1
|
Wang H, Zhang X, Wang D, Jiang Q, Sun Y, Zhao B, Liang Z, Qing G, Jiang B, Zhang L, Zhang Y. Affinity peptide ligands: new tools for chasing non-canonical N-phosphoproteome. Chem Sci 2025:d5sc01557j. [PMID: 40290335 PMCID: PMC12022672 DOI: 10.1039/d5sc01557j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025] Open
Abstract
The enrichment of protein N-phosphorylation encounters substantial challenges due to the inherent instability of the N-P bond, severely impeding the manifestation of its biological functions. Traditional enrichment methods often rely on antibodies, organic solvents and metal ion interactions, which are limited by lack of universality, potential degradation of sample integrity, or reduced selectivity for N-phosphorylation. To overcome these challenges, we innovatively capitalized phage display technology to identify affinity peptides that specifically bind to the N-PO3 group. By functionalizing magnetic nanoparticles with the affinity peptide, we developed a novel, organic solvent- and metal-free enrichment strategy that enhanced both the selectivity and efficiency for all three types of N-phosphopeptide capture under neutral conditions, ensuring superior preservation of sample integrity and allowing more accurate proteomic analysis. This strategy has demonstrated robust enrichment capabilities for both prokaryotic and eukaryotic samples. In HeLa cells, 1995 novel N-phosphorylation sites were identified, representing a substantial increase of 2- to 5-fold in detection depth over previous approaches and significantly expanding the scale of the N-phosphoproteome database. Additionally, it was discovered that N-phosphorylation modification was highly concentrated in the nucleus. By integrating the nuclear isolation technique, 1296 N-phosphorylation sites were identified for the first time, offering new leads for uncovering the functions of N-phosphorylation in nuclear proteins. Finally, in conjunction with the quantitative proteomics method, the dynamic changes in N-phosphorylation modification during the progression of Alzheimer's disease were investigated, providing fresh perspectives on the research of AD pathogenesis. Overall, this work not only presents a new approach for efficient enrichment of N-phosphopeptides but also advances the functional study of N-phosphorylated proteins in physiological and pathological processes.
Collapse
Affiliation(s)
- He Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Xiaoyu Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Dongdong Wang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Qianqian Jiang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
- University of Chinese Academy of Sciences Beijing 100049 China
| | - Yue Sun
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Baofeng Zhao
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Zhen Liang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Guangyan Qing
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Bo Jiang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Lihua Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| | - Yukui Zhang
- State Key Laboratory of Medical Proteomics, National Chromatographic R&A Center, CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences Dalian 116023 China
| |
Collapse
|
2
|
Bilgin N, Hintzen JCJ, Mecinović J. Chemical tools for probing histidine modifications. Chem Commun (Camb) 2025; 61:3805-3820. [PMID: 39936705 DOI: 10.1039/d4cc06586g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Histidine is a unique amino acid with critical roles in protein structure and function, ranging from metal ion binding to enzyme catalysis. Histidine residues in proteins also undergo diverse posttranslational modifications, including methylation, phosphorylation and hydroxylation, by various enzymes, some of them being only recently identified and characterised. In this review, we describe the development of chemical tools for understanding the role of histidine residues in chemical and biological systems. We spotlight the application of histidine analogues in probing biomedically important posttranslational modifications of histidine residues in proteins, and we highlight novel bioconjugation methods that enable chemoselective modifications of histidine residues in peptides and proteins.
Collapse
Affiliation(s)
- Nurgül Bilgin
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| | - Jordi C J Hintzen
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jasmin Mecinović
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, Campusvej 55, 5230 Odense, Denmark.
| |
Collapse
|
3
|
Jedele S, Allegrini B, Guizouarn H, Etchebest C. Investigating the Dynamics of the KCNN4 Channel: From the Determination of the Complete K + Permeation Pathway Across the Channel to Its Opening by PIP2. J Chem Inf Model 2025; 65:2116-2128. [PMID: 39928097 DOI: 10.1021/acs.jcim.4c01711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2025]
Abstract
KCNN4 is a calcium (Ca)-activated potassium channel for which Ca2+ sensitivity is conferred by calmodulin (CaM) that constitutively binds to the channel. Until the main part of the structure bound to CaM has been resolved, in silico studies had used homology models derived from the well characterized transmembrane domain of other K+ channels, limiting the functional investigation to this particular region. Thus, how the regulatory domains of KCNN4 communicate with each other and where the possible gates are located across the complete structure are still not well understood. Here we present for the first time results obtained from the investigation of full-length models of the channel in different conformational states and molecular contexts using classical all-atom molecular dynamic simulations. The simulations covered two activated states (open and closed) and a preactivated state of the channel embedded in a simple membrane model and a model of red blood cell membrane, where the channel is functionally expressed in vivo. Surprisingly, the intracellular domain was refractory to the entrance of K+, whatever the state of the channel was, allowing the K+ ions to enter and exit the channel only through two newly identified restrained diffusion spots. Inside the channel, the K+ flux was controlled by the V282 residue closing the pore region when the CaM N-lobes were not bound. This flux was compatible with the passage of fully or partially hydrated K+, depending on the opening level. Finally, the presence of phosphatidylinositol-4,5-bisphosphate (PIP2), a well-known K+-channel modulator, in a putative binding site of KCNN4 clearly facilitated the opening of the V282 restriction. Thus, in addition to the elucidation of the possible complete K+ permeation pathway throughout KCNN4, our results confirmed the direct activatory role of PIP2, associated with the channel opening, and provide a first insight into the architecture and the behavior of the complete intracellular region of KCNN4.
Collapse
Affiliation(s)
- Stephane Jedele
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| | | | | | - Catherine Etchebest
- Université Paris Cité and Université des Antilles, Inserm, BIGR, F-75015 Paris, France
| |
Collapse
|
4
|
Zhang Z, Patel R, Luo ZQ, Das C. Cryo-EM Detection of AMPylated Histidine Implies Covalent Catalysis in AMPylation Mediated by a Bacterial Effector. J Mol Biol 2025; 437:168917. [PMID: 39694182 DOI: 10.1016/j.jmb.2024.168917] [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: 06/27/2024] [Revised: 12/09/2024] [Accepted: 12/12/2024] [Indexed: 12/20/2024]
Abstract
AMPylation is a post-translational modification (PTM) whereby adenosine monophosphate (AMP) from adenosine triphosphate (ATP) is transferred onto protein hydroxyl groups of serine, threonine, or tyrosine. Recently, an actin-dependent AMPylase namely LnaB from the bacterial pathogen Legionella pneumophila was found to AMPylate phosphate groups of phosphoribosylated ubiquitin and Src family kinases. LnaB represents an evolutionarily distinct family of AMPylases with conserved active site Ser-His-Glu residues. Here, we capture the structure of the LnaB-actin complex in a putative intermediate state via single-particle cryogenic electron microscopy (cryo-EM) and find that the catalytic histidine of LnaB is covalently attached to AMP through a phosphoramidate linkage at the Nδ1 atom. This observation provides direct structural evidence of histidine AMPylation as a PTM and implies the possibility of covalent catalysis in LnaB-mediated AMPylation, a mechanism distinct from known AMPylases. Subsequent biochemical studies confirm the observed AMP binding site and provide additional insights into the catalytic properties of LnaB. Together, our work highlights the power of cryo-EM in capturing labile PTMs and transient species during enzymatic reactions, while opening new avenues of mechanistic investigation into the LnaB family.
Collapse
Affiliation(s)
- Zhengrui Zhang
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Rishi Patel
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA
| | - Zhao-Qing Luo
- Department of Biological Sciences, Purdue Institute for Inflammation, Immunology and Infectious Disease, Purdue University, West Lafayette, IN 47907, USA
| | - Chittaranjan Das
- Department of Chemistry, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
5
|
Zheng Z, Cheng Y, Li P, Heng Tan CS. Covalent Modification of Protein by Chemical Probe in Living Cells for Structural and Interaction Studies. Chembiochem 2025; 26:e202400715. [PMID: 39380164 DOI: 10.1002/cbic.202400715] [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: 08/31/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Cellular activities are predominantly carried out by proteins that can dynamically adopt different structural conformations and differentially interact with other biomolecules according to cellular needs. Chemical probes are small molecules used to selectively interact and modulate the activities of specific proteins to study their functions such as the validation of potential drug targets. The remarkable performance of AlphaFold algorithms in the prediction of protein structures has pivoted interest toward elucidating the intracellular dynamics of protein structural conformation where covalent modification of proteins by chemical probes could be used to shed light upon. However, due to the barrier to entry by cell membrane and the general unfavorable reactive conditions of the intracellular environment, most studies using reactive chemical probes are still conducted on purified proteins and cell lysates. Nevertheless, recent progresses have been made in designing chemical probes with improved membrane permeability, stability and reactivity. This paper surveys the literature on recent advancements in membrane-permeable chemical probes and their applications with protein mass spectrometry for the intracellular studies of protein structural conformations and biomolecular interactions.
Collapse
Affiliation(s)
- Zhenxiang Zheng
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China PR
| | - Yuyu Cheng
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China PR
| | - Pengfei Li
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China PR
| | - Chris Soon Heng Tan
- Department of Chemistry and Research Center for Chemical Biology and Omics Analysis, College of Science, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China PR
| |
Collapse
|
6
|
Thi Hong Van N, Hyun Nam J. Intermediate conductance calcium-activated potassium channel (KCa3.1) in cancer: Emerging roles and therapeutic potentials. Biochem Pharmacol 2024; 230:116573. [PMID: 39396649 DOI: 10.1016/j.bcp.2024.116573] [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: 07/20/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024]
Abstract
The KCa3.1 channel (also known as the KCNN4, IK1, or SK4 channel) is an intermediate-conductance calcium-activated potassium channel that regulates the membrane potential and maintains calcium homeostasis. Recently, KCa3.1 channels have attracted increasing attention because of their diverse roles in various types of cancers. In cancer cells, KCa3.1 channels regulate key processes, including cell proliferation, cell cycle, migration, invasion, tumor microenvironments, and therapy resistance. In addition, abnormal KCa3.1 expression in cancers is utilized to distinguish between tumor and normal tissues, classify cancer stages, and predict patient survival outcomes. This review comprehensively examines the current understanding of the contribution of KCa3.1 channels to tumor formation, metastasis, and its mechanisms. We evaluated the potential of KCa3.1 as a biomarker for cancer diagnosis and prognosis. Finally, we discuss the advances and challenges of applying KCa3.1 modulators in cancer treatment and propose approaches to overcome these obstacles. In summary, this review highlights the importance of this ion channel as a potent therapeutic target and prognostic biomarker of cancer.
