1
|
Diaz-Barreiro A, Cereghetti G, Ortega Sánchez FG, Tonacini J, Talabot-Ayer D, Kieffer-Jaquinod S, Kissling VM, Huard A, Swale C, Knowles TPJ, Couté Y, Peter M, Francés-Monerris A, Palmer G. Oxidation-sensitive cysteines drive IL-38 amyloid formation. Cell Rep 2024; 43:114940. [PMID: 39488827 DOI: 10.1016/j.celrep.2024.114940] [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: 08/19/2023] [Revised: 07/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024] Open
Abstract
Interleukin (IL)-1 family cytokines are essential for host defense at epithelial barriers. The IL-1 family member IL-33 was recently linked to stress granules (SGs). Formation of SGs and other biomolecular condensates is promoted by proteins containing low-complexity regions (LCRs). Computational analysis predicts LCRs in six of the 11 IL-1 family members. Among these, IL-38 contains a long LCR including two amyloid cores. IL-38 localizes to intracellular granules in keratinocytes under oxidative stress (OS) and forms OS-induced amyloid aggregates in cells and in vitro. Interestingly, soluble and aggregated IL-38 are released from keratinocytes in an exosome-enriched extracellular vesicle fraction. Disulfide-bond mapping, in silico modeling, and mutational analysis suggest that oxidation-sensitive cysteines act as redox switches to alter IL-38 conformation and promote its aggregation. Finally, the presence of IL-38 granules in human epidermis facing environmental OS suggests that oxidation-induced amyloidogenesis, as an intrinsic property of IL-38, supports barrier function.
Collapse
Affiliation(s)
- Alejandro Diaz-Barreiro
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland.
| | - Gea Cereghetti
- Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Francisco Gabriel Ortega Sánchez
- IBS Granada, Institute of Biomedical Research, Granada, Spain; Pulmonology Unit, Hospital Universitario Virgen de las Nieves, Granada, Spain; GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Granada, Spain
| | - Jenna Tonacini
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Dominique Talabot-Ayer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | | | - Vera Maria Kissling
- Particles-Biology Interactions Laboratory, Department of Materials Meet Life, Empa (Swiss Federal Laboratories for Materials Science and Technology), St. Gallen, Switzerland
| | - Arnaud Huard
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| | - Christopher Swale
- Institute for Advanced Biosciences (IAB), Team Host-Pathogen Interactions and Immunity to Infection, University Grenoble Alpes, INSERM U1209, CNRS UMR5309, Grenoble, France
| | | | - Yohann Couté
- Université Grenoble Alpes, CEA, INSERM, UA13 BGE, CNRS, CEA, FR2048 Grenoble, France
| | - Matthias Peter
- Institute of Biochemistry, Department of Biology, ETH Zurich, Zurich, Switzerland
| | | | - Gaby Palmer
- Division of Rheumatology, Department of Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Geneva Centre for Inflammation Research, Geneva, Switzerland
| |
Collapse
|
2
|
Tam S, Wear D, Morrone CD, Yu WH. The complexity of extracellular vesicles: Bridging the gap between cellular communication and neuropathology. J Neurochem 2024; 168:2391-2422. [PMID: 38650384 DOI: 10.1111/jnc.16108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/12/2024] [Accepted: 03/31/2024] [Indexed: 04/25/2024]
Abstract
Brain-derived extracellular vesicles (EVs) serve a prominent role in maintaining homeostasis and contributing to pathology in health and disease. This review establishes a crucial link between physiological processes leading to EV biogenesis and their impacts on disease. EVs are involved in the clearance and transport of proteins and nucleic acids, responding to changes in cellular processes associated with neurodegeneration, including autophagic disruption, organellar dysfunction, aging, and other cell stresses. In neurodegenerative disorders (e.g., Alzheimer's disease, Parkinson's disease, etc.), EVs contribute to the spread of pathological proteins like amyloid β, tau, ɑ-synuclein, prions, and TDP-43, exacerbating neurodegeneration and accelerating disease progression. Despite evidence for both neuropathological and neuroprotective effects of EVs, the mechanistic switch between their physiological and pathological functions remains elusive, warranting further research into their involvement in neurodegenerative disease. Moreover, owing to their innate ability to traverse the blood-brain barrier and their ubiquitous nature, EVs emerge as promising candidates for novel diagnostic and therapeutic strategies. The review uniquely positions itself at the intersection of EV cell biology, neurophysiology, and neuropathology, offering insights into the diverse biological roles of EVs in health and disease.
Collapse
Affiliation(s)
- Stephanie Tam
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Darcy Wear
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| | - Christopher D Morrone
- Brain Health Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Wai Haung Yu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Zhang Y, Zhang X, Zhang M, Zhang F, Chen T, Zha J, Shen Q, Wang D, Hou C. Hepatocytes-derived Prdx1 regulates macrophage phenotypes via TLR4 activation in acute liver injury. Int Immunopharmacol 2024; 127:111439. [PMID: 38159556 DOI: 10.1016/j.intimp.2023.111439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Acute liver injury (ALI) is a significant causative factor for multiple hepatic diseases. The excessive inflammatory response triggers proinflammatory immune cells recruitment, infiltration and differentiation, further contributing to inflammatory injuries in liver. As a proinflammatory factor, circulating Peroxiredoxin 1 (Prdx1) is elevated in ALI patients and mice. In this study, through carbon tetrachloride (CCl4) and cecal puncture and ligation (CLP)-induced liver injury mice model, we found hepatocytes-derived Prdx1 expression was increased in ALI. After AAV8-Prdx1-mediated Prdx1 knockdown, CCl4 and CLP-induced ALI was alleviated, along with the reduced proinflammatory cytokines, suppressed myeloid cells recruitment, decreased proportions of hepatic macrophages and neutrophils, restrained proinflammatory macrophage differentiation and infiltration. Mechanistically, hepatocyte-derived Prdx1 regulated macrophages through paracrine activation of the TLR4 signal. Our data support the immune and inflammatory regulatory role of Prdx1 in ALI pathological process to suggest its potential therapeutic application and clinical value.
Collapse
Affiliation(s)
- Yujing Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xinru Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Mingxun Zhang
- The First Affiliated Hospital of University of Science and Technology of China, China
| | - Fanrong Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Tong Chen
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Jingjing Zha
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Qiying Shen
- The First Affiliated Hospital of Anhui Medical University, China.
| | - Dong Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Chao Hou
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| |
Collapse
|
4
|
Kostyak JC, McKenzie SE, Naik UP. The Function of ASK1 in Sepsis and Stress-Induced Disorders. Int J Mol Sci 2023; 25:213. [PMID: 38203381 PMCID: PMC10778746 DOI: 10.3390/ijms25010213] [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: 11/28/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Apoptosis signal-regulating kinase 1 (ASK1) is a serine-threonine kinase that is ubiquitously expressed in nucleated cells and is responsible for the activation of multiple mitogen-activated protein kinases (MAPK) to regulate cell stress. Activation of ASK1 via cellular stress leads to activation of downstream signaling components, activation of transcription factors, and proinflammatory cytokine production. ASK1 is also expressed in anucleate platelets and is a key player in platelet activation as it is important for signaling. Interestingly, the mechanism of ASK1 activation is cell type-dependent. In this review we will explore how ASK1 regulates a variety of cellular processes from innate immune function to thrombosis and hemostasis. We will discuss how ASK1 influences FcγRIIA-mediated platelet reactivity and how that reactivity drives platelet clearance. Furthermore, we will explore the role of ASK1 in thromboxane (TxA2) generation, which highlights differences in the way ASK1 functions in mouse and human platelets.
Collapse
Affiliation(s)
- John C. Kostyak
- Cardeza Center for Hemostasis, Thrombosis, and Vascular Biology, Cardeza Foundation for Hematologic Research, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; (S.E.M.); (U.P.N.)
| | | | | |
Collapse
|
5
|
Vianello E, Persson J, Andersson B, van Veen S, Dias TL, Santoro F, Östensson M, Obudulu O, Agbajogu C, Torkzadeh S, Nakaya HI, Medaglini D, Siegrist CA, Ottenhoff TH, Harandi AM. Global blood miRNA profiling unravels early signatures of immunogenicity of Ebola vaccine rVSVΔG-ZEBOV-GP. iScience 2023; 26:108574. [PMID: 38162033 PMCID: PMC10755791 DOI: 10.1016/j.isci.2023.108574] [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: 04/11/2023] [Revised: 10/27/2023] [Accepted: 11/21/2023] [Indexed: 01/03/2024] Open
Abstract
The vectored Ebola vaccine rVSVΔG-ZEBOV-GP elicits protection against Ebola Virus Disease (EVD). In a study of forty-eight healthy adult volunteers who received either the rVSVΔG-ZEBOV-GP vaccine or placebo, we profiled intracellular microRNAs (miRNAs) from whole blood cells (WB) and circulating miRNAs from serum-derived extracellular vesicles (EV) at baseline and longitudinally following vaccination. Further, we identified early miRNA signatures associated with ZEBOV-specific IgG antibody responses at baseline and up to one year post-vaccination, and pinpointed target mRNA transcripts and pathways correlated to miRNAs whose expression was altered after vaccination by using systems biology approaches. Several miRNAs were differentially expressed (DE) and miRNA signatures predicted high or low IgG ZEBOV-specific antibody levels with high classification performance. The top miRNA discriminators were WB-miR-6810, EV-miR-7151-3p, and EV-miR-4426. An eight-miRNA antibody predictive signature was associated with immune-related target mRNAs and pathways. These findings provide valuable insights into early blood biomarkers associated with rVSVΔG-ZEBOV-GP vaccine-induced IgG antibody responses.
Collapse
Affiliation(s)
- Eleonora Vianello
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Josefine Persson
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Björn Andersson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Suzanne van Veen
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Malin Östensson
- Bioinformatics Core Facility, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ogonna Obudulu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Christopher Agbajogu
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Sara Torkzadeh
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - Donata Medaglini
- Department of Medical Biotechnologies, University of Siena, Italy
| | - Claire-Anne Siegrist
- Centre for Vaccinology, University Hospitals of Geneva and Faculty of Medicine, Geneva, Switzerland
| | - Tom H.M. Ottenhoff
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, the Netherlands
| | - Ali M. Harandi
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Vaccine Evaluation Center, BC Children’s Hospital Research Institute, University of British Columbia, Vancouver, Canada
| |
Collapse
|
6
|
Bai Z, Yin W, Liu R, Tang M, Shi X, Luo C, Xie X. PRDX1 Cys52Ser variant alleviates nonalcoholic steatohepatitis by reducing inflammation in mice. Mol Metab 2023; 76:101789. [PMID: 37562742 PMCID: PMC10470253 DOI: 10.1016/j.molmet.2023.101789] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023] Open
Abstract
OBJECTIVE Peroxiredoxin 1 (PRDX1) is a peroxidase and guards against oxidative stress by scavenging intracellular peroxides, whereas it also has been shown to stimulate inflammatory response by functioning as a chaperone protein. The potential in vivo link between PRDX1's peroxidase activity and its pro-inflammatory activity remains elusive. METHODS We generated peroxidase-dead PRDX1 variant mice by mutating its peroxidatic cysteine at 52 (Cys52) to serine, here referred to as PRDX1Cys52Ser. Trx-TrxR-NADPH coupled activity assay was applied to evaluate the peroxidase activity of global PRDX in PRDX1Cys52Ser variant mice. PRDX1Cys52Ser mice and their wild-type littermates were subjected to western diet or methionine and choline deficient diet feeding. NASH phenotypes were assessed through different analyses including physiological measurements, immunohistochemical staining, and quantitative PCR (qPCR). RNA sequencing, qPCR and western blotting were used to reveal and validate any changes in the signaling pathways responsible for the altered NASH phenotypes observed between WT and PRDX1Cys52Ser variant mice. RESULTS PRDX1Cys52Ser variant mice showed impaired global PRDX peroxidase activity and reduced susceptibility to diet-induced NASH and liver fibrosis. Mechanistically, PRDX1 Cys52Ser variant suppressed NF-κB signaling and STAT1 signaling pathways that are known to promote inflammation and NASH. CONCLUSION The peroxidatic Cys52 of PRDX1 is required for its pro-inflammatory activity in vivo. This study further suggests that PRDX1 may play dual but opposing roles in NASH.
