1
|
Ahmad F, Ahmad S, Srivastav AK, Upadhyay TK, Husain A, Khubaib M, Kang S, Park MN, Kim B, Sharma R. "β-glucan signalling stimulates NOX-2 dependent autophagy and LC-3 associated autophagy (LAP) pathway". Int J Biol Macromol 2024; 282:136520. [PMID: 39401634 DOI: 10.1016/j.ijbiomac.2024.136520] [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: 08/02/2024] [Revised: 10/02/2024] [Accepted: 10/10/2024] [Indexed: 12/19/2024]
Abstract
β-Glucan, a complex polysaccharide derived from fungal and yeast cell walls, plays a crucial role in modulating immune responses through their interaction with receptors such as Dectin-1 and Complement receptor 3 (CR-3). This review provides an in-depth analysis of the molecular mechanisms by which β-glucans activate receptor-mediated signalling pathways, focusing particularly on the LC3-associated phagocytosis (LAP) and autophagy pathways. Hence, we explore how β-glucan receptor engagement stimulates NADPH oxidase 2 (NOX-2), leading to the intracellular production of significant level of reactive oxygen species (ROS) essential for both conventional autophagy and LAP. While significant progress has been made in elucidation of downstream signaling by glucans, the regulation of phago-lysosomal maturation and antigen presentation during LAP induction still remains less explored. This review aims to provide a comprehensive overview of these pathways and their regulation by β-glucans. By consolidating the current knowledge, we seek to highlight how these mechanisms can be leveraged for therapeutic applications, particularly in the context of tuberculosis (TB) management, where β-glucans could serve as host-directed adjuvant therapies to combat drug-resistant strains. Despite major advancements in this field, currently key research gaps still persist, including detailed molecular interactions between β-glucan receptors and NOX-2 and the translation of these findings to in-vivo models and clinical investigations. This review underscores the need for further research to explore the therapeutic potential of β-glucans in managing not only tuberculosis but also other diseases such as cancer, cardiovascular conditions, and metabolic disorders.
Collapse
Affiliation(s)
- Firoz Ahmad
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Physiological Sciences, Oklahoma Centre for Respiratory and Infectious Diseases, Oklahoma State University, OK 74074, United States of America
| | - Shad Ahmad
- Department of Biochemistry, Dr. Ram Manohar Lohia Avadh University, Faizabad 224001, Uttar Pradesh, India
| | - Anurag Kumar Srivastav
- Department of Clinical Immunology & Rheumatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Tarun Kumar Upadhyay
- Department of Life Sciences, Parul Institute of Applied Sciences & Research and Development Cell, Parul University, Vadodara 391760, Gujarat, India
| | - Adil Husain
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India; Department of Biochemistry, Babu Banarasi Das [BBD] College of Dental Sciences BBD University, Lucknow 226028, Uttar Pradesh, India
| | - Mohd Khubaib
- Department of Biosciences, Integral University, Lucknow 226026, Uttar Pradesh, India
| | - Sojin Kang
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul 02447, the Republic of Korea.
| | - Rolee Sharma
- Department of Life Sciences & Biotechnology, CSJM University, Kanpur 228024, Uttar Pradesh, India.
| |
Collapse
|
2
|
Fantone KM, Nothaft H, Son Y, Stecenko AA, Szymanski CM, Rada B. The bacterial serine protease inhibitor ecotin inhibits neutrophil elastase enzymatic activity in cystic fibrosis sputa. Heliyon 2024; 10:e38895. [PMID: 39444402 PMCID: PMC11497391 DOI: 10.1016/j.heliyon.2024.e38895] [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: 05/08/2024] [Revised: 10/01/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024] Open
Abstract
Cystic Fibrosis (CF) airway disease is characterized by impaired mucociliary clearance, chronic, polymicrobial infections and robust, neutrophil-dominated inflammation. Pulmonary disease is the leading cause of morbidity and mortality in people with CF and is due to progressive airflow obstruction and ultimately respiratory failure. One of the earliest abnormalities in CF airway disease is the recruitment of neutrophils to the lungs. Neutrophil activation leads to the release of their intracellular content, including neutrophil elastase (NE), that damages lung tissues in CF. Our goal is to characterize a known bacterial NE inhibitor, ecotin, in the CF airway environment. Our results indicate that ecotins cloned from four Gram-negative bacterial species (Campylobacter rectus, Campylobacter showae, Escherichia coli and Pseudomonas aeruginosa) inhibit NE activity in CF sputum samples in a dose-dependent manner. Although we observed differences in the NE-inhibitory activity of the tested ecotins with the Campylobacter homologs being the most effective in NE inhibition in CF sputa, none of the ecotins impaired the ability of human neutrophils to kill major CF respiratory pathogens, P. aeruginosa or S. aureus, in vitro. Overall, we demonstrate that bacterial ecotins inhibit NE activity in CF sputa without compromising bacterial killing by neutrophils.
Collapse
Affiliation(s)
- Kayla M. Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA, USA
| | - Harald Nothaft
- Department of Medical Microbiology and Immunology, University of Alberta, Katz Group Centre, 6-065, Edmonton, AB, T6G 2E1, Canada
| | - Yeongseo Son
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA, USA
| | - Arlene A. Stecenko
- Division of Pulmonology, Asthma, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, USA
| | - Christine M. Szymanski
- Department of Medical Microbiology and Immunology, University of Alberta, Katz Group Centre, 6-065, Edmonton, AB, T6G 2E1, Canada
- Department of Microbiology and Complex Carbohydrate Research Center, University of Georgia, 315 Riverbend Road, Athens, GA 30602, USA
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA, USA
| |
Collapse
|
3
|
Becker S, Swoboda A, Siemer H, Schimmelpfennig S, Sargin S, Shahin V, Schwab A, Najder K. Membrane potential dynamics of C5a-stimulated neutrophil granulocytes. Pflugers Arch 2024; 476:1007-1018. [PMID: 38613695 PMCID: PMC11139730 DOI: 10.1007/s00424-024-02947-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 04/15/2024]
Abstract
Neutrophil granulocytes play a crucial role in host defense against invading pathogens and in inflammatory diseases. The aim of this study was to elucidate membrane potential dynamics during the initial phase of neutrophil activation and its relation to migration and production of reactive oxygen species (ROS). We performed ROS production measurements of neutrophils from healthy C57BL/6J mice after TNFα-priming and/or C5a stimulation. The actin cytoskeleton was visualized with fluorescence microscopy. Furthermore, we combined migration assays and measurements of membrane potential dynamics after stimulating unprimed and/or TNFα-primed neutrophils with C5a. We show that C5a has a concentration-dependent effect on ROS production and chemokinetic migration. Chemokinetic migration and chemotaxis are impaired at C5a concentrations that induce ROS production. The actin cytoskeleton of unstimulated and of ROS-producing neutrophils is not distributed in a polarized way. Inhibition of the phagocytic NADPH oxidase NOX2 with diphenyleneiodonium (DPI) leads to a polarized distribution of the actin cytoskeleton and rescues chemokinetic migration of primed and C5a-stimulated neutrophils. Moreover, C5a evokes a pronounced depolarization of the cell membrane potential by 86.6 ± 4.2 mV starting from a resting membrane potential of -74.3 ± 0.7 mV. The C5a-induced depolarization occurs almost instantaneously (within less than one minute) in contrast to the more gradually developing depolarization induced by PMA (lag time of 3-4 min). This initial depolarization is accompanied by a decrease of the migration velocity. Collectively, our results show that stimulation with C5a evokes parallel changes in membrane potential dynamics, neutrophil ROS production and motility. Notably, the amplitude of membrane potential dynamics is comparable to that of excitable cells.
Collapse
Affiliation(s)
- Stina Becker
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Aljoscha Swoboda
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Henrik Siemer
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | | | - Sarah Sargin
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Victor Shahin
- Institute of Physiology II, University Hospital Münster, Münster, Germany
| | - Albrecht Schwab
- Institute of Physiology II, University Hospital Münster, Münster, Germany.
| | - Karolina Najder
- Institute of Physiology II, University Hospital Münster, Münster, Germany.
- Department of Anesthesiology, Intensive Care and Pain Medicine, University Hospital Münster, Münster, Germany.
| |
Collapse
|
4
|
Krepuska M, Mayer B, Vitale-Cross L, Myneni VD, Boyajian MK, Németh K, Szalayova I, Cho T, McClain-Caldwell I, Gingerich AD, Han H, Westerman M, Rada B, Mezey É. Bone marrow stromal cell-derived hepcidin has antimicrobial and immunomodulatory activities. Sci Rep 2024; 14:3986. [PMID: 38368463 PMCID: PMC10874407 DOI: 10.1038/s41598-024-54227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/09/2024] [Indexed: 02/19/2024] Open
Abstract
Bone marrow stromal cells (BMSCs) have immunomodulatory activities in numerous species and have been used in clinical trials. BMSCs also make antibacterial agents. Since hepcidin is known to have antimicrobial effects in fish, we wondered if it might also be used as an antimicrobial agent by mammalian BMSCs. In the present study, we show hepcidin expression in both mouse (mBMSC) and human BMSCs (hBMSC). We observed a hBMSC hepcidin-dependent degradation of ferroportin in HEK-293 reporter cells in vitro. In human and mouse bone marrows (BM) we detected hepcidin-positive BMSCs in close proximity to hematopoietic progenitors. The conditioned culture medium of hBMSCs significantly reduced bacterial proliferation that was partially blocked by a hepcidin-neutralizing antibody. Similarly, medium in which hepcidin-deficient (Hamp-/-) mouse BMSCs had been grown was significantly less effective in reducing bacterial counts than the medium of wild-type cells. In a zymosan-induced peritonitis mouse model we found that mBMSC-derived hepcidin reduced the number of invading polymorphonuclear (PMN) cells in the peritoneal cavity. Our results show that BMSC-derived hepcidin has antimicrobial properties in vitro and also reduces inflammation in vivo. We conclude that hepcidin should be added to the expanding arsenal of agents available to BMSCs to fight infections and inflammation.
Collapse
Affiliation(s)
- Miklós Krepuska
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
- Department of Neuroradiology, University Hospital Zürich, Zürich, Switzerland
| | - Balázs Mayer
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
- Stem Cell Laboratory, Department of Dermatology, Venereology and Dermato-Oncology, Semmelweis University, Budapest, Hungary
| | | | - Vamsee D Myneni
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
| | | | - Krisztián Németh
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
- Stem Cell Laboratory, Department of Dermatology, Venereology and Dermato-Oncology, Semmelweis University, Budapest, Hungary
| | | | - Ted Cho
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
| | | | - Aaron D Gingerich
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | | | | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Éva Mezey
- National Institutes of Health, NIDCR, ASCS, Bethesda, MD, USA
| |
Collapse
|
5
|
DeCoursey TE. Transcendent Aspects of Proton Channels. Annu Rev Physiol 2024; 86:357-377. [PMID: 37931166 PMCID: PMC10938948 DOI: 10.1146/annurev-physiol-042222-023242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
A handful of biological proton-selective ion channels exist. Some open at positive or negative membrane potentials, others open at low or high pH, and some are light activated. This review focuses on common features that result from the unique properties of protons. Proton conduction through water or proteins differs qualitatively from that of all other ions. Extraordinary proton selectivity is needed to ensure that protons permeate and other ions do not. Proton selectivity arises from a proton pathway comprising a hydrogen-bonded chain that typically includes at least one titratable amino acid side chain. The enormously diverse functions of proton channels in disparate regions of the phylogenetic tree can be summarized by considering the chemical and electrical consequences of proton flux across membranes. This review discusses examples of cells in which proton efflux serves to increase pHi, decrease pHo, control the membrane potential, generate action potentials, or compensate transmembrane movement of electrical charge.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, Chicago, Illinois, USA;
| |
Collapse
|
6
|
Wisniewski É, Czárán D, Kovács F, Bahurek E, Németh A, Sasvári P, Szanda G, Pettkó-Szandtner A, Klement E, Ligeti E, Csépányi-Kömi R. A novel BRET-Based GAP assay reveals phosphorylation-dependent regulation of the RAC-specific GTPase activating protein ARHGAP25. FASEB J 2022; 36:e22584. [PMID: 36190314 DOI: 10.1096/fj.202200689r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 09/07/2022] [Accepted: 09/20/2022] [Indexed: 11/11/2022]
Abstract
ARHGAP25, a RAC-specific GTPase activating protein (GAP), is an essential regulator of phagocyte effector functions such as phagocytosis, superoxide production, and transendothelial migration. Furthermore, its complex role in tumor behavior has recently been recognized. We previously demonstrated that phosphorylation of serine 363 in ARHGAP25 regulates hematopoietic stem cells and progenitor cells in mouse bone marrow. However, the significance of other potential phosphorylation sites of ARHGAP25 remained unknown. Now, we developed a novel, real-time bioluminescence resonance energy transfer (BRET) assay to monitor the GAP activity of ARHGAP25 in vitro. Using this approach, we revealed that phosphorylation of S363 and S488, but not that of S379-380, controls ARHGAP25's RACGAP activity. On the other hand, we found in granulocyte-differentiated human PLB-985 cells that superoxide production and actin depolymerization are regulated by residues S363 and S379-380. The present data demonstrate the value of our BRET-GAP assay and show that different phosphorylation patterns regulate ARHGAP25's GAP activity and its effect on superoxide production and phagocytosis.
