51
|
Domon H, Terao Y. The Role of Neutrophils and Neutrophil Elastase in Pneumococcal Pneumonia. Front Cell Infect Microbiol 2021; 11:615959. [PMID: 33796475 PMCID: PMC8008068 DOI: 10.3389/fcimb.2021.615959] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 03/01/2021] [Indexed: 12/11/2022] Open
Abstract
Streptococcus pneumoniae, also known as pneumococcus, is a Gram-positive diplococcus and a major human pathogen. This bacterium is a leading cause of bacterial pneumonia, otitis media, meningitis, and septicemia, and is a major cause of morbidity and mortality worldwide. To date, studies on S. pneumoniae have mainly focused on the role of its virulence factors including toxins, cell surface proteins, and capsules. However, accumulating evidence indicates that in addition to these studies, knowledge of host factors and host-pathogen interactions is essential for understanding the pathogenesis of pneumococcal diseases. Recent studies have demonstrated that neutrophil accumulation, which is generally considered to play a critical role in host defense during bacterial infections, can significantly contribute to lung injury and immune subversion, leading to pneumococcal invasion of the bloodstream. Here, we review bacterial and host factors, focusing on the role of neutrophils and their elastase, which contribute to the progression of pneumococcal pneumonia.
Collapse
Affiliation(s)
- Hisanori Domon
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.,Research Center for Advanced Oral Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| |
Collapse
|
52
|
Ostermann L, Maus R, Stolper J, Schütte L, Katsarou K, Tumpara S, Pich A, Mueller C, Janciauskiene S, Welte T, Maus UA. Alpha-1 antitrypsin deficiency impairs lung antibacterial immunity in mice. JCI Insight 2021; 6:140816. [PMID: 33554955 PMCID: PMC7934856 DOI: 10.1172/jci.insight.140816] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/17/2020] [Indexed: 01/05/2023] Open
Abstract
Alpha-1 antitrypsin (AAT) is a major inhibitor of serine proteases in mammals. Therefore, its deficiency leads to protease–antiprotease imbalance and a risk for developing lung emphysema. Although therapy with human plasma-purified AAT attenuates AAT deficiency–related emphysema, its impact on lung antibacterial immunity is poorly defined. Here, we examined the effect of AAT therapy on lung protective immunity in AAT-deficient (KO) mice challenged with Streptococcus pneumoniae. AAT-KO mice were highly susceptible to S. pneumoniae, as determined by severe lobar pneumonia and early mortality. Mechanistically, we found that neutrophil-derived elastase (NE) degraded the opsonophagocytically important collectins, surfactant protein A (SP-A) and D (SP-D), which was accompanied by significantly impaired lung bacterial clearance in S. pneumoniae–infected AAT-KO mice. Treatment of S. pneumoniae–infected AAT-KO mice with human AAT protected SP-A and SP-D from NE-mediated degradation and corrected the pulmonary pathology observed in these mice. Likewise, treatment with Sivelestat, a specific inhibitor of NE, also protected collectins from degradation and significantly decreased bacterial loads in S. pneumoniae–infected AAT-KO mice. Our findings show that NE is responsible for the degradation of lung SP-A and SP-D in AAT-KO mice affecting lung protective immunity in AAT deficiency.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Andreas Pich
- Institute of Toxicology and Core Facility Proteomics, Hannover Medical School, Hannover, Germany
| | - Christian Mueller
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Sabina Janciauskiene
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Tobias Welte
- Clinic for Pneumology, and.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| | - Ulrich A Maus
- Division of Experimental Pneumology.,German Center for Lung Research, partner site BREATH, Hannover, Germany
| |
Collapse
|
53
|
Ulfig A, Leichert LI. The effects of neutrophil-generated hypochlorous acid and other hypohalous acids on host and pathogens. Cell Mol Life Sci 2021; 78:385-414. [PMID: 32661559 PMCID: PMC7873122 DOI: 10.1007/s00018-020-03591-y] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 06/21/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022]
Abstract
Neutrophils are predominant immune cells that protect the human body against infections by deploying sophisticated antimicrobial strategies including phagocytosis of bacteria and neutrophil extracellular trap (NET) formation. Here, we provide an overview of the mechanisms by which neutrophils kill exogenous pathogens before we focus on one particular weapon in their arsenal: the generation of the oxidizing hypohalous acids HOCl, HOBr and HOSCN during the so-called oxidative burst by the enzyme myeloperoxidase. We look at the effects of these hypohalous acids on biological systems in general and proteins in particular and turn our attention to bacterial strategies to survive HOCl stress. HOCl is a strong inducer of protein aggregation, which bacteria can counteract by chaperone-like holdases that bind unfolding proteins without the need for energy in the form of ATP. These chaperones are activated by HOCl through thiol oxidation (Hsp33) or N-chlorination of basic amino acid side-chains (RidA and CnoX) and contribute to bacterial survival during HOCl stress. However, neutrophil-generated hypohalous acids also affect the host system. Recent studies have shown that plasma proteins act not only as sinks for HOCl, but get actively transformed into modulators of the cellular immune response through N-chlorination. N-chlorinated serum albumin can prevent aggregation of proteins, stimulate immune cells, and act as a pro-survival factor for immune cells in the presence of cytotoxic antigens. Finally, we take a look at the emerging role of HOCl as a potential signaling molecule, particularly its role in neutrophil extracellular trap formation.
Collapse
Affiliation(s)
- Agnes Ulfig
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany
| | - Lars I Leichert
- Ruhr University Bochum, Institute for Biochemistry and Pathobiochemistry-Microbial Biochemistry, Universitätsstrasse 150, 44780, Bochum, Germany.
| |
Collapse
|
54
|
Gigon L, Yousefi S, Karaulov A, Simon HU. Mechanisms of toxicity mediated by neutrophil and eosinophil granule proteins. Allergol Int 2021; 70:30-38. [PMID: 33277190 DOI: 10.1016/j.alit.2020.11.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Neutrophils and eosinophils are granulocytes which are characterized by the presence of granules in the cytoplasm. Granules provide a safe storage site for granule proteins that play important roles in the immune function of granulocytes. Upon granulocytes activation, diverse proteins are released from the granules into the extracellular space and contribute to the fight against infections. In this article, we describe granule proteins of both neutrophils and eosinophils able to kill pathogens and review their anticipated mechanism of antimicrobial toxicity. It should be noted that an excess of granules protein release can lead to tissue damage of the host resulting in chronic inflammation and organ dysfunction.