Collapse
Affiliation(s)
- Zhonghao Bai
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Wen Yin
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Rui Liu
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Minglei Tang
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Xiaofeng Shi
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Cheng Luo
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan 528437, China; School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310000, China
| | - Xiangyang Xie
- NHC Key Laboratory of Hormones and Development, Tianjin Key Laboratory of Metabolic Diseases, Chu Hsien-I Memorial Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China.
| |
Collapse
|
7
|
Zhang Z, Zhou P, Liu M, Pei B. Expression And Prognostic Role of PRDX1 In Gastrointestinal Cancers. J Cancer 2023; 14:2895-2907. [PMID: 37781072 PMCID: PMC10539570 DOI: 10.7150/jca.86568] [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: 05/27/2023] [Accepted: 07/23/2023] [Indexed: 10/03/2023] Open
Abstract
Esophageal, gastric, liver, and colorectal cancers represent four prevalent gastrointestinal cancers that pose substantial threats to global health due to their high morbidity and mortality rates. Peroxiredoxin 1 (PRDX1), a significant component of the PRDXs family, primarily functions to counteract the peroxides produced by metabolic activities in the body, thereby maintaining the dynamic equilibrium of peroxides in vivo. Intriguingly, PRDX1 expression correlates strongly with cancer's onset, progression, and prognosis. This study mainly applied bioinformatics methods to analyze PRDX1's expression, diagnosis, and prognosis in gastrointestinal cancers and to summarize current research advancements. Evidence from the bioinformatics database suggested that the high expression of PRDX1 was a prominent characteristic of these four gastrointestinal cancers, with this observation reaching statistical significance. The high expression of PRDX1 in gastrointestinal cancer cells also confirms this result. Notably, the primary alteration in PRDX1 within these cancers is the presence of genetic mutations. PRDX1 demonstrated the highest diagnostic efficacy for colorectal cancer. Nevertheless, elevated PRDX1 levels only significantly diminished the survival time of liver cancer patients, exerting no statistically significant impact on the survival duration of patients afflicted by the other three types of gastrointestinal cancers. Recent research has indicated variability in PRDX1 expression across different cancer types, with high expression being predominantly observed in these four gastrointestinal cancers and, in most instances, unfavorable prognosis. These findings broadly align with the results derived from bioinformatics. This research underscores the high expression of PRDX1 in gastrointestinal cancers, its relevance to the diagnosis and prognosis monitoring of these cancers, and its potential to guide clinical treatment for these cancers.
Collapse
Affiliation(s)
- Zhou Zhang
- Department of Clinical Laboratory, Wuxi Huishan District People's Hospital, Wuxi, Jiangsu Province, 214000, China
| | - Pengli Zhou
- College of Basic Medicine, China Medical University, Shenyang, Liaoning province 110000, China
| | - Mingyue Liu
- Department of Ultrasound, Wuxi No.2 People's Hospital; Jiangnan University Medical Center, Affiliated Wuxi Clinical College of Nantong University, Wuxi, Jiangsu province 214002, China
| | - Bing Pei
- Department of Clinical Laboratory, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, Jiangsu, 223800, China
| |
Collapse
|
8
|
Martinez-Banaclocha MA. Targeting the Cysteine Redox Proteome in Parkinson's Disease: The Role of Glutathione Precursors and Beyond. Antioxidants (Basel) 2023; 12:1373. [PMID: 37507913 PMCID: PMC10376658 DOI: 10.3390/antiox12071373] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
Encouraging recent data on the molecular pathways underlying aging have identified variants and expansions of genes associated with DNA replication and repair, telomere and stem cell maintenance, regulation of the redox microenvironment, and intercellular communication. In addition, cell rejuvenation requires silencing some transcription factors and the activation of pluripotency, indicating that hidden molecular networks must integrate and synchronize all these cellular mechanisms. Therefore, in addition to gene sequence expansions and variations associated with senescence, the optimization of transcriptional regulation and protein crosstalk is essential. The protein cysteinome is crucial in cellular regulation and plays unexpected roles in the aging of complex organisms, which show cumulative somatic mutations, telomere attrition, epigenetic modifications, and oxidative dysregulation, culminating in cellular senescence. The cysteine thiol groups are highly redox-active, allowing high functional versatility as structural disulfides, redox-active disulfides, active-site nucleophiles, proton donors, and metal ligands to participate in multiple regulatory sites in proteins. Also, antioxidant systems control diverse cellular functions, including the transcription machinery, which partially depends on the catalytically active cysteines that can reduce disulfide bonds in numerous target proteins, driving their biological integration. Since we have previously proposed a fundamental role of cysteine-mediated redox deregulation in neurodegeneration, we suggest that cellular rejuvenation of the cysteine redox proteome using GSH precursors, like N-acetyl-cysteine, is an underestimated multitarget therapeutic approach that would be particularly beneficial in Parkinson's disease.
Collapse
|
9
|
Ischemia challenged epicardial adipose tissue stem cells-derived extracellular vesicles alter the gene expression of cardiac fibroblasts to cardiomyocyte like phenotype. Transl Res 2023; 254:54-67. [PMID: 36273744 DOI: 10.1016/j.trsl.2022.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The present study hypothesizes that the ischemic insults activate epicardial adipose tissue-derived stem cells (EATDS) to secrete extracellular vesicles (EVs) packed with regenerative mediators to alter the gene expression in cardiac fibroblasts (CF). EATDS and CF were isolated from hyperlipidemic microswine and EVs were harvested from control, simulated ischemia (ISC) and ischemia-reperfusion (ISC/R) groups. The in vitro interaction between ISC-EVs and CF resulted in the upregulation of cardiomyocyte-specific transcription factors including GATA4, Nkx2.5, IRX4, and TBX5 in CF and the healing marker αSMA and the downregulation of fibroblast biomarkers such as vimentin, FSP1, and podoplanin and the cardiac biomarkers such as troponin-I and connexin-43. These results suggest a cardiomyocyte-like phenotype as confirmed by immunostaining and Western blot. The LC-MS/MS analysis of ISC-EVs LGALS1, PRDX2, and CCL2 to be the potent protein mediators which are intimately involved in versatile regenerative processes and connected with a diverse array of regenerative genes. Moreover, the LGALS1+, PRDX2+, and CCL2+ EATDS phenotypes were deciphered at single cell resolution revealing corresponding sub-populations with superior healing potential. Overall, the findings unveiled the healing potential of EATDS-derived EVs and sub-populations of regenerative EATDS promising novel translational opportunities in improved cardiac healing following ischemic injury.
Collapse
|
10
|
Möller M, Orrico F, Villar S, López AC, Silva N, Donzé M, Thomson L, Denicola A. Oxidants and Antioxidants in the Redox Biochemistry of Human Red Blood Cells. ACS OMEGA 2023; 8:147-168. [PMID: 36643550 PMCID: PMC9835686 DOI: 10.1021/acsomega.2c06768] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/09/2022] [Indexed: 06/01/2023]
Abstract
Red blood cells (RBCs) are exposed to both external and internal sources of oxidants that challenge their integrity and compromise their physiological function and supply of oxygen to tissues. Autoxidation of oxyhemoglobin is the main source of endogenous RBC oxidant production, yielding superoxide radical and then hydrogen peroxide. In addition, potent oxidants from other blood cells and the surrounding endothelium can reach the RBCs. Abundant and efficient enzymatic systems and low molecular weight antioxidants prevent most of the damage to the RBCs and also position the RBCs as a sink of vascular oxidants that allow the body to maintain a healthy circulatory system. Among the antioxidant enzymes, the thiol-dependent peroxidase peroxiredoxin 2, highly abundant in RBCs, is essential to keep the redox balance. A great part of the RBC antioxidant activity is supported by an active glucose metabolism that provides reducing power in the form of NADPH via the pentose phosphate pathway. There are several RBC defects and situations that generate oxidative stress conditions where the defense mechanisms are overwhelmed, and these include glucose-6-phosphate dehydrogenase deficiencies (favism), hemoglobinopathies like sickle cell disease and thalassemia, as well as packed RBCs for transfusion that suffer from storage lesions. These oxidative stress-associated pathologies of the RBCs underline the relevance of redox balance in these anucleated cells that lack a mechanism of DNA-inducible antioxidant response and rely on a complex and robust network of antioxidant systems.
Collapse
Affiliation(s)
- Matias
N. Möller
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Florencia Orrico
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Sebastián
F. Villar
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Ana C. López
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Nicolás Silva
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
- Departamento
de Medicina Transfusional, Hospital de Clínicas, Facultad de
Medicina, Universidad de la República, Montevideo 11600, Uruguay
| | - Marcel Donzé
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| | - Leonor Thomson
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
- Laboratorio
de Enzimología, Instituto de Química Biológica,
Facultad de Ciencias, Universidad de la
República, Montevideo 11400, Uruguay
| | - Ana Denicola
- Laboratorio
de Fisicoquímica Biológica, Instituto de Química
Biológica, Facultad de Ciencias,
Universidad de la República, Montevideo 11400, Uruguay
- Centro
de Investigaciones Biomédicas (CEINBIO), Universidad de la República, Montevideo 11800, Uruguay
| |
Collapse
|
11
|
Liu G, Miao F, Wang Y, Kou J, Yang K, Li W, Xiong C, Zhang F, Wang X, Yan H, Wei C, Zhao C, Yan G. Comparative proteomics analysis of Schistosoma japonicum developed in different Oncomelania snails as intermediate hosts. Front Cell Infect Microbiol 2022; 12:959766. [PMID: 36710964 PMCID: PMC9875565 DOI: 10.3389/fcimb.2022.959766] [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: 06/02/2022] [Accepted: 09/26/2022] [Indexed: 12/23/2022] Open
Abstract
Schistosomiasis is a tropical parasitic disease that seriously endangers humans and animals. In this study, two Oncomelania snails, Oncomelania hupensis (O. hupensis) and Oncomelania weishan (O. weishan), were infected with Schistosoma japonicum (S. japonicum) cercariae during the early period, and ICR mice were subsequently infected with two kinds of miracidia that developed in male and female adult worms. In this study, isobaric tags for relative and absolute quantification (iTRAQ) were used to identify four channels: 113, 115, 117, and 119. A total of 2364 adult schistosome proteins were identified, and 1901 proteins were quantitative. Our results revealed 68 differentially expressed proteins (DEPs) in female adult worms, including 24 upregulated proteins and 44 downregulated proteins, and 55 DEPs in male adult worms, including 25 upregulated proteins and 30 downregulated proteins. LC-MS/MS and bioinformatics analysis indicated that these DEPs are mainly concentrated in cellular composition, molecular function, biological function and catabolism pathways. In summary, this proteomics analysis of adult schistosomes that hatched in two intermediate hosts helps to improve our understanding of the growth and developmental mechanisms of S. japonicum.
Collapse
Affiliation(s)
- Gongzhen Liu
- College of Agriculture and Forestry, Linyi University, Linyi, Shandong Province, China,Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China
| | - Feng Miao
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China,*Correspondence: Feng Miao,
| | - Yongbin Wang
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China
| | - Jingxuan Kou
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China
| | - Kun Yang
- Jiangsu Institutes of Parasitic Diseases, Wuxi, Jiangsu Province, China
| | - Wei Li
- Jiangsu Institutes of Parasitic Diseases, Wuxi, Jiangsu Province, China
| | - Chunrong Xiong
- Jiangsu Institutes of Parasitic Diseases, Wuxi, Jiangsu Province, China
| | - Fengjian Zhang
- Jiangsu Institutes of Parasitic Diseases, Wuxi, Jiangsu Province, China
| | - Xinyao Wang
- Jiangsu Institutes of Parasitic Diseases, Wuxi, Jiangsu Province, China
| | - Haoyun Yan
- Fourth Hospital of Weishan, Jining, Shandong Province, China
| | - Changyin Wei
- Shandong Weishan Center for Disease Prevention and Control, Jining, Shandong Province, China
| | - Changlei Zhao
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China
| | - Ge Yan
- Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining, Shandong Province, China
| |
Collapse
|
12
|
Meng Q, Wang J, Cui J, Li B, Wu S, Yun J, Aschner M, Wang C, Zhang L, Li X, Chen R. Prediction of COPD acute exacerbation in response to air pollution using exosomal circRNA profile and Machine learning. ENVIRONMENT INTERNATIONAL 2022; 168:107469. [PMID: 36041244 PMCID: PMC9939562 DOI: 10.1016/j.envint.2022.107469] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 08/10/2022] [Indexed: 05/11/2023]
Abstract
Ambient fine particulate matter (PM2.5) is linked to an increased risk of chronic obstructive pulmonary disease (COPD) exacerbations, which significantly increase the risk of mortality in COPD patients. Identifying the subtype of COPD patients who are sensitive to environmental aggressions is necessary. Using in vitro and in vivo PM2.5 exposure models, we demonstrate that exosomal hsa_circ_0005045 is upregulated by PM2.5 and binds to the protein cargo peroxiredoxin2, which functionally aggravates hallmarks of COPD by recruiting neutrophil elastase and triggering in situ release of tumor necrosis factor (TNF)-α by inflammatory cells. The biological function of hsa_circ_0005045 associated with aggravation of COPD is validated using exosome-transplantation and conditional circRNA-knockdown murine models. By sorting the major components of PM2.5, we find that PM2.5-bound heavy metals, which are distinguishable from the components of cigarette smoke, trigger the elevation of exosomal hsa_circ_0005045. Finally, using machine learning models in a cohort with 327 COPD patients, the PM2.5 exposure-sensitive COPD patients are characterized by relatively high hsa_circ_0005045 expression, non-smoking, and group C (mMRC 0-1 (or CAT < 10) and ≥ 2 exacerbations (or ≥ 1 exacerbation leading to hospital admission) in the past year). Thus, our results suggest that environmental reduction in PM2.5 emission provides a targeted approach to protecting non-smoking COPD patients against air pollution-related disease exacerbation.
