1
|
Martin‐Ramirez S, Stouthamer J, Smakowska‐Luzan E. More questions than answers: insights into potential cysteine-rich receptor-like kinases redox signalling in Arabidopsis. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2025; 122:e70176. [PMID: 40300149 PMCID: PMC12040379 DOI: 10.1111/tpj.70176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 05/01/2025]
Abstract
Over the past few decades, significant advancements have been made in understanding how plasma-membrane localised receptor kinases (RKs) detect signals and activate responses to various stimuli. Numerous examples of ligand-induced receptor activation mechanisms and their downstream consequences have been characterised in detail. The crucial role of post-translational modifications (PTMs), such as the phosphorylation of receptor kinases, has been demonstrated concerning different cellular responses. Given the diverse structures and architectures of the extracellular domains (ECDs) of RKs, it is probable that various forms of PTMs also play an essential role in receptor activation, including cysteine oxidative modifications triggered by reactive oxygen species (ROS). The function of cysteine oxidative modifications as functional redox switches that modulate protein structure and function has been extensively studied across various multicellular organisms. Based on biochemical and structural characteristics, the family of cysteine-rich receptor-like kinases (CRK) emerges as excellent candidates for proteins regulated in a redox-dependent manner. This review provides a concise overview of cysteine's biochemical and structural properties in its role as a molecular redox switch. Drawing on the currently available literature, we describe how cysteine-redox signalling is maintained, particularly in plant cells. We further focus on extracellular ROS perception and the role of CRKs as promising candidates for ROS sensors in Arabidopsis thaliana. We discuss the structural and biochemical properties of CRKs, their involvement in plant growth and defence processes, and our perspective on why CRKs could be key components of the ROS sensing machinery or ROS sensors, especially regarding the dimerization abilities of CRKs. Finally, we highlight the current challenges in identifying and quantifying cysteine oxidative modifications and propose methods for detecting ROS-modified cysteines that may be promising for investigating the role of CRKs in extracellular ROS perception and signalling.
Collapse
Affiliation(s)
- Sergio Martin‐Ramirez
- Laboratory of BiochemistryWageningen University and ResearchWageningenThe Netherlands
| | - Jente Stouthamer
- Laboratory of BiochemistryWageningen University and ResearchWageningenThe Netherlands
| | | |
Collapse
|
2
|
Braden DC, Adbel-Salam MAL, Asan A, Skoko J, Lu H, Conrads TP, Freeman BA, Schopfer FJ, Saini I, Kuper J, Kisker C, Uboveja A, Tangudu NK, Aird KM, Davis AJ, Neumann CA. Chemoproteomic analysis reveals RECQL4 as a mediator of nitroalkene-dependent double-strand break repair inhibition in cancer. RESEARCH SQUARE 2025:rs.3.rs-6141403. [PMID: 40196015 PMCID: PMC11975020 DOI: 10.21203/rs.3.rs-6141403/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Nitroalkenes are endogenous products generated by the metabolism of unsaturated fatty acids. They are generated under oxidative stress conditions, mediating important anti-inflammatory signaling activities through covalent modification of protein cysteine thiols. Despite being cytoprotective in benign tissue, nitroalkenes display single-agent anti-proliferative activity in breast cancer cells and sensitize them to multiple DNA-damaging agents. Initial mechanistic evidence suggested that nitroalkene anti-cancer activities are partially mediated by inhibition of homologous recombination (HR) through the recombinase RAD51 at Cys319. However, nitroalkenes are multi-target agents, and thus, it is likely that other important DNA repair targets beyond RAD51 are modified by nitroalkenes, contributing to their anti-cancer effects. We, therefore, conducted a global proteomics analysis to address this question. This analysis led to the identification of the recQ helicase RECQL4 with a nitro-alkylation at Cys1052. This modification was further confirmed by click chemistry-based chemoproteomics and determined to be DNA damage-dependent. Functional analyses demonstrated that nitroalkene modification inhibits RECQL4 ATP-dependent helicase activity and disrupts DSB end resection and downstream homology-dependent repair. Furthermore, experiments with C1052S mutant RECQL4 revealed that RECQL4 is a major mediator of nitroalkene effects on end resection, DSB formation, and repair. The evidence presented here denotes RECQL4 as an important nitroalkene target conferring DSB repair inhibition and supports further evaluation of nitroalkenes as therapeutic agents in RECQL4-amplified cancers.
