and Welfare, Tokyo, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Shitsugawa, Toon, Ehime, Japan
| |
Collapse
|
12
|
Wolfe SA, Ma Y, Pilo CA, Chang C, Ghassemian M, Roberts AJ, Lee SR, Gorrie G, Taylor SS, Newton AC. Sex specific disruptions in Protein Kinase Cγ signaling in a mouse model of Spinocerebellar Ataxia Type 14. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.10.637267. [PMID: 39990445 PMCID: PMC11844395 DOI: 10.1101/2025.02.10.637267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Spinocerebellar Ataxia Type 14 (SCA14) is an autosomal dominant neurodegenerative disease caused by mutations in the gene encoding protein kinase C gamma (PKCγ), a Ca 2+ /diacylglycerol (DG)-dependent serine/threonine kinase dominantly expressed in cerebellar Purkinje cells. These mutations impair autoinhibitory constraints to increase the basal activity of the kinase, resulting in deficits in the cerebellum that are not observed upon simple deletion of the gene, and severe ataxia. To better understand the phenotypic impact of aberrant PKCγ signaling in disease pathology, we developed a knock-in murine model of the SCA14 mutation ΔF48 in PKCγ. This fully penetrant mutation is severe in humans and is mechanistically informative as it has high basal activity but is unresponsive to agonist stimulation. Genetic, behavioral, and molecular testing revealed that ΔF48 PKCγ SCA14 mice have ataxia related phenotypes and an altered cerebellar phosphoproteome, effects that are more severe in male mice. Analysis of existing human data reveal that SCA14 has a significantly earlier age of onset for males compared with females. Our data from this clinically relevant mutation suggest that enhanced basal activity of PKCγ is necessary and sufficient to cause ataxia and that treatment strategies to modulate aberrant PKCγ may be particularly beneficial in males. Summary New mouse model of Spinocerebellar Ataxia Type 14 containing a clinically relevant mutation in PKCγ identified underlying drivers of the disease and neuroprotection in females.
Collapse
|
13
|
Irrinki AM, Kaur J, Randhawa B, McFadden R, Snyder C, Truong H, Soohoo D, Hu E, Yu H, Murray BP, Lu B, Kornyeyev D, Irwan ID, Nguyen L, Yang YS, Belzile JP, Schmitz U, Appleby TC, Schultz B, Lalezari J, Deeks S, Cihlar T, Murry JP. Activating PKC-ε induces HIV expression with improved tolerability. PLoS Pathog 2025; 21:e1012874. [PMID: 39913544 PMCID: PMC11801715 DOI: 10.1371/journal.ppat.1012874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 01/01/2025] [Indexed: 02/11/2025] Open
Abstract
Despite suppressive antiretroviral therapy (ART), HIV-1 persists in latent reservoirs that seed new HIV infections if ART is interrupted, necessitating lifelong therapy for people with HIV. Activation of latent HIV during ART could improve recognition and elimination of infected cells by the immune system. Protein kinase C (PKC) isozymes increase HIV transcription and hence are potential latency reversal agents. However, the clinical utility of PKCs for this application is limited due to toxicity, which is poorly understood. Our studies showed that PKC activation with multiple classes of agonists leads to widespread platelet activation, consistent with disseminated intravascular coagulation, at concentrations that were similar to those required for T-cell activation. Differential expression analysis indicated that PKC-ε and PKC-η isoforms are expressed at high levels in human CD4+ T cells but not in platelets. Using structure-based drug design, we developed a novel PKC agonist, C-233, with increased selectivity for PKC-ε. C-233 increased both supernatant HIV RNA and p24 expression ex vivo after treatment of CD4+ T cells from ART-suppressed people with HIV. C-233 was 5-fold more potent for T-cell activation relative to platelet activation. Our studies support the use of structure-based drug design to create selective novel PKC agonists for the safe activation of HIV reservoirs and improved tolerability.
Collapse
Affiliation(s)
- Alivelu M. Irrinki
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Jasmine Kaur
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Bally Randhawa
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Ryan McFadden
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Chelsea Snyder
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Hoa Truong
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Daniel Soohoo
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Eric Hu
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Helen Yu
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Bernard P. Murray
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Bing Lu
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Dmytro Kornyeyev
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Ishak Darryl Irwan
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Lan Nguyen
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Yu-San Yang
- Gilead Sciences, Inc., Foster City, California, United States of America
| | | | - Uli Schmitz
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Todd C. Appleby
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Brian Schultz
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Jay Lalezari
- Quest Clinical Research, San Francisco, California, United States of America
| | - Steven Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, California, United States of America
| | - Tomas Cihlar
- Gilead Sciences, Inc., Foster City, California, United States of America
| | - Jeffrey P. Murry
- Gilead Sciences, Inc., Foster City, California, United States of America
| |
Collapse
|
14
|
Wu C, Feng H, Tian M, Chu B, Liu X, Zeng S, Wang Y, Wang H, Hou S, Liang Q. Identification and validation of diagnostic genes IFI44 and IRF9 in insomnia-associated autoimmune uveitis. Front Immunol 2025; 16:1519371. [PMID: 39958336 PMCID: PMC11825768 DOI: 10.3389/fimmu.2025.1519371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 02/18/2025] Open
Abstract
Objective Insomnia is increasingly recognized as a significant factor in the development of various autoimmune diseases, including autoimmune uveitis (AU). We investigated insomnia-associated genes that may contribute to AU pathogenesis and sought to identify potential biomarkers for insomnia-associated AU. Methods Microarray data related to insomnia and AU were downloaded from the Gene Expression Omnibus (GEO) database and analyzed. The GEO2R tool was used to identify differentially expressed genes (DEGs) that were common between insomnia and AU. Weighted gene co-expression network analysis (WGCNA), protein-protein interaction (PPI), functional enrichment, and CMap analyses were then performed to identify pathogenic genes, underlying mechanisms, and potential therapeutic drugs for insomnia-associated AU. Least absolute shrinkage and selection operator regression was employed to screen for candidate biomarkers, and their diagnostic performance was evaluated using receiver operating characteristic (ROC) curves and quantitative polymerase chain reaction (qPCR). Finally, molecular docking was applied to verify binding activities. Results We identified 138 up-regulated and 85 down-regulated DEGs that were common to insomnia and AU. PPI network analysis highlighted 10 key genes, CMap analysis identified 30 compounds, and WGCNA revealed 54 key genes and 30 compounds. Intersection of the above-mentioned key genes and compounds identified six genes and five compounds. After verification by qPCR and ROC curve analysis, IFI44 and IRF9 were confirmed as hub genes. Finally, two compounds were selected based on docking scores of less than -7 kcal/mol. Conclusion Our study demonstrated involvement of the viral response in both insomnia and AU and identified the diagnostic significance of IFI44 and IRF9 in these conditions. These findings provide novel insights for future diagnostic and therapeutic strategies to treat insomnia-associated AU.
Collapse
Affiliation(s)
- Chao Wu
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hui Feng
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Meng Tian
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Baorui Chu
- Department of Ophthalmology, Qilu Hospital, Shandong University, Jinan, China
| | - Xianyang Liu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shuhao Zeng
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yakun Wang
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Shengping Hou
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qingfeng Liang
- Beijing Institute of Ophthalmology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Gentry ZO, McAteer OD, Hamad JL, Moran JA, Kim JT, Marsden MD, Zack JA, Wender PA. Synthesis and preclinical evaluation of tigilanol tiglate analogs as latency-reversing agents for the eradication of HIV. SCIENCE ADVANCES 2025; 11:eads1911. [PMID: 39854456 PMCID: PMC11778240 DOI: 10.1126/sciadv.ads1911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025]
Abstract
Tigilanol tiglate (EBC-46) is a selective modulator of protein kinase C (PKC) isoforms that is Food and Drug Administration (FDA) approved for the treatment of mast cell tumors in canines with up to an 88% cure rate. Recently, it has been FDA approved for the treatment of soft tissue sarcomas in humans. The role of EBC-46 and, especially, its analogs in efforts to eradicate HIV, treat neurological and cardiovascular disorders, or enhance antigen density in antigen-targeted chimeric antigen receptor-T cell and chimeric antigen receptor-natural killer cell immunotherapies has not been reported. Enabled by our previously reported scalable synthesis of EBC-46, we report herein the systematic design, synthesis, and evaluation of EBC-46 analogs, including those inaccessible from the natural source and their PKC affinities, ability to translocate PKC, nuclear factor κB activity, and efficacy in reversing HIV latency in Jurkat-Latency cells. Leading analogs show exceptional PKC affinities, isoform selectivities, and functional activities, serving as promising candidates for therapeutic applications.
Collapse
Affiliation(s)
- Zachary O. Gentry
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Owen D. McAteer
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Jennifer L. Hamad
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Jose A. Moran
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Jocelyn T. Kim
- Department of Medicine, Division of Infectious Diseases, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew D. Marsden
- Department of Microbiology & Molecular Genetics, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
- Department of Medicine (Division of Infectious Diseases), School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Jerome A. Zack
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Hematology and Oncology, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Paul A. Wender
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
16
|
Ong Q, Yi Lim CJ, Liao Y, Tze-Yang Ng J, Rachel Lim LT, Yi Koh SX, En Chan S, Yu Ying PL, Lim H, Ye CR, Wang LC, Ler SG, Sobota RM, Sing Tan Y, Shulman GI, Yang X, Han W. Optogenetic control of Protein Kinase C-epsilon activity reveals its intrinsic signaling properties with spatiotemporal resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.06.631444. [PMID: 39829776 PMCID: PMC11741287 DOI: 10.1101/2025.01.06.631444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The regulation of PKC epsilon (PKCε) and its downstream effects is still not fully understood, making it challenging to develop targeted therapies or interventions. A more precise tool that enables spatiotemporal control of PKCε activity is thus required. Here, we describe a photo-activatable optogenetic PKCε probe (Opto-PKCε) consisting of an engineered PKCε catalytic domain and a blue-light inducible dimerization domain. Molecular dynamics and AlphaFold simulations enable rationalization of the dark-light activity of the optogenetic probe. We first characterize the binding partners of Opto-PKCε, which are similar to those of PKCε. Subsequent validation of the Opto-PKCε tool is performed with phosphoproteome analysis, which reveals that only PKCε substrates are phosphorylated upon light activation. Opto-PKCε could be engineered for recruitment to specific subcellular locations. Activation of Opto-PKCε in isolated hepatocytes reveals its sustained activation at the plasma membrane is required for its phosphorylation of the insulin receptor at Thr1160. In addition, Opto-PKCε recruitment to the mitochondria results in its lowering of the spare respiratory capacity through phosphorylation of complex I NDUFS4. These results demonstrate that Opto-PKCε may have broad applications for the studies of PKCε signaling with high specificity and spatiotemporal resolution.