Collapse
Affiliation(s)
- Qingtao Meng
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Jiajia Wang
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Jian Cui
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, Medical School of Southeast University, 87, Ding Jia Qiao Road, Nanjing 210009, China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Bin Li
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Shenshen Wu
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China
| | - Jun Yun
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Forchheimer 209, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Chengshuo Wang
- Department of Otolaryngology, Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing 100730, China; Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing 100005, China
| | - Luo Zhang
- Department of Allergy, Beijing TongRen Hospital, Capital Medical University, Beijing, 100005, China; Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otolaryngology, Beijing China; Department of Otolaryngology Head and Neck Surgery, Beijing TongRen Hospital, Capital Medical University, Beijing 100005, China.
| | - Xiaobo Li
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China; Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Rui Chen
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, PR China; School of Public Health, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing 100069, PR China; Institute for Chemical Carcinogenesis, Guangzhou Medical University, Guangzhou 511436, PR China.
| |
Collapse
|
13
|
Changing Perspectives from Oxidative Stress to Redox Signaling-Extracellular Redox Control in Translational Medicine. Antioxidants (Basel) 2022; 11:antiox11061181. [PMID: 35740078 PMCID: PMC9228063 DOI: 10.3390/antiox11061181] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/07/2022] Open
Abstract
Extensive research has changed the understanding of oxidative stress that has been linked to every major disease. Today we distinguish oxidative eu- and distress, acknowledging that redox modifications are crucial for signal transduction in the form of specific thiol switches. Long underestimated, reactive species and redox proteins of the Thioredoxin (Trx) family are indeed essential for physiological processes. Moreover, extracellular redox proteins, low molecular weight thiols and thiol switches affect signal transduction and cell–cell communication. Here, we highlight the impact of extracellular redox regulation for health, intermediate pathophenotypes and disease. Of note, recent advances allow the analysis of redox changes in body fluids without using invasive and expensive techniques. With this new knowledge in redox biochemistry, translational strategies can lead to innovative new preventive and diagnostic tools and treatments in life sciences and medicine.
Collapse
|
14
|
Reciprocal Regulation of Shh Trafficking and H2O2 Levels via a Noncanonical BOC-Rac1 Pathway. Antioxidants (Basel) 2022; 11:antiox11040718. [PMID: 35453403 PMCID: PMC9025708 DOI: 10.3390/antiox11040718] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/25/2022] [Accepted: 04/03/2022] [Indexed: 11/21/2022] Open
Abstract
Among molecules that bridge environment, cell metabolism, and cell signaling, hydrogen peroxide (H2O2) recently appeared as an emerging but central player. Its level depends on cell metabolism and environment and was recently shown to play key roles during embryogenesis, contrasting with its long-established role in disease progression. We decided to explore whether the secreted morphogen Sonic hedgehog (Shh), known to be essential in a variety of biological processes ranging from embryonic development to adult tissue homeostasis and cancers, was part of these interactions. Here, we report that H2O2 levels control key steps of Shh delivery in cell culture: increased levels reduce primary secretion, stimulate endocytosis and accelerate delivery to recipient cells; in addition, physiological in vivo modulation of H2O2 levels changes Shh distribution and tissue patterning. Moreover, a feedback loop exists in which Shh trafficking controls H2O2 synthesis via a non-canonical BOC-Rac1 pathway, leading to cytoneme growth. Our findings reveal that Shh directly impacts its own distribution, thus providing a molecular explanation for the robustness of morphogenesis to both environmental insults and individual variability.
Collapse
|
15
|
Mujammami M, Rafiullah M, Alfadda AA, Akkour K, Alanazi IO, Masood A, Musambil M, Alhalal H, Arafah M, Rahman AMA, Benabdelkamel H. Proteomic Analysis of Endometrial Cancer Tissues from Patients with Type 2 Diabetes Mellitus. Life (Basel) 2022; 12:life12040491. [PMID: 35454982 PMCID: PMC9030544 DOI: 10.3390/life12040491] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Endometrial cancer (EC) is the most common form of gynecological cancer. Type 2 diabetes mellitus is associated with an increased risk of EC. Currently, no proteomic studies have investigated the role of diabetes in endometrial cancers from clinical samples. The present study aims to elucidate the molecular link between diabetes and EC using a proteomic approach. Endometrial tissue samples were obtained from age-matched patients (EC Diabetic and EC Non-Diabetic) during surgery. Untargeted proteomic analysis of the endometrial tissues was carried out using a two-dimensional difference in gel electrophoresis (2D-DIGE) coupled with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI TOF). A total of 53 proteins were identified, with a significant difference in abundance (analysis of variance (ANOVA) test, p ≤ 0.05; fold-change ≥ 1.5) between the two groups, among which 30 were upregulated and 23 downregulated in the EC Diabetic group compared to EC Non-Diabetic. The significantly upregulated proteins included peroxiredoxin-1, vinculin, endoplasmin, annexin A5, calreticulin, and serotransferrin. The significantly downregulated proteins were myosin regulatory light polypeptide 9, Retinol dehydrogenase 12, protein WWC3, intraflagellar transport protein 88 homolog, superoxide dismutase [Cu-Zn], and retinal dehydrogenase 1. The network pathway was related to connective tissue disorder, developmental disorder, and hereditary disorder, with the identified proteins centered around dysregulation of ERK1/2 and F Actin signaling pathways. Cancer-associated protein alterations such as upregulation of peroxiredoxin-1, annexin 5, and iNOS, and downregulation of RDH12, retinaldehyde dehydrogenase 1, SOD1, and MYL 9, were found in the EC tissues of the diabetic group. Differential expression of proteins linked to cancer metastasis, such as the upregulation of vinculin and endoplasmin and downregulation of WWC3 and IFT88, was seen in the patients with diabetes. Calreticulin and alpha-enolase, which might have a role in the interplay between diabetes and EC, need further investigation.
Collapse
Affiliation(s)
- Muhammad Mujammami
- University Diabetes Center, King Saud University Medical City, King Saud University, Riyadh 11461, Saudi Arabia;
- Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| | - Mohamed Rafiullah
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
| | - Assim A. Alfadda
- Department of Medicine, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
- Strategic Center for Diabetes Research, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia;
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.M.); (M.M.)
| | - Khalid Akkour
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.)
| | - Ibrahim O. Alanazi
- The National Center for Biotechnology (NCB), Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Afshan Masood
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.M.); (M.M.)
| | - Mohthash Musambil
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.M.); (M.M.)
| | - Hani Alhalal
- Obstetrics and Gynecology Department, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (K.A.); (H.A.)
| | - Maria Arafah
- Department of Pathology, College of Medicine, King Saud University, King Saud University Medical City, Riyadh 11461, Saudi Arabia;
| | - Anas M. Abdel Rahman
- Metabolomics Section, Center for Genome Medicine, Department of Clinical Genomics, King Faisal Specialist Hospital and Research Centre (KFSHRC), Riyadh 11211, Saudi Arabia;
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Hicham Benabdelkamel
- Proteomics Resource Unit, Obesity Research Center, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (A.M.); (M.M.)
- Correspondence:
| |
Collapse
|
16
|
Martinez-Banaclocha M. N-Acetyl-Cysteine: Modulating the Cysteine Redox Proteome in Neurodegenerative Diseases. Antioxidants (Basel) 2022; 11:antiox11020416. [PMID: 35204298 PMCID: PMC8869501 DOI: 10.3390/antiox11020416] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/13/2022] [Accepted: 02/16/2022] [Indexed: 12/14/2022] Open
Abstract
In the last twenty years, significant progress in understanding the pathophysiology of age-associated neurodegenerative diseases has been made. However, the prevention and treatment of these diseases remain without clinically significant therapeutic advancement. While we still hope for some potential genetic therapeutic approaches, the current reality is far from substantial progress. With this state of the issue, emphasis should be placed on early diagnosis and prompt intervention in patients with increased risk of neurodegenerative diseases to slow down their progression, poor prognosis, and decreasing quality of life. Accordingly, it is urgent to implement interventions addressing the psychosocial and biochemical disturbances we know are central in managing the evolution of these disorders. Genomic and proteomic studies have shown the high molecular intricacy in neurodegenerative diseases, involving a broad spectrum of cellular pathways underlying disease progression. Recent investigations indicate that the dysregulation of the sensitive-cysteine proteome may be a concurrent pathogenic mechanism contributing to the pathophysiology of major neurodegenerative diseases, opening new therapeutic opportunities. Considering the incidence and prevalence of these disorders and their already significant burden in Western societies, they will become a real pandemic in the following decades. Therefore, we propose large-scale investigations, in selected groups of people over 40 years of age with decreased blood glutathione levels, comorbidities, and/or mild cognitive impairment, to evaluate supplementation of the diet with low doses of N-acetyl-cysteine, a promising and well-tolerated therapeutic agent suitable for long-term use.
Collapse
|
17
|
Aki T, Unuma K, Uemura K. The Role of Peroxiredoxins in the Regulation of Sepsis. Antioxidants (Basel) 2022; 11:antiox11010126. [PMID: 35052630 PMCID: PMC8773135 DOI: 10.3390/antiox11010126] [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: 12/12/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 02/01/2023] Open
Abstract
Oxidative stress, a result of a disturbance in redox homeostasis, is considered to be one of the main aggravating events in the pathogenesis of immune disorders. Peroxiredoxins (Prdxs) are an enzyme family that catalyzes the reduction of peroxides, including hydrogen peroxide, lipid peroxides, and nitrogen peroxides. Although the maintenance of cellular redox homeostasis through Prdxs is essential for surviving in adverse environments, Prdxs also participate in the regulation of cellular signal transduction by modulating the activities of a panel of molecules involved in the signal transduction process. Although Prdxs were discovered as intracellular anti-oxidative enzymes, recent research has revealed that Prdxs also play important roles in the extracellular milieu. Indeed, Prdxs have been shown to have the capacity to activate immune cells through ligation with innate immune receptors such as toll-like receptors (TLRs). In this review, we will summarize the intracellular as well as extracellular roles of Prdxs for and against the pathogenesis of inflammatory disorders including sepsis, hemorrhagic shock, and drug-induced liver injury.
Collapse
|
18
|
Jaiswal S, Joshi B, Chen J, Wang F, Dame MK, Spence JR, Newsome GM, Katz EL, Shah YM, Ramakrishnan SK, Li G, Lee M, Appelman HD, Kuick R, Wang TD. Membrane Bound Peroxiredoxin-1 Serves as a Biomarker for In Vivo Detection of Sessile Serrated Adenomas. Antioxid Redox Signal 2022; 36:39-56. [PMID: 34409853 PMCID: PMC8792500 DOI: 10.1089/ars.2020.8244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Aim: Sessile serrated adenomas (SSAs) are premalignant lesions driven by the BRAFV600E mutation to give rise to colorectal cancers (CRCs). They are often missed during white light colonoscopy because of their subtle appearance. Previously, a fluorescently labeled 7mer peptide KCCFPAQ was shown to detect SSAs in vivo. We aim to identify the target of this peptide. Results: Peroxiredoxin-1 (Prdx1) was identified as the binding partner of the peptide ligand. In vitro binding assays and immunofluorescence staining of human colon specimens ex vivo supported this result. Prdx1 was overexpressed on the membrane of cells with the BRAFV600E mutation, and this effect was dependent on oxidative stress. RKO cells harboring the BRAFV600E mutation and human SSA specimens showed higher oxidative stress as well as elevated levels of Prdx1 on the cell membrane. Innovation and Conclusion: These results suggest that Prdx1 is overexpressed on the cell surface in the presence of oxidative stress and can serve as an imaging biomarker for in vivo detection of SSAs. Antioxid. Redox Signal. 36, 39-56.
