1
|
Azzouz D, Palaniyar N. How Do ROS Induce NETosis? Oxidative DNA Damage, DNA Repair, and Chromatin Decondensation. Biomolecules 2024; 14:1307. [PMID: 39456240 PMCID: PMC11505619 DOI: 10.3390/biom14101307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024] Open
Abstract
Neutrophil extracellular traps (NETs) are intricate, DNA-based, web-like structures adorned with cytotoxic proteins. They play a crucial role in antimicrobial defense but are also implicated in autoimmune diseases and tissue injury. The process of NET formation, known as NETosis, is a regulated cell death mechanism that involves the release of these structures and is unique to neutrophils. NETosis is heavily dependent on the production of reactive oxygen species (ROS), which can be generated either through NADPH oxidase (NOX) or mitochondrial pathways, leading to NOX-dependent or NOX-independent NETosis, respectively. Recent research has revealed an intricate interplay between ROS production, DNA repair, and NET formation in different contexts. UV radiation can trigger a combined process of NETosis and apoptosis, known as apoNETosis, driven by mitochondrial ROS and DNA repair. Similarly, in calcium ionophore-induced NETosis, both ROS and DNA repair are key components, but only play a partial role. In the case of bacterial infections, the early stages of DNA repair are pivotal. Interestingly, in serum-free conditions, spontaneous NETosis occurs through NOX-derived ROS, with early-stage DNA repair inhibition halting the process, while late-stage inhibition increases it. The intricate balance between DNA repair processes and ROS production appears to be a critical factor in regulating NET formation, with different pathways being activated depending on the nature of the stimulus. These findings not only deepen our understanding of the mechanisms behind NETosis but also suggest potential therapeutic targets for conditions where NETs contribute to disease pathology.
Collapse
Affiliation(s)
- Dhia Azzouz
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
2
|
Kwan A, Mcdermott-Brown I, Muthana M. Proliferating Cell Nuclear Antigen in the Era of Oncolytic Virotherapy. Viruses 2024; 16:1264. [PMID: 39205238 PMCID: PMC11359830 DOI: 10.3390/v16081264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/01/2024] [Accepted: 07/22/2024] [Indexed: 09/04/2024] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is a well-documented accessory protein of DNA repair and replication. It belongs to the sliding clamp family of proteins that encircle DNA and acts as a mobile docking platform for interacting proteins to mount and perform their metabolic tasks. PCNA presence is ubiquitous to all cells, and when located in the nucleus it plays a role in DNA replication and repair, cell cycle control and apoptosis in proliferating cells. It also plays a crucial role in the infectivity of some viruses, such as herpes simplex viruses (HSVs). However, more recently it has been found in the cytoplasm of immune cells such as neutrophils and macrophages where it has been shown to be involved in the development of a pro-inflammatory state. PCNA is also expressed on the surface of certain cancer cells and can play a role in preventing immune cells from killing tumours, as well as being associated with cancer virulence. Given the growing interest in oncolytic viruses (OVs) as a novel cancer therapeutic, this review considers the role of PCNA in healthy, cancerous, and immune cells to gain an understanding of how PCNA targeted therapy and oncolytic virotherapy may interact in the future.
Collapse
Affiliation(s)
| | | | - Munitta Muthana
- Medical School, University of Sheffield, Beech Hill Road, Sheffield S10 2RX, UK; (A.K.)
| |
Collapse
|
3
|
Liu D, Zuo R, Liu W, He Y, Wang Y, Yue P, Gong W, Cui J, Zhu F, Luo Y, Qi L, Guo Y, Chen L, Li G, Liu Z, Chen P, Guo H. DNAJC24 acts directly with PCNA and promotes malignant progression of LUAD by activating phosphorylation of AKT. FASEB J 2024; 38:e23630. [PMID: 38713100 DOI: 10.1096/fj.202300667rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 03/09/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
Heat shock proteins (HSPs) are a group of highly conserved proteins found in a wide range of organisms. In recent years, members of the HSP family were overexpressed in various tumors and widely involved in oncogenesis, tumor development, and therapeutic resistance. In our previous study, DNAJC24, a member of the DNAJ/HSP40 family of HSPs, was found to be closely associated with the malignant phenotype of hepatocellular carcinoma. However, its relationship with other malignancies needs to be further explored. Herein, we demonstrated that DNAJC24 exhibited upregulated expression in LUAD tissue samples and predicted poor survival in LUAD patients. The upregulation of DNAJC24 expression promoted proliferation and invasion of LUAD cells in A549 and NCI-H1299 cell lines. Further studies revealed that DNAJC24 could regulate the PI3K/AKT signaling pathway by affecting AKT phosphorylation. In addition, a series of experiments such as Co-IP and mass spectrometry confirmed that DNAJC24 could directly interact with PCNA and promoted the malignant phenotypic transformation of LUAD. In conclusion, our results suggested that DNAJC24 played an important role in the progression of LUAD and may serve as a specific prognostic biomarker for LUAD patients. The DNAJC24/PCNA/AKT axis may be a potential target for future individualized and precise treatment of LUAD patients.
Collapse
Affiliation(s)
- Dongming Liu
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Ran Zuo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wei Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yuchao He
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yu Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ping Yue
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Wenchen Gong
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jinfang Cui
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Fuyi Zhu
- Department of Oncology Surgery, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yi Luo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Lisha Qi
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Yan Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Cancer Biobank of Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Liwei Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Guangtao Li
- Department of Hepatobiliary Cancer, Liver Cancer Research Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Zhiyong Liu
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Peng Chen
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Thoracic Oncology, LUAD Diagnosis and Treatment Center, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hua Guo
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin, China
- Department of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| |
Collapse
|
4
|
Søgaard CK, Otterlei M. Targeting proliferating cell nuclear antigen (PCNA) for cancer therapy. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2024; 100:209-246. [PMID: 39034053 DOI: 10.1016/bs.apha.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Proliferating cell nuclear antigen (PCNA) is an essential scaffold protein in many cellular processes. It is best known for its role as a DNA sliding clamp and processivity factor during DNA replication, which has been extensively reviewed by others. However, the importance of PCNA extends beyond its DNA-associated functions in DNA replication, chromatin remodelling, DNA repair and DNA damage tolerance (DDT), as new non-canonical roles of PCNA in the cytosol have recently been identified. These include roles in the regulation of immune evasion, apoptosis, metabolism, and cellular signalling. The diverse roles of PCNA are largely mediated by its myriad protein interactions, and its centrality to cellular processes makes PCNA a valid therapeutic anticancer target. PCNA is expressed in all cells and plays an essential role in normal cellular homeostasis; therefore, the main challenge in targeting PCNA is to selectively kill cancer cells while avoiding unacceptable toxicity to healthy cells. This chapter focuses on the stress-related roles of PCNA, and how targeting these PCNA roles can be exploited in cancer therapy.
Collapse
Affiliation(s)
- Caroline K Søgaard
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway; APIM Therapeutics A/S, Trondheim, Norway.
| |
Collapse
|
5
|
Ferrão Maciel-Fiuza M, Rengel BD, Wachholz GE, do Amaral Gomes J, de Oliveira MR, Kowalski TW, Roehe PM, Luiz Vianna FS, Schüler-Faccini L, Mayer FQ, Varela APM, Fraga LR. New candidate genes potentially involved in Zika virus teratogenesis. Comput Biol Med 2024; 173:108259. [PMID: 38522248 DOI: 10.1016/j.compbiomed.2024.108259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 02/15/2024] [Accepted: 03/06/2024] [Indexed: 03/26/2024]
Abstract
Despite efforts to elucidate Zika virus (ZIKV) teratogenesis, still several issues remain unresolved, particularly on the molecular mechanisms behind the pathogenesis of Congenital Zika Syndrome (CZS). To answer this question, we used bioinformatics tools, animal experiments and human gene expression analysis to investigate genes related to brain development potentially involved in CZS. Searches in databases for genes related to brain development and CZS were performed, and a protein interaction network was created. The expression of these genes was analyzed in a CZS animal model and secondary gene expression analysis (DGE) was performed in human cells exposed to ZIKV. A total of 2610 genes were identified in the databases, of which 1013 were connected. By applying centrality statistics of the global network, 36 candidate genes were identified, which, after selection resulted in nine genes. Gene expression analysis revealed distinctive expression patterns for PRKDC, PCNA, ATM, SMC3 as well as for FGF8 and SHH in the CZS model. Furthermore, DGE analysis altered expression of ATM, PRKDC, PCNA. In conclusion, systems biology are helpful tools to identify candidate genes to be validated in vitro and in vivo. PRKDC, PCNA, ATM, SMC3, FGF8 and SHH have altered expression in ZIKV-induced brain malformations.
Collapse
Affiliation(s)
- Miriãn Ferrão Maciel-Fiuza
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Bruna Duarte Rengel
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Gabriela Elis Wachholz
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Julia do Amaral Gomes
- Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Maikel Rosa de Oliveira
- Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Thayne Woycinck Kowalski
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Bioinformatics Core, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Centro Universitário CESUCA, Cachoeirinha, Brazil
| | - Paulo Michel Roehe
- Department of Microbiology, Immunology and Parasitology, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Sales Luiz Vianna
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Lavínia Schüler-Faccini
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Instituto Nacional de Genética Médica Populacional, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Fabiana Quoos Mayer
- Graduate Program in Molecular and Cellular Biology, Biotechnology Center, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Ana Paula Muterle Varela
- Graduate Program in Biosciences, Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil.
| | - Lucas Rosa Fraga
- Genomics Medicine Laboratory, Center of Experimental Research, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil; Department of Morphological Sciences, Institute of Health Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Graduate Program in Medicine: Medical Sciences, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil; Teratogen Information System, Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.
| |
Collapse
|
6
|
Allen LAH. PCNA at the crossroads of human neutrophil activation, metabolism, and survival. J Leukoc Biol 2024; 115:201-204. [PMID: 38057160 DOI: 10.1093/jleuko/qiad153] [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: 10/30/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/08/2023] Open
Abstract
The proliferating cell nuclear antigen scaffold differentially binds hexokinase, procaspase-9, and p47phox to regulate neutrophil metabolism, viability and activation state.
Collapse
Affiliation(s)
- Lee-Ann H Allen
- Department of Molecular Microbiology and Immunology, One Hospital Drive, M616B Medical Sciences Building, University of Missouri, Columbia, MO 65212, United States
- Research Division, Harry S. Truman Memorial Veterans' Hospital, 800 Hospital Drive, Columbia, MO 65201, United States
| |
Collapse
|
7
|
Aymonnier K, Bosetta E, Leborgne NGF, Ullmer A, Le Gall M, De Chiara A, Salnot V, Many S, Scapini P, Wicks I, Chatfield S, Martin KR, Witko-Sarsat V. G-CSF reshapes the cytosolic PCNA scaffold and modulates glycolysis in neutrophils. J Leukoc Biol 2024; 115:205-221. [PMID: 37824822 DOI: 10.1093/jleuko/qiad122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 08/22/2023] [Accepted: 09/18/2023] [Indexed: 10/14/2023] Open
Abstract
Cytosolic proliferating cell nuclear antigen (PCNA) is involved in neutrophil survival and function, in which it acts as a scaffold and associates with proteins involved in apoptosis, NADPH oxidase activation, cytoskeletal dynamics, and metabolism. While the PCNA interactome has been characterized in neutrophils under homeostatic conditions, less is known about neutrophil PCNA in pathophysiological contexts. Granulocyte colony-stimulating factor (G-CSF) is a cytokine produced in response to inflammatory stimuli that regulates many aspects of neutrophil biology. Here, we used isolated normal-density neutrophils from G-CSF-treated haemopoietic stem cell donors (GDs) as a model to understand the role of PCNA during inflammation. Proteomic analysis of the neutrophil cytosol revealed significant differences between GDs and healthy donors (HDs). PCNA was one of the most upregulated proteins in GDs, and the PCNA interactome was significantly different in GDs compared with HDs. Importantly, while PCNA associated with almost all enzymes involved in glycolysis in HDs, these associations were decreased in GDs. Functionally, neutrophils from GDs had a significant increase in glycolysis compared with HDs. Using p21 competitor peptides, we showed that PCNA negatively regulates neutrophil glycolysis in HDs but had no effect on GD neutrophils. These data demonstrate that G-CSF alters the PCNA scaffold, affecting interactions with key glycolytic enzymes, and thus regulates glycolysis, the main energy pathway utilized by neutrophils. By this selective control of glycolysis, PCNA can organize neutrophils functionality in parallel with other PCNA mechanisms of prolonged survival. PCNA may therefore be instrumental in the reprogramming that neutrophils undergo in inflammatory or tumoral settings.