Collapse
|
55
|
Bai X, Hippensteel J, Leavitt A, Maloney JP, Beckham D, Garcia C, Li Q, Freed BM, Ordway D, Sandhaus RA, Chan ED. Hypothesis: Alpha-1-antitrypsin is a promising treatment option for COVID-19. Med Hypotheses 2021; 146:110394. [PMID: 33239231 PMCID: PMC7659642 DOI: 10.1016/j.mehy.2020.110394] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/12/2020] [Accepted: 11/06/2020] [Indexed: 01/08/2023]
Abstract
No definitive treatment for COVID-19 exists although promising results have been reported with remdesivir and glucocorticoids. Short of a truly effective preventive or curative vaccine against SARS-CoV-2, it is becoming increasingly clear that multiple pathophysiologic processes seen with COVID-19 as well as SARS-CoV-2 itself should be targeted. Because alpha-1-antitrypsin (AAT) embraces a panoply of biologic activities that may antagonize several pathophysiologic mechanisms induced by SARS-CoV-2, we hypothesize that this naturally occurring molecule is a promising agent to ameliorate COVID-19. We posit at least seven different mechanisms by which AAT may alleviate COVID-19. First, AAT is a serine protease inhibitor (SERPIN) shown to inhibit TMPRSS-2, the host serine protease that cleaves the spike protein of SARS-CoV-2, a necessary preparatory step for the virus to bind its cell surface receptor ACE2 to gain intracellular entry. Second, AAT has anti-viral activity against other RNA viruses HIV and influenza as well as induces autophagy, a known host effector mechanism against MERS-CoV, a related coronavirus that causes the Middle East Respiratory Syndrome. Third, AAT has potent anti-inflammatory properties, in part through inhibiting both nuclear factor-kappa B (NFκB) activation and ADAM17 (also known as tumor necrosis factor-alpha converting enzyme), and thus may dampen the hyper-inflammatory response of COVID-19. Fourth, AAT inhibits neutrophil elastase, a serine protease that helps recruit potentially injurious neutrophils and implicated in acute lung injury. AAT inhibition of ADAM17 also prevents shedding of ACE2 and hence may preserve ACE2 inhibition of bradykinin, reducing the ability of bradykinin to cause a capillary leak in COVID-19. Fifth, AAT inhibits thrombin, and venous thromboembolism and in situ microthrombi and macrothrombi are increasingly implicated in COVID-19. Sixth, AAT inhibition of elastase can antagonize the formation of neutrophil extracellular traps (NETs), a complex extracellular structure comprised of neutrophil-derived DNA, histones, and proteases, and implicated in the immunothrombosis of COVID-19; indeed, AAT has been shown to change the shape and adherence of non-COVID-19-related NETs. Seventh, AAT inhibition of endothelial cell apoptosis may limit the endothelial injury linked to severe COVID-19-associated acute lung injury, multi-organ dysfunction, and pre-eclampsia-like syndrome seen in gravid women. Furthermore, because both NETs formation and the presence of anti-phospholipid antibodies are increased in both COVID-19 and non-COVID pre-eclampsia, it suggests a similar vascular pathogenesis in both disorders. As a final point, AAT has an excellent safety profile when administered to patients with AAT deficiency and is dosed intravenously once weekly but also comes in an inhaled preparation. Thus, AAT is an appealing drug candidate to treat COVID-19 and should be studied.
Collapse
Affiliation(s)
- Xiyuan Bai
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA; Departments of Academic Affairs and Medicine, National Jewish Health, Denver, CO, USA; Division of Pulmonary Sciences and Critical Care Medicine, USA
| | - Joseph Hippensteel
- Division of Pulmonary Sciences and Critical Care Medicine, USA; Denver Health, Denver, CO, USA
| | | | - James P Maloney
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA; Division of Pulmonary Sciences and Critical Care Medicine, USA
| | - David Beckham
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA; Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Cindy Garcia
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| | - Qing Li
- Departments of Academic Affairs and Medicine, National Jewish Health, Denver, CO, USA; School of Public Health, San Diego State University, San Diego, CA, USA
| | - Brian M Freed
- Department of Immunology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Diane Ordway
- Department of Microbiology, Immunlogy, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - Robert A Sandhaus
- Departments of Academic Affairs and Medicine, National Jewish Health, Denver, CO, USA
| | - Edward D Chan
- Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA; Departments of Academic Affairs and Medicine, National Jewish Health, Denver, CO, USA; Division of Pulmonary Sciences and Critical Care Medicine, USA.
| |
Collapse
|
56
|
El Amri C. Serine Protease Inhibitors to Treat Lung Inflammatory Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:215-226. [PMID: 34019272 DOI: 10.1007/978-3-030-68748-9_13] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Lung is a vital organ that ensures breathing function. It provides the essential interface of air filtering providing oxygen to the whole body and eliminating carbon dioxide in the blood; because of its exposure to the external environment, it is fall prey to many exogenous elements, such as pathogens, especially viral infections or environmental toxins and chemicals. These exogenous actors in addition to intrinsic disorders lead to important inflammatory responses that compromise lung tissue and normal functioning. Serine proteases regulating inflammation responses are versatile enzymes, usually involved in pro-inflammatory cytokines or other molecular mediator's production and activation of immune cells. In this chapter, an overview on major serine proteases in airway inflammation as therapeutic targets and their clinically relevant inhibitors is provided. Recent updates on serine protease inhibitors in the context of the COVID-19 pandemic are summarized.
Collapse
Affiliation(s)
- Chahrazade El Amri
- Sorbonne Université, Faculty of Sciences and Engineering, IBPS, UMR 8256 CNRS-UPMC, ERL INSERM U1164, Biological Adaptation and Ageing, Paris, France.
| |
Collapse
|
57
|
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: 6] [Impact Index Per Article: 1.2] [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
|
58
|
Crespo M, Gonzalez-Teran B, Nikolic I, Mora A, Folgueira C, Rodríguez E, Leiva-Vega L, Pintor-Chocano A, Fernández-Chacón M, Ruiz-Garrido I, Cicuéndez B, Tomás-Loba A, A-Gonzalez N, Caballero-Molano A, Beiroa D, Hernández-Cosido L, Torres JL, Kennedy NJ, Davis RJ, Benedito R, Marcos M, Nogueiras R, Hidalgo A, Matesanz N, Leiva M, Sabio G. Neutrophil infiltration regulates clock-gene expression to organize daily hepatic metabolism. eLife 2020; 9:59258. [PMID: 33287957 PMCID: PMC7723411 DOI: 10.7554/elife.59258] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 11/04/2020] [Indexed: 12/20/2022] Open
Abstract
Liver metabolism follows diurnal fluctuations through the modulation of molecular clock genes. Disruption of this molecular clock can result in metabolic disease but its potential regulation by immune cells remains unexplored. Here, we demonstrated that in steady state, neutrophils infiltrated the mouse liver following a circadian pattern and regulated hepatocyte clock-genes by neutrophil elastase (NE) secretion. NE signals through c-Jun NH2-terminal kinase (JNK) inhibiting fibroblast growth factor 21 (FGF21) and activating Bmal1 expression in the hepatocyte. Interestingly, mice with neutropenia, defective neutrophil infiltration or lacking elastase were protected against steatosis correlating with lower JNK activation, reduced Bmal1 and increased FGF21 expression, together with decreased lipogenesis in the liver. Lastly, using a cohort of human samples we found a direct correlation between JNK activation, NE levels and Bmal1 expression in the liver. This study demonstrates that neutrophils contribute to the maintenance of daily hepatic homeostasis through the regulation of the NE/JNK/Bmal1 axis. Every day, the body's biological processes work to an internal clock known as the circadian rhythm. This rhythm is controlled by ‘clock genes’ that are switched on or off by daily physical and environmental cues, such as changes in light levels. These daily rhythms are very finely tuned, and disturbances can lead to serious health problems, such as diabetes or high blood pressure. The ability of the body to cycle through the circadian rhythm each day is heavily influenced by the clock of one key organ: the liver. This organ plays a critical role in converting food and drink into energy. There is evidence that neutrophils – white blood cells that protect the body by being the first response to inflammation – can influence how the liver performs its role in obese people, by for example, releasing a protein called elastase. Additionally, the levels of neutrophils circulating in the blood change following a daily pattern. Crespo, González-Terán et al. wondered whether neutrophils enter the liver at specific times of the day to control liver’s daily rhythm. Crespo, González-Terán et al. revealed that neutrophils visit the liver in a pattern that peaks when it gets light and dips when it gets dark by counting the number of neutrophils in the livers of mice at different times of the day. During these visits, neutrophils secreted elastase, which activated a protein called JNK in the cells of the mice’s liver. This subsequently blocked the activity of another protein, FGF21, which led to the activation of the genes that allow cells to make fat molecules for storage. JNK activation also switched on the clock gene, Bmal1, ultimately causing fat to build up in the mice’s liver. Crespo, González-Terán et al. also found that, in samples from human livers, the levels of elastase, the activity of JNK, and whether the Bmal1 gene was switched on were tightly linked. This suggests that neutrophils may be controlling the liver’s rhythm in humans the same way they do in mice. Overall, this research shows that neutrophils can control and reset the liver's daily rhythm using a precisely co-ordinated series of molecular changes. These insights into the liver's molecular clock suggest that elastase, JNK and BmaI1 may represent new therapeutic targets for drugs or smart medicines to treat metabolic diseases such as diabetes or high blood pressure.