Collapse
Affiliation(s)
- Éva Wisniewski
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Domonkos Czárán
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Fanni Kovács
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Enikő Bahurek
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Afrodité Németh
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Péter Sasvári
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Gergő Szanda
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Eva Klement
- Laboratory of Proteomics Research, Biological Research Centre, Szeged, Hungary.,Single Cell Omics ACF, Hungarian Centre of Excellence for Molecular Medicine, Szeged, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | |
Collapse
|
7
|
Anwar MM, Sah R, Shrestha S, Ozaki A, Roy N, Fathah Z, Rodriguez-Morales AJ. Disengaging the COVID-19 Clutch as a Discerning Eye Over the Inflammatory Circuit During SARS-CoV-2 Infection. Inflammation 2022; 45:1875-1894. [PMID: 35639261 PMCID: PMC9153229 DOI: 10.1007/s10753-022-01674-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 01/08/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes the cytokine release syndrome (CRS) and leads to multiorgan dysfunction. Mitochondrial dynamics are fundamental to protect against environmental insults, but they are highly susceptible to viral infections. Defective mitochondria are potential sources of reactive oxygen species (ROS). Infection with SARS-CoV-2 damages mitochondria, alters autophagy, reduces nitric oxide (NO), and increases both nicotinamide adenine dinucleotide phosphate oxidases (NOX) and ROS. Patients with coronavirus disease 2019 (COVID-19) exhibited activated toll-like receptors (TLRs) and the Nucleotide-binding and oligomerization domain (NOD-), leucine-rich repeat (LRR-), pyrin domain-containing protein 3 (NLRP3) inflammasome. The activation of TLRs and NLRP3 by SARS-CoV-2 induces interleukin 6 (IL-6), IL-1β, IL-18, and lactate dehydrogenase (LDH). Herein, we outline the inflammatory circuit of COVID-19 and what occurs behind the scene, the interplay of NOX/ROS and their role in hypoxia and thrombosis, and the important role of ROS scavengers to reduce COVID-19-related inflammation.
Collapse
Affiliation(s)
- Mohammed Moustapha Anwar
- Department of Biotechnology, Institute of Graduate Studies and Research (IGSR), Alexandria University, Alexandria, Egypt.
| | - Ranjit Sah
- Tribhuvan University Institute of Medicine, Kathmandu, Nepal
| | - Sunil Shrestha
- Department of Pharmaceutical and Health Service Research, Nepal Health Research and Innovation Foundation, Lalitpur, Nepal
| | - Akihiko Ozaki
- Department of Breast Surgery, Jyoban Hospital of Tokiwa Foundation, Iwaki, Japan
- Medical Governance Research Institute, Tokyo, Japan
| | - Namrata Roy
- SRM University, SRM Nagar, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Zareena Fathah
- Kings College London, London, UK
- College of Medicine and Health Sciences, United Arab University, Abu Dhabi, United Arab Emirates
| | - Alfonso J Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundacion Universitaria Autonoma de Las Americas, Pereira, Risaralda, Colombia.
- Institución Universitaria Visión de Las Americas, Pereira, Risaralda, Colombia.
- Faculty of Health Sciences, Universidad Cientifica del Sur, Lima, Peru.
- School of Medicine, Universidad Privada Franz Tamayo (UNIFRANZ), Cochabamba, Bolivia.
| |
Collapse
|
8
|
Infections in G6PD-Deficient Hospitalized Patients—Prevalence, Risk Factors, and Related Mortality. Antibiotics (Basel) 2022; 11:antibiotics11070934. [PMID: 35884188 PMCID: PMC9312035 DOI: 10.3390/antibiotics11070934] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 02/04/2023] Open
Abstract
G6PD deficiency is a genetic disease that weakens the immune system and renders affected individuals susceptible to infections. In the Sultanate of Oman resides a high number of recorded G6PD cases due to widespread consanguineous marriage, which may reach 25% of the population. We studied the infection patterns and risk factors for mortality to provide antimicrobial stewardship recommendations for these patients. After obtaining ethical approval, a registry of recorded cases was consulted retrospectively to include G6PD-deficient adult patients admitted to Suhar hospital over 5 years with microbiologically confirmed infections. Patient demographics, health-related information, infection causes, treatment, and clinical outcomes were studied. Data were analyzed to describe infection patterns and risk factors. Several variables, including underlying comorbidities and hospitalization details, such as length of stay, admission to critical care unit, blood transfusion, or exposure to an invasive procedure, were statistically associated with the acquisition of multidrug-resistant and hospital-acquired infections. Meanwhile, these infections were associated with a high mortality rate (28%), significantly associated with the patient’s health status and earlier exposure to antimicrobial treatment due to previous bacterial infection. The high prevalence of G6PD deficiency among the Omani population should alert practitioners to take early action when dealing with such cases during infection that requires hospitalization. Strict infection control measures, Gram-negative empiric coverage, hospital discharge as early as possible, and potent targeted antimicrobial therapy in this patient population can ameliorate the treatment outcomes and should be emphasized by the antimicrobial stewardship team.
Collapse
|
9
|
Andresen S, Fantone K, Chapla D, Rada B, Moremen KW, Pierce M, Szymanski CM. Human Intelectin-1 Promotes Cellular Attachment and Neutrophil Killing of Streptococcus pneumoniae in a Serotype-Dependent Manner. Infect Immun 2022; 90:e0068221. [PMID: 35499339 PMCID: PMC9119095 DOI: 10.1128/iai.00682-21] [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: 12/25/2021] [Accepted: 04/10/2022] [Indexed: 11/20/2022] Open
Abstract
Human intelectin-1 (hIntL-1) is a secreted glycoprotein capable of binding exocyclic 1,2-diols within surface glycans of human pathogens such as Streptococcus pneumoniae, Vibrio cholerae, and Helicobacter pylori. For the latter, lectin binding was shown to cause bacterial agglutination and increased phagocytosis, suggesting a role for hIntL-1 in pathogen surveillance. In this study, we investigated the interactions between hIntL-1 and S. pneumoniae, the leading cause of bacterial pneumonia. We show that hIntL-1 also agglutinates S. pneumoniae serotype 43, which displays an exocyclic 1,2-diol moiety in its capsular polysaccharide but is unable to kill in a complement-dependent manner or to promote bacterial killing by peripheral blood mononuclear cells. In contrast, hIntL-1 not only significantly increases serotype-specific S. pneumoniae killing by neutrophils but also enhances the attachment of these bacteria to A549 lung epithelial cells. Taken together, our results suggest that hIntL-1 participates in host surveillance through microbe sequestration and enhanced targeting to neutrophils.
Collapse
Affiliation(s)
- Silke Andresen
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| | - Kayla Fantone
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Digantkumar Chapla
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, USA
| | - Kelley W. Moremen
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Michael Pierce
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
- Department of Biochemistry and Molecular Biology, University of Georgia, Athens, Georgia, USA
| | - Christine M. Szymanski
- Department of Microbiology, University of Georgia, Athens, Georgia, USA
- Complex Carbohydrate Research Center, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
10
|
Shalaby MA, Anwar MM, Saeed H. Nanomaterials for application in wound Healing: current state-of-the-art and future perspectives. JOURNAL OF POLYMER RESEARCH 2022. [DOI: 10.1007/s10965-021-02870-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
AbstractNanoparticles are the gateway to the new era in drug delivery of biocompatible agents. Several products have emerged from nanomaterials in quest of developing practical wound healing dressings that are nonantigenic, antishear stress, and gas-exchange permeable. Numerous studies have isolated and characterised various wound healing nanomaterials and nanoproducts. The electrospinning of natural and synthetic materials produces fine products that can be mixed with other wound healing medications and herbs. Various produced nanomaterials are highly influential in wound healing experimental models and can be used commercially as well. This article reviewed the current state-of-the-art and briefly specified the future concerns regarding the different systems of nanomaterials in wound healing (i.e., inorganic nanomaterials, organic and hybrid nanomaterials, and nanofibers). This review may be a comprehensive guidance to help health care professionals identify the proper wound healing materials to avoid the usual wound complications.
Collapse
|
11
|
Droste A, Chaves G, Stein S, Trzmiel A, Schweizer M, Karl H, Musset B. Zinc accelerates respiratory burst termination in human PMN. Redox Biol 2021; 47:102133. [PMID: 34562872 PMCID: PMC8476447 DOI: 10.1016/j.redox.2021.102133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/09/2021] [Accepted: 09/12/2021] [Indexed: 11/25/2022] Open
Abstract
The respiratory burst of phagocytes is essential for human survival. Innate immune defence against pathogens relies strongly on reactive oxygen species (ROS) production by the NADPH oxidase (NOX2). ROS kill pathogens while the translocation of electrons across the plasma membrane via NOX2 depolarizes the cell. Simultaneously, protons are released into the cytosol. Here, we compare freshly isolated human polymorphonuclear leukocytes (PMN) to the granulocytes-like cell line PLB 985. We are recording ROS production while inhibiting the charge compensating and pH regulating voltage-gated proton channel (HV1). The data suggests that human PMN and the PLB 985 generate ROS via a general mechanism, consistent of NOX2 and HV1. Additionally, we advanced a mathematical model based on the biophysical properties of NOX2 and HV1. Our results strongly suggest the essential interconnection of HV1 and NOX2 during the respiratory burst of phagocytes. Zinc chelation during the time course of the experiments postulates that zinc leads to an irreversible termination of the respiratory burst over time. Flow cytometry shows cell death triggered by high zinc concentrations and PMA. Our data might help to elucidate the complex interaction of proteins during the respiratory burst and contribute to decipher its termination.
Collapse
Affiliation(s)
- Annika Droste
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Department of Gynecology and Obstetrics, Johannes Gutenberg University, Mainz, Germany
| | - Gustavo Chaves
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany
| | - Stefan Stein
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Annette Trzmiel
- Flow Cytometry Unit, Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt, Germany
| | - Matthias Schweizer
- Federal Institute for Vaccines and Biomedicines, Paul-Ehrlich-Institut, Langen, Germany
| | - Hubert Karl
- Department efi, Technische Hochschule Nürnberg Georg Simon Ohm, Nuremberg, Germany
| | - Boris Musset
- Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Nuremberg, Germany; Center of Physiology, Pathophysiology and Biophysics, Paracelsus Medical University, Salzburg, Austria.
| |
Collapse
|
12
|
Szeifert V, Kolonics F, Bartos B, Khamari D, Vági P, Barna L, Ligeti E, Lőrincz ÁM. Mac-1 Receptor Clustering Initiates Production of Pro-Inflammatory, Antibacterial Extracellular Vesicles From Neutrophils. Front Immunol 2021; 12:671995. [PMID: 34456905 PMCID: PMC8397541 DOI: 10.3389/fimmu.2021.671995] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 07/26/2021] [Indexed: 12/18/2022] Open
Abstract
Depending on the prevailing environmental conditions, neutrophilic granulocytes release extracellular vesicles (EV) which have either anti-inflammatory effects on other neutrophils or pro-inflammatory and antibacterial effects. In the present study we investigated the molecular mechanisms underlying the biogenesis of functionally heterogenic EVs. We show that selective stimulation of Mac-1 integrin (complement receptor 3) by specific ligands initiates the generation of EVs which are able to impair bacterial growth and to induce the secretion of the pro-inflammatory cytokine IL-8 (aEV). However, direct Mac-1 stimulation results in aEV release only if neutrophils were activated on ligand coated surfaces whereas soluble ligands are ineffective. Using total internal reflection fluorescence (TIRF) microcopy, an increased clustering of Mac-1 molecules could be visualized in neutrophils added to C3bi coated surfaces; moreover antibody induced cluster formation triggers aEV release as well. Mac-1 induced production of aEV apparently necessitates a strong calcium signal as it fully depends on the presence of extracellular calcium. However, initiation of a strong calcium signal by an ionophore only results the generation of EV devoid of any antibacterial or pro-inflammatory effect. Our results thus demonstrate that stimulation and clustering of Mac-1 is necessary and sufficient for initiation of aEV biogenesis. In contrast, an intracellular calcium signal is necessary but by itself not sufficient for the production of antibacterial and pro-inflammatory EVs.
Collapse
Affiliation(s)
| | - Ferenc Kolonics
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Balázs Bartos
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics, Cell- and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Pál Vági
- Nikon Center of Excellence, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Barna
- Nikon Center of Excellence, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Ákos M Lőrincz
- Department of Physiology, Semmelweis University, Budapest, Hungary.,Second Department of Internal Medicine, Szent György Hospital, Székesfehérvár, Hungary
| |
Collapse
|
13
|
Kienle K, Glaser KM, Eickhoff S, Mihlan M, Knöpper K, Reátegui E, Epple MW, Gunzer M, Baumeister R, Tarrant TK, Germain RN, Irimia D, Kastenmüller W, Lämmermann T. Neutrophils self-limit swarming to contain bacterial growth in vivo. Science 2021; 372:372/6548/eabe7729. [PMID: 34140358 DOI: 10.1126/science.abe7729] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 04/29/2021] [Indexed: 12/30/2022]
Abstract
Neutrophils communicate with each other to form swarms in infected organs. Coordination of this population response is critical for the elimination of bacteria and fungi. Using transgenic mice, we found that neutrophils have evolved an intrinsic mechanism to self-limit swarming and avoid uncontrolled aggregation during inflammation. G protein-coupled receptor (GPCR) desensitization acts as a negative feedback control to stop migration of neutrophils when they sense high concentrations of self-secreted attractants that initially amplify swarming. Interference with this process allows neutrophils to scan larger tissue areas for microbes. Unexpectedly, this does not benefit bacterial clearance as containment of proliferating bacteria by neutrophil clusters becomes impeded. Our data reveal how autosignaling stops self-organized swarming behavior and how the finely tuned balance of neutrophil chemotaxis and arrest counteracts bacterial escape.
