1
|
Dall'Agnese A, Zheng MM, Moreno S, Platt JM, Hoang AT, Kannan D, Dall'Agnese G, Overholt KJ, Sagi I, Hannett NM, Erb H, Corradin O, Chakraborty AK, Lee TI, Young RA. Proteolethargy is a pathogenic mechanism in chronic disease. Cell 2025; 188:207-221.e30. [PMID: 39610243 PMCID: PMC11724756 DOI: 10.1016/j.cell.2024.10.051] [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: 03/16/2024] [Revised: 08/07/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
The pathogenic mechanisms of many diseases are well understood at the molecular level, but there are prevalent syndromes associated with pathogenic signaling, such as diabetes and chronic inflammation, where our understanding is more limited. Here, we report that pathogenic signaling suppresses the mobility of a spectrum of proteins that play essential roles in cellular functions known to be dysregulated in these chronic diseases. The reduced protein mobility, which we call proteolethargy, was linked to cysteine residues in the affected proteins and signaling-related increases in excess reactive oxygen species. Diverse pathogenic stimuli, including hyperglycemia, dyslipidemia, and inflammation, produce similar reduced protein mobility phenotypes. We propose that proteolethargy is an overlooked cellular mechanism that may account for various pathogenic features of diverse chronic diseases.
Collapse
Affiliation(s)
| | - Ming M Zheng
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Shannon Moreno
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jesse M Platt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - An T Hoang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Deepti Kannan
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Kalon J Overholt
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Ido Sagi
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Nancy M Hannett
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Hailey Erb
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Olivia Corradin
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Arup K Chakraborty
- Department of Physics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Institute of Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02139, USA
| | - Tong Ihn Lee
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA.
| | - Richard A Young
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
2
|
Hu Y, Li H, Zhang X, Song Y, Liu J, Pu J, Wen S, Xu H, Xin H, Wang B, Yang S. Identification of two repurposed drugs targeting GSDMD oligomerization interface I to block pyroptosis. Cell Chem Biol 2024; 31:2024-2038.e7. [PMID: 39486414 DOI: 10.1016/j.chembiol.2024.10.002] [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: 04/30/2024] [Revised: 08/14/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
As an executor of pyroptosis, gasdermin D (GSDMD) plays a critical role in inflammatory diseases and cancer. Thus, GSDMD is currently being widely explored as a drug target. Existing inhibitors targeting GSDMD, such as necrosulfonamide, disulfiram, and fumarate, primarily prevent pyroptosis by modifying human/mouse C191/C192 in the N-terminal fragment of GSDMD. However, cysteine modification can prevent the function of important proteins or enzymes, thereby leading to adverse reactions. Here, we chose an alternative key intervention site for GSDMD activation, which is located at the oligomerization interface I of its pore-forming structure. Through high-throughput virtual and experimental screening and in combination with efficacy and pharmacological validation, we have identified two safe, specific "repurposed drugs" that potently suppress GSDMD-mediated pyroptosis. Moreover, the candidates exhibited synergistic therapeutic effects of "1 + 1>2" in murine sepsis and tumorigenesis models. These recently identified GSDMD inhibitors hold great promise for clinical translation in the development of anti-inflammatory and anti-cancer immunotherapies.
Collapse
Affiliation(s)
- Yingchao Hu
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Honghui Li
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China; Department of Clinical Laboratory, The Third Affiliated Hospital of Nanchang University (The First Hospital of Nanchang), Nanchang 330008, Jiangxi, P.R. China
| | - Xiangyu Zhang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yuxian Song
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Jun Liu
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Jie Pu
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Shuang Wen
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Hongyang Xu
- Department of Critical Care Medicine, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi 214000, China.
| | - Hongliang Xin
- Department of Pharmaceutics, School of Pharmacy, Nanjing Medical University, 101 Longmian Road, Jiangning, Nanjing 211166, China.
| | - Bingwei Wang
- Department of Pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China.
| | - Shuo Yang
- The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Department of Immunology, Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
3
|
Domingo-Relloso A, Riffo-Campos AL, Zhao N, Ayala G, Haack K, Manterola C, Rhoades DA, Umans JG, Fallin MD, Herreros-Martinez M, Pollan M, Boerwinkle E, Platz EA, Jones MR, Bressler J, Joehanes R, Ryan CP, Gonzalez JR, Levy D, Belsky DW, Cole SA, Michaud DS, Navas-Acien A, Tellez-Plaza M. Multicohort Epigenome-Wide Association Study of All-Cause Cardiovascular Disease and Cancer Incidence: A Cardio-Oncology Approach. JACC CardioOncol 2024; 6:731-742. [PMID: 39479324 PMCID: PMC11520201 DOI: 10.1016/j.jaccao.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Background Emerging evidence reveals a complex relationship between cardiovascular disease (CVD) and cancer, which share common risk factors and biological pathways. Objectives The aim of this study was to evaluate common epigenetic signatures for CVD and cancer incidence in 3 ethnically diverse cohorts: Native Americans from the SHS (Strong Heart Study), European Americans from the FHS (Framingham Heart Study), and European Americans and African Americans from the ARIC (Atherosclerosis Risk In Communities) study. Methods A 2-stage strategy was used that included first conducting untargeted epigenome-wide association studies for each cohort and then running targeted models in the union set of identified differentially methylated positions (DMPs). We also explored potential molecular pathways by conducting a bioinformatics analysis. Results Common DMPs were identified across all populations. In a subsequent meta-analysis, 3 and 1 of those DMPs were statistically significant for CVD only and both cancer and CVD, respectively. No meta-analyzed DMPs were statistically significant for cancer only. The enrichment analysis pointed to interconnected biological pathways involved in cancer and CVD. In the DrugBank database, elements related to 1-carbon metabolism and cancer and CVD medications were identified as potential drugs for target gene products. In an additional analysis restricted to the 950 SHS participants who developed incident CVD, the C index for incident cancer increased from 0.618 (95% CI: 0.570-0.672) to 0.971 (95% CI: 0.963-0.978) when adjusting the models for the combined cancer and CVD DMPs identified in the other cohorts. Conclusions These results point to molecular pathways and potential treatments for precision prevention of CVD and cancer. Screening based on common epigenetic signatures of incident CVD and cancer may help identify patients with newly diagnosed CVD at increased cancer risk.
Collapse
Affiliation(s)
- Arce Domingo-Relloso
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA
- Integrative Epidemiology Group, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
- Department of Chronic Disease Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Angela L. Riffo-Campos
- Universidad de La Frontera, Ph.D. Program in Medical Sciences; and Millennium Nucleus on Sociomedicine (SocioMed), Temuco, Chile
- Department of Computer Science, Universidad de Valencia, Valencia, Spain
- Center for Cancer Prevention and Control, Santiago, Chile
| | - Naisi Zhao
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Guillermo Ayala
- Department of Computer Science, Universidad de Valencia, Valencia, Spain
| | - Karin Haack
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Carlos Manterola
- Universidad de La Frontera, Ph.D. Program in Medical Sciences; and Millennium Nucleus on Sociomedicine (SocioMed), Temuco, Chile
| | - Dorothy A. Rhoades
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | | | - M Daniele Fallin
- Emory University Rollins School of Public Health, Atlanta, Georgia, USA
| | | | - Marina Pollan
- Department of Chronic Disease Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
| | - Eric Boerwinkle
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Miranda R. Jones
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, and Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA
| | - Jan Bressler
- The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Roby Joehanes
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Calen P. Ryan
- Columbia Aging Center, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Juan R. Gonzalez
- Bioinformatics and Genetic Epidemiology Unit, Instituto de Salud Global, Barcelona, Spain
| | - Daniel Levy
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
- Framingham Heart Study, Framingham, Massachusetts, USA
| | - Daniel W. Belsky
- Department of Epidemiology, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, Texas, USA
| | - Dominique S. Michaud
- Department of Public Health & Community Medicine, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ana Navas-Acien
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, New York, USA
| | - Maria Tellez-Plaza
- Integrative Epidemiology Group, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
- Department of Chronic Disease Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
4
|
Monaghan RM, Naylor RW, Flatman D, Kasher PR, Williams SG, Keavney BD. FLT4 causes developmental disorders of the cardiovascular and lymphovascular systems via pleiotropic molecular mechanisms. Cardiovasc Res 2024; 120:1164-1176. [PMID: 38713105 PMCID: PMC11368125 DOI: 10.1093/cvr/cvae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/08/2024] Open
Abstract
AIMS Rare, deleterious genetic variants in FLT4 are associated with Tetralogy of Fallot (TOF), the most common cyanotic congenital heart disease. The distinct genetic variants in FLT4 are also an established cause of Milroy disease, the most prevalent form of primary hereditary lymphoedema. The phenotypic features of these two conditions are non-overlapping, implying pleiotropic cellular mechanisms during development. METHODS AND RESULTS In this study, we show that FLT4 variants identified in patients with TOF, when expressed in primary human endothelial cells, cause aggregation of FLT4 protein in the perinuclear endoplasmic reticulum, activating proteostatic and metabolic signalling, whereas lymphoedema-associated FLT4 variants and wild-type (WT) FLT4 do not. FLT4 TOF variants display characteristic gene expression profiles in key developmental signalling pathways, revealing a role for FLT4 in cardiogenesis distinct from its role in lymphatic development. Inhibition of proteostatic signalling abrogates these effects, identifying potential avenues for therapeutic intervention. Depletion of flt4 in zebrafish caused cardiac phenotypes of reduced heart size and altered heart looping. These phenotypes were rescued with coinjection of WT human FLT4 mRNA, but incompletely or not at all by mRNA harbouring FLT4 TOF variants. CONCLUSION Taken together, we identify a pathogenic mechanism for FLT4 variants predisposing to TOF that is distinct from the known dominant negative mechanism of Milroy-causative variants. FLT4 variants give rise to conditions of the two circulatory subdivisions of the vascular system via distinct developmental pleiotropic molecular mechanisms.
Collapse
Affiliation(s)
- Richard M Monaghan
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
| | - Richard W Naylor
- Wellcome Centre for Cell Matrix Research, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester, M13 9PN, UK
| | - Daisy Flatman
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Paul R Kasher
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, University of Manchester, Oxford Road, Manchester, M13 9PT, UK
| | - Simon G Williams
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
| | - Bernard D Keavney
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, Manchester Academic Health Science Centre, University of Manchester, 5th Floor, AV Hill Building, Oxford Road, Manchester, M13 9NT, UK
- Manchester Heart Institute, Manchester University NHS Foundation Trust, Oxford Road, M13 9WL, UK
| |
Collapse
|
5
|
Guberovic I, Frezza C. Functional implications of fumarate-induced cysteine succination. Trends Biochem Sci 2024; 49:775-790. [PMID: 38876954 DOI: 10.1016/j.tibs.2024.05.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/29/2024] [Accepted: 05/10/2024] [Indexed: 06/16/2024]
Abstract
Mutations in metabolic enzymes are associated with hereditary and sporadic forms of cancer. For example, loss-of-function mutations affecting fumarate hydratase (FH), the tricarboxylic acid (TCA) cycle enzyme, result in the accumulation of millimolar levels of fumarate that cause an aggressive form of kidney cancer. A distinct feature of fumarate is its ability to spontaneously react with thiol groups of cysteines in a chemical reaction termed succination. Although succination of a few proteins has been causally implicated in the molecular features of FH-deficient cancers, the stoichiometry, wider functional consequences, and contribution of succination to disease development remain largely unexplored. We discuss the functional implications of fumarate-induced succination in FH-deficient cells, the available methodologies, and the current challenges in studying this post-translational modification.