Collapse
Affiliation(s)
- Karen Aymonnier
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Enzo Bosetta
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Nathan G F Leborgne
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Audrey Ullmer
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Morgane Le Gall
- Proteom'IC facility, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du Faubourg Saint Jacques, Paris F-75014, France
| | - Alessia De Chiara
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Virginie Salnot
- Proteom'IC facility, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du Faubourg Saint Jacques, Paris F-75014, France
| | - Souganya Many
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| | - Patrizia Scapini
- Department of General Pathology, University of Verona, Verona 37134, Italy
| | - Ian Wicks
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
- Department of Rheumatology, Royal Melbourne Hospital, Grattan Street, Parkville, Victoria 3050, Australia
| | - Simon Chatfield
- Department of Rheumatology, Royal Melbourne Hospital, Grattan Street, Parkville, Victoria 3050, Australia
| | - Katherine R Martin
- Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Grattan Street, Parkville, Victoria 3010, Australia
| | - Véronique Witko-Sarsat
- Department of Immunology, Institut Cochin, Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique Unité Mixte de Recherche 8104, Université Paris Cité, 27 rue du faubourg Saint Jacques, Paris F-75014, France
| |
Collapse
|
8
|
Azzouz D, Palaniyar N. Mitochondrial ROS and base excision repair steps leading to DNA nick formation drive ultraviolet induced-NETosis. Front Immunol 2023; 14:1198716. [PMID: 37350954 PMCID: PMC10282603 DOI: 10.3389/fimmu.2023.1198716] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Reactive oxygen species (ROS) is essential for neutrophil extracellular trap formation (NETosis), and generated either by NADPH oxidases (e.g., during infections) or mitochondria (e.g., sterile injury) in neutrophils. We recently showed that ultraviolet (UV) radiation, a sterile injury-inducing agent, dose-dependently induced mitochondrial ROS generation, and increasing levels of ROS shifted the neutrophil death from apoptosis to NETosis. Nevertheless, how ROS executes UV-induced NETosis is unknown. In this study, we first confirmed that UV doses used in our experiments generated mitochondrial ROS, and the inhibition of mitochondrial ROS suppressed NETosis (Mitosox, SYTOX, immunocytochemistry, imaging). Next, we showed that UV irradiation extensively oxidized DNA, by confocal imaging of 8-oxyguanine (8-oxoG) in NETs. Immunofluorescence microscopy further showed that a DNA repair protein, proliferating cell nuclear antigen, was widely distributed throughout the DNA, indicating that the DNA repair machinery was active throughout the genome during UV-induced NETosis. Inhibition of specific steps of base excision repair (BER) pathway showed that steps leading up to DNA nick formation, but not the later steps, suppressed UV-induced NETosis. In summary, this study shows that (i) high levels of mitochondrial ROS produced following UV irradiation induces extensive oxidative DNA damage, and (ii) early steps of the BER pathway leading to DNA nicking results in chromatin decondensation and NETosis. Collectively, these findings reveal how ROS induces NOX-independent NETosis, and also a novel biological mechanism for UV irradiation- and -mitochondrial ROS-mediated NETosis.
Collapse
Affiliation(s)
- Dhia Azzouz
- Translational Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Nades Palaniyar
- Translational Medicine, Peter Gilgan Center for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
9
|
de Oliveira Formiga R, Amaral FC, Souza CF, Mendes DAGB, Wanderley CWS, Lorenzini CB, Santos AA, Antônia J, Faria LF, Natale CC, Paula NM, Silva PCS, Fonseca FR, Aires L, Heck N, Starick MR, Queiroz‐Junior CM, Santos FRS, de Souza FRO, Costa VV, Barroso SPC, Morrot A, Van Weyenbergh J, Sordi R, Alisson‐Silva F, Cunha FQ, Rocha EL, Chollet‐Martin S, Hurtado‐Nedelec MM, Martin C, Burgel P, Mansur DS, Maurici R, Macauley MS, Báfica A, Witko‐Sarsat V, Spiller F. Neuraminidase is a host-directed approach to regulate neutrophil responses in sepsis and COVID-19. Br J Pharmacol 2023; 180:1460-1481. [PMID: 36526272 PMCID: PMC9877938 DOI: 10.1111/bph.16013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 07/29/2022] [Accepted: 08/16/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE Neutrophil overstimulation plays a crucial role in tissue damage during severe infections. Because pathogen-derived neuraminidase (NEU) stimulates neutrophils, we investigated whether host NEU can be targeted to regulate the neutrophil dysregulation observed in severe infections. EXPERIMENTAL APPROACH The effects of NEU inhibitors on lipopolysaccharide (LPS)-stimulated neutrophils from healthy donors or COVID-19 patients were determined by evaluating the shedding of surface sialic acids, cell activation, and reactive oxygen species (ROS) production. Re-analysis of single-cell RNA sequencing of respiratory tract samples from COVID-19 patients also was carried out. The effects of oseltamivir on sepsis and betacoronavirus-induced acute lung injury were evaluated in murine models. KEY RESULTS Oseltamivir and zanamivir constrained host NEU activity, surface sialic acid release, cell activation, and ROS production by LPS-activated human neutrophils. Mechanistically, LPS increased the interaction of NEU1 with matrix metalloproteinase 9 (MMP-9). Inhibition of MMP-9 prevented LPS-induced NEU activity and neutrophil response. In vivo, treatment with oseltamivir fine-tuned neutrophil migration and improved infection control as well as host survival in peritonitis and pneumonia sepsis. NEU1 also is highly expressed in neutrophils from COVID-19 patients, and treatment of whole-blood samples from these patients with either oseltamivir or zanamivir reduced neutrophil overactivation. Oseltamivir treatment of intranasally infected mice with the mouse hepatitis coronavirus 3 (MHV-3) decreased lung neutrophil infiltration, viral load, and tissue damage. CONCLUSION AND IMPLICATIONS These findings suggest that interplay of NEU1-MMP-9 induces neutrophil overactivation. In vivo, NEU may serve as a host-directed target to dampen neutrophil dysfunction during severe infections.
Collapse
Affiliation(s)
- Rodrigo de Oliveira Formiga
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Flávia C. Amaral
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Camila F. Souza
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Daniel A. G. B. Mendes
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Carlos W. S. Wanderley
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Cristina B. Lorenzini
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Adara A. Santos
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Juliana Antônia
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Lucas F. Faria
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Caio C. Natale
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicholas M. Paula
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Priscila C. S. Silva
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Fernanda R. Fonseca
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Luan Aires
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Nicoli Heck
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Márick R. Starick
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Celso M. Queiroz‐Junior
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Felipe R. S. Santos
- Department of Biochemistry and Immunology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Filipe R. O. de Souza
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Vivian V. Costa
- Department of Morphology, Institute of Biological SciencesFederal University of Minas GeraisBelo HorizonteBrazil
| | - Shana P. C. Barroso
- Molecular Biology Laboratory, Institute of Biomedical ResearchMarcilio Dias Naval Hospital, Navy of BrazilRio de JaneiroBrazil
| | - Alexandre Morrot
- Tuberculosis Research Laboratory, Faculty of MedicineFederal University of Rio de JaneiroRio de JaneiroBrazil
- Immunoparasitology LaboratoryOswaldo Cruz Foundation (FIOCRUZ)Rio de JaneiroBrazil
| | - Johan Van Weyenbergh
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Laboratory for Clinical and Epidemiological VirologyKU LeuvenLeuvenBelgium
| | - Regina Sordi
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Frederico Alisson‐Silva
- Department of Immunology, Paulo de Goes Institute of MicrobiologyFederal University of Rio de JaneiroRio de JaneiroBrazil
| | - Fernando Q. Cunha
- Department of Pharmacology, School of Medicine of Ribeirão PretoUniversity of São PauloRibeirão PretoBrazil
| | - Edroaldo L. Rocha
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Sylvie Chollet‐Martin
- INSERM UMR 996, ‘Infammation, Microbiome and Immunosurveillance’, Faculty of PharmacyUniversité Paris‐SaclayChâtenay‐MalabryFrance
| | | | - Clémence Martin
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Pierre‐Régis Burgel
- Université de Paris, Institut Cochin, INSERM U1016, CNRSParisFrance
- Department of PneumologyAP‐HP, Hôpital CochinParisFrance
| | - Daniel S. Mansur
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | - Rosemeri Maurici
- Department of Clinical MedicineFederal University of Santa CatarinaFlorianópolisBrazil
| | - Matthew S. Macauley
- Department of Chemistry, Department of Medical Microbiology and ImmunologyUniversity of AlbertaEdmontonAlbertaCanada
| | - André Báfica
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| | | | - Fernando Spiller
- Department of PharmacologyFederal University of Santa CatarinaFlorianópolisBrazil
- Laboratory of Immunobiology, Department of Microbiology, Immunology and ParasitologyFederal University of Santa CatarinaFlorianópolisBrazil
| |
Collapse
|
10
|
ATX-101, a cell-penetrating protein targeting PCNA, can be safely administered as intravenous infusion in patients and shows clinical activity in a Phase 1 study. Oncogene 2023; 42:541-544. [PMID: 36564469 PMCID: PMC9918429 DOI: 10.1038/s41388-022-02582-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
Proliferating Cell Nuclear Antigen (PCNA) is a highly conserved protein essential for DNA replication, repair and scaffold functions in the cytosol. Specific inhibition of PCNA in cancer cells is an attractive anti-cancer strategy. ATX-101 is a first-in-class drug targeting PCNA, primarily in cellular stress regulation. Multiple in vivo and in vitro investigations demonstrated anti-cancer activity of ATX-101 in many tumor types and a potentiating effect on the activity of anti-cancer therapies. Healthy cells were less affected. Based on preclinical data, a clinical phase 1 study was initiated. Twenty-five patients with progressive, late-stage solid tumors were treated with weekly ATX-101 infusions at four dose levels (20, 30, 45, 60 mg/m2). ATX-101 showed a favorable safety profile supporting that vital cellular functions are not compromised in healthy cells. Mild and moderate infusion-related reactions were observed in 64% of patients. ATX-101 was quickly cleared from blood with elimination half-lives of less than 30 min at all dose levels, probably due to both, a quick cell penetration and peptide digestion in serum, as demonstrated in vivo. No tumor responses were observed but stable disease was seen in 70% of the efficacy population (n = 20). Further studies have been initiated to provide evidence of efficacy. Trial registration numbers: ANZCTR 375262 and ANZCTR 375319.
Collapse
|
11
|
PCNA regulates primary metabolism by scaffolding metabolic enzymes. Oncogene 2023; 42:613-624. [PMID: 36564470 PMCID: PMC9937922 DOI: 10.1038/s41388-022-02579-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 12/08/2022] [Accepted: 12/13/2022] [Indexed: 12/24/2022]
Abstract
The essential roles of proliferating cell nuclear antigen (PCNA) as a scaffold protein in DNA replication and repair are well established, while its cytosolic roles are less explored. Two metabolic enzymes, alpha-enolase (ENO1) and 6-phosphogluconate dehydrogenase (6PGD), both contain PCNA interacting motifs. Mutation of the PCNA interacting motif APIM in ENO1 (F423A) impaired its binding to PCNA and resulted in reduced cellular levels of ENO1 protein, reduced growth rate, reduced glucose consumption, and reduced activation of AKT. Metabolome and signalome analysis reveal large consequences of impairing the direct interaction between PCNA and ENO1. Metabolites above ENO1 in glycolysis accumulated while lower glycolytic and TCA cycle metabolite pools decreased in the APIM-mutated cells; however, their overall energetic status were similar to parental cells. Treating haematological cancer cells or activated primary monocytes with a PCNA targeting peptide drug containing APIM (ATX-101) also lead to a metabolic shift characterized by reduced glycolytic rate. In addition, we show that ATX-101 treatments reduced the ENO1 - PCNA interaction, the ENO1, GAPDH and 6PGD protein levels, as well as the 6PGD activity. Here we report for the first time that PCNA acts as a scaffold for metabolic enzymes, and thereby act as a direct regulator of primary metabolism.
Collapse
|
12
|
Bölükbaş F, Öznurlu Y. Determining the effects of in ovo administration of monosodium glutamate on the embryonic development of brain in chickens. Neurotoxicology 2023; 94:87-97. [PMID: 36400230 DOI: 10.1016/j.neuro.2022.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 10/24/2022] [Accepted: 11/14/2022] [Indexed: 11/17/2022]
Abstract
Monosodium glutamate (MSG) is a popular flavor enhancer largely used in the food industry. Although numerous studies have reported the neurotoxic effects of MSG on humans and animals, there is limited information about how it affects embryonic brain development. Thus, this study aimed to determine the effects of in ovo administered MSG on embryonic brain development in chickens. For this purpose, 410 fertilized chicken eggs were divided into 5 groups as control, distilled water, 0.12, 0.6 and 1.2 mg/g egg MSG, and injections were performed via the egg yolk. On days 15, 18, and 21 of the incubation period, brain tissue samples were taken from all embryos and chicks. The mortality rates of MSG-treated groups were significantly higher than those of the control and distilled water groups. The MSG-treated groups showed embryonic growth retardation and various structural abnormalities such as abdominal hernia, unilateral anophthalmia, hemorrhage, brain malformation, and the curling of legs and fingers. The relative embryo and body weights of the MSG-treated groups were significantly lower than those of the control group on incubation days 18 and 21. Histopathological evaluations revealed that MSG caused histopathological changes such as necrosis, neuronophagia, and gliosis in brain on incubation days 15, 18, and 21. There was a significant increase in the number of necrotic neurons in the MSG-treated groups compared to the control and distilled water groups in the hyperpallium, optic tectum and hippocampus regions. Proliferating cell nuclear antigen (PCNA) positive cells in brain were found in the hyperpallium, optic tectum, and hippocampus regions; there were more PCNA(+) immunoreactive cells in MSG-treated groups than in control and distilled water groups. In conclusion, it was determined that in ovo MSG administered could adversely affect embryonic growth and development in addition to causing necrosis in the neurons in the developing brain.