Collapse
Affiliation(s)
- Sangeeta Jaiswal
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Bishnu Joshi
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jing Chen
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fa Wang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Michael K Dame
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason R Spence
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gina M Newsome
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Erica L Katz
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Yatrik M Shah
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sadeesh K Ramakrishnan
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gaoming Li
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Miki Lee
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Henry D Appelman
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Rork Kuick
- Department of Biostatistics, and University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas D Wang
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, Michigan, USA.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Ramos AP, Sebinelli HG, Ciancaglini P, Rosato N, Mebarek S, Buchet R, Millán JL, Bottini M. The functional role of soluble proteins acquired by extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e34. [PMID: 38938684 PMCID: PMC11080634 DOI: 10.1002/jex2.34] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 06/29/2024]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed nanosized particles released by all cell types during physiological as well as pathophysiological processes to carry out diverse biological functions, including acting as sources of cellular dumping, signalosomes and mineralisation nanoreactors. The ability of EVs to perform specific biological functions is due to their biochemical machinery. Among the components of the EVs' biochemical machinery, surface proteins are of critical functional significance as they mediate the interactions of EVs with components of the extracellular milieu, the extracellular matrix and neighbouring cells. Surface proteins are thought to be native, that is, pre-assembled on the EVs' surface by the parent cells before the vesicles are released. However, numerous pieces of evidence have suggested that soluble proteins are acquired by the EVs' surface from the extracellular milieu and further modulate the biological functions of EVs during innate and adaptive immune responses, autoimmune disorders, complement activation, coagulation, viral infection and biomineralisation. Herein, we will describe the methods currently used to identify the EVs' surface proteins and discuss recent knowledge on the functional relevance of the soluble proteins acquired by EVs.
Collapse
Affiliation(s)
- Ana Paula Ramos
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Heitor Gobbi Sebinelli
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Pietro Ciancaglini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Nicola Rosato
- Dipartimento di Medicina SperimentaleUniversita’ di Roma “Tor Vergata”RomeItaly
| | - Saida Mebarek
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | - Rene Buchet
- ICBMS UMR CNRS 5246UFR BiosciencesUniversité Lyon 1Villeurbanne CedexFrance
| | | | - Massimo Bottini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão PretoUniversidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
- Sanford Burnham PrebysLa JollaCaliforniaUSA
| |
Collapse
|
20
|
Cheng X, Fu Z, Xie W, Zhu L, Meng J. Preoperative circulating peroxiredoxin 1 levels as a predictor of non-alcoholic fatty liver disease remission after laparoscopic bariatric surgery. Front Endocrinol (Lausanne) 2022; 13:1072513. [PMID: 36619535 PMCID: PMC9810748 DOI: 10.3389/fendo.2022.1072513] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is associated with obesity and insulin resistance and can be improved after bariatric surgery. Circulating Peroxiredoxin 1 (Prdx1) protein was reported to regulate energy metabolism and inflammation. This study aimed to investigate the roles of serum prdx1 in NAFLD patients with obesity undergoing LSG and to develop a prognostic model to predict the remission of severe NAFLD. METHODS The data of 93 participants from a tertiary hospital were assessed. Before laparoscopic sleeve gastrectomy (LSG) and three months after LSG, anthropometric parameters, laboratory biochemical data, and abdominal B-ultrasound results were collected, and their hepatic steatosis index (HSI) and triglyceride-glucose index (TyG) were calculated. A NAFLD improvement (NAFLD-I) nomogram prediction model was constructed using the least absolute shrinkage and selection operator (LASSO) regression and multiple regression, and its predictive ability was verified in a validation cohort. RESULTS The baseline Prdx1 (OR: 0.887, 95% CI: 0.816-0.963, p=0.004), preoperative TyG (OR: 8.207, 95% CI: 1.903-35.394, p=0.005) and HSI (OR: 0.861, 95% CI: 0.765-0.969, p=0.013) levels were independently associated with NAFLD-I at three months after LSG in NAFLD patients with obesity. In the primary and validation cohorts, the area under the receiver operating characteristic (AUC) of the developed nomogram model was 0.891 and 0.878, respectively. The preoperative circulating Prdx1 levels of NAFLD patients with obesity were significantly reduced after LSG (25.32 [18.99-30.88] vs. 23.34 [15.86-26.42], p=0.001). Prdx1 was related to obesity and hepatic steatosis based on correlation analysis. CONCLUSION The nomogram based on preoperative serum prdx1, HSI and TyG could be an effective tool for predicting remission of severe NAFLD after LSG.
Collapse
Affiliation(s)
- Xiaoyun Cheng
- Department of Pulmonary and Critical Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China
| | - Zhibing Fu
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xie
- Department of Cardiology, Xiangya Hospital, Central South University, Changsha, China
| | - Liyong Zhu
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China
- *Correspondence: Jie Meng, ; Liyong Zhu,
| | - Jie Meng
- Department of Pulmonary and Critical Care Medicine, Xiangya Hospital of Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Organ Fibrosis, Central South University, Changsha, Hunan, China
- *Correspondence: Jie Meng, ; Liyong Zhu,
| |
Collapse
|
21
|
Huang Y, Driedonks TA, Cheng L, Rajapaksha H, Routenberg DA, Nagaraj R, Redding J, Arab T, Powell BH, Pletniková O, Troncoso JC, Zheng L, Hill AF, Mahairaki V, Witwer KW. Brain Tissue-Derived Extracellular Vesicles in Alzheimer's Disease Display Altered Key Protein Levels Including Cell Type-Specific Markers. J Alzheimers Dis 2022; 90:1057-1072. [PMID: 36213994 PMCID: PMC9741741 DOI: 10.3233/jad-220322] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Brain tissue-derived extracellular vesicles (bdEVs) play neurodegenerative and protective roles, including in Alzheimer's disease (AD). Extracellular vesicles (EVs) may also leave the brain to betray the state of the CNS in the periphery. Only a few studies have profiled the proteome of bdEVs and source brain tissue. Additionally, studies focusing on bdEV cell type-specific surface markers are rare. OBJECTIVE We aimed to reveal the pathological mechanisms inside the brain by profiling the tissue and bdEV proteomes in AD patients. In addition, to indicate targets for capturing and molecular profiling of bdEVs in the periphery, CNS cell-specific markers were profiled on the intact bdEV surface. METHODS bdEVs were separated and followed by EV counting and sizing. Brain tissue and bdEVs from age-matched AD patients and controls were then proteomically profiled. Total tau (t-tau), phosphorylated tau (p-tau), and antioxidant peroxiredoxins (PRDX) 1 and 6 were measured by immunoassay in an independent bdEV separation. Neuron, microglia, astrocyte, and endothelia markers were detected on intact EVs by multiplexed ELISA. RESULTS Overall, concentration of recovered bdEVs was not affected by AD. Proteome differences between AD and control were more pronounced for bdEVs than for brain tissue. Levels of t-tau, p-tau, PRDX1, and PRDX6 were significantly elevated in AD bdEVs compared with controls. Release of certain cell-specific bdEV markers was increased in AD. CONCLUSION Several bdEV proteins are involved in AD mechanisms and may be used for disease monitoring. The identified CNS cell markers may be useful tools for peripheral bdEV capture.
Collapse
Affiliation(s)
- Yiyao Huang
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tom A.P. Driedonks
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lesley Cheng
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | - Harinda Rajapaksha
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| | | | | | - Javier Redding
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Tanina Arab
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Bonita H. Powell
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Olga Pletniková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Andrew F. Hill
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
- Institute of Health and Sport, Victoria University, Melbourne, VIC, Australia
| | - Vasiliki Mahairaki
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Richman Family Precision Medicine Center of Excellence in Alzheimer’s Disease, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
22
|
Khodamoradi K, Golan R, Dullea A, Ramasamy R. Exosomes as Potential Biomarkers for Erectile Dysfunction, Varicocele, and Testicular Injury. Sex Med Rev 2021; 10:311-322. [PMID: 34838504 DOI: 10.1016/j.sxmr.2021.10.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Optimal male reproductive health is dependent upon critical mediators of cell-cell communication: exosomes or extracellular vesicles. These vesicles are nano-sized particles released into a variety of bodily fluids, such as blood and semen. Exosomes are highly stable and can carry genetic and other molecules, including DNA, RNA, and proteins, which provide information about their origin cells. OBJECTIVE To identify exosomes as potential biomarkers or therapeutic mediators in male sexual and reproductive disorders like erectile dysfunction (ED), varicocele, and testicular injury. METHODS A PubMed search was performed to highlight all articles available relating to exosomes and extracellular vesicles in the pathogenesis of different male sexual and reproductive disorders, and their importance in clinical use as both diagnostic markers and potential therapeutic mediators. RESULTS Various male reproductive system disorders, such as ED, varicocele, and testicular injury, are linked to increased or decreased levels of exosomes. Exosomes have a higher number of molecules such as DNA, RNA, and proteins, which can give a more precise and comprehensive result when compared to other biomarkers. Exosomes can be considered as plausible diagnostic biomarkers for male sexual and reproductive diseases, with considerable advantages over other diagnostic procedures such as invasive tissue biopsy. Exosomes can carry cargo such certain drugs and therapeutic molecules making them a promising therapeutic approach. Several studies have begun to test treating various male sexual reproductive disorders with exosomes. CONCLUSION Exosomes deliver many components that can regulate gene expression and target signaling pathways. Understanding how extracellular vesicles can be utilized as biomarkers in diagnosing men, particularly those with idiopathic erectile dysfunction, will not only aid in diagnosis but also help with making therapeutic targets. Khodamoradi K, Golan R, Dullea A, et al. Exosomes as Potential Biomarkers for Erectile Dysfunction, Varicocele, and Testicular Injury. Sex Med Rev 2021;XX:XXX-XXX.
Collapse
Affiliation(s)
- Kajal Khodamoradi
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Roei Golan
- Departement of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL, USA
| | - Alexandra Dullea
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Ranjith Ramasamy
- Department of Urology, Miller School of Medicine, University of Miami, Miami, FL, USA.
| |
Collapse
|
23
|
Extracellular Vesicles under Oxidative Stress Conditions: Biological Properties and Physiological Roles. Cells 2021; 10:cells10071763. [PMID: 34359933 PMCID: PMC8306565 DOI: 10.3390/cells10071763] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Under physio-pathological conditions, cells release membrane-surrounded structures named Extracellular Vesicles (EVs), which convey their molecular cargo to neighboring or distant cells influencing their metabolism. Besides their involvement in the intercellular communication, EVs might represent a tool used by cells to eliminate unnecessary/toxic material. Here, we revised the literature exploring the link between EVs and redox biology. The first proof of this link derives from evidence demonstrating that EVs from healthy cells protect target cells from oxidative insults through the transfer of antioxidants. Oxidative stress conditions influence the release and the molecular cargo of EVs that, in turn, modulate the redox status of target cells. Oxidative stress-related EVs exert both beneficial or harmful effects, as they can carry antioxidants or ROS-generating enzymes and oxidized molecules. As mediators of cell-to-cell communication, EVs are also implicated in the pathophysiology of oxidative stress-related diseases. The review found evidence that numerous studies speculated on the role of EVs in redox signaling and oxidative stress-related pathologies, but few of them unraveled molecular mechanisms behind this complex link. Thus, the purpose of this review is to report and discuss this evidence, highlighting that the analysis of the molecular content of oxidative stress-released EVs (reminiscent of the redox status of originating cells), is a starting point for the use of EVs as diagnostic and therapeutic tools in oxidative stress-related diseases.
Collapse
|
24
|
Peroxiredoxins-The Underrated Actors during Virus-Induced Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10060977. [PMID: 34207367 PMCID: PMC8234473 DOI: 10.3390/antiox10060977] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/09/2021] [Accepted: 06/15/2021] [Indexed: 12/19/2022] Open
Abstract
Enhanced production of reactive oxygen species (ROS) triggered by various stimuli, including viral infections, has attributed much attention in the past years. It has been shown that different viruses that cause acute or chronic diseases induce oxidative stress in infected cells and dysregulate antioxidant its antioxidant capacity. However, most studies focused on catalase and superoxide dismutases, whereas a family of peroxiredoxins (Prdx), the most effective peroxide scavengers, were given little or no attention. In the current review, we demonstrate that peroxiredoxins scavenge hydrogen and organic peroxides at their physiological concentrations at various cell compartments, unlike many other antioxidant enzymes, and discuss their recycling. We also provide data on the regulation of their expression by various transcription factors, as they can be compared with the imprint of viruses on transcriptional machinery. Next, we discuss the involvement of peroxiredoxins in transferring signals from ROS on specific proteins by promoting the oxidation of target cysteine groups, as well as briefly demonstrate evidence of nonenzymatic, chaperone, functions of Prdx. Finally, we give an account of the current state of research of peroxiredoxins for various viruses. These data clearly show that Prdx have not been given proper attention despite all the achievements in general redox biology.