Collapse
Affiliation(s)
- Iva Guberovic
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Christian Frezza
- Institute for Metabolomics in Ageing, Cluster of Excellence Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany; Institute of Genetics, Faculty of Mathematics and Natural Sciences, Faculty of Medicine, University of Cologne, Cologne, Germany.
| |
Collapse
|
6
|
Percio A, Cicchinelli M, Masci D, Summo M, Urbani A, Greco V. Oxidative Cysteine Post Translational Modifications Drive the Redox Code Underlying Neurodegeneration and Amyotrophic Lateral Sclerosis. Antioxidants (Basel) 2024; 13:883. [PMID: 39199129 PMCID: PMC11351139 DOI: 10.3390/antiox13080883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 07/17/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Redox dysregulation, an imbalance between oxidants and antioxidants, is crucial in the pathogenesis of various neurodegenerative diseases. Within this context, the "redoxome" encompasses the network of redox molecules collaborating to maintain cellular redox balance and signaling. Among these, cysteine-sensitive proteins are fundamental for this homeostasis. Due to their reactive thiol groups, cysteine (Cys) residues are particularly susceptible to oxidative post-translational modifications (PTMs) induced by free radicals (reactive oxygen, nitrogen, and sulfur species) which profoundly affect protein functions. Cys-PTMs, forming what is referred to as "cysteinet" in the redox proteome, are essential for redox signaling in both physiological and pathological conditions, including neurodegeneration. Such modifications significantly influence protein misfolding and aggregation, key hallmarks of neurodegenerative diseases such as Alzheimer's, Parkinson's, and notably, amyotrophic lateral sclerosis (ALS). This review aims to explore the complex landscape of cysteine PTMs in the cellular redox environment, elucidating their impact on neurodegeneration at protein level. By investigating specific cysteine-sensitive proteins and the regulatory networks involved, particular emphasis is placed on the link between redox dysregulation and ALS, highlighting this pathology as a prime example of a neurodegenerative disease wherein such redox dysregulation is a distinct hallmark.
Collapse
Affiliation(s)
- Anna Percio
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Michela Cicchinelli
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
| | - Mariagrazia Summo
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| | - Viviana Greco
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (A.P.); (M.C.); (D.M.); (M.S.); (A.U.)
- Department of Laboratory Diagnostic and Infectious Diseases, Unity of Chemistry, Biochemistry and Clinical Molecular Biology, Fondazione Policlinico Universitario Agostino Gemelli-IRCCS, 00168 Rome, Italy
| |
Collapse
|
7
|
Chigozie AE, Ravikumar A, Yang X, Tamilselvan G, Deng Y, Arunjegan A, Li X, Hu Z, Zhang Z. A metal-phenolic coordination framework nanozyme exhibits dual enzyme mimicking activity and its application is effective for colorimetric detection of biomolecules. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:3530-3538. [PMID: 38779841 DOI: 10.1039/d4ay00689e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Biomolecules play vital roles in many biological processes and diseases, making their identification crucial. Herein, we present a colorimetric sensing method for detecting biomolecules like cysteine (Cys), homocysteine (Hcy), and glutathione (GSH). This approach is based on a reaction system whereby colorless 3,3',5,5'-tetramethylbenzidine (TMB) undergoes catalytic oxidation to form blue-colored oxidized TMB (ox-TMB) in the presence of hydrogen peroxide (H2O2), utilizing the peroxidase and catalase-mimicking activities of metal-phenolic coordination frameworks (MPNs) of Cu-TA, Co-TA, and Fe-TA nanospheres. The Fe-TA nanospheres demonstrated superior activity, more active sites and enhanced electron transport. Under optimal conditions, the Fe-TA nanospheres were used for the detection of biomolecules. When present, biomolecules inhibit the reaction between TMB and H2O2, causing various colorimetric responses at low detection limits of 0.382, 0.776 and 0.750 μM for Cys, Hcy and GSH. Furthermore, it was successfully applied to real water samples with good recovery results. The developed sensor not only offers a rapid, portable, and user-friendly technique for multi-target analysis of biomolecules at low concentrations but also expands the potential uses of MPNs for other targets in the environmental field.
Collapse
Affiliation(s)
- Aham Emmanuel Chigozie
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, Nigeria
| | - A Ravikumar
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Xiaofeng Yang
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - G Tamilselvan
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Yibin Deng
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
- Key Laboratory of Clinical Molecular Diagnosis and Research for High Incidence Diseases in Western Guangxi, Guangxi, 533000, China
| | - A Arunjegan
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Xuesong Li
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Zhang Hu
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Zhen Zhang
- School of the Environment and Safety Engineering, Jiangsu University, Zhenjiang 212013, China.
- Center for Medical Laboratory Science, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, China.
- Key Laboratory of Clinical Molecular Diagnosis and Research for High Incidence Diseases in Western Guangxi, Guangxi, 533000, China
| |
Collapse
|
8
|
Arron HE, Marsh BD, Kell DB, Khan MA, Jaeger BR, Pretorius E. Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: the biology of a neglected disease. Front Immunol 2024; 15:1386607. [PMID: 38887284 PMCID: PMC11180809 DOI: 10.3389/fimmu.2024.1386607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/11/2024] [Indexed: 06/20/2024] Open
Abstract
Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is a chronic, debilitating disease characterised by a wide range of symptoms that severely impact all aspects of life. Despite its significant prevalence, ME/CFS remains one of the most understudied and misunderstood conditions in modern medicine. ME/CFS lacks standardised diagnostic criteria owing to variations in both inclusion and exclusion criteria across different diagnostic guidelines, and furthermore, there are currently no effective treatments available. Moving beyond the traditional fragmented perspectives that have limited our understanding and management of the disease, our analysis of current information on ME/CFS represents a significant paradigm shift by synthesising the disease's multifactorial origins into a cohesive model. We discuss how ME/CFS emerges from an intricate web of genetic vulnerabilities and environmental triggers, notably viral infections, leading to a complex series of pathological responses including immune dysregulation, chronic inflammation, gut dysbiosis, and metabolic disturbances. This comprehensive model not only advances our understanding of ME/CFS's pathophysiology but also opens new avenues for research and potential therapeutic strategies. By integrating these disparate elements, our work emphasises the necessity of a holistic approach to diagnosing, researching, and treating ME/CFS, urging the scientific community to reconsider the disease's complexity and the multifaceted approach required for its study and management.
Collapse
Affiliation(s)
- Hayley E. Arron
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
| | - Benjamin D. Marsh
- MRCPCH Consultant Paediatric Neurodisability, Exeter, Devon, United Kingdom
| | - Douglas B. Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | - M. Asad Khan
- Directorate of Respiratory Medicine, Manchester University Hospitals, Wythenshawe Hospital, Manchester, United Kingdom
| | - Beate R. Jaeger
- Long COVID department, Clinic St Georg, Bad Aibling, Germany
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, South Africa
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| |
Collapse
|
9
|
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
|
10
|
Goncalves D, Duy DL, Peffer S, Morano KA. Cytoplasmic redox imbalance in the thioredoxin system activates Hsf1 and results in hyperaccumulation of the sequestrase Hsp42 with misfolded proteins. Mol Biol Cell 2024; 35:ar53. [PMID: 38381577 PMCID: PMC11064659 DOI: 10.1091/mbc.e23-07-0296] [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: 07/31/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 02/23/2024] Open
Abstract
Cells employ multiple systems to maintain homeostasis when experiencing environmental stress. For example, the folding of nascent polypeptides is exquisitely sensitive to proteotoxic stressors including heat, pH, and oxidative stress, and is safeguarded by a network of protein chaperones that concentrate potentially toxic misfolded proteins into transient assemblies to promote folding or degradation. The redox environment itself is buffered by both cytosolic and organellar thioredoxin and glutathione pathways. How these systems are linked is poorly understood. Here, we determine that specific disruption of the cytosolic thioredoxin system resulted in constitutive activation of the heat shock response in Saccharomyces cerevisiae and accumulation of the sequestrase Hsp42 into an exaggerated and persistent juxtanuclear quality control (JUNQ) compartment. Terminally misfolded proteins also accumulated in this compartment in thioredoxin reductase (TRR1)-deficient cells, despite apparently normal formation and dissolution of transient cytoplasmic quality control (CytoQ) bodies during heat shock. Notably, cells lacking TRR1 and HSP42 exhibited severe synthetic slow growth exacerbated by oxidative stress, signifying a critical role for Hsp42 under redox-challenged conditions. Finally, we demonstrated that Hsp42 localization patterns in trr1∆ cells mimic those observed in chronically aging and glucose-starved cells, linking nutrient depletion and redox imbalance with management of misfolded proteins via a process of long-term sequestration.
Collapse
Affiliation(s)
- Davi Goncalves
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX 77030
| | - Duong Long Duy
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX 77030
| | - Sara Peffer
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX 77030
- Microbiology and Infectious Disease Program, MD Anderson UTHealth Graduate School at UTHealth Houston, Houston, TX 77030
| | - Kevin A. Morano
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX 77030
| |
Collapse
|
11
|
Héja L, Simon Á, Kardos J. Simulation of gap junction formation reveals critical role of Cys disulfide redox state in connexin hemichannel docking. Cell Commun Signal 2024; 22:185. [PMID: 38500186 PMCID: PMC10949817 DOI: 10.1186/s12964-023-01439-z] [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: 08/11/2023] [Accepted: 12/12/2023] [Indexed: 03/20/2024] Open
Abstract
Video Abstract.
Collapse
Affiliation(s)
- László Héja
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary.
| | - Ágnes Simon
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary
| | - Julianna Kardos
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Magyar Tudósok Körútja 2, 1117, Budapest, Hungary
| |
Collapse
|
12
|
Mora-Zenil J, Morán J. ROS produced by NOX promote the neurite growth in a PI3K/Akt independent manner. J Neurosci Res 2024; 102:e25259. [PMID: 37840360 DOI: 10.1002/jnr.25259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 05/17/2023] [Accepted: 09/28/2023] [Indexed: 10/17/2023]
Abstract
Reactive oxygen species (ROS) function as signaling molecules in several physiologic and pathologic processes. In central nervous system, ROS are critical for differentiation, migration, polarization, and neurite growth. These actions are mediated by reversible oxidation of target proteins. On the other hand, PI3K/Akt signaling pathway is susceptible to be modulated by ROS and it has been implicated in neurite growth. In this study, we evaluated the participation of ROS in the neurite growth of cultured rat cerebellar granule neurons (CGN), as well as the possible regulation of the PI3K/Akt pathway by ROS during neurite outgrowth. For this purpose, CGN were treated with cellular or mitochondrial antioxidants, or an NOX inhibitor and neurite growth was evaluated. Moreover, to assess the participation Akt in this process, the p-Akt levels were measured in CGN treated with antioxidants or a NOX inhibitor. The effect of antioxidants on the neurite growth in the presence of a PI3K inhibitor was also measured. We found that cellular antioxidants and the NOX inhibitor decreased the neurite growth, but not the mitochondrial antioxidant. Interestingly, the antioxidants increased the p-Akt levels; however, the effect of antioxidants on neurite growth was no dependent on the Akt activity since the inhibitor of PI3K did not modify the antioxidant action on neurite growth. Our results show that the PI3K/Akt pathway participates in neurite growth and that ROS produced by NOX could function as signals in this process; however, this action is not mediated by a redox regulation of Akt activity.
Collapse
Affiliation(s)
- Janeth Mora-Zenil
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Julio Morán
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
13
|
Song B, Yang P, Zhang S. Cell fate regulation governed by p53: Friends or reversible foes in cancer therapy. Cancer Commun (Lond) 2024; 44:297-360. [PMID: 38311377 PMCID: PMC10958678 DOI: 10.1002/cac2.12520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 01/03/2024] [Accepted: 01/11/2024] [Indexed: 02/10/2024] Open
Abstract
Cancer is a leading cause of death worldwide. Targeted therapies aimed at key oncogenic driver mutations in combination with chemotherapy and radiotherapy as well as immunotherapy have benefited cancer patients considerably. Tumor protein p53 (TP53), a crucial tumor suppressor gene encoding p53, regulates numerous downstream genes and cellular phenotypes in response to various stressors. The affected genes are involved in diverse processes, including cell cycle arrest, DNA repair, cellular senescence, metabolic homeostasis, apoptosis, and autophagy. However, accumulating recent studies have continued to reveal novel and unexpected functions of p53 in governing the fate of tumors, for example, functions in ferroptosis, immunity, the tumor microenvironment and microbiome metabolism. Among the possibilities, the evolutionary plasticity of p53 is the most controversial, partially due to the dizzying array of biological functions that have been attributed to different regulatory mechanisms of p53 signaling. Nearly 40 years after its discovery, this key tumor suppressor remains somewhat enigmatic. The intricate and diverse functions of p53 in regulating cell fate during cancer treatment are only the tip of the iceberg with respect to its equally complicated structural biology, which has been painstakingly revealed. Additionally, TP53 mutation is one of the most significant genetic alterations in cancer, contributing to rapid cancer cell growth and tumor progression. Here, we summarized recent advances that implicate altered p53 in modulating the response to various cancer therapies, including chemotherapy, radiotherapy, and immunotherapy. Furthermore, we also discussed potential strategies for targeting p53 as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Bin Song
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ping Yang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
| | - Shuyu Zhang
- Laboratory of Radiation MedicineWest China Second University HospitalSichuan UniversityChengduSichuanP. R. China
- The Second Affiliated Hospital of Chengdu Medical CollegeChina National Nuclear Corporation 416 HospitalChengduSichuanP. R. China
- Laboratory of Radiation MedicineNHC Key Laboratory of Nuclear Technology Medical TransformationWest China School of Basic Medical Sciences & Forensic MedicineSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
14
|
Pérez-Sala D, Quinlan RA. The redox-responsive roles of intermediate filaments in cellular stress detection, integration and mitigation. Curr Opin Cell Biol 2024; 86:102283. [PMID: 37989035 DOI: 10.1016/j.ceb.2023.102283] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/18/2023] [Accepted: 10/29/2023] [Indexed: 11/23/2023]
Abstract
Intermediate filaments are critical for cell and tissue homeostasis and for stress responses. Cytoplasmic intermediate filaments form versatile and dynamic assemblies that interconnect cellular organelles, participate in signaling and protect cells and tissues against stress. Here we have focused on their involvement in redox signaling and oxidative stress, which arises in numerous pathophysiological situations. We pay special attention to type III intermediate filaments, mainly vimentin, because it provides a physical interface for redox signaling, stress responses and mechanosensing. Vimentin possesses a single cysteine residue that is a target for multiple oxidants and electrophiles. This conserved residue fine tunes vimentin assembly, response to oxidative stress and crosstalk with other cellular structures. Here we integrate evidence from the intermediate filament and redox biology fields to propose intermediate filaments as redox sentinel networks of the cell. To support this, we appraise how vimentin detects and orchestrates cellular responses to oxidative and electrophilic stress.