Collapse
Affiliation(s)
- Qunxiang Ong
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- These authors contributed equally
- Lead contact
| | - Crystal Jing Yi Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- These authors contributed equally
| | - Yilie Liao
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
- Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Justin Tze-Yang Ng
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Ler Ting Rachel Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shernys Xuan Yi Koh
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Sher En Chan
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Pheobe Lee Yu Ying
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Huijun Lim
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Chen Rui Ye
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Loo Chien Wang
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Siok Ghee Ler
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Radoslaw M Sobota
- Functional Proteomics Laboratory, SingMass National Laboratory, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| | - Yaw Sing Tan
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Gerald I Shulman
- Departments of Internal Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520, Howard Hughes Medical Institute, Chevy Chase, MD
| | - Xiaoyong Yang
- Departments of Comparative Medicine and Cellular & Molecular Physiology, Yale School of Medicine, New Haven, CT 06520
| | - Weiping Han
- Laboratory of Metabolic Medicine, Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
17
|
Wu J, Jonniya NA, Hirakis SP, Olivieri C, Veglia G, Kornev AP, Taylor SS. Role of the αC-β4 loop in protein kinase structure and dynamics. eLife 2024; 12:RP91980. [PMID: 39630082 PMCID: PMC11616992 DOI: 10.7554/elife.91980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
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.
Collapse
Affiliation(s)
- Jian Wu
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Nisha A Jonniya
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Sophia P Hirakis
- Department of Chemistry and Biochemistry, University of California, San DiegoSan DiegoUnited States
| | - Cristina Olivieri
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of MinnesotaMinneapolisUnited States
- Department of Chemistry, University of MinnesotaMinneapolisUnited States
| | - Alexandr P Kornev
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
| | - Susan S Taylor
- Department of Pharmacology, University of California, San DiegoSan DiegoUnited States
- Department of Chemistry and Biochemistry, University of California, San DiegoSan DiegoUnited States
| |
Collapse
|
18
|
Chen X, Chu F, Sunchen S, Li J, Zhang M, Xu F, Dong H. TRPV4 couples with NCX1 to mediate glucose-dependent glucagon-like peptide-1 release and improve glucose homeostasis. J Physiol 2024; 602:6827-6847. [PMID: 39573816 DOI: 10.1113/jp287092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/30/2024] [Indexed: 12/18/2024] Open
Abstract
Although glucose, as a secretagogue of intestinal hormone, can stimulate glucagon-like peptide 1 (GLP-1) release, it has not been fully elucidated how glucose triggers GLP-1 release from enteroendocrine cells (EECs). Here, we investigated the regulatory mechanisms of glucose-induced Ca2+-dependent GLP-1 release from EECs. STC-1 cells that possess many features of native intestinal EECs were used. The expression of TRPV4 channels and Na+/Ca2+ exchanger 1 (NCX1) in STC-1 was analysed by immunocytochemistry. Calcium and sodium imaging, and patch clamp were applied, and GLP-1 was detected using quantitative PCR, western blot and enzyme-linked immunosorbent assays. Glucose markedly induced Na+ and Ca2+ signalling in STC-1 cells. The glucose-induced Ca2+ signalling was significantly attenuated by selective blockers of the voltage-gated Ca2+ channels (VGCC), ryanodine receptors and InsP3 receptors. Most importantly, glucose-induced Ca2+ signalling was significantly attenuated by the selective blockers of TRPV4 and NCX1. Moreover, the physical and functional couplings of TRPV4 and NCX1 were demonstrated in STC-1 cells, and they promoted glucose-mediated Ca2+ signalling to upregulate expression and release of GLP-1 via Ca2+-sensitive PKCα. Finally, the selective TRPV4 activator improved glucose tolerance in an oral glucose tolerance test in mice, but the selective blockers of TRPV4 and NCX1 attenuated glucose-induced intestinal GLP-1 release. We demonstrate a coupling of TRPV4 and NCX1 in EECs to regulate glucose-stimulated intestinal GLP-1 release via a novel TRPV4/NCX1/Ca2+/PKCα axis. Targeting this axis may provide new therapeutic potentials for glycometabolic diseases. KEY POINTS: Glucagon-like peptide 1 (GLP-1) secreted primarily from intestinal L cells in response to meals plays a critical role in maintaining glucose homeostasis. Physical and functional couplings of TRPV4 and NCX1 are pivotal in glucose-stimulated GLP-1 release via a novel TRPV4/NCX1/Ca2+/PKCα axis. Since this axis is involved in glucose homeostasis, our findings may provide new potential drug targets for prevention/treatment of glycometabolic diseases.
Collapse
Affiliation(s)
- Xiongying Chen
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Fenglan Chu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Sijin Sunchen
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Junhui Li
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Mengting Zhang
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| | - Feng Xu
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
| | - Hui Dong
- Department of Pediatric Intensive Care Unit, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatric Metabolism and Inflammatory Diseases, Chongqing, China
- Department of Pharmacology, School of Pharmacy, Qingdao University Medical College, Qingdao, China
| |
Collapse
|
19
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
20
|
Burton JC, Royer F, Grimsey NJ. Spatiotemporal control of kinases and the biomolecular tools to trace activity. J Biol Chem 2024; 300:107846. [PMID: 39362469 PMCID: PMC11550616 DOI: 10.1016/j.jbc.2024.107846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 10/05/2024] Open
Abstract
The delicate balance of cell physiology is implicitly tied to the expression and activation of proteins. Post-translational modifications offer a tool to dynamically switch protein activity on and off to orchestrate a wide range of protein-protein interactions to tune signal transduction during cellular homeostasis and pathological responses. There is a growing acknowledgment that subcellular locations of kinases define the spatial network of potential scaffolds, adaptors, and substrates. These highly ordered and localized biomolecular microdomains confer a spatially distinct bias in the outcomes of kinase activity. Furthermore, they may hold essential clues to the underlying mechanisms that promote disease. Developing tools to dissect the spatiotemporal activation of kinases is critical to reveal these mechanisms and promote the development of spatially targeted kinase inhibitors. Here, we discuss the spatial regulation of kinases, the tools used to detect their activity, and their potential impact on human health.
Collapse
Affiliation(s)
- Jeremy C Burton
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA
| | - Fredejah Royer
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA
| | - Neil J Grimsey
- Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia Athens, Athens, Georgia, USA.
| |
Collapse
|
21
|
Murter BM, Robinson SC, Banerjee H, Lau L, Uche UN, Szymczak-Workman AL, Kane LP. Downregulation of PIK3IP1/TrIP on T cells is controlled by TCR signal strength, PKC, and metalloprotease-mediated cleavage. J Biol Chem 2024; 300:107930. [PMID: 39454954 PMCID: PMC11615590 DOI: 10.1016/j.jbc.2024.107930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/15/2024] [Accepted: 10/19/2024] [Indexed: 10/28/2024] Open
Abstract
The protein known as PI3K-interacting protein (PIK3IP1), or transmembrane inhibitor of PI3K (TrIP), is highly expressed by T cells and can modulate PI3K activity in these cells. Several studies have also revealed that TrIP is rapidly downregulated following T cell activation. However, it is unclear how this downregulation is controlled. Using a novel monoclonal antibody that robustly stains cell-surface TrIP, we demonstrate that TrIP is lost from the surface of activated T cells in a manner dependent on the strength of signaling through the T cell receptor and specific downstream signaling pathways, in particular classical PKC isoforms. TrIP expression returns by 24 h after stimulation, suggesting that it may play a role in resetting T cell receptor signaling at later time points. We also provide evidence that ADAM family proteases are required for both constitutive and stimulation-induced downregulation of TrIP in T cells. Finally, by expressing truncated forms of TrIP in cells, we identify the region in the extracellular stalk domain of TrIP that is targeted for proteolytic cleavage.
Collapse
Affiliation(s)
- Benjamin M Murter
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sean C Robinson
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Hridesh Banerjee
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Louis Lau
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA; Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Uzodinma N Uche
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Lawrence P Kane
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
22
|
Murter BM, Robinson SC, Banerjee H, Lau L, Uche UU, Szymczak-Workman AL, Kane LP. Downregulation of PIK3IP1/TrIP on T cells is controlled by TCR signal strength, PKC and metalloprotease-mediated cleavage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.29.591680. [PMID: 38746242 PMCID: PMC11092459 DOI: 10.1101/2024.04.29.591680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The protein known as PI3K-interacting protein (PIK3IP1), or transmembrane inhibitor of PI3K (TrIP), is highly expressed by T cells and can modulate PI3K activity in these cells. Several studies have also revealed that TrIP is rapidly downregulated following T cell activation. However, it is unclear as to how this downregulation is controlled. Using a novel monoclonal antibody that robustly stains cell-surface TrIP, we demonstrate that TrIP is lost from the surface of activated T cells in a manner dependent on the strength of signaling through the T cell receptor (TCR) and specific downstream signaling pathways, in particular classical PKC isoforms. TrIP expression returns by 24 hours after stimulation, suggesting that it may play a role in resetting TCR signaling at later time points. We also provide evidence that ADAM family proteases are required for both constitutive and stimulation-induced downregulation of TrIP in T cells. Finally, by expressing truncated forms of TrIP in cells, we identify the region in the extracellular stalk domain of TrIP that is targeted for proteolytic cleavage.
Collapse
Affiliation(s)
- Benjamin M. Murter
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Sean C. Robinson
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Hridesh Banerjee
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Louis Lau
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
- Graduate Program in Microbiology and Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | - Uzodinma U. Uche
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| | | | - Lawrence P. Kane
- Dept. of Immunology, University of Pittsburgh, Pittsburgh PA, USA
| |
Collapse
|
23
|
Mustafa A, Shabbir M, Badshah Y, Khan K, Abid F, Trembley JH, Afsar T, Almajwal A, Razak S. Genetic polymorphism in untranslated regions of PRKCZ influences mRNA structure, stability and binding sites. BMC Cancer 2024; 24:1147. [PMID: 39272077 PMCID: PMC11401371 DOI: 10.1186/s12885-024-12900-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Variations in untranslated regions (UTR) alter regulatory pathways impacting phenotype, disease onset, and course of disease. Protein kinase C Zeta (PRKCZ), a serine-threonine kinase, is implicated in cardiovascular, neurological and oncological disorders. Due to limited research on PRKCZ, this study aimed to investigate the impact of UTR genetic variants' on binding sites for transcription factors and miRNA. RNA secondary structure, eQTLs, and variation tolerance analysis were also part of the study. METHODS The data related to PRKCZ gene variants was downloaded from the Ensembl genome browser, COSMIC and gnomAD. The RegulomeDB database was used to assess the functional impact of 5' UTR and 3'UTR variants. The analysis of the transcription binding sites (TFBS) was done through the Alibaba tool, and the Kyoto Encyclopaedia of Genes and Genomes (KEGG) was employed to identify pathways associated with PRKCZ. To predict the effect of variants on microRNA binding sites, PolymiRTS was utilized for 3' UTR variants, and the SNPinfo tool was used for 5' UTR variants. RESULTS The results obtained indicated that a total of 24 variants present in the 3' UTR and 25 variants present in the 5' UTR were most detrimental. TFBS analysis revealed that 5' UTR variants added YY1, repressor, and Oct1, whereas 3' UTR variants added AP-2alpha, AhR, Da, GR, and USF binding sites. The study predicted TFs that influenced PRKCZ expression. RNA secondary structure analysis showed that eight 5' UTR and six 3' UTR altered the RNA structure by either removal or addition of the stem-loop. The microRNA binding site analysis highlighted that seven 3' UTR and one 5' UTR variant altered the conserved site and also created new binding sites. eQTLs analysis showed that one variant was associated with PRKCZ expression in the lung and thyroid. The variation tolerance analysis revealed that PRKCZ was an intolerant gene. CONCLUSION This study laid the groundwork for future studies aimed at targeting PRKCZ as a therapeutic target.