Collapse
|
25
|
Martins-Marques T, Rodriguez-Sinovas A, Girao H. Cellular crosstalk in cardioprotection: Where and when do reactive oxygen species play a role? Free Radic Biol Med 2021; 169:397-409. [PMID: 33892116 DOI: 10.1016/j.freeradbiomed.2021.03.044] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 03/14/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022]
Abstract
A well-balanced intercellular communication between the different cells within the heart is vital for the maintenance of cardiac homeostasis and function. Despite remarkable advances on disease management and treatment, acute myocardial infarction remains the major cause of morbidity and mortality worldwide. Gold standard reperfusion strategies, namely primary percutaneous coronary intervention, are crucial to preserve heart function. However, reestablishment of blood flow and oxygen levels to the infarcted area are also associated with an accumulation of reactive oxygen species (ROS), leading to oxidative damage and cardiomyocyte death, a phenomenon termed myocardial reperfusion injury. In addition, ROS signaling has been demonstrated to regulate multiple biological pathways, including cell differentiation and intercellular communication. Given the importance of cell-cell crosstalk in the coordinated response after cell injury, in this review, we will discuss the impact of ROS in the different forms of inter- and intracellular communication, as well as the role of gap junctions, tunneling nanotubes and extracellular vesicles in the propagation of oxidative damage in cardiac diseases, particularly in the context of ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Tania Martins-Marques
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal
| | - Antonio Rodriguez-Sinovas
- Cardiovascular Diseases Research Group, Department of Cardiology, Vall D'Hebron Institut de Recerca (VHIR), Vall D'Hebron Hospital Universitari, Vall D'Hebron Barcelona Hospital Campus, Passeig Vall D'Hebron, 119-129, 08035, Barcelona, Spain; Departament de Medicina, Universitat Autònoma de Barcelona, 08193, Bellaterra, Spain; Centro de Investigación Biomédica en Red Sobre Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Henrique Girao
- Univ Coimbra, Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, Coimbra, Portugal; Univ Coimbra, Center for Innovative Biomedicine and Biotechnology (CIBB), Coimbra, Portugal; Clinical Academic Centre of Coimbra (CACC), Coimbra, Portugal.
| |
Collapse
|
26
|
Caserta S, Ghezzi P. Release of redox enzymes and micro-RNAs in extracellular vesicles, during infection and inflammation. Free Radic Biol Med 2021; 169:248-257. [PMID: 33862160 DOI: 10.1016/j.freeradbiomed.2021.04.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/27/2021] [Accepted: 04/08/2021] [Indexed: 12/11/2022]
Abstract
Many studies reported that redox enzymes, particularly thioredoxin and peroxiredoxin, can be released by cells and act as soluble mediators in immunity. Recently, it became clear that peroxiredoxins can be secreted via the exosome-release route, yet it remains unclear how this exactly happens and why. This review will first introduce briefly the possible redox states of protein cysteines and the role of redox enzymes in their regulation. We will then discuss the studies on the extracellular forms of some of these enzymes, their association with exosomes/extracellular vesicles and with exosome micro-RNAs (miRNAs)/mRNAs involved in oxidative processes, relevant in infection and inflammation.
Collapse
Affiliation(s)
- Stefano Caserta
- Department of Biomedical Sciences, Hardy Building, The University of Hull, Hull, HU6 7RX, United Kingdom
| | - Pietro Ghezzi
- Department of Clinical Experimental Medicine, Brighton & Sussex Medical School, Brighton, BN19RY, United Kingdom.
| |
Collapse
|
27
|
Kim H, Rhee SJ, Lee H, Han D, Lee TY, Kim M, Kim EY, Kwon JS, Shin H, Kim H, Ahn YM, Ha K. Identification of altered protein expression in major depressive disorder and bipolar disorder patients using liquid chromatography-tandem mass spectrometry. Psychiatry Res 2021; 299:113850. [PMID: 33711561 DOI: 10.1016/j.psychres.2021.113850] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 02/28/2021] [Indexed: 01/07/2023]
Abstract
Emerging high-throughput proteomic technologies have recently been considered as a powerful means of identifying substrates involved in mood disorders. We performed proteomic profiling using liquid chromatography-tandem mass spectrometry to identify dysregulated proteins in plasma samples of 42 and 45 patients with major depressive disorder (MDD) and bipolar disorder (BD), respectively, in comparison to 51 healthy controls (HCs). Fourteen and six proteins in MDD and BD patients, respectively, were differentially expressed compared to HCs, among which coagulation factor XIII A chain (F13A1), platelet basic protein (PPBP), platelet facor 4 (PF4), glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and thymosin beta-4 (TMSB4X) were altered in both disorders. For proteins dysregulated in both, except F13A1, higher fold changes were observed in MDD than in BD patients. These findings may help identify candidate biomarkers of mood disorders and elucidate their underlying pathophysiology and biochemical abnormalities.
Collapse
Affiliation(s)
- Hyeyoung Kim
- Department of Psychiatry, Inha University Hospital, Incheon, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sang Jin Rhee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyunju Lee
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Tae Young Lee
- Department of Neuropsychiatry, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Minah Kim
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eun Young Kim
- Department of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jun Soo Kwon
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea; Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul, Republic of Korea
| | - Hyunsuk Shin
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Hyeyoon Kim
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea; Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yong Min Ahn
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| | - Kyooseob Ha
- Department of Psychiatry, Seoul National University College of Medicine, Seoul, Republic of Korea; Department of Neuropsychiatry, Seoul National University Hospital, Seoul, Republic of Korea; Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Elko EA, Manuel AM, White S, Zito E, van der Vliet A, Anathy V, Janssen-Heininger YMW. Oxidation of peroxiredoxin-4 induces oligomerization and promotes interaction with proteins governing protein folding and endoplasmic reticulum stress. J Biol Chem 2021; 296:100665. [PMID: 33895140 PMCID: PMC8141880 DOI: 10.1016/j.jbc.2021.100665] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
Peroxiredoxins (PRDXs) catalyze the reduction of hydrogen peroxide (H2O2). PRDX4 is the only peroxiredoxin located within the endoplasmic reticulum (ER) and is the most highly expressed H2O2 scavenger in the ER. PRDX4 has emerged as an important player in numerous diseases, such as fibrosis and metabolic syndromes, and its overoxidation is a potential indicator of ER redox stress. It is unclear how overoxidation of PRDX4 governs its oligomerization state and interacting partners. Herein, we addressed these questions via nonreducing Western blots, mass spectrometry, and site-directed mutagenesis. We report that the oxidation of PRDX4 in lung epithelial cells treated with tertbutyl hydroperoxide caused a shift of PRDX4 from monomer/dimer to high molecular weight (HMW) species, which contain PRDX4 modified with sulfonic acid residues (PRDX4-SO3), as well as of a complement of ER-associated proteins, including protein disulfide isomerases important in protein folding, thioredoxin domain-containing protein 5, and heat shock protein A5, a key regulator of the ER stress response. Mutation of any of the four cysteines in PRDX4 altered the HMW species in response to tertbutyl hydroperoxide as well as the secretion of PRDX4. We also demonstrate that the expression of ER oxidoreductase 1 alpha, which generates H2O2 in the ER, increased PRDX4 HMW formation and secretion. These results suggest a link between SO3 modification in the formation of HMW PRDX4 complexes in cells, whereas the association of key regulators of ER homeostasis with HMW oxidized PRDX4 point to a putative role of PRDX4 in regulating ER stress responses.
Collapse
Affiliation(s)
- Evan A Elko
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sheryl White
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Ester Zito
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | | |
Collapse
|
29
|
Buckens H, Pirenne S, Achouri Y, Baldan J, Dahou H, Bouwens L, Lemaigre FP, Jacquemin P, Assi M. Peroxiredoxin-I Sustains Inflammation During Pancreatitis. Cell Mol Gastroenterol Hepatol 2021; 12:741-743. [PMID: 33813037 PMCID: PMC8267543 DOI: 10.1016/j.jcmgh.2021.03.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/23/2021] [Accepted: 03/24/2021] [Indexed: 01/27/2023]
Affiliation(s)
- Hortense Buckens
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Sophie Pirenne
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Younes Achouri
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium,Transgene Technology Platform, Université Catholique de Louvain, Brussels, Belgium
| | - Jonathan Baldan
- Cell Differentiation Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | - Hajar Dahou
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Luc Bouwens
- Cell Differentiation Laboratory, Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Patrick Jacquemin
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium,Correspondence Address correspondence to: Patrick Jacquemin, PhD and Mohamad Assi, PhD. de Duve Institute, 75 Avenue Hippocrate, 1200, Brussels, Belgium.
| | - Mohamad Assi
- de Duve Institute, Université Catholique de Louvain, Brussels, Belgium,Correspondence Address correspondence to: Patrick Jacquemin, PhD and Mohamad Assi, PhD. de Duve Institute, 75 Avenue Hippocrate, 1200, Brussels, Belgium.
| |
Collapse
|
30
|
Benáková Š, Holendová B, Plecitá-Hlavatá L. Redox Homeostasis in Pancreatic β-Cells: From Development to Failure. Antioxidants (Basel) 2021; 10:antiox10040526. [PMID: 33801681 PMCID: PMC8065646 DOI: 10.3390/antiox10040526] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/23/2021] [Accepted: 03/25/2021] [Indexed: 12/16/2022] Open
Abstract
Redox status is a key determinant in the fate of β-cell. These cells are not primarily detoxifying and thus do not possess extensive antioxidant defense machinery. However, they show a wide range of redox regulating proteins, such as peroxiredoxins, thioredoxins or thioredoxin reductases, etc., being functionally compartmentalized within the cells. They keep fragile redox homeostasis and serve as messengers and amplifiers of redox signaling. β-cells require proper redox signaling already in cell ontogenesis during the development of mature β-cells from their progenitors. We bring details about redox-regulated signaling pathways and transcription factors being essential for proper differentiation and maturation of functional β-cells and their proliferation and insulin expression/maturation. We briefly highlight the targets of redox signaling in the insulin secretory pathway and focus more on possible targets of extracellular redox signaling through secreted thioredoxin1 and thioredoxin reductase1. Tuned redox homeostasis can switch upon chronic pathological insults towards the dysfunction of β-cells and to glucose intolerance. These are characteristics of type 2 diabetes, which is often linked to chronic nutritional overload being nowadays a pandemic feature of lifestyle. Overcharged β-cell metabolism causes pressure on proteostasis in the endoplasmic reticulum, mainly due to increased demand on insulin synthesis, which establishes unfolded protein response and insulin misfolding along with excessive hydrogen peroxide production. This together with redox dysbalance in cytoplasm and mitochondria due to enhanced nutritional pressure impact β-cell redox homeostasis and establish prooxidative metabolism. This can further affect β-cell communication in pancreatic islets through gap junctions. In parallel, peripheral tissues losing insulin sensitivity and overall impairment of glucose tolerance and gut microbiota establish local proinflammatory signaling and later systemic metainflammation, i.e., low chronic inflammation prooxidative properties, which target β-cells leading to their dedifferentiation, dysfunction and eventually cell death.
Collapse
Affiliation(s)
- Štěpánka Benáková
- Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, 142 20 Prague 4, Czech Republic; (Š.B.); (B.H.)
- First Faculty of Medicine, Charles University, Katerinska 1660/32, 121 08 Prague, Czech Republic
| | - Blanka Holendová
- Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, 142 20 Prague 4, Czech Republic; (Š.B.); (B.H.)
| | - Lydie Plecitá-Hlavatá
- Department of Mitochondrial Physiology, Institute of Physiology, Czech Academy of Sciences, 142 20 Prague 4, Czech Republic; (Š.B.); (B.H.)
- Department of Mitochondrial Physiology, Czech Academy of Sciences, Videnska 1083, 142 20 Prague 4, Czech Republic
- Correspondence: ; Tel.: +420-296-442-285
| |
Collapse
|
31
|
Coagulation factor IX analysis in bioreactor cell culture supernatant predicts quality of the purified product. Commun Biol 2021; 4:390. [PMID: 33758337 PMCID: PMC7988164 DOI: 10.1038/s42003-021-01903-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Coagulation factor IX (FIX) is a complex post-translationally modified human serum glycoprotein and high-value biopharmaceutical. The quality of recombinant FIX (rFIX), especially complete γ-carboxylation, is critical for rFIX clinical efficacy. Bioreactor operating conditions can impact rFIX production and post-translational modifications (PTMs). With the goal of optimizing rFIX production, we developed a suite of Data Independent Acquisition Mass Spectrometry (DIA-MS) proteomics methods and used these to investigate rFIX yield, γ-carboxylation, other PTMs, and host cell proteins during bioreactor culture and after purification. We detail the dynamics of site-specific PTM occupancy and structure on rFIX during production, which correlated with the efficiency of purification and the quality of the purified product. We identified new PTMs in rFIX near the GLA domain which could impact rFIX GLA-dependent purification and function. Our workflows are applicable to other biologics and expression systems, and should aid in the optimization and quality control of upstream and downstream bioprocesses.