Collapse
Affiliation(s)
- María Crespo
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | - Ivana Nikolic
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Cintia Folgueira
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Elena Rodríguez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Luis Leiva-Vega
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | | | - Irene Ruiz-Garrido
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Beatriz Cicuéndez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Antonia Tomás-Loba
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Noelia A-Gonzalez
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | | | - Daniel Beiroa
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.,CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Lourdes Hernández-Cosido
- Department of General Surgery, University Hospital of Salamanca-IBSAL, Department of Surgery, University of Salamanca, Salamanca, Spain
| | - Jorge L Torres
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Norman J Kennedy
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Roger J Davis
- Howard Hughes Medical Institute and Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, United States
| | - Rui Benedito
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Miguel Marcos
- Department of Internal Medicine, University Hospital of Salamanca-IBSAL, Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Ruben Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Santiago de Compostela, Spain.,CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
| | - Andrés Hidalgo
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Nuria Matesanz
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Magdalena Leiva
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares Carlos (CNIC), Madrid, Spain
| |
Collapse
|
59
|
Tran NT, Graf R, Wulf-Goldenberg A, Stecklum M, Strauß G, Kühn R, Kocks C, Rajewsky K, Chu VT. CRISPR-Cas9-Mediated ELANE Mutation Correction in Hematopoietic Stem and Progenitor Cells to Treat Severe Congenital Neutropenia. Mol Ther 2020; 28:2621-2634. [PMID: 32822592 PMCID: PMC7704744 DOI: 10.1016/j.ymthe.2020.08.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 07/09/2020] [Accepted: 08/05/2020] [Indexed: 01/08/2023] Open
Abstract
Severe congenital neutropenia (SCN) is a monogenic disorder. SCN patients are prone to recurrent life-threatening infections. The main causes of SCN are autosomal dominant mutations in the ELANE gene that lead to a block in neutrophil differentiation. In this study, we use CRISPR-Cas9 ribonucleoproteins and adeno-associated virus (AAV)6 as a donor template delivery system to repair the ELANEL172P mutation in SCN patient-derived hematopoietic stem and progenitor cells (HSPCs). We used a single guide RNA (sgRNA) specifically targeting the mutant allele, and an sgRNA targeting exon 4 of ELANE. Using the latter sgRNA, ∼34% of the known ELANE mutations can in principle be repaired. We achieved gene correction efficiencies of up to 40% (with sgELANE-ex4) and 56% (with sgELANE-L172P) in the SCN patient-derived HSPCs. Gene repair restored neutrophil differentiation in vitro and in vivo upon HSPC transplantation into humanized mice. Mature edited neutrophils expressed normal elastase levels and behaved normally in functional assays. Thus, we provide a proof of principle for using CRISPR-Cas9 to correct ELANE mutations in patient-derived HSPCs, which may translate into gene therapy for SCN.
Collapse
Affiliation(s)
- Ngoc Tung Tran
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Robin Graf
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | | | | | | | - Ralf Kühn
- iPS Cell Based Disease Modeling, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Christine Kocks
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; Transgenics, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany
| | - Klaus Rajewsky
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| | - Van Trung Chu
- Immune Regulation and Cancer, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany; iPS Cell Based Disease Modeling, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 13125 Berlin, Germany.
| |
Collapse
|
60
|
Alhede M, Alhede M, Qvortrup K, Kragh KN, Jensen PØ, Stewart PS, Bjarnsholt T. The origin of extracellular DNA in bacterial biofilm infections in vivo. Pathog Dis 2020; 78:5810662. [PMID: 32196074 PMCID: PMC7150582 DOI: 10.1093/femspd/ftaa018] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 03/19/2020] [Indexed: 02/03/2023] Open
Abstract
Extracellular DNA (eDNA) plays an important role in both the aggregation of bacteria and in the interaction of the resulting biofilms with polymorphonuclear leukocytes (PMNs) during an inflammatory response. Here, transmission electron and confocal scanning laser microscopy were used to examine the interaction between biofilms of Pseudomonas aeruginosa and PMNs in a murine implant model and in lung tissue from chronically infected cystic fibrosis patients. PNA FISH, DNA staining, labeling of PMN DNA with a thymidine analogue and immunohistochemistry were applied to localize bacteria, eDNA, PMN-derived eDNA, PMN-derived histone H3 (H3), neutrophil elastase (NE) and citrullinated H3 (citH3). Host-derived eDNA was observed surrounding bacterial biofilms but not within the biofilms. H3 localized to the lining of biofilms while NE was found throughout biofilms. CitH3, a marker for neutrophil extracellular traps (NETs) was detected only sporadically indicating that most host-derived eDNA in vivo was not a result of NETosis. Together these observations show that, in these in vivo biofilm infections with P. aeruginosa, the majority of eDNA is found external to the biofilm and derives from the host.
Collapse
Affiliation(s)
- Maria Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Morten Alhede
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Klaus Qvortrup
- CFIM/Department of Biomedical Sciences, University of Copenhagen, Blegdmasvej 3, DK-2200 Copenhagen N., Denmark
| | - Kasper Nørskov Kragh
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark
| | - Peter Østrup Jensen
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark.,Department of Clinical Microbiology, H:S Rigshospitalet, Juliane Maries Vej 22, DK-2100 Copenhagen Ø., Denmark
| | - Philip Shook Stewart
- Center for Biofilm Engineering, Montana State University, 366 Barnard Hall, P.O. Box 173980, Bozeman, MT 59717-3980, USA
| | - Thomas Bjarnsholt
- Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N., Denmark.,Department of Clinical Microbiology, H:S Rigshospitalet, Juliane Maries Vej 22, DK-2100 Copenhagen Ø., Denmark
| |
Collapse
|
61
|
Mahor D, Kumari V, Vashisht K, Galgalekar R, Samarth RM, Mishra PK, Banerjee N, Dixit R, Saluja R, De S, Pandey KC. Elevated serum matrix metalloprotease (MMP-2) as a candidate biomarker for stable COPD. BMC Pulm Med 2020; 20:302. [PMID: 33198714 PMCID: PMC7670729 DOI: 10.1186/s12890-020-01323-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 10/21/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The increasing trend of Chronic Obstructive Pulmonary Disease (COPD) in becoming the third leading cause of deaths by 2020 is of great concern, globally as well as in India. Dysregulation of protease/anti-protease balance in COPD has been reported to cause tissue destruction, inflammation and airway remodelling; which are peculiar characteristics of COPD. Therefore, it is imperative to explore various serum proteases involved in COPD pathogenesis, as candidate biomarkers. COPD and Asthma often have overlapping symptoms and therefore involvement of certain proteases in their pathogenesis would render accurate diagnosis of COPD to be difficult. METHODS Serum samples from controls, COPD and Asthma patients were collected after requisite institutional ethics committee approvals. The preliminary analysis qualitatively and quantitatively analyzed various serum proteases by ELISA and mass spectrometry techniques. In order to identify a distinct biomarker of COPD, serum neutrophil elastase (NE) and matrix metalloprotease-2 (MMP-2) from COPD and Asthma patients were compared; as these proteases tend to have overlapping activities in both the diseases. A quantitative analysis of the reactive oxygen species (ROS) in the serum of controls and COPD patients was also performed. Statistical analysis for estimation of p-values was performed using unpaired t-test with 95% confidence interval. RESULTS Amongst the significantly elevated proteases in COPD patients vs the controls- neutrophil elastase (NE) [P < 0.0241], caspase-7 [P < 0.0001] and matrix metalloprotease-2 (MMP-2) [P < 0.0001] were observed, along with increased levels of reactive oxygen species (ROS) [P < 0.0001]. The serum dipeptidyl peptidase-IV (DPP-IV) [P < 0.0010) concentration was found to be decreased in COPD patients as compared to controls. Interestingly, a distinct elevation of MMP-2 was observed only in COPD patients, but not in Asthma, as compared to controls. Mass spectrometry analysis further identified significant alterations (fold-change) in various proteases (carboxy peptidase, MMP-2 and human leukocyte elastase), anti-proteases (Preg. zone protein, α-2 macroglobulin, peptidase inhibitor) and signalling mediators (cytokine suppressor- SOCS-3). CONCLUSION The preliminary study of various serum proteases in stable COPD patients distinctly identified elevated MMP-2 as a candidate biomarker for COPD, subject to its validation in large cohort studies.