Collapse
Affiliation(s)
- Nhung Thi Hong Van
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea
| | - Joo Hyun Nam
- Department of Physiology, Dongguk University College of Medicine, Gyeongju 38066, Republic of Korea; Channelopathy Research Center (CRC), Dongguk University College of Medicine, Goyang 10326, Republic of Korea.
| |
Collapse
|
7
|
Martyn GD, Kalagiri R, Veggiani G, Stanfield RL, Choudhuri I, Sala M, Meisenhelder J, Chen C, Biswas A, Levy RM, Lyumkis D, Wilson IA, Hunter T, Sidhu SS. Using phage display for rational engineering of a higher affinity humanized 3'phosphohistidine-specific antibody. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621849. [PMID: 39574610 PMCID: PMC11580931 DOI: 10.1101/2024.11.04.621849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2024]
Abstract
Histidine phosphorylation (pHis) is a non-canonical post-translational modification (PTM) that is historically understudied due to a lack of robust reagents that are required for its investigation, such as high affinity pHis-specific antibodies. Engineering pHis-specific antibodies is very challenging due to the labile nature of the phosphoramidate (P-N) bond and the stringent requirements for selective recognition of the two isoforms, 1-phosphohistidine (1-pHis) and 3-phosphohistidine (3-pHis). Here, we present a strategy for in vitro engineering of antibodies for detection of native 3-pHis targets. Specifically, we humanized the rabbit SC44-8 anti-3-pTza (a stable 3-pHis mimetic) mAb into a scaffold (herein referred to as hSC44) that was suitable for phage display. We then constructed six unique Fab phage-displayed libraries using the hSC44 scaffold and selected high affinity 3-pHis binders. Our selection strategy was carefully designed to enrich antibodies that bound 3-pHis with high affinity and had specificity for 3-pHis versus 3-pTza. hSC44.20N32F L , the best engineered antibody, has an ∼10-fold higher affinity for 3-pHis than the parental hSC44. Eleven new Fab structures, including the first reported antibody-pHis peptide structures were solved by X-ray crystallography. Structural and quantum mechanical calculations provided molecular insights into 3-pHis and 3-pTza discrimination by different hSC44 variants and their affinity increase obtained through in vitro engineering. Furthermore, we demonstrate the utility of these newly developed high-affinity 3-pHis-specific antibodies for recognition of pHis proteins in mammalian cells by immunoblotting and immunofluorescence staining. Overall, our work describes a general method for engineering PTM-specific antibodies and provides a set of novel antibodies for further investigations of the role of 3-pHis in cell biology. Significance Statement Histidine phosphorylation is an elusive PTM whose role in mammalian cell biology is largely unknown due to the lack of robust tools and methods for its analysis. Here we report the development of antibodies with unprecedented affinity and specificity towards 3-pHis and present the first crystal structures of a pHis peptide in complex with an antibody. Finally, we show how these antibodies can be used in standard molecular biology workflows to investigate pHis-dependent biology.
Collapse
Affiliation(s)
- Gregory D. Martyn
- School of Pharmacy, University of Waterloo, 10 Victoria St A, Kitchener, ON, N2G 1C5, Canada
| | - Rajasree Kalagiri
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Gianluca Veggiani
- School of Pharmacy, University of Waterloo, 10 Victoria St A, Kitchener, ON, N2G 1C5, Canada
- Division of Biotechnology and Molecular Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
- Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Robyn L. Stanfield
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, 92037, USA
| | - Indrani Choudhuri
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Margaux Sala
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Jill Meisenhelder
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Chao Chen
- School of Pharmacy, University of Waterloo, 10 Victoria St A, Kitchener, ON, N2G 1C5, Canada
| | - Avik Biswas
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
- Department of Physics, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ronald M. Levy
- Center for Biophysics and Computational Biology, and Department of Chemistry, Temple University, Philadelphia, PA, 19122, USA
| | - Dmitry Lyumkis
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Ian A. Wilson
- Department of Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA, 92037, USA
- The Skaggs Institute for Chemical Biology, Scripps Research, La Jolla, California, 92037, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, 92037, USA
| | - Sachdev S. Sidhu
- School of Pharmacy, University of Waterloo, 10 Victoria St A, Kitchener, ON, N2G 1C5, Canada
| |
Collapse
|
8
|
Zhai Y, Zhang X, Chen Z, Yan D, Zhu L, Zhang Z, Wang X, Tian K, Huang Y, Yang X, Sun W, Wang D, Tsai YH, Luo T, Li G. Global profiling of functional histidines in live cells using small-molecule photosensitizer and chemical probe relay labelling. Nat Chem 2024; 16:1546-1557. [PMID: 38834725 DOI: 10.1038/s41557-024-01545-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 04/26/2024] [Indexed: 06/06/2024]
Abstract
Recent advances in chemical proteomics have focused on developing chemical probes that react with nucleophilic amino acid residues. Although histidine is an attractive candidate due to its importance in enzymatic catalysis, metal binding and protein-protein interaction, its moderate nucleophilicity poses challenges. Its modification is frequently influenced by cysteine and lysine, which results in poor selectivity and narrow proteome coverage. Here we report a singlet oxygen and chemical probe relay labelling method that achieves high selectivity towards histidine. Libraries of small-molecule photosensitizers and chemical probes were screened to optimize histidine labelling, enabling histidine profiling in live cells with around 7,200 unique sites. Using NMR spectroscopy and X-ray crystallography, we characterized the reaction mechanism and the structures of the resulting products. We then applied this method to discover unannotated histidine sites key to enzymatic activity and metal binding in select metalloproteins. This method also revealed the accessibility change of histidine mediated by protein-protein interaction that influences select protein subcellular localization, underscoring its capability in discovering functional histidines.
Collapse
Affiliation(s)
- Yansheng Zhai
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xinyu Zhang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- State Key Laboratory of Crop Stress Biology for Arid Areas, College of Life Sciences, Northwest A & F University, Yangling, China
| | - Zijing Chen
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Ministry of Education and Beijing National Laboratory for Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | | | - Lin Zhu
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Zhe Zhang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xianghe Wang
- Synthetic and Functional Biomolecules Center, Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
| | - Kailu Tian
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yan Huang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Xi Yang
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Wen Sun
- State Key Laboratory of Fine Chemicals, Frontiers Science Center for Smart Materials Oriented Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Dong Wang
- Shenzhen University, Shenzhen, China
| | - Yu-Hsuan Tsai
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Tuoping Luo
- Key Laboratory of Bioorganic Chemistry and Molecular Engineering, Ministry of Education and Beijing National Laboratory for Molecular Science, College of Chemistry and Molecular Engineering, Peking University, Beijing, China
- Institute of Molecular Physiology, Shenzhen Bay Laboratory, Shenzhen, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Gang Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
9
|
Gary CR, Acharige NPN, Oyewumi TO, Pflum MKH. Kinase-catalyzed biotinylation for discovery and validation of substrates to multispecificity kinases NME1 and NME2. J Biol Chem 2024; 300:107588. [PMID: 39032654 PMCID: PMC11375270 DOI: 10.1016/j.jbc.2024.107588] [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: 09/18/2023] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Protein phosphorylation by kinases regulates mammalian cell functions, such as growth, division, and signal transduction. Among human kinases, NME1 and NME2 are associated with metastatic tumor suppression but remain understudied due to the lack of tools to monitor their cellular substrates. In particular, NME1 and NME2 are multispecificity kinases phosphorylating serine, threonine, histidine, and aspartic acid residues of substrate proteins, and the heat and acid sensitivity of phosphohistidine and phosphoaspartate complicates substrate discovery and validation. To provide new substrate monitoring tools, we established the γ-phosphate-modified ATP analog, ATP-biotin, as a cosubstrate for phosphorylbiotinylation of NME1 and NME2 cellular substrates. Building upon this ATP-biotin compatibility, the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method enabled validation of a known substrate and the discovery of seven NME1 and three NME2 substrates. Given the paucity of methods to study kinase substrates, ATP-biotin and the Kinase-catalyzed Biotinylation with Inactivated Lysates for Discovery of Substrates method are valuable tools to characterize the roles of NME1 and NME2 in human cell biology.
Collapse
Affiliation(s)
- Chelsea R Gary
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA
| | | | | | - Mary Kay H Pflum
- Department of Chemistry, Wayne State University, Detroit, Michigan, USA.
| |
Collapse
|
10
|
Ning J, Sala M, Reina J, Kalagiri R, Hunter T, McCullough BS. Histidine Phosphorylation: Protein Kinases and Phosphatases. Int J Mol Sci 2024; 25:7975. [PMID: 39063217 PMCID: PMC11277029 DOI: 10.3390/ijms25147975] [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: 06/07/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Phosphohistidine (pHis) is a reversible protein post-translational modification (PTM) that is currently poorly understood. The P-N bond in pHis is heat and acid-sensitive, making it more challenging to study than the canonical phosphoamino acids pSer, pThr, and pTyr. As advancements in the development of tools to study pHis have been made, the roles of pHis in cells are slowly being revealed. To date, a handful of enzymes responsible for controlling this modification have been identified, including the histidine kinases NME1 and NME2, as well as the phosphohistidine phosphatases PHPT1, LHPP, and PGAM5. These tools have also identified the substrates of these enzymes, granting new insights into previously unknown regulatory mechanisms. Here, we discuss the cellular function of pHis and how it is regulated on known pHis-containing proteins, as well as cellular mechanisms that regulate the activity of the pHis kinases and phosphatases themselves. We further discuss the role of the pHis kinases and phosphatases as potential tumor promoters or suppressors. Finally, we give an overview of various tools and methods currently used to study pHis biology. Given their breadth of functions, unraveling the role of pHis in mammalian systems promises radical new insights into existing and unexplored areas of cell biology.
Collapse
Affiliation(s)
- Jia Ning
- Correspondence: (J.N.); (B.S.M.)
| | | | | | | | | | - Brandon S. McCullough
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; (M.S.); (J.R.); (R.K.); (T.H.)
| |
Collapse
|
11
|
Cozzolino M, Panyi G. Intracellular acidity impedes KCa3.1 activation by Riluzole and SKA-31. Front Pharmacol 2024; 15:1380655. [PMID: 38638868 PMCID: PMC11024243 DOI: 10.3389/fphar.2024.1380655] [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: 02/01/2024] [Accepted: 03/05/2024] [Indexed: 04/20/2024] Open
Abstract
Background The unique microenvironment in tumors inhibits the normal functioning of tumor-infiltrating lymphocytes, leading to immune evasion and cancer progression. Over-activation of KCa3.1 using positive modulators has been proposed to rescue the anti-tumor response. One of the key characteristics of the tumor microenvironment is extracellular acidity. Herein, we analyzed how intra- and extracellular pH affects K+ currents through KCa3.1 and if the potency of two of its positive modulators, Riluzole and SKA-31, is pH sensitive. Methods Whole-cell patch-clamp was used to measure KCa3.1 currents either in activated human peripheral lymphocytes or in CHO cells transiently transfected with either the H192A mutant or wild-type hKCa3.1 in combination with T79D-Calmodulin, or with KCa2.2. Results We found that changes in the intra- and extracellular pH minimally influenced the KCa3.1-mediated K+ current. Extracellular pH, in the range of 6.0-8.0, does not interfere with the capacity of Riluzole and SKA-31 to robustly activate the K+ currents through KCa3.1. Contrariwise, an acidic intracellular solution causes a slow, but irreversible loss of potency of both the activators. Using different protocols of perfusion and depolarization we demonstrated that the loss of potency is strictly time and pH-dependent and that this peculiar effect can be observed with a structurally similar channel KCa2.2. While two different point mutations of both KCa3.1 (H192A) and its associated protein Calmodulin (T79D) do not limit the effect of acidity, increasing the cytosolic Ca2+ concentration to saturating levels eliminated the loss-of-potency phenotype. Conclusion Based on our data we conclude that KCa3.1 currents are not sensitive the either the intracellular or the extracellular pH in the physiological and pathophysiological range. However, intracellular acidosis in T cells residing in the tumor microenvironment could hinder the potentiating effect of KCa3.1 positive modulators administered to boost their activity. Further research is warranted both to clarify the molecular interactions between the modulators and KCa3.1 at different intracellular pH conditions and to define whether this loss of potency can be observed in cancer models as well.