Collapse
Affiliation(s)
- Aneela Mustafa
- Department of Healthcare BiotechnologyAtta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, 44000, Pakistan
| | - Maria Shabbir
- Department of Healthcare BiotechnologyAtta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, 44000, Pakistan.
| | - Yasmin Badshah
- Department of Healthcare BiotechnologyAtta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, 44000, Pakistan
| | | | - Fizzah Abid
- Department of Healthcare BiotechnologyAtta-Ur-Rahman School of Applied Biosciences (ASAB), National University of Sciences and Technology (NUST), Sector H-12, Islamabad, 44000, Pakistan
| | - Janeen H Trembley
- Minneapolis VA Health Care System Research Service, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Tayyaba Afsar
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Ali Almajwal
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia
| | - Suhail Razak
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
24
|
Yanagita RC, Suzuki Y, Kawanami Y, Hanaki Y, Irie K. Effect of phenolic-hydroxy-group incorporation on the biological activity of a simplified aplysiatoxin analog with an (R)-(-)-carvone-based core. Biosci Biotechnol Biochem 2024; 88:992-998. [PMID: 38936828 DOI: 10.1093/bbb/zbae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 06/22/2024] [Indexed: 06/29/2024]
Abstract
We synthesized a phenolic hydroxy group-bearing version (1) of a simplified analog of aplysiatoxin comprising a carvone-based conformation-controlling unit. Thereafter, we evaluated its antiproliferative activity against human cancer cell lines and its binding affinity to protein kinase C (PKC) isozymes. The antiproliferative activity and PKC-binding ability increased with the introduction of the phenolic hydroxy group. The results of molecular dynamics simulations and subsequent relative binding free-energy calculations conducted using an alchemical transformation procedure showed that the phenolic hydroxy group in 1 could form a hydrogen bond with a phospholipid and the PKC. The former hydrogen bonding formation facilitated the partitioning of the compound from water to the phospholipid membrane and the latter compensated for the loss of hydrogen bond with the phospholipid upon binding to the PKC. This information may facilitate the development of rational design methods for PKC ligands with additional hydrogen bonding groups.
Collapse
Affiliation(s)
- Ryo C Yanagita
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Yoshiyuki Suzuki
- Division of Applied Biological and Rare Sugar Sciences, Graduate School of Agriculture, Kagawa University, Kagawa, Japan
| | - Yasuhiro Kawanami
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Yusuke Hanaki
- Department of Applied Biological Science, Faculty of Agriculture, Kagawa University, Kagawa, Japan
| | - Kazuhiro Irie
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto, Japan
- Research and Development Promotion Organization, Doshisha University, Kyotanabe, Japan
| |
Collapse
|
25
|
Fernández-Moncada I, Lavanco G, Fundazuri UB, Bollmohr N, Mountadem S, Dalla Tor T, Hachaguer P, Julio-Kalajzic F, Gisquet D, Serrat R, Bellocchio L, Cannich A, Fortunato-Marsol B, Nasu Y, Campbell RE, Drago F, Cannizzaro C, Ferreira G, Bouzier-Sore AK, Pellerin L, Bolaños JP, Bonvento G, Barros LF, Oliet SHR, Panatier A, Marsicano G. A lactate-dependent shift of glycolysis mediates synaptic and cognitive processes in male mice. Nat Commun 2024; 15:6842. [PMID: 39122700 PMCID: PMC11316019 DOI: 10.1038/s41467-024-51008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 07/16/2024] [Indexed: 08/12/2024] Open
Abstract
Astrocytes control brain activity via both metabolic processes and gliotransmission, but the physiological links between these functions are scantly known. Here we show that endogenous activation of astrocyte type-1 cannabinoid (CB1) receptors determines a shift of glycolysis towards the lactate-dependent production of D-serine, thereby gating synaptic and cognitive functions in male mice. Mutant mice lacking the CB1 receptor gene in astrocytes (GFAP-CB1-KO) are impaired in novel object recognition (NOR) memory. This phenotype is rescued by the gliotransmitter D-serine, by its precursor L-serine, and also by lactate and 3,5-DHBA, an agonist of the lactate receptor HCAR1. Such lactate-dependent effect is abolished when the astrocyte-specific phosphorylated-pathway (PP), which diverts glycolysis towards L-serine synthesis, is blocked. Consistently, lactate and 3,5-DHBA promoted the co-agonist binding site occupancy of CA1 post-synaptic NMDA receptors in hippocampal slices in a PP-dependent manner. Thus, a tight cross-talk between astrocytic energy metabolism and gliotransmission determines synaptic and cognitive processes.
Collapse
Affiliation(s)
| | - Gianluca Lavanco
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, ''G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Unai B Fundazuri
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Nasrin Bollmohr
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Sarah Mountadem
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Tommaso Dalla Tor
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Pauline Hachaguer
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Doriane Gisquet
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Roman Serrat
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Luigi Bellocchio
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Astrid Cannich
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | | | - Yusuke Nasu
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- PRESTO, Japan Science and Technology Agency, Chiyoda-ku, Tokyo, Japan
| | - Robert E Campbell
- Department of Chemistry, School of Science, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- CERVO Brain Research Center and Department of Biochemistry, Microbiology, and Bioinformatics, Université Laval, Québec City, QC, Canada
| | - Filippo Drago
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Catania, Italy
| | - Carla Cannizzaro
- Department of Biomedicine, Neuroscience and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Guillaume Ferreira
- Univ. Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000, Bordeaux, France
| | - Anne-Karine Bouzier-Sore
- Univ. Bordeaux, CNRS, Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536, F-33000, Bordeaux, France
| | - Luc Pellerin
- Université de Poitiers et CHU de Poitiers, INSERM, IRMETIST, U1313, Poitiers, France
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), Universidad de Salamanca, CSIC, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Hospital Universitario de Salamanca, Salamanca, Spain
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), Madrid, Spain
| | - Gilles Bonvento
- Universite Paris-Saclay, CEA, CNRS, MIRCen, Laboratoire des Maladies Neurodegeneratives, Fontenay-aux-Roses, France
| | - L Felipe Barros
- Centro de Estudios Cientificos, Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Stephane H R Oliet
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Aude Panatier
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France
| | - Giovanni Marsicano
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, F-33000, Bordeaux, France.
| |
Collapse
|
26
|
Lim WL, Gaunt JR, Tan JM, Zainolabidin N, Bansal VA, Lye YM, Ch'ng TH. CREB-regulated transcription during glycogen synthesis in astrocytes. Sci Rep 2024; 14:17942. [PMID: 39095513 PMCID: PMC11297295 DOI: 10.1038/s41598-024-67976-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 07/18/2024] [Indexed: 08/04/2024] Open
Abstract
Glycogen storage, conversion and utilization in astrocytes play an important role in brain energy metabolism. The conversion of glycogen to lactate through glycolysis occurs through the coordinated activities of various enzymes and inhibition of this process can impair different brain processes including formation of long-lasting memories. To replenish depleted glycogen stores, astrocytes undergo glycogen synthesis, a cellular process that has been shown to require transcription and translation during specific stimulation paradigms. However, the detail nuclear signaling mechanisms and transcriptional regulation during glycogen synthesis in astrocytes remains to be explored. In this report, we study the molecular mechanisms of vasoactive intestinal peptide (VIP)-induced glycogen synthesis in astrocytes. VIP is a potent neuropeptide that triggers glycogenolysis followed by glycogen synthesis in astrocytes. We show evidence that VIP-induced glycogen synthesis requires CREB-mediated transcription that is calcium dependent and requires conventional Protein Kinase C but not Protein Kinase A. In parallel to CREB activation, we demonstrate that VIP also triggers nuclear accumulation of the CREB coactivator CRTC2 in astrocytic nuclei. Transcriptome profiles of VIP-induced astrocytes identified robust CREB transcription, including a subset of genes linked to glucose and glycogen metabolism. Finally, we demonstrate that VIP-induced glycogen synthesis shares similar as well as distinct molecular signatures with glucose-induced glycogen synthesis, including the requirement of CREB-mediated transcription. Overall, our data demonstrates the importance of CREB-mediated transcription in astrocytes during stimulus-driven glycogenesis.
Collapse
Affiliation(s)
- Wei Lee Lim
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Jessica Ruth Gaunt
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Jia Min Tan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Norliyana Zainolabidin
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Vibhavari Aysha Bansal
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Yi Ming Lye
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore
| | - Toh Hean Ch'ng
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Clinical Science Building, 11 Mandalay Road, 10-01-01M, Singapore, 308232, Singapore.
- School of Biological Science, Nanyang Technological University, Singapore, 636551, Singapore.
| |
Collapse
|
27
|
Gross LZF, Winkel AF, Galceran F, Schulze JO, Fröhner W, Cämmerer S, Zeuzem S, Engel M, Leroux AE, Biondi RM. Molecular insights into the regulatory landscape of PKC-related kinase-2 (PRK2/PKN2) using targeted small compounds. J Biol Chem 2024; 300:107550. [PMID: 39002682 PMCID: PMC11357854 DOI: 10.1016/j.jbc.2024.107550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/07/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024] Open
Abstract
The PKC-related kinases (PRKs, also termed PKNs) are important in cell migration, cancer, hepatitis C infection, and nutrient sensing. They belong to a group of protein kinases called AGC kinases that share common features like a C-terminal extension to the catalytic domain comprising a hydrophobic motif. PRKs are regulated by N-terminal domains, a pseudosubstrate sequence, Rho-binding domains, and a C2 domain involved in inhibition and dimerization, while Rho and lipids are activators. We investigated the allosteric regulation of PRK2 and its interaction with its upstream kinase PDK1 using a chemical biology approach. We confirmed the phosphoinositide-dependent protein kinase 1 (PDK1)-interacting fragment (PIF)-mediated docking interaction of PRK2 with PDK1 and showed that this interaction can be modulated allosterically. We showed that the polypeptide PIFtide and a small compound binding to the PIF-pocket of PRK2 were allosteric activators, by displacing the pseudosubstrate PKL region from the active site. In addition, a small compound binding to the PIF-pocket allosterically inhibited the catalytic activity of PRK2. Together, we confirmed the docking interaction and allostery between PRK2 and PDK1 and described an allosteric communication between the PIF-pocket and the active site of PRK2, both modulating the conformation of the ATP-binding site and the pseudosubstrate PKL-binding site. Our study highlights the allosteric modulation of the activity and the conformation of PRK2 in addition to the existence of at least two different complexes between PRK2 and its upstream kinase PDK1. Finally, the study highlights the potential for developing allosteric drugs to modulate PRK2 kinase conformations and catalytic activity.