Collapse
Affiliation(s)
- Dennis C Braden
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Mostafa A L Adbel-Salam
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Alparslan Asan
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - John Skoko
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Huiming Lu
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX75390, USA
| | - Thomas P Conrads
- Gynecologic Cancer Center of Excellence, Department of Gynecologic Surgery and Obstetrics, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA; Women's Health Integrated Research Center, Women's Service Line, Inova Health System, Annandale, VA 22003, United States; Murtha Cancer Center Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Walter Reed National Military Medical Center, Bethesda, MD 20889, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine (C3M) University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Heart, Lung, Blood, and Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA; Pittsburgh Liver Research Center (PLRC), University of Pittsburgh, Pittsburgh, PA, USA; Center for Metabolism and Mitochondrial Medicine (C3M) University of Pittsburgh, Pittsburgh, PA, USA
| | - Ishu Saini
- Rudolf-Virchow-Zentrum-Center for Integrative and Translational Bioimaging Institute for Structural Biology, University of Würzburg, Würzburg, Germany
| | - Jochen Kuper
- Rudolf-Virchow-Zentrum-Center for Integrative and Translational Bioimaging Institute for Structural Biology, University of Würzburg, Würzburg, Germany
| | - Caroline Kisker
- Rudolf-Virchow-Zentrum-Center for Integrative and Translational Bioimaging Institute for Structural Biology, University of Würzburg, Würzburg, Germany
| | - Apoorva Uboveja
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Naveen K Tangudu
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Katherine M Aird
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Anthony J Davis
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX75390, USA
| | - Carola A Neumann
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA; Women's Cancer Research Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA, 15213, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| |
Collapse
|
3
|
Handegard V, Lunde PK, Frisk M, Seynnes O, Ørtenblad N, Louch WE, Paulsen G, Raastad T. Myofiber structure, sarcoplasmic reticulum Ca 2+ handling, and contractile function after muscle-damaging exercise in humans. Physiol Rep 2025; 13:e70204. [PMID: 39895015 PMCID: PMC11788311 DOI: 10.14814/phy2.70204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/18/2024] [Accepted: 01/06/2025] [Indexed: 02/04/2025] Open
Abstract
Exercise-induced muscle damage (EIMD) is characterized by a severe and prolonged decline in force-generating capacity. However, the precise cellular mechanisms underlying the observed long-lasting decline in force-generating capacity associated with EIMD are still unclear. We investigated in vivo force generation and ex vivo Ca2+-activated force generation, Ca2+ sensitivity, and myofiber Ca2+ handling systems (SR and t-tubules) in human biceps brachii before and 2, 48, and 96 h after eccentrically muscle-damaging contractions and in non-exercised control arm. The force-generating capacity declined by 50 ± 13% 3 h after exercise and was still not recovered after 96 h. The force-Ca relationship of skinned myofibers revealed an impaired maximal Ca2+-activated force in MHC I-fibers, but not MHC II-fibers 48 h after exercise. Further, Ca2+ sensitivity was increased in MHC II-fibers, which was reversed after incubation with a strong reductant. There was a biphasic increase in SERCA sulfonylation, and a parallel reduction in the SR Ca2+ uptake rate, with no effects on SR vesicle leak or SR vesicle Ca2+ release rate. T-tubules showed a progressive increase in the density of longitudinal tubules by 96 h after exercise. In conclusion, MHC II-fiber Ca2+ sensitivity was increased 48 h after exercise, attributed to changes in the REDOX status. 96 h after exercise SR vesicle Ca2+ uptake was impaired, and an increased number of longitudinal tubules were observed. These alterations may contribute to the impaired force generation evident at the late stage of recovery.