Collapse
Affiliation(s)
| | - Gyorgy Panyi
- Department of Biophysics and Cell Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
12
|
Wang H, Gaston R, Ahmed KT, Dudley GB, Barrios AM. Derivatives of the Fungal Natural Product Illudalic Acid Inhibit the Activity of Protein Histidine Phosphatase PHPT1. ChemMedChem 2023; 18:e202300187. [PMID: 37267298 PMCID: PMC10443188 DOI: 10.1002/cmdc.202300187] [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: 04/05/2023] [Revised: 05/30/2023] [Accepted: 05/31/2023] [Indexed: 06/04/2023]
Abstract
PHPT1 is a protein histidine phosphatase that has been implicated in several disease pathways, but the chemical tools necessary to study the biological roles of this enzyme and investigate its utility as a therapeutic target have yet to be developed. To this end, the discovery of PHPT1 inhibitors is an area of significant interest. Here, we report an investigation of illudalic acid and illudalic acid analog-based inhibition of PHPT1 activity. Four of the seven analogs investigated had IC50 values below 5 μM, with the most potent compound (IA1-8H2) exhibiting an IC50 value of 3.4±0.7 μM. Interestingly, these compounds appear to be non-covalent, non-competitive inhibitors of PHPT1 activity, in contrast to other recently reported PHPT1 inhibitors. Mutating the three cysteine residues to alanine has no effect on inhibition, indicating that cysteine is not critical for interactions between inhibitor and enzyme.
Collapse
Affiliation(s)
- Hanfei Wang
- Department of Medicinal Chemistry, University of Utah College of Pharmacy, Salt Lake City, UT 84112
| | - Robert Gaston
- Department of Chemistry, West Virginia University, Morgantown, WV 26506
| | - Kh Tanvir Ahmed
- Department of Chemistry, West Virginia University, Morgantown, WV 26506
| | - Gregory B. Dudley
- Department of Chemistry, West Virginia University, Morgantown, WV 26506
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah College of Pharmacy, Salt Lake City, UT 84112
| |
Collapse
|
13
|
Soret B, Hense J, Lüdtke S, Thale I, Schwab A, Düfer M. Pancreatic K Ca3.1 channels in health and disease. Biol Chem 2023; 404:339-353. [PMID: 36571487 DOI: 10.1515/hsz-2022-0232] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 11/24/2022] [Indexed: 12/27/2022]
Abstract
Ion channels play an important role for regulation of the exocrine and the endocrine pancreas. This review focuses on the Ca2+-regulated K+ channel KCa3.1, encoded by the KCNN4 gene, which is present in both parts of the pancreas. In the islets of Langerhans, KCa3.1 channels are involved in the regulation of membrane potential oscillations characterizing nutrient-stimulated islet activity. Channel upregulation is induced by gluco- or lipotoxic conditions and might contribute to micro-inflammation and impaired insulin release in type 2 diabetes mellitus as well as to diabetes-associated renal and vascular complications. In the exocrine pancreas KCa3.1 channels are expressed in acinar and ductal cells. They are thought to play a role for anion secretion during digestion but their physiological role has not been fully elucidated yet. Pancreatic carcinoma, especially pancreatic ductal adenocarcinoma (PDAC), is associated with drastic overexpression of KCa3.1. For pharmacological targeting of KCa3.1 channels, we are discussing the possible benefits KCa3.1 channel inhibitors might provide in the context of diabetes mellitus and pancreatic cancer, respectively. We are also giving a perspective for the use of a fluorescently labeled derivative of the KCa3.1 blocker senicapoc as a tool to monitor channel distribution in pancreatic tissue. In summary, modulating KCa3.1 channel activity is a useful strategy for exo-and endocrine pancreatic disease but further studies are needed to evaluate its clinical suitability.
Collapse
Affiliation(s)
- Benjamin Soret
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Jurek Hense
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Simon Lüdtke
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| | - Insa Thale
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Corrensstraße 48, D-48149 Münster, Germany
| | - Albrecht Schwab
- University of Münster, Institute of Physiology II, Robert-Koch-Straße 27b, D-48149 Münster, Germany
| | - Martina Düfer
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, Department of Pharmacology, Corrensstraße 48, D-48149 Münster, Germany
| |
Collapse
|
14
|
Lin D, Li L, Chen WB, Chen J, Ren D, Zheng ZH, Zhao C, Zhong Y, Luo B, Jing H, Chen P, Zou S, Lai X, Zhou T, Ding N, Li L, Pan BX, Fei E. LHPP, a risk factor for major depressive disorder, regulates stress-induced depression-like behaviors through its histidine phosphatase activity. Mol Psychiatry 2023; 28:908-918. [PMID: 36460727 DOI: 10.1038/s41380-022-01893-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 11/14/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022]
Abstract
Histidine phosphorylation (pHis), occurring on the histidine of substrate proteins, is a hidden phosphoproteome that is poorly characterized in mammals. LHPP (phospholysine phosphohistidine inorganic pyrophosphate phosphatase) is one of the histidine phosphatases and its encoding gene was recently identified as a susceptibility gene for major depressive disorder (MDD). However, little is known about how LHPP or pHis contributes to depression. Here, by using integrative approaches of genetics, behavior and electrophysiology, we observed that LHPP in the medial prefrontal cortex (mPFC) was essential in preventing stress-induced depression-like behaviors. While genetic deletion of LHPP per se failed to affect the mice's depression-like behaviors, it markedly augmented the behaviors upon chronic social defeat stress (CSDS). This augmentation could be recapitulated by the local deletion of LHPP in mPFC. By contrast, overexpressing LHPP in mPFC increased the mice's resilience against CSDS, suggesting a critical role of mPFC LHPP in stress-induced depression. We further found that LHPP deficiency increased the levels of histidine kinases (NME1/2) and global pHis in the cortex, and decreased glutamatergic transmission in mPFC upon CSDS. NME1/2 served as substrates of LHPP, with the Aspartic acid 17 (D17), Threonine 54 (T54), or D214 residue within LHPP being critical for its phosphatase activity. Finally, reintroducing LHPP, but not LHPP phosphatase-dead mutants, into the mPFC of LHPP-deficient mice reversed their behavioral and synaptic deficits upon CSDS. Together, these results demonstrate a critical role of LHPP in regulating stress-related depression and provide novel insight into the pathogenesis of MDD.
Collapse
Affiliation(s)
- Dong Lin
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Luhui Li
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Wen-Bing Chen
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Jiang Chen
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Zhi-Heng Zheng
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Changqin Zhao
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Yanzi Zhong
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Bin Luo
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Hongyang Jing
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Peng Chen
- Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Suqi Zou
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Xinsheng Lai
- School of Life Sciences, Nanchang University, Nanchang, 330031, China.,Institute of Life Science, Nanchang University, Nanchang, 330031, China
| | - Tian Zhou
- School of Basic Medical Sciences, Nanchang University, Nanchang, 330031, China
| | - Ning Ding
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Lei Li
- School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Bing-Xing Pan
- School of Life Sciences, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang, 330031, China. .,Institute of Life Science, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
15
|
Sakai Y, Hanafusa H, Hisamoto N, Matsumoto K. Histidine dephosphorylation of the Gβ protein GPB-1 promotes axon regeneration in C. elegans. EMBO Rep 2022; 23:e55076. [PMID: 36278516 PMCID: PMC9724660 DOI: 10.15252/embr.202255076] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 12/12/2022] Open
Abstract
Histidine phosphorylation is an emerging noncanonical protein phosphorylation in animals, yet its physiological role remains largely unexplored. The protein histidine phosphatase (PHPT1) was recently identified for the first time in mammals. Here, we report that PHIP-1, an ortholog of PHPT1 in Caenorhabditis elegans, promotes axon regeneration by dephosphorylating GPB-1 Gβ at His-266 and inactivating GOA-1 Goα signaling, a negative regulator of axon regeneration. Overexpression of the histidine kinase NDK-1 also inhibits axon regeneration via GPB-1 His-266 phosphorylation. Thus, His-phosphorylation plays an antiregenerative role in C. elegans. Furthermore, we identify a conserved UNC-51/ULK kinase that functions in autophagy as a PHIP-1-binding protein. We demonstrate that UNC-51 phosphorylates PHIP-1 at Ser-112 and activates its catalytic activity and that this phosphorylation is required for PHIP-1-mediated axon regeneration. This study reveals a molecular link from ULK to protein histidine phosphatase, which facilitates axon regeneration by inhibiting trimeric G protein signaling.
Collapse
Affiliation(s)
- Yoshiki Sakai
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Hiroshi Hanafusa
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Naoki Hisamoto
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| | - Kunihiro Matsumoto
- Division of Biological Science, Graduate School of ScienceNagoya UniversityNagoyaJapan
| |
Collapse
|
16
|
Lin Y, Zhao YJ, Zhang HL, Hao WJ, Zhu RD, Wang Y, Hu W, Zhou RP. Regulatory role of KCa3.1 in immune cell function and its emerging association with rheumatoid arthritis. Front Immunol 2022; 13:997621. [PMID: 36275686 PMCID: PMC9580404 DOI: 10.3389/fimmu.2022.997621] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/16/2022] [Indexed: 11/25/2022] Open
Abstract
Rheumatoid arthritis (RA) is a common autoimmune disease characterized by chronic inflammation. Immune dysfunction is an essential mechanism in the pathogenesis of RA and directly linked to synovial inflammation and cartilage/bone destruction. Intermediate conductance Ca2+-activated K+ channel (KCa3.1) is considered a significant regulator of proliferation, differentiation, and migration of immune cells by mediating Ca2+ signal transduction. Earlier studies have demonstrated abnormal activation of KCa3.1 in the peripheral blood and articular synovium of RA patients. Moreover, knockout of KCa3.1 reduced the severity of synovial inflammation and cartilage damage to a significant extent in a mouse collagen antibody-induced arthritis (CAIA) model. Accumulating evidence implicates KCa3.1 as a potential therapeutic target for RA. Here, we provide an overview of the KCa3.1 channel and its pharmacological properties, discuss the significance of KCa3.1 in immune cells and feasibility as a drug target for modulating the immune balance, and highlight its emerging role in pathological progression of RA.