Collapse
Affiliation(s)
- Ferhan Bölükbaş
- Department of Histology and Embryology, Faculty of Medicine, Aksaray University, Aksaray, Turkey.
| | - Yasemin Öznurlu
- Department of Histology and Embryology, Faculty of Veterinary Medicine, University of Selcuk, Konya, Turkey
| |
Collapse
|
13
|
Azzouz D, Palaniyar N. ROS and DNA repair in spontaneous versus agonist-induced NETosis: Context matters. Front Immunol 2022; 13:1033815. [PMID: 36426351 PMCID: PMC9679651 DOI: 10.3389/fimmu.2022.1033815] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/19/2022] [Indexed: 04/14/2024] Open
Abstract
Reactive oxygen species (ROS) is essential for neutrophil extracellular trap formation (NETosis). Nevertheless, how ROS induces NETosis at baseline and during neutrophil activation is unknown. Although neutrophils carry DNA transcription, replication and repair machineries, their relevance in the short-lived mature neutrophils that carry pre-synthesized proteins has remained a mystery for decades. Our recent studies show that (i) NETosis-inducing agonists promote NETosis-specific kinase activation, genome-wide transcription that helps to decondense chromatin, and (ii) excess ROS produced by NADPH oxidase activating agonists generate genome-wide 8-oxy-guanine (8-OG), and the initial steps of DNA repair are needed to decondense chromatin in these cells. These steps require DNA repair proteins necessary for the assembly and nicking at the damaged DNA sites (poly ADP ribose polymerase PARP, apurinic endonuclease APE1 and DNA ligase), but not the enzymes that mediate the repair DNA synthesis (Proliferating cell nuclear antigen (PCNA) and DNA Polymerases). In this study, we show that (i) similar to agonist-induced NETosis, inhibition of early steps of oxidative DNA damage repair proteins suppresses spontaneous NETosis, but (ii) the inhibition of late stage repair proteins DNA polymerases and PCNA drastically promotes baseline NETosis. Hence, in the absence of excessive ROS generation and neutrophil activation, DNA repair mediated by PCNA and DNA polymerases is essential to prevent chromatin decondensation and spontaneous NETosis. These findings indicate that ROS, oxidative DNA damage, transcription and DNA repair differentially regulate spontaneous and agonist-induced NETosis. Therefore, context matters.
Collapse
Affiliation(s)
- Dhia Azzouz
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Nades Palaniyar
- Program in Translational Medicine, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, Canada
- Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Institute of Medical Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
14
|
Mulye M, Singh MI, Jain V. From Processivity to Genome Maintenance: The Many Roles of Sliding Clamps. Genes (Basel) 2022; 13:2058. [PMID: 36360296 PMCID: PMC9690074 DOI: 10.3390/genes13112058] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 07/30/2023] Open
Abstract
Sliding clamps play a pivotal role in the process of replication by increasing the processivity of the replicative polymerase. They also serve as an interacting platform for a plethora of other proteins, which have an important role in other DNA metabolic processes, including DNA repair. In other words, clamps have evolved, as has been correctly referred to, into a mobile "tool-belt" on the DNA, and provide a platform for several proteins that are involved in maintaining genome integrity. Because of the central role played by the sliding clamp in various processes, its study becomes essential and relevant in understanding these processes and exploring the protein as an important drug target. In this review, we provide an updated report on the functioning, interactions, and moonlighting roles of the sliding clamps in various organisms and its utilization as a drug target.
Collapse
Affiliation(s)
- Meenakshi Mulye
- Correspondence: (M.M.); (V.J.); Tel.: +91-755-269-1425 (V.J.); Fax: +91-755-269-2392 (V.J.)
| | | | - Vikas Jain
- Correspondence: (M.M.); (V.J.); Tel.: +91-755-269-1425 (V.J.); Fax: +91-755-269-2392 (V.J.)
| |
Collapse
|
15
|
Matouk AI, El-Daly M, Habib HA, Senousy S, Naguib Abdel Hafez SM, Kasem AW, Almalki WH, Alzahrani A, Alshehri A, Ahmed ASF. Protective effects of menthol against sepsis-induced hepatic injury: Role of mediators of hepatic inflammation, apoptosis, and regeneration. Front Pharmacol 2022; 13:952337. [PMID: 36120368 PMCID: PMC9476320 DOI: 10.3389/fphar.2022.952337] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Liver dysfunction in sepsis is a major complication that amplifies multiple organ failure and increases the risk of death. Inflammation and oxidative stress are the main mediators in the pathophysiology of sepsis. Therefore, we investigated the role of menthol, a natural antioxidant, against sepsis-induced liver injury in female Wistar rats. Sepsis was induced by cecal ligation and puncture (CLP). Menthol (100 mg/kg) was given intragastric 2 h after CLP. Blood samples and liver tissues were collected 24 h after surgery. Menthol significantly (p < 0.05) attenuated the sepsis-induced elevation in serum liver enzymes and improved the hepatic histopathological changes. Menthol treatment significantly (p < 0.05) decreased hepatic levels of tumor necrosis factor-alpha, malondialdehyde, total nitrite, and cleaved caspase-3. It restored the hepatic levels of superoxide dismutase and reduced glutathione. Additionally, menthol significantly (p < 0.05) increased hepatic levels of B-cell lymphoma 2 (Bcl-2); an anti-apoptotic factor, and proliferating cell nuclear antigen (PCNA), a biomarker of regeneration and survival. Our results showed the therapeutic potential of menthol against liver injury induced by sepsis.
Collapse
Affiliation(s)
- Asmaa I. Matouk
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Heba A. Habib
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Shaymaa Senousy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | | | - AlShaimaa W. Kasem
- Department of Histology and Cell Biology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Abdulaziz Alzahrani
- Department of Pharmacology and Toxicology, College of Clinical Pharmacy, AlBaha University, Al Bahah, Saudi Arabia
| | - Ahmed Alshehri
- Department of Pharmacology and Toxicology, College of Clinical Pharmacy, AlBaha University, Al Bahah, Saudi Arabia
| | - Al-Shaimaa F. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
- *Correspondence: Al-Shaimaa F. Ahmed,
| |
Collapse
|
16
|
Anter A, Ahmed ASF, Hammad ASA, Almalki WH, Abdel Hafez SMN, Kasem AW, El-Moselhy MA, Alrabia MW, Ibrahim ARN, El-Daly M. The Severity of Acute Kidney and Lung Injuries Induced by Cecal Ligation and Puncture Is Attenuated by Menthol: Role of Proliferating Cell Nuclear Antigen and Apoptotic Markers. Front Med (Lausanne) 2022; 9:904286. [PMID: 35814769 PMCID: PMC9260148 DOI: 10.3389/fmed.2022.904286] [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: 03/25/2022] [Accepted: 05/16/2022] [Indexed: 11/24/2022] Open
Abstract
Objective Sepsis-induced acute lung injury (ALI) and acute kidney injury (AKI) are major causes of mortality. Menthol is a natural compound that has anti-inflammatory and antioxidative actions. Since exaggerated inflammatory and oxidative stress are characteristics of sepsis, the aim of this study was to evaluate the effect of menthol against sepsis-induced mortality, ALI, and AKI. Methods The cecal ligation and puncture (CLP) procedure was employed as a model of sepsis. Rats were grouped into sham, sham-Menthol, CLP, and CLP-Menthol (100 mg/kg, p.o). Key Findings A survival study showed that menthol enhanced the survival after sepsis from 0% in septic group to 30%. Septic rats developed histological evidence of ALI and AKI. Menthol markedly suppressed sepsis induced elevation of tissue TNF-a, ameliorated sepsis-induced cleavage of caspase-3 and restored the antiapoptotic marker Bcl2. Significance We introduced a role of the proliferating cell nuclear antigen (PCNA) in these tissues with a possible link to the damage induced by sepsis. PCNA level was markedly reduced in septic animals and menthol ameliorated this effect. Our data provide novel evidence that menthol protects against organ damage and decreases mortality in experimental sepsis.
Collapse
Affiliation(s)
- Aliaa Anter
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Al-Shaimaa F. Ahmed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
- *Correspondence: Al-Shaimaa F. Ahmed,
| | - Asmaa S. A. Hammad
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - AlShaimaa W. Kasem
- Department of Pathology, Faculty of Medicine, Minia University, Minya, Egypt
| | - Mohamed A. El-Moselhy
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
- Department of Clinical Pharmacy and Pharmacology, Ibn Sina National College for Medical Studies, Jeddah, Saudi Arabia
| | - Mohammad W. Alrabia
- Department of Microbiology and Medical Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed R. N. Ibrahim
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minya, Egypt
| | - Mahmoud El-Daly
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Minia University, Minya, Egypt
| |
Collapse
|
17
|
Zambalde ÉP, Pavan ICB, Mancini MCS, Severino MB, Scudero OB, Morelli AP, Amorim MR, Bispo-dos-Santos K, Góis MM, Toledo-Teixeira DA, Parise PL, Mauad T, Dolhnikoff M, Saldiva PHN, Marques-Souza H, Proenca-Modena JL, Ventura AM, Simabuco FM. Characterization of the Interaction Between SARS-CoV-2 Membrane Protein (M) and Proliferating Cell Nuclear Antigen (PCNA) as a Potential Therapeutic Target. Front Cell Infect Microbiol 2022; 12:849017. [PMID: 35677658 PMCID: PMC9168989 DOI: 10.3389/fcimb.2022.849017] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 04/25/2022] [Indexed: 12/21/2022] Open
Abstract
SARS-CoV-2 is an emerging virus from the Coronaviridae family and is responsible for the ongoing COVID-19 pandemic. In this work, we explored the previously reported SARS-CoV-2 structural membrane protein (M) interaction with human Proliferating Cell Nuclear Antigen (PCNA). The M protein is responsible for maintaining virion shape, and PCNA is a marker of DNA damage which is essential for DNA replication and repair. We validated the M-PCNA interaction through immunoprecipitation, immunofluorescence co-localization, and PLA (Proximity Ligation Assay). In cells infected with SARS-CoV-2 or transfected with M protein, using immunofluorescence and cell fractioning, we documented a reallocation of PCNA from the nucleus to the cytoplasm and the increase of PCNA and γH2AX (another DNA damage marker) expression. We also observed an increase in PCNA and γH2AX expression in the lung of a COVID-19 patient by immunohistochemistry. In addition, the inhibition of PCNA translocation by PCNA I1 and Verdinexor led to a reduction of plaque formation in an in vitro assay. We, therefore, propose that the transport of PCNA to the cytoplasm and its association with M could be a virus strategy to manipulate cell functions and may be considered a target for COVID-19 therapy.
Collapse
Affiliation(s)
- Érika Pereira Zambalde
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Isadora Carolina Betim Pavan
- Laboratory of Signaling Mechanisms, School of Pharmaceutical Sciences, University of Campinas, (Unicamp), Campinas, Brazil
| | - Mariana Camargo Silva Mancini
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Matheus Brandemarte Severino
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Orlando Bonito Scudero
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Ana Paula Morelli
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Mariene Ribeiro Amorim
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Karina Bispo-dos-Santos
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Mariana Marcela Góis
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| | - Daniel A. Toledo-Teixeira
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Pierina Lorencini Parise
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Thais Mauad
- São Paulo University Medical School, Department of Pathology, University of São Paulo (USP), São Paulo, Brazil
| | - Marisa Dolhnikoff
- São Paulo University Medical School, Department of Pathology, University of São Paulo (USP), São Paulo, Brazil
| | | | | | - José Luiz Proenca-Modena
- Laboratory of Emerging Viruses (LEVE), Department of Genetics, Evolution, Microbiology and Immunology, Institute of Biology, University of Campinas (Unicamp), Campinas, SP, Brazil
- Experimental Medicine Research Cluster, University of Campinas (Unicamp), Campinas, Brazil
- Hub of Global Health (HGH), University of Campinas (Unicamp), Campinas, Brazil
| | - Armando Morais Ventura
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health, School of Applied Sciences, University of Campinas (Unicamp), Limeira, Brazil
| |
Collapse
|
18
|
Filep JG. Targeting Neutrophils for Promoting the Resolution of Inflammation. Front Immunol 2022; 13:866747. [PMID: 35371088 PMCID: PMC8966391 DOI: 10.3389/fimmu.2022.866747] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 02/21/2022] [Indexed: 12/15/2022] Open
Abstract
Acute inflammation is a localized and self-limited innate host-defense mechanism against invading pathogens and tissue injury. Neutrophils, the most abundant immune cells in humans, play pivotal roles in host defense by eradicating invading pathogens and debris. Ideally, elimination of the offending insult prompts repair and return to homeostasis. However, the neutrophils` powerful weaponry to combat microbes can also cause tissue damage and neutrophil-driven inflammation is a unifying mechanism for many diseases. For timely resolution of inflammation, in addition to stopping neutrophil recruitment, emigrated neutrophils need to be disarmed and removed from the affected site. Accumulating evidence documents the phenotypic and functional versatility of neutrophils far beyond their antimicrobial functions. Hence, understanding the receptors that integrate opposing cues and checkpoints that determine the fate of neutrophils in inflamed tissues provides insight into the mechanisms that distinguish protective and dysregulated, excessive inflammation and govern resolution. This review aims to provide a brief overview and update with key points from recent advances on neutrophil heterogeneity, functional versatility and signaling, and discusses challenges and emerging therapeutic approaches that target neutrophils to enhance the resolution of inflammation.