Collapse
Affiliation(s)
| | - Angelika F Winkel
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | | | - Jörg O Schulze
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | - Wolfgang Fröhner
- Department of Pharmaceutical and Medicinal Chemistry, University of Saarland, Saarbrücken, Germany
| | - Simon Cämmerer
- Department of Pharmaceutical and Medicinal Chemistry, University of Saarland, Saarbrücken, Germany
| | - Stefan Zeuzem
- Department of Internal Medicine I, Universitätsklinikum Frankfurt, Frankfurt, Germany
| | - Matthias Engel
- Department of Pharmaceutical and Medicinal Chemistry, University of Saarland, Saarbrücken, Germany
| | | | - Ricardo M Biondi
- IBioBA-CONICET-MPSP, Buenos Aires, Argentina; Department of Internal Medicine I, Universitätsklinikum Frankfurt, Frankfurt, Germany.
| |
Collapse
|
28
|
Huang Y, Chen C, Liu Y, Tan B, Xiang Q, Chen Q, Wang Y, Yang W, He J, Zhou D, Wang Y, Gao K, Zheng D, Zhai R. Downregulation of tRF-Cys-GCA-029 by hyperglycemia promotes tumorigenesis and glycolysis of diabetic breast cancer through upregulating PRKCG translation. Breast Cancer Res 2024; 26:117. [PMID: 39039568 PMCID: PMC11265092 DOI: 10.1186/s13058-024-01870-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/08/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Diabetes mellitus (DM) affects up to one-third of breast cancer (BC) patients. Patients with co-existing BC and DM (BC-DM) have worsened BC prognosis. Nevertheless, the molecular mechanisms orchestrating BC-DM prognosis remain poorly understood. tRNA-derived fragments (tRFs) have been shown to regulate cancer progression. However, the biological role of tRFs in BC-DM has not been explored. METHODS tRF levels in tumor tissues and cells were detected by tRF sequencing and qRT-PCR. The effects of tRF on BC cell malignancy were assessed under euglycemic and hyperglycemic conditions in vitro. Metabolic changes were assessed by lactate, pyruvate, and extracellular acidification rate (ECAR) assays. Diabetic animal model was used to evaluate the impacts of tRF on BC tumor growth. RNA-sequencing (RNA-seq), qRT-PCR, Western blot, polysome profiling, luciferase reporter assay, and rescue experiments were performed to explore the regulatory mechanisms of tRF in BC-DM. RESULTS We identified that tRF-Cys-GCA-029 was downregulated in BC-DM tissues and under hyperglycemia conditions in BC cells. Functionally, downregulation of tRF-Cys-GCA-029 promoted BC cell proliferation and migration in a glucose level-dependent manner. tRF-Cys-GCA-029 knockdown also enhanced glycolysis metabolism in BC cells, indicated by increasing lactate/pyruvate production and ECAR levels. Notably, injection of tRF-Cys-GCA-029 mimic significantly suppressed BC tumor growth in diabetic-mice. Mechanistically, tRF-Cys-GCA-029 regulated BC cell malignancy and glycolysis via interacting with PRKCG in two ways: binding to the coding sequence (CDS) of PRKCG mRNA to regulate its transcription and altering polysomal PRKCG mRNA expression to modify its translation. CONCLUSIONS Hyperglycemia-downregulated tRF-Cys-GCA-029 enhances the malignancy and glycolysis of BC cells. tRF-Cys-GCA-029-PRKCG-glycolysis axis may be a potential therapeutic target against BC-DM.
Collapse
Affiliation(s)
- Yongyi Huang
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Cheng Chen
- Department of Cell Biology, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Yang Liu
- Department of Surgery, Cancer Hospital of Harbin Medical University, Harbin, 150081, China
| | - Binbin Tan
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Qin Xiang
- Department of Cell Biology, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Qianqian Chen
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Yiling Wang
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Wenhan Yang
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Jingsong He
- Department of Breast Surgery, Peking University Shenzhen Hospital, 1120 Lianhua Road, Shenzhen, 518036, China
| | - Duanyang Zhou
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Yuting Wang
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China
| | - Kaiping Gao
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China.
| | - Duo Zheng
- Department of Cell Biology, Shenzhen University Medical School, Shenzhen, 518055, China.
| | - Rihong Zhai
- School of Public Health, Guangdong Key Laboratory for Genome Stability & Disease Prevention, International Cancer Center, Shenzhen University Medical School, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Wang D, Li Y, Li G, Liu M, Zhou Z, Wu M, Song S, Bian Y, Dong J, Li X, Du Y, Zhang T, Shi Y. Inhibition of PKC-δ retards kidney fibrosis via inhibiting cGAS-STING signaling pathway in mice. Cell Death Discov 2024; 10:314. [PMID: 38972937 PMCID: PMC11228024 DOI: 10.1038/s41420-024-02087-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 06/17/2024] [Accepted: 07/02/2024] [Indexed: 07/09/2024] Open
Abstract
Kidney fibrosis is considered to be the ultimate aggregation pathway of chronic kidney disease (CKD), but its underlying mechanism remains elusive. Protein kinase C-delta (PKC-δ) plays critical roles in the control of growth, differentiation, and apoptosis. In this study, we found that PKC-δ was highly upregulated in human biopsy samples and mouse kidneys with fibrosis. Rottlerin, a PKC-δ inhibitor, alleviated unilateral ureteral ligation (UUO)-induced kidney fibrosis, inflammation, VDAC1 expression, and cGAS-STING signaling pathway activation. Adeno-associated virus 9 (AAV9)-mediated VDAC1 silencing or VBIT-12, a VDAC1 inhibitor, attenuated renal injury, inflammation, and activation of cGAS-STING signaling pathway in UUO mouse model. Genetic and pharmacologic inhibition of STING relieved renal fibrosis and inflammation in UUO mice. In vitro, hypoxia resulted in PKC-δ phosphorylation, VDAC1 oligomerization, and activation of cGAS-STING signaling pathway in HK-2 cells. Inhibition of PKC-δ, VDAC1 or STING alleviated hypoxia-induced fibrotic and inflammatory responses in HK-2 cells, respectively. Mechanistically, PKC-δ activation induced mitochondrial membrane VDAC1 oligomerization via direct binding VDAC1, followed by the mitochondrial DNA (mtDNA) release into the cytoplasm, and subsequent activated cGAS-STING signaling pathway, which contributed to the inflammation leading to fibrosis. In conclusion, this study has indicated for the first time that PKC-δ is an important regulator in kidney fibrosis by promoting cGAS-STING signaling pathway which mediated by VDAC1. PKC-δ may be useful for treating renal fibrosis and subsequent CKD.
Collapse
Affiliation(s)
- Dongyun Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yue Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Guiying Li
- Department of Nephrology, Affiliated Hospital of Hebei University of Engineering, Handan, 056000, China
| | - Mengyu Liu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Zihui Zhou
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Ming Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Shan Song
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yawei Bian
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Jiajia Dong
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Xinran Li
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Yunxia Du
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
| | - Tao Zhang
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China
- Department of Nephrology, Third Hospital of Hebei Medical University, Shijiazhuang, 050051, China
| | - Yonghong Shi
- Department of Pathology, Hebei Medical University, Shijiazhuang, 050017, China.
- Hebei Key Laboratory of Kidney Disease, Shijiazhuang, 050017, China.
| |
Collapse
|
30
|
Xiang Y, Naik S, Zhao L, Shi J, Ke H. Emerging phosphodiesterase inhibitors for treatment of neurodegenerative diseases. Med Res Rev 2024; 44:1404-1445. [PMID: 38279990 DOI: 10.1002/med.22017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/13/2023] [Accepted: 01/09/2024] [Indexed: 01/29/2024]
Abstract
Neurodegenerative diseases (NDs) cause progressive loss of neuron structure and ultimately lead to neuronal cell death. Since the available drugs show only limited symptomatic relief, NDs are currently considered as incurable. This review will illustrate the principal roles of the signaling systems of cyclic adenosine and guanosine 3',5'-monophosphates (cAMP and cGMP) in the neuronal functions, and summarize expression/activity changes of the associated enzymes in the ND patients, including cyclases, protein kinases, and phosphodiesterases (PDEs). As the sole enzymes hydrolyzing cAMP and cGMP, PDEs are logical targets for modification of neurodegeneration. We will focus on PDE inhibitors and their potentials as disease-modifying therapeutics for the treatment of Alzheimer's disease, Parkinson's disease, and Huntington's disease. For the overlapped but distinct contributions of cAMP and cGMP to NDs, we hypothesize that dual PDE inhibitors, which simultaneously regulate both cAMP and cGMP signaling pathways, may have complementary and synergistic effects on modifying neurodegeneration and thus represent a new direction on the discovery of ND drugs.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Swapna Naik
- Department of Pharmacology, Yale Cancer Biology Institute, Yale University, West Haven, Connecticut, USA
| | - Liyun Zhao
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
31
|
Inderwiedenstraße L, Kienitz MC. Angiotensin receptors and α 1B-adrenergic receptors regulate native IK (ACh) and phosphorylation-deficient GIRK4 (S418A) channels through different PKC isoforms. Pflugers Arch 2024; 476:1041-1064. [PMID: 38658400 DOI: 10.1007/s00424-024-02966-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/03/2024] [Accepted: 04/17/2024] [Indexed: 04/26/2024]
Abstract
Signaling of G protein-activated inwardly rectifying K+ (GIRK) channels is an important mechanism of the parasympathetic regulation of the heart rate and cardiac excitability. GIRK channels are inhibited during stimulation of Gq-coupled receptors (GqPCRs) by depletion of phosphatidyl-4,5-bisphosphate (PIP2) and/or channel phosphorylation by protein kinase C (PKC). The GqPCR-dependent modulation of GIRK currents in terms of specific PKC isoform activation was analyzed in voltage-clamp experiments in rat atrial myocytes and in CHO or HEK 293 cells. By using specific PKC inhibitors, we identified the receptor-activated PKC isoforms that contribute to phenylephrine- and angiotensin-induced GIRK channel inhibition. We demonstrate that the cPKC isoform PKCα significantly contributes to GIRK inhibition during stimulation of wildtype α1B-adrenergic receptors (α1B-ARs). Deletion of the α1B-AR serine residues S396 and S400 results in a preferential regulation of GIRK activity by PKCβ. As a novel finding, we report that the AT1-receptor-induced GIRK inhibition depends on the activation of the nPKC isoform PKCε whereas PKCα and PKCβ do not mainly participate in the angiotensin-mediated GIRK reduction. Expression of the dominant negative (DN) PKCε prolonged the onset of GIRK inhibition and significantly reduced AT1-R desensitization, indicating that PKCε regulates both GIRK channel activity and the strength of the receptor signal via a negative feedback mechanism. The serine residue S418 represents an important phosphorylation site for PKCε in the GIRK4 subunit. To analyze the functional impact of this PKC phosphorylation site for receptor-specific GIRK channel modulation, we monitored the activity of a phosphorylation-deficient (GIRK4 (S418A)) GIRK4 channel mutant during stimulation of α1B-ARs or AT1-receptors. Mutation of S418 did not impede α1B-AR-mediated GIRK inhibition, suggesting that S418 within the GIRK4 subunit is not subject to PKCα-induced phosphorylation. Furthermore, activation of angiotensin receptors induced pronounced GIRK4 (S418A) channel inhibition, excluding that this phosphorylation site contributes to the AT1-R-induced GIRK reduction. Instead, phosphorylation of S418 has a facilitative effect on GIRK activity that was abolished in the GIRK4 (S418A) mutant. To summarize, the present study shows that the receptor-dependent regulation of atrial GIRK channels is attributed to the GqPCR-specific activation of different PKC isoforms. Receptor-specific activated PKC isoforms target distinct phosphorylation sites within the GIRK4 subunit, resulting in differential regulation of GIRK channel activity with either facilitative or inhibitory effects on GIRK currents.