Collapse
Affiliation(s)
- Yi Lin
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
| | - Hai-Lin Zhang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wen-Juan Hao
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Ren-Di Zhu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Yan Wang
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- *Correspondence: Wei Hu, ; Ren-Peng Zhou,
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
- *Correspondence: Wei Hu, ; Ren-Peng Zhou,
| |
Collapse
|
17
|
Altowyan MS, Khalil SMSM, Al-Wahaib D, Barakat A, Soliman SM, Ali AE, Elbadawy HA. Synthesis of a Novel Unexpected Cu(II)–Thiazolidine Complex—X-ray Structure, Hirshfeld Surface Analysis, and Biological Studies. Molecules 2022; 27:molecules27144583. [PMID: 35889457 PMCID: PMC9319961 DOI: 10.3390/molecules27144583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/12/2022] [Accepted: 07/13/2022] [Indexed: 11/26/2022] Open
Abstract
An unexpected trinuclear Cu(II)–thiazolidine complex has been synthesized by mixing CuCl2·2H2O with the Schiff base ligand, 1-(((4,5-dihydrothiazol-2-yl)ethylidene)hydrazono)methyl)phenol L, in ethanol. Unexpectedly, the reaction proceeded via the hydrolysis of the Schiff base L, followed by cyclization to afford 3-methyl-5,6-dihydrothiazolo[3,2-c][1,2,3]triazole (La), then complexation with the Cu(II) salt, forming the trinuclear [Cu3(La)4(Cl)6] complex. The complex was characterized by means of FTIR spectra, elemental analysis, and X-ray crystallography. In the trinuclear [Cu3(La)4(Cl)6] complex, there are two crystallographically independent hexa- and penta-coordinated Cu(II) sites, where the thiazolidine ligand La units act as a monodentate ligand and a linker between the Cu(II) centers. The crystal packing of the [Cu3(La)4(Cl)6] complex is primarily affected by the weak non-covalent C-H∙∙∙Cl interactions. In accordance with Hirshfeld surface analysis, the Cl∙∙∙H, H∙∙∙H, S∙∙∙H, and N∙∙∙H percentages are 31.9%, 27.2%, 13.5%, and 9.9%, respectively. X-ray photoelectron spectroscopy confirmed the oxidation state of copper as Cu(II), as well as the presence of two different coordination environments around copper centers. The complex showed interesting antibacterial activity against the Gram-positive bacteria S. subtilis, with MIC = 9.7 µg/mL compared to MIC = 4.8 µg/mL for the control, gentamycin. Moreover, the Cu(II) complex showed an equal MIC (312.5 µg/mL) against C. albicans compared to ketoconazole. It also exhibits a very promising inhibitory activity against colon carcinoma (IC50 = 3.75 ± 0.43 µg/mL).
Collapse
Affiliation(s)
- Mezna Saleh Altowyan
- Department of Chemistry, College of Science, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia;
| | - Samar M. S. M. Khalil
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt; (S.M.S.M.K.); (S.M.S.)
| | - Dhuha Al-Wahaib
- Chemistry Department, Faculty of Science, Kuwait University, Kuwait City 13060, Kuwait;
| | - Assem Barakat
- Department of Chemistry, College of Science, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Saied M. Soliman
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt; (S.M.S.M.K.); (S.M.S.)
| | - Ali Eldissouky Ali
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt; (S.M.S.M.K.); (S.M.S.)
- Correspondence: (A.E.A.); (H.A.E.)
| | - Hemmat A. Elbadawy
- Chemistry Department, Faculty of Science, Alexandria University, P.O. Box 426, Alexandria 21321, Egypt; (S.M.S.M.K.); (S.M.S.)
- Correspondence: (A.E.A.); (H.A.E.)
| |
Collapse
|
18
|
Xiao S, Baik LS, Shang X, Carlson JR. Meeting a threat of the Anthropocene: Taste avoidance of metal ions by Drosophila. Proc Natl Acad Sci U S A 2022; 119:e2204238119. [PMID: 35700364 PMCID: PMC9231609 DOI: 10.1073/pnas.2204238119] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/06/2022] [Indexed: 11/18/2022] Open
Abstract
The Anthropocene Epoch poses a critical challenge for organisms: they must cope with new threats at a rapid rate. These threats include toxic chemical compounds released into the environment by human activities. Here, we examine elevated concentrations of heavy metal ions as an example of anthropogenic stressors. We find that the fruit fly Drosophila avoids nine metal ions when present at elevated concentrations that the flies experienced rarely, if ever, until the Anthropocene. We characterize the avoidance of feeding and egg laying on metal ions, and we identify receptors, neurons, and taste organs that contribute to this avoidance. Different subsets of taste receptors, including members of both Ir (Ionotropic receptor) and Gr (Gustatory receptor) families contribute to the avoidance of different metal ions. We find that metal ions activate certain bitter-sensing neurons and inhibit sugar-sensing neurons. Some behavioral responses are mediated largely through neurons of the pharynx. Feeding avoidance remains stable over 10 generations of exposure to copper and zinc ions. Some responses to metal ions are conserved across diverse dipteran species, including the mosquito Aedes albopictus. Our results suggest mechanisms that may be essential to insects as they face challenges from environmental changes in the Anthropocene.
Collapse
Affiliation(s)
- Shuke Xiao
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Lisa S. Baik
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Xueying Shang
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - John R. Carlson
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| |
Collapse
|
19
|
Hunter T. A journey from phosphotyrosine to phosphohistidine and beyond. Mol Cell 2022; 82:2190-2200. [PMID: 35654043 DOI: 10.1016/j.molcel.2022.05.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 10/18/2022]
Abstract
Protein phosphorylation is a reversible post-translational modification. Nine of the 20 natural amino acids in proteins can be phosphorylated, but most of what we know about the roles of protein phosphorylation has come from studies of serine, threonine, and tyrosine phosphorylation. Much less is understood about the phosphorylation of histidine, lysine, arginine, cysteine, aspartate, and glutamate, so-called non-canonical phosphorylations. Phosphohistidine (pHis) was discovered 60 years ago as a mitochondrial enzyme intermediate; since then, evidence for the existence of histidine kinases and phosphohistidine phosphatases has emerged, together with examples where protein function is regulated by reversible histidine phosphorylation. pHis is chemically unstable and has thus been challenging to study. However, the recent development of tools for studying pHis has accelerated our understanding of the multifaceted functions of histidine phosphorylation, revealing a large number of proteins that are phosphorylated on histidine and implicating pHis in a wide range of cellular processes.
Collapse
Affiliation(s)
- Tony Hunter
- Molecular Cell Biology, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
20
|
Channelopathy-causing mutations in the S 45A/S 45B and HA/HB helices of K Ca2.3 and K Ca3.1 channels alter their apparent Ca 2+ sensitivity. Cell Calcium 2022; 102:102538. [PMID: 35030515 PMCID: PMC8844225 DOI: 10.1016/j.ceca.2022.102538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/11/2022]
Abstract
Small- and intermediate-conductance Ca2+-activated potassium (KCa2.x and KCa3.1, also called SK and IK) channels are activated exclusively by a Ca2+-calmodulin gating mechanism. Wild-type KCa2.3 channels have a Ca2+ EC50 value of ∼0.3 μM, while the apparent Ca2+ sensitivity of wild-type KCa3.1 channels is ∼0.27 μM. Heterozygous genetic mutations of KCa2.3 channels have been associated with Zimmermann-Laband syndrome and idiopathic noncirrhotic portal hypertension, while KCa3.1 channel mutations were reported in hereditary xerocytosis patients. KCa2.3_S436C and KCa2.3_V450L channels with mutations in the S45A/S45B helices exhibited hypersensitivity to Ca2+. The corresponding mutations in KCa3.1 channels also elevated the apparent Ca2+ sensitivity. KCa3.1_S314P, KCa3.1_A322V and KCa3.1_R352H channels with mutations in the HA/HB helices are hypersensitive to Ca2+, whereas KCa2.3 channels with the equivalent mutations are not. The different effects of the equivalent mutations in the HA/HB helices on the apparent Ca2+ sensitivity of KCa2.3 and KCa3.1 channels may imply distinct modulation of the two channel subtypes by the HA/HB helices. AP14145 reduced the apparent Ca2+ sensitivity of the hypersensitive mutant KCa2.3 channels, suggesting the potential therapeutic usefulness of negative gating modulators.
Collapse
|
21
|
Zn(II)-DPA functionalized graphene oxide two-dimensional nanocomposites for N-phosphoproteins enrichment. Talanta 2022; 243:123384. [DOI: 10.1016/j.talanta.2022.123384] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/07/2022] [Accepted: 03/11/2022] [Indexed: 11/15/2022]
|
22
|
Caglioti C, Palazzetti F, Monarca L, Lobello R, Ceccarini MR, Iannitti RG, Russo R, Ragonese F, Pennetta C, De Luca A, Codini M, Fioretti B. LY294002 Inhibits Intermediate Conductance Calcium-Activated Potassium (KCa3.1) Current in Human Glioblastoma Cells. Front Physiol 2022; 12:790922. [PMID: 35069252 PMCID: PMC8782274 DOI: 10.3389/fphys.2021.790922] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Glioblastomas (GBs) are among the most common tumors with high malignancy and invasiveness of the central nervous system. Several alterations in protein kinase and ion channel activity are involved to maintain the malignancy. Among them, phosphatidylinositol 3-kinase (PI3K) activity and intermediate conductance calcium-activated potassium (KCa3.1) current are involved in several aspects of GB biology. By using the electrophysiological approach and noise analysis, we observed that KCa3.1 channel activity is LY294002-sensitive and Wortmannin-resistant in accordance with the involvement of PI3K class IIβ (PI3KC2β). This modulation was observed also during the endogenous activation of KCa3.1 current with histamine. The principal action of PI3KC2β regulation was the reduction of open probability in intracellular free calcium saturating concentration. An explanation based on the “three-gate” model of the KCa3.1 channel by PI3KC2β was proposed. Based on the roles of KCa3.1 and PI3KC2β in GB biology, a therapeutic implication was suggested to prevent chemo- and radioresistance mechanisms.
Collapse
Affiliation(s)
- Concetta Caglioti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | - Federico Palazzetti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy.,Department of Medicine, Perugia Medical School, University of Perugia, Perugia, Italy
| | | | | | | | - Roberta Russo
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Chiara Pennetta
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Antonella De Luca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
The many ways that nature has exploited the unusual structural and chemical properties of phosphohistidine for use in proteins. Biochem J 2021; 478:3575-3596. [PMID: 34624072 DOI: 10.1042/bcj20210533] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 01/12/2023]
Abstract
Histidine phosphorylation is an important and ubiquitous post-translational modification. Histidine undergoes phosphorylation on either of the nitrogens in its imidazole side chain, giving rise to 1- and 3- phosphohistidine (pHis) isomers, each having a phosphoramidate linkage that is labile at high temperatures and low pH, in contrast with stable phosphomonoester protein modifications. While all organisms routinely use pHis as an enzyme intermediate, prokaryotes, lower eukaryotes and plants also use it for signal transduction. However, research to uncover additional roles for pHis in higher eukaryotes is still at a nascent stage. Since the discovery of pHis in 1962, progress in this field has been relatively slow, in part due to a lack of the tools and techniques necessary to study this labile modification. However, in the past ten years the development of phosphoproteomic techniques to detect phosphohistidine (pHis), and methods to synthesize stable pHis analogues, which enabled the development of anti-phosphohistidine (pHis) antibodies, have accelerated our understanding. Recent studies that employed anti-pHis antibodies and other advanced techniques have contributed to a rapid expansion in our knowledge of histidine phosphorylation. In this review, we examine the varied roles of pHis-containing proteins from a chemical and structural perspective, and present an overview of recent developments in pHis proteomics and antibody development.