Collapse
Affiliation(s)
- Korbinian Kienle
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Katharina M Glaser
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Sarah Eickhoff
- Institute of Systems Immunology, University of Würzburg, Max Planck Research Group, Würzburg, Germany
| | - Michael Mihlan
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Konrad Knöpper
- Institute of Systems Immunology, University of Würzburg, Max Planck Research Group, Würzburg, Germany
| | - Eduardo Reátegui
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston, MA, USA.,William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, USA
| | - Maximilian W Epple
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.,International Max Planck Research School for Immunobiology, Epigenetics and Metabolism (IMPRS-IEM), Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany.,Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V., Dortmund, Germany
| | - Ralf Baumeister
- Bioinformatics and Molecular Genetics, Faculty of Biology, Centre for Biochemistry and Molecular Cell Research, Faculty of Medicine, Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Teresa K Tarrant
- Division of Rheumatology and Immunology, Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Ronald N Germain
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, Bethesda, MD, USA
| | - Daniel Irimia
- Center for Engineering in Medicine, Massachusetts General Hospital, Harvard Medical School, Shriners Hospital for Children, Boston, MA, USA
| | - Wolfgang Kastenmüller
- Institute of Systems Immunology, University of Würzburg, Max Planck Research Group, Würzburg, Germany
| | - Tim Lämmermann
- Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany.
| |
Collapse
|
14
|
Fantone K, Tucker SL, Miller A, Yadav R, Bernardy EE, Fricker R, Stecenko AA, Goldberg JB, Rada B. Cystic Fibrosis Sputum Impairs the Ability of Neutrophils to Kill Staphylococcus aureus. Pathogens 2021; 10:pathogens10060703. [PMID: 34200034 PMCID: PMC8229215 DOI: 10.3390/pathogens10060703] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) airway disease is characterized by chronic microbial infections and infiltration of inflammatory polymorphonuclear (PMN) granulocytes. Staphylococcus aureus (S. aureus) is a major lung pathogen in CF that persists despite the presence of PMNs and has been associated with CF lung function decline. While PMNs represent the main mechanism of the immune system to kill S. aureus, it remains largely unknown why PMNs fail to eliminate S. aureus in CF. The goal of this study was to observe how the CF airway environment affects S. aureus killing by PMNs. PMNs were isolated from the blood of healthy volunteers and CF patients. Clinical isolates of S. aureus were obtained from the airways of CF patients. The results show that PMNs from healthy volunteers were able to kill all CF isolates and laboratory strains of S. aureus tested in vitro. The extent of killing varied among strains. When PMNs were pretreated with supernatants of CF sputum, S. aureus killing was significantly inhibited suggesting that the CF airway environment compromises PMN antibacterial functions. CF blood PMNs were capable of killing S. aureus. Although bacterial killing was inhibited with CF sputum, PMN binding and phagocytosis of S. aureus was not diminished. The S. aureus-induced respiratory burst and neutrophil extracellular trap release from PMNs also remained uninhibited by CF sputum. In summary, our data demonstrate that the CF airway environment limits killing of S. aureus by PMNs and provides a new in vitro experimental model to study this phenomenon and its mechanism.
Collapse
Affiliation(s)
- Kayla Fantone
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Samantha L. Tucker
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Arthur Miller
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Ruchi Yadav
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Eryn E. Bernardy
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Rachel Fricker
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
| | - Arlene A. Stecenko
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Joanna B. Goldberg
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, GA 30322, USA; (E.E.B.); (A.A.S.); (J.B.G.)
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, GA 30602, USA; (K.F.); (S.L.T.); (A.M.); (R.Y.); (R.F.)
- Correspondence:
| |
Collapse
|
15
|
Yan Y, Li Y, Lv M, Li W, Shi HN. Role of p40 phox in host defense against Citrobacter rodentium infection. FEBS Open Bio 2021; 11:1476-1486. [PMID: 33780601 PMCID: PMC8091579 DOI: 10.1002/2211-5463.13155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 03/11/2021] [Accepted: 03/26/2021] [Indexed: 11/25/2022] Open
Abstract
NADPH oxidase (NOX) is a membrane-bound enzyme complex that generates reactive oxygen species (ROS). Mutations in NOX subunit genes have been implicated in the pathogenesis of inflammatory bowel disease (IBD), indicating a crucial role for ROS in regulating host immune responses. In this study, we utilize genetically deficient mice to investigate whether defects in p40phox , one subunit of NOX, impair host immune response in the intestine and aggravate disease in an infection-based (Citrobacter rodentium) model of colitis. We show that p40phox deficiency does not increase susceptibility of mice to C. rodentium infection, as no differences in body weight loss, bacterial clearance, colonic pathology, cytokine production, or immune cell recruitment were observed between p40phox-/- and wild-type mice. Interestingly, higher IL-10 levels were observed in the supernatants of MLN cells and splenocytes isolated from infected p40phox -deficient mice. Further, a higher expression level of inducible nitric oxide synthase (iNOS) was also noted in mice lacking p40phox . In contrast to wild-type mice, p40phox-/- mice exhibited greater NO production after LPS or bacterial antigen re-stimulation. These results suggest that p40phox-/- mice do not develop worsened colitis. While the precise mechanisms are unclear, it may involve the observed alteration in cytokine responses and enhancement in levels of iNOS and NO.
Collapse
Affiliation(s)
- Yanyun Yan
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationCollege of Life SciencesHunan Normal UniversityChangshaChina
| | - Yali Li
- Hunan Provincial Key Laboratory of Animal Intestinal Function and RegulationCollege of Life SciencesHunan Normal UniversityChangshaChina
| | | | | | - Hai Ning Shi
- Mucosal Immunology and Biology Research CenterHarvard Medical SchoolMassachusetts General HospitalCharlestownMAUSA
| |
Collapse
|
16
|
Acute lung injury after exchange transfusion in two newborns with Glucose-6-phosphate dehydrogenase deficiency. Transfus Apher Sci 2021; 60:103133. [PMID: 33846094 DOI: 10.1016/j.transci.2021.103133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 04/01/2021] [Indexed: 11/21/2022]
Abstract
Transfusion-related lung injury (TRALI) is a condition that develops suddenly within the first six hours after a blood transfusion and it is one of the most important causes of blood transfusion-related mortality. There are few data in the literature about TRALI in the neonatal period. We present two newborn patients who developed TRALI after exchange transfusion due to high bilirubin levels. Our first case was a late preterm LGA baby and was on CPAP. The baby was intubated due to sudden deterioration after the exchange transfusion. Our second case was born at term and, an exchange transfusion was performed on the 5th day of life. He developed respiratory distress unexpectedly soon after the exchange transfusion and was intubated. Glucose-6- phosphate dehydrogenase (G6PD) deficiency was detected in both of our cases. We wanted to emphasize that TRALI should be considered in the differential diagnosis of respiratory distress that develops soon after a transfusion in the newborn period and to draw attention to that TRALI may develop more frequently in patients with G6PD deficiency.
Collapse
|
17
|
Thomas C, Nothaft H, Yadav R, Fodor C, Alemka A, Oni O, Bell M, Rada B, Szymanski CM. Characterization of ecotin homologs from Campylobacter rectus and Campylobacter showae. PLoS One 2020; 15:e0244031. [PMID: 33378351 PMCID: PMC7773321 DOI: 10.1371/journal.pone.0244031] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 12/01/2020] [Indexed: 12/18/2022] Open
Abstract
Ecotin, first described in Escherichia coli, is a potent
inhibitor of a broad range of serine proteases including those typically
released by the innate immune system such as neutrophil elastase (NE). Here we
describe the identification of ecotin orthologs in various
Campylobacter species, including Campylobacter
rectus and Campylobacter showae residing in the
oral cavity and implicated in the development and progression of periodontal
disease in humans. To investigate the function of these ecotins in
vitro, the orthologs from C.
rectus and C. showae were
recombinantly expressed and purified from E.
coli. Using CmeA degradation/protection assays,
fluorescence resonance energy transfer and NE activity assays, we found that
ecotins from C. rectus and C.
showae inhibit NE, factor Xa and trypsin, but not the
Campylobacter jejuni serine protease HtrA or its ortholog
in E. coli, DegP. To further evaluate ecotin
function in vivo, an E. coli
ecotin-deficient mutant was complemented with the C.
rectus and C. showae
homologs. Using a neutrophil killing assay, we demonstrate that the low survival
rate of the E. coli ecotin-deficient mutant
can be rescued upon expression of ecotins from C.
rectus and C. showae. In
addition, the C. rectus and
C. showae ecotins partially compensate for
loss of N-glycosylation and increased protease susceptibility in the related
pathogen, Campylobacter jejuni, thus implicating a similar role
for these proteins in the native host to cope with the protease-rich environment
of the oral cavity.
Collapse
Affiliation(s)
- Cody Thomas
- Department of Microbiology and Complex Carbohydrate Research Center,
University of Georgia, Athens, Georgia, United States of
America
| | - Harald Nothaft
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Ruchi Yadav
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Christopher Fodor
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Abofu Alemka
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
| | - Oluwadamilola Oni
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Michael Bell
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens,
Georgia, United States of America
| | - Christine M. Szymanski
- Department of Microbiology and Complex Carbohydrate Research Center,
University of Georgia, Athens, Georgia, United States of
America
- Department of Biological Sciences, University of Alberta, Edmonton,
Alberta, Canada
- * E-mail:
| |
Collapse
|
18
|
Williams TL, Rada B, Tandon E, Gestal MC. "NETs and EETs, a Whole Web of Mess". Microorganisms 2020; 8:E1925. [PMID: 33291570 PMCID: PMC7761834 DOI: 10.3390/microorganisms8121925] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/13/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes that have very distinct functions. Neutrophils are first responders to external threats, and they use different mechanisms to control pathogens. Phagocytosis, reactive oxygen species, and neutrophil extracellular traps (NETs) are some of the mechanisms that neutrophils utilize to fight pathogens. Although there is some controversy as to whether NETs are in fact beneficial or detrimental to the host, it mainly depends on the biological context. NETs can contribute to disease pathogenesis in certain types of diseases, while they are also undeniably critical components of the innate immune response. On the contrary, the role of eosinophils during host immune responses remains to be better elucidated. Eosinophils play an important role during helminthic infections and allergic responses. Eosinophils can function as effector cells in viral respiratory infections, gut bacterial infections, and as modulators of immune responses by driving the balance between Th1 and Th2 responses. In particular, eosinophils have biological activities that appear to be quite similar to those of neutrophils. Both possess bactericidal activity, can activate proinflammatory responses, can modulate adaptive immune responses, can form extracellular traps, and can be beneficial or detrimental to the host according to the underlying pathology. In this review we compare these two cell types with a focus on highlighting their numerous similarities related to extracellular traps.
Collapse
Affiliation(s)
- Tyler L. Williams
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Balázs Rada
- Department of Infectious Diseases, University of Georgia, Athens, GA 30302, USA;
| | - Eshaan Tandon
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| | - Monica C. Gestal
- Department of Microbiology and Immunology, Louisiana State University (LSU), Health Science Center, Shreveport, LA 71103, USA; (T.L.W.); (E.T.)
| |
Collapse
|
19
|
Kolonics F, Kajdácsi E, Farkas VJ, Veres DS, Khamari D, Kittel Á, Merchant ML, McLeish KR, Lőrincz ÁM, Ligeti E. Neutrophils produce proinflammatory or anti-inflammatory extracellular vesicles depending on the environmental conditions. J Leukoc Biol 2020; 109:793-806. [PMID: 32946637 PMCID: PMC8851677 DOI: 10.1002/jlb.3a0320-210r] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/30/2020] [Accepted: 08/21/2020] [Indexed: 12/15/2022] Open
Abstract
Extracellular vesicles (EVs) are important elements of intercellular communication. A plethora of different, occasionally even opposite, physiologic and pathologic effects have been attributed to these vesicles in the last decade. A direct comparison of individual observations is however hampered by the significant differences in the way of elicitation, collection, handling, and storage of the investigated vesicles. In the current work, we carried out a careful comparative study on 3, previously characterized types of EVs produced by neutrophilic granulocytes. We investigated in parallel the modulation of multiple blood-related cells and functions by medium-sized vesicles. We show that EVs released from resting neutrophils exert anti-inflammatory action by reducing production of reactive oxygen species (ROS) and cytokine release from neutrophils. In contrast, vesicles generated upon encounter of neutrophils with opsonized particles rather promote proinflammatory processes as they increase production of ROS and cytokine secretion from neutrophils and activate endothelial cells. EVs released from apoptosing cells were mainly active in promoting coagulation. We thus propose that EVs are “custom made,” acquiring selective capacities depending on environmental factors prevailing at the time of their biogenesis.