Collapse
Affiliation(s)
- Leonie Inderwiedenstraße
- Department for Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Universitätsstrasse 150, 44801, Bochum, Germany
| | - Marie-Cécile Kienitz
- Department for Cellular and Translational Physiology, Institute of Physiology, Ruhr University Bochum, Universitätsstrasse 150, 44801, Bochum, Germany.
| |
Collapse
|
32
|
Hodapp SJ, Gravel N, Kannan N, Newton AC. Cancer-associated mutations in protein kinase C theta are loss-of-function. Biochem J 2024; 481:759-775. [PMID: 38752473 PMCID: PMC11346454 DOI: 10.1042/bcj20240148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/11/2024]
Abstract
The Ca2+-independent, but diacylglycerol-regulated, novel protein kinase C (PKC) theta (θ) is highly expressed in hematopoietic cells where it participates in immune signaling and platelet function. Mounting evidence suggests that PKCθ may be involved in cancer, particularly blood cancers, breast cancer, and gastrointestinal stromal tumors, yet how to target this kinase (as an oncogene or as a tumor suppressor) has not been established. Here, we examine the effect of four cancer-associated mutations, R145H/C in the autoinhibitory pseudosubstrate, E161K in the regulatory C1A domain, and R635W in the regulatory C-terminal tail, on the cellular activity and stability of PKCθ. Live-cell imaging studies using the genetically-encoded fluorescence resonance energy transfer-based reporter for PKC activity, C kinase activity reporter 2 (CKAR2), revealed that the pseudosubstrate and C1A domain mutations impaired autoinhibition to increase basal signaling. This impaired autoinhibition resulted in decreased stability of the protein, consistent with the well-characterized behavior of Ca2+-regulated PKC isozymes wherein mutations that impair autoinhibition are paradoxically loss-of-function because the mutant protein is degraded. In marked contrast, the C-terminal tail mutation resulted in enhanced autoinhibition and enhanced stability. Thus, the examined mutations were loss-of-function by different mechanisms: mutations that impaired autoinhibition promoted the degradation of PKC, and those that enhanced autoinhibition stabilized an inactive PKC. Supporting a general loss-of-function of PKCθ in cancer, bioinformatics analysis revealed that protein levels of PKCθ are reduced in diverse cancers, including lung, renal, head and neck, and pancreatic. Our results reveal that PKCθ function is lost in cancer.
Collapse
Affiliation(s)
- Stefanie J. Hodapp
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
- Biomedical Sciences Graduate Program, University of California, San Diego, La Jolla, CA 92093, U.S.A
| | - Nathan Gravel
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Natarajan Kannan
- Department of Biochemistry and Molecular Biology and Institute of Bioinformatics, University of Georgia, Athens, GA 30602, U.S.A
| | - Alexandra C. Newton
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093, U.S.A
| |
Collapse
|
33
|
Li W, Ye C, He M, Ko WKW, Cheng CHK, Chan YW, Wong AOL. Differential involvement of cAMP/PKA-, PLC/PKC- and Ca 2+/calmodulin-dependent pathways in GnRH-induced prolactin secretion and gene expression in grass carp pituitary cells. Front Endocrinol (Lausanne) 2024; 15:1399274. [PMID: 38894746 PMCID: PMC11183098 DOI: 10.3389/fendo.2024.1399274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Gonadotropin-releasing hormone (GnRH) is a key stimulator for gonadotropin secretion in the pituitary and its pivotal role in reproduction is well conserved in vertebrates. In fish models, GnRH can also induce prolactin (PRL) release, but little is known for the corresponding effect on PRL gene expression as well as the post-receptor signalling involved. Using grass carp as a model, the functional role of GnRH and its underlying signal transduction for PRL regulation were examined at the pituitary level. Using laser capture microdissection coupled with RT-PCR, GnRH receptor expression could be located in carp lactotrophs. In primary cell culture prepared from grass carp pituitaries, the native forms of GnRH, GnRH2 and GnRH3, as well as the GnRH agonist [D-Arg6, Pro9, NEt]-sGnRH were all effective in elevating PRL secretion, PRL mRNA level, PRL cell content and total production. In pituitary cells prepared from the rostral pars distalis, the region in the carp pituitary enriched with lactotrophs, GnRH not only increased cAMP synthesis with parallel CREB phosphorylation and nuclear translocation but also induced a rapid rise in cytosolic Ca2+ by Ca2+ influx via L-type voltage-sensitive Ca2+ channel (VSCC) with subsequent CaM expression and NFAT2 dephosphorylation. In carp pituitary cells prepared from whole pituitaries, GnRH-induced PRL secretion was reduced/negated by inhibiting cAMP/PKA, PLC/PKC and Ca2+/CaM/CaMK-II pathways but not the signalling events via IP3 and CaN/NFAT. The corresponding effect on PRL mRNA expression, however, was blocked by inhibiting cAMP/PKA/CREB/CBP and Ca2+/CaM/CaN/NFAT2 signalling but not PLC/IP3/PKC pathway. At the pituitary cell level, activation of cAMP/PKA pathway could also induce CaM expression and Ca2+ influx via VSCC with parallel rises in PRL release and gene expression in a Ca2+/CaM-dependent manner. These findings, as a whole, suggest that the cAMP/PKA-, PLC/PKC- and Ca2+/CaM-dependent cascades are differentially involved in GnRH-induced PRL secretion and PRL transcript expression in carp lactotrophs. During the process, a functional crosstalk between the cAMP/PKA- and Ca2+/CaM-dependent pathways may occur with PRL release linked with CaMK-II and PKC activation and PRL gene transcription caused by nuclear action of CREB/CBP and CaN/NFAT2 signalling.
Collapse
Affiliation(s)
- Wensheng Li
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- School of Life Sciences, Sun Yat-Sen University, Guangzhou, China
| | - Cheng Ye
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mulan He
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Wendy K. W. Ko
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Christopher H. K. Cheng
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ying Wai Chan
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Anderson O. L. Wong
- School of Biological Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
34
|
Shanmukha S, Godfrey WH, Gharibani P, Lee JJ, Guo Y, Deng X, Wender PA, Kornberg MD, Kim PM. TPPB modulates PKC activity to attenuate neuroinflammation and ameliorate experimental multiple sclerosis. Front Cell Neurosci 2024; 18:1373557. [PMID: 38841204 PMCID: PMC11150779 DOI: 10.3389/fncel.2024.1373557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2024] [Accepted: 04/29/2024] [Indexed: 06/07/2024] Open
Abstract
Protein kinase C (PKC) plays a key role in modulating the activities of the innate immune cells of the central nervous system (CNS). A delicate balance between pro-inflammatory and regenerative activities by microglia and CNS-associated macrophages is necessary for the proper functioning of the CNS. Thus, a maladaptive activation of these CNS innate immune cells results in neurodegeneration and demyelination associated with various neurologic disorders, such as multiple sclerosis (MS) and Alzheimer's disease. Prior studies have demonstrated that modulation of PKC activity by bryostatin-1 (bryo-1) and its analogs (bryologs) attenuates the pro-inflammatory processes by microglia/CNS macrophages and alleviates the neurologic symptoms in experimental autoimmune encephalomyelitis (EAE), an MS animal model. Here, we demonstrate that (2S,5S)-(E,E)-8-(5-(4-(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPPB), a structurally distinct PKC modulator, has a similar effect to bryo-1 on CNS innate immune cells both in vitro and in vivo, attenuating neuroinflammation and resulting in CNS regeneration and repair. This study identifies a new structural class of PKC modulators, which can therapeutically target CNS innate immunity as a strategy to treat neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Wesley H. Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Judy J. Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul A. Wender
- Departments of Chemistry and Chemical and Systems Biology, Stanford University, Stanford, CA, United States
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Paul M. Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
35
|
Maldonado E, Canobra P, Oyarce M, Urbina F, Miralles VJ, Tapia JC, Castillo C, Solari A. In Vitro Identification of Phosphorylation Sites on TcPolβ by Protein Kinases TcCK1, TcCK2, TcAUK1, and TcPKC1 and Effect of Phorbol Ester on Activation by TcPKC of TcPolβ in Trypanosoma cruzi Epimastigotes. Microorganisms 2024; 12:907. [PMID: 38792752 PMCID: PMC11124317 DOI: 10.3390/microorganisms12050907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 04/06/2024] [Accepted: 04/17/2024] [Indexed: 05/26/2024] Open
Abstract
Chagas disease is caused by the single-flagellated protozoan Trypanosoma cruzi, which affects several million people worldwide. Understanding the signal transduction pathways involved in this parasite's growth, adaptation, and differentiation is crucial. Understanding the basic mechanisms of signal transduction in T. cruzi could help to develop new drugs to treat the disease caused by these protozoa. In the present work, we have demonstrated that Fetal Calf Serum (FCS) can quickly increase the levels of both phosphorylated and unphosphorylated forms of T. cruzi DNA polymerase beta (TcPolβ) in tissue-cultured trypomastigotes. The in vitro phosphorylation sites on TcPolβ by protein kinases TcCK1, TcCK2, TcAUK1, and TcPKC1 have been identified by Mass Spectrometry (MS) analysis and with antibodies against phosphor Ser-Thr-Tyr. MS analysis indicated that these protein kinases can phosphorylate Ser and Thr residues on several sites on TcPolβ. Unexpectedly, it was found that TcCK1 and TcPKC1 can phosphorylate a different Tyr residue on TcPolβ. By using a specific anti-phosphor Tyr monoclonal antibody, it was determined that TcCK1 can be in vitro autophosphorylated on Tyr residues. In vitro and in vivo studies showed that phorbol 12-myristate 13-acetate (PMA) can activate the PKC to stimulate the TcPolβ phosphorylation and enzymatic activity in T. cruzi epimastigotes.