Collapse
Affiliation(s)
- V. Handegard
- Department of Physical PerformanceNorwegian School of Sport SciencesOsloNorway
| | - P. K. Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOsloNorway
| | - M. Frisk
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOsloNorway
| | - O. Seynnes
- Department of Physical PerformanceNorwegian School of Sport SciencesOsloNorway
| | - N. Ørtenblad
- Department of Sports Science and Clinical BiomechanicsUniversity of Southern DenmarkOdenseDenmark
| | - W. E. Louch
- Institute for Experimental Medical Research, Oslo University Hospital and University of OsloOsloNorway
| | - G. Paulsen
- Department of Physical PerformanceNorwegian School of Sport SciencesOsloNorway
| | - T. Raastad
- Department of Physical PerformanceNorwegian School of Sport SciencesOsloNorway
| |
Collapse
|
4
|
Sies H, Mailloux RJ, Jakob U. Fundamentals of redox regulation in biology. Nat Rev Mol Cell Biol 2024; 25:701-719. [PMID: 38689066 PMCID: PMC11921270 DOI: 10.1038/s41580-024-00730-2] [Citation(s) in RCA: 93] [Impact Index Per Article: 93.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/02/2024]
Abstract
Oxidation-reduction (redox) reactions are central to the existence of life. Reactive species of oxygen, nitrogen and sulfur mediate redox control of a wide range of essential cellular processes. Yet, excessive levels of oxidants are associated with ageing and many diseases, including cardiological and neurodegenerative diseases, and cancer. Hence, maintaining the fine-tuned steady-state balance of reactive species production and removal is essential. Here, we discuss new insights into the dynamic maintenance of redox homeostasis (that is, redox homeodynamics) and the principles underlying biological redox organization, termed the 'redox code'. We survey how redox changes result in stress responses by hormesis mechanisms, and how the lifelong cumulative exposure to environmental agents, termed the 'exposome', is communicated to cells through redox signals. Better understanding of the molecular and cellular basis of redox biology will guide novel redox medicine approaches aimed at preventing and treating diseases associated with disturbed redox regulation.
Collapse
Affiliation(s)
- Helmut Sies
- Institute for Biochemistry and Molecular Biology I, Faculty of Medicine, Heinrich Heine University Düsseldorf, Düsseldorf, Germany.
- Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany.
| | - Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Sainte-Anne-de-Bellevue, Quebec, Canada.
| | - Ursula Jakob
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Calabrese V, Osakabe N, Siracusa R, Modafferi S, Di Paola R, Cuzzocrea S, Jacob UM, Fritsch T, Abdelhameed AS, Rashan L, Wenzel U, Franceschi C, Calabrese EJ. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech Ageing Dev 2024; 220:111960. [PMID: 38971236 DOI: 10.1016/j.mad.2024.111960] [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: 04/27/2024] [Revised: 06/20/2024] [Accepted: 06/25/2024] [Indexed: 07/08/2024]
Abstract
Neurodegenerative diseases have multifactorial pathogenesis, mainly involving neuroinflammatory processes. Finding drugs able to treat these diseases, expecially because for most of these diseases there are no effective drugs, and the current drugs cause undesired side effects, represent a crucial point. Most in vivo and in vitro studies have been concentrated on various aspects related to neurons (e.g. neuroprotection), however, there has not been focus on the prevention of early stages involving glial cell activation and neuroinflammation. Recently, it has been demonstrated that nutritional phytochemicals including polyphenols, the main active constituents of the Mediterranean diet, maintain redox balance and neuroprotection through the activation of hormetic vitagene pathway. Recent lipidomics data from our laboratory indicate mushrooms as strong nutritional neuronutrients with strongly activity against neuroinflammation in Meniere' diseaseas, a model of cochleovestibular neural degeneration, as well as in animal model of traumatic brain injury, or rotenone induced parkinson's disease. Moreover, Hidrox®, an aqueous extract of olive containing hydroxytyrosol, and Boswellia, acting as Nrf2 activators, promote resilience by enhancing the redox potential, and thus, regulate through hormetic mechanisms, cellular stress response mechanisms., Thus, modulation of cellular stress pathways, in particular vitagenes system, may be an innovative approach for therapeutic intervention in neurodegenerative disorders.
Collapse
Affiliation(s)
- Vittorio Calabrese
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Naomi Osakabe
- Department of Bioscience and Engineering, Shibaura Institute Technology, Tokyo, Japan.