Collapse
|
24
|
Mathis CL, Barrios AM. Histidine phosphorylation in metalloprotein binding sites. J Inorg Biochem 2021; 225:111606. [PMID: 34555600 DOI: 10.1016/j.jinorgbio.2021.111606] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/30/2021] [Accepted: 09/09/2021] [Indexed: 11/26/2022]
Abstract
Post-translational modifications (PTMs) are invaluable regulatory tools for the control of catalytic functionality, protein-protein interactions, and signaling pathways. Historically, the study of phosphorylation as a PTM has been focused on serine, threonine, and tyrosine residues. In contrast, the significance of mammalian histidine phosphorylation remains largely unexplored. This gap in knowledge regarding the molecular basis for histidine phosphorylation as a regulatory agent exists in part because of the relative instability of phosphorylated histidine as compared with phosphorylated serine, threonine and tyrosine. However, the unique metal binding abilities of histidine make it one of the most common metal coordinating ligands in nature, and it is interesting to consider how phosphorylation would change the metal coordinating ability of histidine, and consequently, the properties of the phosphorylated metalloprotein. In this review, we examine eleven metalloproteins that have been shown to undergo reversible histidine phosphorylation at or near their metal binding sites. These proteins are described with respect to their biological activity and structure, with a particular emphasis on how phosphohistidine may tune the primary coordination sphere and protein conformation. Furthermore, several common methods, challenges, and limitations of studying sensitive, high affinity metalloproteins are discussed.
Collapse
Affiliation(s)
- Cheryl L Mathis
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, United States
| | - Amy M Barrios
- Department of Medicinal Chemistry, College of Pharmacy, University of Utah, Salt Lake City, UT 84112, United States.
| |
Collapse
|
25
|
Gawali VS, Chimote AA, Newton HS, Feria-Garzón MG, Chirra M, Janssen EM, Wise-Draper TM, Conforti L. Immune Checkpoint Inhibitors Regulate K + Channel Activity in Cytotoxic T Lymphocytes of Head and Neck Cancer Patients. Front Pharmacol 2021; 12:742862. [PMID: 34512366 PMCID: PMC8429813 DOI: 10.3389/fphar.2021.742862] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/16/2021] [Indexed: 12/27/2022] Open
Abstract
Programmed death receptor-1 (PD-1) and its ligand (PD-L1) interaction negatively regulates T cell function in head and neck squamous cell carcinoma (HNSCC). Overexpression of PD-1 reduces intracellular Ca2+ fluxes, and thereby T cell effector functions. In HNSCC patients, PD-1 blockade increases KCa3.1 and Kv1.3 activity along with Ca2+ signaling and mobility in CD8+ peripheral blood T cells (PBTs). The mechanism by which PD-L1/PD-1 interaction regulates ion channel function is not known. We investigated the effects of blocking PD-1 and PD-L1 on ion channel functions and intracellular Ca2+ signaling in CD8+ PBTs of HNSCC patients and healthy donors (HDs) using single-cell electrophysiology and live microscopy. Anti-PD-1 and anti-PD-L1 antibodies increase KCa3.1 and Kv1.3 function in CD8+ PBTs of HNSCC patients. Anti-PD-1 treatment increases Ca2+ fluxes in a subset of HSNCC patients. In CD8+ PBTs of HDs, exposure to PD-L1 reduces KCa3.1 activity and Ca2+ signaling, which were restored by anti-PD-1 treatment. The PD-L1-induced inhibition of KCa3.1 channels was rescued by the intracellular application of the PI3 kinase modulator phosphatidylinositol 3-phosphate (PI3P) in patch-clamp experiments. In HNSCC CD8+ PBTs, anti-PD-1 treatment did not affect the expression of KCa3.1, Kv1.3, Ca2+ release activated Ca2+ (CRAC) channels, and markers of cell activation (CD69) and exhaustion (LAG-3 and TIM-3). Our data show that immune checkpoint blockade improves T cell function by increasing KCa3.1 and Kv1.3 channel activity in HNSCC patients.
Collapse
Affiliation(s)
- Vaibhavkumar S Gawali
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Ameet A Chimote
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Hannah S Newton
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Manuel G Feria-Garzón
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Martina Chirra
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| | - Edith M Janssen
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Immunology, Janssen Research and Development, Spring House, PA, United States
| | - Trisha M Wise-Draper
- Department of Internal Medicine, Division of Hematology Oncology, University of Cincinnati, Cincinnati, OH, United States
| | - Laura Conforti
- Department of Internal Medicine, Division of Nephrology, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
26
|
Abstract
Metals are vital for life as they are necessary for essential biological processes. Traditionally, metals are categorized as either dynamic signals or static cofactors. Redox-inactive metals such as calcium (Ca), potassium (K), sodium (Na), and zinc (Zn) signal through large fluctuations in their metal-ion pools. In contrast, redox-active transition metals such as copper (Cu) and iron (Fe) drive catalysis and are largely characterized as static cofactors that must be buried and protected within the active sites of proteins, due to their ability to generate damaging reactive-oxygen species through Fenton chemistry. Cu has largely been studied as a static cofactor in fundamental processes from cellular respiration to pigmentation, working through cytochrome c oxidase and tyrosinase, respectively. However, within the last decade, a new paradigm in nutrient sensing and protein regulation - termed 'metalloallostery' - has emerged, expanding the repertoire of Cu beyond the catalytic proteins to dynamic signaling molecules essential for cellular processes that impact normal physiology and disease states. In this Primer we introduce both the 'traditional' and emerging roles for Cu in biology and the many ways in which Cu intersects with human health.
Collapse
Affiliation(s)
- Tiffany Tsang
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Caroline I Davis
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Donita C Brady
- Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
27
|
Structural basis for differential recognition of phosphohistidine-containing peptides by 1-pHis and 3-pHis monoclonal antibodies. Proc Natl Acad Sci U S A 2021; 118:2010644118. [PMID: 33547238 PMCID: PMC8017925 DOI: 10.1073/pnas.2010644118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In 2015, monoclonal antibodies (mAbs) that selectively recognize the 1-pHis or 3-pHis isoforms of phosphohistidine were developed by immunizing rabbits with degenerate Ala/Gly peptides containing the nonhydrolyzable phosphohistidine (pHis) analog- phosphotriazolylalanine (pTza). Here, we report structures of five rabbit mAbs bound to cognate pTza peptides: SC1-1 and SC50-3 that recognize 1-pHis, and their 3-pHis-specific counterparts, SC39-4, SC44-8, and SC56-2. These cocrystal structures provide insights into the binding modes of the pTza phosphate group that are distinct for the 1- and 3-pHis mAbs with the selectivity arising from specific contacts with the phosphate group and triazolyl ring. The mode of phosphate recognition in the 3-pHis mAbs recapitulates the Walker A motif, as present in kinases. The complementarity-determining regions (CDRs) of four of the Fabs interact with the peptide backbone rather than peptide side chains, thus conferring sequence independence, whereas SC44-8 shows a proclivity for binding a GpHAGA motif mediated by a sterically complementary CDRL3 loop. Specific hydrogen bonding with the triazolyl ring precludes recognition of pTyr and other phosphoamino acids by these mAbs. Kinetic binding experiments reveal that the affinity of pHis mAbs for pHis and pTza peptides is submicromolar. Bound pHis mAbs also shield the pHis peptides from rapid dephosphorylation. The epitope-paratope interactions illustrate how these anti-pHis antibodies are useful for a wide range of research techniques and this structural information can be utilized to improve the specificity and affinity of these antibodies toward a variety of pHis substrates to understand the role of histidine phosphorylation in healthy and diseased states.
Collapse
|
28
|
Schulte JE, Roggiani M, Shi H, Zhu J, Goulian M. The phosphohistidine phosphatase SixA dephosphorylates the phosphocarrier NPr. J Biol Chem 2020; 296:100090. [PMID: 33199374 PMCID: PMC7948535 DOI: 10.1074/jbc.ra120.015121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 10/28/2020] [Accepted: 11/16/2020] [Indexed: 01/17/2023] Open
Abstract
Histidine phosphorylation is a posttranslational modification that alters protein function and also serves as an intermediate of phosphoryl transfer. Although phosphohistidine is relatively unstable, enzymatic dephosphorylation of this residue is apparently needed in some contexts, since both prokaryotic and eukaryotic phosphohistidine phosphatases have been reported. Here we identify the mechanism by which a bacterial phosphohistidine phosphatase dephosphorylates the nitrogen-related phosphotransferase system, a broadly conserved bacterial pathway that controls diverse metabolic processes. We show that the phosphatase SixA dephosphorylates the phosphocarrier protein NPr and that the reaction proceeds through phosphoryl transfer from a histidine on NPr to a histidine on SixA. In addition, we show that Escherichia coli lacking SixA are outcompeted by wild-type E. coli in the context of commensal colonization of the mouse intestine. Notably, this colonization defect requires NPr and is distinct from a previously identified in vitro growth defect associated with dysregulation of the nitrogen-related phosphotransferase system. The widespread conservation of SixA, and its coincidence with the phosphotransferase system studied here, suggests that this dephosphorylation mechanism may be conserved in other bacteria.
Collapse
Affiliation(s)
- Jane E Schulte
- Graduate Group in Biochemistry and Molecular Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Manuela Roggiani
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Hui Shi
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; College of Food Science, Southwest University, Beibei, Chongqing, China
| | - Jun Zhu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mark Goulian
- Department of Biology, University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Physics & Astronomy, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
29
|
Adam K, Ning J, Reina J, Hunter T. NME/NM23/NDPK and Histidine Phosphorylation. Int J Mol Sci 2020; 21:E5848. [PMID: 32823988 PMCID: PMC7461546 DOI: 10.3390/ijms21165848] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
The NME (Non-metastatic) family members, also known as NDPKs (nucleoside diphosphate kinases), were originally identified and studied for their nucleoside diphosphate kinase activities. This family of kinases is extremely well conserved through evolution, being found in prokaryotes and eukaryotes, but also diverges enough to create a range of complexity, with homologous members having distinct functions in cells. In addition to nucleoside diphosphate kinase activity, some family members are reported to possess protein-histidine kinase activity, which, because of the lability of phosphohistidine, has been difficult to study due to the experimental challenges and lack of molecular tools. However, over the past few years, new methods to investigate this unstable modification and histidine kinase activity have been reported and scientific interest in this area is growing rapidly. This review presents a global overview of our current knowledge of the NME family and histidine phosphorylation, highlighting the underappreciated protein-histidine kinase activity of NME family members, specifically in human cells. In parallel, information about the structural and functional aspects of the NME family, and the knowns and unknowns of histidine kinase involvement in cell signaling are summarized.