Collapse
Affiliation(s)
- Edio Maldonado
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Paz Canobra
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Matías Oyarce
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Fabiola Urbina
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Vicente J. Miralles
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, 46110 Valencia, Spain;
| | - Julio C. Tapia
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| | - Christian Castillo
- Programa de Anatomía y Biología del Desarrollo, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
| | - Aldo Solari
- Programa de Biología Celular y Molecular, Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile; (P.C.); (M.O.); (F.U.); (J.C.T.)
| |
Collapse
|
36
|
Su Q, Zhang J, Lin W, Zhang JF, Newton AC, Mehta S, Yang J, Zhang J. Sensitive Fluorescent Biosensor Reveals Differential Subcellular Regulation of PKC. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.29.587373. [PMID: 38586003 PMCID: PMC10996667 DOI: 10.1101/2024.03.29.587373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
The protein kinase C (PKC) family of serine/threonine kinases, which consist of three distinctly regulated subfamilies, have long been established as critical for a variety of cellular functions. However, how PKC enzymes are regulated at different subcellular locations, particularly at emerging signaling hubs such as the ER, lysosome, and Par signaling complexes, is unclear. Here, we present a sensitive Excitation Ratiometric (ExRai) C Kinase Activity Reporter (ExRai-CKAR2) that enables the detection of minute changes in subcellular PKC activity. Using ExRai-CKAR2 in conjunction with an enhanced diacylglycerol (DAG) biosensor capable of detecting intracellular DAG dynamics, we uncover the differential regulation of PKC isoforms at distinct subcellular locations. We find that G-protein coupled receptor (GPCR) stimulation triggers sustained PKC activity at the ER and lysosomes, primarily mediated by Ca2+ sensitive conventional PKC (cPKC) and novel PKC (nPKC), respectively, with nPKC showing high basal activity due to elevated basal DAG levels on lysosome membranes. The high sensitivity of ExRai-CKAR2, targeted to either the cytosol or Par-complexes, further enabled us to detect previously inaccessible endogenous atypical PKC (aPKC) activity in 3D organoids. Taken together, ExRai-CKAR2 is a powerful tool for interrogating PKC regulation in response to physiological stimuli.
Collapse
Affiliation(s)
- Qi Su
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Zhang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Wei Lin
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jin-Fan Zhang
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| | - Alexandra C Newton
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Sohum Mehta
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jing Yang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Jin Zhang
- Department of Pharmacology, School of Medicine, University of California San Diego, La Jolla, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
37
|
Nadel G, Yao Z, Hacohen-Lev-Ran A, Wainstein E, Maik-Rachline G, Ziv T, Naor Z, Admon A, Seger R. Phosphorylation of PP2Ac by PKC is a key regulatory step in the PP2A-switch-dependent AKT dephosphorylation that leads to apoptosis. Cell Commun Signal 2024; 22:154. [PMID: 38419089 PMCID: PMC10900696 DOI: 10.1186/s12964-024-01536-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 02/17/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND Although GqPCR activation often leads to cell survival by activating the PI3K/AKT pathway, it was previously shown that in several cell types AKT activity is reduced and leads to JNK activation and apoptosis. The mechanism of AKT inactivation in these cells involves an IGBP1-coupled PP2Ac switch that induces the dephosphorylation and inactivation of both PI3K and AKT. However, the machinery involved in the initiation of PP2A switch is not known. METHODS We used phospho-mass spectrometry to identify the phosphorylation site of PP2Ac, and raised specific antibodies to follow the regulation of this phosphorylation. Other phosphorylations were monitored by commercial antibodies. In addition, we used coimmunoprecipitation and proximity ligation assays to follow protein-protein interactions. Apoptosis was detected by a TUNEL assay as well as PARP1 cleavage using SDS-PAGE and Western blotting. RESULTS We identified Ser24 as a phosphorylation site in PP2Ac. The phosphorylation is mediated mainly by classical PKCs (PKCα and PKCβ) but not by novel PKCs (PKCδ and PKCε). By replacing the phosphorylated residue with either unphosphorylatable or phosphomimetic residues (S24A and S24E), we found that this phosphorylation event is necessary and sufficient to mediate the PP2A switch, which ultimately induces AKT inactivation, and a robust JNK-dependent apoptosis. CONCLUSION Our results show that the PP2A switch is induced by PKC-mediated phosphorylation of Ser24-PP2Ac and that this phosphorylation leads to apoptosis upon GqPCR induction of various cells. We propose that this mechanism may provide an unexpected way to treat some cancer types or problems in the endocrine machinery.
Collapse
Affiliation(s)
- Guy Nadel
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Zhong Yao
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Avital Hacohen-Lev-Ran
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Ehud Wainstein
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Galia Maik-Rachline
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel
| | - Tamar Ziv
- Smoler Proteomic Center, Technion-Israel Institute of Technology, Haifa, Israel
| | - Zvi Naor
- Department of Biochemistry and Molecular Biology, Tel Aviv University, Tel Aviv, Israel
| | - Arie Admon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Rony Seger
- Department of Immunology and Regenerative Biology, the Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
38
|
Zong P, Feng J, Legere N, Li Y, Yue Z, Li CX, Mori Y, Miller B, Hao B, Yue L. TRPM2 enhances ischemic excitotoxicity by associating with PKCγ. Cell Rep 2024; 43:113722. [PMID: 38308841 PMCID: PMC11023021 DOI: 10.1016/j.celrep.2024.113722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/30/2023] [Accepted: 01/13/2024] [Indexed: 02/05/2024] Open
Abstract
N-methyl-D-aspartate receptor (NMDAR)-mediated glutamate excitotoxicity significantly contributes to ischemic neuronal death and post-recanalization infarction expansion. Despite tremendous efforts, targeting NMDARs has proven unsuccessful in clinical trials for mitigating brain injury. Here, we show the discovery of an interaction motif for transient receptor potential melastatin 2 (TRPM2) and protein kinase Cγ (PKCγ) association and demonstrate that TRPM2-PKCγ uncoupling is an effective therapeutic strategy for attenuating NMDAR-mediated excitotoxicity in ischemic stroke. We demonstrate that the TRPM2-PKCγ interaction allows TRPM2-mediated Ca2+ influx to promote PKCγ activation, which subsequently enhances TRPM2-induced potentiation of extrasynaptic NMDAR (esNMDAR) activity. By identifying the PKCγ binding motif on TRPM2 (M2PBM), which directly associates with the C2 domain of PKCγ, an interfering peptide (TAT-M2PBM) is developed to disrupt TRPM2-PKCγ interaction without compromising PKCγ function. M2PBM deletion or TRPM2-PKCγ dissociation abolishes both TRPM2-PKCγ and TRPM2-esNMDAR couplings, resulting in reduced excitotoxic neuronal death and attenuated ischemic brain injury.
Collapse
Affiliation(s)
- Pengyu Zong
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA; Institute for the Brain and Cognitive Sciences, University of Connecticut, 337 Mansfield Road, Unit 1272, Storrs, CT 06269, USA
| | - Jianlin Feng
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Nicholas Legere
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Genetics and Genome Sciences, UConn Health, Farmington, CT 06030, USA
| | - Yunfeng Li
- Department of Molecular Biology and Biophysics, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Zhichao Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Cindy X Li
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA; Institute for the Brain and Cognitive Sciences, University of Connecticut, 337 Mansfield Road, Unit 1272, Storrs, CT 06269, USA
| | - Yasuo Mori
- Laboratory of Molecular Biology, Department of Synthetic Chemistry and Biological Chemistry, Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Barbara Miller
- Departments of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, P.O. Box 850, Hershey, PA 17033, USA
| | - Bing Hao
- Department of Molecular Biology and Biophysics, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA
| | - Lixia Yue
- Department of Cell Biology, Calhoun Cardiology Center, University of Connecticut School of Medicine (UConn Health), Farmington, CT 06030, USA.
| |
Collapse
|
39
|
Deng Y, Hou Z, Li Y, Yi M, Wu Y, Zheng Y, Yang F, Zhong G, Hao Q, Zhai Z, Wang M, Ma X, Kang H, Ji F, Dong C, Liu H, Dai Z. Superbinder based phosphoproteomic landscape revealed PRKCD_pY313 mediates the activation of Src and p38 MAPK to promote TNBC progression. Cell Commun Signal 2024; 22:115. [PMID: 38347536 PMCID: PMC10860301 DOI: 10.1186/s12964-024-01487-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 01/11/2024] [Indexed: 02/15/2024] Open
Abstract
Phosphorylation proteomics is the basis for the study of abnormally activated kinase signaling pathways in breast cancer, which facilitates the discovery of new oncogenic agents and drives the discovery of potential targets for early diagnosis and therapy of breast cancer. In this study, we have explored the aberrantly active kinases in breast cancer development and to elucidate the role of PRKCD_pY313 in triple negative breast cancer (TNBC) progression. We collected 47 pairs of breast cancer and paired far-cancer normal tissues and analyzed phosphorylated tyrosine (pY) peptides by Superbinder resin and further enriched the phosphorylated serine/threonine (pS/pT) peptides using TiO2 columns. We mapped the kinases activity of different subtypes of breast cancer and identified PRKCD_pY313 was upregulated in TNBC cell lines. Gain-of-function assay revealed that PRKCD_pY313 facilitated the proliferation, enhanced invasion, accelerated metastasis, increased the mitochondrial membrane potential and reduced ROS level of TNBC cell lines, while Y313F mutation and low PRKCD_pY313 reversed these effects. Furthermore, PRKCD_pY313 significantly upregulated Src_pY419 and p38_pT180/pY182, while low PRKCD_pY313 and PRKCD_Y313F had opposite effects. Dasatinib significantly inhibited the growth of PRKCD_pY313 overexpression cells, and this effect could be enhanced by Adezmapimod. In nude mice xenograft model, PRKCD_pY313 significantly promoted tumor progression, accompanied by increased levels of Ki-67, Bcl-xl and Vimentin, and decreased levels of Bad, cleaved caspase 3 and ZO1, which was opposite to the trend of Y313F group. Collectively, the heterogeneity of phosphorylation exists in different molecular subtypes of breast cancer. PRKCD_pY313 activates Src and accelerates TNBC progression, which could be inhibited by Dasatinib.
Collapse
Affiliation(s)
- Yujiao Deng
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhanwu Hou
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yizhen Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ming Yi
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yi Zheng
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fei Yang
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guansheng Zhong
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhen Zhai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaobin Ma
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fanpu Ji
- Department of Infectious Diseases, The Second Affiliated Hospital of Xian Jiaotong University, Xi'an, China
| | - Chenfang Dong
- Department of Pathology and Pathophysiology, Department of Colorectal Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, The Second Affiliated Hospital, Ministry of Education, Zhejiang University School of Medicine, Hangzhou, China.
- Zhejiang Key Laboratory for Disease Proteomics, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| | - Huadong Liu
- Center for Mitochondrial Biology and Medicine & Douglas C. Wallace Institute for Mitochondrial and Epigenetic Information Sciences, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.