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | - Sergio Modafferi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Rosanna Di Paola
- Department of Veterinary Sciences, University of Messina, Messina 98168, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina 98166, Italy
| | | | | | - Ali S Abdelhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| | - Luay Rashan
- Biodiversity Unit, Dhofar University, Salalah, Oman
| | - Uwe Wenzel
- Institut für Ernährungswissenschaft, Justus Liebig Universitat Giessen, Germany
| | | | - Edward J Calabrese
- Department of Environmental Health Sciences, Morrill I, N344, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
6
|
Anjo SI, He Z, Hussain Z, Farooq A, McIntyre A, Laughton CA, Carvalho AN, Finelli MJ. Protein Oxidative Modifications in Neurodegenerative Diseases: From Advances in Detection and Modelling to Their Use as Disease Biomarkers. Antioxidants (Basel) 2024; 13:681. [PMID: 38929122 PMCID: PMC11200609 DOI: 10.3390/antiox13060681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/26/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
Oxidation-reduction post-translational modifications (redox-PTMs) are chemical alterations to amino acids of proteins. Redox-PTMs participate in the regulation of protein conformation, localization and function, acting as signalling effectors that impact many essential biochemical processes in the cells. Crucially, the dysregulation of redox-PTMs of proteins has been implicated in the pathophysiology of numerous human diseases, including neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. This review aims to highlight the current gaps in knowledge in the field of redox-PTMs biology and to explore new methodological advances in proteomics and computational modelling that will pave the way for a better understanding of the role and therapeutic potential of redox-PTMs of proteins in neurodegenerative diseases. Here, we summarize the main types of redox-PTMs of proteins while providing examples of their occurrence in neurodegenerative diseases and an overview of the state-of-the-art methods used for their detection. We explore the potential of novel computational modelling approaches as essential tools to obtain insights into the precise role of redox-PTMs in regulating protein structure and function. We also discuss the complex crosstalk between various PTMs that occur in living cells. Finally, we argue that redox-PTMs of proteins could be used in the future as diagnosis and prognosis biomarkers for neurodegenerative diseases.
Collapse
Affiliation(s)
- Sandra I. Anjo
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3004-517 Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789 Coimbra, Portugal
| | - Zhicheng He
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Zohaib Hussain
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Aruba Farooq
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Alan McIntyre
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| | - Charles A. Laughton
- Biodiscovery Institute, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | - Andreia Neves Carvalho
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Mattéa J. Finelli
- Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK
| |
Collapse
|
7
|
Ujcikova H, Lee YS, Roubalova L, Svoboda P. The impact of multifunctional enkephalin analogs and morphine on the protein changes in crude membrane fractions isolated from the rat brain cortex and hippocampus. Peptides 2024; 174:171165. [PMID: 38307418 DOI: 10.1016/j.peptides.2024.171165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/12/2024] [Accepted: 01/29/2024] [Indexed: 02/04/2024]
Abstract
Endogenous opioid peptides serve as potent analgesics through the opioid receptor (OR) activation. However, they often suffer from poor metabolic stability, low lipophilicity, and low blood-brain barrier permeability. Researchers have developed many strategies to overcome the drawbacks of current pain medications and unwanted biological effects produced by the interaction with opioid receptors. Here, we tested multifunctional enkephalin analogs LYS739 (MOR/DOR agonist and KOR partial antagonist) and LYS744 (MOR/DOR agonist and KOR full antagonist) under in vivo conditions in comparison with MOR agonist, morphine. We applied 2D electrophoretic resolution to investigate differences in proteome profiles of crude membrane (CM) fractions isolated from the rat brain cortex and hippocampus exposed to the drugs (10 mg/kg, seven days). Our results have shown that treatment with analog LYS739 induced the most protein changes in cortical and hippocampal samples. The identified proteins were mainly associated with energy metabolism, cell shape and movement, apoptosis, protein folding, regulation of redox homeostasis, and signal transduction. Among these, the isoform of mitochondrial ATP synthase subunit beta (ATP5F1B) was the only protein upregulation in the hippocampus but not in the brain cortex. Contrarily, the administration of analog LYS744 caused a small number of protein alterations in both brain parts. Our results indicate that the KOR full antagonism, together with MOR/DOR agonism of multifunctional opioid ligands, can be beneficial in treating chronic pain states by reducing changes in protein expression levels but retaining analgesic efficacy.