Collapse
Affiliation(s)
- Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas Margarita Salas, C.S.I.C., 28040 Madrid, Spain.
| | - Roy A Quinlan
- Department of Biosciences, University of Durham, Upper Mountjoy Science Site, Durham, United Kingdom; Biophysical Sciences Institute, University of Durham, Durham, United Kingdom; Department of Biological Structure, University of Washington, Seattle, WA, United States.
| |
Collapse
|
15
|
Perluigi M, Di Domenico F, Butterfield DA. Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol Rev 2024; 104:103-197. [PMID: 37843394 PMCID: PMC11281823 DOI: 10.1152/physrev.00030.2022] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 03/30/2023] [Accepted: 05/24/2023] [Indexed: 10/17/2023] Open
Abstract
Alzheimer disease (AD) is associated with multiple etiologies and pathological mechanisms, among which oxidative stress (OS) appears as a major determinant. Intriguingly, OS arises in various pathways regulating brain functions, and it seems to link different hypotheses and mechanisms of AD neuropathology with high fidelity. The brain is particularly vulnerable to oxidative damage, mainly because of its unique lipid composition, resulting in an amplified cascade of redox reactions that target several cellular components/functions ultimately leading to neurodegeneration. The present review highlights the "OS hypothesis of AD," including amyloid beta-peptide-associated mechanisms, the role of lipid and protein oxidation unraveled by redox proteomics, and the antioxidant strategies that have been investigated to modulate the progression of AD. Collected studies from our groups and others have contributed to unraveling the close relationships between perturbation of redox homeostasis in the brain and AD neuropathology by elucidating redox-regulated events potentially involved in both the pathogenesis and progression of AD. However, the complexity of AD pathological mechanisms requires an in-depth understanding of several major intracellular pathways affecting redox homeostasis and relevant for brain functions. This understanding is crucial to developing pharmacological strategies targeting OS-mediated toxicity that may potentially contribute to slow AD progression as well as improve the quality of life of persons with this severe dementing disorder.
Collapse
Affiliation(s)
- Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli," Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, Rome, Italy
| | - D Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States
| |
Collapse
|
16
|
Rajan S, Yoon HS. Covalent ligands of nuclear receptors. Eur J Med Chem 2023; 261:115869. [PMID: 37857142 DOI: 10.1016/j.ejmech.2023.115869] [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: 07/20/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 10/21/2023]
Abstract
Nuclear receptors (NRs) are ligand-induced transcriptional factors implicated in several physiological pathways. Naïve ligands bind to their cognate receptors and modulate gene expression as agonists or antagonists. It has been observed that some ligands bind via covalent bonding with the NR Ligand Binding Domain (LBD) residues. While many such instances have been known since the 1980s, a consolidated account of these ligands and their interactions with NR-LBD is yet to be documented. To negate this, we have culled out the human NR-LBDs that form a covalent attachment with ligands. According to the study, 16 of the 48 human NRs have been targeted by covalent ligands. It was found that conserved cysteines prone to covalent attachment are predominantly located in NR-LBD helices 3 and 11. These conserved cysteines are also observed in many of the remaining NRs, which can be probed for their reactivity. Thus, the structural insights into NR-LBD interactions with covalent ligands presented here would aid drug discovery efforts targeting NRs.
Collapse
Affiliation(s)
- Sreekanth Rajan
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore
| | - Ho Sup Yoon
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, 637551, Singapore; College of Pharmacy, CHA University, 120 Haeryong-ro, Pocheon-si, Gyeonggi-do, 11160, Republic of Korea; CHA Advanced Research Institute, 335 Pangyo-ro, Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
17
|
Yan T, Julio AR, Villanueva M, Jones AE, Ball AB, Boatner LM, Turmon AC, Nguyễn KB, Yen SL, Desai HS, Divakaruni AS, Backus KM. Proximity-labeling chemoproteomics defines the subcellular cysteinome and inflammation-responsive mitochondrial redoxome. Cell Chem Biol 2023; 30:811-827.e7. [PMID: 37419112 PMCID: PMC10510412 DOI: 10.1016/j.chembiol.2023.06.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/01/2023] [Accepted: 06/07/2023] [Indexed: 07/09/2023]
Abstract
Proteinaceous cysteines function as essential sensors of cellular redox state. Consequently, defining the cysteine redoxome is a key challenge for functional proteomic studies. While proteome-wide inventories of cysteine oxidation state are readily achieved using established, widely adopted proteomic methods such as OxICAT, Biotin Switch, and SP3-Rox, these methods typically assay bulk proteomes and therefore fail to capture protein localization-dependent oxidative modifications. Here we establish the local cysteine capture (Cys-LoC) and local cysteine oxidation (Cys-LOx) methods, which together yield compartment-specific cysteine capture and quantitation of cysteine oxidation state. Benchmarking of the Cys-LoC method across a panel of subcellular compartments revealed more than 3,500 cysteines not previously captured by whole-cell proteomic analysis. Application of the Cys-LOx method to LPS-stimulated immortalized murine bone marrow-derived macrophages (iBMDM), revealed previously unidentified, mitochondrially localized cysteine oxidative modifications upon pro-inflammatory activation, including those associated with oxidative mitochondrial metabolism.
Collapse
Affiliation(s)
- Tianyang Yan
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095, USA
| | - Ashley R Julio
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095, USA
| | - Miranda Villanueva
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA
| | - Anthony E Jones
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los A ngeles, CA 90095, USA
| | - Andréa B Ball
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los A ngeles, CA 90095, USA
| | - Lisa M Boatner
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095, USA
| | - Alexandra C Turmon
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095, USA
| | - Kaitlyn B Nguyễn
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los A ngeles, CA 90095, USA
| | - Stephanie L Yen
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Heta S Desai
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA
| | - Ajit S Divakaruni
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los A ngeles, CA 90095, USA
| | - Keriann M Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, Los Angeles, CA 90095, USA; DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
18
|
Gonçalves D, Peffer S, Morano KA. Cytoplasmic redox imbalance in the thioredoxin system activates Hsf1 and results in hyperaccumulation of the sequestrase Hsp42 with misfolded proteins. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.26.546610. [PMID: 37425817 PMCID: PMC10327208 DOI: 10.1101/2023.06.26.546610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Cells employ multiple systems to maintain homeostasis when experiencing environmental stress. For example, the folding of nascent polypeptides is exquisitely sensitive to proteotoxic stressors including heat, pH and oxidative stress, and is safeguarded by a network of protein chaperones that concentrate potentially toxic misfolded proteins into transient assemblies to promote folding or degradation. The redox environment itself is buffered by both cytosolic and organellar thioredoxin and glutathione pathways. How these systems are linked is poorly understood. Here, we determine that specific disruption of the cytosolic thioredoxin system resulted in constitutive activation of the heat shock response in Saccharomyces cerevisiae and accumulation of the sequestrase Hsp42 into an exaggerated and persistent juxtanuclear quality control (JUNQ) compartment. Terminally misfolded proteins also accumulated in this compartment in thioredoxin reductase (TRR1)-deficient cells, despite apparently normal formation and dissolution of transient cytoplasmic quality control (CytoQ) bodies during heat shock. Notably, cells lacking TRR1 and HSP42 exhibited severe synthetic slow growth exacerbated by oxidative stress, signifying a critical role for Hsp42 under redox-challenged conditions. Finally, we demonstrated that Hsp42 localization patterns in trr1∆ cells mimic those observed in chronically aging and glucose-starved cells, linking nutrient depletion and redox imbalance with management of misfolded proteins via a mechanism of long-term sequestration.
Collapse
Affiliation(s)
- Davi Gonçalves
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX USA
- Current address: Cemvita Factory, Houston, TX USA
| | - Sara Peffer
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX USA
- MD Anderson UTHealth Graduate School at UTHealth Houston, Houston, TX USA
- Current address: Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX USA
| | - Kevin A. Morano
- Department of Microbiology and Molecular Genetics, McGovern Medical School at UTHealth Houston, Houston, TX USA
| |
Collapse
|
19
|
Du W, Gong XL, Tian Y, Zhu X, Peng Y, Wang YW. Coumarin-Based Fluorescence Probe for Differentiated Detection of Biothiols and Its Bioimaging in Cells. BIOSENSORS 2023; 13:bios13040447. [PMID: 37185522 PMCID: PMC10136212 DOI: 10.3390/bios13040447] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 05/17/2023]
Abstract
In this work, a coumarin derivative, SWJT-14, was synthesized as a fluorescence probe to distinguish cysteine (Cys), homocysteine (Hcy) and glutathione (GSH) in aqueous solutions. The detection limit of Cys, Hcy and GSH for the probe was 0.02 μM, 0.42 μM and 0.92 μM, respectively, which was lower than biothiols in cells. The probe reacted with biothiols to generate different products with different conjugated structures. Additionally, it could distinguish Cys, Hcy and GSH using fluorescence and UV-Vis spectra. The detection mechanism was confirmed by MS. SWJT-14 was successfully used in cellular experiments and detected both endogenous and exogenous biothiols.