Collapse
Affiliation(s)
- János G Filep
- Department of Pathology and Cell Biology, University of Montreal, Montreal, QC, Canada.,Research Center, Maisonneuve-Rosemont Hospital, Montreal, QC, Canada
| |
Collapse
|
19
|
Yang S, Xiao Y, Du Y, Chen J, Ni Q, Guo X, Xue G, Xie X. Diagnostic and Prognostic Value of Neutrophil Extracellular Trap Levels in Patients With Acute Aortic Dissection. Front Cardiovasc Med 2022; 8:683445. [PMID: 35242817 PMCID: PMC8885526 DOI: 10.3389/fcvm.2021.683445] [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: 03/21/2021] [Accepted: 09/10/2021] [Indexed: 11/13/2022] Open
Abstract
Background Acute aortic dissection (AAD) is a fatal disease demanding prompt diagnosis and proper treatment. There is a lack of serum markers that can effectively assist diagnosis and predict prognosis of AAD patients. Methods Ninety-six AAD patients were enrolled in this study, and 249 patients with chest pain due to acute myocardial infarction, pulmonary embolism, intramural hematoma, angina or other causes and 80 healthy controls were included as control group and healthy control group. Demographics, biochemical and hematological data and risk factors were recorded as baseline characteristics. The 1-year follow-up data were collected and analyzed. The diagnostic performance and ability to predict disease severity and prognosis of NET components in serum and aortic tissue were evaluated. Results Circulating NET markers, citH3 (citrullination of histone 3), cell-free DNA (cfDNA) and nucleosomes, had good diagnostic value for AAD, with superior diagnostic performance to D-dimer in discriminating patients with chest pain due to other reasons in the emergency department. Circulating NET marker levels (i.e., citH3, cfDNA and nucleosomes) of AAD patients were significantly higher than that of control group and healthy control group. In addition, circulating NET markers levels were closely associated with the disease severity, in-hospital death and 1-year survival of AAD patients. Systolic blood pressure < 90 mmHg and serum citH3 levels were identified as independent risk factors for 1-year survival of AAD patients. Excessive NET components (i.e., neutrophil elastase and citH3) in the aortic tissue of AAD patient were significantly higher than that of healthy donor aortic tissue. The expression levels of granules and nuclear NET components were significantly higher in aortic tissue from AAD patients than controls. Conclusions Circulating NET markers, citH3, cfDNA and nucleosomes, have significant diagnostic value and predictive value of disease severity and prognosis of AAD patients. The NETs components may constitute a useful diagnostic and prognostic marker in AAD patients.
Collapse
Affiliation(s)
- Shuofei Yang
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yongsheng Xiao
- Department of Vascular Surgery, Tianjin 4th Centre Hospital, The Fourth Central Hospital Affiliated to Nankai University, The Fourth Center Clinical College of Tianjin Medical University, Tianjin, China
| | - Yuanfeng Du
- Department of Neurosurgery, School of Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University, Hangzhou, China
| | - Jiaquan Chen
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Qihong Ni
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Xiangjiang Guo
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Guanhua Xue
- Department of Vascular Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
- *Correspondence: Guanhua Xue
| | - Xupin Xie
- Department of Vascular Surgery, School of Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University, Hangzhou, China
- Xupin Xie
| |
Collapse
|
20
|
Gravina GL, Colapietro A, Mancini A, Rossetti A, Martellucci S, Ventura L, Di Franco M, Marampon F, Mattei V, Biordi LA, Otterlei M, Festuccia C. ATX-101, a Peptide Targeting PCNA, Has Antitumor Efficacy Alone or in Combination with Radiotherapy in Murine Models of Human Glioblastoma. Cancers (Basel) 2022; 14:289. [PMID: 35053455 PMCID: PMC8773508 DOI: 10.3390/cancers14020289] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/31/2021] [Accepted: 01/03/2022] [Indexed: 02/01/2023] Open
Abstract
Cell proliferation requires the orchestrated actions of a myriad of proteins regulating DNA replication, DNA repair and damage tolerance, and cell cycle. Proliferating cell nuclear antigen (PCNA) is a master regulator which interacts with multiple proteins functioning in these processes, and this makes PCNA an attractive target in anticancer therapies. Here, we show that a cell-penetrating peptide containing the AlkB homolog 2 PCNA-interacting motif (APIM), ATX-101, has antitumor activity in a panel of human glioblastoma multiforme (GBM) cell lines and patient-derived glioma-initiating cells (GICs). Their sensitivity to ATX-101 was not related to cellular levels of PCNA, or p53, PTEN, or MGMT status. However, ATX-101 reduced Akt/mTOR and DNA-PKcs signaling, and a correlation between high Akt activation and sensitivity for ATX-101 was found. ATX-101 increased the levels of γH2AX, DNA fragmentation, and apoptosis when combined with radiotherapy (RT). In line with the in vitro results, ATX-101 strongly reduced tumor growth in two subcutaneous xenografts and two orthotopic GBM models, both as a single agent and in combination with RT. The ability of ATX-101 to sensitize cells to RT is promising for further development of this compound for use in GBM.
Collapse
Affiliation(s)
- Giovanni Luca Gravina
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiation Oncology, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Alessandro Colapietro
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.M.); (A.R.)
| | - Andrea Mancini
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.M.); (A.R.)
| | - Alessandra Rossetti
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.M.); (A.R.)
| | - Stefano Martellucci
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Cellular Pathology, University of L’Aquila, 67100 L’Aquila, Italy;
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, 02100 Rieti, Italy;
| | - Luca Ventura
- Division of Pathology, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.V.); (M.D.F.)
| | - Martina Di Franco
- Division of Pathology, San Salvatore Hospital, 67100 L’Aquila, Italy; (L.V.); (M.D.F.)
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, 00100 Rome, Italy;
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, 02100 Rieti, Italy;
| | - Leda Assunta Biordi
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Medical Oncology, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Marit Otterlei
- APIM Therapeutics A/S, N-7100 Rissa, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), N-7006 Trondheim, Norway
| | - Claudio Festuccia
- Department of Biotechnological and Applied Clinical Sciences, Laboratory of Radiobiology, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.M.); (A.R.)
| |
Collapse
|
21
|
Martin KR, Wong HL, Witko-Sarsat V, Wicks IP. G-CSF - A double edge sword in neutrophil mediated immunity. Semin Immunol 2021; 54:101516. [PMID: 34728120 DOI: 10.1016/j.smim.2021.101516] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 10/23/2021] [Indexed: 11/15/2022]
Abstract
Neutrophils are vital for the innate immune system's control of pathogens and neutrophil deficiency can render the host susceptible to life-threatening infections. Neutrophil responses must also be tightly regulated because excessive production, recruitment or activation of neutrophils can cause tissue damage in both acute and chronic inflammatory diseases. Granulocyte colony stimulating factor (G-CSF) is a key regulator of neutrophil biology, from production, differentiation, and release of neutrophil precursors in the bone marrow (BM) to modulating the function of mature neutrophils outside of the BM, particularly at sites of inflammation. G-CSF acts by binding to its cognate cell surface receptor on target cells, causing the activation of intracellular signalling pathways mediating the proliferation, differentiation, function, and survival of cells in the neutrophil lineage. Studies in humans and mice demonstrate that G-CSF contributes to protecting the host against infection, but conversely, it can play a deleterious role in inflammatory diseases. As such, neutrophils and the G-CSF pathway may provide novel therapeutic targets. This review will focus on understanding the role G-CSF plays in the balance between effective neutrophil mediated host defence versus neutrophil-mediated inflammation and tissue damage in various inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Katherine R Martin
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Huon L Wong
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia
| | | | - Ian P Wicks
- WEHI, 1G Royal Parade, Parkville, Victoria, 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia.
| |
Collapse
|
22
|
Structural and Functional Alterations in Mitochondria-Associated Membranes (MAMs) and in Mitochondria Activate Stress Response Mechanisms in an In Vitro Model of Alzheimer's Disease. Biomedicines 2021; 9:biomedicines9080881. [PMID: 34440085 PMCID: PMC8389659 DOI: 10.3390/biomedicines9080881] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/21/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by the accumulation of extracellular plaques composed by amyloid-β (Aβ) and intracellular neurofibrillary tangles of hyperphosphorylated tau. AD-related neurodegenerative mechanisms involve early changes of mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) and impairment of cellular events modulated by these subcellular domains. In this study, we characterized the structural and functional alterations at MAM, mitochondria, and ER/microsomes in a mouse neuroblastoma cell line (N2A) overexpressing the human amyloid precursor protein (APP) with the familial Swedish mutation (APPswe). Proteins levels were determined by Western blot, ER-mitochondria contacts were quantified by transmission electron microscopy, and Ca2+ homeostasis and mitochondria function were analyzed using fluorescent probes and Seahorse assays. In this in vitro AD model, we found APP accumulated in MAM and mitochondria, and altered levels of proteins implicated in ER-mitochondria tethering, Ca2+ signaling, mitochondrial dynamics, biogenesis and protein import, as well as in the stress response. Moreover, we observed a decreased number of close ER-mitochondria contacts, activation of the ER unfolded protein response, reduced Ca2+ transfer from ER to mitochondria, and impaired mitochondrial function. Together, these results demonstrate that several subcellular alterations occur in AD-like neuronal cells, which supports that the defective ER-mitochondria crosstalk is an important player in AD physiopathology.
Collapse
|
23
|
ROS induces NETosis by oxidizing DNA and initiating DNA repair. Cell Death Discov 2021; 7:113. [PMID: 34001856 PMCID: PMC8128883 DOI: 10.1038/s41420-021-00491-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 03/22/2021] [Accepted: 04/23/2021] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are essential for neutrophil extracellular trap (NET) formation or NETosis. Nevertheless, how ROS induces NETosis is unknown. Neutrophil activation induces excess ROS production and a meaningless genome-wide transcription to facilitate chromatin decondensation. Here we show that the induction of NADPH oxidase-dependent NETosis leads to extensive DNA damage, and the subsequent translocation of proliferating cell nuclear antigen (PCNA), a key DNA repair protein, stored in the cytoplasm into the nucleus. During the activation of NETosis (e.g., by phorbol myristate acetate, Escherichia coli LPS, Staphylococcus aureus (RN4220), or Pseudomonas aeruginosa), preventing the DNA-repair-complex assembly leading to nick formation that decondenses chromatin causes the suppression of NETosis (e.g., by inhibitors to, or knockdown of, Apurinic endonuclease APE1, poly ADP ribose polymerase PARP, and DNA ligase). The remaining repair steps involving polymerase activity and PCNA interactions with DNA polymerases β/δ do not suppress agonist-induced NETosis. Therefore, excess ROS produced during neutrophil activation induces NETosis by inducing extensive DNA damage (e.g., oxidising guanine to 8-oxoguanine), and the subsequent DNA repair pathway, leading to chromatin decondensation.
Collapse
|
24
|
Babushkina NP, Postrigan AE, Kucher AN. Involvement of Variants in the Genes Encoding BRCA1-Associated Genome Surveillance Complex (BASC) in the Development of Human Common Diseases. Mol Biol 2021. [DOI: 10.1134/s0026893321020047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
25
|
Lima TS, Mallya S, Jankeel A, Messaoudi I, Lodoen MB. Toxoplasma gondii Extends the Life Span of Infected Human Neutrophils by Inducing Cytosolic PCNA and Blocking Activation of Apoptotic Caspases. mBio 2021; 12:e02031-20. [PMID: 33500339 PMCID: PMC7858050 DOI: 10.1128/mbio.02031-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 12/01/2020] [Indexed: 11/20/2022] Open
Abstract
Toxoplasma gondii is an intracellular protozoan parasite that has the remarkable ability to infect and replicate in neutrophils, immune cells with an arsenal of antimicrobial effector mechanisms. We report that T. gondii infection extends the life span of primary human peripheral blood neutrophils by delaying spontaneous apoptosis, serum starvation-induced apoptosis, and tumor necrosis alpha (TNF-α)-mediated apoptosis. T. gondii blockade of apoptosis was associated with an inhibition of processing and activation of the apoptotic caspases caspase-8 and -3, decreased phosphatidylserine exposure on the plasma membrane, and reduced cell death. We performed a global transcriptome analysis of T. gondii-infected peripheral blood neutrophils using RNA sequencing (RNA-Seq) and identified gene expression changes associated with DNA replication and DNA repair pathways, which in mature neutrophils are indicative of changes in regulators of cell survival. Consistent with the RNA-Seq data, T. gondii infection upregulated transcript and protein expression of PCNA, which is found in the cytosol of human neutrophils, where it functions as a key inhibitor of apoptotic pro-caspases. Infection of neutrophils resulted in increased interaction of PCNA with pro-caspase-3. Inhibition of this interaction with an AlkB homologue 2 PCNA-interacting motif (APIM) peptide reversed the infection-induced delay in cell death. Taken together, these findings indicate a novel strategy by which T. gondii manipulates cell life span in primary human neutrophils, which may allow the parasite to maintain an intracellular replicative niche and avoid immune clearance.IMPORTANCEToxoplasma gondii is an obligate intracellular parasite that can cause life-threatening disease in immunocompromised individuals and in the developing fetus. Interestingly, T. gondii has evolved strategies to successfully manipulate the host immune system to establish a productive infection and evade host defense mechanisms. Although it is well documented that neutrophils are mobilized during acute T. gondii infection and infiltrate the site of infection, these cells can also be actively infected by T. gondii and serve as a replicative niche for the parasite. However, there is a limited understanding of the molecular processes occurring within T. gondii-infected neutrophils. This study reveals that T. gondii extends the life span of human neutrophils by inducing the expression of PCNA, which prevents activation of apoptotic caspases, thus delaying apoptosis. This strategy may allow the parasite to preserve its replicative intracellular niche.