- Department of Breast Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
40
|
Shanmukha S, Godfrey WH, Gharibani P, Lee JJ, Guo Y, Deng X, Wender PA, Kornberg MD, Kim PM. TPPB modulates PKC activity to attenuate neuroinflammation and ameliorate experimental multiple sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.02.578637. [PMID: 38370818 PMCID: PMC10871289 DOI: 10.1101/2024.02.02.578637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Protein kinase C (PKC) plays a key role in modulating the activities of the innate immune cells of the central nervous system (CNS). A delicate balance between pro-inflammatory and regenerative activities by microglia and CNS-associated macrophages is necessary for the proper functioning of the CNS. Thus, a maladaptive activation of these CNS innate immune cells results in neurodegeneration and demyelination associated with various neurologic disorders, such as multiple sclerosis (MS) and Alzheimer's disease. Prior studies have demonstrated that modulation of PKC activity by bryostatin-1 (bryo-1) and its analogs (bryologs) attenuates the pro-inflammatory processes by microglia/CNS macrophages and alleviates the neurologic symptoms in experimental autoimmune encephalomyelitis (EAE), an MS animal model. Here, we demonstrate that (2S,5S)-(E,E)-8-(5-(4(trifluoromethyl)phenyl)-2,4-pentadienoylamino)benzolactam (TPPB), a structurally distinct PKC modulator, has a similar effect to bryo-1 on CNS innate immune cells both in vitro and in vivo, attenuating neuroinflammation and resulting in CNS regeneration and repair. This study identifies a new structural class of PKC modulators, which can therapeutically target CNS innate immunity as a strategy to treat neuroinflammatory and neurodegenerative disorders.
Collapse
Affiliation(s)
- Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Wesley H. Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Judy J. Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine; Baltimore, Maryland, 21287, USA
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Paul A. Wender
- Departments of Chemistry and of Chemical and Systems Biology, Stanford University, Stanford, California, 94305, USA
| | - Michael D. Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| | - Paul M. Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland, 21287, USA
| |
Collapse
|
41
|
Seino K, Nakano T, Tanaka T, Hozumi Y, Topham MK, Goto K, Iseki K. Ablation of DGKα facilitates α-smooth muscle actin expression via the Smad and PKCδ signaling pathways during the acute phase of CCl 4 -induced hepatic injury. FEBS Open Bio 2024; 14:300-308. [PMID: 38105414 PMCID: PMC10839370 DOI: 10.1002/2211-5463.13749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/31/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Expression of α-smooth muscle actin (αSMA) is constitutive in vascular smooth muscle cells, but is induced in nonmuscle cells such as hepatic stellate cells (HSCs). HSCs play important roles in both physiological homeostasis and pathological response. HSC activation is characterized by αSMA expression, which is regulated by the TGFβ-induced Smad pathway. Recently, protein kinase C (PKC) was identified to regulate αSMA expression. Diacylglycerol kinase (DGK) metabolizes a second-messenger DG, thereby controlling components of DG-mediated signaling, such as PKC. In the present study we aimed to investigate the putative role of DGKα in αSMA expression. Use of a cellular model indicated that the DGK inhibitor R59949 promotes αSMA expression and PKCδ phosphorylation. It also facilitates Smad2 phosphorylation after 30 min of TGFβ stimulation. Furthermore, immunocytochemical analysis revealed that DGK inhibitor pretreatment without TGFβ stimulation engenders αSMA expression in a granular pattern, whereas DGK inhibitor pretreatment plus TGFβ stimulation significantly induces αSMA incorporation in stress fibers. Through animal model experiments, we observed that DGKα-knockout mice exhibit increased expression of αSMA in the liver after 48 h of carbon tetrachloride injection, together with enhanced phosphorylation levels of Smad2 and PKCδ. Together, these findings suggest that DGKα negatively regulates αSMA expression by acting on the Smad and PKCδ signaling pathways, which differentially regulate stress fiber incorporation and protein expression of αSMA, respectively.
Collapse
Affiliation(s)
- Keiko Seino
- Department of Anatomy and Cell BiologyYamagata University School of MedicineJapan
| | - Tomoyuki Nakano
- Department of Anatomy and Cell BiologyYamagata University School of MedicineJapan
| | - Toshiaki Tanaka
- Department of Anatomy and Cell BiologyYamagata University School of MedicineJapan
| | - Yasukazu Hozumi
- Department of Cell Biology and MorphologyAkita University Graduate School of MedicineJapan
| | | | - Kaoru Goto
- Department of Anatomy and Cell BiologyYamagata University School of MedicineJapan
| | - Ken Iseki
- Department of Emergency and Critical Care Medicine, School of MedicineFukushima Medical UniversityFukushimaJapan
| |
Collapse
|
42
|
Li W, Shen MY, Liu RB, Zhang JY, Li RY, Wang GG. Deletion of protein kinase C θ attenuates hepatic ischemia/reperfusion injury and further elucidates its mechanism in pathophysiology. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:1323-1330. [PMID: 39229579 PMCID: PMC11366945 DOI: 10.22038/ijbms.2024.77365.16730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 04/06/2024] [Indexed: 09/05/2024]
Abstract
Objectives Hepatic ischemia-reperfusion (HIR) is a severe process in pathophysiology that occurs clinically in hepatectomy, and hepatic transplantations. The present study aimed to investigate the effect of PKC θ deletion against HIR injury and elucidate its mechanism in pathophysiology. Materials and Methods HIR injury was induced in wild-type and PKC θ deletion mice treated with or without heme. The ALT and AST levels were determined to evaluate liver function. HIR injury was observed via histological examination. Oxidative stress and inflammatory response markers, and their signaling pathways were detected. Results The study found that PKC θ knockout decreased serum AST and ALT levels when compared to the WT mice. Furthermore, heme treatment significantly reduced the ALT and AST levels of the PKC θ deletion mice compared with the untreated PKC θ deletion mice. PKC θ deletion markedly elevated superoxide dismutase activity in the liver tissue, reduced malondialdehyde content in the tissue, and the serum TNF-α and IL-6 levels compared with the WT mice. Heme treatment was observed to elevate the activity of SOD and reduced MDA content and serum of TNF-α and IL 6 in the PKC θ deletion animals. Meanwhile, heme treatment increased HO-1 and Nrf 2 protein expression, and reduced the levels of TLR4, phosphorylated NF-κB, and IKB-α. Conclusion These findings suggested that PKC θ deletion ameliorates HIR, and heme treatment further improves HIR, which is related to regulation of PKC θ deletion on Nrf 2/HO-1 and TLR4/NF-κB/IKB α pathway.
Collapse
Affiliation(s)
- Wei Li
- Department of Pathophysiology, Wannan Medical College, Wuhu, China
| | - Meng-Yuan Shen
- School of Medical Imaging, Wannan Medical College, Wuhu, China
| | - Ruo-Bing Liu
- School of Clinical Medicine, Wannan Medical College, Wuhu, China
| | - Jun-Yang Zhang
- School of Medical Imaging, Wannan Medical College, Wuhu, China
| | - Rong-Yu Li
- Department of Immunology, Wannan Medical College, Wuhu, China
| | - Guo-Guang Wang
- Department of Pathophysiology, Wannan Medical College, Wuhu, China
| |
Collapse
|
43
|
Silnitsky S, Rubin SJS, Zerihun M, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases. Int J Mol Sci 2023; 24:17600. [PMID: 38139428 PMCID: PMC10743896 DOI: 10.3390/ijms242417600] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor-kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II).
Collapse
Affiliation(s)
- Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| |
Collapse
|
44
|
Leis HJ, Windischhofer W. Phospholipase D1 activity is crucial for cytosolic phospholipase A 2 -dependent prostaglandin E 2 formation in murine osteoblastic MC3T3-E1 cells. Prostaglandins Leukot Essent Fatty Acids 2023; 198-199:102592. [PMID: 37951067 DOI: 10.1016/j.plefa.2023.102592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/23/2023] [Accepted: 10/27/2023] [Indexed: 11/13/2023]
Abstract
In bone, prostaglandin E2 (PGE2) is highly osteogenic and formed by osteoblasts, a key modulatory event in the regulation of bone cell activity. MC3T3-E1 cells are widely used as an in vitro model of osteoblast function. It is still not clear which pathways contribute to the release of AA in these cells. In this study we have focussed on the contribution of phospholipase D (PLD) enzymes to osteoblastic PGE2 formation after stimulation with endothelin-1 (ET-1). Using specific inhibitors of PLD1 and PLD2 we could show that PGE2 formation was strictly dependent on PLD1 but not PLD2 activity and cytosolic phospholipase A2 (cPLA2) was activated by triggering through PLD1. We have identified diacyl glycerol (DAG) as a possible effector molecule which may serve as a triggering signal for PKC activation and subsequent cPLA2 phosphorylation.
Collapse
Affiliation(s)
- Hans Jörg Leis
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, University Hospital of Youth and Adolescence Medicine, Medical University of Graz, Auenbruggerplatz 34/2, Graz A-8036, Austria.
| | - Werner Windischhofer
- Research Unit of Analytical Mass Spectrometry, Cell Biology and Biochemistry of Inborn Errors of Metabolism, University Hospital of Youth and Adolescence Medicine, Medical University of Graz, Auenbruggerplatz 34/2, Graz A-8036, Austria
| |
Collapse
|
45
|
Noguchi S, Kajimoto T, Kumamoto T, Shingai M, Narasaki S, Urabe T, Imamura S, Harada K, Hide I, Tanaka S, Yanase Y, Nakamura SI, Tsutsumi YM, Sakai N. Features and mechanisms of propofol-induced protein kinase C (PKC) translocation and activation in living cells. Front Pharmacol 2023; 14:1284586. [PMID: 38026993 PMCID: PMC10662334 DOI: 10.3389/fphar.2023.1284586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Background and purpose: In this study, we aimed to elucidate the action mechanisms of propofol, particularly those underlying propofol-induced protein kinase C (PKC) translocation. Experimental approach: Various PKCs fused with green fluorescent protein (PKC-GFP) or other GFP-fused proteins were expressed in HeLa cells, and their propofol-induced dynamics were observed using confocal laser scanning microscopy. Propofol-induced PKC activation in cells was estimated using the C kinase activity receptor (CKAR), an indicator of intracellular PKC activation. We also examined PKC translocation using isomers and derivatives of propofol to identify the crucial structural motifs involved in this process. Key results: Propofol persistently translocated PKCα conventional PKCs and PKCδ from novel PKCs (nPKCs) to the plasma membrane (PM). Propofol translocated PKCδ and PKCη of nPKCs to the Golgi apparatus and endoplasmic reticulum, respectively. Propofol also induced the nuclear translocation of PKCζ of atypical PKCs or proteins other than PKCs, such that the protein concentration inside and outside the nucleus became uniform. CKAR analysis revealed that propofol activated PKC in the PM and Golgi apparatus. Moreover, tests using isomers and derivatives of propofol predicted that the structural motifs important for the induction of PKC and nuclear translocation are different. Conclusion and implications: Propofol induced the subtype-specific intracellular translocation of PKCs and activated PKCs. Additionally, propofol induced the nuclear translocation of PKCs and other proteins, probably by altering the permeability of the nuclear envelope. Interestingly, propofol-induced PKC and nuclear translocation may occur via different mechanisms. Our findings provide insights into the action mechanisms of propofol.