Collapse
Affiliation(s)
- Durga Mahor
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Vandana Kumari
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Kapil Vashisht
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Ruma Galgalekar
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | | | - Pradyumna K Mishra
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Nalok Banerjee
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Rajnikant Dixit
- ICMR-National Institute of Malaria Research, New Delhi, India
| | - Rohit Saluja
- All India Institute of Medical Sciences, Bibinagar, Hyderabad, India
- All India Institute of Medical Sciences, Bhopal, India
| | - Sajal De
- ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Kailash C Pandey
- ICMR-National Institute for Research in Environmental Health, Bhopal, India.
- ICMR-National Institute of Malaria Research, New Delhi, India.
| |
Collapse
|
62
|
Hoste E, Maueröder C, van Hove L, Catrysse L, Vikkula HK, Sze M, Maes B, Karjosukarso D, Martens L, Gonçalves A, Parthoens E, Roelandt R, Declercq W, Fuentes I, Palisson F, Gonzalez S, Salas-Alanis JC, Boon L, Huebener P, Mulder KW, Ravichandran K, Saeys Y, Schwabe RF, van Loo G. Epithelial HMGB1 Delays Skin Wound Healing and Drives Tumor Initiation by Priming Neutrophils for NET Formation. Cell Rep 2020; 29:2689-2701.e4. [PMID: 31775038 DOI: 10.1016/j.celrep.2019.10.104] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 09/06/2019] [Accepted: 10/25/2019] [Indexed: 01/03/2023] Open
Abstract
Regenerative responses predispose tissues to tumor formation by largely unknown mechanisms. High-mobility group box 1 (HMGB1) is a danger-associated molecular pattern contributing to inflammatory pathologies. We show that HMGB1 derived from keratinocytes, but not myeloid cells, delays cutaneous wound healing and drives tumor formation. In wounds of mice lacking HMGB1 selectively in keratinocytes, a marked reduction in neutrophil extracellular trap (NET) formation is observed. Pharmacological targeting of HMGB1 or NETs prevents skin tumorigenesis and accelerates wound regeneration. HMGB1-dependent NET formation and skin tumorigenesis is orchestrated by tumor necrosis factor (TNF) and requires RIPK1 kinase activity. NETs are present in the microenvironment of keratinocyte-derived tumors in mice and lesional and tumor skin of patients suffering from recessive dystrophic epidermolysis bullosa, a disease in which skin blistering predisposes to tumorigenesis. We conclude that tumorigenicity of the wound microenvironment depends on epithelial-derived HMGB1 regulating NET formation, thereby establishing a mechanism linking reparative inflammation to tumor initiation.
Collapse
Affiliation(s)
- Esther Hoste
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| | - Christian Maueröder
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Lisette van Hove
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Leen Catrysse
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Hanna-Kaisa Vikkula
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Mozes Sze
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Bastiaan Maes
- VIB Center for Inflammation Research, 9052 Ghent, Belgium
| | - Dyah Karjosukarso
- Department of Molecular Developmental Biology, Radboud University, 6525 XZ Nijmegen, the Netherlands
| | - Liesbet Martens
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Amanda Gonçalves
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; VIB Bio-Imaging Core, 9052 Ghent, Belgium
| | - Eef Parthoens
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium; VIB Bio-Imaging Core, 9052 Ghent, Belgium
| | - Ria Roelandt
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Wim Declercq
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Ignacia Fuentes
- Fundación DEBRA Chile, Santiago, Chile; Centro de Genetica y Genomica, Clinica Allemana, Universidad de Desarrollo, Santiago, Chile
| | - Francis Palisson
- Fundación DEBRA Chile, Santiago, Chile; Facultad de Medicina, Universidad de Desarrollo, Santiago, Chile
| | - Sergio Gonzalez
- Departemento de Patología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Louis Boon
- Bioceros, 3584 CM Utrecht, the Netherlands
| | - Peter Huebener
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Klaas Willem Mulder
- Department of Molecular Developmental Biology, Radboud University, 6525 XZ Nijmegen, the Netherlands
| | - Kodi Ravichandran
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium
| | - Yvan Saeys
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Applied Mathematics, Computer Sciences and Statistics, Ghent University, 9052 Ghent, Belgium
| | | | - Geert van Loo
- VIB Center for Inflammation Research, 9052 Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, 9052 Ghent, Belgium.
| |
Collapse
|
63
|
Ahmad J, Ikram S, Hafeez AB, Durdagi S. Physics-driven identification of clinically approved and investigation drugs against human neutrophil serine protease 4 (NSP4): A virtual drug repurposing study. J Mol Graph Model 2020; 101:107744. [PMID: 33032202 DOI: 10.1016/j.jmgm.2020.107744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/19/2023]
Abstract
Neutrophils synthesize four immune associated serine proteases: Cathepsin G (CTSG), Elastase (ELANE), Proteinase 3 (PRTN3) and Neutrophil Serine Protease 4 (NSP4). While previously considered to be immune modulators, overexpression of neutrophil serine proteases correlates with various disease conditions. Therefore, identifying novel small molecules that can potentially control or inhibit the proteolytic activity of these proteases is crucial to revert or temper the aggravated disease phenotype. To the best of our knowledge, although there is limited data for inhibitors of other neutrophil protease members, there is no previous clinical study of a synthetic small molecule inhibitor targeting NSP4. In this study, an integrated molecular modeling algorithm was performed within a virtual drug repurposing study to identify novel inhibitors for NSP4, using clinically approved and investigation drugs library (∼8000 compounds). Based on our rigorous filtration, we found that following molecules Becatecarin, Iogulamide, Delprostenate and Iralukast are predicted to block the activity of NSP4 by interacting with core catalytic residues. The selected ligands were energetically more favorable compared to the reference molecule. The result of this study identifies promising molecules as potential lead candidates.