Collapse
Affiliation(s)
| | | | | | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA; (K.A.); (J.N.); (J.R.)
| |
Collapse
|
30
|
Manfroni G, Ragonese F, Monarca L, Astolfi A, Mancinelli L, Iannitti RG, Bastioli F, Barreca ML, Cecchetti V, Fioretti B. New Insights on KCa3.1 Channel Modulation. Curr Pharm Des 2020; 26:2096-2101. [PMID: 32175839 DOI: 10.2174/1381612826666200316152645] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/11/2020] [Indexed: 11/22/2022]
Abstract
The human intermediate conductance calcium-activated potassium channel, KCa3.1, is involved in several pathophysiological conditions playing a critical role in cell secretory machinery and calcium signalling. The recent cryo-EM analysis provides new insights for understanding the modulation by both endogenous and pharmacological agents. A typical feature of this channel is the low open probability in saturating calcium concentrations and its modulation by potassium channel openers (KCOs), such as benzo imidazolone 1-EBIO, without changing calcium-dependent activation. In this paper, we proposed a model of KCOs action in the modulation of channel activity. The KCa3.1 channel has a very rich pharmacological profile with several classes of molecules that selectively interact with different binding sites of the channel. Among them, benzo imidazolones can be openers (positive modulators such as 1-EBIO, DC-EBIO) or blockers (negative modulators such as NS1619). Through computation modelling techniques, we identified the 1,4-benzothiazin-3-one as a promising scaffold to develop new KCa3.1 channel modulators. Further studies are needed to explore the potential use of 1-4 benzothiazine- 3-one in KCa3.1 modulation and its pharmacological application.
Collapse
Affiliation(s)
- Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Francesco Ragonese
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy.,Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Lorenzo Monarca
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy.,Department of Experimental Medicine, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy
| | - Andrea Astolfi
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Loretta Mancinelli
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| | | | | | - Maria L Barreca
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, Via del Liceo, 1-06123-Perugia (PG), Italy
| | - Bernard Fioretti
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via Elce di Sotto 8, 06123 Perugia, Italy
| |
Collapse
|
31
|
Nucleoside Diphosphate Kinase B Contributes to Arrhythmogenesis in Human-Induced Pluripotent Stem Cell-Derived Cardiomyocytes from a Patient with Arrhythmogenic Right Ventricular Cardiomyopathy. J Clin Med 2020; 9:jcm9020486. [PMID: 32050722 PMCID: PMC7073527 DOI: 10.3390/jcm9020486] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 01/30/2020] [Accepted: 02/05/2020] [Indexed: 12/29/2022] Open
Abstract
Background: Arrhythmogenic right ventricular cardiomyopathy (ARVC) is a rare, inheritable cardiac disorder characterized by ventricular tachyarrhythmias, progressive loss of cardiomyocytes with fibrofatty replacement and sudden cardiac death. The exact underlying mechanisms are unclear. Methods: This study investigated the possible roles of nucleoside diphosphate kinase B (NDPK-B) and SK4 channels in the arrhythmogenesis of ARVC by using human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Results: In hiPSC-CMs from a patient with ARVC, the expression levels of NDPK-B and SK4 channels were upregulated, the cell automaticity was increased and the occurrence rate of arrhythmic events was enhanced. Recombinant NDPK-B applied into hiPSC-CMs from either healthy donors or the patient enhanced SK4 channel current (ISK4), cell automaticity and the occurrence of arrhythmic events, whereas protein histidine phosphatase 1 (PHP-1), a counter actor of NDPK-B, prevented the NDPK-B effect. Application of PHP-1 alone or a SK4 channel blocker also reduced cell automaticity and arrhythmic events. Conclusion: This study demonstrated that the elevated NDPK-B expression, via activating SK4 channels, contributes to arrhythmogenesis in ARVC, and hence, NDPK-B may be a potential therapeutic target for treating arrhythmias in patients with ARVC.
Collapse
|
32
|
Roach KM, Bradding P. Ca 2+ signalling in fibroblasts and the therapeutic potential of K Ca3.1 channel blockers in fibrotic diseases. Br J Pharmacol 2020; 177:1003-1024. [PMID: 31758702 DOI: 10.1111/bph.14939] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 10/23/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
The role of Ca2+ signalling in fibroblasts is of great interest in fibrosis-related diseases. Intracellular free Ca2+ ([Ca2+ ]i ) is a ubiquitous secondary messenger, regulating a number of cellular functions such as secretion, metabolism, differentiation, proliferation and contraction. The intermediate conductance Ca2+ -activated K+ channel KCa 3.1 is pivotal in Ca2+ signalling and plays a central role in fibroblast processes including cell activation, migration and proliferation through the regulation of cell membrane potential. Evidence from a number of approaches demonstrates that KCa 3.1 plays an important role in the development of many fibrotic diseases, including idiopathic pulmonary, renal tubulointerstitial fibrosis and cardiovascular disease. The KCa 3.1 selective blocker senicapoc was well tolerated in clinical trials for sickle cell disease, raising the possibility of rapid translation to the clinic for people suffering from pathological fibrosis. This review after analysing all the data, concludes that targeting KCa 3.1 should be a high priority for human fibrotic disease.
Collapse
Affiliation(s)
- Katy M Roach
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| | - Peter Bradding
- Institute for Lung Health, Department of Respiratory Sciences, University of Leicester, Leicester, UK
| |
Collapse
|
33
|
She G, Ren YJ, Wang Y, Hou MC, Wang HF, Gou W, Lai BC, Lei T, Du XJ, Deng XL. K Ca3.1 Channels Promote Cardiac Fibrosis Through Mediating Inflammation and Differentiation of Monocytes Into Myofibroblasts in Angiotensin II -Treated Rats. J Am Heart Assoc 2020; 8:e010418. [PMID: 30563389 PMCID: PMC6405723 DOI: 10.1161/jaha.118.010418] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Cardiac fibrosis is a core pathological process associated with heart failure. The recruitment and differentiation of primitive fibroblast precursor cells of bone marrow origin play a critical role in pathological interstitial cardiac fibrosis. The KC a3.1 channels are expressed in both ventricular fibroblasts and circulating mononuclear cells in rats and are upregulated by angiotensin II . We hypothesized that KC a3.1 channels mediate the inflammatory microenvironment in the heart, promoting the infiltrated bone marrow-derived circulating mononuclear cells to differentiate into myofibroblasts, leading to myocardial fibrosis. Methods and Results We established a cardiac fibrosis model in rats by infusing angiotensin II to evaluate the impact of the specific KC a3.1 channel blocker TRAM -34 on cardiac fibrosis. At the same time, mouse CD 4+ T cells and rat circulating mononuclear cells were separated to investigate the underlying mechanism of the TRAM -34 anti-cardiac fibrosis effect. TRAM -34 significantly attenuated cardiac fibrosis and the inflammatory reaction and reduced the number of fibroblast precursor cells and myofibroblasts. Inhibition of KC a3.1 channels suppressed angiotensin II -stimulated expression and secretion of interleukin-4 and interleukin-13 in CD 4+ T cells and interleukin-4- or interleukin-13-induced differentiation of monocytes into fibrocytes. Conclusions KC a3.1 channels facilitate myocardial inflammation and the differentiation of bone marrow-derived monocytes into myofibroblasts in cardiac fibrosis caused by angiotensin II infusion.
Collapse
Affiliation(s)
- Gang She
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Yu-Jie Ren
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,5 Department of Pathology Xi'an Guangren Hospital Affiliated to Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Yan Wang
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Meng-Chen Hou
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Hui-Fang Wang
- 5 Department of Pathology Xi'an Guangren Hospital Affiliated to Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Wei Gou
- 3 Basic Experiment Teaching Center School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Bao-Chang Lai
- 4 Cardiovascular Research Centre School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Ting Lei
- 2 Department of Pathology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| | - Xiao-Jun Du
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,6 Baker Heart and Diabetes Institute Melbourne Victoria Australia
| | - Xiu-Ling Deng
- 1 Department of Physiology and Pathophysiology School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China.,4 Cardiovascular Research Centre School of Basic Medical Sciences Xi'an Jiaotong University Health Science Center Xi'an Shaanxi China
| |
Collapse
|
34
|
Tomlinson LJ, Clubbs Coldron AKM, Eyers PA, Eyers CE. Determination of Phosphohistidine Stoichiometry in Histidine Kinases by Intact Mass Spectrometry. Methods Mol Biol 2020; 2077:83-91. [PMID: 31707653 DOI: 10.1007/978-1-4939-9884-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Protein histidine phosphorylation has largely remained unexplored due to the challenges of analyzing relatively unstable phosphohistidine-containing proteins. We describe a procedure for determining the stoichiometry of histidine phosphorylation on the human histidine kinases NME1 and NME2 by intact mass spectrometry under conditions that retain this acid-labile protein modification. By characterizing these two model histidine protein kinases in the absence and presence of a suitable phosphate donor, the stoichiometry of histidine phosphorylation can be determined. The described method can be readily adapted for the analysis of other proteins containing phosphohistidine.
Collapse
Affiliation(s)
- Lauren J Tomlinson
- Department of Biochemistry, Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Alice K M Clubbs Coldron
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Claire E Eyers
- Department of Biochemistry, Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
35
|
Kalagiri R, Adam K, Hunter T. Empirical Evidence of Cellular Histidine Phosphorylation by Immunoblotting Using pHis mAbs. Methods Mol Biol 2020; 2077:181-191. [PMID: 31707659 PMCID: PMC9828874 DOI: 10.1007/978-1-4939-9884-5_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Immunoblotting is a ubiquitous immunological technique that aids in detecting and quantifying proteins (including those of lower abundance) and their posttranslational modifications such as phosphorylation, acetylation, ubiquitylation, and sumoylation. The technique involves electrophoretically separating proteins on an SDS-PAGE gel, transferring them onto a PVDF (or nitrocellulose) membrane and probing with specific antibodies. Here we describe an immunoblotting technique for detecting cellular phosphohistidine, a labile posttranslational modification, by optimizing experimental conditions such that the labile phosphohistidine signal is conserved throughout the experiment.
Collapse
Affiliation(s)
- Rajasree Kalagiri
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kevin Adam
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA.
| |
Collapse
|
36
|
Immunohistochemistry (IHC): Chromogenic Detection of 3-Phosphohistidine Proteins in Formaldehyde-Fixed, Frozen Mouse Liver Tissue Sections. Methods Mol Biol 2020; 2077:193-208. [PMID: 31707660 PMCID: PMC9828869 DOI: 10.1007/978-1-4939-9884-5_13] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The development of antibodies that specifically detect histidine-phosphorylated proteins is a recent achievement and allows potential roles of histidine phosphorylated proteins in pathological and physiological conditions to be characterized. Immunohistochemical analyses enable the detection of proteins in tissues and can reveal alterations to the quantity and/or localization of these proteins through comparisons of normal and diseased specimens. However, the sensitivity of phosphohistidine modifications to phosphatases, acidic pH, and elevated temperatures poses unique challenges to the detection process and requires a protocol that bypasses traditional procedures utilizing paraffin-embedding and antigen-retrieval methods. Here, we detail a method for a brief fixation by 4% (v/v) paraformaldehyde on freshly collected tissues in the presence of PhosSTOP to block phosphatase activity, followed by a float on sucrose to protect the tissue prior to freezing. Specimens are then embedded in a cryopreservation medium in molds and frozen using an isoflurane, dry ice bath to best preserve the tissue morphology and phosphohistidine signal. We validate this technique in normal mouse liver using SC44-1, a monoclonal anti-3-pHis antibody used to uncover a role for a protein histidine phosphatase as a tumor suppressor in the liver. Furthermore, we demonstrate that the antibody signal can be eliminated by preincubating SC44-1 with a peptide treated with phosphoramidate to phosphorylate histidine residues. Thus, we present an IHC protocol suitable for specific detection of 3-phosphohistidine proteins in mouse liver tissue, and suggest that this can be used as a starting point for optimization of IHC using other phosphohistidine antibodies or in other tissue types, generating information that will enhance our understanding of phosphohistidine in models of disease.