Collapse
Affiliation(s)
- Ferenc Kolonics
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Erika Kajdácsi
- Research Laboratory of the 3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Veronika J Farkas
- Department of Medical Biochemistry, Semmelweis University, Budapest, Hungary
| | - Dániel S Veres
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Delaram Khamari
- Department of Genetics and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, Eötvös Loránd Research Network (ELRN), Budapest, Hungary
| | - Michael L Merchant
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Kenneth R McLeish
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Ákos M Lőrincz
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Gingerich AD, Doja F, Thomason R, Tóth E, Bradshaw JL, Douglass MV, McDaniel LS, Rada B. Oxidative killing of encapsulated and nonencapsulated Streptococcus pneumoniae by lactoperoxidase-generated hypothiocyanite. PLoS One 2020; 15:e0236389. [PMID: 32730276 PMCID: PMC7392276 DOI: 10.1371/journal.pone.0236389] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 07/06/2020] [Indexed: 01/04/2023] Open
Abstract
Streptococcus pneumoniae (Pneumococcus) infections affect millions of people worldwide, cause serious mortality and represent a major economic burden. Despite recent successes due to pneumococcal vaccination and antibiotic use, Pneumococcus remains a significant medical problem. Airway epithelial cells, the primary responders to pneumococcal infection, orchestrate an extracellular antimicrobial system consisting of lactoperoxidase (LPO), thiocyanate anion and hydrogen peroxide (H2O2). LPO oxidizes thiocyanate using H2O2 into the final product hypothiocyanite that has antimicrobial effects against a wide range of microorganisms. However, hypothiocyanite’s effect on Pneumococcus has never been studied. Our aim was to determine whether hypothiocyanite can kill S. pneumoniae. Bactericidal activity was measured in a cell-free in vitro system by determining the number of surviving pneumococci via colony forming units on agar plates, while bacteriostatic activity was assessed by measuring optical density of bacteria in liquid cultures. Our results indicate that hypothiocyanite generated by LPO exerted robust killing of both encapsulated and nonencapsulated pneumococcal strains. Killing of S. pneumoniae by a commercially available hypothiocyanite-generating product was even more pronounced than that achieved with laboratory reagents. Catalase, an H2O2 scavenger, inhibited killing of pneumococcal by hypothiocyanite under all circumstances. Furthermore, the presence of the bacterial capsule or lytA-dependent autolysis had no effect on hypothiocyanite-mediated killing of pneumococci. On the contrary, a pneumococcal mutant deficient in pyruvate oxidase (main bacterial H2O2 source) had enhanced susceptibility to hypothiocyanite compared to its wild-type strain. Overall, results shown here indicate that numerous pneumococcal strains are susceptible to LPO-generated hypothiocyanite.
Collapse
Affiliation(s)
- Aaron D. Gingerich
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Fayhaa Doja
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Rachel Thomason
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Eszter Tóth
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Jessica L. Bradshaw
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Martin V. Douglass
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
| | - Larry S. McDaniel
- Department of Microbiology and Immunology, University of Mississippi Medical Center, Jackson, Mississippi, United States of America
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, The University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
21
|
Cyr JL, Gawriluk TR, Kimani JM, Rada B, Watford WT, Kiama SG, Seifert AW, Ezenwa VO. Regeneration-Competent and -Incompetent Murids Differ in Neutrophil Quantity and Function. Integr Comp Biol 2020; 59:1138-1149. [PMID: 30989211 DOI: 10.1093/icb/icz023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Regeneration is rare in mammals, but spiny mice (Acomys spp.) naturally regenerate skin and ear holes. Inflammation is thought to inhibit regeneration during wound healing, but aspects of inflammation contribute to both regeneration and pathogen defense. We compared neutrophil traits among uninjured, regeneration-competent (Acomys: A. cahirinus, A. kempi, A. percivali) and -incompetent (Mus musculus: Swiss Webster, wild-caught strains) murids to test for constitutive differences in neutrophil quantity and function between these groups. Neutrophil quantity differed significantly among species. In blood, Acomys had lower percentages of circulating neutrophils than Mus; and in bone marrow, Acomys had higher percentages of band neutrophils and lower percentages of segmented neutrophils. Functionally, Acomys and Mus neutrophils did not differ in their ability to migrate or produce reactive oxygen species, but Acomys neutrophils phagocytosed more fungal zymosan. Despite this enhanced phagocytosis activity, Acomys neutrophils were not more effective than Mus neutrophils at killing Escherichia coli. Interestingly, whole blood bacteria killing was dominated by serum in Acomys versus neutrophils only or neutrophils and serum in Mus, suggesting that Acomys primarily rely on serum to kill bacteria whereas Mus do not. These subtle differences in neutrophil traits may allow regeneration-competent species to offset damaging effects of inflammation without compromising pathogen defense.
Collapse
Affiliation(s)
- Jennifer L Cyr
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Thomas R Gawriluk
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA
| | - John M Kimani
- Department of Veterinary Anatomy and Physiology, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Wendy T Watford
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Stephen G Kiama
- Department of Veterinary Anatomy and Physiology, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY 40506, USA.,Department of Veterinary Anatomy and Physiology, University of Nairobi, P.O. Box 30197-00100, Nairobi, Kenya
| | - Vanessa O Ezenwa
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA.,Odum School of Ecology, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
22
|
Lőrincz ÁM, Szeifert V, Bartos B, Szombath D, Mócsai A, Ligeti E. Different Calcium and Src Family Kinase Signaling in Mac-1 Dependent Phagocytosis and Extracellular Vesicle Generation. Front Immunol 2019; 10:2942. [PMID: 31921192 PMCID: PMC6928112 DOI: 10.3389/fimmu.2019.02942] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Accepted: 11/29/2019] [Indexed: 01/18/2023] Open
Abstract
Encountering opsonized particles by neutrophils results in phagocytosis of the particle and generation of extracellular vesicles with antibacterial property (aEV). The aim of the present study is to compare the involvement of different receptors and receptor-proximal signaling pathways in these two parallel processes. Investigating human neutrophils from peripheral blood, we show that complement receptors are decisive for both processes whereas immunoglobulin binding Fc receptors (FcR) only participate moderately in phagocytosis and pattern recognition receptors induce mild EV production but only minimal phagocytosis. Studying bone marrow derived neutrophils of genetically modified animals we verify that the involved complement receptor is CR3, also known as the β2 integrin Mac-1. We show that genetic deletion of the adaptor molecules FcRγ chain or DAP12 does not influence either process, suggesting potential redundant function. Combined absence of the Src family kinases Hck, Fgr, and Lyn drastically impairs phagocytosis but does not influence aEV production. In contrast, deletion of PLCγ2 has no influence on phagocytosis, but reduces aEV formation. In accord with the essential role of PLCγ2, aEV biogenesis both from murine and from human neutrophils is dependent on presence of extracellular calcium. Absence of external calcium prevented the generation of antibacterial EVs, whereas the spontaneous EV formation was not influenced. We thus show that phagocytosis and biogenesis of antibacterial EVs are independent processes and proceed on different signaling pathways although the same receptor plays the critical role in both. Our data reveal the possibility in neutrophilic granulocytes to modulate aEV production without disturbing the phagocytic process.
Collapse
Affiliation(s)
- Ákos M Lőrincz
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Balázs Bartos
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Dávid Szombath
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
23
|
Lőrincz ÁM, Bartos B, Szombath D, Szeifert V, Timár CI, Turiák L, Drahos L, Kittel Á, Veres DS, Kolonics F, Mócsai A, Ligeti E. Role of Mac-1 integrin in generation of extracellular vesicles with antibacterial capacity from neutrophilic granulocytes. J Extracell Vesicles 2019; 9:1698889. [PMID: 31853340 PMCID: PMC6913618 DOI: 10.1080/20013078.2019.1698889] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 10/01/2019] [Accepted: 11/19/2019] [Indexed: 12/13/2022] Open
Abstract
Production of extracellular vesicles (EVs) involved in intercellular communication is a common capacity of most cell types. Upon encountering opsonized microorganisms, neutrophilic granulocytes release EVs that compromise bacterial growth. We carried out a systematic investigation of the involvement of potential opsonin receptors in EV-generation from human and murine neutrophils. Applying flow cytometric, proteomic and functional analysis as well as using genetically modified mice, we demonstrate that formation of antibacterial EVs depends upon stimulation of the multifunctional Mac-1 integrin complex, also called as complement receptor 3 (CR3), whereas activation of immunoglobulin binding Fc receptors or pattern recognition receptors alone or in combination is ineffective. Mac-1/CR3 stimulation and downstream tyrosine kinase signalling affect both the numbers, the cargo content and the antibacterial capacity of the produced vesicles. In contrast, Mac-1/CR3 signalling is not required for spontaneous EV formation, clearly indicating the existence of separate molecular pathways in EV biogenesis. We propose that EVs are “tailor-made” with different composition and functional properties depending on the environmental circumstances.
Collapse
Affiliation(s)
- Ákos M Lőrincz
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Balázs Bartos
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Dávid Szombath
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | | | - Csaba I Timár
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Lilla Turiák
- MS Proteomics Research Group, Research Centre for Natural Science, Hungarian Academy of Sciences, Budapest, Hungary
| | - László Drahos
- MS Proteomics Research Group, Research Centre for Natural Science, Hungarian Academy of Sciences, Budapest, Hungary
| | - Ágnes Kittel
- Experimental Research Institute of Hungarian Academy of Sciences, Budapest, Hungary
| | - Dániel S Veres
- Department of Biophysics and Radiation Biology, Semmelweis University, Budapest, Hungary
| | - Ferenc Kolonics
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Attila Mócsai
- Department of Physiology, Semmelweis University, Budapest, Hungary
| | - Erzsébet Ligeti
- Department of Physiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
24
|
Yen WC, Wu YH, Wu CC, Lin HR, Stern A, Chen SH, Shu JC, Tsun-Yee Chiu D. Impaired inflammasome activation and bacterial clearance in G6PD deficiency due to defective NOX/p38 MAPK/AP-1 redox signaling. Redox Biol 2019; 28:101363. [PMID: 31707353 PMCID: PMC6854078 DOI: 10.1016/j.redox.2019.101363] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/18/2019] [Accepted: 10/25/2019] [Indexed: 01/11/2023] Open
Abstract
Glucose-6-phosphate dehydrogenase (G6PD) is the rate-limiting enzyme of the pentose phosphate pathway that modulates cellular redox homeostasis via the regeneration of NADPH. G6PD-deficient cells have a reduced ability to induce the innate immune response, thus increasing host susceptibility to pathogen infections. An important part of the immune response is the activation of the inflammasome. G6PD-deficient peripheral blood mononuclear cells (PBMCs) from patients and human monocytic (THP-1) cells were used as models to investigate whether G6PD modulates inflammasome activation. A decreased expression of IL-1β was observed in both G6PD-deficient PBMCs and PMA-primed G6PD-knockdown (G6PD-kd) THP-1 cells upon lipopolysaccharide (LPS)/adenosine triphosphate (ATP) or LPS/nigericin stimulation. The pro-IL-1β expression of THP-1 cells was decreased by G6PD knockdown at the transcriptional and translational levels in an investigation of the expression of the inflammasome subunits. The phosphorylation of p38 MAPK and downstream c-Fos expression were decreased upon G6PD knockdown, accompanied by decreased AP-1 translocation into the nucleus. Impaired inflammasome activation in G6PD-kd THP-1 cells was mediated by a decrease in the production of reactive oxygen species (ROS) by NOX signaling, while treatment with hydrogen peroxide (H2O2) enhanced inflammasome activation in G6PD-kd THP-1 cells. G6PD knockdown decreased Staphylococcus aureus and Escherichia coli clearance in G6PD-kd THP-1 cells and G6PD-deficient PBMCs following inflammasome activation. These findings support the notion that enhanced pathogen susceptibility in G6PD deficiency is, in part, due to an altered redox signaling, which adversely affects inflammasome activation and the bactericidal response.