Collapse
Affiliation(s)
- Jamshaid Ahmad
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan.
| | - Saima Ikram
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan; Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey
| | - Ahmer Bin Hafeez
- Center of Biotechnology & Microbiology, University of Peshawar, Pakistan
| | - Serdar Durdagi
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahcesehir University, Istanbul, Turkey.
| |
Collapse
|
64
|
Lee JH, Liu A, Park JH, Kato H, Hao Q, Zhang X, Zhou L, Lee JW. Therapeutic Effects of Hyaluronic Acid in Peritonitis-Induced Sepsis in Mice. Shock 2020; 54:488-497. [PMID: 31977961 PMCID: PMC7369239 DOI: 10.1097/shk.0000000000001512] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Intra-abdominal infection is the second most common cause of sepsis, and the mortality rate from abdominal sepsis remains high. High molecular weight (HMW) hyaluronic acid (HA) has been studied in sterile injury models as an anti-inflammatory and anti-permeability agent. This study evaluated the therapeutic effects of intraperitoneal HMW HA administration in mice with peritonitis-induced sepsis. Sepsis was induced in C57BL/6 mice by cecal ligation and puncture (CLP), followed 4 h later by an intraperitoneal injection of HMW HA (20 mg/kg) solution or phosphate buffered saline (PBS). Survival, physiological data, organ injury, bacterial burden, and inflammatory cytokine levels were assessed in the CLP mice. To assess the effect of HA on macrophage phagocytosis activity, RAW264.7 cells, primed with lipopolysaccharide, were exposed with either PBS or HMW HA (500 μg/mL) prior to exposure to 10 CFU of E coli bacteria. HMW HA instillation significantly improved blood oxygenation, lung histology, and survival in CLP mice. Inflammatory cytokine levels in the plasma and bacterial burdens in the lung and spleen were significantly decreased by HA administration at 24 h after CLP. At 6 h after CLP, HA significantly decreased bacterial burden in the peritoneal lavage fluid. HMW HA administration significantly increased E coli bacterial phagocytosis by RAW264.7 cells in part through increased phosphorylation of ezrin/radixin/moesin, a known downstream target of CD44 (a HA receptor); ezrin inhibition abolished the enhanced phagocytosis by RAW264.7 cells induced by HA. Intraperitoneal administration of HMW HA had therapeutic effects against CLP-induced sepsis in terms of suppressing inflammation and increasing antimicrobial activity.
Collapse
Affiliation(s)
- Jae Hoon Lee
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
- Department of Anesthesiology and Pain Medicine, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Airan Liu
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Jeong-Hyun Park
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Hideya Kato
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Qi Hao
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Xiwen Zhang
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Li Zhou
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| | - Jae-Woo Lee
- Department of Anesthesiology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
65
|
Cantin AM, Ouellet C, Cloutier A, McDonald PP. Airway Mucins Inhibit Oxidative and Non-Oxidative Bacterial Killing by Human Neutrophils. Front Pharmacol 2020; 11:554353. [PMID: 33101020 PMCID: PMC7554606 DOI: 10.3389/fphar.2020.554353] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/11/2020] [Indexed: 11/13/2022] Open
Abstract
Neutrophil killing of bacteria is mediated by oxidative and non-oxidative mechanisms. Oxidants are generated through the NADPH oxidase complex, whereas antimicrobial proteins and peptides rank amongst non-oxidative host defenses. Mucus hypersecretion, deficient hydration and poor clearance from the airways are prominent features of cystic fibrosis (CF) lung disease. CF airways are commonly infected by Pseudomonas aeruginosa and Burkholderia cepacia complex bacteria. Whereas the former bacterium is highly sensitive to non-oxidative killing, the latter is only killed if the oxidative burst is intact. Despite an abundance of neutrophils, both pathogens thrive in CF airway secretions. In this study, we report that secreted mucins protect these CF pathogens against host defenses. Mucins were purified from CF sputum and from the saliva of healthy volunteers. Whereas mucins did not alter the phagocytosis of Pseudomonas aeruginosa and Burkholderia cenocepacia by neutrophils, they completely suppressed bacterial killing. Accordingly, mucins markedly inhibited non-oxidative bacterial killing by neutrophil granule extracts, or by lysozyme and the cationic peptide, human β defensin-2 (HBD2). Mucins also suppressed the neutrophil oxidative burst through a charge-dependent mechanism that could be reversed by the cationic aminoglycoside, tobramycin. Our data indicate that airway mucins protect Gram-negative bacteria against neutrophil killing by suppressing the oxidative burst and inhibiting the bactericidal capacity of cationic proteins and peptides. Mucin hypersecretion, dehydration, stasis and anionic charge represent key therapeutic targets for improving host defenses and airway inflammation in CF and other muco-secretory airway diseases.
Collapse
Affiliation(s)
- André M. Cantin
- Pulmonary Division, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | | | | |
Collapse
|
66
|
Neutrophil elastase promotes macrophage cell adhesion and cytokine production through the integrin-Src kinases pathway. Sci Rep 2020; 10:15874. [PMID: 32981934 PMCID: PMC7522083 DOI: 10.1038/s41598-020-72667-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/26/2020] [Indexed: 01/08/2023] Open
Abstract
There are a number of respiratory diseases characterized by the presence of excess neutrophil elastase (NE) activity in tissues, including cystic fibrosis and chronic obstructive pulmonary disease (COPD). NE is considered a primary contributor to disease development, but the precise mechanism has yet to be fully determined. We hypothesized that NE alters the function of macrophages (Mɸ) which play a critical role in many physiological processes in healthy lungs. We demonstrate that monocyte-derived Mɸ exposed to NE releases active matrix metalloproteinases (MMPs), increase expression of pro-inflammatory cytokines TNFα, IL-1β, and IL-8, and reduce capacity to phagocytose bacteria. Changes in Mɸ function following NE treatment were accompanied by increased adhesion and cytoskeleton re-arrangement, indicating the possibility of integrin involvement. To support this observation, we demonstrate that NE induces phosphorylation of kinases from the Src kinase family, a hallmark of integrin signaling activation. Moreover, pretreatment of Mɸ with a specific Src kinase inhibitor, PP2 completely prevents NE-induced pro-inflammatory cytokine production. Taken together these findings indicate that NE participates in lung destruction not only through direct proteolytic degradation of matrix proteins, but also through activation of Mɸ inflammatory and proteolytic functions.
Collapse
|
67
|
Sebina I, Phipps S. The Contribution of Neutrophils to the Pathogenesis of RSV Bronchiolitis. Viruses 2020; 12:E808. [PMID: 32726921 PMCID: PMC7472258 DOI: 10.3390/v12080808] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/22/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Acute viral bronchiolitis causes significant mortality in the developing world, is the number one cause of infant hospitalisation in the developed world, and is associated with the later development of chronic lung diseases such as asthma. A vaccine against respiratory syncytial virus (RSV), the leading cause of viral bronchiolitis in infancy, remains elusive, and hence new therapeutic modalities are needed to limit disease severity. However, much remains unknown about the underlying pathogenic mechanisms. Neutrophilic inflammation is the predominant phenotype observed in infants with both mild and severe disease, however, a clear understanding of the beneficial and deleterious effects of neutrophils is lacking. In this review, we describe the multifaceted roles of neutrophils in host defence and antiviral immunity, consider their contribution to bronchiolitis pathogenesis, and discuss whether new approaches that target neutrophil effector functions will be suitable for treating severe RSV bronchiolitis.
Collapse
Affiliation(s)
- Ismail Sebina
- Respiratory Immunology Laboratory, QIMR Berghofer Medical Research Institute, Herston 4006, Australia;
| | | |
Collapse
|
68
|
Abstract
Neutrophils are critical to innate immunity, including host defense against bacterial and fungal infections. They achieve their host defense role by phagocytosing pathogens, secreting their granules full of cytotoxic enzymes, or expelling neutrophil extracellular traps (NETs) during the process of NETosis. NETs are weblike DNA structures decorated with histones and antimicrobial proteins released by activated neutrophils. Initially described as a means for neutrophils to neutralize pathogens, NET release also occurs in sterile inflammation, promotes thrombosis, and can mediate tissue damage. To effectively manipulate this double-edged sword to fight a particular disease, researchers must work toward understanding the mechanisms driving NETosis. Such understanding would allow the generation of new drugs to promote or prevent NETosis as needed. While knowledge regarding the (patho)physiological roles of NETosis is accumulating, little is known about the cellular and biophysical bases of this process. In this review, we describe and discuss our current knowledge of the molecular, cellular, and biophysical mechanisms mediating NET release as well as open questions in the field.