Collapse
|
32
|
Martinez-Banaclocha M. Proteomic Complexity in Parkinson's Disease: A Redox Signaling Perspective of the Pathophysiology and Progression. Neuroscience 2020; 453:287-300. [PMID: 33212217 DOI: 10.1016/j.neuroscience.2020.11.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/19/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a prevalent age-related neurodegenerative disorder that results in the progressive impairment of motor and cognitive functions. The majority of PD cases are sporadic, and only 5% of patients are associated with mutations in a few genes, which cause the early onset or familial PD. Environmental toxic substances and the individual genetic susceptibility play a role in sporadic cases, but despite significant efforts to treat and prevent the disease, the pathophysiological mechanisms leading to its onset and progress are not fully understood. In the last decade, genomic and proteomic studies have shown an increasing molecular complexity of sporadic PD, suggesting that a broad spectrum of biochemical pathways underlie its progression. Recent investigations and the literature review suggest the potential role of deregulation of the sensitive-cysteine proteome as a convergent pathogenic mechanism that may contribute to this complexity, opening new therapeutic opportunities.
Collapse
|
33
|
Thioredoxin 1 is upregulated in the bone and bone marrow following experimental myocardial infarction: evidence for a remote organ response. Histochem Cell Biol 2020; 155:89-99. [PMID: 33161477 PMCID: PMC7847876 DOI: 10.1007/s00418-020-01939-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/27/2020] [Indexed: 10/31/2022]
Abstract
Ischemia and reperfusion events, such as myocardial infarction (MI), are reported to induce remote organ damage severely compromising patient outcomes. Tissue survival and functional restoration relies on the activation of endogenous redox regulatory systems such as the oxidoreductases of the thioredoxin (Trx) family. Trxs and peroxiredoxins (Prxs) are essential for the redox regulation of protein thiol groups and for the reduction of hydrogen peroxide, respectively. Here, we determined whether experimental MI induces changes in Trxs and Prxs in the heart as well as in secondary organs. Levels and localization of Trx1, TrxR1, Trx2, Prx1, and Prx2 were analyzed in the femur, vertebrae, and kidneys of rats following MI or sham surgery. Trx1 levels were significantly increased in the heart (P = 0.0017) and femur (P < 0.0001) of MI animals. In the femur and lumbar vertebrae, Trx1 upregulation was detected in bone-lining cells, osteoblasts, megakaryocytes, and other hematopoietic cells. Serum levels of Trx1 increased significantly 2 days after MI compared to sham animals (P = 0.0085). Differential regulation of Trx1 in the bone was also detected by immunohistochemistry 1 month after MI. N-Acetyl-cysteine treatment over a period of 1 month induced a significant reduction of Trx1 levels in the bone of MI rats compared to sham and to MI vehicle. This study provides first evidence that MI induces remote organ upregulation of the redox protein Trx1 in the bone, as a response to ischemia-reperfusion injury in the heart.
Collapse
|
34
|
Hanschmann EM, Berndt C, Hecker C, Garn H, Bertrams W, Lillig CH, Hudemann C. Glutaredoxin 2 Reduces Asthma-Like Acute Airway Inflammation in Mice. Front Immunol 2020; 11:561724. [PMID: 33224135 PMCID: PMC7670054 DOI: 10.3389/fimmu.2020.561724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 10/12/2020] [Indexed: 01/06/2023] Open
Abstract
Endogenous redox systems not only counteract oxidative damage induced by high levels of hydroxyl radicals (OH·) under pathological conditions, but also shape redox signaling as a key player in the regulation of physiological processes. Second messengers like hydrogen peroxide and nitric oxide, as well as redox enzymes of the Thioredoxin (Trx) family, including Trxs, glutaredoxins (Grxs), and peroxiredoxins (Prxs) modulate reversible, oxidative modifications of proteins. Thereby redox regulation is part of various cellular processes such as the immune response and Trx proteins have been linked in different disorders including inflammatory diseases. Here, we have analyzed the protein distribution of representative oxidoreductases of the Trx fold protein family—Trx1, Grx1, Grx2, and Prx2—in a murine model of allergic asthma bronchiale, as well as their potential therapeutic impact on type-2 driven airway inflammation. Ovalbumin (OVA) sensitization and challenge using the type-2 prone Balb/c mouse strain resulted in increased levels of all investigated proteins in distinct cellular patterns. While concomitant treatment with Grx1 and Prx2 did not show any therapeutic impact on the outcome of the disease, Grx2 or Trx1 treatment before and during the OVA challenge phase displayed pronounced protective effects on the manifestation of allergic airway inflammation. Eosinophil numbers and the type-2 cytokine IL-5 were significantly reduced while lung function parameters profoundly improved. The number of macrophages in the bronchoalveolar lavage (BAL) did not change significantly, however, the release of nitric oxide that was linked to airway inflammation was successfully prevented by enzymatically active Grx2 ex vivo. The Grx2 Cys-X-X-Ser mutant that facilitates de-/glutathionylation, but does not catalyze dithiol/disulfide exchange lost the ability to protect from airway hyper reactivity and to decrease NO release by macrophages, however, it reduced the number of infiltrating immune cells and IL-5 release. Altogether, this study demonstrates that specific redox proteins and particular enzyme activities protect against inflammatory damage. During OVA-induced allergic airway inflammation, administration of Grx2 exerts beneficial and thus potentially therapeutic effects.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, Germany
| | - Holger Garn
- Biochemical Pharmacological Center (BPC)-Translational Inflammation Research Division, Philips Universität Marburg, Member of the German Center for Lung Research (DZL) and the Universities of Giessen and Marburg Lung Center (UGMLC), Marburg, Germany
| | - W Bertrams
- Institute for Lung Research, Universities of Giessen and Marburg Lung Center, Philipps-University Marburg, Member of the German Center for Lung Research (DZL), Marburg, Germany
| | - Christopher H Lillig
- Institute for Medical Biochemistry and Molecular Biology, University of Greifswald, Greifswald, Germany
| | - Christoph Hudemann
- Department of Dermatology and Allergology, Philipps Universität Marburg, Marburg, Germany.,Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
35
|
Lorenzen I, Eble JA, Hanschmann EM. Thiol switches in membrane proteins - Extracellular redox regulation in cell biology. Biol Chem 2020; 402:253-269. [PMID: 33108336 DOI: 10.1515/hsz-2020-0266] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/17/2020] [Indexed: 12/11/2022]
Abstract
Redox-mediated signal transduction depends on the enzymatic production of second messengers such as hydrogen peroxide, nitric oxide and hydrogen sulfite, as well as specific, reversible redox modifications of cysteine-residues in proteins. So-called thiol switches induce for instance conformational changes in specific proteins that regulate cellular pathways e.g., cell metabolism, proliferation, migration, gene expression and inflammation. Reduction, oxidation and disulfide isomerization are controlled by oxidoreductases of the thioredoxin family, including thioredoxins, glutaredoxins, peroxiredoxins and protein dsisulfide isomerases. These proteins are located in different cellular compartments, interact with substrates and catalyze specific reactions. Interestingly, some of these proteins are released by cells. Their extracellular functions and generally extracellular redox control have been widely underestimated. Here, we give an insight into extracellular redox signaling, extracellular thiol switches and their regulation by secreted oxidoreductases and thiol-isomerases, a topic whose importance has been scarcely studied so far, likely due to methodological limitations. We focus on the secreted redox proteins and characterized thiol switches in the ectodomains of membrane proteins, such as integrins and the metalloprotease ADAM17, which are among the best-characterized proteins and discuss their underlying mechanisms and biological implications.
Collapse
Affiliation(s)
- Inken Lorenzen
- Centre of Biochemistry and Molecular Biology, Structural Biology, Christian-Albrecht University of Kiel, Am Botanischen Garten 1-9, D-24118Kiel, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Waldeyerstr. 15, D-48149Münster, Germany
| | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich Heine University Düsseldorf, Life Science Center, Merowingerplatz 1a, D-40225Düsseldorf, Germany
| |
Collapse
|
36
|
Zhang Y, Deng Y, Yang X, Xue H, Lang Y. The Relationship Between Protein S-Nitrosylation and Human Diseases: A Review. Neurochem Res 2020; 45:2815-2827. [PMID: 32984933 DOI: 10.1007/s11064-020-03136-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/18/2020] [Accepted: 09/19/2020] [Indexed: 01/12/2023]
Abstract
S-nitrosylation (SNO) is a covalent post-translational oxidative modification. The reaction is the nitroso group (-NO) to a reactive cysteine thiol within a protein to form the SNO. In recent years, a variety of proteins in human body have been found to undergo thiol nitrosylation under specific conditions. Protein SNO, which is closely related to cardiovascular disease, Parkinson's syndrome, Alzheimer's disease and tumors, plays an important role in regulatory mechanism of protein function in both physiological and pathological pathways, such as in cellular homeostasis and metabolism. This review discusses possible molecular mechanisms protein SNO modification, such as the role of NO in vivo and the formation mechanism of SNO, with particular emphasis on mechanisms utilized by SNO to cause certain diseases of human. Importantly, the effect of SNO on diseases is multifaceted and multi-channel, and its critical value in vivo is not well defined. Intracellular redox environment is also a key factor affecting its level. Therefore, we should pay more attention to the equilibrium relationship between SNO and denitrosylation pathway in the future researches. These findings provide theoretical support for the improvement or treatment of diseases from the point of view of SNO.
Collapse
Affiliation(s)
- Yadi Zhang
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, No. 180 Wusidong Road, Baoding, 071002, People's Republic of China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, People's Republic of China
| | - Yuzhen Deng
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, No. 180 Wusidong Road, Baoding, 071002, People's Republic of China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, People's Republic of China
| | - Xiaoxi Yang
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, No. 180 Wusidong Road, Baoding, 071002, People's Republic of China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, People's Republic of China
| | - Hongmei Xue
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, No. 180 Wusidong Road, Baoding, 071002, People's Republic of China.,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, People's Republic of China
| | - Yumiao Lang
- Key Laboratory of Public Health Safety of Hebei Province, College of Public Health, Hebei University, No. 180 Wusidong Road, Baoding, 071002, People's Republic of China. .,Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, People's Republic of China.
| |
Collapse
|
37
|
Jeong SJ, Cho MJ, Ko NY, Kim S, Jung IH, Min JK, Lee SH, Park JG, Oh GT. Deficiency of peroxiredoxin 2 exacerbates angiotensin II-induced abdominal aortic aneurysm. Exp Mol Med 2020; 52:1587-1601. [PMID: 32929220 PMCID: PMC8080566 DOI: 10.1038/s12276-020-00498-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/21/2020] [Accepted: 06/29/2020] [Indexed: 01/02/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is an inflammatory vascular disease characterized by structural deterioration of the aorta caused by inflammation and oxidative stress, leading to aortic dilatation and rupture. Peroxiredoxin 2 (PRDX2), an antioxidant enzyme, has been reported as a potential negative regulator of inflammatory vascular diseases, and it has been identified as a protein that is increased in patients with ruptured AAA compared to patients with nonruptured AAA. In this study, we demonstrated that PRDX2 was a pivotal factor involved in the inhibition of AAA progression. PRDX2 levels were increased in AAA compared with those in normal aortas in both humans and mice. Ultrasound imaging revealed that the loss of PRDX2 accelerated the development of AAA in the early stages and increased AAA incidence in mice infused with angiotensin II (Ang II). Prdx2−/− mice infused with Ang II exhibited increased aortic dilatation and maximal aortic diameter without a change in blood pressure. Structural deterioration of the aortas from Prdx2−/− mice infused with Ang II was associated with increases in the degradation of elastin, oxidative stress, and intramural thrombi caused by microhemorrhages, immature neovessels, and the activation of matrix metalloproteinases compared to that observed in controls. Moreover, an increase in inflammatory responses, including the production of cell adhesion molecules and the accumulation of inflammatory cells and proinflammatory cytokines due to PRDX2 deficiency, accelerated Ang II-induced AAA progression. Our data confirm that PRDX2 plays a role as a negative regulator of the pathological process of AAA and suggest that increasing PRDX2 activity may be a novel strategy for the prevention and treatment of AAA. An enzyme with antioxidant properties may provide a biomarker and therapeutic agent to help treat abdominal aortic aneurysm (AAA). AAA involves the structural deterioration of the aorta through chronic inflammation and oxidative stress, and can trigger life-threatening artery rupture. An antioxidant enzyme called peroxiredoxin 2 (PRDX2) is increased in patients with ruptures, but whether its role in AAA is beneficial or detrimental is unclear. Goo Taeg Oh at the Ewha Womans University in Seoul, Jong-Gil Park at the Korea Research Institute of Bioscience and Biotechnology, Daejeon, South Korea, and co-workers examined the effect of PRDX2 on AAA progression. PRDX2 suppressed structural damage in mice, limiting artery dilation and protein degradation. Loss of PRDX2 accelerated AAA development. Measuring levels of PRDX2 may indicate AAA severity in patients, while boosting the enzyme could repair aortic damage.