Collapse
Affiliation(s)
- Wei Du
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Xiu-Lin Gong
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Yang Tian
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Xi Zhu
- Department of Neurology, The Third People's Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu 610031, China
| | - Yu Peng
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| | - Ya-Wen Wang
- School of Life Science and Engineering, School of Chemistry, Southwest Jiaotong University, Chengdu 610031, China
| |
Collapse
|
20
|
Azemin WA, Alias N, Ali AM, Shamsir MS. In silico analysis prediction of HepTH1-5 as a potential therapeutic agent by targeting tumour suppressor protein networks. J Biomol Struct Dyn 2023; 41:1141-1167. [PMID: 34935583 DOI: 10.1080/07391102.2021.2017349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Many studies reported that the activation of tumour suppressor protein, p53 induced the human hepcidin expression. However, its expression decreased when p53 was silenced in human hepatoma cells. Contrary to Tilapia hepcidin TH1-5, HepTH1-5 was previously reported to trigger the p53 activation through the molecular docking approach. The INhibitor of Growth (ING) family members are also shown to directly interact with p53 and promote cell cycle arrest, senescence, apoptosis and participate in DNA replication and DNA damage responses to suppress the tumour initiation and progression. However, the interrelation between INGs and HepTH1-5 remains unknown. Therefore, this study aims to identify the mechanism and their protein interactions using in silico approaches. The finding revealed that HepTH1-5 and its ligands had interacted mostly on hotspot residues of ING proteins which involved in histone modifications via acetylation, phosphorylation, and methylation. This proves that HepTH1-5 might implicate in an apoptosis signalling pathway and preserve the protein structure and function of INGs by reducing the perturbation of histone binding upon oxidative stress response. This study would provide theoretical guidance for the design and experimental studies to decipher the role of HepTH1-5 as a potential therapeutic agent for cancer therapy. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Wan-Atirah Azemin
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia.,Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia
| | - Nadiawati Alias
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Abdul Manaf Ali
- Faculty of Bioresources and Food Industry, School of Agriculture Science and Biotechnology, Universiti Sultan Zainal Abidin, Besut, Malaysia
| | - Mohd Shahir Shamsir
- Faculty of Science, Bioinformatics Research Group (BIRG), Department of Biosciences, Universiti Teknologi Malaysia, Skudai, Malaysia.,Faculty of Applied Sciences and Technology, Universiti Tun Hussein Onn Malaysia, Pagoh Higher Education Hub, Muar, Malaysia
| |
Collapse
|
21
|
Yan T, Julio AR, Villanueva M, Jones AE, Ball AB, Boatner LM, Turmon AC, Yen SL, Desai HS, Divakaruni AS, Backus KM. Proximity-labeling chemoproteomics defines the subcellular cysteinome and inflammation-responsive mitochondrial redoxome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.22.525042. [PMID: 36711448 PMCID: PMC9882296 DOI: 10.1101/2023.01.22.525042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Proteinaceous cysteines function as essential sensors of cellular redox state. Consequently, defining the cysteine redoxome is a key challenge for functional proteomic studies. While proteome-wide inventories of cysteine oxidation state are readily achieved using established, widely adopted proteomic methods such as OxiCat, Biotin Switch, and SP3-Rox, they typically assay bulk proteomes and therefore fail to capture protein localization-dependent oxidative modifications. To obviate requirements for laborious biochemical fractionation, here, we develop and apply an unprecedented two step cysteine capture method to establish the Local Cysteine Capture (Cys-LoC), and Local Cysteine Oxidation (Cys-LOx) methods, which together yield compartment-specific cysteine capture and quantitation of cysteine oxidation state. Benchmarking of the Cys-LoC method across a panel of subcellular compartments revealed more than 3,500 cysteines not previously captured by whole cell proteomic analysis. Application of the Cys-LOx method to LPS stimulated murine immortalized bone marrow-derived macrophages (iBMDM), revealed previously unidentified mitochondria-specific inflammation-induced cysteine oxidative modifications including those associated with oxidative phosphorylation. These findings shed light on post-translational mechanisms regulating mitochondrial function during the cellular innate immune response.
Collapse
Affiliation(s)
- Tianyang Yan
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
| | - Ashley R. Julio
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
| | - Miranda Villanueva
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
| | - Anthony E. Jones
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Andréa B. Ball
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Lisa M. Boatner
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
| | - Alexandra C. Turmon
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
| | - Stephanie L. Yen
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Heta S. Desai
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
| | - Ajit S. Divakaruni
- Department of Molecular and Medical Pharmacology, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - Keriann M. Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, UCLA, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, 90095, USA
- DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, CA, 90095, USA
| |
Collapse
|
22
|
Lopes IS, Cassas F, Veiga TAM, de Oliveira Silva FR, Courrol LC. Synthesis and Characterization of Eugenia uniflora L. Silver Nanoparticles and L-Cysteine Sensor Application. Chem Biodivers 2023; 20:e202200787. [PMID: 36420909 DOI: 10.1002/cbdv.202200787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/21/2022] [Accepted: 11/24/2022] [Indexed: 11/25/2022]
Abstract
L-Cysteine (Cys) is a non-essential sulfur-containing amino acid, crucial for protein synthesis, detoxification, and several metabolic functions. Cys is widely used in the agricultural, food, cosmetic, and pharmaceutical industries. So, a suitable sensitive and selective sensing approach is of great interest, and a low-cost sensor would be necessary. This article presents silver nanoparticles (EuAgNPs) synthesized by a green synthesis method using Eugenia uniflora L. extracts and photoreduction. The nanoparticles were characterized by UV/VIS, transmission electron microscopy, high-performance liquid chromatography (HPLC), FTIR, and Zeta potential. With the addition of Cys in the EuAgNPs solution, the terminal thiol part of L-cysteine binds on the surface of nanoparticles through Ag-S bond. The EuAgNPs and CysAgNPs coexist until flavonoids bound the amino group of Cys, enhancing the red color of solutions. The EuAgNPs provided selectivity to detect Cys among other amino acids, and its detection limit was found to be 3.8 nM. The sensor has the advantages of low-cost synthesis, fast response, high selectivity, and sensitivity.
Collapse
Affiliation(s)
- Isabela Santos Lopes
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Física, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | - Fernando Cassas
- Programa de Pós-Graduação em Biologia Química, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | - Thiago André Moura Veiga
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Química, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| | | | - Lilia Coronato Courrol
- Instituto de Ciências Ambientais, Químicas e Farmacêuticas, Departamento de Física, Universidade Federal de São Paulo, Diadema, São Paulo, Brazil
| |
Collapse
|
23
|
Qureshi F, Hahn J. Towards the Development of a Diagnostic Test for Autism Spectrum Disorder: Big Data Meets Metabolomics. CAN J CHEM ENG 2023; 101:9-17. [PMID: 36591338 PMCID: PMC9799131 DOI: 10.1002/cjce.24594] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/06/2022] [Indexed: 01/05/2023]
Abstract
Autism spectrum disorder (ASD) is defined as a neurodevelopmental disorder which results in impairments in social communications and interactions as well as repetitive behaviors. Despite current estimates showing that approximately 2.2% of children are affected in the United States, relatively little about ASD pathophysiology is known in part due to the highly heterogenous presentation of the disorder. Given the limited knowledge into the biological mechanisms governing its etiology, the diagnosis of ASD is performed exclusively based on an individual's behavior assessed by a clinician through psychometric tools. Although there is no readily available biochemical test for ASD diagnosis, multivariate statistical methods show considerable potential for effectively leveraging multiple biochemical measurements for classification and characterization purposes. In this work, markers associated with the folate dependent one-carbon metabolism and transulfuration (FOCM/TS) pathways analyzed via both Fisher Discriminant Analysis and Support Vector Machine showed strong capability to distinguish between ASD and TD cohorts. Furthermore, using Kernel Partial Least Squares regression it was possible to assess some degree of behavioral severity from metabolomic data. While the results presented need to be replicated in independent future studies, they represent a promising avenue for uncovering clinically relevant ASD biomarkers.
Collapse
Affiliation(s)
- Fatir Qureshi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy NY 12180
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy NY 12180
| | - Juergen Hahn
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy NY 12180
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy NY 12180
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy NY 12180
| |
Collapse
|
24
|
Baroni L, Abreu-Filho PG, Pereira LM, Nagl M, Yatsuda AP. Recombinant actin-depolymerizing factor of the apicomplexan Neospora caninum (NcADF) is susceptible to oxidation. Front Cell Infect Microbiol 2022; 12:952720. [PMID: 36601306 PMCID: PMC9806845 DOI: 10.3389/fcimb.2022.952720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 09/21/2022] [Indexed: 12/24/2022] Open
Abstract
Neospora caninum is a member of Apicomplexa Phylum and the causative agent of neosporosis, a disease responsible for abortions in cattle. Apicomplexan parasites have a limited set of actin-binding proteins conducting the regulation of the dynamics of nonconventional actin. The parasite actin-based motility is implicated in the parasite invasion process in the host cell. Once no commercial strategy for the neosporosis control is available, the interference in the parasite actin function may result in novel drug targets. Actin-depolymerization factor (ADF) is a member of the ADF/cofilin family, primarily known for its function in actin severing and depolymerization. ADF/cofilins are versatile proteins modulated by different mechanisms, including reduction and oxidation. In apicomplexan parasites, the mechanisms involved in the modulation of ADF function are barely explored and the effects of oxidation in the protein are unknown so far. In this study, we used the oxidants N-chlorotaurine (NCT) and H2O2 to investigate the susceptibility of the recombinant N. caninum ADF (NcADF) to oxidation. After exposing the protein to either NCT or H2O2, the dimerization status and cysteine residue oxidation were determined. Also, the interference of NcADF oxidation in the interaction with actin was assessed. The treatment of the recombinant protein with oxidants reversibly induced the production of dimers, indicating that disulfide bonds between NcADF cysteine residues were formed. In addition, the exposure of NcADF to NCT resulted in more efficient oxidation of the cysteine residues compared to H2O2. Finally, the oxidation of NcADF by NCT reduced the ability of actin-binding and altered the function of NcADF in actin polymerization. Altogether, our results clearly show that recombinant NcADF is sensitive to redox conditions, indicating that the function of this protein in cellular processes involving actin dynamics may be modulated by oxidation.
Collapse
Affiliation(s)
- Luciana Baroni
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Péricles Gama Abreu-Filho
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Luiz Miguel Pereira
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| | - Markus Nagl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ana Patricia Yatsuda
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil,*Correspondence: Ana Patricia Yatsuda,
| |
Collapse
|
25
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
26
|
Lee J, Roh JL. SLC7A11 as a Gateway of Metabolic Perturbation and Ferroptosis Vulnerability in Cancer. Antioxidants (Basel) 2022; 11:antiox11122444. [PMID: 36552652 PMCID: PMC9774303 DOI: 10.3390/antiox11122444] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
SLC7A11 is a cell transmembrane protein composing the light chain of system xc-, transporting extracellular cystine into cells for cysteine production and GSH biosynthesis. SLC7A11 is a critical gateway for redox homeostasis by maintaining the cellular levels of GSH that counter cellular oxidative stress and suppress ferroptosis. SLC7A11 is overexpressed in various human cancers and regulates tumor development, proliferation, metastasis, microenvironment, and treatment resistance. Upregulation of SLC7A11 in cancers is needed to adapt to high oxidative stress microenvironments and maintain cellular redox homeostasis. High basal ROS levels and SLC7A11 dependences in cancer cells render them vulnerable to further oxidative stress. Therefore, cyst(e)ine depletion may be an effective new strategy for cancer treatment. However, the effectiveness of the SLC7A11 inhibitors or cyst(e)inase has been established in many preclinical studies but has not reached the stage of clinical trials for cancer patients. A better understanding of cysteine and SLC7A11 functions regulating and interacting with redox-active proteins and their substrates could be a promising strategy for cancer treatment. Therefore, this review intends to understand the role of cysteine in antioxidant and redox signaling, the regulators of cysteine bioavailability in cancer, the role of SLC7A11 linking cysteine redox signaling in cancer metabolism and targeting SLC7A11 for novel cancer therapeutics.
Collapse
Affiliation(s)
- Jaewang Lee
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam 13496, Republic of Korea
| | - Jong-Lyel Roh
- Department of Otorhinolaryngology-Head and Neck Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Republic of Korea
- Department of Biomedical Science, General Graduate School, CHA University, Seongnam 13496, Republic of Korea
- Correspondence: ; Tel.: +82-31-780-2988
| |
Collapse
|
27
|
Abstract
The term "scavengome" refers to the chemical space of all the metabolites that may be formed from an antioxidant upon scavenging reactive oxygen or nitrogen species (ROS/RNS). This chemical space covers a wide variety of free radical metabolites with drug discovery potential. It is very rich in structures representing an increased chemical complexity as compared to the parent antioxidant: a wide range of unusual heterocyclic structures, new CC bonds, etc. may be formed. Further, in a biological environment, this increased chemical complexity is directly translated from the localized conditions of oxidative stress that determines the amounts and types of ROS/RNS present. Biomimetic oxidative chemistry provides an excellent tool to model chemical reactions between antioxidants and ROS/RNS. In this chapter, we provide an overview on the known metabolites obtained by biomimetic oxidation of a few selected natural antioxidants, i.e., a stilbene (resveratrol), a pair of hydroxycinnamates (caffeic acid and methyl caffeate), and a flavonol (quercetin), and discuss the drug discovery perspectives of the related chemical space.