Collapse
Affiliation(s)
- Tatiane S Lima
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Sharmila Mallya
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Allen Jankeel
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| | - Melissa B Lodoen
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, California, USA
- Institute for Immunology, University of California, Irvine, Irvine, California, USA
| |
Collapse
|
26
|
Mulcahy ME, O'Brien EC, O'Keeffe KM, Vozza EG, Leddy N, McLoughlin RM. Manipulation of Autophagy and Apoptosis Facilitates Intracellular Survival of Staphylococcus aureus in Human Neutrophils. Front Immunol 2020; 11:565545. [PMID: 33262756 PMCID: PMC7686353 DOI: 10.3389/fimmu.2020.565545] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 10/15/2020] [Indexed: 01/13/2023] Open
Abstract
Polymorphonuclear neutrophils (PMN) are critical for first line innate immune defence against Staphylococcus aureus. Mature circulating PMN maintain a short half-life ending in constitutive apoptotic cell death. This makes them unlikely candidates as a bacterial intracellular niche. However, there is significant evidence to suggest that S. aureus can survive intracellularly within PMN and this contributes to persistence and dissemination during infection. The precise mechanism by which S. aureus parasitizes these cells remains to be established. Herein we propose a novel mechanism by which S. aureus subverts both autophagy and apoptosis in PMN in order to maintain an intracellular survival niche during infection. Intracellular survival of S. aureus within primary human PMN was associated with an accumulation of the autophagic flux markers LC3-II and p62, while inhibition of the autophagy pathway led to a significant reduction in intracellular survival of bacteria. This intracellular survival of S. aureus was coupled with a delay in neutrophil apoptosis as well as increased expression of several anti-apoptotic factors. Importantly, blocking autophagy in infected PMN partially restored levels of apoptosis to that of uninfected PMN, suggesting a connection between the autophagic and apoptotic pathways during intracellular survival. These results provide a novel mechanism for S. aureus intracellular survival and suggest that S. aureus may be subverting crosstalk between the autophagic and apoptosis pathways in order to maintain an intracellular niche within human PMN.
Collapse
Affiliation(s)
- Michelle E Mulcahy
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Eóin C O'Brien
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Kate M O'Keeffe
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Emilio G Vozza
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Neal Leddy
- bioTEM, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Rachel M McLoughlin
- Host-Pathogen Interactions Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
27
|
Cardano M, Tribioli C, Prosperi E. Targeting Proliferating Cell Nuclear Antigen (PCNA) as an Effective Strategy to Inhibit Tumor Cell Proliferation. Curr Cancer Drug Targets 2020; 20:240-252. [PMID: 31951183 DOI: 10.2174/1568009620666200115162814] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 12/12/2019] [Accepted: 12/18/2019] [Indexed: 12/20/2022]
Abstract
Targeting highly proliferating cells is an important issue for many types of aggressive tumors. Proliferating Cell Nuclear Antigen (PCNA) is an essential protein that participates in a variety of processes of DNA metabolism, including DNA replication and repair, chromatin organization and transcription and sister chromatid cohesion. In addition, PCNA is involved in cell survival, and possibly in pathways of energy metabolism, such as glycolysis. Thus, the possibility of targeting this protein for chemotherapy against highly proliferating malignancies is under active investigation. Currently, approaches to treat cells with agents targeting PCNA rely on the use of small molecules or on peptides that either bind to PCNA, or act as a competitor of interacting partners. Here, we describe the status of the art in the development of agents targeting PCNA and discuss their application in different types of tumor cell lines and in animal model systems.
Collapse
Affiliation(s)
- Miriana Cardano
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Carla Tribioli
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| | - Ennio Prosperi
- Istituto di Genetica Molecolare del C.N.R. "Luca Cavalli-Sforza", Pavia- 27100, Italy
| |
Collapse
|
28
|
Wu L, Saxena S, Goel P, Prajapati DR, Wang C, Singh RK. Breast Cancer Cell-Neutrophil Interactions Enhance Neutrophil Survival and Pro-Tumorigenic Activities. Cancers (Basel) 2020; 12:E2884. [PMID: 33049964 PMCID: PMC7599756 DOI: 10.3390/cancers12102884] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 10/02/2020] [Accepted: 10/05/2020] [Indexed: 12/14/2022] Open
Abstract
Breast cancer remains the most prevalent cancer in women with limited treatment options for patients suffering from therapy-resistance and metastatic disease. Neutrophils play an important role in breast cancer progression and metastasis. We examined the pro-tumorigenic nature of the breast cancer cell-neutrophil interactions and delineated the differences in neutrophil properties between the chemotherapy-resistant and the parent tumor microenvironment. Our data demonstrated that high neutrophil infiltration is associated with disease aggressiveness and therapy resistance. In the human breast cancer dataset, expression of neutrophil-related signature gene expression was higher in tumors from therapy-resistant patients than therapy-sensitive patients. We observed that breast cancer-derived factors significantly enhanced neutrophil survival, polarization, and pro-inflammatory cytokine expression. Breast cancer cell-derived supernatant treated neutrophils significantly expressed high levels of interleukin-1β (IL-1β), CC-chemokine ligand-2-4 (CCL2, CCL3, CCL4), inducible nitric oxide synthase (iNOS), and matrix metallopeptidase-9 (MMP9), and formed extracellular traps (NETs). Moreover, neutrophils showed increased secretion of MMP9 when cultured with the supernatant of chemotherapy-resistant Cl66-Doxorubicin (Cl66-Dox) and Cl66-Paclitaxel (Cl66-Pac) cells in comparison with the supernatant of Cl66-parent cells. Together, these data suggest an important role of breast cancer cell-neutrophil interactions in regulating pro-tumor characteristics in neutrophils and its modulation by therapy resistance.
Collapse
Affiliation(s)
- Lingyun Wu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
- Vascular Biology and Therapeutics Program and Department of Pharmacology, Yale University, Yale School of Medicine, New Haven, CT 06520-8089, USA
| | - Sugandha Saxena
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
| | - Paran Goel
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
| | - Dipakkumar R. Prajapati
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
| | - Cheng Wang
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
| | - Rakesh K. Singh
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 UNMC, Omaha, NE 68198-5900, USA; (L.W.); (S.S.); (P.G.); (D.R.P.); (C.W.)
| |
Collapse
|
29
|
Tang D, Liu X, Chen K, Li Z, Dai Y, Xu J, Zhang HT, Gao X, Liu L. Cytoplasmic PCNA is located in the actin belt and involved in osteoclast differentiation. Aging (Albany NY) 2020; 12:13297-13317. [PMID: 32597793 PMCID: PMC7377826 DOI: 10.18632/aging.103434] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 05/25/2020] [Indexed: 12/18/2022]
Abstract
Osteoporosis (OP) is an age-related osteolytic disease and characterized by low bone mass and more prone to fracture due to active osteoclasts. Proliferating cell nuclear antigen (PCNA) has been long identified as a nuclear protein playing critical roles in the regulation of DNA replication and repair. Recently, a few studies have demonstrated the cytoplasmic localization of PCNA and its function associated with apoptosis in neutrophil and neuroblastoma cells. However, the involvement of PCNA, including the cytoplasmic PCNA, in the osteoclast differentiation remains unclear. In the present study, we show that PCNA is translocated from nucleus to cytoplasm during the RANKL-induced osteoclast differentiation, and localized in the actin belt of mature osteoclast. Knockdown of PCNA significantly affected the integrity of actin belt, the formation of multinucleated osteoclasts, the expression of osteoclast-specific genes, and the in vitro bone resorption. Interactomic study has revealed β-actin as the major interacting partner of the cytoplasmic PCNA, suggesting that cytoplasmic PCNA might play a critical role in the differentiation of osteoclast through regulation of actin-cytoskeleton remodeling. Taken together, our results demonstrate the critical role of cytoplasmic PCNA during the process of osteoclast differentiation, and provided a potential therapeutic target for treatment of osteoclast-related bone diseases.
Collapse
Affiliation(s)
- Donge Tang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China.,Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Xiaohui Liu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Kezhi Chen
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Zhipeng Li
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Yong Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital Southern University of Science and Technology, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth 6009, Western Australia, Australia
| | - Huan-Tian Zhang
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China.,Institute of Orthopedic Diseases and Department of Bone and Joint Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510630, Guangdong, China
| | - Xuejuan Gao
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and MOE Key Laboratory of Tumor Molecular Biology, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
30
|
O Bortolazzo F, D Lucke L, de Oliveira Fujii L, Marqueti RDC, Vieira Ramos G, Theodoro V, Bombeiro AL, Felonato M, A Dalia R, D Carneiro G, Pontes Vicente C, A M Esquisatto M, A S Mendonça F, T Dos Santos GM, R Pimentel E, de Aro AA. Microcurrent and adipose-derived stem cells modulate genes expression involved in the structural recovery of transected tendon of rats. FASEB J 2020; 34:10011-10026. [PMID: 32558993 DOI: 10.1096/fj.201902942rr] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/02/2020] [Accepted: 05/05/2020] [Indexed: 01/30/2023]
Abstract
Tendon injuries are common and have a high incidence of re-rupture that can cause loss of functionality. Therapies with adipose-derived stem cells (ASC) and the microcurrent (low-intensity electrical stimulation) application present promising effects on the tissue repair. We analyzed the expression of genes and the participation of some molecules potentially involved in the structural recovery of the Achilles tendon of rats, in response to the application of both therapies, isolated and combined. The tendons were distributed in five groups: normal (N), transected (T), transected and ASC (C) or microcurrent (M) or with ASC, and microcurrent (MC). Microcurrent therapy was beneficial for tendon repair, as it was observed a statistically significant increase in the organization of the collagen fibers, with involvement of the TNC, CTGF, FN, FMDO, and COL3A1 genes as well as PCNA, IL-10, and TNF-α. ASC therapy significantly increased the TNC and FMDO genes expression with no changes in the molecular organization of collagen. With the association of therapies, a significant greater collagen fibers organization was observed with involvement of the FMOD gene. The therapies did not affect the expression of COL1A1, SMAD2, SMAD3, MKX, and EGR1 genes, nor did they influence the amount of collagen I and III, caspase-3, tenomodulin (Tnmd), and hydroxyproline. In conclusion, the application of the microcurrent isolated or associated with ASC increased the organization of the collagen fibers, which can result in a greater biomechanical resistance in relation to the tendons treated only with ASC. Future studies will be needed to demonstrate the biological effects of these therapies on the functional recovery of injured tendons.
Collapse
Affiliation(s)
- Fernanda O Bortolazzo
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Letícia D Lucke
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Lucas de Oliveira Fujii
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Rita de Cassia Marqueti
- Graduate Program of rehabilitation science and Graduate Program of Sciences and Technology of Health and Rehabilitation Sciences, University of Brasilia (UnB), Brasília, Brazil
| | | | - Viviane Theodoro
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - André L Bombeiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Maíra Felonato
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Rodrigo A Dalia
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Giane D Carneiro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Cristina Pontes Vicente
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Marcelo A M Esquisatto
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Fernanda A S Mendonça
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Gláucia Maria T Dos Santos
- Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| | - Edson R Pimentel
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil
| | - Andrea A de Aro
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas-UNICAMP, São Paulo, Brazil.,Biomedical Sciences Graduate Program, University Center of Herminio Ometto Foundation/FHO, São Paulo, Brazil
| |
Collapse
|
31
|
A new insight into protein-protein interactions and the effect of conformational alterations in PCNA. Int J Biol Macromol 2020; 148:999-1009. [DOI: 10.1016/j.ijbiomac.2020.01.212] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 01/21/2020] [Accepted: 01/21/2020] [Indexed: 12/14/2022]
|
32
|
Cheng H, Cao X, Min X, Zhang X, Kong Q, Mao Q, Li R, Xue B, Fang L, Liu L, Ding Z. Heat-Shock protein A12A is a novel PCNA-binding protein and promotes hepatocellular carcinoma growth. FEBS J 2020; 287:5464-5477. [PMID: 32128976 DOI: 10.1111/febs.15276] [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: 09/09/2019] [Revised: 02/03/2020] [Accepted: 03/02/2020] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death. Proliferating cell nuclear antigen (PCNA) plays a pivotal role in cancer development and progression. However, the long-term dismal prognosis of HCC mandates more investigation to identify novel regulators in HCC pathogenesis. Heat-shock protein A12A (HSPA12A) encodes a novel member of the HSP70 family. Here, we report that HCC cells showed increased HSPA12A expression, and overexpression of HSPA12A promoted HCC growth and angiogenesis in mice. Gain- and loss-of-functional studies demonstrated that the proliferation of HCC HepG2 cells, as well as β-catenin expression and nuclear translocation, was promoted by HSPA12A overexpression, but in turn suppressed by HSPA12A knockdown. HSPA12A did not impact PCNA expression; however, mass spectrometry and co-immunoprecipitation immunoblotting analysis revealed that HSPA12A directly binds to PCNA and promotes its trimerization, which is an essential functional conformation of PCNA for carcinogenesis. Importantly, PCNA inhibition by PCNA-I1 reversed the HSPA12A-mediated HepG2 cell differentiation. These findings indicate that HSPA12A is a novel regulator of HCC cell proliferation and tumor growth through binding to PCNA for its trimerization. HSPA12A inhibition might represent a viable strategy for the management of HCC in humans.