Collapse
Affiliation(s)
- Hana Ujcikova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic.
| | - Yeon Sun Lee
- Department of Pharmacology, University of Arizona, Tucson, AZ 85724, USA
| | - Lenka Roubalova
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| | - Petr Svoboda
- Laboratory of Neurochemistry, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, Prague 4 14200, Czech Republic
| |
Collapse
|
8
|
Porta EOJ. Mapping the Evolution of Activity-Based Protein Profiling: A Bibliometric Review. Adv Pharm Bull 2023; 13:639-645. [PMID: 38022804 PMCID: PMC10676541 DOI: 10.34172/apb.2023.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/24/2023] [Accepted: 05/17/2023] [Indexed: 12/01/2023] Open
Abstract
Activity-based protein profiling (ABPP) is a chemoproteomic approach that employs small-molecule probes to directly evaluate protein functionality within complex proteomes. This technology has proven to be a potent strategy for mapping ligandable sites in organisms and has significantly impacted drug discovery processes by enabling the development of highly selective small-molecule inhibitors and the identification of new therapeutic molecular targets. Despite being nearly a quarter of a century old as a chemoproteomic tool, ABPP has yet to undergo a bibliometric analysis. In order to gauge its scholarly impact and evolution, a bibliometric analysis was performed, comparing all 1919 reported articles with the articles published in the last five years. Through a comprehensive data analysis, including a 5-step workflow, the most influential articles were identified, and their bibliometric parameters were determined. The 1919 analyzed articles span from 1999 to 2022, providing a comprehensive overview of the historical and current state of ABPP research. This analysis presents, for the first time, the characteristics of the most influential ABPP articles, offering valuable insight into the research conducted in this field and its potential future directions. The findings underscore the crucial role of ABPP in drug discovery and novel therapeutic target identification, as well as the need for continued advancements in the development of novel chemical probes and proteomic technologies to further expand the utility of ABPP.
Collapse
|
9
|
Jain M, Dhariwal R, Patil N, Ojha S, Tendulkar R, Tendulkar M, Dhanda PS, Yadav A, Kaushik P. Unveiling the Molecular Footprint: Proteome-Based Biomarkers for Alzheimer's Disease. Proteomes 2023; 11:33. [PMID: 37873875 PMCID: PMC10594437 DOI: 10.3390/proteomes11040033] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 10/25/2023] Open
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by progressive cognitive decline and memory loss. Early and accurate diagnosis of AD is crucial for implementing timely interventions and developing effective therapeutic strategies. Proteome-based biomarkers have emerged as promising tools for AD diagnosis and prognosis due to their ability to reflect disease-specific molecular alterations. There is of great significance for biomarkers in AD diagnosis and management. It emphasizes the limitations of existing diagnostic approaches and the need for reliable and accessible biomarkers. Proteomics, a field that comprehensively analyzes the entire protein complement of cells, tissues, or bio fluids, is presented as a powerful tool for identifying AD biomarkers. There is a diverse range of proteomic approaches employed in AD research, including mass spectrometry, two-dimensional gel electrophoresis, and protein microarrays. The challenges associated with identifying reliable biomarkers, such as sample heterogeneity and the dynamic nature of the disease. There are well-known proteins implicated in AD pathogenesis, such as amyloid-beta peptides, tau protein, Apo lipoprotein E, and clusterin, as well as inflammatory markers and complement proteins. Validation and clinical utility of proteome-based biomarkers are addressing the challenges involved in validation studies and the diagnostic accuracy of these biomarkers. There is great potential in monitoring disease progression and response to treatment, thereby aiding in personalized medicine approaches for AD patients. There is a great role for bioinformatics and data analysis in proteomics for AD biomarker research and the importance of data preprocessing, statistical analysis, pathway analysis, and integration of multi-omics data for a comprehensive understanding of AD pathophysiology. In conclusion, proteome-based biomarkers hold great promise in the field of AD research. They provide valuable insights into disease mechanisms, aid in early diagnosis, and facilitate personalized treatment strategies. However, further research and validation studies are necessary to harness the full potential of proteome-based biomarkers in clinical practice.