Collapse
Affiliation(s)
- Attila Hunyadi
- Institute of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary; Interdisciplinary Centre for Natural Products, University of Szeged, Szeged, Hungary.
| | - Orinhamhe G Agbadua
- Institute of Pharmacognosy, Interdisciplinary Excellence Centre, University of Szeged, Szeged, Hungary
| | - Gábor Takács
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Budapest, Hungary; Mcule.com Ltd., Budapest, Hungary
| | - Gyorgy T Balogh
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Budapest, Hungary; Department of Pharmacodynamics and Biopharmacy, University of Szeged, Szeged, Hungary
| |
Collapse
|
28
|
Micovic K, Satkunarajah T, Carnet A, Hurst M, Viirre R, Olson MF. Synthesis and Use of the Bifunctional Sulfenic Acid Probe BCN-E-BCN for In Vitro and Cell-Based Assays of Protein Oxidation. Curr Protoc 2022; 2:e559. [PMID: 36200822 PMCID: PMC11648817 DOI: 10.1002/cpz1.559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The reversible oxidation of cysteine thiol groups to sulfenic acid by reactive oxygen species (ROS) such as hydrogen peroxide can impact protein function, activity, and localization. As a consequence, ROS have profound effects on cell functions including proliferation, differentiation, and survival. Furthermore, there are clear associations between the effects of ROS on cells and the etiology of several diseases including cancer and neurodegeneration. In spite of the importance of cysteine sulfenylation as a validated post-translational modification, its labile nature impedes efficient and reproducible detection of proteins with cysteine sulfenic acid residues. To overcome this challenge, we developed a novel cell-permeable bifunctional reagent, consisting of two linked bicyclo[6.1.0]nonyne (BCN) moieties coupled with a short ethylenediamine-derived linker (BCN-E-BCN) that enables the detection of sulfenylated proteins in vitro and in intact cells. The two symmetrical BCN groups allow protein sulfenic acids to be selectively tagged with a BCN at one end while allowing for copper-free click chemistry with azide-tagged reagents of the opposite BCN. In this protocol, the synthesis of BCN-E-BCN and its use to detect cysteine sulfenic acids will be detailed. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Copper-mediated cyclopropanation of 1,5-cyclooctadiene Basic Protocol 2: Synthesis of endo- and exo-bicyclononyne Basic Protocol 3: Synthesis of endo-BCN-E-BCN Basic Protocol 4: BCN-E-BCN treatment of wild-type and cysteine-deficient mutant recombinant cofilin protein Basic Protocol 5: BCN-E-BCN labeling in live cells Basic Protocol 6: Western blotting and visualization of BCN-E-BCN-labeled samples.
Collapse
Affiliation(s)
- Katarina Micovic
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Thershan Satkunarajah
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Alexandre Carnet
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Mackenzie Hurst
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Russell Viirre
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| | - Michael F. Olson
- Department of Chemistry and BiologyToronto Metropolitan UniversityTorontoOntarioCanada
| |
Collapse
|
29
|
Smythers AL, Bhatnagar N, Ha C, Majumdar P, McConnell EW, Mohanasundaram B, Hicks LM, Pandey S. Abscisic acid-controlled redox proteome of Arabidopsis and its regulation by heterotrimeric Gβ protein. THE NEW PHYTOLOGIST 2022; 236:447-463. [PMID: 35766993 DOI: 10.1111/nph.18348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/18/2022] [Indexed: 06/15/2023]
Abstract
The plant hormone abscisic acid (ABA) plays crucial roles in regulation of stress responses and growth modulation. Heterotrimeric G-proteins are key mediators of ABA responses. Both ABA and G-proteins have also been implicated in intracellular redox regulation; however, the extent to which reversible protein oxidation manipulates ABA and/or G-protein signaling remains uncharacterized. To probe the role of reversible protein oxidation in plant stress response and its dependence on G-proteins, we determined the ABA-dependent reversible redoxome of wild-type and Gβ-protein null mutant agb1 of Arabidopsis. We quantified 6891 uniquely oxidized cysteine-containing peptides, 923 of which show significant changes in oxidation following ABA treatment. The majority of these changes required the presence of G-proteins. Divergent pathways including primary metabolism, reactive oxygen species response, translation and photosynthesis exhibited both ABA- and G-protein-dependent redox changes, many of which occurred on proteins not previously linked to them. We report the most comprehensive ABA-dependent plant redoxome and uncover a complex network of reversible oxidations that allow ABA and G-proteins to rapidly adjust cellular signaling to adapt to changing environments. Physiological validation of a subset of these observations suggests that functional G-proteins are required to maintain intracellular redox homeostasis and fully execute plant stress responses.
Collapse
Affiliation(s)
- Amanda L Smythers
- The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Chien Ha
- Donald Danforth Plant Science Center, St Louis, MO, 63132, USA
| | | | - Evan W McConnell
- The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | | | - Leslie M Hicks
- The University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Sona Pandey
- Donald Danforth Plant Science Center, St Louis, MO, 63132, USA
| |
Collapse
|
30
|
Janciauskiene S, Tumpara S, Schebb NH, Buettner FFR, Mainka M, Sivaraman K, Immenschuh S, Grau V, Welte T, Olejnicka B. Indirect effect of alpha-1-antitrypsin on endotoxin-induced IL-1β secretion from human PBMCs. Front Pharmacol 2022; 13:995869. [PMID: 36249781 PMCID: PMC9564231 DOI: 10.3389/fphar.2022.995869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 09/13/2022] [Indexed: 11/16/2022] Open
Abstract
Human alpha-1-antitrypsin (AAT) encoded by the SERPINA1 gene, is an acute phase glycoprotein that regulates inflammatory responses via both protease inhibitory and non-inhibitory activities. We previously reported that AAT controls ATP-induced IL-1β release from human mononuclear cells by stimulating the release of small bioactive molecules. In the current study, we aimed to elucidate the identity of these putative effectors released from human PBMCs in response to AAT, which may inhibit the LPS-induced release of IL-1β. We pre-incubated human PBMCs alone or with different preparations of AAT (4 mg/ml) for 30 min at 37°C, 5% CO2, and collected cell supernatants filtered through centrifugal filters (cutoff 3 kDa) to eliminate AAT and other high molecular weight substances. Supernatants passed through the filters were used to culture PBMCs isolated from the autologous or a heterologous donors with or without adding LPS (1 μg/ml) for 6 h. Unexpectedly, supernatants from PBMCs pre-incubated with AAT (Zemaira®), but not with other AAT preparations tested or with oxidized AAT (Zemaira®), lowered the LPS-induced release of IL-1β by about 25%–60% without affecting IL1B mRNA. The reversed-phase liquid chromatography coupled with mass spectrometry did not confirm the hypothesis that small pro-resolving lipid mediators released from PBMCs after exposure to AAT (Zemaira®) are responsible for lowering the LPS-induced IL-1β release. Distinctively from other AAT preparations, AAT (Zemaira®) and supernatants from PBMCs pre-treated with this protein contained high levels of total thiols. In line, mass spectrometry analysis revealed that AAT (Zemaira®) protein contains freer Cys232 than AAT (Prolastin®). Our data show that a free Cys232 in AAT is required for controlling LPS-induced IL-1β release from human PBMCs. Further studies characterizing AAT preparations used to treat patients with inherited AAT deficiency remains of clinical importance.
Collapse
Affiliation(s)
- Sabina Janciauskiene
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Department of Experimental Medicine, Lund University, Lund, Sweden
- *Correspondence: Sabina Janciauskiene,
| | - Srinu Tumpara
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Falk F. R. Buettner
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Kokilavani Sivaraman
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany
| | - Veronika Grau
- Laboratory of Experimental Surgery, Department of General and Thoracic Surgery, Justus-Liebig-University Giessen, German Center for Lung Research, Giessen, Germany
| | - Tobias Welte
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
| | - Beata Olejnicka
- Department of Respiratory Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover, Germany
- Department of Experimental Medicine, Lund University, Lund, Sweden
| |
Collapse
|
31
|
Mazmanian K, Chen T, Sargsyan K, Lim C. From quantum-derived principles underlying cysteine reactivity to combating the COVID-19 pandemic. WILEY INTERDISCIPLINARY REVIEWS. COMPUTATIONAL MOLECULAR SCIENCE 2022; 12:e1607. [PMID: 35600063 PMCID: PMC9111396 DOI: 10.1002/wcms.1607] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/31/2022] [Accepted: 02/13/2022] [Indexed: 12/20/2022]
Abstract
The COVID-19 pandemic poses a challenge in coming up with quick and effective means to counter its cause, the SARS-CoV-2. Here, we show how the key factors governing cysteine reactivity in proteins derived from combined quantum mechanical/continuum calculations led to a novel multi-targeting strategy against SARS-CoV-2, in contrast to developing potent drugs/vaccines against a single viral target such as the spike protein. Specifically, they led to the discovery of reactive cysteines in evolutionary conserved Zn2+-sites in several SARS-CoV-2 proteins that are crucial for viral polypeptide proteolysis as well as viral RNA synthesis, proofreading, and modification. These conserved, reactive cysteines, both free and Zn2+-bound, can be targeted using the same Zn-ejector drug (disulfiram/ebselen), which enables the use of broad-spectrum anti-virals that would otherwise be removed by the virus's proofreading mechanism. Our strategy of targeting multiple, conserved viral proteins that operate at different stages of the virus life cycle using a Zn-ejector drug combined with other broad-spectrum anti-viral drug(s) could enhance the barrier to drug resistance and antiviral effects, as compared to each drug alone. Since these functionally important nonstructural proteins containing reactive cysteines are highly conserved among coronaviruses, our proposed strategy has the potential to tackle future coronaviruses. This article is categorized under:Structure and Mechanism > Reaction Mechanisms and CatalysisStructure and Mechanism > Computational Biochemistry and BiophysicsElectronic Structure Theory > Density Functional Theory.
Collapse
Affiliation(s)
| | - Ting Chen
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - Karen Sargsyan
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
| | - Carmay Lim
- Institute of Biomedical Sciences Academia Sinica Taipei Taiwan
- Department of Chemistry National Tsing Hua University Hsinchu Taiwan
| |
Collapse
|
32
|
Chiang FF, Chao TH, Huang SC, Cheng CH, Tseng YY, Huang YC. Cysteine Regulates Oxidative Stress and Glutathione-Related Antioxidative Capacity before and after Colorectal Tumor Resection. Int J Mol Sci 2022; 23:ijms23179581. [PMID: 36076975 PMCID: PMC9455234 DOI: 10.3390/ijms23179581] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 11/16/2022] Open
Abstract
Cysteine might scavenge free radicals and is a limiting substrate for the cellular synthesis of glutathione (GSH). We investigated the association of cysteine with oxidative stress and GSH-related antioxidant capacity in colorectal cancer (CRC) patients. Plasma samples were drawn from 66 patients 1 day before (pre-resection) and 4 weeks after resection (post-resection). Tumor and adjacent normal tissues were collected. We measured levels of plasma and tissue cysteine, homocysteine, oxidative stress indicators (malondialdehyde, MDA; advanced oxidation protein products, AOPP), GSH, and antioxidant enzyme activities. After tumor resection, patients had significantly higher levels of plasma cysteine, homocysteine, MDA, AOPP, and GSH-related antioxidant enzyme activities when compared with pre-resection. Levels of cysteine, homocysteine, AOPP and all antioxidant capacity indicators in tumor tissue were significantly higher than those levels in the adjacent normal tissue. Plasma cysteine levels measured at pre-resection were positively associated with MDA levels in the tumor and in the adjacent normal tissues. Cysteine levels in tumor and adjacent normal tissues were significantly associated with tissue levels of homocysteine, almost as indicators of oxidative stress and antioxidant capacities. Cysteine in the circulation was likely utilized to mediate GSH-related antioxidant capacity and further cope with increased oxidative stress in tumor and adjacent normal tissues.
Collapse
Affiliation(s)
- Feng-Fan Chiang
- Division of Colorectal Surgery, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
- Department of Food and Nutrition, Providence University, Taichung 43301, Taiwan
| | - Te-Hsin Chao
- Chiayi & Wanqiao Branch, Taichung Veterans General Hospital, Chiayi 60090, Taiwan
| | - Shih-Chien Huang
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chien-Hsiang Cheng
- Department of Respiratory Therapy, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Yu-Yao Tseng
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yi-Chia Huang
- Department of Nutrition, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Nutrition, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence:
| |
Collapse
|
33
|
Ning Q, Li J. DLF-Sul: a multi-module deep learning framework for prediction of S-sulfinylation sites in proteins. Brief Bioinform 2022; 23:6658856. [PMID: 35945138 DOI: 10.1093/bib/bbac323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/16/2022] [Accepted: 07/18/2022] [Indexed: 11/14/2022] Open
Abstract
Protein S-sulfinylation is an important posttranslational modification that regulates a variety of cell and protein functions. This modification has been linked to signal transduction, redox homeostasis and neuronal transmission in studies. Therefore, identification of S-sulfinylation sites is crucial to understanding its structure and function, which is critical in cell biology and human diseases. In this study, we propose a multi-module deep learning framework named DLF-Sul for identification of S-sulfinylation sites in proteins. First, three types of features are extracted including binary encoding, BLOSUM62 and amino acid index. Then, sequential features are further extracted based on these three types of features using bidirectional long short-term memory network. Next, multi-head self-attention mechanism is utilized to filter the effective attribute information, and residual connection helps to reduce information loss. Furthermore, convolutional neural network is employed to extract local deep features information. Finally, fully connected layers acts as classifier that map samples to corresponding label. Performance metrics on independent test set, including sensitivity, specificity, accuracy, Matthews correlation coefficient and area under curve, reach 91.80%, 92.36%, 92.08%, 0.8416 and 96.40%, respectively. The results show that DLF-Sul is an effective tool for predicting S-sulfinylation sites. The source code is available on the website https://github.com/ningq669/DLF-Sul.