Collapse
Affiliation(s)
- Hao Cheng
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Xiaofei Cao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Xinxu Min
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Xiaojin Zhang
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, China
| | - Qiuyue Kong
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Qian Mao
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Rongrong Li
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| | - Bin Xue
- State Key Laboratory of Pharmaceutical Biotechnology, Jiangsu Key Laboratory of Molecular Medicine, School of Medicine, Nanjing University, China
| | - Lei Fang
- Medical School, Nanjing University, China
| | - Li Liu
- Department of Geriatrics, Jiangsu Provincial Key Laboratory of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, China.,Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, China
| | - Zhengnian Ding
- Department of Anesthesiology, First Affiliated Hospital of Nanjing Medical University, China
| |
Collapse
|
33
|
Stromal Cell-Derived Factor-1 Enhances the Therapeutic Effects of Human Endometrial Regenerative Cells in a Mouse Sepsis Model. Stem Cells Int 2020; 2020:4820543. [PMID: 32256608 PMCID: PMC7103048 DOI: 10.1155/2020/4820543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Revised: 02/09/2020] [Accepted: 02/25/2020] [Indexed: 12/17/2022] Open
Abstract
Endometrial regenerative cells (ERCs) are mesenchymal-like stromal cells obtained from human menstrual blood, whose positive therapeutic effects have been validated in several experimental models. Stromal cell-derived factor-1 (SDF-1), the ligand for CXCR4, plays an important role in the migration of mesenchymal stromal cells. The purpose of this study was to investigate the role of the SDF-1/CXCR4 pathway in the therapeutic effects of ERCs in a mouse sepsis model. Through preexperiment and confirmation, wild-type C57BL/6 mice were intraperitoneally injected with 10 mg/kg lipopolysaccharide (LPS). The therapeutic effects of ERCs with different pretreatments were evaluated by assessing sepsis-related symptoms, detecting tissue damage and measuring levels of inflammatory and oxidative stress-related factors. The in vitro experiments demonstrated that there was a much higher CXCR4 expression on ERCs when they were cocultured with SDF-1. The ex vivo experiment results showed that SDF-1 expression significantly increased in mouse tissues. Further experiments also confirmed that, compared with the unmodified ERC treatment group, SDF-1 pretreatment significantly enhanced the therapeutic effects of ERCs on alleviating sepsis symptoms, ameliorating pathological changes, reducing Bax level, and increasing Bcl-2 and PCNA expressions in mouse liver tissues. Furthermore, it was also found that SDF-1-pretreated ERCs contributed to reducing the levels of proinflammatory cytokines (TNF-α, IL-1β) and increasing the levels of anti-inflammatory factors (IL-4, IL10) in mouse serum, liver, and lung. Moreover, SDF-1-pretreated ERCs could also significantly decrease the levels of iNOS and MDA and increase the expression of Nrf2, HO-1, and SOD in liver tissues. Taken together, these results indicate that SDF-1 pretreatment plays a key role in improving the therapeutic effects of ERCs in alleviating sepsis-related symptoms, reducing tissue damage, regulating inflammatory imbalance, and relieving oxidative stress in a mouse sepsis model, which provides more possibilities for the clinical application of ERCs in sepsis and relevant diseases.
Collapse
|
34
|
Lee CC, Wang JW, Leu WM, Huang YT, Huang YW, Hsu YH, Meng M. Proliferating Cell Nuclear Antigen Suppresses RNA Replication of Bamboo Mosaic Virus through an Interaction with the Viral Genome. J Virol 2019; 93:e00961-19. [PMID: 31511381 PMCID: PMC6819918 DOI: 10.1128/jvi.00961-19] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 08/30/2019] [Indexed: 11/20/2022] Open
Abstract
Bamboo mosaic virus (BaMV), a member of the Potexvirus genus, has a monopartite positive-strand RNA genome on which five open reading frames (ORFs) are organized. ORF1 encodes a 155-kDa nonstructural protein (REPBaMV) that plays a core function in replication/transcription of the viral genome. To find out cellular factors modulating the replication efficiency of BaMV, a putative REPBaMV-associated protein complex from Nicotiana benthamiana leaf was isolated on an SDS-PAGE gel, and a few proteins preferentially associated with REPBaMV were identified by tandem mass spectrometry. Among them, proliferating cell nuclear antigen (PCNA) was particularly noted. Overexpression of PCNA strongly suppressed the accumulation of BaMV coat protein and RNAs in leaf protoplasts. In addition, PCNA exhibited an inhibitory effect on BaMV polymerase activity. A pulldown assay confirmed a binding capability of PCNA toward BaMV genomic RNA. Mutations at D41 or F114 residues, which are critical for PCNA to function in nuclear DNA replication and repair, disabled PCNA from binding BaMV genomic RNA as well as suppressing BaMV replication. This suggests that PCNA bound to the viral RNA may interfere with the formation of a potent replication complex or block the replication process. Interestingly, BaMV is almost invisible in the newly emerging leaves where PCNA is actively expressed. Accordingly, PCNA is probably one of the factors restricting the proliferation of BaMV in young leaves. Foxtail mosaic virus and Potato virus X were also suppressed by PCNA in the protoplast experiment, suggesting a general inhibitory effect of PCNA on the replication of potexviruses.IMPORTANCE Knowing the dynamic interplay between plant RNA viruses and their host is a basic step toward first understanding how the viruses survive the plant defense mechanisms and second gaining knowledge of pathogenic control in the field. This study found that plant proliferating cell nuclear antigen (PCNA) imposes a strong inhibition on the replication of several potexviruses, including Bamboo mosaic virus, Foxtail mosaic virus, and Potato virus X Based on the tests on Bamboo mosaic virus, PCNA is able to bind the viral genomic RNA, and this binding is a prerequisite for the protein to suppress the virus replication. This study also suggests that PCNA plays an important role in restricting the proliferation of potexviruses in the rapidly dividing tissues of plants.
Collapse
Affiliation(s)
- Cheng-Cheng Lee
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Jhih-Wei Wang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Wei-Ming Leu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Yu-Ting Huang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Ying-Wen Huang
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Yau-Heiu Hsu
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Menghsiao Meng
- Graduate Institute of Biotechnology, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
35
|
Lu S, Dong Z. Additive effects of a small molecular PCNA inhibitor PCNA-I1S and DNA damaging agents on growth inhibition and DNA damage in prostate and lung cancer cells. PLoS One 2019; 14:e0223894. [PMID: 31600334 PMCID: PMC6786632 DOI: 10.1371/journal.pone.0223894] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 10/01/2019] [Indexed: 12/15/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is essential for DNA replication and repair, and cell growth and survival. Previously, we identified a novel class of small molecules that bind directly to PCNA, stabilize PCNA trimer structure, reduce chromatin-associated PCNA, selectively inhibit tumor cell growth, and induce apoptosis. The purpose of this study was to investigate the combinatorial effects of lead compound PCNA-I1S with DNA damaging agents on cell growth, DNA damage, and DNA repair in four lines of human prostate and lung cancer cells. The DNA damage agents used in the study include ionizing radiation source cesium-137 (Cs-137), chemotherapy drug cisplatin (cisPt), ultraviolet-C (UV-C), and oxidative compound H2O2. DNA damage was assessed using immunofluorescent staining of γH2AX and the Comet assay. The homologous recombination repair (HRR) was determined using a plasmid-based HRR reporter assay and the nucleotide excision repair (NER) was indirectly examined by the removal of UV-induced cyclobutane pyrimidine dimers (CPD). We found that PCNA-I1S inhibited cell growth in a dose-dependent manner and significantly enhanced the cell growth inhibition induced by pretreatment with DNA damaging agents Cs-137 irradiation, UV-C, and cisPt. However, the additive growth inhibitory effects were not observed in cells pre-treated with PCNA-I1S, followed by treatment with cisPt. H2O2 enhanced the level of chromatin-bound PCNA in quiescent cells, which was attenuated by PCNA-I1S. DNA damage was induced in cells treated with either PCNA-I1S or cisPt alone and was significantly elevated in cells exposed to the combination of PCNA-I1S and cisPt. Finally, PCNA-I1S attenuated repair of DNA double strand breaks (DSBs) by HRR and the removal of CPD by NER. These data suggest that targeting PCNA with PCNA-I1S may provide a novel approach for enhancing the efficacy of chemotherapy and radiation therapy in treatment of human prostate and lung cancer.
Collapse
Affiliation(s)
- Shan Lu
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
| | - Zhongyun Dong
- Division of Hematology-Oncology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, Ohio, United States of America
- * E-mail:
| |
Collapse
|
36
|
Mo A, Wang J, Yuan M, Zhao D, Gu Z, Liu Y, Huang H, Yuan YC. Effect of sub-chronic dietary L-selenomethionine exposure on reproductive performance of Red Swamp Crayfish, (Procambarus clarkii). ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 253:749-758. [PMID: 31344537 DOI: 10.1016/j.envpol.2019.07.082] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/18/2019] [Accepted: 07/15/2019] [Indexed: 06/10/2023]
Abstract
The effect of selenium (Se) on the reproductive system has been investigated in both humans and vertebrates, but few studies of female fertility and reproduction in invertebrate have been reported. This study is aimed to investigate the effect of SeMet on growth performance and reproductive system after crayfish were fed with graded levels of dietary SeMet (0, 1.49, 3.29, 10.02, 30.27 or 59.8 μg Se/g dry weight) for 60 days. Crayfish treated with the high levels of SeMet (10.02, 30.27 and 59.76 μg Se/g) exhibited decreasing FW and CL in both male and female. Interestingly, Se accumulation was higher in ovary than in other tissues, suggesting that ovary may serve as a target organ for Se accumulation. We found that dietary Se concentration of 10.02 μg Se/g significantly improved the spawning rate, promoted the synchronized spawning, and up-regulated the expressions of mRNA of cdc2 and vitellogenin, with significantly increased E2 and VTG concentrations in hemolymph of female crayfish. However, a marked decrease of the E2 contents and spawning rate was observed in the groups treated with 30.27 and 59.76 μg Se/g diets. In conclusion, the results of this study indicated that the Se had maximum accumulation in ovary, affecting the reproductive capacity by intervening the expression of cdc2 and vitellogenin in the reproductive system. The LOAEL to induce FW was observed in crayfish fed with 10.02 μg Se/g diet, and its value can cause toxicity within the range of natural concentration, so the addition of Se in the feed should be within 10.02 μg Se/g.
Collapse
Affiliation(s)
- Aijie Mo
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jianghua Wang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Mingrui Yuan
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Dengxiao Zhao
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Zemao Gu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Ya Liu
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Hongying Huang
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yong Chao Yuan
- College of Fisheries, Key Lab of Freshwater Animal Breeding, Ministry of Agriculture, Huazhong Agricultural University, Wuhan, Hubei 430070, China; Shuangshui Shuanglu Institute, Huazhong Agricultural University, Wuhan 430070, China; National Demonstration Center for Experimental Aquaculture Education, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
37
|
Ohayon D, De Chiara A, Dang PMC, Thieblemont N, Chatfield S, Marzaioli V, Burgener SS, Mocek J, Candalh C, Pintard C, Tacnet-Delorme P, Renault G, Lagoutte I, Favier M, Walker F, Hurtado-Nedelec M, Desplancq D, Weiss E, Benarafa C, Housset D, Marie JC, Frachet P, El-Benna J, Witko-Sarsat V. Cytosolic PCNA interacts with p47phox and controls NADPH oxidase NOX2 activation in neutrophils. J Exp Med 2019; 216:2669-2687. [PMID: 31492810 PMCID: PMC6829599 DOI: 10.1084/jem.20180371] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 06/17/2019] [Accepted: 08/16/2019] [Indexed: 12/29/2022] Open
Abstract
This study describes a novel function of cytosolic proliferating cell nuclear antigen (PCNA) in the control of neutrophil NADPH oxidase, a complex pivotal for ROS generation in inflammation. Inhibition of neutrophil PCNA results in a potent antiinflammatory effect in colitis. Neutrophils produce high levels of reactive oxygen species (ROS) by NADPH oxidase that are crucial for host defense but can lead to tissue injury when produced in excess. We previously described that proliferating cell nuclear antigen (PCNA), a nuclear scaffolding protein pivotal in DNA synthesis, controls neutrophil survival through its cytosolic association with procaspases. We herein showed that PCNA associated with p47phox, a key subunit of NADPH oxidase, and that this association regulated ROS production. Surface plasmon resonance and crystallography techniques demonstrated that the interdomain-connecting loop of PCNA interacted directly with the phox homology (PX) domain of the p47phox. PCNA inhibition by competing peptides or by T2AA, a small-molecule PCNA inhibitor, decreased NADPH oxidase activation in vitro. Furthermore, T2AA provided a therapeutic benefit in mice during trinitro-benzene-sulfonic acid (TNBS)–induced colitis by decreasing oxidative stress, accelerating mucosal repair, and promoting the resolution of inflammation. Our data suggest that targeting PCNA in inflammatory neutrophils holds promise as a multifaceted antiinflammatory strategy.
Collapse
Affiliation(s)
- Delphine Ohayon
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Alessia De Chiara
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Pham My-Chan Dang
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Nathalie Thieblemont
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Simon Chatfield
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Viviana Marzaioli
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Sabrina Sofia Burgener
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Julie Mocek
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Céline Candalh
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Coralie Pintard
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Pascale Tacnet-Delorme
- Université Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut de Biologie Structurale, Grenoble, France
| | - Gilles Renault
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Isabelle Lagoutte
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | - Maryline Favier
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France.,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| | | | - Margarita Hurtado-Nedelec
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Dominique Desplancq
- Ecole Supérieure de Biotechnologie de Strasbourg, Centre National de la Recherche Scientifique UMR 7242, Université de Strasbourg, Strasbourg, France
| | - Etienne Weiss
- Ecole Supérieure de Biotechnologie de Strasbourg, Centre National de la Recherche Scientifique UMR 7242, Université de Strasbourg, Strasbourg, France
| | - Charaf Benarafa
- Institute of Virology and Immunology, Mittelhäusern, Switzerland.,Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Dominique Housset
- Université Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut de Biologie Structurale, Grenoble, France
| | - Jean-Claude Marie
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Philippe Frachet
- Université Grenoble Alpes, Commissariat à l'Énergie Atomique et aux Énergies Alternatives, Centre National de la Recherche Scientifique, Institut de Biologie Structurale, Grenoble, France
| | - Jamel El-Benna
- LabEx Inflamex, Sorbonne Paris Cité, Paris, France.,Institut National de la Santé et de la Recherche Médicale U1149, Centre National de la Recherche Scientifique ERL8252, Centre de Recherche sur l'Inflammation, Université Paris Diderot, Faculté de Médecine, Site Xavier Bichat, Paris, France
| | - Véronique Witko-Sarsat
- Institut National de la Santé et de la Recherche Médicale U1016, Centre National de la Recherche Scientifique UMR 8104, Université Paris-Descartes, Cochin Institute, Paris, France .,LabEx Inflamex, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
38
|
Denes V, Hideg O, Nyisztor Z, Lakk M, Godri Z, Berta G, Geck P, Gabriel R. The Neuroprotective Peptide PACAP1-38 Contributes to Horizontal Cell Development in Postnatal Rat Retina. Invest Ophthalmol Vis Sci 2019; 60:770-778. [PMID: 30795011 DOI: 10.1167/iovs.18-25719] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose PACAP1-38, a member of the secretin/glucagon superfamily, is expressed in the developing retina with documented neuroprotective effects. However, its function in retinal cell differentiation has yet to be elucidated. Our goals, therefore, were to identify PAC1 expressing cells morphologically, investigate the PACAP1-38 action functionally, and establish PACAP1-38 regulated events developmentally during the first postnatal week in rat retina. Methods P1 retinal sections or whole mounts of Wistar rats were used to reveal PAC1 and calbindin immunoreactive structures. P1, P3, or P7 pups were injected intravitreally with 100 pmol PACAP1-38. Tissues were harvested 24 hours post-treatment, then processed for calbindin immunohistochemistry to determine horizontal cell number, or 6, 12, 24 hours post-treatment for real-time PCR and immunoblots to detect PCNA expression. To localize proliferating cells, anti-PCNA antibody was applied. Results We showed various PAC1 expressing cells in RPE, NBL, and GCL in P1 retina including calbindin positive horizontal cells. We found that PACAP1-38 induced a marked cell number increase at P3 and P7 and showed upregulated cell proliferation as its mechanism; however, it was ineffective at P1. PACAP1-38 induced proliferative cells localized in the NBL, and double-marker studies demonstrated that the induced proliferative cells were horizontal cells. Conclusions PACAP1-38 appears to act in retinal differentiation by inducing mitosis selectively in a time and cell specific manner through PAC1. The control of horizontal cell proliferation raises the novel possibilities that (1) PACAP1-38 may be a major player in retinal patterning and (2) PACAP signaling may be critical in retinoblastoma.