Collapse
Affiliation(s)
- Hawa Racine Thiam
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| | - Siu Ling Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232
| | - Denisa D Wagner
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts 02115, USA.,Department of Pediatrics, Harvard Medical School, Boston, Massachusetts 02115, USA.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, Massachusetts 02115, USA
| | - Clare M Waterman
- Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, Maryland 20892, USA; ,
| |
Collapse
|
69
|
Reihill J, Moffitt K, Douglas L, Stuart Elborn J, Jones A, Lorraine Martin S. Sputum trypsin-like protease activity relates to clinical outcome in cystic fibrosis. J Cyst Fibros 2020; 19:647-653. [DOI: 10.1016/j.jcf.2019.12.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/18/2019] [Accepted: 12/27/2019] [Indexed: 10/25/2022]
|
70
|
Burgener SS, Schroder K. Neutrophil Extracellular Traps in Host Defense. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a037028. [PMID: 31767647 DOI: 10.1101/cshperspect.a037028] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Neutrophils are produced in the bone marrow and then patrol blood vessels from which they can be rapidly recruited to a site of infection. Neutrophils bind, engulf, and efficiently kill invading microbes via a suite of defense mechanisms. Diverse extracellular and intracellular microbes induce neutrophils to extrude neutrophil extracellular traps (NETs) through the process of NETosis. Here, we review the signaling mechanisms and cell biology underpinning the key NETosis pathways during infection and the antimicrobial functions of NETs in host defense.
Collapse
Affiliation(s)
- Sabrina Sofia Burgener
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| | - Kate Schroder
- Institute for Molecular Bioscience (IMB), and IMB Centre for Inflammation and Disease Research, The University of Queensland, St Lucia 4072, Australia
| |
Collapse
|
71
|
Jia Z, Han HH, Sedgwick AC, Williams GT, Gwynne L, Brewster JT, Bull SD, Jenkins ATA, He XP, Schönherr H, Sessler JL, James TD. Protein Encapsulation: A Nanocarrier Approach to the Fluorescence Imaging of an Enzyme-Based Biomarker. Front Chem 2020; 8:389. [PMID: 32582623 PMCID: PMC7283737 DOI: 10.3389/fchem.2020.00389] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/14/2020] [Indexed: 01/01/2023] Open
Abstract
Here, we report a new pentafluoropropanamido rhodamine fluorescent probe (ACS-HNE) that allows for the selective detection of neutrophil elastase (NE). ACS-HNE displayed high sensitivity, with a low limit of detection (<5.3 nM), and excellent selectivity toward elastase over other relevant biological analytes and enzymes. The comparatively poor solubility and cell permeability of neat ACS-HNE was improved by creating an ACS-HNE-albumin complex; this approach allowed for improvements in the in situ visualization of elastase activity in RAW 264.7 cells relative to ACS-HNE alone. The present study thus serves to demonstrate a simple universal strategy that may be used to overcome cell impermeability and solubility limitations, and to prepare probes suitable for the cellular imaging of enzymatic activity in vitro.
Collapse
Affiliation(s)
- Zhiyuan Jia
- Department of Chemistry and Biology, Physical Chemistry & Research Center of Micro- and Nanochemistry and Engineering (Cμ), University of Siegen, Siegen, Germany
| | - Hai-Hao Han
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Adam C Sedgwick
- Department of Chemistry, The University of Texas at Austin, Austin, TX, United States
| | | | - Lauren Gwynne
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - James T Brewster
- Department of Chemistry, The University of Texas at Austin, Austin, TX, United States
| | - Steven D Bull
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - A Toby A Jenkins
- Department of Chemistry, University of Bath, Bath, United Kingdom
| | - Xiao-Peng He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular Engineering, Feringa Nobel Prize Scientist Joint Research Center, School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Holger Schönherr
- Department of Chemistry and Biology, Physical Chemistry & Research Center of Micro- and Nanochemistry and Engineering (Cμ), University of Siegen, Siegen, Germany
| | - Jonathan L Sessler
- Department of Chemistry, The University of Texas at Austin, Austin, TX, United States
| | - Tony D James
- Department of Chemistry, University of Bath, Bath, United Kingdom
| |
Collapse
|
72
|
Jackson-Jones LH, Smith P, Portman JR, Magalhaes MS, Mylonas KJ, Vermeren MM, Nixon M, Henderson BEP, Dobie R, Vermeren S, Denby L, Henderson NC, Mole DJ, Bénézech C. Stromal Cells Covering Omental Fat-Associated Lymphoid Clusters Trigger Formation of Neutrophil Aggregates to Capture Peritoneal Contaminants. Immunity 2020; 52:700-715.e6. [PMID: 32294409 PMCID: PMC7156918 DOI: 10.1016/j.immuni.2020.03.011] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 02/11/2020] [Accepted: 03/18/2020] [Indexed: 12/19/2022]
Abstract
The omentum is a visceral adipose tissue rich in fat-associated lymphoid clusters (FALCs) that collects peritoneal contaminants and provides a first layer of immunological defense within the abdomen. Here, we investigated the mechanisms that mediate the capture of peritoneal contaminants during peritonitis. Single-cell RNA sequencing and spatial analysis of omental stromal cells revealed that the surface of FALCs were covered by CXCL1+ mesothelial cells, which we termed FALC cover cells. Blockade of CXCL1 inhibited the recruitment and aggregation of neutrophils at FALCs during zymosan-induced peritonitis. Inhibition of protein arginine deiminase 4, an enzyme important for the release of neutrophil extracellular traps, abolished neutrophil aggregation and the capture of peritoneal contaminants by omental FALCs. Analysis of omental samples from patients with acute appendicitis confirmed neutrophil recruitment and bacterial capture at FALCs. Thus, specialized omental mesothelial cells coordinate the recruitment and aggregation of neutrophils to capture peritoneal contaminants.
Collapse
Affiliation(s)
- Lucy Helen Jackson-Jones
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK; Division of Biomedical and Life Sciences, Lancaster University, Lancaster LA1 4YQ, UK.
| | - Peter Smith
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | | | - Katie Jude Mylonas
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK; Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Mark Nixon
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | | | - Ross Dobie
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Sonja Vermeren
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Laura Denby
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Neil Cowan Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK; MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Damian James Mole
- Centre for Inflammation Research, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Cécile Bénézech
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH16 4TJ, UK.
| |
Collapse
|
73
|
Transposon Mutagenesis Screen of Klebsiella pneumoniae Identifies Multiple Genes Important for Resisting Antimicrobial Activities of Neutrophils in Mice. Infect Immun 2020; 88:IAI.00034-20. [PMID: 31988174 DOI: 10.1128/iai.00034-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 12/25/2022] Open
Abstract
Klebsiella pneumoniae is a Gram-negative bacterial pathogen that causes a range of infections, including pneumonias, urinary tract infections, and septicemia, in otherwise healthy and immunocompromised patients. K. pneumoniae has become an increasing concern due to the rise and spread of antibiotic-resistant and hypervirulent strains. However, its virulence determinants remain understudied. To identify novel K. pneumoniae virulence factors needed to cause pneumonia, a high-throughput screen was performed with an arrayed library of over 13,000 K. pneumoniae transposon insertion mutants in the lungs of wild-type (WT) and neutropenic mice using transposon sequencing (Tn-seq). Insertions in 166 genes resulted in K. pneumoniae mutants that were significantly less fit in the lungs of WT mice than in those of neutropenic mice. Of these, mutants with insertions in 51 genes still had significant defects in neutropenic mice, while mutants with insertions in 52 genes recovered significantly. In vitro screens using a minilibrary of K. pneumoniae transposon mutants identified putative functions for a subset of these genes, including in capsule content and resistance to reactive oxygen and nitrogen species. Lung infections in mice confirmed roles in K. pneumoniae virulence for the ΔdedA, ΔdsbC, ΔgntR, Δwzm-wzt, ΔyaaA, and ΔycgE mutants, all of which were defective in either capsule content or growth in reactive oxygen or nitrogen species. The fitness of the ΔdedA, ΔdsbC, ΔgntR, ΔyaaA, and ΔycgE mutants was higher in neutropenic mouse lungs, indicating that these genes encode proteins that protect K. pneumoniae against neutrophil-related effector functions.