Collapse
Affiliation(s)
- Qiao Ning
- Information Science and Technology College, Dalian Maritime University, Dalian 116026, China
| | - Jinmou Li
- Information Science and Technology College, Dalian Maritime University, Dalian 116026, China
| |
Collapse
|
34
|
Apeland T, Ushakova A, Mansoor MA, Furriol J, Jonsson G, Marti HP. Association of redox and inflammation-related biomarkers with prognosis in IgA nephropathy: A prospective observational study. Free Radic Biol Med 2022; 188:62-70. [PMID: 35716825 DOI: 10.1016/j.freeradbiomed.2022.06.224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/23/2022] [Accepted: 06/12/2022] [Indexed: 12/01/2022]
Abstract
BACKGROUND IgA nephropathy (IGAN) has a variable prognosis. Risk stratification tools are usually based on clinical parameters combined with histologic Oxford-MEST-C score. Circulating redox- and inflammation-related biomarkers may be related to histological changes in IGAN. Therefore, we studied the performance of these biomarkers in predicting the rate of GFR-loss in IGAN. METHODS This was an observational prospective study. Fifty-seven stable patients with IGAN were examined at baseline and after a mean observational time of 5.9 ± 1.1 years. The main outcome measure was eGFR-loss per year with predefined groups, stable (<1.5 ml/min/1,73 m2/year, intermediate (between 1.5 and 2.5), and progressive (>2.5). RESULTS Fifteen patients were in the progressive, 11 in the intermediate, and 31 in the stable groups. Positive relationships were detected between eGFR-loss per year and baseline nitrate, oxidized free cysteine, parathyroid hormone, APRIL, TNFR1, CD30, chitinase 3, and LIF-5. The progressive group had elevated concentrations of these markers plus AOPP and osteopontin. Through ROC analysis, it was observed that AOPP, oxidized free cysteine, TNFR1, osteopontin, and LIF-5 had the best ability to identify progressive vs. non-progressive diseases. The combination of urinary albumin/creatinine ratio with AOPP and TNFR1 significantly improved the ability to identify progressive eGFR decline with ROC AUC 95% (adjusted 85%). CONCLUSIONS We found prognostic biomarkers related to the rate of eGFR-loss in IGAN. These biomarkers may help identify patients at risk of progressive disease. AOPP, oxidized free cysteine, TNFR1, and osteopontin are promising prognostic biomarkers in IGAN, however, further validation studies are needed.
Collapse
Affiliation(s)
- Terje Apeland
- Department of Medicine, Stavanger University Hospital, Stavanger, Norway.
| | - Anastasia Ushakova
- Department of Research, Stavanger University Hospital, Stavanger, Norway
| | - Mohammad A Mansoor
- Department of Natural Sciences, University of Agder, Kristiansand, Norway
| | - Jessica Furriol
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Grete Jonsson
- Department of Medical Biochemistry, Stavanger University Hospital, Stavanger, Norway
| | - Hans-Peter Marti
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
35
|
Benske TM, Mu TW, Wang YJ. Protein quality control of N-methyl-D-aspartate receptors. Front Cell Neurosci 2022; 16:907560. [PMID: 35936491 PMCID: PMC9352929 DOI: 10.3389/fncel.2022.907560] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/29/2022] [Indexed: 12/23/2022] Open
Abstract
N-methyl-D-aspartate receptors (NMDARs) are glutamate-gated cation channels that mediate excitatory neurotransmission and are critical for synaptic development and plasticity in the mammalian central nervous system (CNS). Functional NMDARs typically form via the heterotetrameric assembly of GluN1 and GluN2 subunits. Variants within GRIN genes are implicated in various neurodevelopmental and neuropsychiatric disorders. Due to the significance of NMDAR subunit composition for regional and developmental signaling at synapses, properly folded receptors must reach the plasma membrane for their function. This review focuses on the protein quality control of NMDARs. Specifically, we review the quality control mechanisms that ensure receptors are correctly folded and assembled within the endoplasmic reticulum (ER) and trafficked to the plasma membrane. Further, we discuss disease-associated variants that have shown disrupted NMDAR surface expression and function. Finally, we discuss potential targeted pharmacological and therapeutic approaches to ameliorate disease phenotypes by enhancing the expression and surface trafficking of subunits harboring disease-associated variants, thereby increasing their incorporation into functional receptors.
Collapse
Affiliation(s)
| | - Ting-Wei Mu
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Ya-Juan Wang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH, United States
| |
Collapse
|
36
|
Medlock AE, Dailey HA. New Avenues of Heme Synthesis Regulation. Int J Mol Sci 2022; 23:ijms23137467. [PMID: 35806474 PMCID: PMC9267699 DOI: 10.3390/ijms23137467] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/30/2022] [Accepted: 07/02/2022] [Indexed: 02/04/2023] Open
Abstract
During erythropoiesis, there is an enormous demand for the synthesis of the essential cofactor of hemoglobin, heme. Heme is synthesized de novo via an eight enzyme-catalyzed pathway within each developing erythroid cell. A large body of data exists to explain the transcriptional regulation of the heme biosynthesis enzymes, but until recently much less was known about alternate forms of regulation that would allow the massive production of heme without depleting cellular metabolites. Herein, we review new studies focused on the regulation of heme synthesis via carbon flux for porphyrin synthesis to post-translations modifications (PTMs) that regulate individual enzymes. These PTMs include cofactor regulation, phosphorylation, succinylation, and glutathionylation. Additionally discussed is the role of the immunometabolite itaconate and its connection to heme synthesis and the anemia of chronic disease. These recent studies provide new avenues to regulate heme synthesis for the treatment of diseases including anemias and porphyrias.
Collapse
Affiliation(s)
- Amy E. Medlock
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Augusta University/University of Georgia Medical Partnership, University of Georgia, Athens, GA 30602, USA
- Correspondence: (A.E.M.); (H.A.D.)
| | - Harry A. Dailey
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Department of Microbiology, University of Georgia, Athens, GA 30602, USA
- Correspondence: (A.E.M.); (H.A.D.)
| |
Collapse
|
37
|
Obi CD, Bhuiyan T, Dailey HA, Medlock AE. Ferrochelatase: Mapping the Intersection of Iron and Porphyrin Metabolism in the Mitochondria. Front Cell Dev Biol 2022; 10:894591. [PMID: 35646904 PMCID: PMC9133952 DOI: 10.3389/fcell.2022.894591] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/14/2022] [Indexed: 12/29/2022] Open
Abstract
Porphyrin and iron are ubiquitous and essential for sustaining life in virtually all living organisms. Unlike iron, which exists in many forms, porphyrin macrocycles are mostly functional as metal complexes. The iron-containing porphyrin, heme, serves as a prosthetic group in a wide array of metabolic pathways; including respiratory cytochromes, hemoglobin, cytochrome P450s, catalases, and other hemoproteins. Despite playing crucial roles in many biological processes, heme, iron, and porphyrin intermediates are potentially cytotoxic. Thus, the intersection of porphyrin and iron metabolism at heme synthesis, and intracellular trafficking of heme and its porphyrin precursors are tightly regulated processes. In this review, we discuss recent advances in understanding the physiological dynamics of eukaryotic ferrochelatase, a mitochondrially localized metalloenzyme. Ferrochelatase catalyzes the terminal step of heme biosynthesis, the insertion of ferrous iron into protoporphyrin IX to produce heme. In most eukaryotes, except plants, ferrochelatase is localized to the mitochondrial matrix, where substrates are delivered and heme is synthesized for trafficking to multiple cellular locales. Herein, we delve into the structural and functional features of ferrochelatase, as well as its metabolic regulation in the mitochondria. We discuss the regulation of ferrochelatase via post-translational modifications, transportation of substrates and product across the mitochondrial membrane, protein-protein interactions, inhibition by small-molecule inhibitors, and ferrochelatase in protozoal parasites. Overall, this review presents insight on mitochondrial heme homeostasis from the perspective of ferrochelatase.
Collapse
Affiliation(s)
- Chibuike David Obi
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
| | - Tawhid Bhuiyan
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
| | - Harry A. Dailey
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
- Department of Microbiology, University of Georgia, Athens, GA, United States
| | - Amy E. Medlock
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA, United States
- Augusta University/University of Georgia Medical Partnership, University of Georgia, Athens, GA, United States
| |
Collapse
|
38
|
Matsuo Y. Introducing Thioredoxin-Related Transmembrane Proteins: Emerging Roles of Human TMX and Clinical Implications. Antioxid Redox Signal 2022; 36:984-1000. [PMID: 34465218 PMCID: PMC9127828 DOI: 10.1089/ars.2021.0187] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: The presence of a large number of thioredoxin superfamily members suggests a complex mechanism of redox-based regulation in mammalian cells. However, whether these members are functionally redundant or play separate and distinct roles in each cellular compartment remains to be elucidated. Recent Advances: In the mammalian endoplasmic reticulum (ER), ∼20 thioredoxin-like proteins have been identified. Most ER oxidoreductases are soluble proteins located in the luminal compartment, whereas a small family of five thioredoxin-related transmembrane proteins (TMX) also reside in the ER membrane and play crucial roles with specialized functions. Critical Issues: In addition to the predicted function of ER protein quality control, several independent studies have suggested the diverse roles of TMX family proteins in the regulation of cellular processes, including calcium homeostasis, bioenergetics, and thiol-disulfide exchange in the extracellular space. Moreover, recent studies have provided evidence of their involvement in the pathogenesis of various diseases. Future Directions: Extensive research is required to unravel the physiological roles of TMX family proteins. Given that membrane-associated proteins are prime targets for drug discovery in a variety of human diseases, expanding our knowledge on the mechanistic details of TMX action on the cell membrane will provide the molecular basis for developing novel diagnostic and therapeutic approaches as a potent molecular target in a clinical setting. Antioxid. Redox Signal. 36, 984-1000.
Collapse
Affiliation(s)
- Yoshiyuki Matsuo
- Department of Human Stress Response Science, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka, Japan
| |
Collapse
|
39
|
Desai HS, Yan T, Yu F, Sun AW, Villanueva M, Nesvizhskii AI, Backus KM. SP3-Enabled Rapid and High Coverage Chemoproteomic Identification of Cell-State-Dependent Redox-Sensitive Cysteines. Mol Cell Proteomics 2022; 21:100218. [PMID: 35219905 PMCID: PMC9010637 DOI: 10.1016/j.mcpro.2022.100218] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 02/17/2022] [Accepted: 02/22/2022] [Indexed: 02/07/2023] Open
Abstract
Proteinaceous cysteine residues act as privileged sensors of oxidative stress. As reactive oxygen and nitrogen species have been implicated in numerous pathophysiological processes, deciphering which cysteines are sensitive to oxidative modification and the specific nature of these modifications is essential to understanding protein and cellular function in health and disease. While established mass spectrometry-based proteomic platforms have improved our understanding of the redox proteome, the widespread adoption of these methods is often hindered by complex sample preparation workflows, prohibitive cost of isotopic labeling reagents, and requirements for custom data analysis workflows. Here, we present the SP3-Rox redox proteomics method that combines tailored low cost isotopically labeled capture reagents with SP3 sample cleanup to achieve high throughput and high coverage proteome-wide identification of redox-sensitive cysteines. By implementing a customized workflow in the free FragPipe computational pipeline, we achieve accurate MS1-based quantitation, including for peptides containing multiple cysteine residues. Application of the SP3-Rox method to cellular proteomes identified cysteines sensitive to the oxidative stressor GSNO and cysteine oxidation state changes that occur during T cell activation. High-coverage Cys oxidation state quantification using custom isotopic probes. FragPipe-IonQuant accurately quantifies Cys labeling comparably to Skyline. PTMProphet enables site-of-labeling localization for multi-Cys–containing peptides. SP3-Rox identifies changes in Cys oxidation during T cell activation.