Collapse
Affiliation(s)
- Se-Jin Jeong
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Ji Cho
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Na Young Ko
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - Sinai Kim
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea
| | - In-Hyuk Jung
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Republic of Korea
| | - Sang Hak Lee
- Division of Cardiology, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Korea.,Cardiovascular Research Institute, Yonsei University College of Medicine, Seoul, Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience & Biotechnology, Daejeon, Republic of Korea.
| | - Goo Taeg Oh
- Immune and Vascular Cell Network Research Center, National Creative Initiatives, Department of Life Sciences, Ewha Womans University, Seoul, Republic of Korea.
| |
Collapse
|
38
|
Finelli MJ. Redox Post-translational Modifications of Protein Thiols in Brain Aging and Neurodegenerative Conditions-Focus on S-Nitrosation. Front Aging Neurosci 2020; 12:254. [PMID: 33088270 PMCID: PMC7497228 DOI: 10.3389/fnagi.2020.00254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species and reactive nitrogen species (RONS) are by-products of aerobic metabolism. RONS trigger a signaling cascade that can be transduced through oxidation-reduction (redox)-based post-translational modifications (redox PTMs) of protein thiols. This redox signaling is essential for normal cellular physiology and coordinately regulates the function of redox-sensitive proteins. It plays a particularly important role in the brain, which is a major producer of RONS. Aberrant redox PTMs of protein thiols can impair protein function and are associated with several diseases. This mini review article aims to evaluate the role of redox PTMs of protein thiols, in particular S-nitrosation, in brain aging, and in neurodegenerative diseases. It also discusses the potential of using redox-based therapeutic approaches for neurodegenerative conditions.
Collapse
Affiliation(s)
- Mattéa J Finelli
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
39
|
Mullen L, Mengozzi M, Hanschmann EM, Alberts B, Ghezzi P. How the redox state regulates immunity. Free Radic Biol Med 2020; 157:3-14. [PMID: 31899344 DOI: 10.1016/j.freeradbiomed.2019.12.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022]
Abstract
Oxidative stress is defined as an imbalance between the levels of reactive oxygen species (ROS) and antioxidant defences. The view of oxidative stress as a cause of cell damage has evolved over the past few decades to a much more nuanced view of the role of oxidative changes in cell physiology. This is no more evident than in the field of immunity, where oxidative changes are now known to regulate many aspects of the immune response, and inflammatory pathways in particular. Our understanding of redox regulation of immunity now encompasses not only increases in reactive oxygen and nitrogen species, but also changes in the activities of oxidoreductase enzymes. These enzymes are important regulators of immune pathways both via changes in their redox activity, but also via other more recently identified cytokine-like functions. The emerging picture of redox regulation of immune pathways is one of increasing complexity and while therapeutic targeting of the redox environment to treat inflammatory disease is a possibility, any such strategy is likely to be more nuanced than simply inhibiting ROS production.
Collapse
Affiliation(s)
- Lisa Mullen
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | | | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany
| | - Ben Alberts
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Falmer, Brighton, UK.
| |
Collapse
|
40
|
Tanaka LY, Oliveira PVS, Laurindo FRM. Peri/Epicellular Thiol Oxidoreductases as Mediators of Extracellular Redox Signaling. Antioxid Redox Signal 2020; 33:280-307. [PMID: 31910038 DOI: 10.1089/ars.2019.8012] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Significance: Supracellular redox networks regulating cell-extracellular matrix (ECM) and organ system architecture merge with structural and functional (catalytic or allosteric) properties of disulfide bonds. This review addresses emerging evidence that exported thiol oxidoreductases (TORs), such as thioredoxin, protein disulfide isomerases (PDIs), quiescin sulfhydryl oxidases (QSOX)1, and peroxiredoxins, composing a peri/epicellular (pec)TOR pool, mediate relevant signaling. pecTOR functions depend mainly on kinetic and spatial regulation of thiol-disulfide exchange reactions governed by redox potentials, which are modulated by exported intracellular low-molecular-weight thiols, together conferring signal specificity. Recent Advances: pecTOR redox-modulates several targets including integrins, ECM proteins, surface molecules, and plasma components, although clear-cut documentation of direct effects is lacking in many cases. TOR catalytic pathways, displaying common patterns, culminate in substrate thiol reduction, oxidation, or isomerization. Peroxiredoxins act as redox/peroxide sensors, contrary to PDIs, which are likely substrate-targeted redox modulators. Emerging evidence suggests important pecTOR roles in patho(physio)logical processes, including blood coagulation, vascular remodeling, mechanosensing, endothelial function, immune responses, and inflammation. Critical Issues: Effects of pecPDIs supporting thrombosis/platelet activation have been well documented and reached the clinical arena. Roles of pecPDIA1 in vascular remodeling/mechanosensing are also emerging. Extracellular thioredoxin and pecPDIs redox-regulate immunoinflammation. Routes of TOR externalization remain elusive and appear to involve Golgi-independent routes. pecTORs are particularly accessible drug targets. Future Directions: Further understanding mechanisms of thiol redox reactions and developing assays for assessing pecTOR redox activities remain important research avenues. Also, addressing pecTORs as disease markers and achieving more efficient/specific drugs for pecTOR modulation are major perspectives for diagnostic/therapeutic improvements.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Percillia V S Oliveira
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
41
|
The GABARAP Co-Secretome Identified by APEX2-GABARAP Proximity Labelling of Extracellular Vesicles. Cells 2020; 9:cells9061468. [PMID: 32560054 PMCID: PMC7349886 DOI: 10.3390/cells9061468] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/05/2020] [Accepted: 06/12/2020] [Indexed: 01/07/2023] Open
Abstract
The autophagy-related ATG8 protein GABARAP has not only been shown to be involved in the cellular self-degradation process called autophagy but also fulfils functions in intracellular trafficking processes such as receptor transport to the plasma membrane. Notably, available mass spectrometry data suggest that GABARAP is also secreted into extracellular vesicles (EVs). Here, we confirm this finding by the immunoblotting of EVs isolated from cell culture supernatants and human blood serum using specific anti-GABARAP antibodies. To investigate the mechanism by which GABARAP is secreted, we applied proximity labelling, a method for studying the direct environment of a protein of interest in a confined cellular compartment. By expressing an engineered peroxidase (APEX2)-tagged variant of GABARAP—which, like endogenous GABARAP, was present in EVs prepared from HEK293 cells—we demonstrate the applicability of APEX2-based proximity labelling to EVs. The biotinylated protein pool which contains the APEX2-GABARAP co-secretome contained not only known GABARAP interaction partners but also proteins that were found in APEX2-GABARAP’s proximity inside of autophagosomes in an independent study. All in all, we not only introduce a versatile tool for co-secretome analysis in general but also uncover the first details about autophagy-based pathways as possible biogenesis mechanisms of GABARAP-containing EVs.
Collapse
|
42
|
Peroxiredoxin-1 aggravates lipopolysaccharide-induced septic shock via promoting inflammation. Biochem Biophys Res Commun 2020; 527:861-865. [PMID: 32423810 DOI: 10.1016/j.bbrc.2020.04.149] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 04/29/2020] [Indexed: 01/06/2023]
Abstract
Septic shock induced by lipopolysaccharide (LPS) is characterized by serious systemic inflammatory response and robust production of pro-inflammatory cytokines from activated macrophages. Damage-associated molecular patterns (DAMPs) secreted by activated macrophages are key contributors to septic shock. However, the current knowledge on those DAMPs that promote inflammatory response under LPS-induced septic shock remains poorly understood. Here, we report that Peroxiredoxin 1 (Prdx1) plays a detrimental role in LPS-induced septic shock. Intraperitoneal injection of LPS elicited a progressive course of septic shock in mice, which was characterized by significant lethality along with robust production of cytokines (IL-1β, IL-6 and TNF-α). Removal of Prdx1 strongly protected mice from LPS-induced death, and decreased IL-1β, IL-6 and TNF-α productions. Additionally, primary macrophages deficient in Prdx1 are less able to produce much more IL-1β, IL-6 and TNF-α. Collectively, we provide a demonstration for Prdx1 contributing to LPS-induced septic shock likely via promoting inflammation.
Collapse
|
43
|
Dhanasiri A, Chen X, Dahle D, Siriyappagouder P, Fæste CK, Fernandes JMO. Dietary inclusion of plant ingredients induces epigenetic changes in the intestine of zebrafish. Epigenetics 2020; 15:1035-1051. [PMID: 32223500 DOI: 10.1080/15592294.2020.1747777] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Epigenetic modifications, such as DNA methylation, can be regulated by nutrition and dietary factors. There has been a large increase in the use of sustainable plant-based protein sources in fish feed due to limitations of fishmeal resources, which are needed to sustain a rapidly growing aquaculture industry. With this major transition from marine ingredients to plant-based diets, fish are abruptly introduced to changes in dietary composition and exposed to a variety of phytochemicals, some of which known to cause epigenetic changes in mammals. However, the effect of plant ingredients on the epigenome of fish is barely understood. In the present study, the nutriepigenomic effects of the addition of pea, soy, and wheat gluten protein concentrate to aquafeeds were investigated using zebrafish as a model. A genome-wide analysis of DNA methylation patterns was performed by reduced representation bisulphite sequencing to examine global epigenetic alterations in the mid intestine after a 42-day feeding trial. We found that inclusion of 30% of wheat gluten, pea and soy protein concentrate in the diet induced epigenetic changes in the mid intestine of zebrafish. A large number of genes and intergenic regions were differentially methylated with plant-based diets. The genes concerned were related to immunity, NF-κB system, ubiquitin-proteasome pathway, MAPK pathway, and the antioxidant defence system. Epigenetic regulation of several biological processes, including neurogenesis, cell adhesion, response to stress and immunity was also observed. Ultimately, the observed epigenetic changes may enable zebrafish to rapidly regulate inflammation and maintain intestinal homoeostasis when fed plant protein-based diets.
Collapse
Affiliation(s)
- Anusha Dhanasiri
- Faculty of Biosciences and Aquaculture, Nord University , Bodø, Norway.,Department of Paraclinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences (NMBU) , Oslo, Norway
| | - Xianquan Chen
- Faculty of Biosciences and Aquaculture, Nord University , Bodø, Norway.,School of Life Sciences, Sun Yat-Sen University , Guangzhou, PR China
| | - Dalia Dahle
- Faculty of Biosciences and Aquaculture, Nord University , Bodø, Norway
| | | | - Christiane K Fæste
- Toxinology Research Group, Norwegian Veterinary Institute , Oslo, Norway
| | | |
Collapse
|
44
|
Anjo SI, Dos Santos PV, Rosado L, Baltazar G, Baldeiras I, Pires D, Gomes A, Januário C, Castelo-Branco M, Grãos M, Manadas B. A different vision of translational research in biomarker discovery: a pilot study on circulatory mitochondrial proteins as Parkinson's disease potential biomarkers. Transl Neurodegener 2020; 9:11. [PMID: 32266064 PMCID: PMC7118951 DOI: 10.1186/s40035-020-00188-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 03/11/2020] [Indexed: 12/15/2022] Open
Abstract
Background The identification of circulating biomarkers that closely correlate with Parkinson’s Disease (PD) has failed several times in the past. Nevertheless, in this pilot study, a translational approach was conducted, allowing the evaluation of the plasma levels of two mitochondrial-related proteins, whose combination leads to a robust model with potential diagnostic value to discriminate the PD patients from matched controls. Methods The proposed translational approach was initiated by the analysis of secretomes from cells cultured under control or well-defined oxidative stress conditions, followed by the identification of proteins related to PD pathologic mechanisms that were altered between the two states. This pipeline was further translated into the analysis of undepleted plasma samples from 28 control and 31 PD patients. Results From the secretome analysis, several mitochondria-related proteins were found to be differentially released between control and stress conditions and to be able to distinguish the two secretomes. Similarly, two mitochondrial-related proteins were found to be significantly changed in a PD cohort compared to matched controls. Moreover, a linear discriminant model with potential diagnostic value to discriminate PD patients was obtained using the combination of these two proteins. Both proteins are associated with apoptotic mitochondrial changes, which may correspond to potential indicators of cell death. Moreover, one of these proteins, the VPS35 protein, was reported in plasma for the first time, and its quantification was only possible due to its previous identification in the secretome analysis. Conclusions In this work, an adaptation of a translational pipeline for biomarker selection was presented and transposed to neurological diseases, in the present case Parkinson’s Disease. The novelty and success of this pilot study may arise from the combination of: i) a translational research pipeline, where plasma samples are interrogated using knowledge previously obtained from the evaluation of cells’ secretome under oxidative stress; ii) the combined used of statistical analysis and an informed selection of candidates based on their link with relevant disease mechanisms, and iii) the use of SWATH-MS, an untargeted MS method that allows a complete record of the analyzed samples and a targeted data extraction of the quantitative values of proteins previously identified.