Collapse
Affiliation(s)
- Wei-Chen Yen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Chih-Ching Wu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Molecular Medicine Research Center, Chang Gung University, Tao-Yuan, Taiwan; Department of Otolaryngology - Head & Neck Surgery, Chang Gung Memorial Hospital, Tao-Yuan, Taiwan
| | - Hsin-Ru Lin
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Arnold Stern
- New York University School of Medicine, New York, NY, USA
| | - Shih-Hsiang Chen
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Jwu-Ching Shu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Daniel Tsun-Yee Chiu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan; Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
| |
Collapse
|
25
|
Rocco-Machado N, Cosentino-Gomes D, Nascimento MT, Paes-Vieira L, Khan YA, Mittra B, Andrews NW, Meyer-Fernandes JR. Leishmania amazonensis ferric iron reductase (LFR1) is a bifunctional enzyme: Unveiling a NADPH oxidase activity. Free Radic Biol Med 2019; 143:341-353. [PMID: 31446054 DOI: 10.1016/j.freeradbiomed.2019.08.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/21/2019] [Indexed: 01/26/2023]
Abstract
Leishmania amazonensis is one of leishmaniasis' causative agents, a disease that has no cure and leads to the appearance of cutaneous lesions. Recently, our group showed that heme activates a Na+/K+ ATPase in these parasites through a signaling cascade involving hydrogen peroxide (H2O2) generation. Heme has a pro-oxidant activity and signaling capacity, but the mechanism by which this molecule increases H2O2 levels in L. amazonensis has not been elucidated. Here we investigated the source of H2O2 stimulated by heme, ruling out the participation of mitochondria and raising the possibility of a role for a NADPH oxidase (Nox) activity. Despite the absence of a classical Nox sequence in trypanosomatid genomes, L. amazonensis expresses a surface ferric iron reductase (LFR1). Interestingly, Nox enzymes are thought to have evolved from ferric iron reductases because they share same core domain and are very similar in structure. The main difference is that Nox catalyses electron flow from NADPH to oxygen, generating reactive oxygen species (ROS), while ferric iron reductase promotes electron flow to ferric iron, generating ferrous iron. Using L. amazonensis overexpressing or knockout for LFR1 and heterologous expression of LFR1 in mammalian embryonic kidney (HEK 293) cells, we show that this enzyme is bifunctional, being able to generate both ferrous iron and H2O2. It was previously described that protozoans knockout for LFR1 have their differentiation to virulent forms (amastigote and metacyclic promastigote) impaired. In this work, we observed that LFR1 overexpression stimulates protozoan differentiation to amastigote forms, reinforcing the importance of this enzyme in L. amazonensis life cycle regulation. Thus, we not only identified a new source of ROS production in Leishmania, but also described, for the first time, an enzyme with both ferric iron reductase and Nox activities.
Collapse
Affiliation(s)
- N Rocco-Machado
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| | - D Cosentino-Gomes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of Chemistry, Department of Biochemistry, Universidade Federal Rural do Rio de Janeiro, Seropédica, RJ, Brazil
| | - M T Nascimento
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| | - L Paes-Vieira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil
| | - Y A Khan
- Department of Cell Biology and Molecular Genetics, University of Maryland, 20742, College Park, MD, United States
| | - B Mittra
- Department of Cell Biology and Molecular Genetics, University of Maryland, 20742, College Park, MD, United States
| | - N W Andrews
- Department of Cell Biology and Molecular Genetics, University of Maryland, 20742, College Park, MD, United States
| | - J R Meyer-Fernandes
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro (UFRJ), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil; Institute of National Science and Technology of Structural Biology and Bioimage (INCTBEB), CCS, Cidade Universitária, Ilha do Fundão, 21941-590, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
26
|
Abstract
Immune cell populations determine the balance between ongoing damage and repair following tissue injury. Cells responding to a tissue-damaged environment have significant bioenergetic and biosynthetic needs. In addition to supporting these needs, metabolic pathways govern the function of pro-repair immune cells, including regulatory T cells and tissue macrophages. In this Review, we explore how specific features of the tissue-damaged environment such as hypoxia, oxidative stress, and nutrient depletion serve as metabolic cues to promote or impair the reparative functions of immune cell populations. Hypoxia, mitochondrial DNA stress, and altered redox balance each contribute to mechanisms regulating the response to tissue damage. For example, hypoxia induces changes in regulatory T cell and macrophage metabolic profiles, including generation of 2-hydroxyglutarate, which inhibits demethylase reactions to modulate cell fate and function. Reactive oxygen species abundant in oxidative environments cause damage to mitochondrial DNA, initiating signaling pathways that likewise control pro-repair cell function. Nutrient depletion following tissue damage also affects pro-repair cell function through metabolic signaling pathways, specifically those sensitive to the redox state of the cell. The study of immunometabolism as an immediate sensor and regulator of the tissue-damaged environment provides opportunities to consider mechanisms that facilitate healthy repair of tissue injury.
Collapse
|
27
|
The function of TRP channels in neutrophil granulocytes. Pflugers Arch 2018; 470:1017-1033. [PMID: 29717355 DOI: 10.1007/s00424-018-2146-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 04/09/2018] [Accepted: 04/13/2018] [Indexed: 02/07/2023]
Abstract
Neutrophil granulocytes are exposed to widely varying microenvironmental conditions when pursuing their physiological or pathophysiological functions such as fighting invading bacteria or infiltrating cancer tissue. Examples for harsh environmental challenges include among others mechanical shear stress during the recruitment from the vasculature or the hypoxic and acidotic conditions within the tumor microenvironment. Chemokine gradients, reactive oxygen species, pressure, matrix elasticity, and temperature can be added to the list of potential challenges. Transient receptor potential (TRP) channels serve as cellular sensors since they respond to many of the abovementioned environmental stimuli. The present review investigates the role of TRP channels in neutrophil granulocytes and their role in regulating and adapting neutrophil function to microenvironmental cues. Following a brief description of neutrophil functions, we provide an overview of the electrophysiological characterization of neutrophilic ion channels. We then summarize the function of individual TRP channels in neutrophil granulocytes with a focus on TRPC6 and TRPM2 channels. We close the review by discussing the impact of the tumor microenvironment of pancreatic ductal adenocarcinoma (PDAC) on neutrophil granulocytes. Since neutrophil infiltration into PDAC tissue contributes to disease progression, we propose neutrophilic TRP channel blockade as a potential therapeutic option.
Collapse
|
28
|
DeCoursey TE. Voltage and pH sensing by the voltage-gated proton channel, H V1. J R Soc Interface 2018; 15:20180108. [PMID: 29643227 PMCID: PMC5938591 DOI: 10.1098/rsif.2018.0108] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 03/19/2018] [Indexed: 12/15/2022] Open
Abstract
Voltage-gated proton channels are unique ion channels, membrane proteins that allow protons but no other ions to cross cell membranes. They are found in diverse species, from unicellular marine life to humans. In all cells, their function requires that they open and conduct current only under certain conditions, typically when the electrochemical gradient for protons is outwards. Consequently, these proteins behave like rectifiers, conducting protons out of cells. Their activity has electrical consequences and also changes the pH on both sides of the membrane. Here we summarize what is known about the way these proteins sense the membrane potential and the pH inside and outside the cell. Currently, it is hypothesized that membrane potential is sensed by permanently charged arginines (with very high pKa) within the protein, which results in parts of the protein moving to produce a conduction pathway. The mechanism of pH sensing appears to involve titratable side chains of particular amino acids. For this purpose their pKa needs to be within the operational pH range. We propose a 'counter-charge' model for pH sensing in which electrostatic interactions within the protein are selectively disrupted by protonation of internally or externally accessible groups.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Physiology & Biophysics, Rush University, 1750 West Harrison, Chicago, IL 60612, USA
| |
Collapse
|
29
|
Lőrincz ÁM, Szeifert V, Bartos B, Ligeti E. New flow cytometry-based method for the assessment of the antibacterial effect of immune cells and subcellular particles. J Leukoc Biol 2018. [DOI: 10.1002/jlb.4ta0817-317r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
- Ákos M. Lőrincz
- Department of Physiology; Semmelweis University; Budapest Hungary
| | | | - Balázs Bartos
- Department of Physiology; Semmelweis University; Budapest Hungary
| | - Erzsébet Ligeti
- Department of Physiology; Semmelweis University; Budapest Hungary
| |
Collapse
|
30
|
Shin B, Park C, Imlay JA, Park W. 4-Hydroxybenzaldehyde sensitizes Acinetobacter baumannii to amphenicols. Appl Microbiol Biotechnol 2018; 102:2323-2335. [PMID: 29387955 DOI: 10.1007/s00253-018-8791-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 01/02/2018] [Accepted: 01/16/2018] [Indexed: 12/21/2022]
Abstract
Bacterial metabolism modulated by environmental chemicals could alter antibiotic susceptibility. 4-Hydroxybenzaldehyde (4-HBA), which cannot support the growth of Acinetobacter baumannii, exhibited synergism only with amphenicol antibiotics including chloramphenicol (CAM) and thiamphenicol. Interestingly, this synergistic effect was not observed with other growth-supporting, structurally similar compounds such as 4-hydroxybenzoate. Transcriptomic analysis demonstrated that genes involved in protocatechuate metabolism (pca genes) and osmotic stress (bet genes) were significantly upregulated by 4-HBA and CAM treatment. The 14C-labeled CAM influx was lower in a pcaK1 (encoding a transporter of protocatechuate) deletion mutant and was higher in the pcaK1 overexpressing cells relative to that in the wild type upon 4-HBA treatment. Our kinetic data using 14C-labeled CAM clearly showed that CAM uptake is possibly through facilitated diffusion. Deletion of pcaK1 did not result in the elimination of CAM influx, indicating that CAM does not enter only through PcaK1. The amount of 4-HBA in the culture supernatant was, however, unaffected during the test conditions, validating that it was not metabolized by the bacteria. CAM resistant A. baumannii cells derived by serial passages through CAM-amended media exhibited lower level of pcaK1 gene expression. These results led us to conclude that the activation of PcaK1 transporter is probably linked to cellular CAM susceptibility. This is the first report showing a relationship between CAM influx and aromatic compound metabolism in A. baumannii.
Collapse
Affiliation(s)
- Bora Shin
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Chulwoo Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - James A Imlay
- Department of Microbiology, University of Illinois, Urbana, IL, 61801, USA
| | - Woojun Park
- Laboratory of Molecular Environmental Microbiology, Department of Environmental Sciences and Ecological Engineering, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
31
|
Buvelot H, Posfay-Barbe KM, Linder P, Schrenzel J, Krause KH. Staphylococcus aureus, phagocyte NADPH oxidase and chronic granulomatous disease. FEMS Microbiol Rev 2017; 41:139-157. [PMID: 27965320 DOI: 10.1093/femsre/fuw042] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2016] [Indexed: 11/14/2022] Open
Abstract
Dysfunction of phagocytes is a relevant risk factor for staphylococcal infection. The most common hereditary phagocyte dysfunction is chronic granulomatous disease (CGD), characterized by impaired generation of reactive oxygen species (ROS) due to loss of function mutations within the phagocyte NADPH oxidase NOX2. Phagocytes ROS generation is fundamental to eliminate pathogens and to regulate the inflammatory response to infection. CGD is characterized by recurrent and severe bacterial and fungal infections, with Staphylococcus aureus as the most frequent pathogen, and skin and lung abscesses as the most common clinical entities. Staphylococcus aureus infection may occur in virtually any human host, presumably because of the many virulence factors of the bacterium. However, in the presence of functional NOX2, staphylococcal infections remain rare and are mainly linked to breaches of the skin barrier. In contrast, in patients with CGD, S. aureus readily survives and frequently causes clinically apparent disease. Astonishingly, little is known why S. aureus, which possesses a wide range of antioxidant enzymes (e.g. catalase, SOD), is particularly sensitive to control through NOX2. In this review, we will evaluate the discovery of CGD and our present knowledge of the role of NOX2 in S. aureus infection.
Collapse
Affiliation(s)
- Helene Buvelot
- Division of General Internal Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Klara M Posfay-Barbe
- Paediatric Infectious Diseases Unit, Department of Paediatrics, University Hospitals of Geneva, 1205 Geneva and Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Patrick Linder
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| | - Jacques Schrenzel
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland
| | - Karl-Heinz Krause
- Divisions of Infectious Diseases and Laboratory Medicine, Geneva University Hospitals, CH-1211 Geneva 4, Switzerland.,Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1206 Geneva, Switzerland
| |
Collapse
|
32
|
Metallothioneins: Emerging Modulators in Immunity and Infection. Int J Mol Sci 2017; 18:ijms18102197. [PMID: 29065550 PMCID: PMC5666878 DOI: 10.3390/ijms18102197] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/14/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Metallothioneins (MTs) are a family of metal-binding proteins virtually expressed in all organisms including prokaryotes, lower eukaryotes, invertebrates and mammals. These proteins regulate homeostasis of zinc (Zn) and copper (Cu), mitigate heavy metal poisoning, and alleviate superoxide stress. In recent years, MTs have emerged as an important, yet largely underappreciated, component of the immune system. Innate and adaptive immune cells regulate MTs in response to stress stimuli, cytokine signals and microbial challenge. Modulation of MTs in these cells in turn regulates metal ion release, transport and distribution, cellular redox status, enzyme function and cell signaling. While it is well established that the host strictly regulates availability of metal ions during microbial pathogenesis, we are only recently beginning to unravel the interplay between metal-regulatory pathways and immunological defenses. In this perspective, investigation of mechanisms that leverage the potential of MTs to orchestrate inflammatory responses and antimicrobial defenses has gained momentum. The purpose of this review, therefore, is to illumine the role of MTs in immune regulation. We discuss the mechanisms of MT induction and signaling in immune cells and explore the therapeutic potential of the MT-Zn axis in bolstering immune defenses against pathogens.