Collapse
Affiliation(s)
- Soma Noguchi
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Taketoshi Kajimoto
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Takuya Kumamoto
- Department of Synthetic Organic Chemistry, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masashi Shingai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Soshi Narasaki
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tomoaki Urabe
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Serika Imamura
- Department of Dental Anesthesiology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Sigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Yuhki Yanase
- Department of Pharmacotherapy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shun-Ichi Nakamura
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yasuo M. Tsutsumi
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
46
|
Onken MD, Erdmann-Gilmore P, Zhang Q, Thapa K, King E, Kaltenbronn KM, Noda SE, Makepeace CM, Goldfarb D, Babur Ö, Townsend RR, Blumer KJ. Protein Kinase Signaling Networks Driven by Oncogenic Gq/11 in Uveal Melanoma Identified by Phosphoproteomic and Bioinformatic Analyses. Mol Cell Proteomics 2023; 22:100649. [PMID: 37730182 PMCID: PMC10616553 DOI: 10.1016/j.mcpro.2023.100649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 08/22/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023] Open
Abstract
Metastatic uveal melanoma (UM) patients typically survive only 2 to 3 years because effective therapy does not yet exist. Here, to facilitate the discovery of therapeutic targets in UM, we have identified protein kinase signaling mechanisms elicited by the drivers in 90% of UM tumors: mutant constitutively active G protein α-subunits encoded by GNAQ (Gq) or GNA11 (G11). We used the highly specific Gq/11 inhibitor FR900359 (FR) to elucidate signaling networks that drive proliferation, metabolic reprogramming, and dedifferentiation of UM cells. We determined the effects of FR on the proteome and phosphoproteome of UM cells as indicated by bioinformatic analyses with CausalPath and site-specific gene set enrichment analysis. We found that inhibition of oncogenic Gq/11 caused deactivation of PKC, Erk, and the cyclin-dependent kinases CDK1 and CDK2 that drive proliferation. Inhibition of oncogenic Gq/11 in UM cells with low metastatic risk relieved inhibitory phosphorylation of polycomb-repressive complex subunits that regulate melanocytic redifferentiation. Site-specific gene set enrichment analysis, unsupervised analysis, and functional studies indicated that mTORC1 and 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 2 drive metabolic reprogramming in UM cells. Together, these results identified protein kinase signaling networks driven by oncogenic Gq/11 that regulate critical aspects of UM cell biology and provide targets for therapeutic investigation.
Collapse
Affiliation(s)
- Michael D Onken
- Department of Biochemistry and Molecular Biophysics, Washington University in St Louis, St Louis, Missouri, USA.
| | | | - Qiang Zhang
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kisan Thapa
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - Emily King
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Kevin M Kaltenbronn
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Sarah E Noda
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Carol M Makepeace
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Dennis Goldfarb
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA
| | - Özgün Babur
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts, USA
| | - R Reid Townsend
- Department of Medicine, Washington University in St Louis, St Louis, Missouri, USA
| | - Kendall J Blumer
- Department of Cell Biology and Physiology, Washington University in St Louis, St Louis, Missouri, USA.
| |
Collapse
|
47
|
Saiz-Baggetto S, Dolz-Edo L, Méndez E, García-Bolufer P, Marí M, Bañó MC, Fariñas I, Morante-Redolat JM, Igual JC, Quilis I. A Multimodel Study of the Role of Novel PKC Isoforms in the DNA Integrity Checkpoint. Int J Mol Sci 2023; 24:15796. [PMID: 37958781 PMCID: PMC10650207 DOI: 10.3390/ijms242115796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 10/22/2023] [Accepted: 10/29/2023] [Indexed: 11/15/2023] Open
Abstract
The protein kinase C (PKC) family plays important regulatory roles in numerous cellular processes. Saccharomyces cerevisiae contains a single PKC, Pkc1, whereas in mammals, the PKC family comprises nine isoforms. Both Pkc1 and the novel isoform PKCδ are involved in the control of DNA integrity checkpoint activation, demonstrating that this mechanism is conserved from yeast to mammals. To explore the function of PKCδ in a non-tumor cell line, we employed CRISPR-Cas9 technology to obtain PKCδ knocked-out mouse embryonic stem cells (mESCs). This model demonstrated that the absence of PKCδ reduced the activation of the effector kinase CHK1, although it suggested that other isoform(s) might contribute to this function. Therefore, we used yeast to study the ability of each single PKC isoform to activate the DNA integrity checkpoint. Our analysis identified that PKCθ, the closest isoform to PKCδ, was also able to perform this function, although with less efficiency. Then, by generating truncated and mutant versions in key residues, we uncovered differences between the activation mechanisms of PKCδ and PKCθ and identified their essential domains. Our work strongly supports the role of PKC as a key player in the DNA integrity checkpoint pathway and highlights the advantages of combining distinct research models.
Collapse
Affiliation(s)
- Sara Saiz-Baggetto
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
| | - Laura Dolz-Edo
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
- Departament de Biologia Cellular, Biologia Funcional i Antropologia Física, Universitat de València, 46100 Burjassot, Spain
| | - Ester Méndez
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
| | - Pau García-Bolufer
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
- Departament de Biologia Cellular, Biologia Funcional i Antropologia Física, Universitat de València, 46100 Burjassot, Spain
| | - Miquel Marí
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
- Departament de Biologia Cellular, Biologia Funcional i Antropologia Física, Universitat de València, 46100 Burjassot, Spain
| | - M. Carmen Bañó
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
| | - Isabel Fariñas
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
- Departament de Biologia Cellular, Biologia Funcional i Antropologia Física, Universitat de València, 46100 Burjassot, Spain
| | - José Manuel Morante-Redolat
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
- Departament de Biologia Cellular, Biologia Funcional i Antropologia Física, Universitat de València, 46100 Burjassot, Spain
| | - J. Carlos Igual
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
| | - Inma Quilis
- Departament de Bioquímica i Biologia Molecular, Universitat de València, 46100 Burjassot, Spain; (S.S.-B.); (L.D.-E.); (M.C.B.)
- Institut de Biotecnologia i Biomedicina (BIOTECMED), Universitat de València, 46100 Burjassot, Spain (I.F.); (J.M.M.-R.)
| |
Collapse
|
48
|
Wu J, Jonniya NA, Hirakis SP, Olivieri C, Veglia G, Kornev AP, Taylor SS. Protein Kinase Structure and Dynamics: Role of the αC-β4 Loop. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.31.555822. [PMID: 37693538 PMCID: PMC10491255 DOI: 10.1101/2023.08.31.555822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
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.
Collapse
Affiliation(s)
- Jian Wu
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037-0654, USA
| | - Nisha A. Jonniya
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037-0654, USA
| | - Sophia P. Hirakis
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92037-0654, USA
| | - Cristina Olivieri
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, MN 55455, USA
| | - Gianluigi Veglia
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, MN 55455, USA
- Department of Chemistry, University of Minnesota, MN 55455, USA
| | - Alexandr P. Kornev
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037-0654, USA
| | - Susan S. Taylor
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92037-0654, USA
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92037-0654, USA
| |
Collapse
|
49
|
Leroux AE, Biondi RM. The choreography of protein kinase PDK1 and its diverse substrate dance partners. Biochem J 2023; 480:1503-1532. [PMID: 37792325 DOI: 10.1042/bcj20220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
The protein kinase PDK1 phosphorylates at least 24 distinct substrates, all of which belong to the AGC protein kinase group. Some substrates, such as conventional PKCs, undergo phosphorylation by PDK1 during their synthesis and subsequently get activated by DAG and Calcium. On the other hand, other substrates, including members of the Akt/PKB, S6K, SGK, and RSK families, undergo phosphorylation and activation downstream of PI3-kinase signaling. This review presents two accepted molecular mechanisms that determine the precise and timely phosphorylation of different substrates by PDK1. The first mechanism involves the colocalization of PDK1 with Akt/PKB in the presence of PIP3. The second mechanism involves the regulated docking interaction between the hydrophobic motif (HM) of substrates and the PIF-pocket of PDK1. This interaction, in trans, is equivalent to the molecular mechanism that governs the activity of AGC kinases through their HMs intramolecularly. PDK1 has been instrumental in illustrating the bi-directional allosteric communication between the PIF-pocket and the ATP-binding site and the potential of the system for drug discovery. PDK1's interaction with substrates is not solely regulated by the substrates themselves. Recent research indicates that full-length PDK1 can adopt various conformations based on the positioning of the PH domain relative to the catalytic domain. These distinct conformations of full-length PDK1 can influence the interaction and phosphorylation of substrates. Finally, we critically discuss recent findings proposing that PIP3 can directly regulate the activity of PDK1, which contradicts extensive in vitro and in vivo studies conducted over the years.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| |
Collapse
|
50
|
Pei J, Cong Q. Computational analysis of regulatory regions in human protein kinases. Protein Sci 2023; 32:e4764. [PMID: 37632170 PMCID: PMC10503413 DOI: 10.1002/pro.4764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/08/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
Eukaryotic proteins often feature modular domain structures comprising globular domains that are connected by linker regions and intrinsically disordered regions that may contain important functional motifs. The intramolecular interactions of globular domains and nonglobular regions can play critical roles in different aspects of protein function. However, studying these interactions and their regulatory roles can be challenging due to the flexibility of nonglobular regions, the long insertions separating interacting modules, and the transient nature of some interactions. Obtaining the experimental structures of multiple domains and functional regions is more difficult than determining the structures of individual globular domains. High-quality structural models generated by AlphaFold offer a unique opportunity to study intramolecular interactions in eukaryotic proteins. In this study, we systematically explored intramolecular interactions between human protein kinase domains (KDs) and potential regulatory regions, including globular domains, N- and C-terminal tails, long insertions, and distal nonglobular regions. Our analysis identified intramolecular interactions between human KDs and 35 different types of globular domains, exhibiting a variety of interaction modes that could contribute to orthosteric or allosteric regulation of kinase activity. We also identified prevalent interactions between human KDs and their flanking regions (N- and C-terminal tails). These interactions exhibit group-specific characteristics and can vary within each specific kinase group. Although long-range interactions between KDs and nonglobular regions are relatively rare, structural details of these interactions offer new insights into the regulation mechanisms of several kinases, such as HASPIN, MAPK7, MAPK15, and SIK1B.
Collapse
Affiliation(s)
- Jimin Pei
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| | - Qian Cong
- Eugene McDermott Center for Human Growth and DevelopmentUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Department of BiophysicsUniversity of Texas Southwestern Medical CenterDallasTexasUSA
- Harold C. Simmons Comprehensive Cancer CenterUniversity of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|