Collapse
Affiliation(s)
- Sandra I Anjo
- 1CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,2Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Luiza Rosado
- 4Centro Hospitalar Cova da Beira, E.P.E, Covilhã, Portugal
| | - Graça Baltazar
- 3Centro de Investigação em Ciências da Saúde (CICS-UBI), Universidade da Beira Interior, Covilhã, Portugal
| | - Inês Baldeiras
- 1CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,2Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,5Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Diana Pires
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
| | - Andreia Gomes
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
| | - Cristina Januário
- 5Neurology Department, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Miguel Castelo-Branco
- Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), Coimbra, Portugal
| | - Mário Grãos
- 1CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,7Biocant, Biotechnology Transfer Association, Cantanhede, Portugal.,8Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| | - Bruno Manadas
- 1CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,8Institute for Interdisciplinary Research, University of Coimbra (IIIUC), Coimbra, Portugal
| |
Collapse
|
45
|
Borras C, Mas-Bargues C, Sanz-Ros J, Román-Domínguez A, Gimeno-Mallench L, Inglés M, Gambini J, Viña J. Extracellular vesicles and redox modulation in aging. Free Radic Biol Med 2020; 149:44-50. [PMID: 31783096 DOI: 10.1016/j.freeradbiomed.2019.11.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are nowadays known to be mediators of cell-to-cell communication involved in physiological and pathological processes. The current expectation is their use as specific biomarkers and therapeutic tools due to their inner characteristics. However, several investigations still need to be done before we can use them in the clinic. First, their categorization is still under debate, although an accurate classification of EVs subtypes should be based on physical characteristics, biochemical composition or condition description of the cell of origin. Second, EVs carry lipids, proteins and nucleic acids that can induce epigenetic modifications on target cells. These cargos, as well as EVs biogenesis, shedding and uptake is both ageing and redox sensitive. More specifically, senescence and oxidative stress increase EVs release, and their altered content can trigger antioxidant but also prooxidant responses in target cells thereby modulating the redox status. Further analysis would help to asses EVs role in the development and progression of oxidative stress-related pathologies. In this review we aimed to summarize the current knowledge on EVs and their involvement in redox modulation on age-related pathologies. We also discuss future directions and prospective that could be performed to improve EVs usage as biomarkers or therapeutic tools.
Collapse
Affiliation(s)
- Consuelo Borras
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain.
| | - Cristina Mas-Bargues
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Jorge Sanz-Ros
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Aurora Román-Domínguez
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Lucia Gimeno-Mallench
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Marta Inglés
- Freshage Research Group. Department of Physiotherapy. Faculty of Physiotherapy. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Juan Gambini
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| | - Jose Viña
- Freshage Research Group. Department of Physiology. Faculty of Medicine. University of Valencia. CIBERFES. INCLIVA, Avenida Blasco Ibañez, 15, Valencia, Spain
| |
Collapse
|
46
|
ASK1 inhibition: a therapeutic strategy with multi-system benefits. J Mol Med (Berl) 2020; 98:335-348. [PMID: 32060587 PMCID: PMC7080683 DOI: 10.1007/s00109-020-01878-y] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 12/18/2019] [Accepted: 01/15/2020] [Indexed: 12/11/2022]
Abstract
p38 mitogen-activated protein kinases (P38α and β) and c-Jun N-terminal kinases (JNK1, 2, and 3) are key mediators of the cellular stress response. However, prolonged P38 and JNK signalling is associated with damaging inflammatory responses, reactive oxygen species-induced cell death, and fibrosis in multiple tissues, such as the kidney, liver, central nervous system, and cardiopulmonary systems. These responses are associated with many human diseases, including arthritis, dementia, and multiple organ dysfunctions. Attempts to prevent P38- and JNK-mediated disease using small molecule inhibitors of P38 or JNK have generally been unsuccessful. However, apoptosis signal-regulating kinase 1 (ASK1), an upstream regulator of P38 and JNK, has emerged as an alternative drug target for limiting P38- and JNK-mediated disease. Within this review, we compile the evidence that ASK1 mediates damaging cellular responses via prolonged P38 or JNK activation. We discuss the potential benefits of ASK1 inhibition as a therapeutic and summarise the studies that have tested the effects of ASK1 inhibition in cell and animal disease models, in addition to human clinical trials for a variety of disorders.
Collapse
|
47
|
Peroxiredoxin 5 Silencing Sensitizes Dopaminergic Neuronal Cells to Rotenone via DNA Damage-Triggered ATM/p53/PUMA Signaling-Mediated Apoptosis. Cells 2019; 9:cells9010022. [PMID: 31861721 PMCID: PMC7016837 DOI: 10.3390/cells9010022] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/11/2022] Open
Abstract
Peroxiredoxins (Prxs) are a family of thioredoxin peroxidases. Accumulating evidence suggests that changes in the expression of Prxs may be involved in neurodegenerative diseases pathology. However, the expression and function of Prxs in Parkinson’s disease (PD) remains unclear. Here, we showed that Prx5 was the most downregulated of the six Prx subtypes in dopaminergic (DA) neurons in rotenone-induced cellular and rat models of PD, suggesting possible roles in regulating their survival. Depletion of Prx5 sensitized SH-SY5Y DA neuronal cells to rotenone-induced apoptosis. The extent of mitochondrial membrane potential collapse, cytochrome c release, and caspase activation was increased by Prx5 loss. Furthermore, Prx5 knockdown enhanced the induction of PUMA by rotenone through a p53-dependent mechanism. Using RNA interference approaches, we demonstrated that the p53/PUMA signaling was essential for Prx5 silencing-exacerbated mitochondria-driven apoptosis. Additionally, downregulation of Prx5 augmented rotenone-induced DNA damage manifested as induction of phosphorylated histone H2AX (γ-H2AX) and activation of ataxia telangiectasia mutated (ATM) kinase. The pharmacological inactivation of ATM revealed that ATM was integral to p53 activation by DNA damage. These findings provided a novel link between Prx5 and DNA damage-triggered ATM/p53/PUMA signaling in a rotenone-induced PD model. Thus, Prx5 might play an important role in protection against rotenone-induced DA neurodegeneration.
Collapse
|
48
|
Ishida YI, Ichinowatari Y, Nishimoto S, Koike S, Ishii K, Ogasawara Y. Differential oxidation processes of peroxiredoxin 2 dependent on the reaction with several peroxides in human red blood cells. Biochem Biophys Res Commun 2019; 518:685-690. [PMID: 31472963 DOI: 10.1016/j.bbrc.2019.08.108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 08/20/2019] [Indexed: 11/18/2022]
Abstract
Peroxiredoxins (Prxs) detoxify hydrogen peroxide (H2O2), peroxynitrite, and various organic hydroperoxides. However, the differential oxidative status of Prxs reacted with each peroxide remains unclear. In the present study, we focused on the oxidative alteration of Prxs and demonstrated that, in human red blood cells (RBCs), peroxiredoxin 2 (Prx2) is readily reactive with H2O2, forming disulfide dimers, but was not easily hyperoxidized. In contrast, Prx2 was highly sensitive to the relatively hydrophobic oxidants, such as tert-butyl hydroperoxide (t-BHP) and cumene hydroperoxide. These peroxides hyperoxidized Prx2 into oxidatively damaged forms in RBCs. The t-BHP treatment formed hyperoxidized Prx2 in a dose-dependent manner. When organic hydroperoxide-treated RBC lysates were subjected to reverse-phase high performance liquid chromatography, two peaks derived from hyperoxidized Prx2 appeared along with the decrease of that corresponding to native Prx2. Liquid chromatography-tandem mass spectrometry analysis clearly showed that hyperoxidation to sulfonic acid (-SO3H) at Cys-51 residue was more advanced in a newfound hyperoxidized Prx2 compared to another hydrophobic hyperoxidized form previously identified. These results indicate that irreversible hyperoxidation of the Prx2 monomer in RBCs was easily caused by organic hydroperoxide but not H2O2. Thus, it is important to detect the hyperoxidation of Prx2 into sulfinic or sulfonic acid derivates of Cys-51 because hyperoxidized Prx2 is a potential marker of oxidative injury caused by organic hydroperoxides in human RBCs.
Collapse
Affiliation(s)
- Yo-Ichi Ishida
- Departments of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yuko Ichinowatari
- Departments of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Shoichi Nishimoto
- Departments of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Shin Koike
- Departments of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Kazuyuki Ishii
- Departments of Hygienic Chemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan
| | - Yuki Ogasawara
- Departments of Analytical Biochemistry, Meiji Pharmaceutical University, 2-522-1 Noshio, Kiyose, Tokyo, 204-8588, Japan.
| |
Collapse
|
49
|
Abruzzo PM, Matté A, Bolotta A, Federti E, Ghezzo A, Guarnieri T, Marini M, Posar A, Siciliano A, De Franceschi L, Visconti P. Plasma peroxiredoxin changes and inflammatory cytokines support the involvement of neuro-inflammation and oxidative stress in Autism Spectrum Disorder. J Transl Med 2019; 17:332. [PMID: 31578139 PMCID: PMC6775664 DOI: 10.1186/s12967-019-2076-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 09/21/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND It has been established that children with Autism Spectrum Disorders (ASD) are affected by oxidative stress, the origin of which is still under investigation. In the present work, we evaluated inflammatory and pro-oxidant soluble signature in non-syndromic ASD and age-matched typically developing (TD) control children. METHODS We analyzed leukocyte gene expression of inflammatory cytokines and inflammation/oxidative-stress related molecules in 21 ASD and 20 TD children. Moreover, in another-comparable-group of non-syndromic ASD (N = 22) and TD (N = 21) children, we analyzed for the first time the protein expression of the four members of the antioxidant enzyme family of peroxiredoxins (Prx) in both erythrocyte membranes and in plasma. RESULTS The gene expression of IL6 and of HSP70i, a stress protein, was increased in ASD children. Moreover, gene expression of many inflammatory cytokines and inflammation/oxidative stress-related proteins correlated with clinical features, and appeared to be linked by a complex network of inter-correlations involving the Aryl Hydrocarbon Receptor signaling pathway. In addition, when the study of inter-correlations within the expression pattern of these molecules was extended to include the healthy subjects, the intrinsic physiological relationships of the inflammatory/oxidative stress network emerged. Plasma levels of Prx2 and Prx5 were remarkably increased in ASD compared to healthy controls, while no significant differences were found in red cell Prx levels. CONCLUSIONS Previous findings reported elevated inflammatory cytokines in the plasma of ASD children, without clearly pointing to the presence of neuro-inflammation. On the other hand, the finding of microglia activation in autoptic specimens was clearly suggesting the presence of neuro-inflammation in ASD. Given the role of peroxiredoxins in the protection of brain cells against oxidative stress, the whole of our results, using peripheral data collected in living patients, support the involvement of neuro-inflammation in ASD, and generate a rational for neuro-inflammation as a possible therapeutic target and for plasma Prx5 as a novel indicator of ASD severity.
Collapse
Affiliation(s)
- P M Abruzzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy
| | - A Matté
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - A Bolotta
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy.,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy
| | - E Federti
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - A Ghezzo
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy
| | - T Guarnieri
- Department of Biological, Geological and Environmental Sciences, University of Bologna, Bologna, Italy
| | - M Marini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy. .,IRCCS Fondazione Don Carlo Gnocchi, Via A. Capecelatro, 66, 20148, Milan, Italy.
| | - A Posar
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Ugo Foscolo 7, 40123, Bologna, Italy.,Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura, 3, 40139, Bologna, Italy
| | - A Siciliano
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - L De Franceschi
- Department of Medicine, University of Verona Medical School, Verona, Italy
| | - P Visconti
- Child Neurology and Psychiatry Unit, IRCCS Istituto delle Scienze Neurologiche di Bologna, Via Altura, 3, 40139, Bologna, Italy
| |
Collapse
|
50
|
Ingram S, Mengozzi M, Heikal L, Mullen L, Ghezzi P. Inflammation-induced reactive nitrogen species cause proteasomal degradation of dimeric peroxiredoxin-1 in a mouse macrophage cell line. Free Radic Res 2019; 53:875-881. [DOI: 10.1080/10715762.2019.1637863] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Sonia Ingram
- Brighton & Sussex Medical School, Trafford Centre, Brighton, UK
| | | | - Lamia Heikal
- Brighton & Sussex Medical School, Trafford Centre, Brighton, UK
| | - Lisa Mullen
- Brighton & Sussex Medical School, Trafford Centre, Brighton, UK
| | - Pietro Ghezzi
- Brighton & Sussex Medical School, Trafford Centre, Brighton, UK
| |
Collapse
|