Collapse
|
33
|
Hyperbaric oxygen therapy augments tobramycin efficacy in experimental Staphylococcus aureus endocarditis. Int J Antimicrob Agents 2017; 50:406-412. [DOI: 10.1016/j.ijantimicag.2017.04.025] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/24/2017] [Accepted: 04/05/2017] [Indexed: 12/14/2022]
|
34
|
Nguyen GT, Green ER, Mecsas J. Neutrophils to the ROScue: Mechanisms of NADPH Oxidase Activation and Bacterial Resistance. Front Cell Infect Microbiol 2017; 7:373. [PMID: 28890882 PMCID: PMC5574878 DOI: 10.3389/fcimb.2017.00373] [Citation(s) in RCA: 474] [Impact Index Per Article: 59.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/02/2017] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen species (ROS) generated by NADPH oxidase play an important role in antimicrobial host defense and inflammation. Their deficiency in humans results in recurrent and severe bacterial infections, while their unregulated release leads to pathology from excessive inflammation. The release of high concentrations of ROS aids in clearance of invading bacteria. Localization of ROS release to phagosomes containing pathogens limits tissue damage. Host immune cells, like neutrophils, also known as PMNs, will release large amounts of ROS at the site of infection following the activation of surface receptors. The binding of ligands to G-protein-coupled receptors (GPCRs), toll-like receptors, and cytokine receptors can prime PMNs for a more robust response if additional signals are encountered. Meanwhile, activation of Fc and integrin directly induces high levels of ROS production. Additionally, GPCRs that bind to the bacterial-peptide analog fMLP, a neutrophil chemoattractant, can both prime cells and trigger low levels of ROS production. Engagement of these receptors initiates intracellular signaling pathways, resulting in activation of downstream effector proteins, assembly of the NADPH oxidase complex, and ultimately, the production of ROS by this complex. Within PMNs, ROS released by the NADPH oxidase complex can activate granular proteases and induce the formation of neutrophil extracellular traps (NETs). Additionally, ROS can cross the membranes of bacterial pathogens and damage their nucleic acids, proteins, and cell membranes. Consequently, in order to establish infections, bacterial pathogens employ various strategies to prevent restriction by PMN-derived ROS or downstream consequences of ROS production. Some pathogens are able to directly prevent the oxidative burst of phagocytes using secreted effector proteins or toxins that interfere with translocation of the NADPH oxidase complex or signaling pathways needed for its activation. Nonetheless, these pathogens often rely on repair and detoxifying proteins in addition to these secreted effectors and toxins in order to resist mammalian sources of ROS. This suggests that pathogens have both intrinsic and extrinsic mechanisms to avoid restriction by PMN-derived ROS. Here, we review mechanisms of oxidative burst in PMNs in response to bacterial infections, as well as the mechanisms by which bacterial pathogens thwart restriction by ROS to survive under conditions of oxidative stress.
Collapse
Affiliation(s)
- Giang T Nguyen
- Graduate Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts UniversityBoston, MA, United States
| | - Erin R Green
- Department of Molecular Biology and Microbiology, Tufts University School of MedicineBoston, MA, United States
| | - Joan Mecsas
- Graduate Program in Immunology, Sackler School of Graduate Biomedical Sciences, Tufts UniversityBoston, MA, United States.,Department of Molecular Biology and Microbiology, Tufts University School of MedicineBoston, MA, United States
| |
Collapse
|
35
|
DeCoursey TE. The intimate and controversial relationship between voltage-gated proton channels and the phagocyte NADPH oxidase. Immunol Rev 2017; 273:194-218. [PMID: 27558336 DOI: 10.1111/imr.12437] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
One of the most fascinating and exciting periods in my scientific career entailed dissecting the symbiotic relationship between two membrane transporters, the Nicotinamide adenine dinucleotide phosphate reduced form (NADPH) oxidase complex and voltage-gated proton channels (HV 1). By the time I entered this field, there had already been substantial progress toward understanding NADPH oxidase, but HV 1 were known only to a tiny handful of cognoscenti around the world. Having identified the first proton currents in mammalian cells in 1991, I needed to find a clear function for these molecules if the work was to become fundable. The then-recent discoveries of Henderson, Chappell, and colleagues in 1987-1988 that led them to hypothesize interactions of both molecules during the respiratory burst of phagocytes provided an excellent opportunity. In a nutshell, both transporters function by moving electrical charge across the membrane: NADPH oxidase moves electrons and HV 1 moves protons. The consequences of electrogenic NADPH oxidase activity on both membrane potential and pH strongly self-limit this enzyme. Fortunately, both consequences specifically activate HV 1, and HV 1 activity counteracts both consequences, a kind of yin-yang relationship. Notwithstanding a decade starting in 1995 when many believed the opposite, these are two separate molecules that function independently despite their being functionally interdependent in phagocytes. The relationship between NADPH oxidase and HV 1 has become a paradigm that somewhat surprisingly has now extended well beyond the phagocyte NADPH oxidase - an industrial strength producer of reactive oxygen species (ROS) - to myriad other cells that produce orders of magnitude less ROS for signaling purposes. These cells with their seven NADPH oxidase (NOX) isoforms provide a vast realm of mechanistic obscurity that will occupy future studies for years to come.
Collapse
Affiliation(s)
- Thomas E DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University, Chicago, IL, USA
| |
Collapse
|
36
|
Interactions between Neutrophils and Pseudomonas aeruginosa in Cystic Fibrosis. Pathogens 2017; 6:pathogens6010010. [PMID: 28282951 PMCID: PMC5371898 DOI: 10.3390/pathogens6010010] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/03/2017] [Indexed: 12/23/2022] Open
Abstract
Cystic fibrosis (CF) affects 70,000 patients worldwide. Morbidity and mortality in CF is largely caused by lung complications due to the triad of impaired mucociliary clearance, microbial infections and chronic inflammation. Cystic fibrosis airway inflammation is mediated by robust infiltration of polymorphonuclear neutrophil granulocytes (PMNs, neutrophils). Neutrophils are not capable of clearing lung infections and contribute to tissue damage by releasing their dangerous cargo. Pseudomonas aeruginosa is an opportunistic pathogen causing infections in immunocompromised individuals. P. aeruginosa is a main respiratory pathogen in CF infecting most patients. Although PMNs are key to attack and clear P. aeruginosa in immunocompetent individuals, PMNs fail to do so in CF. Understanding why neutrophils cannot clear P. aeruginosa in CF is essential to design novel therapies. This review provides an overview of the antimicrobial mechanisms by which PMNs attack and eliminate P. aeruginosa. It also summarizes current advances in our understanding of why PMNs are incapable of clearing P. aeruginosa and how this bacterium adapts to and resists PMN-mediated killing in the airways of CF patients chronically infected with P. aeruginosa.
Collapse
|
37
|
Foote JR, Behe P, Frampton M, Levine AP, Segal AW. An Exploration of Charge Compensating Ion Channels across the Phagocytic Vacuole of Neutrophils. Front Pharmacol 2017; 8:94. [PMID: 28293191 PMCID: PMC5329019 DOI: 10.3389/fphar.2017.00094] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 02/13/2017] [Indexed: 11/13/2022] Open
Abstract
Neutrophils phagocytosing bacteria and fungi exhibit a burst of non-mitochondrial respiration that is required to kill and digest the engulfed microbes. This respiration is accomplished by the movement of electrons across the wall of the phagocytic vacuole by the neutrophil NADPH oxidase, NOX2. In this study, we have attempted to identify the non-proton ion channels or transporters involved in charge compensation by examining the effect of inhibitors on vacuolar pH and cross-sectional area, and on oxygen consumption. The chloride channel inhibitors 4-[(2-Butyl-6,7-dichloro-2-cyclopentyl-2,3-dihydro-1-oxo-1H-inden-5-yl)oxy]butanoic acid (DCPIB) and flufenamic acid (FFA) were the most effective inhibitors of alkalinisation in human neutrophil vacuoles, suggesting an efflux of chloride from the vacuole. The proton channel inhibitor, zinc (Zn2+), combined with DCPIB caused more vacuolar swelling than either compound alone, suggesting the conductance of osmotically active cations into the vacuole. Support for cation influx was provided by the broad-spectrum cation transport inhibitors anandamide and quinidine which inhibited vacuolar alkalinisation and swelling when applied with zinc. Oxygen consumption was generally unaffected by these anion or cation inhibitors alone, but when combined with Zn2+ it was dramatically reduced, suggesting that multiple channels in combination can compensate the charge. In an attempt to identify specific channels, we tested neutrophils from knock-out mouse models including CLIC1, ClC3, ClC4, ClC7, KCC3, KCNQ1, KCNE3, KCNJ15, TRPC1/3/5/6, TRPA1/TRPV1, TRPM2, and TRPV2, and double knockouts of CLIC1, ClC3, KCC3, TRPM2, and KCNQ1 with HVCN1, and humans with channelopathies involving BEST1, ClC7, CFTR, and MCOLN1. No gross abnormalities in vacuolar pH or area were found in any of these cells suggesting that we had not tested the correct channel, or that there is redundancy in the system. The respiratory burst was suppressed in the KCC3-/- and enhanced in the CLIC1-/- cells, but was normal in all others, including ClC3-/-. These results suggest charge compensation by a chloride conductance out of the vacuole and by cation/s into it. The identity of these channels remains to be established.
Collapse
Affiliation(s)
- Juliet R Foote
- Division of Medicine, Centre for Molecular Medicine, University College London London, UK
| | - Philippe Behe
- Division of Medicine, Centre for Molecular Medicine, University College London London, UK
| | - Mathew Frampton
- Division of Medicine, Centre for Molecular Medicine, University College London London, UK
| | - Adam P Levine
- Division of Medicine, Centre for Molecular Medicine, University College London London, UK
| | - Anthony W Segal
- Division of Medicine, Centre for Molecular Medicine, University College London London, UK
| |
Collapse
|
38
|
Behnen M, Möller S, Brozek A, Klinger M, Laskay T. Extracellular Acidification Inhibits the ROS-Dependent Formation of Neutrophil Extracellular Traps. Front Immunol 2017; 8:184. [PMID: 28293240 PMCID: PMC5329032 DOI: 10.3389/fimmu.2017.00184] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/09/2017] [Indexed: 11/13/2022] Open
Abstract
The inflammatory microenvironment is commonly characterized by extracellular acidosis (pH < 7.35). Sensitivity to pH, CO2 or bicarbonate concentrations allows neutrophils to react to changes in their environment and to detect inflamed areas in the tissue. One important antimicrobial effector mechanism is the production of neutrophil extracellular traps (NETs), which are released during a programmed reactive oxygen species (ROS)-dependent cell death, the so-called NETosis. Although several functions of neutrophils have been analyzed under acidic conditions, the effect of extracellular acidosis on NETosis remains mainly unexplored and the available experimental results are contradictory. We performed a comprehensive study with the aim to elucidate the effect of extracellular acidosis on ROS-dependent NETosis of primary human neutrophils and to identify the underlying mechanisms. The study was performed in parallel in a CO2–bicabonate-buffered culture medium, which mimics in vivo conditions, and under HEPES-buffered conditions to verify the effect of pH independent of CO2 or bicarbonate. We could clearly show that extracellular acidosis (pH 6.5, 6.0, and 5.5) and intracellular acidification inhibit the release of ROS-dependent NETs upon stimulation of neutrophils with phorbol myristate acetate and immobilized immune complexes. Moreover, our findings suggest that the diminished NET release is a consequence of reduced ROS production and diminished glycolysis of neutrophils under acidic conditions. It was suggested previously that neutrophils can sense the border of inflamed tissue by the pH gradient and that a drop in pH serves as an indicator for the progress of inflammation. Following this hypothesis, our data indicate that an acidic inflammatory environment results in inhibition of extracellular operating effector mechanisms of neutrophils such as release of ROS and NETs. This way the release of toxic components and tissue damage can be avoided. However, we observed that major antimicrobial effector mechanisms such as phagocytosis and the killing of pathogens by neutrophils remain functional under acidic conditions.
Collapse
Affiliation(s)
- Martina Behnen
- Department for Infectious Diseases and Microbiology, University of Lübeck , Lübeck , Germany
| | - Sonja Möller
- Department for Infectious Diseases and Microbiology, University of Lübeck , Lübeck , Germany
| | - Antonia Brozek
- Department for Infectious Diseases and Microbiology, University of Lübeck , Lübeck , Germany
| | | | - Tamás Laskay
- Department for Infectious Diseases and Microbiology, University of Lübeck , Lübeck , Germany
| |
Collapse
|
39
|
Sil P, Hayes CP, Reaves BJ, Breen P, Quinn S, Sokolove J, Rada B. P2Y6 Receptor Antagonist MRS2578 Inhibits Neutrophil Activation and Aggregated Neutrophil Extracellular Trap Formation Induced by Gout-Associated Monosodium Urate Crystals. THE JOURNAL OF IMMUNOLOGY 2016; 198:428-442. [PMID: 27903742 DOI: 10.4049/jimmunol.1600766] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 10/23/2016] [Indexed: 12/27/2022]
Abstract
Human neutrophils (polymorphonuclear leukocytes [PMNs]) generate inflammatory responses within the joints of gout patients upon encountering monosodium urate (MSU) crystals. Neutrophil extracellular traps (NETs) are found abundantly in the synovial fluid of gout patients. The detailed mechanism of MSU crystal-induced NET formation remains unknown. Our goal was to shed light on possible roles of purinergic signaling and neutrophil migration in mediating NET formation induced by MSU crystals. Interaction of human neutrophils with MSU crystals was evaluated by high-throughput live imaging using confocal microscopy. We quantitated NET levels in gout synovial fluid supernatants and detected enzymatically active neutrophil primary granule enzymes, myeloperoxidase, and human neutrophil elastase. Suramin and PPADS, general P2Y receptor blockers, and MRS2578, an inhibitor of the purinergic P2Y6 receptor, blocked NET formation triggered by MSU crystals. AR-C25118925XX (P2Y2 antagonist) did not inhibit MSU crystal-stimulated NET release. Live imaging of PMNs showed that MRS2578 represses neutrophil migration and blocked characteristic formation of MSU crystal-NET aggregates called aggregated NETs. Interestingly, the store-operated calcium entry channel inhibitor (SK&F96365) also reduced MSU crystal-induced NET release. Our results indicate that the P2Y6/store-operated calcium entry/IL-8 axis is involved in MSU crystal-induced aggregated NET formation, but MRS2578 could have additional effects affecting PMN migration. The work presented in the present study could lead to a better understanding of gouty joint inflammation and help improve the treatment and care of gout patients.