Collapse
|
74
|
Haynes WA, Haddon DJ, Diep VK, Khatri A, Bongen E, Yiu G, Balboni I, Bolen CR, Mao R, Utz PJ, Khatri P. Integrated, multicohort analysis reveals unified signature of systemic lupus erythematosus. JCI Insight 2020; 5:122312. [PMID: 31971918 DOI: 10.1172/jci.insight.122312] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 01/17/2020] [Indexed: 12/27/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that follows an unpredictable disease course and affects multiple organs and tissues. We performed an integrated, multicohort analysis of 7,471 transcriptomic profiles from 40 independent studies to identify robust gene expression changes associated with SLE. We identified a 93-gene signature (SLE MetaSignature) that is differentially expressed in the blood of patients with SLE compared with healthy volunteers; distinguishes SLE from other autoimmune, inflammatory, and infectious diseases; and persists across diverse tissues and cell types. The SLE MetaSignature correlated significantly with disease activity and other clinical measures of inflammation. We prospectively validated the SLE MetaSignature in an independent cohort of pediatric patients with SLE using a microfluidic quantitative PCR (qPCR) array. We found that 14 of the 93 genes in the SLE MetaSignature were independent of IFN-induced and neutrophil-related transcriptional profiles that have previously been associated with SLE. Pathway analysis revealed dysregulation associated with nucleic acid biosynthesis and immunometabolism in SLE. We further refined a neutropoiesis signature and identified underappreciated transcripts related to immune cells and oxidative stress. In our multicohort, transcriptomic analysis has uncovered underappreciated genes and pathways associated with SLE pathogenesis, with the potential to advance clinical diagnosis, biomarker development, and targeted therapeutics for SLE.
Collapse
Affiliation(s)
- Winston A Haynes
- Institute for Immunity, Transplantation and Infection.,Division of Biomedical Informatics Research
| | - D James Haddon
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Vivian K Diep
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Avani Khatri
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Erika Bongen
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Gloria Yiu
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Imelda Balboni
- Division of Allergy, Immunology and Rheumatology, Department of Pediatrics, Stanford University School of Medicine, Stanford, California, USA
| | | | - Rong Mao
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Paul J Utz
- Institute for Immunity, Transplantation and Infection.,Division of Immunology and Rheumatology, Department of Medicine, and
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection.,Division of Biomedical Informatics Research
| |
Collapse
|
75
|
Chellappan DK, Yee LW, Xuan KY, Kunalan K, Rou LC, Jean LS, Ying LY, Wie LX, Chellian J, Mehta M, Satija S, Singh SK, Gulati M, Dureja H, Da Silva MW, Tambuwala MM, Gupta G, Paudel KR, Wadhwa R, Hansbro PM, Dua K. Targeting neutrophils using novel drug delivery systems in chronic respiratory diseases. Drug Dev Res 2020; 81:419-436. [PMID: 32048757 DOI: 10.1002/ddr.21648] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/21/2020] [Accepted: 01/25/2020] [Indexed: 12/29/2022]
Abstract
Neutrophils are essential effector cells of immune system for clearing the extracellular pathogens during inflammation and immune reactions. Neutrophils play a major role in chronic respiratory diseases. In respiratory diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, lung cancer and others, there occurs extreme infiltration and activation of neutrophils followed by a cascade of events like oxidative stress and dysregulated cellular proteins that eventually result in apoptosis and tissue damage. Dysregulation of neutrophil effector functions including delayed neutropil apoptosis, increased neutrophil extracellular traps in the pathogenesis of asthma, and chronic obstructive pulmonary disease enable neutrophils as a potential therapeutic target. Accounting to their role in pathogenesis, neutrophils present as an excellent therapeutic target for the treatment of chronic respiratory diseases. This review highlights the current status and the emerging trends in novel drug delivery systems such as nanoparticles, liposomes, microspheres, and other newer nanosystems that can target neutrophils and their molecular pathways, in the airways against infections, inflammation, and cancer. These drug delivery systems are promising in providing sustained drug delivery, reduced therapeutic dose, improved patient compliance, and reduced drug toxicity. In addition, the review also discusses emerging strategies and the future perspectives in neutrophil-based therapy.
Collapse
Affiliation(s)
- Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Lim W Yee
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kong Y Xuan
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Kishen Kunalan
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Lim C Rou
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Leong S Jean
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Lee Y Ying
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Lee X Wie
- School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Meenu Mehta
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Saurabh Satija
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Sachin K Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Harish Dureja
- Department of Pharmaceutical Sciences, Maharishi Dayanand University, Rohtak, Haryana, India
| | - Mateus Webba Da Silva
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, County Londonderry, Northern Ireland, United Kingdom
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, County Londonderry, Northern Ireland, United Kingdom
| | - Gaurav Gupta
- School of Phamacy, Suresh Gyan Vihar University, Jaipur, India
| | - Keshav R Paudel
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia
| | - Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, New South Wales, Australia
| | - Philip M Hansbro
- School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia.,Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute, Sydney, New South Wales, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, New South Wales, Australia.,Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, New South Wales, Australia.,School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India
| |
Collapse
|
76
|
Alpha-1 Antitrypsin-A Target for MicroRNA-Based Therapeutic Development for Cystic Fibrosis. Int J Mol Sci 2020; 21:ijms21030836. [PMID: 32012925 PMCID: PMC7037267 DOI: 10.3390/ijms21030836] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023] Open
Abstract
Cystic fibrosis (CF) is an autosomal recessive genetic disorder arising from mutations to the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Disruption to normal ion homeostasis in the airway results in impaired mucociliary clearance, leaving the lung more vulnerable to recurrent and chronic bacterial infections. The CF lung endures an excess of neutrophilic inflammation, and whilst neutrophil serine proteases are a crucial part of the innate host defence to infection, a surplus of neutrophil elastase (NE) is understood to create a net destructive effect. Alpha-1 antitrypsin (A1AT) is a key antiprotease in the control of NE protease activity but is ineffective in the CF lung due to the huge imbalance of NE levels. Therapeutic strategies to boost levels of protective antiproteases such as A1AT in the lung remain an attractive research strategy to limit the damage from excess protease activity. microRNAs are small non-coding RNA molecules that bind specific cognate sequences to inhibit expression of target mRNAs. The inhibition of miRNAs which target the SERPINA1 (A1AT-encoding gene) mRNA represents a novel therapeutic approach for CF inflammation. This could involve the delivery of antagomirs that bind and sequester the target miRNA, or target site blockers that bind miRNA recognition elements within the target mRNA to prevent miRNA interaction. Therefore, miRNA targeted therapies offer an alternative strategy to drive endogenous A1AT production and thus supplement the antiprotease shield of the CF lung.
Collapse
|
77
|
Stackowicz J, Jönsson F, Reber LL. Mouse Models and Tools for the in vivo Study of Neutrophils. Front Immunol 2020; 10:3130. [PMID: 32038641 PMCID: PMC6985372 DOI: 10.3389/fimmu.2019.03130] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 12/23/2019] [Indexed: 12/21/2022] Open
Abstract
Neutrophils are the most abundant leukocytes in human blood and critical actors of the immune system. Many neutrophil functions and facets of their activity in vivo were revealed by studying genetically modified mice or by tracking fluorescent neutrophils in animals using imaging approaches. Assessing the roles of neutrophils can be challenging, especially when exact molecular pathways are questioned or disease states are interrogated that alter normal neutrophil homeostasis. This review discusses the main in vivo models for the study of neutrophils, their advantages and limitations. The side-by-side comparison underlines the necessity to carefully choose the right model(s) to answer a given scientific question, and exhibit caveats that need to be taken into account when designing experimental procedures. Collectively, this review suggests that at least two models should be employed to legitimately conclude on neutrophil functions.