Collapse
|
37
|
Clubbs Coldron AKM, Byrne DP, Eyers PA. Analysis of 1- and 3-Phosphohistidine (pHis) Protein Modification Using Model Enzymes Expressed in Bacteria. Methods Mol Biol 2020; 2077:63-81. [PMID: 31707652 DOI: 10.1007/978-1-4939-9884-5_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Despite the discovery of protein histidine (His) phosphorylation nearly six decades ago, difficulties in measuring and quantifying this unstable post-translational modification (PTM) have limited its mechanistic analysis in prokaryotic and eukaryotic signaling. Here, we describe reliable procedures for affinity purification, cofactor-binding analysis and antibody-based detection of phosphohistidine (pHis), on the putative human His kinases NME1 (NDPK-A) and NME2 (NDPK-B) and the glycolytic phosphoglycerate mutase PGAM1. By exploiting isomer-specific monoclonal N1-pHis and N3-pHis antibodies, we describe robust protocols for immunological detection and isomer discrimination of site-specific pHis, including N3-pHis on His 11 of PGAM1.
Collapse
Affiliation(s)
- Alice K M Clubbs Coldron
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Dominic P Byrne
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK.
| |
Collapse
|
38
|
Hardman G, Perkins S, Brownridge PJ, Clarke CJ, Byrne DP, Campbell AE, Kalyuzhnyy A, Myall A, Eyers PA, Jones AR, Eyers CE. Strong anion exchange-mediated phosphoproteomics reveals extensive human non-canonical phosphorylation. EMBO J 2019; 38:e100847. [PMID: 31433507 PMCID: PMC6826212 DOI: 10.15252/embj.2018100847] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/18/2022] Open
Abstract
Phosphorylation is a key regulator of protein function under (patho)physiological conditions, and defining site-specific phosphorylation is essential to understand basic and disease biology. In vertebrates, the investigative focus has primarily been on serine, threonine and tyrosine phosphorylation, but mounting evidence suggests that phosphorylation of other "non-canonical" amino acids also regulates critical aspects of cell biology. However, standard methods of phosphoprotein characterisation are largely unsuitable for the analysis of non-canonical phosphorylation due to their relative instability under acidic conditions and/or elevated temperature. Consequently, the complete landscape of phosphorylation remains unexplored. Here, we report an unbiased phosphopeptide enrichment strategy based on strong anion exchange (SAX) chromatography (UPAX), which permits identification of histidine (His), arginine (Arg), lysine (Lys), aspartate (Asp), glutamate (Glu) and cysteine (Cys) phosphorylation sites on human proteins by mass spectrometry-based phosphoproteomics. Remarkably, under basal conditions, and having accounted for false site localisation probabilities, the number of unique non-canonical phosphosites is approximately one-third of the number of observed canonical phosphosites. Our resource reveals the previously unappreciated diversity of protein phosphorylation in human cells, and opens up avenues for high-throughput exploration of non-canonical phosphorylation in all organisms.
Collapse
Affiliation(s)
- Gemma Hardman
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Simon Perkins
- Department of Comparative and Functional Genomics, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Philip J Brownridge
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Christopher J Clarke
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Dominic P Byrne
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Amy E Campbell
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Anton Kalyuzhnyy
- Department of Comparative and Functional Genomics, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Ashleigh Myall
- Department of Comparative and Functional Genomics, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Patrick A Eyers
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Andrew R Jones
- Department of Comparative and Functional Genomics, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - Claire E Eyers
- Centre for Proteome Research, Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| |
Collapse
|
39
|
Petkowski JJ, Bains W, Seager S. Natural Products Containing 'Rare' Organophosphorus Functional Groups. Molecules 2019; 24:E866. [PMID: 30823503 PMCID: PMC6429109 DOI: 10.3390/molecules24050866] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/13/2019] [Accepted: 02/22/2019] [Indexed: 12/25/2022] Open
Abstract
Phosphorous-containing molecules are essential constituents of all living cells. While the phosphate functional group is very common in small molecule natural products, nucleic acids, and as chemical modification in protein and peptides, phosphorous can form P⁻N (phosphoramidate), P⁻S (phosphorothioate), and P⁻C (e.g., phosphonate and phosphinate) linkages. While rare, these moieties play critical roles in many processes and in all forms of life. In this review we thoroughly categorize P⁻N, P⁻S, and P⁻C natural organophosphorus compounds. Information on biological source, biological activity, and biosynthesis is included, if known. This review also summarizes the role of phosphorylation on unusual amino acids in proteins (N- and S-phosphorylation) and reviews the natural phosphorothioate (P⁻S) and phosphoramidate (P⁻N) modifications of DNA and nucleotides with an emphasis on their role in the metabolism of the cell. We challenge the commonly held notion that nonphosphate organophosphorus functional groups are an oddity of biochemistry, with no central role in the metabolism of the cell. We postulate that the extent of utilization of some phosphorus groups by life, especially those containing P⁻N bonds, is likely severely underestimated and has been largely overlooked, mainly due to the technological limitations in their detection and analysis.
Collapse
Affiliation(s)
- Janusz J Petkowski
- Department of Earth, Atmospheric, and Planetary Sciences, Massachusetts Institute of Technology, 77 Mass. Ave., Cambridge, MA 02139, USA.
| | - William Bains
- Rufus Scientific, 37 The Moor, Melbourn, Royston, Herts SG8 6ED, UK.
| | - Sara Seager
- Department of Earth, Atmospheric, and Planetary Sciences, Massachusetts Institute of Technology, 77 Mass. Ave., Cambridge, MA 02139, USA.
- Department of Physics, Massachusetts Institute of Technology, 77 Mass. Ave., Cambridge, MA 02139, USA.
- Department of Aeronautics and Astronautics, Massachusetts Institute of Technology, 77 Mass. Ave., Cambridge, MA 02139, USA.
| |
Collapse
|
40
|
Cancer-Associated Intermediate Conductance Ca 2+-Activated K⁺ Channel K Ca3.1. Cancers (Basel) 2019; 11:cancers11010109. [PMID: 30658505 PMCID: PMC6357066 DOI: 10.3390/cancers11010109] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 01/10/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Several tumor entities have been reported to overexpress KCa3.1 potassium channels due to epigenetic, transcriptional, or post-translational modifications. By modulating membrane potential, cell volume, or Ca2+ signaling, KCa3.1 has been proposed to exert pivotal oncogenic functions in tumorigenesis, malignant progression, metastasis, and therapy resistance. Moreover, KCa3.1 is expressed by tumor-promoting stroma cells such as fibroblasts and the tumor vasculature suggesting a role of KCa3.1 in the adaptation of the tumor microenvironment. Combined, this features KCa3.1 as a candidate target for innovative anti-cancer therapy. However, immune cells also express KCa3.1 thereby contributing to T cell activation. Thus, any strategy targeting KCa3.1 in anti-cancer therapy may also modulate anti-tumor immune activity and/or immunosuppression. The present review article highlights the potential of KCa3.1 as an anti-tumor target providing an overview of the current knowledge on its function in tumor pathogenesis with emphasis on vasculo- and angiogenesis as well as anti-cancer immune responses.
Collapse
|
41
|
Gohla A. Do metabolic HAD phosphatases moonlight as protein phosphatases? BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:153-166. [DOI: 10.1016/j.bbamcr.2018.07.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 07/12/2018] [Indexed: 12/14/2022]
|
42
|
Lu S, Wang Y. Nonmetabolic functions of metabolic enzymes in cancer development. Cancer Commun (Lond) 2018; 38:63. [PMID: 30367676 PMCID: PMC6235390 DOI: 10.1186/s40880-018-0336-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 10/21/2018] [Indexed: 12/28/2022] Open
Abstract
Metabolism is a fundamental biological process composed of a series of reactions catalyzed by metabolic enzymes. Emerging evidence demonstrates that the aberrant signaling in cancer cells induces nonmetabolic functions of metabolic enzymes in many instrumental cellular activities, which involve metabolic enzyme-mediated protein post-translational modifications, such as phosphorylation, acetylation, and succinylation. In the most well-researched literatures, metabolic enzymes phosphorylate proteins rather than their metabolites as substrates. Some metabolic enzymes have altered subcellular localization, which allows their metabolic products to directly participate in nonmetabolic activities. This review discusses how these findings have deepened our understanding on enzymes originally classified as metabolic enzymes, by highlighting the nonmetabolic functions of several metabolic enzymes responsible for the development of cancer, and evaluates the potential for targeting these functions in cancer treatment.
Collapse
Affiliation(s)
- Sean Lu
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yugang Wang
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
43
|
Kardos J, Héja L, Simon Á, Jablonkai I, Kovács R, Jemnitz K. Copper signalling: causes and consequences. Cell Commun Signal 2018; 16:71. [PMID: 30348177 PMCID: PMC6198518 DOI: 10.1186/s12964-018-0277-3] [Citation(s) in RCA: 127] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 09/24/2018] [Indexed: 12/18/2022] Open
Abstract
Copper-containing enzymes perform fundamental functions by activating dioxygen (O2) and therefore allowing chemical energy-transfer for aerobic metabolism. The copper-dependence of O2 transport, metabolism and production of signalling molecules are supported by molecular systems that regulate and preserve tightly-bound static and weakly-bound dynamic cellular copper pools. Disruption of the reducing intracellular environment, characterized by glutathione shortage and ambient Cu(II) abundance drives oxidative stress and interferes with the bidirectional, copper-dependent communication between neurons and astrocytes, eventually leading to various brain disease forms. A deeper understanding of of the regulatory effects of copper on neuro-glia coupling via polyamine metabolism may reveal novel copper signalling functions and new directions for therapeutic intervention in brain disorders associated with aberrant copper metabolism.