Collapse
Affiliation(s)
- Mukul Jain
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Rupal Dhariwal
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Nil Patil
- Cell and Developmental Biology Laboratory, Research and Development Cell, Parul University, Vadodara 391760, India; (R.D.); (N.P.)
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Sandhya Ojha
- Department of Life Sciences, Parul Institute of Applied Sciences, Parul University, Vadodara 391760, India;
| | - Reshma Tendulkar
- Vivekanand Education Society, College of Pharmacy, Chembur, Mumbai 400071, India;
| | - Mugdha Tendulkar
- Sardar Vallabhbhai Patel College of Science, Mira Rd (East), Thane 400071, India;
| | | | - Alpa Yadav
- Department of Botany, Indira Gandhi University, Meerpur, Rewari 122502, India;
| | - Prashant Kaushik
- Instituto de Conservacióny Mejora de la Agrodiversidad Valenciana, Universitat Politècnica de València, 46022 Valencia, Spain
| |
Collapse
|
10
|
Gowthami N, Pursotham N, Dey G, Ghose V, Sathe G, Pruthi N, Shukla D, Gayathri N, Santhoshkumar R, Padmanabhan B, Chandramohan V, Mahadevan A, Srinivas Bharath MM. Neuroanatomical zones of human traumatic brain injury reveal significant differences in protein profile and protein oxidation: Implications for secondary injury events. J Neurochem 2023; 167:218-247. [PMID: 37694499 DOI: 10.1111/jnc.15953] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/03/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023]
Abstract
Traumatic brain injury (TBI) causes significant neurological deficits and long-term degenerative changes. Primary injury in TBI entails distinct neuroanatomical zones, i.e., contusion (Ct) and pericontusion (PC). Their dynamic expansion could contribute to unpredictable neurological deterioration in patients. Molecular characterization of these zones compared with away from contusion (AC) zone is invaluable for TBI management. Using proteomics-based approach, we were able to distinguish Ct, PC and AC zones in human TBI brains. Ct was associated with structural changes (blood-brain barrier (BBB) disruption, neuroinflammation, axonal injury, demyelination and ferroptosis), while PC was associated with initial events of secondary injury (glutamate excitotoxicity, glial activation, accumulation of cytoskeleton proteins, oxidative stress, endocytosis) and AC displayed mitochondrial dysfunction that could contribute to secondary injury events and trigger long-term degenerative changes. Phosphoproteome analysis in these zones revealed that certain differentially phosphorylated proteins synergistically contribute to the injury events along with the differentially expressed proteins. Non-synaptic mitochondria (ns-mito) was associated with relatively more differentially expressed proteins (DEPs) compared to synaptosomes (Syn), while the latter displayed increased protein oxidation including tryptophan (Trp) oxidation. Proteomic analysis of immunocaptured complex I (CI) from Syn revealed increased Trp oxidation in Ct > PC > AC (vs. control). Oxidized W272 in the ND1 subunit of CI, revealed local conformational changes in ND1 and the neighboring subunits, as indicated by molecular dynamics simulation (MDS). Taken together, neuroanatomical zones in TBI show distinct protein profile and protein oxidation representing different primary and secondary injury events with potential implications for TBI pathology and neurological status of the patients.