Collapse
Affiliation(s)
- Heta S Desai
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, California, USA; Molecular Biology Institute, UCLA, Los Angeles, California, USA
| | - Tianyang Yan
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, California, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, California, USA
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Alexander W Sun
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Miranda Villanueva
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, California, USA; Molecular Biology Institute, UCLA, Los Angeles, California, USA
| | - Alexey I Nesvizhskii
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan, USA; Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Keriann M Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA, Los Angeles, California, USA; Department of Chemistry and Biochemistry, UCLA, Los Angeles, California, USA; Molecular Biology Institute, UCLA, Los Angeles, California, USA; DOE Institute for Genomics and Proteomics, UCLA, Los Angeles, California, USA; Jonsson Comprehensive Cancer Center, UCLA, Los Angeles, California, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA, Los Angeles, California, USA.
| |
Collapse
|
40
|
Cartee NMP, Lee SJ, Young KZ, Zhang X, Wang MM. Trans-Reduction of Cerebral Small Vessel Disease Proteins by Notch-Derived EGF-like Sequences. Int J Mol Sci 2022; 23:ijms23073671. [PMID: 35409031 PMCID: PMC9115637 DOI: 10.3390/ijms23073671] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/23/2022] [Accepted: 02/28/2022] [Indexed: 02/05/2023] Open
Abstract
Cysteine oxidation states of extracellular proteins participate in functional regulation and in disease pathophysiology. In the most common inherited dementia, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), mutations in NOTCH3 that alter extracellular cysteine number have implicated NOTCH3 cysteine states as potential triggers of cerebral vascular smooth muscle cytopathology. In this report, we describe a novel property of the second EGF-like domain of NOTCH3: its capacity to alter the cysteine redox state of the NOTCH3 ectodomain. Synthetic peptides corresponding to this sequence (NOTCH3 N-terminal fragment 2, NTF2) readily reduce NOTCH3 N-terminal ectodomain polypeptides in a dose- and time-dependent fashion. Furthermore, NTF2 preferentially reduces regional domains of NOTCH3 with the highest intensity against EGF-like domains 12–15. This process requires cysteine residues of NTF2 and is also capable of targeting selected extracellular proteins that include TSP2 and CTSH. CADASIL mutations in NOTCH3 increase susceptibility to NTF2-facilitated reduction and to trans-reduction by NOTCH3 produced in cells. Moreover, NTF2 forms complexes with the NOTCH3 ectodomain, and cleaved NOTCH3 co-localizes with the NOTCH3 ectodomain in cerebral arteries of CADASIL patients. The potential for NTF2 to reduce vascular proteins and the enhanced preference for it to trans-reduce mutant NOTCH3 implicate a role for protein trans-reduction in cerebrovascular pathological states such as CADASIL.
Collapse
Affiliation(s)
- Naw May Pearl Cartee
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; (N.M.P.C.); (S.J.L.); (K.Z.Y.); (X.Z.)
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Soo Jung Lee
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; (N.M.P.C.); (S.J.L.); (K.Z.Y.); (X.Z.)
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Kelly Z. Young
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; (N.M.P.C.); (S.J.L.); (K.Z.Y.); (X.Z.)
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Xiaojie Zhang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; (N.M.P.C.); (S.J.L.); (K.Z.Y.); (X.Z.)
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
| | - Michael M. Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA; (N.M.P.C.); (S.J.L.); (K.Z.Y.); (X.Z.)
- Neurology Service, VA Ann Arbor Healthcare System, Ann Arbor, MI 48105, USA
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
- Correspondence: ; Tel.: +1-734-936-9075; Fax: +1-734-936-8813
| |
Collapse
|
41
|
Héja L, Simon Á, Szabó Z, Kardos J. Connexons Coupling to Gap Junction Channel: Potential Role for Extracellular Protein Stabilization Centers. Biomolecules 2021; 12:biom12010049. [PMID: 35053197 PMCID: PMC8773650 DOI: 10.3390/biom12010049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/21/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022] Open
Abstract
Connexin (Cx) proteins establish intercellular gap junction channels (Cx GJCs) through coupling of two apposed hexameric Cx hemichannels (Cx HCs, connexons). Pre- and post-GJ interfaces consist of extracellular EL1 and EL2 loops, each with three conserved cysteines. Previously, we reported that known peptide inhibitors, mimicking a variety of Cx43 sequences, appear non-selective when binding to homomeric Cx43 vs. Cx36 GJC homology model subtypes. In pursuit of finding potentially Cx subtype-specific inhibitors of connexon-connexon coupling, we aimed at to understand better how the GJ interface is formed. Here we report on the discovery of Cx GJC subtype-specific protein stabilization centers (SCs) featuring GJ interface architecture. First, the Cx43 GJC homology model, embedded in two opposed membrane bilayers, has been devised. Next, we endorsed the fluctuation dynamics of SCs of the interface domain of Cx43 GJC by applying standard molecular dynamics under open and closed cystine disulfide bond (CS-SC) preconditions. The simulations confirmed the major role of the unique trans-GJ SC pattern comprising conserved (55N, 56T) and non-conserved (57Q) residues of the apposed EL1 loops in the stabilization of the GJC complex. Importantly, clusters of SC patterns residing close to the GJ interface domain appear to orient the interface formation via the numerous SCs between EL1 and EL2. These include central 54CS-S198C or 61CS-S192C contacts with residues 53R, 54C, 55N, 197D, 199F or 64V, 191P, respectively. In addition, we revealed that GJC interface formation is favoured when the psi dihedral angle of the nearby 193P residue is stable around 180° and the interface SCs disappear when this angle moves to the 0° to −45° range. The potential of the association of non-conserved residues with SC motifs in connexon-connexon coupling makes the development of Cx subtype-specific inhibitors viable.
Collapse
|
42
|
Liu Y, Li L, Timani K, White C, He JJ. Tip110 Expression Facilitates the Release of HEXIM1 and pTEFb from the 7SK Ribonucleoprotein Complex Involving Regulation of the Intracellular Redox Level. Aging Dis 2021; 12:2113-2124. [PMID: 34881089 PMCID: PMC8612609 DOI: 10.14336/ad.2021.0528] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 05/28/2021] [Indexed: 11/18/2022] Open
Abstract
HIV-1 Tat-interacting protein of 110 kDa (Tip110; p110nrb/SART3) has been identified to be important for HIV gene transcription and several host gene expression. In this study, we showed that Tip110 was present in the 7SK snRNP through direct binding to MEPCE, a component of the 7SK snRNP complex. In addition, we found a positive association between Tip110 expression, change of HEXIM1 from dimer/oligomer to monomer, and release of HEXIM1 and P-TEFb from the 7SK snRNP complex. A similar association was also noted specifically in nuclear matrix as well as in chromatin where the free HEXIM1 and 7SK snRNP-bound HEXIM1 are located. Moreover, we demonstrated that Tip110 expression was linked to the glutathione metabolic pathway and the intracellular redox level, which in turn regulated HEXIM1 dimerization/oligomerization. Lastly, we performed the FRET microscopic analysis and confirmed the direct relationship between Tip110 expression and HEXIM1 dimerization/oligomerization in vivo. Taken together, these results identified a new mechanism governing HEXIM1 dimerization/oligomerization and the release of HEXIM1 and P-TEFb from the 7SK snRNP complex. These results also yield new insights to the roles of Tip110 in HIV gene transcription and replication.
Collapse
Affiliation(s)
- Ying Liu
- 1Department of Microbiology and Immunology.,2Center for Cancer Cell Biology, Immunology and Infection, and
| | - Lu Li
- 1Department of Microbiology and Immunology.,2Center for Cancer Cell Biology, Immunology and Infection, and
| | - Khalid Timani
- 1Department of Microbiology and Immunology.,2Center for Cancer Cell Biology, Immunology and Infection, and
| | - Carl White
- 2Center for Cancer Cell Biology, Immunology and Infection, and.,3Department of Physiology and Biophysics, Rosalind Franklin University of Medicine and Science Chicago Medical School, 3333 Green Bay Road, North Chicago, IL 60064, USA
| | - Johnny J He
- 1Department of Microbiology and Immunology.,2Center for Cancer Cell Biology, Immunology and Infection, and
| |
Collapse
|
43
|
Yang QQ, Ji N, Zhan Y, Tian QQ, Cai ZD, Lu XL, He W. Rational design of a new near-infrared fluorophore and apply to the detection and imaging study of cysteine and thiophenol. Anal Chim Acta 2021; 1186:339116. [PMID: 34756262 DOI: 10.1016/j.aca.2021.339116] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 09/18/2021] [Accepted: 09/24/2021] [Indexed: 12/23/2022]
Abstract
The development of a near-infrared fluorophore with excellent fluorescence performance, a large Stokes shift, and good biocompatibility has become a focus in the field of fluorescence imaging in recent years. Based on quantum chemistry calculations and reasonable molecular design strategies, a new NIR fluorophore was developed and characterized by simple synthesis, easy structural modification, and a large Stokes shift (105 nm). Furthermore, two new "activatable" fluorescent probes QN-Cys and QN-DNP were synthesized using a simple structural modification. The probe QN-Cys can recognize Cys with high sensitivity (LOD = 128 nM) and high selectivity, and its fluorescence intensity has a good linear relationship with the Cys concentration in the range of 5-35 μM. Furthermore, probe QN-Cys can effectively distinguish Cys from Hcy and GSH, and was successfully applied to the detection and imaging of Cys in human serum, cells, and zebrafish. The probe QN-DNP showed a good specific and sensitive (LOD = 78 nM) fluorescence response to thiophenol, and its fluorescence intensity has a good linear relationship with the thiophenol concentration in the range of 5-30 μM. Furthermore, it was successfully applied to detect thiophenol in real water samples with good recoveries (97-102%), and image thiophenol in living cells, zebrafish and mice. Notebly, the QN-DNP probe could be applied to visualize the distribution of thiophenol in the mice.
Collapse
Affiliation(s)
- Qing-Qing Yang
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China
| | - Nan Ji
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China
| | - Yu Zhan
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China
| | - Qin-Qin Tian
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China
| | - Ze-Dong Cai
- Department of Pharmaceutics, School of Pharmacy, The Fourth Military Medical University, Xi'an, 710032, PR China
| | - Xian-Lin Lu
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China
| | - Wei He
- Department of Chemistry, School of Pharmacy, The Fourth Military Medical University, 169 Changle West Road, Xi'an, 710032, PR China.
| |
Collapse
|
44
|
Algieri C, Trombetti F, Pagliarani A, Ventrella V, Nesci S. The mitochondrial F 1F O-ATPase exploits the dithiol redox state to modulate the permeability transition pore. Arch Biochem Biophys 2021; 712:109027. [PMID: 34520732 DOI: 10.1016/j.abb.2021.109027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 01/02/2023]
Abstract
The dithiol reagents phenylarsine oxide (PAO) and dibromobimane (DBrB) have opposite effects on the F1FO-ATPase activity. PAO 20% increases ATP hydrolysis at 50 μM when the enzyme activity is activated by the natural cofactor Mg2+ and at 150 μM when it is activated by Ca2+. The PAO-driven F1FO-ATPase activation is reverted to the basal activity by 50 μM dithiothreitol (DTE). Conversely, 300 μM DBrB decreases the F1FO-ATPase activity by 25% when activated by Mg2+ and by 50% when activated by Ca2+. In both cases, the F1FO-ATPase inhibition by DBrB is insensitive to DTE. The mitochondrial permeability transition pore (mPTP) formation, related to the Ca2+-dependent F1FO-ATPase activity, is stimulated by PAO and desensitized by DBrB. Since PAO and DBrB apparently form adducts with different cysteine couples, the results highlight the crucial role of cross-linking of vicinal dithiols on the F1FO-ATPase, with (ir)reversible redox states, in the mPTP modulation.