Collapse
Affiliation(s)
- Payel Sil
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Craig P Hayes
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Barbara J Reaves
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602
| | - Patrick Breen
- Institute of Bioinformatics, University of Georgia, Athens, GA 30602
| | - Shannon Quinn
- Department of Computer Science, Franklin College of Arts and Sciences, University of Georgia, Athens, 30602 GA
| | - Jeremy Sokolove
- Stanford University School of Medicine, Stanford, CA 94305; and.,Internal Medicine and Rheumatology, VA Palo Alto Health Care System, Palo Alto, CA 94034
| | - Balázs Rada
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, GA 30602;
| |
Collapse
|
40
|
Floyd M, Winn M, Cullen C, Sil P, Chassaing B, Yoo DG, Gewirtz AT, Goldberg JB, McCarter LL, Rada B. Swimming Motility Mediates the Formation of Neutrophil Extracellular Traps Induced by Flagellated Pseudomonas aeruginosa. PLoS Pathog 2016; 12:e1005987. [PMID: 27855208 PMCID: PMC5113990 DOI: 10.1371/journal.ppat.1005987] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 10/10/2016] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen causing severe infections often characterized by robust neutrophilic infiltration. Neutrophils provide the first line of defense against P. aeruginosa. Aside from their defense conferred by phagocytic activity, neutrophils also release neutrophil extracellular traps (NETs) to immobilize bacteria. Although NET formation is an important antimicrobial process, the details of its mechanism are largely unknown. The identity of the main components of P. aeruginosa responsible for triggering NET formation is unclear. In this study, our focus was to identify the main bacterial factors mediating NET formation and to gain insight into the underlying mechanism. We found that P. aeruginosa in its exponential growth phase promoted strong NET formation in human neutrophils while its NET-inducing ability dramatically decreased at later stages of bacterial growth. We identified the flagellum as the primary component of P. aeruginosa responsible for inducing NET extrusion as flagellum-deficient bacteria remained seriously impaired in triggering NET formation. Purified P. aeruginosa flagellin, the monomeric component of the flagellum, does not stimulate NET formation in human neutrophils. P. aeruginosa-induced NET formation is independent of the flagellum-sensing receptors TLR5 and NLRC4 in both human and mouse neutrophils. Interestingly, we found that flagellar motility, not flagellum binding to neutrophils per se, mediates NET release induced by flagellated bacteria. Immotile, flagellar motor-deficient bacterial strains producing paralyzed flagella did not induce NET formation. Forced contact between immotile P. aeruginosa and neutrophils restored their NET-inducing ability. Both the motAB and motCD genetic loci encoding flagellar motor genes contribute to maximal NET release; however the motCD genes play a more important role. Phagocytosis of P. aeruginosa and superoxide production by neutrophils were also largely dependent upon a functional flagellum. Taken together, the flagellum is herein presented for the first time as the main organelle of planktonic bacteria responsible for mediating NET release. Furthermore, flagellar motility, rather than binding of the flagellum to flagellum-sensing receptors on host cells, is required for P. aeruginosa to induce NET release.
Collapse
Affiliation(s)
- Madison Floyd
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
| | - Matthew Winn
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
| | - Christian Cullen
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
| | - Payel Sil
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
| | - Benoit Chassaing
- Center for Inflammation, Immunity, & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Dae-goon Yoo
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
| | - Andrew T. Gewirtz
- Center for Inflammation, Immunity, & Infection, Institute for Biomedical Sciences, Georgia State University, Atlanta, Georgia, United States of America
| | - Joanna B. Goldberg
- Division of Pulmonology, Allergy/Immunology, Cystic Fibrosis and Sleep, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Linda L. McCarter
- Carver College of Medicine, Department of Microbiology, The University of Iowa, Iowa City, Iowa, United States of America
| | - Balázs Rada
- College of Veterinary Medicine, Department of Infectious Diseases, The University of Georgia, Athens, Georgia, United States of America
- * E-mail:
| |
Collapse
|
41
|
Ella K, Csépányi-Kömi R, Káldi K. Circadian regulation of human peripheral neutrophils. Brain Behav Immun 2016; 57:209-221. [PMID: 27132055 DOI: 10.1016/j.bbi.2016.04.016] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 04/13/2016] [Accepted: 04/26/2016] [Indexed: 01/12/2023] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood. Beside being essential responders in bacterial and fungal infections, they also contribute to tissue reactions in many autoimmune and inflammatory diseases. Although several immune responses linked to neutrophil functions have been described to be rhythmic, the mechanism of the circadian regulation of these cells is still not understood. Characterization of the time-of-day-specific control of neutrophil responsiveness could help to better understand the pathomechanism of these inflammatory responses and design effective chronotherapy. Here we report that the time-dependent expression of core clock components in human neutrophils characteristically differs from that in mononuclear cells. Both the low expression and the reduced nuclear accumulation of the essential clock protein BMAL1 suggest that the molecular oscillator is down-regulated in neutrophils. By following the expression of the maturation marker Cxcr4 and morphological attributes (side-scattering properties and nuclear segmentation), we found that the distribution of young and aged cells within the peripheral neutrophil pool displays a daily rhythm. In addition, we detected synchronous fluctuations in the plasma level of the CXCR4 ligand CXCL12, an important regulator of cell trafficking within the bone marrow. We found that expression of another maturation marker, the core component of the superoxide generating NADPH oxidase, and parallelly, the superoxide producing capacity of neutrophils were also dependent on the time of the day. In line with this, number of opsonized bacteria engulfed by neutrophils also showed time-dependent differences, supporting that clearance of pathogens shows a daily rhythm. We suggest that maturation-dependent changes in neutrophil responsiveness rather than the cellular autonomous clock are involved in the daily regulation of human neutrophil functions.
Collapse
Affiliation(s)
- Krisztina Ella
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47, Budapest H-1094, Hungary
| | - Roland Csépányi-Kömi
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47, Budapest H-1094, Hungary
| | - Krisztina Káldi
- Department of Physiology, Semmelweis University, Tűzoltó u. 37-47, Budapest H-1094, Hungary.
| |
Collapse
|
42
|
Siler U, Romao S, Tejera E, Pastukhov O, Kuzmenko E, Valencia RG, Meda Spaccamela V, Belohradsky BH, Speer O, Schmugge M, Kohne E, Hoenig M, Freihorst J, Schulz AS, Reichenbach J. Severe glucose-6-phosphate dehydrogenase deficiency leads to susceptibility to infection and absent NETosis. J Allergy Clin Immunol 2016; 139:212-219.e3. [PMID: 27458052 DOI: 10.1016/j.jaci.2016.04.041] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 03/23/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Glucose-6-phosphate dehydrogenase (G6PD) deficiency is the most common enzymatic disorder of red blood cells in human subjects, causing hemolytic anemia linked to impaired nicotinamide adenine dinucleotide phosphate (NADPH) production and imbalanced redox homeostasis in erythrocytes. Because G6PD is expressed by a variety of hematologic and nonhematologic cells, a broader clinical phenotype could be postulated in G6PD-deficient patients. We describe 3 brothers with severe G6PD deficiency and susceptibility to bacterial infection. OBJECTIVE We sought to study the molecular pathophysiology leading to susceptibility to infection in 3 siblings with severe G6PD deficiency. METHODS Blood samples of 3 patients with severe G6PD deficiency were analyzed for G6PD enzyme activity, cellular oxidized nicotinamide adenine dinucleotide phosphate/NADPH levels, phagocytic reactive oxygen species production, neutrophil extracellular trap (NET) formation, and neutrophil elastase translocation. RESULTS In these 3 brothers strongly reduced NADPH oxidase function was found in granulocytes, leading to impaired NET formation. Defective NET formation has thus far been only observed in patients with the NADPH oxidase deficiency chronic granulomatous disease, who require antibiotic and antimycotic prophylaxis to prevent life-threatening bacterial and fungal infections. CONCLUSION Because severe G6PD deficiency can be a phenocopy of chronic granulomatous disease with regard to the cellular and clinical phenotype, careful evaluation of neutrophil function seems mandatory in these patients to decide on appropriate anti-infective preventive measures. Determining the level of G6PD enzyme activity should be followed by analysis of reactive oxygen species production and NET formation to decide on required antibiotic and antimycotic prophylaxis.
Collapse
Affiliation(s)
- Ulrich Siler
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Susana Romao
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Emilio Tejera
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Oleksandr Pastukhov
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Elena Kuzmenko
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Rocio G Valencia
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Virginia Meda Spaccamela
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Bernd H Belohradsky
- Division of Infectious Diseases and Immunology, Dr. von Haunersches Kinderspital, University Childrens Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Oliver Speer
- Division of Hematology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Markus Schmugge
- Division of Hematology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland
| | - Elisabeth Kohne
- Department of Pediatrics and Adolescent Medicine, University Medical Centre Ulm, Ulm, Germany
| | - Manfred Hoenig
- Department of Pediatrics and Adolescent Medicine, University Medical Centre Ulm, Ulm, Germany
| | | | - Ansgar S Schulz
- Department of Pediatrics and Adolescent Medicine, University Medical Centre Ulm, Ulm, Germany
| | - Janine Reichenbach
- Division of Immunology, University Children's Hospital and Children's Research Centre, Zurich, Switzerland; Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland; Centre for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland; Swiss Center for Regenerative Medicine, University of Zurich, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
43
|
Naqvi AR, Fordham JB, Nares S. MicroRNA target Fc receptors to regulate Ab-dependent Ag uptake in primary macrophages and dendritic cells. Innate Immun 2016; 22:510-21. [PMID: 27449126 DOI: 10.1177/1753425916661042] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 06/24/2016] [Indexed: 12/23/2022] Open
Abstract
Phagocytosis commences with particle internalization and culminates with the activation of innate and adaptive immune responses. However, the role of miRNAs in phagocytosis remains largely unknown. In this study, we examined the role of miR-24, miR-30b and miR-142-3p in Ab Fc receptor (FcR)-mediated phagocytosis by macrophages (MΦ) and dendritic cells (DC). The expression of these miRNAs was reduced following phagocytosis of both IgG-opsonized beads and Escherichia coli, indicating their regulatory role in the process. Further, overexpression of these miRNAs impaired the uptake of IgG-coated latex beads, which corroborated the reduced secretion of the pro-inflammatory cytokines TNF-α and IL-8 and down-regulation of PKC-α, as well as superoxide-generating enzyme NADPH oxidase 2 expression level. Mechanistically, MΦ and DC transfected with miRNA mimics show marked reduction in expression of FcRs including FCGR2A, FcɛR1G and FCER2. We show that FcɛR1G expression is not affected at the transcription level, rather it is post-transcriptionally regulated by miR-30b. Finally, we demonstrate that siRNA-mediated knockdown of FcɛR1G leads to reduced uptake of IgG-opsonized beads, indicating its involvement on Ab-mediated phagocytosis. These results uncover miR-24, miR-30b and miR-142-3p as an essential component of FcR-mediated phagocytosis and associated innate immune responses.
Collapse
Affiliation(s)
- Afsar Raza Naqvi
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jezrom B Fordham
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| | - Salvador Nares
- Department of Periodontics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
44
|
A common theme in extracellular fluids of beetles: extracellular superoxide dismutases crucial for balancing ROS in response to microbial challenge. Sci Rep 2016; 6:24082. [PMID: 27068683 PMCID: PMC4828634 DOI: 10.1038/srep24082] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 03/15/2016] [Indexed: 12/14/2022] Open
Abstract
Extracellular Cu/Zn superoxide dismutases (SODs) are critical for balancing the level of reactive oxygen species in the extracellular matrix of eukaryotes. In the present study we have detected constitutive SOD activity in the haemolymph and defensive secretions of different leaf beetle species. Exemplarily, we have chosen the mustard leaf beetle, Phaedon cochleariae, as representative model organism to investigate the role of extracellular SODs in antimicrobial defence. Qualitative and quantitative proteome analyses resulted in the identification of two extracellular Cu/Zn SODs in the haemolymph and one in the defensive secretions of juvenile P. cochleariae. Furthermore, quantitative expression studies indicated fat body tissue and defensive glands as the main synthesis sites of these SODs. Silencing of the two SODs revealed one of them, PcSOD3.1, as the only relevant enzyme facilitating SOD activity in haemolymph and defensive secretions in vivo. Upon challenge with the entomopathogenic fungus, Metarhizium anisopliae, PcSOD3.1-deficient larvae exhibited a significantly higher mortality compared to other SOD-silenced groups. Hence, our results serve as a basis for further research on SOD regulated host-pathogen interactions. In defensive secretions PcSOD3.1-silencing affected neither deterrent production nor activity against fungal growth. Instead, we propose another antifungal mechanism based on MRJP/yellow proteins in the defensive exudates.