Collapse
Affiliation(s)
- Viktoria Denes
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Orsolya Hideg
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Zsolt Nyisztor
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Monika Lakk
- Department of Ophthalmology and Visual Sciences, School of Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Zoltan Godri
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| | - Gergely Berta
- Department of Medical Biology and Central Electron Microscope Laboratory, Medical School, University of Pécs, Pécs, Hungary
| | - Peter Geck
- Department of Immunology, School of Medicine, Tufts University, Boston, Massachusetts, United States
| | - Robert Gabriel
- Department of Experimental Zoology and Neurobiology, University of Pécs, Pécs, Hungary
| |
Collapse
|
39
|
Kowalska E, Bartnicki F, Fujisawa R, Bonarek P, Hermanowicz P, Tsurimoto T, Muszynska K, Strzalka W. Inhibition of DNA replication by an anti-PCNA aptamer/PCNA complex. Nucleic Acids Res 2019; 46:25-41. [PMID: 29186524 PMCID: PMC5758903 DOI: 10.1093/nar/gkx1184] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 11/13/2017] [Indexed: 12/29/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is a multifunctional protein present in the nuclei of eukaryotic cells that plays an important role as a component of the DNA replication machinery, as well as DNA repair systems. PCNA was recently proposed as a potential non-oncogenic target for anti-cancer therapy. In this study, using the Systematic Evolution of Ligands by EXponential enrichment (SELEX) method, we developed a short DNA aptamer that binds human PCNA. In the presence of PCNA, the anti-PCNA aptamer inhibited the activity of human DNA polymerase δ and ϵ at nM concentrations. Moreover, PCNA protected the anti-PCNA aptamer against the exonucleolytic activity of these DNA polymerases. Investigation of the mechanism of anti-PCNA aptamer-dependent inhibition of DNA replication revealed that the aptamer did not block formation, but was a component of PCNA/DNA polymerase δ or ϵ complexes. Additionally, the anti-PCNA aptamer competed with the primer-template DNA for binding to the PCNA/DNA polymerase δ or ϵ complex. Based on the observations, a model of anti-PCNA aptamer/PCNA complex-dependent inhibition of DNA replication was proposed.
Collapse
Affiliation(s)
- Ewa Kowalska
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland
| | - Filip Bartnicki
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland
| | - Ryo Fujisawa
- Department of Biology, Faculty of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Piotr Bonarek
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland
| | - Pawel Hermanowicz
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland.,Laboratory of Photobiology, Malopolska Centre of Biotechnology, Jagiellonian University, Gronostajowa 7A, Krakow 30-387, Poland
| | - Toshiki Tsurimoto
- Department of Biology, Faculty of Science, Kyushu University, 744 Motooka, Nishi-ku, Fukuoka 819-0395, Japan
| | - Klaudia Muszynska
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland
| | - Wojciech Strzalka
- Department of Plant Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, Krakow 30-387, Poland
| |
Collapse
|
40
|
Kundu K, Ghosh S, Sarkar R, Edri A, Brusilovsky M, Gershoni-Yahalom O, Yossef R, Shemesh A, Soria JC, Lazar V, Joshua BZ, Campbell KS, Elkabets M, Porgador A. Inhibition of the NKp44-PCNA Immune Checkpoint Using a mAb to PCNA. Cancer Immunol Res 2019; 7:1120-1134. [PMID: 31164357 DOI: 10.1158/2326-6066.cir-19-0023] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/09/2019] [Accepted: 05/30/2019] [Indexed: 02/05/2023]
Abstract
mAb-based blocking of the immune checkpoints involving the CTLA4-B7 and PD1-PDL1 inhibitory axes enhance T-cell-based adaptive immune responses in patients with cancer. We show here that antitumor responses by natural killer (NK) cells can be enhanced by a checkpoint-blocking mAb, 14-25-9, which we developed against proliferating cell nuclear antigen (PCNA). PCNA is expressed on the surface of cancer cells and acts as an inhibitory ligand for the NK-cell receptor, NKp44-isoform1. We tested for cytoplasmic- and membrane-associated PCNA by FACS- and ImageStream-based staining of cell lines and IHC of human cancer formalin fixed, paraffin embedded tissues. The mAb, 14-25-9, inhibited binding of chimeric NKp44 receptor to PCNA and mostly stained the cytoplasm and membrane of tumor cells, whereas commercial antibody (clone PC10) stained nuclear PCNA. NK functions were measured using ELISA-based IFNγ secretion assays and FACS-based killing assays. The NK92-NKp44-1 cell line and primary human NK cells showed increased IFNγ release upon coincubation with mAb 14-25-9 and various solid tumor cell lines and leukemias. Treatment with 14-25-9 also increased NK cytotoxic activity. In vivo efficacy was evaluated on patient-derived xenografts (PDX)-bearing NSG mice. In PDX-bearing mice, intravenous administration of mAb 14-25-9 increased degranulation (CD107a expression) of intratumorally injected patient autologous or allogeneic NK cells, as well as inhibited tumor growth when treated long term. Our study describes a mAb against the NKp44-PCNA innate immune checkpoint that can enhance NK-cell antitumor activity both in vitro and in vivo.
Collapse
Affiliation(s)
- Kiran Kundu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Susmita Ghosh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rhitajit Sarkar
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishay Edri
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Michael Brusilovsky
- Division of Allergy and Immunology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Orly Gershoni-Yahalom
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rami Yossef
- Surgery Branch, NCI, NIH, Bethesda, Maryland
| | - Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | | | - Vladimir Lazar
- Worldwide Innovative Network (WIN) Association - WIN Consortium, Villejuif, France
| | - Ben-Zion Joshua
- Department of Otolaryngology-Head and Neck Surgery, Soroka Medical Center and Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kerry S Campbell
- Institute for Cancer Research, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel. .,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
41
|
Abstract
Community-acquired pneumonia (CAP) is a leading cause of morbidity and mortality worldwide. Despite broad literature including basic and translational scientific studies, many gaps in our understanding of host-pathogen interactions remain. In this review, pathogen virulence factors that drive lung infection and injury are discussed in relation to their associated host immune pathways. CAP epidemiology is considered, with a focus on Staphylococcus aureus and Streptococcus pneumoniae as primary pathogens. Bacterial factors involved in nasal colonization and subsequent virulence are illuminated. A particular emphasis is placed on bacterial pore-forming toxins, host cell death, and inflammasome activation. Identified host-pathogen interactions are then examined by linking pathogen factors to aberrant host response pathways in the context of acute lung injury in both primary and secondary infection. While much is known regarding bacterial virulence and host immune responses, CAP management is still limited to mostly supportive care. It is likely that improvements in therapy will be derived from combinatorial targeting of both pathogen virulence factors and host immunomodulation.
Collapse
|
42
|
Zeng Z, Liu W, Benton CB, Konoplev S, Lu H, Wang RY, Chen J, Shpall E, Baggerly KA, Champlin R, Konopleva M. Proteomic Profiling of Signaling Networks Modulated by G-CSF/Plerixafor/Busulfan-Fludarabine Conditioning in Acute Myeloid Leukemia Patients in Remission or with Active Disease prior to Allogeneic Stem Cell Transplantation. Acta Haematol 2019; 142:176-184. [PMID: 31112940 DOI: 10.1159/000495456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/07/2023]
Abstract
To characterize intracellular signaling in peripheral blood (PB) cells of acute myeloid leukemia (AML) patients undergoing pretransplant conditioning with CXCR4 inhibitor plerixafor, granulocyte colony-stimulating factor (G-CSF), and busulfan plus fludarabine (Bu+Flu) chemotherapy, we profiled 153 proteins in 33 functional groups using reverse phase protein array. CXCR4 inhibition mobilized AML progenitors and clonal AML cells, and this was associated with molecular markers of cell cycle progression. G-CSF/plerixafor and G-CSF/plerixafor/Bu+Flu modulated distinct signaling networks in AML blasts of patients undergoing conditioning with active disease compared to nonleukemic PB cells of patients in remission. We identified AML-specific proteins that remained aberrantly expressed after chemotherapy, representing putative chemoresistance markers in AML.
Collapse
Affiliation(s)
- Zhihong Zeng
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Wenbin Liu
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Christopher B Benton
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sergej Konoplev
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Hongbo Lu
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rui-Yu Wang
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Julianne Chen
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Keith A Baggerly
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Marina Konopleva
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA,
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA,
| |
Collapse
|
43
|
Zolbin MM, Mamillapalli R, Nematian SE, Goetz TG, Taylor HS. Adipocyte alterations in endometriosis: reduced numbers of stem cells and microRNA induced alterations in adipocyte metabolic gene expression. Reprod Biol Endocrinol 2019; 17:36. [PMID: 30982470 PMCID: PMC6463663 DOI: 10.1186/s12958-019-0480-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 04/02/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Endometriosis is an estrogen dependent, inflammatory disorder occurring in 5-10% of reproductive-aged women. Women with endometriosis have a lower body mass index (BMI) and decreased body fat compared to those without the disease, yet few studies have focused on the metabolic abnormalities in adipose tissue in women with endometriosis. Previously, we identified microRNAs that are differentially expressed in endometriosis and altered in the serum of women with the disease. Here we explore the effect of endometriosis on fat tissue and identified a role for endometriosis-related microRNAs in fat metabolism and a reduction in adipocyte stem cell number. METHODS Primary adipocyte cells cultured from 20 patients with and without endometriosis were transfected with mimics and inhibitors of microRNAs 342-3p or Let 7b-5p to model the status of these microRNAs in endometriosis. RNA was extracted for gene expression analysis by qRT-PCR. PCNA expression was used as a marker of adipocyte proliferation. Endometriosis was induced experimentally in 9-week old female C57BL/6 mice and after 10 months fat tissue was harvested from both the subcutaneous (inguinal) and visceral (mesenteric) tissue. Adipose-derived mesenchymal stem cells in fat tissue were characterized in both endometriosis and non-endometriosis mice by FACS analysis. RESULTS Gene expression analysis showed that endometriosis altered the expression of Cebpa, Cebpb, Ppar-γ, leptin, adiponectin, IL-6, and HSL, which are involved in driving brown adipocyte differentiation, appetite, insulin sensitivity and fat metabolism. Each gene was regulated by an alteration in microRNA expression known to occur in endometriosis. Analysis of the stem cell content of adipose tissue in a mouse model of endometriosis demonstrated a reduced number of adipocyte stem cells. CONCLUSIONS We demonstrate that microRNAs Let-7b and miR-342-3p affected metabolic gene expression significantly in adipocytes of women with endometriosis. Similarly, there is a reduction in the adipose stem cell population in a mouse model of endometriosis. Taken together these data suggest that endometriosis alters BMI in part through an effect on adipocytes and fat metabolism.