Collapse
Affiliation(s)
- Cristina Algieri
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra, 50, Ozzano Emilia, Bologna, 40064, Italy
| | - Fabiana Trombetti
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra, 50, Ozzano Emilia, Bologna, 40064, Italy
| | - Alessandra Pagliarani
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra, 50, Ozzano Emilia, Bologna, 40064, Italy
| | - Vittoria Ventrella
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra, 50, Ozzano Emilia, Bologna, 40064, Italy
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Via Tolara di Sopra, 50, Ozzano Emilia, Bologna, 40064, Italy.
| |
Collapse
|
45
|
Butturini E, Butera G, Pacchiana R, Carcereri de Prati A, Mariotto S, Donadelli M. Redox Sensitive Cysteine Residues as Crucial Regulators of Wild-Type and Mutant p53 Isoforms. Cells 2021; 10:cells10113149. [PMID: 34831372 PMCID: PMC8618966 DOI: 10.3390/cells10113149] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/25/2022] Open
Abstract
The wild-type protein p53 plays a key role in preventing the formation of neoplasms by controlling cell growth. However, in more than a half of all cancers, the TP53 gene has missense mutations that appear during tumorigenesis. In most cases, the mutated gene encodes a full-length protein with the substitution of a single amino acid, resulting in structural and functional changes and acquiring an oncogenic role. This dual role of the wild-type protein and the mutated isoforms is also evident in the regulation of the redox state of the cell, with antioxidant and prooxidant functions, respectively. In this review, we introduce a new concept of the p53 protein by discussing its sensitivity to the cellular redox state. In particular, we focus on the discussion of structural and functional changes following post-translational modifications of redox-sensitive cysteine residues, which are also responsible for interacting with zinc ions for proper structural folding. We will also discuss therapeutic opportunities using small molecules targeting cysteines capable of modifying the structure and function of the p53 mutant isoforms in view of possible anticancer therapies for patients possessing the mutation in the TP53 gene.
Collapse
Affiliation(s)
| | | | | | | | - Sofia Mariotto
- Correspondence: (S.M.); (M.D.); Tel.: +39-045-8027167 (S.M.); +39-045-8027281 (M.D.)
| | - Massimo Donadelli
- Correspondence: (S.M.); (M.D.); Tel.: +39-045-8027167 (S.M.); +39-045-8027281 (M.D.)
| |
Collapse
|
46
|
Evolutionary Aspects of the Oxido-Reductive Network of Methylglyoxal. J Mol Evol 2021; 89:618-638. [PMID: 34718825 DOI: 10.1007/s00239-021-10031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
In the chemoautotrophic theory for the origin of life, offered as an alternative to broth theory, the archaic reductive citric acid cycle operating without enzymes is in the center. The non-enzymatic (methyl)glyoxalase pathway has been suggested to be the anaplerotic route for the reductive citric acid cycle. In the recent years, much has been learned about methylglyoxal, but its importance in the metabolic machinery is still uncovered. If methylglyoxal had been essential participant of the early stage of evolution, then it is a legitimate question whether it might have played a role in the early oxido-reduction network, too. Therefore, an oxido-reduction network of methylglyoxal that might have functioned under ancient circumstances without enzymes was constructed and analyzed by virtue of group contribution method. Taking methylglyoxal as input material, it turned out that the evolutionary value of reactions and biomolecules were not similar. Glycerol, glycerate, and tartonate, the output components, were conserved to different degrees. Although the tartonate route was similarly favorable from energetic point of view, its intermediates are almost not present in extant biochemistry. The presence of two carboxyl or aldehyde groups, or their combination in tricarbons of the constructed network seemed disadvantageous for selection, and the inductive effect, resulting in an asymmetry in electron cloud of chemicals, might have been important. The evolutionary role for cysteine, H2S, and formaldehyde in the emergence of high-energy bonds in the form of thioesters and in Fe-S cluster formation as well as in imidazole synthesis was shown to bridge the gap between prebiotic chemistry and contemporary biochemistry. Overall, the ideas developed here represent an approach fitting to chemoautotrophic origin of life and implying to the role of methylglyoxal in triose formation. The proposed network is expected to have an impact upon how one may think of prebiological chemical processes on methylglyoxal, too. Finally, along the evolutionary time line, the network functioning without enzymes is situated between the formation of simple organic compounds and primeval cells, being closer to the former and well preceding the last common metabolic ancestor developed after primitive cells emerged.
Collapse
|
47
|
Spears RJ, McMahon C, Chudasama V. Cysteine protecting groups: applications in peptide and protein science. Chem Soc Rev 2021; 50:11098-11155. [PMID: 34605832 DOI: 10.1039/d1cs00271f] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protecting group chemistry for the cysteine thiol group has enabled a vast array of peptide and protein chemistry over the last several decades. Increasingly sophisticated strategies for the protection, and subsequent deprotection, of cysteine have been developed, facilitating synthesis of complex disulfide-rich peptides, semisynthesis of proteins, and peptide/protein labelling in vitro and in vivo. In this review, we analyse and discuss the 60+ individual protecting groups reported for cysteine, highlighting their applications in peptide synthesis and protein science.
Collapse
Affiliation(s)
| | - Clíona McMahon
- Department of Chemistry, University College London, London, UK.
| | - Vijay Chudasama
- Department of Chemistry, University College London, London, UK.
| |
Collapse
|
48
|
McLean JT, Benny A, Nolan MD, Swinand G, Scanlan EM. Cysteinyl radicals in chemical synthesis and in nature. Chem Soc Rev 2021; 50:10857-10894. [PMID: 34397045 DOI: 10.1039/d1cs00254f] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nature harnesses the unique properties of cysteinyl radical intermediates for a diverse range of essential biological transformations including DNA biosynthesis and repair, metabolism, and biological photochemistry. In parallel, the synthetic accessibility and redox chemistry of cysteinyl radicals renders them versatile reactive intermediates for use in a vast array of synthetic applications such as lipidation, glycosylation and fluorescent labelling of proteins, peptide macrocyclization and stapling, desulfurisation of peptides and proteins, and development of novel therapeutics. This review provides the reader with an overview of the role of cysteinyl radical intermediates in both chemical synthesis and biological systems, with a critical focus on mechanistic details. Direct insights from biological systems, where applied to chemical synthesis, are highlighted and potential avenues from nature which are yet to be explored synthetically are presented.
Collapse
Affiliation(s)
- Joshua T McLean
- Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse St., Dublin, D02 R590, Ireland.
| | - Alby Benny
- Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse St., Dublin, D02 R590, Ireland.
| | - Mark D Nolan
- Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse St., Dublin, D02 R590, Ireland.
| | - Glenna Swinand
- Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse St., Dublin, D02 R590, Ireland.
| | - Eoin M Scanlan
- Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse St., Dublin, D02 R590, Ireland.
| |
Collapse
|
49
|
Vitry J, Paré G, Murru A, Charest-Morin X, Maaroufi H, McLeish KR, Naccache PH, Fernandes MJ. Regulation of the Expression, Oligomerisation and Signaling of the Inhibitory Receptor CLEC12A by Cysteine Residues in the Stalk Region. Int J Mol Sci 2021; 22:ijms221910207. [PMID: 34638548 PMCID: PMC8508511 DOI: 10.3390/ijms221910207] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/11/2021] [Accepted: 09/15/2021] [Indexed: 01/26/2023] Open
Abstract
CLEC12A is a myeloid inhibitory receptor that negatively regulates inflammation in mouse models of autoimmune and autoinflammatory arthritis. Reduced CLEC12A expression enhances myeloid cell activation and inflammation in CLEC12A knock-out mice with collagen antibody-induced or gout-like arthritis. Similarly to other C-type lectin receptors, CLEC12A harbours a stalk domain between its ligand binding and transmembrane domains. While it is presumed that the cysteines in the stalk domain have multimerisation properties, their role in CLEC12A expression and/or signaling remain unknown. We thus used site-directed mutagenesis to determine whether the stalk domain cysteines play a role in CLEC12A expression, internalisation, oligomerisation, and/or signaling. Mutation of C118 blocks CLEC12A transport through the secretory pathway diminishing its cell-surface expression. In contrast, mutating C130 does not affect CLEC12A cell-surface expression but increases its oligomerisation, inducing ligand-independent phosphorylation of the receptor. Moreover, we provide evidence that CLEC12A dimerisation is regulated in a redox-dependent manner. We also show that antibody-induced CLEC12A cross-linking induces flotillin oligomerisation in insoluble membrane domains in which CLEC12A signals. Taken together, these data indicate that the stalk cysteines in CLEC12A differentially modulate this inhibitory receptor’s expression, oligomerisation and signaling, suggestive of the regulation of CLEC12A in a redox-dependent manner during inflammation.
Collapse
Affiliation(s)
- Julien Vitry
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Guillaume Paré
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Andréa Murru
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Xavier Charest-Morin
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Halim Maaroufi
- Institute of Integrative Biology and Systems, Laval University, Québec, QC G1V 0A6, Canada;
| | - Kenneth R. McLeish
- Department of Medicine, University of Louisville School of Medicine, Louisville, KY 40206, USA;
| | - Paul H. Naccache
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
| | - Maria J. Fernandes
- CHU de Québec Research Center, Division of Infectious Diseases and Immunology, Laval University, Québec, QC G1V 4G2, Canada; (J.V.); (G.P.); (A.M.); (P.H.N.)
- Department of Microbiology-Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Québec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-418-656-4141 (ext. 46106)
| |
Collapse
|
50
|
Ma X, Zhang J, Zhang C, Yang X, Yu A, Huang Y, Zhang S, Ouyang G. Targeting Enrichment and Correlation Studies of Glutathione and Homocysteine in IgAVN Patient Urine Based on a Core-Shell Zr-Based Metal-Organic Framework. ACS APPLIED MATERIALS & INTERFACES 2021; 13:40070-40078. [PMID: 34387999 DOI: 10.1021/acsami.1c09967] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Aminothiols are closely related to chronic kidney disease, but little is known regarding levels of related aminothiols in the urine of immunoglobulin A vasculitis with nephritis (IgAVN) patients. Herein, a well-defined core-shell Zr-based metal-organic framework (Zr-MOF) composite SiO2@50Benz-Cys was constructed as a mercury ion affinity material via a solvent-assisted ligand exchange strategy for the selective extraction and enrichment of low-concentration aminothiols in IgAVN patient urine. SiO2@50Benz-Cys was competent to enrich the total glutathione (GSH) and total homocysteine (Hcy) in virtue of the excellent affinity after chelation with mercury ions. The extraction efficiencies were closely related to the pH, dithiothreitol amount, and the dose of functional Zr-MOF. Coupled with HPLC-MS/MS in optimized conditions, GSH and Hcy were determined with low detection limits of 0.5 and 1 nmol L-1, respectively. The recoveries of GSH and Hcy for the urine sample at three spiked levels were in the range of 85.3-105% and 79.5-103%, which showed good precision and accuracy. Benefiting from the matrix interference elimination in the process of extraction, the simultaneous detection of aminothiols in the urine of the healthy group and immunoglobulin A vasculitis (IgAV) and IgAVN patients was successfully carried out, suggesting that the Zr-MOF and the robust method together provided a potential application in the analysis of urinary biomolecules. The analysis of variance (ANOVA) showed that the levels of GSH and Hcy had significant differences between the patients and the control. This work is very valuable as it provides a better understanding of concentration alterations of GSH and Hcy in urine involved with IgAVN for clinical research.
Collapse
Affiliation(s)
- Xue Ma
- College of Chemistry, Key Laboratory of Molecular Sensing and Harmful Substances Detection Technology, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| | - Jinghua Zhang
- College of Medicine, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| | - Chong Zhang
- College of Chemistry, Key Laboratory of Molecular Sensing and Harmful Substances Detection Technology, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| | - Xiaoqing Yang
- Henan University of Chinese Medicine, Zhengzhou 450008, P. R. China
| | - Ajuan Yu
- College of Chemistry, Key Laboratory of Molecular Sensing and Harmful Substances Detection Technology, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| | - Yanjie Huang
- Henan University of Chinese Medicine, Zhengzhou 450008, P. R. China
| | - Shusheng Zhang
- Center of Advanced Analysis and Gene Sequencing, Key Laboratory of Molecular Sensing and Harmful Substances Detection Technology, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| | - Gangfeng Ouyang
- Center of Advanced Analysis and Gene Sequencing, Key Laboratory of Molecular Sensing and Harmful Substances Detection Technology, Zhengzhou University, Kexue Avenue 100, Zhengzhou, Henan 450001, P. R. China
| |
Collapse
|