Collapse
|
45
|
Abstract
Neutrophils are essential for killing bacteria and other microorganisms, and they also have a significant role in regulating the inflammatory response. Stimulated neutrophils activate their NADPH oxidase (NOX2) to generate large amounts of superoxide, which acts as a precursor of hydrogen peroxide and other reactive oxygen species that are generated by their heme enzyme myeloperoxidase. When neutrophils engulf bacteria they enclose them in small vesicles (phagosomes) into which superoxide is released by activated NOX2 on the internalized neutrophil membrane. The superoxide dismutates to hydrogen peroxide, which is used by myeloperoxidase to generate other oxidants, including the highly microbicidal species hypochlorous acid. NOX activation occurs at other sites in the cell, where it is considered to have a regulatory function. Neutrophils also release oxidants, which can modify extracellular targets and affect the function of neighboring cells. We discuss the identity and chemical properties of the specific oxidants produced by neutrophils in different situations, and what is known about oxidative mechanisms of microbial killing, inflammatory tissue damage, and signaling.
Collapse
Affiliation(s)
- Christine C Winterbourn
- Centre for Free Radical Research, Department of Pathology, University of Otago, Christchurch 8011, New Zealand; , ,
| | - Anthony J Kettle
- Centre for Free Radical Research, Department of Pathology, University of Otago, Christchurch 8011, New Zealand; , ,
| | - Mark B Hampton
- Centre for Free Radical Research, Department of Pathology, University of Otago, Christchurch 8011, New Zealand; , ,
| |
Collapse
|
46
|
Henríquez C, Riquelme TT, Vera D, Julio-Kalajzić F, Ehrenfeld P, Melvin JE, Figueroa CD, Sarmiento J, Flores CA. The calcium-activated potassium channel KCa3.1 plays a central role in the chemotactic response of mammalian neutrophils. Acta Physiol (Oxf) 2016; 216:132-45. [PMID: 26138196 DOI: 10.1111/apha.12548] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/13/2015] [Accepted: 06/24/2015] [Indexed: 12/27/2022]
Abstract
AIM Neutrophils are the first cells to arrive at sites of injury. Nevertheless, many inflammatory diseases are characterized by an uncontrolled infiltration and action of these cells. Cell migration depends on volume changes that are governed by ion channel activity, but potassium channels in neutrophil have not been clearly identified. We aim to test whether KCa3.1 participates in neutrophil migration and other relevant functions of the cell. METHODS Cytometer and confocal measurements to determine changes in cell volume were used. Cells isolated from human, mouse and horse were tested for KCa3.1-dependent chemotaxis. Chemokinetics, calcium handling and release of reactive oxygen species were measured to determine the role of KCa3.1 in those processes. A mouse model was used to test for neutrophil recruitment after acute lung injury in vivo. RESULTS We show for the first time that KCa3.1 is expressed in mammalian neutrophils. When the channel is inhibited by a pharmacological blocker or by genetic silencing, it profoundly affects cell volume regulation, and chemotactic and chemokinetic properties of the cells. We also demonstrated that pharmacological inhibition of KCa3.1 did not affect calcium entry or reactive oxygen species production in neutrophils. Using a mouse model of acute lung injury, we observed that Kca3.1(-/-) mice are significantly less effective at recruiting neutrophils into the site of inflammation. CONCLUSIONS These results demonstrate that KCa3.1 channels are key actors in the migration capacity of neutrophils, and its inhibition did not affect other relevant cellular functions.
Collapse
Affiliation(s)
- C. Henríquez
- Instituto de Farmacología; Facultad de Medicina Veterinaria; Universidad Austral de Chile; Valdivia Chile
| | | | - D. Vera
- Centro de Estudios Científicos (CECs); Valdivia Chile
| | - F. Julio-Kalajzić
- Centro de Estudios Científicos (CECs); Valdivia Chile
- Pontificia Universidad Católica de Valparaíso; Valparaíso Chile
| | - P. Ehrenfeld
- Institutos de Anatomía; Histología y Patología; Universidad Austral de Chile; Valdivia Chile
| | - J. E. Melvin
- Secretory Mechanisms and Dysfunction Section; National Institute of Dental and Craniofacial Research; National Institutes of Health; Bethesda MD USA
| | - C. D. Figueroa
- Institutos de Anatomía; Histología y Patología; Universidad Austral de Chile; Valdivia Chile
| | - J. Sarmiento
- Instituto de Fisiología; Facultad de Medicina; Universidad Austral de Chile; Valdivia Chile
| | - C. A. Flores
- Centro de Estudios Científicos (CECs); Valdivia Chile
| |
Collapse
|
47
|
Seredenina T, Demaurex N, Krause KH. Voltage-Gated Proton Channels as Novel Drug Targets: From NADPH Oxidase Regulation to Sperm Biology. Antioxid Redox Signal 2015; 23:490-513. [PMID: 24483328 PMCID: PMC4543398 DOI: 10.1089/ars.2013.5806] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
SIGNIFICANCE Voltage-gated proton channels are increasingly implicated in cellular proton homeostasis. Proton currents were originally identified in snail neurons less than 40 years ago, and subsequently shown to play an important auxiliary role in the functioning of reactive oxygen species (ROS)-generating nicotinamide adenine dinucleotide phosphate (NADPH) oxidases. Molecular identification of voltage-gated proton channels was achieved less than 10 years ago. Interestingly, so far, only one gene coding for voltage-gated proton channels has been identified, namely hydrogen voltage-gated channel 1 (HVCN1), which codes for the HV1 proton channel protein. Over the last years, the first picture of putative physiological functions of HV1 has been emerging. RECENT ADVANCES The best-studied role remains charge and pH compensation during the respiratory burst of the phagocyte NADPH oxidase (NOX). Strong evidence for a role of HV1 is also emerging in sperm biology, but the relationship with the sperm NOX5 remains unclear. Probably in many instances, HV1 functions independently of NOX: for example in snail neurons, basophils, osteoclasts, and cancer cells. CRITICAL ISSUES Generally, ion channels are good drug targets; however, this feature has so far not been exploited for HV1, and hitherto no inhibitors compatible with clinical use exist. However, there are emerging indications for HV1 inhibitors, ranging from diseases with a strong activation of the phagocyte NOX (e.g., stroke) to infertility, osteoporosis, and cancer. FUTURE DIRECTIONS Clinically useful HV1-active drugs should be developed and might become interesting drugs of the future.
Collapse
Affiliation(s)
- Tamara Seredenina
- 1 Department of Pathology and Immunology, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland
| | - Nicolas Demaurex
- 2 Department of Cellular Physiology and Metabolism, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland
| | - Karl-Heinz Krause
- 1 Department of Pathology and Immunology, Geneva University Medical Faculty , Centre Médical Universitaire, Geneva, Switzerland .,3 Department of Genetic and Laboratory Medicine, Geneva University Hospitals , Centre Médical Universitaire, Geneva, Switzerland
| |
Collapse
|
48
|
Abstract
The main properties of the voltage-gated proton channel (HV1) are described in this review, along with what is known about how the channel protein structure accomplishes its functions. Just as protons are unique among ions, proton channels are unique among ion channels. Their four transmembrane helices sense voltage and the pH gradient and conduct protons exclusively. Selectivity is achieved by the unique ability of H3O(+) to protonate an Asp-Arg salt bridge. Pathognomonic sensitivity of gating to the pH gradient ensures HV1 channel opening only when acid extrusion will result, which is crucial to most of its biological functions. An exception occurs in dinoflagellates in which influx of H(+) through HV1 triggers the bioluminescent flash. Pharmacological interventions that promise to ameliorate cancer, asthma, brain damage in ischemic stroke, Alzheimer's disease, autoimmune diseases, and numerous other conditions await future progress.
Collapse
Affiliation(s)
- Thomas E. DeCoursey
- Department of Molecular Biophysics and Physiology, Rush University, 1750 West Harrison, Chicago IL, 60612 USA
| |
Collapse
|
49
|
Tay HL, Kaiko GE, Plank M, Li J, Maltby S, Essilfie AT, Jarnicki A, Yang M, Mattes J, Hansbro PM, Foster PS. Antagonism of miR-328 increases the antimicrobial function of macrophages and neutrophils and rapid clearance of non-typeable Haemophilus influenzae (NTHi) from infected lung. PLoS Pathog 2015; 11:e1004549. [PMID: 25894560 PMCID: PMC4404141 DOI: 10.1371/journal.ppat.1004549] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 11/01/2014] [Indexed: 11/23/2022] Open
Abstract
Pathogenic bacterial infections of the lung are life threatening and underpin chronic lung diseases. Current treatments are often ineffective potentially due to increasing antibiotic resistance and impairment of innate immunity by disease processes and steroid therapy. Manipulation miRNA directly regulating anti-microbial machinery of the innate immune system may boost host defence responses. Here we demonstrate that miR-328 is a key element of the host response to pulmonary infection with non-typeable haemophilus influenzae and pharmacological inhibition in mouse and human macrophages augments phagocytosis, the production of reactive oxygen species, and microbicidal activity. Moreover, inhibition of miR-328 in respiratory models of infection, steroid-induced immunosuppression, and smoke-induced emphysema enhances bacterial clearance. Thus, miRNA pathways can be targeted in the lung to enhance host defence against a clinically relevant microbial infection and offer a potential new anti-microbial approach for the treatment of respiratory diseases. MicroRNAs regulate pathogen recognition pathways by modulating translation. In the immune system, miRNAs have been identified as important regulators of gene expression programs, which regulate differentiation, growth and function of innate and adaptive immune cells. Using miRNA microarray, we demonstrated that lung miRNAs were differentially expressed following non-typeable Haemophilus Influenzae (NTHi) infection in mice. To study the role of a specific miRNA in macrophages, we used antagomir (chemically modified single-stranded RNA analogues, complementary to the target miRNA) to block miRNA function. Interestingly, inhibition of microRNA-328 in mouse and human macrophages increases microbicidal activity by amplifying phagocytosis and production of reactive oxygen species. Inhibition of mR-328 in the lung enhanced bacterial clearance in mouse models of immunosuppression and emphysema. Our study provides proof of principle that miRNA pathways can be targeted in the lung and offer a potential new anti-microbial approach for the treatment of respiratory infection.
Collapse
Affiliation(s)
- Hock L. Tay
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Gerard E. Kaiko
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Maximilian Plank
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - JingJing Li
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Steven Maltby
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Ama-Tawiah Essilfie
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Andrew Jarnicki
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | | | - Joerg Mattes
- Priority Research Centre for Asthma and Respiratory Disease, Discipline of Paediatrics and Child Health, School of Medicine and Public Health, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Paul S. Foster
- Priority Research Centre for Asthma and Respiratory Disease, Department of Microbiology and Immunology, School of Pharmacy and Biomedical Sciences, Faculty of Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
- * E-mail:
| |
Collapse
|
50
|
Levine AP, Duchen MR, de Villiers S, Rich PR, Segal AW. Alkalinity of neutrophil phagocytic vacuoles is modulated by HVCN1 and has consequences for myeloperoxidase activity. PLoS One 2015; 10:e0125906. [PMID: 25885273 PMCID: PMC4401748 DOI: 10.1371/journal.pone.0125906] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2014] [Accepted: 03/21/2015] [Indexed: 12/03/2022] Open
Abstract
The NADPH oxidase of neutrophils, essential for innate immunity, passes electrons across the phagocytic membrane to form superoxide in the phagocytic vacuole. Activity of the oxidase requires that charge movements across the vacuolar membrane are balanced. Using the pH indicator SNARF, we measured changes in pH in the phagocytic vacuole and cytosol of neutrophils. In human cells, the vacuolar pH rose to ~9, and the cytosol acidified slightly. By contrast, in Hvcn1 knock out mouse neutrophils, the vacuolar pH rose above 11, vacuoles swelled, and the cytosol acidified excessively, demonstrating that ordinarily this channel plays an important role in charge compensation. Proton extrusion was not diminished in Hvcn1-/- mouse neutrophils arguing against its role in maintaining pH homeostasis across the plasma membrane. Conditions in the vacuole are optimal for bacterial killing by the neutral proteases, cathepsin G and elastase, and not by myeloperoxidase, activity of which was unphysiologically low at alkaline pH.
Collapse
Affiliation(s)
- Adam P. Levine
- Division of Medicine, University College London, London, United Kingdom
| | - Michael R. Duchen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Simon de Villiers
- Glynn Laboratory of Bioenergetics, Department of Biology, University College London, London, United Kingdom
| | - Peter R. Rich
- Glynn Laboratory of Bioenergetics, Department of Biology, University College London, London, United Kingdom
| | - Anthony W. Segal
- Division of Medicine, University College London, London, United Kingdom
- * E-mail:
| |
Collapse
|