Collapse
Affiliation(s)
- Niya Gowthami
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Nithya Pursotham
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Gourav Dey
- Proteomics and Bioinformatics Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
- Institute of Bioinformatics, Bengaluru, India
| | - Vivek Ghose
- Proteomics and Bioinformatics Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
- Institute of Bioinformatics, Bengaluru, India
| | - Gajanan Sathe
- Proteomics and Bioinformatics Laboratory, Neurobiology Research Center, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
- Institute of Bioinformatics, Bengaluru, India
| | - Nupur Pruthi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Narayanappa Gayathri
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Rashmi Santhoshkumar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Balasundaram Padmanabhan
- Department of Biophysics, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - Vivek Chandramohan
- Department of Biotechnology, Siddaganga Institute of Technology (SIT), Tumakuru, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, Karnataka, India
| |
Collapse
|
11
|
Zhong Q, Xiao X, Qiu Y, Xu Z, Chen C, Chong B, Zhao X, Hai S, Li S, An Z, Dai L. Protein posttranslational modifications in health and diseases: Functions, regulatory mechanisms, and therapeutic implications. MedComm (Beijing) 2023; 4:e261. [PMID: 37143582 PMCID: PMC10152985 DOI: 10.1002/mco2.261] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 03/26/2023] [Accepted: 03/27/2023] [Indexed: 05/06/2023] Open
Abstract
Protein posttranslational modifications (PTMs) refer to the breaking or generation of covalent bonds on the backbones or amino acid side chains of proteins and expand the diversity of proteins, which provides the basis for the emergence of organismal complexity. To date, more than 650 types of protein modifications, such as the most well-known phosphorylation, ubiquitination, glycosylation, methylation, SUMOylation, short-chain and long-chain acylation modifications, redox modifications, and irreversible modifications, have been described, and the inventory is still increasing. By changing the protein conformation, localization, activity, stability, charges, and interactions with other biomolecules, PTMs ultimately alter the phenotypes and biological processes of cells. The homeostasis of protein modifications is important to human health. Abnormal PTMs may cause changes in protein properties and loss of protein functions, which are closely related to the occurrence and development of various diseases. In this review, we systematically introduce the characteristics, regulatory mechanisms, and functions of various PTMs in health and diseases. In addition, the therapeutic prospects in various diseases by targeting PTMs and associated regulatory enzymes are also summarized. This work will deepen the understanding of protein modifications in health and diseases and promote the discovery of diagnostic and prognostic markers and drug targets for diseases.
Collapse
Affiliation(s)
- Qian Zhong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xina Xiao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Yijie Qiu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhiqiang Xu
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Chunyu Chen
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Baochen Chong
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Xinjun Zhao
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shan Hai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Shuangqing Li
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Zhenmei An
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| | - Lunzhi Dai
- Department of Endocrinology and MetabolismGeneral Practice Ward/International Medical Center WardGeneral Practice Medical Center and National Clinical Research Center for GeriatricsState Key Laboratory of BiotherapyWest China Hospital, Sichuan UniversityChengduChina
| |
Collapse
|
12
|
Shellaiah M, Sun KW. Review on Carbon Dot-Based Fluorescent Detection of Biothiols. BIOSENSORS 2023; 13:335. [PMID: 36979547 PMCID: PMC10046571 DOI: 10.3390/bios13030335] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/01/2023] [Accepted: 03/01/2023] [Indexed: 06/18/2023]
Abstract
Biothiols, such as cysteine (Cys), homocysteine (Hcy), and glutathione (GSH), play a vital role in gene expression, maintaining redox homeostasis, reducing damages caused by free radicals/toxins, etc. Likewise, abnormal levels of biothiols can lead to severe diseases, such as Alzheimer's disease (AD), neurotoxicity, hair depigmentation, liver/skin damage, etc. To quantify the biothiols in a biological system, numerous low-toxic probes, such as fluorescent quantum dots, emissive organic probes, composited nanomaterials, etc., have been reported with real-time applications. Among these fluorescent probes, carbon-dots (CDs) have become attractive for biothiols quantification because of advantages of easy synthesis, nano-size, crystalline properties, low-toxicity, and real-time applicability. A CDs-based biothiols assay can be achieved by fluorescent "Turn-On" and "Turn-Off" responses via direct binding, metal complex-mediated detection, composite enhanced interaction, reaction-based reports, and so forth. To date, the availability of a review focused on fluorescent CDs-based biothiols detection with information on recent trends, mechanistic aspects, linear ranges, LODs, and real applications is lacking, which allows us to deliver this comprehensive review. This review delivers valuable information on reported carbon-dots-based biothiols assays, the underlying mechanism, their applications, probe/CDs selection, sensory requirement, merits, limitations, and future scopes.