Collapse
Affiliation(s)
- Masoumeh Majidi Zolbin
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 310 Cedar Street, New Haven, CT, 06520, USA
| | - Ramanaiah Mamillapalli
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 310 Cedar Street, New Haven, CT, 06520, USA.
| | - Sepide E Nematian
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 310 Cedar Street, New Haven, CT, 06520, USA
| | - Teddy G Goetz
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 310 Cedar Street, New Haven, CT, 06520, USA
| | - Hugh S Taylor
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, 310 Cedar Street, New Haven, CT, 06520, USA
| |
Collapse
|
44
|
Potey PM, Rossi AG, Lucas CD, Dorward DA. Neutrophils in the initiation and resolution of acute pulmonary inflammation: understanding biological function and therapeutic potential. J Pathol 2019; 247:672-685. [PMID: 30570146 PMCID: PMC6492013 DOI: 10.1002/path.5221] [Citation(s) in RCA: 148] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is the often fatal sequelae of a broad range of precipitating conditions. Despite decades of intensive research and clinical trials there remain no therapies in routine clinical practice that target the dysregulated and overwhelming inflammatory response that characterises ARDS. Neutrophils play a central role in the initiation, propagation and resolution of this complex inflammatory environment by migrating into the lung and executing a variety of pro-inflammatory functions. These include degranulation with liberation of bactericidal proteins, release of cytokines and reactive oxygen species as well as production of neutrophil extracellular traps. Although these functions are advantageous in clearing bacterial infection, the consequence of associated tissue damage, the contribution to worsening acute inflammation and prolonged neutrophil lifespan at sites of inflammation are deleterious. In this review, the importance of the neutrophil will be considered, together with discussion of recent advances in understanding neutrophil function and the factors that influence them throughout the phases of inflammation in ARDS. From a better understanding of neutrophils in this context, potential therapeutic targets are identified and discussed. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Philippe Md Potey
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Adriano G Rossi
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher D Lucas
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David A Dorward
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
45
|
Li P, Wang Q, Wang H. MicroRNA-204 inhibits the proliferation, migration and invasion of human lung cancer cells by targeting PCNA-1 and inhibits tumor growth in vivo. Int J Mol Med 2018; 43:1149-1156. [PMID: 30628638 PMCID: PMC6365052 DOI: 10.3892/ijmm.2018.4044] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 09/28/2018] [Indexed: 01/10/2023] Open
Abstract
Lung cancer accounts for ~20% of the total cancer-associated mortalities worldwide. Lung cancer is often diagnosed at advanced stages and is therefore difficult to treat. The biomarkers for diagnosis of lung cancer are limited and unreliable. In addition, the lack of availability of efficient chemotherapeutic agents and targets has resulted in limitations in the successful treatment of lung cancer. Previously, microRNAs (miRNA/miR) have been implicated in the onset and development of several types of cancer. The expression of miRNAs is often dysregulated in cancer cells; therefore, they are considered important therapeutic targets and agents. The present study examined the expression of miR-204 in 4 different lung cancer cell lines and 1 normal cell line. The results revealed that miR-204 was significantly downregulated (4–8-fold) in all the cancer cell lines (P<0.05). Overexpression of miR-204 in A549 lung cancer cells inhibited the proliferative, migratory and invasive capabilities of the lung cancer cells. Furthermore, miR-204 overexpression also induced apoptosis in the A549 lung cancer cells. Bioinformatics analysis revealed proliferating cell nuclear antigen 1 (PCNA-1) to be a potential target of miR-204. The reverse transcription quantitative polymerase chain reaction analysis revealed that PCNA-1 was significantly upregulated (up to 5-fold) in the lung cancer cells (P<0.05), and the over-expression of miR-204 caused the downregulation of PCNA-1 in A549 lung cancer cells. Silencing of PCNA-1 in A549 cells exerted similar effects to that of miR-204 overexpression on the proliferative, migratory and invasive capabilities of A549 lung cancer cells. Additionally, the suppression of miR-204 in A549 cells transfected with Si-PCNA-1 did not rescue the effects of PCNA-1 silencing on cell proliferation, migration or invasion. Conversely, the overexpression of PCNA-1 in A549 cells transfected with miR-204 mimics promoted the proliferation, migration and invasion of lung cancer cells. Furthermore, overexpression of miR-204 in xenograft tumors significantly inhibited their growth. Taken together, these results indicated that miR-204 regulates the proliferative, migratory and invasive capabilities of lung cancer cells by targeting PCNA-1.
Collapse
Affiliation(s)
- Ping Li
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Qingan Wang
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| | - Haining Wang
- Department of Thoracic Surgery, Shengli Oilfield Central Hospital, Dongying, Shandong 257034, P.R. China
| |
Collapse
|
46
|
Altieri AS, Kelman Z. DNA Sliding Clamps as Therapeutic Targets. Front Mol Biosci 2018; 5:87. [PMID: 30406112 PMCID: PMC6204406 DOI: 10.3389/fmolb.2018.00087] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Accepted: 09/10/2018] [Indexed: 01/12/2023] Open
Abstract
Chromosomal DNA replication is achieved by an assembly of multi-protein complexes at the replication fork. DNA sliding clamps play an important role in this assembly and are essential for cell viability. Inhibitors of bacterial (β-clamp) and eukaryal DNA clamps, proliferating cell nuclear antigen (PCNA), have been explored for use as antibacterial and anti-cancer drugs, respectively. Inhibitors for bacterial β-clamps include modified peptides, small molecule inhibitors, natural products, and modified non-steroidal anti-inflammatory drugs. Targeting eukaryotic PCNA sliding clamp in its role in replication can be complicated by undesired effects on healthy cells. Some success has been seen in the design of peptide inhibitors, however, other research has focused on targeting PCNA molecules that are modified in diseased states. These inhibitors that are targeted to PCNA involved in DNA repair can sensitize cancer cells to existing anti-cancer therapeutics, and a DNA aptamer has also been shown to inhibit PCNA. In this review, studies in the use of both bacterial and eukaryotic sliding clamps as therapeutic targets are summarized.
Collapse
Affiliation(s)
- Amanda S Altieri
- Institute for Bioscience and Biotechnology Research, University of Maryland and the National Institute of Standards and Technology, Rockville, MD, United States
| | - Zvi Kelman
- Institute for Bioscience and Biotechnology Research, University of Maryland and the National Institute of Standards and Technology, Rockville, MD, United States.,Biomolecular Labeling Laboratory, Institute for Bioscience and Biotechnology Research, National Institute of Standards and Technology, Rockville, MD, United States
| |
Collapse
|
47
|
Chatfield SM, Thieblemont N, Witko-Sarsat V. Expanding Neutrophil Horizons: New Concepts in Inflammation. J Innate Immun 2018; 10:422-431. [PMID: 30257246 DOI: 10.1159/000493101] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 08/20/2018] [Indexed: 12/16/2022] Open
Abstract
Research into neutrophil biology in the last 10 years has uncovered a number of unexpected aspects of this still mysterious innate immune cell. Advances in technology have allowed visualisation of neutrophil trafficking to sites of inflammation, and, remarkably, neutrophils have been observed to depart from the scene in what has been termed reverse migration. There has also been increasing appreciation of the heterogeneity of neutrophils with ongoing categorisation of neutrophil subsets, including myeloid-derived suppressor cells and low-density granulocytes. Newly recognised neutrophil functions include the ability to release novel immune mediators such as extracellular DNA and microvesicles. Finally, studies of neutrophil cell death, both apoptotic and non-apoptotic, have revealed remarkable differences compared to other cell types. This review will highlight important discoveries in these facets of neutrophil biology and how the new findings will inform treatment of diseases where neutrophils are implicated.
Collapse
|
48
|
Søgaard CK, Blindheim A, Røst LM, Petrović V, Nepal A, Bachke S, Liabakk NB, Gederaas OA, Viset T, Arum CJ, Bruheim P, Otterlei M. "Two hits - one stone"; increased efficacy of cisplatin-based therapies by targeting PCNA's role in both DNA repair and cellular signaling. Oncotarget 2018; 9:32448-32465. [PMID: 30197755 PMCID: PMC6126690 DOI: 10.18632/oncotarget.25963] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/31/2018] [Indexed: 01/08/2023] Open
Abstract
Low response rate and rapid development of resistance against commonly used chemotherapeutic regimes demand new multi-targeting anti-cancer strategies. In this study, we target the stress-related roles of the scaffold protein PCNA with a cell-penetrating peptide containing the PCNA-interacting motif APIM. The APIM-peptide increased the efficacy of cisplatin-based therapies in a muscle-invasive bladder cancer (MIBC) solid tumor model in rat and in bladder cancer (BC) cell lines. By combining multiple omics-levels, from gene expression to proteome/kinome and metabolome, we revealed a unique downregulation of the EGFR/ERBB2 and PI3K/Akt/mTOR pathways in the APIM-peptide-cisplatin combination treated cells. Additionally, the combination treatment reduced the expression of anti-apoptotic proteins and proteins involved in development of resistance to cisplatin. Concurrently, we observed increased levels of DNA breaks in combination treated cells, suggesting that the APIM-peptide impaired PCNA - DNA repair protein interactions and reduced the efficacy of repair. This was also seen in cisplatin-resistant cells, which notably was re-sensitized to cisplatin by the APIM-peptide. Our data indicate that the increased efficacy of cisplatin treatment is mediated both via downregulation of known oncogenic signaling pathways and inhibition of DNA repair/translesion synthesis (TLS), thus the APIM-peptide hits both nuclear and cytosolic functions of PCNA. The novel multi-targeting strategy of the APIM-peptide could potentially improve the efficacy of chemotherapeutic regiments for treatment of MIBC, and likely other solid tumors.
Collapse
Affiliation(s)
- Caroline Krogh Søgaard
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Augun Blindheim
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Urology and Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Lisa M Røst
- Department of Biotechnology and Food Science, NTNU, Trondheim, Norway
| | - Voin Petrović
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Anala Nepal
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Siri Bachke
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Nina-Beate Liabakk
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Odrun A Gederaas
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Trond Viset
- Department of Pathology, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Carl-Jørgen Arum
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Department of Urology and Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway
| | - Per Bruheim
- Department of Biotechnology and Food Science, NTNU, Trondheim, Norway
| | - Marit Otterlei
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.,Clinic of Surgery, St. Olavs Hospital, Trondheim University Hospital, Trondheim, Norway.,APIM Therapeutics A/S, Trondheim, Norway
| |
Collapse
|
49
|
Olaisen C, Kvitvang HFN, Lee S, Almaas E, Bruheim P, Drabløs F, Otterlei M. The role of PCNA as a scaffold protein in cellular signaling is functionally conserved between yeast and humans. FEBS Open Bio 2018; 8:1135-1145. [PMID: 29988559 PMCID: PMC6026702 DOI: 10.1002/2211-5463.12442] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 02/19/2018] [Accepted: 05/01/2018] [Indexed: 12/11/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA), a member of the highly conserved DNA sliding clamp family, is an essential protein for cellular processes including DNA replication and repair. A large number of proteins from higher eukaryotes contain one of two PCNA-interacting motifs: PCNA-interacting protein box (PIP box) and AlkB homologue 2 PCNA-interacting motif (APIM). APIM has been shown to be especially important during cellular stress. PIP box is known to be functionally conserved in yeast, and here, we show that this is also the case for APIM. Several of the 84 APIM-containing yeast proteins are associated with cellular signaling as hub proteins, which are able to interact with a large number of other proteins. Cellular signaling is highly conserved throughout evolution, and we recently suggested a novel role for PCNA as a scaffold protein in cellular signaling in human cells. A cell-penetrating peptide containing the APIM sequence increases the sensitivity toward the chemotherapeutic agent cisplatin in both yeast and human cells, and both yeast and human cells become hypersensitive when the Hog1/p38 MAPK pathway is blocked. These results suggest that the interactions between APIM-containing signaling proteins and PCNA during the DNA damage response is evolutionary conserved between yeast and mammals and that PCNA has a role in cellular signaling also in yeast.
Collapse
Affiliation(s)
- Camilla Olaisen
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Hans Fredrik N. Kvitvang
- Department of Biotechnology and Food ScienceFaculty of Natural SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Sungmin Lee
- Department of Biotechnology and Food ScienceFaculty of Natural SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Eivind Almaas
- Department of Biotechnology and Food ScienceFaculty of Natural SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Per Bruheim
- Department of Biotechnology and Food ScienceFaculty of Natural SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Finn Drabløs
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| | - Marit Otterlei
- Department of Clinical and Molecular MedicineFaculty of Medicine and Health SciencesNorwegian University of Science and Technology (NTNU)TrondheimNorway
| |
Collapse
|
50
|
Shemesh A, Kundu K, Peleg R, Yossef R, Kaplanov I, Ghosh S, Khrapunsky Y, Gershoni-Yahalom O, Rabinski T, Cerwenka A, Atlas R, Porgador A. NKp44-Derived Peptide Binds Proliferating Cell Nuclear Antigen and Mediates Tumor Cell Death. Front Immunol 2018; 9:1114. [PMID: 29875773 PMCID: PMC5974751 DOI: 10.3389/fimmu.2018.01114] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 05/03/2018] [Indexed: 11/21/2022] Open
Abstract
Proliferating cell nuclear antigen (PCNA) is considered as a hub protein and is a key regulator of DNA replication, repair, cell cycle control, and apoptosis. PCNA is overexpressed in many cancer types, and PCNA overexpression is correlated with cancer virulence. Membrane-associated PCNA is a ligand for the NKp44 (NCR2) innate immune receptor. The purpose of this study was to characterize the PCNA-binding site within NKp44. We have identified NKp44-derived linear peptide (pep8), which can specifically interact with PCNA and partly block the NKp44–PCNA interaction. We then tested whether NKp44-derived pep8 (NKp44-pep8) fused to cell-penetrating peptides (CPPs) can be employed for targeting the intracellular PCNA for the purpose of anticancer therapy. Treatment of tumor cells with NKp44-pep8, fused to R11-NLS cell-penetrating peptide (R11-NLS-pep8), reduced cell viability and promoted cell death, in various murine and human cancer cell lines. Administration of R11-NLS-pep8 to tumor-bearing mice suppressed tumor growth in the 4T1 breast cancer and the B16 melanoma in vivo models. We therefore identified the NKp44 binding site to PCNA and further developed an NKp44-peptide-based agent that can inhibit tumor growth through interfering with the function of intracellular PCNA in the tumor cell.
Collapse
Affiliation(s)
- Avishai Shemesh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kiran Kundu
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Refael Peleg
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Rami Yossef
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, United States
| | - Irena Kaplanov
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Susmita Ghosh
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Yana Khrapunsky
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Orly Gershoni-Yahalom
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tatiana Rabinski
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Adelheid Cerwenka
- Innate Immunity Group, German Cancer Research Center and Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| | - Roee Atlas
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|