Collapse
Affiliation(s)
- Julianna Kardos
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - László Héja
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Ágnes Simon
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - István Jablonkai
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| | - Richard Kovács
- Institute of Neurophysiology, Charité-Universitätsmedizin, Berlin, Germany
| | - Katalin Jemnitz
- Functional Pharmacology Research Group, Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar Tudósok körútja 2, Budapest, 1117 Hungary
| |
Collapse
|
44
|
Ohya S, Kito H. Ca 2+-Activated K + Channel K Ca3.1 as a Therapeutic Target for Immune Disorders. Biol Pharm Bull 2018; 41:1158-1163. [PMID: 30068864 DOI: 10.1248/bpb.b18-00078] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In lymphoid and myeloid cells, membrane hyperpolarization by the opening of K+ channels increases the activity of Ca2+ release-activated Ca2+ (CRAC) channels and transient receptor potential (TRP) Ca2+ channels. The intermediate-conductance Ca2+-activated K+ channel KCa3.1 plays an important role in cell proliferation, differentiation, migration, and cytokine production in innate and adaptive immune systems. KCa3.1 is therefore an attractive therapeutic target for allergic, inflammatory, and autoimmune disorders. In the past several years, studies have provided new insights into 1) KCa3.1 pharmacology and its auxiliary regulators; 2) post-transcriptional and proteasomal regulation of KCa3.1; 3) KCa3.1 as a regulator of immune cell migration, cytokine production, and phenotypic polarization; 4) the role of KCa3.1 in the phosphorylation and nuclear translocation of Smad2/3; and 5) KCa3.1 as a therapeutic target for cancer immunotherapy. In this review, we have assembled a comprehensive overview of current research on the physiological and pathophysiological significance of KCa3.1 in the immune system.
Collapse
Affiliation(s)
- Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University
| |
Collapse
|
45
|
Ji T, Corbalán-García S, Hubbard SR. Crystal structure of the C-terminal four-helix bundle of the potassium channel KCa3.1. PLoS One 2018; 13:e0199942. [PMID: 29953543 PMCID: PMC6023178 DOI: 10.1371/journal.pone.0199942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/15/2018] [Indexed: 11/18/2022] Open
Abstract
KCa3.1 (also known as SK4 or IK1) is a mammalian intermediate-conductance potassium channel that plays a critical role in the activation of T cells, B cells, and mast cells, effluxing potassium ions to maintain a negative membrane potential for influxing calcium ions. KCa3.1 shares primary sequence similarity with three other (low-conductance) potassium channels: KCa2.1, KCa2.2, and KCa2.3 (also known as SK1–3). These four homotetrameric channels bind calmodulin (CaM) in the cytoplasmic region, and calcium binding to CaM triggers channel activation. Unique to KCa3.1, activation also requires phosphorylation of a single histidine residue, His358, in the cytoplasmic region, which relieves copper-mediated inhibition of the channel. Near the cytoplasmic C-terminus of KCa3.1 (and KCa2.1–2.3), secondary-structure analysis predicts the presence of a coiled-coil/heptad repeat. Here, we report the crystal structure of the C-terminal coiled-coil region of KCa3.1, which forms a parallel four-helix bundle, consistent with the tetrameric nature of the channel. Interestingly, the four copies of a histidine residue, His389, in an ‘a’ position within the heptad repeat, are observed to bind a copper ion along the four-fold axis of the bundle. These results suggest that His358, the inhibitory histidine in KCa3.1, might coordinate a copper ion through a similar binding mode.
Collapse
Affiliation(s)
- Tianyang Ji
- Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States of America
| | - Senena Corbalán-García
- Department of Biochemistry and Molecular Biology-A, School of Veterinary, Regional Campus of International Excellence "Campus Mare Nostrum", Biomedical Research Institute of Murcia (IMIB-Arrixaca), University of Murcia, Murcia, Spain
| | - Stevan R. Hubbard
- Kimmel Center for Biology and Medicine of the Skirball Institute, Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, United States of America
- * E-mail:
| |
Collapse
|
46
|
McCullough BS, Barrios AM. Facile, Fluorogenic Assay for Protein Histidine Phosphatase Activity. Biochemistry 2018; 57:2584-2589. [DOI: 10.1021/acs.biochem.8b00278] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Affiliation(s)
- Brandon S. McCullough
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| | - Amy M. Barrios
- Department of Medicinal Chemistry, University of Utah, Salt Lake City, Utah 84112, United States
| |
Collapse
|
47
|
Lu Z, Hunter T. Metabolic Kinases Moonlighting as Protein Kinases. Trends Biochem Sci 2018; 43:301-310. [PMID: 29463470 PMCID: PMC5879014 DOI: 10.1016/j.tibs.2018.01.006] [Citation(s) in RCA: 176] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 01/15/2018] [Accepted: 01/25/2018] [Indexed: 12/17/2022]
Abstract
Protein kinases regulate every aspect of cellular activity, whereas metabolic enzymes are responsible for energy production and catabolic and anabolic processes. Emerging evidence demonstrates that some metabolic enzymes, such as pyruvate kinase M2 (PKM2), phosphoglycerate kinase 1 (PGK1), ketohexokinase (KHK) isoform A (KHK-A), hexokinase (HK), and nucleoside diphosphate kinase 1 and 2 (NME1/2), that phosphorylate soluble metabolites can also function as protein kinases and phosphorylate a variety of protein substrates to regulate the Warburg effect, gene expression, cell cycle progression and proliferation, apoptosis, autophagy, exosome secretion, T cell activation, iron transport, ion channel opening, and many other fundamental cellular functions. The elevated protein kinase functions of these moonlighting metabolic enzymes in tumor development make them promising therapeutic targets for cancer.
Collapse
Affiliation(s)
- Zhimin Lu
- Brain Tumor Center and Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; Cancer Biology Program, MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, The University of Texas, Houston, TX 77030, USA.
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA 92037, USA.
| |
Collapse
|
48
|
Makwana MV, Muimo R, Jackson RF. Advances in development of new tools for the study of phosphohistidine. J Transl Med 2018; 98:291-303. [PMID: 29200202 DOI: 10.1038/labinvest.2017.126] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 08/27/2017] [Accepted: 09/03/2017] [Indexed: 01/04/2023] Open
Abstract
Protein phosphorylation is an important post-translational modification that is an integral part of cellular function. The O-phosphorylated amino-acid residues, such as phosphoserine (pSer), phosphothreonine (pThr) and phosphotyrosine (pTyr), have dominated the literature while the acid labile N-linked phosphorylated amino acids, such as phosphohistidine (pHis), have largely been historically overlooked because of the acidic conditions routinely used in amino-acid detection and analysis. This review highlights some misinterpretations that have arisen in the existing literature, pinpoints outstanding questions and potential future directions to clarify the role of pHis in mammalian signalling systems. Particular emphasis is placed on pHis isomerization and the hybrid functionality for both pHis and pTyr of the proposed τ-pHis analogue bearing the triazole residue.
Collapse
Affiliation(s)
- Mehul V Makwana
- Department of Chemistry, University of Sheffield, Sheffield S3 7HF, UK.,Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK
| | - Richmond Muimo
- Department of Infection, Immunity and Cardiovascular Disease, Medical School, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
49
|
Adam K, Hunter T. Histidine kinases and the missing phosphoproteome from prokaryotes to eukaryotes. J Transl Med 2018; 98:233-247. [PMID: 29058706 PMCID: PMC5815933 DOI: 10.1038/labinvest.2017.118] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 08/16/2017] [Accepted: 08/31/2017] [Indexed: 12/20/2022] Open
Abstract
Protein phosphorylation is the most common type of post-translational modification in eukaryotes. The phosphoproteome is defined as the complete set of experimentally detectable phosphorylation sites present in a cell's proteome under various conditions. However, we are still far from identifying all the phosphorylation sites in a cell mainly due to the lack of information about phosphorylation events involving residues other than Ser, Thr and Tyr. Four types of phosphate-protein linkage exist and these generate nine different phosphoresidues-pSer, pThr, pTyr, pHis, pLys, pArg, pAsp, pGlu and pCys. Most of the effort in studying protein phosphorylation has been focused on Ser, Thr and Tyr phosphorylation. The recent development of 1- and 3-pHis monoclonal antibodies promises to increase our understanding of His phosphorylation and the kinases and phosphatases involved. Several His kinases are well defined in prokaryotes, especially those involved in two-component system (TCS) signaling. However, in higher eukaryotes, NM23, a protein originally characterized as a nucleoside diphosphate kinase, is the only characterized protein-histidine kinase. This ubiquitous and conserved His kinase autophosphorylates its active site His, and transfers this phosphate either onto a nucleoside diphosphate or onto a protein His residue. Studies of NM23 protein targets using newly developed anti-pHis antibodies will surely help illuminate the elusive His phosphorylation-based signaling pathways. This review discusses the role that the NM23/NME/NDPK phosphotransferase has, how the addition of the pHis phosphoproteome will expand the phosphoproteome and make His phosphorylation part of the global phosphorylation world. It also summarizes why our understanding of phosphorylation is still largely restricted to the acid stable phosphoproteome, and highlights the study of NM23 histidine kinase as an entrée into the world of histidine phosphorylation.
Collapse
Affiliation(s)
- Kevin Adam
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Tony Hunter
- Molecular and Cell Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
50
|
Sforna L, Megaro A, Pessia M, Franciolini F, Catacuzzeno L. Structure, Gating and Basic Functions of the Ca2+-activated K Channel of Intermediate Conductance. Curr Neuropharmacol 2018; 16:608-617. [PMID: 28875832 PMCID: PMC5997868 DOI: 10.2174/1570159x15666170830122402] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/21/2017] [Accepted: 07/22/2017] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The KCa3.1 channel is the intermediate-conductance member of the Ca2+- activated K channel superfamily. It is widely expressed in excitable and non-excitable cells, where it plays a major role in a number of cell functions. This paper aims at illustrating the main structural, biophysical and modulatory properties of the KCa3.1 channel, and providing an account of experimental data on its role in volume regulation and Ca2+ signals. METHODS Research and online content related to the structure, structure/function relationship, and physiological role of the KCa3.1 channel are reviewed. RESULTS Expressed in excitable and non-excitable cells, the KCa3.1 channel is voltage independent, its opening being exclusively gated by the binding of intracellular Ca2+ to calmodulin, a Ca2+- binding protein constitutively associated with the C-terminus of each KCa3.1 channel α subunit. The KCa3.1 channel activates upon high affinity Ca2+ binding, and in highly coordinated fashion giving steep Hill functions and relatively low EC50 values (100-350 nM). This high Ca2+ sensitivity is physiologically modulated by closely associated kinases and phosphatases. The KCa3.1 channel is normally activated by global Ca2+ signals as resulting from Ca2+ released from intracellular stores, or by the refilling influx through store operated Ca2+ channels, but cases of strict functional coupling with Ca2+-selective channels are also found. KCa3.1 channels are highly expressed in many types of cells, where they play major roles in cell migration and death. The control of these complex cellular processes is achieved by KCa3.1 channel regulation of the driving force for Ca2+ entry from the extracellular medium, and by mediating the K+ efflux required for cell volume control. CONCLUSION Much work remains to be done to fully understand the structure/function relationship of the KCa3.1 channels. Hopefully, this effort will provide the basis for a beneficial modulation of channel activity under pathological conditions.
Collapse
Affiliation(s)
| | | | | | - Fabio Franciolini
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| | - Luigi Catacuzzeno
- Address correspondence to these authors at the Department of Chemistry, Biology and Biotechnology, University of Perugia, Via Pascoli, 8-06123, Perugia; Tel: 39.075.585.5751; E-mails: and
| |
Collapse
|