Collapse
Affiliation(s)
| | - Kien Wen Sun
- Department of Applied Chemistry, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| |
Collapse
|
13
|
LI J, WANG G, YE M, QIN H. [Advances in applications of activity-based chemical probes in the characterization of amino acid reactivities]. Se Pu 2023; 41:14-23. [PMID: 36633073 PMCID: PMC9837674 DOI: 10.3724/sp.j.1123.2022.05013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Indexed: 01/13/2023] Open
Abstract
The discovery of novel drug targets enhances the development of novel drugs, and the discovery of novel target proteins depends on highly accurate high-throughput methods of analyzing drug-protein interactions. Protein expression levels, spatial localization, and structural differences directly affect pharmacodynamics. To date, >20000 proteins have been discovered in the human proteome by the genome and proteome projects via gene and protein sequencing. Understanding the biological functions of proteins is critical in identifying and regulating biological processes, with most remaining unidentified. Until recently, >85% of proteins were considered undruggable, mainly because of the lack of binding pockets and active sites targeted by small molecules. Therefore, characterization of the reactive sites of amino acids based on proteomic hierarchy is the key to novel drug design. Recently, with the rapid development of mass spectrometry (MS), the study of drug-target protein interactions based on proteomics technology has been considerably promoted. Activity-based protein profiling (ABPP) is an active chemical probe-based method of detecting functional enzymes and drug targets in complex samples. Compared with classical proteomics strategies, ABPP is based mainly on protein activity. It has been successfully utilized to characterize the activities of numerous protease families with crucial biological functions, such as serine hydrolases, protein kinases, glycosidases, and metalloenzymes. It has also been used to identify key enzymes that are closely related to diseases and develop covalent inhibitors for use in disease treatment. The technology used in proteome analysis ranges from gel electrophoresis to high-throughput MS due to the progress of MS technology. ABPP strategies combined with chemical probe labeling and quantitative MS enable the characterization of amino acid activity, which may enhance the discovery of novel drug targets and the development of lead compounds. Amino acid residues play critical roles in protein structures and functions, and covalent drugs targeting these amino acids are effective in treating numerous diseases. There are 20 main types of natural amino acids, with different reactivities, in the proteins in the human body. In addition, the proteins and amino acids are affected by the spatial microenvironment, leading to significant differences in their spatial reactivities. The key in evaluating the reactivities of amino acids via ABPP is to select those with high reactivities. The core of the ABPP strategy is the use of chemical probes to label amino acid sites that exhibit higher activities in certain environments. The activity-based probe (ABP) at the core of ABPP consists of three components: reactive, reporter groups and a linker. The reactive group is the basis of the ABP and anchors the drug target via strong forces, such as covalent bonds. The reaction exhibits a high specificity and conversion rate and should display a good biocompatibility. Activity probes based on different amino acid residues have been developed, and the screening of amino acid activity combined with isotope labeling is a new focus of research. Currently, different types of ABPs have been developed to target amino acids and characterize amino acid reactivity, such as cysteine labeled with an electrophilic iodoacetamide probe and lysine labeled with activated esters. ABPP facilitates the discovery of potentially therapeutic protein targets, the screening of lead compounds, and the identification of drug targets, thus aiding the design of novel drugs. This review focuses on the development of ABPP methods and the progress in the screening of amino acid reactivity using ABPs, which should be promising methods for use in designing targeted drugs with covalent interactions.
Collapse
|
14
|
Mass spectrometry analysis of S-nitrosylation of proteins and its role in cancer, cardiovascular and neurodegenerative diseases. Trends Analyt Chem 2022. [DOI: 10.1016/j.trac.2022.116625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
15
|
Ontario ML, Siracusa R, Modafferi S, Scuto M, Sciuto S, Greco V, Bertuccio MP, Salinaro AT, Crea R, Calabrese EJ, Di Paola R, Calabrese V. POTENTIAL PREVENTION AND TREATMENT OF NEURODEGENERATIVE DISORDERS BY OLIVE POLYPHENOLS AND HYDROX. Mech Ageing Dev 2022; 203:111637. [DOI: 10.1016/j.mad.2022.111637] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 12/15/2022]
|