Collapse
Affiliation(s)
- Julien Stackowicz
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Sorbonne Université, Collège Doctoral, Paris, France
| | - Friederike Jönsson
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France
| | - Laurent L Reber
- Institut Pasteur, Department of Immunology, Unit of Antibodies in Therapy and Pathology, UMR INSERM 1222, Paris, France.,Center for Pathophysiology Toulouse-Purpan (CPTP), UMR 1043, University of Toulouse, INSERM, CNRS, Toulouse, France
| |
Collapse
|
78
|
Bonaventura A, Vecchié A, Abbate A, Montecucco F. Neutrophil Extracellular Traps and Cardiovascular Diseases: An Update. Cells 2020; 9:231. [PMID: 31963447 PMCID: PMC7016588 DOI: 10.3390/cells9010231] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/15/2022] Open
Abstract
Neutrophil extracellular traps (NETs) are formed by decondensed chromatin, histones, and neutrophil granular proteins and have a role in entrapping microbial pathogens. NETs, however, have pro-thrombotic properties by stimulating fibrin deposition, and increased NET levels correlate with larger infarct size and predict major adverse cardiovascular (CV) events. NETs have been involved also in the pathogenesis of diabetes, as high glucose levels were found to induce NETosis. Accordingly, NETs have been described as drivers of diabetic complications, such as diabetic wound and diabetic retinopathy. Inflammasomes are macromolecular structures involved in the release of pro-inflammatory mediators, such as interleukin-1, which is a key mediator in CV diseases. A crosstalk between the inflammasome and NETs is known for some rheumatologic diseases, while this link is still under investigation and not completely understood in CV diseases. In this review, we summarized the most recent updates about the role of NETs in acute myocardial infarction and metabolic diseases and provided an overview on the relationship between NET and inflammasome activities in rheumatologic diseases, speculating a possible link between these two entities also in CV diseases.
Collapse
Affiliation(s)
- Aldo Bonaventura
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy
| | - Alessandra Vecchié
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy
| | - Antonio Abbate
- Pauley Heart Center, Division of Cardiology, Department of Internal Medicine, Virginia Commonwealth University, 1200 E Marshall St, Richmond, VA 23298, USA; (A.V.); (A.A.)
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, viale Benedetto XV 6, 16132 Genoa, Italy;
- IRCCS Ospedale Policlinico San Martino Genova—Italian Cardiovascular Network, Largo R. Benzi 10, 16132 Genoa, Italy
| |
Collapse
|
79
|
Ueno K, Yanagihara N, Otani Y, Shimizu K, Kinjo Y, Miyazaki Y. Neutrophil-mediated antifungal activity against highly virulent Cryptococcus gattii strain R265. Med Mycol 2020; 57:1046-1054. [PMID: 30668754 DOI: 10.1093/mmy/myy153] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/10/2018] [Accepted: 12/13/2018] [Indexed: 01/08/2023] Open
Abstract
Vaccine-induced immune responses, including neutrophil, macrophage, and T-cell responses, ameliorate cryptococcosis caused by Cryptococcus gattii. However, whether neutrophils can exert fungicidal activity against C. gattii remains to be elucidated. Therefore, in this study, we investigated the neutrophil-mediated fungicidal effect against C. gattii R265 in vitro and compared it to the related fungal pathogen, Cryptococcus neoformans standard strain H99. We found that neutrophils recognized, phagocytosed, and killed C. gattii R265 in the presence of fresh mouse serum. This antifungal effect required phagocytosis and serine protease activity but not nicotinamide adenine dinucleotide phosphate oxidase activity. We also demonstrated that C. gattii R265 was more resistant to oxidative and nitrosative stress than C. neoformans H99. Together, these findings indicate that neutrophils can exert fungicidal activity against highly virulent C. gattii, at least under in vitro conditions.
Collapse
Affiliation(s)
- Keigo Ueno
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| | - Nao Yanagihara
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.,Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Yoshiko Otani
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.,Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Kiminori Shimizu
- Department of Biological Science and Technology, Faculty of Industrial Science and Technology, Tokyo University of Science, 6-3-1 Niijuku, Katsushika-ku, Tokyo 125-8585, Japan
| | - Yuki Kinjo
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan.,Department of Bacteriology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan.,Jikei Center for Biofilm Science and Technology, The Jikei University School of Medicine, 3-25-8 Nishi-Shimbashi, Minato-ku, Tokyo 105-8461, Japan
| | - Yoshitsugu Miyazaki
- Department of Chemotherapy and Mycoses, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo 162-8640, Japan
| |
Collapse
|
80
|
Thibault D, Jensen PA, Wood S, Qabar C, Clark S, Shainheit MG, Isberg RR, van Opijnen T. Droplet Tn-Seq combines microfluidics with Tn-Seq for identifying complex single-cell phenotypes. Nat Commun 2019; 10:5729. [PMID: 31844066 PMCID: PMC6914776 DOI: 10.1038/s41467-019-13719-9] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
While Tn-Seq is a powerful tool to determine genome-wide bacterial fitness in high-throughput, culturing transposon-mutant libraries in pools can mask community or other complex single-cell phenotypes. Droplet Tn-Seq (dTn-Seq) solves this problem by microfluidics facilitated encapsulation of individual transposon mutants into growth medium-in-oil droplets, thereby enabling isolated growth, free from the influence of the population. Here we describe and validate microfluidic chip design, production, encapsulation, and dTn-Seq sample preparation. We determine that 1-3% of mutants in Streptococcus pneumoniae have a different fitness when grown in isolation and show how dTn-Seq can help identify leads for gene function, including those involved in hyper-competence, processing of alpha-1-acid glycoprotein, sensitivity against the human leukocyte elastase and microcolony formation. Additionally, we show dTn-Seq compatibility with microscopy, FACS and investigations of bacterial cell-to-cell and bacteria-host cell interactions. dTn-Seq reduces costs and retains the advantages of Tn-Seq, while expanding the method's original applicability.
Collapse
Affiliation(s)
- Derek Thibault
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Paul A Jensen
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
- Department of Bioengineering and Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Stephen Wood
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA
| | - Christine Qabar
- Department of Biological Sciences, Towson University, Towson, MD, 21252, USA
| | - Stacie Clark
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Mara G Shainheit
- Department of Biological Sciences, Towson University, Towson, MD, 21252, USA
| | - Ralph R Isberg
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Tim van Opijnen
- Biology Department, Boston College, Chestnut Hill, MA, 02467, USA.
| |
Collapse
|
81
|
Masucci MT, Minopoli M, Carriero MV. Tumor Associated Neutrophils. Their Role in Tumorigenesis, Metastasis, Prognosis and Therapy. Front Oncol 2019; 9:1146. [PMID: 31799175 PMCID: PMC6874146 DOI: 10.3389/fonc.2019.01146] [Citation(s) in RCA: 403] [Impact Index Per Article: 67.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/15/2019] [Indexed: 12/18/2022] Open
Abstract
Tumor Associated Neutrophils (TANs) are engaged into the tumor microenvironment by cytokines and chemokines, can be distinguished according to their activation and cytokine status and effects on tumor cell growing in N1 and N2 TANs. N1 TANs exert an antitumor activity, by direct or indirect cytotoxicity. N2 TANs stimulate immunosuppression, tumor growth, angiogenesis and metastasis by DNA instability, or by cytokines and chemokines release. In tumor patients, either a high number of TANs and Neutrophil-to-Lymphocyte Ratio (NLR) do correlate with poor prognosis, and, so far, TAN counts and NLR can be regarded as biomarkers. Owing to the pivotal role of TANs in stimulating tumor progression, therapeutic strategies to target TANs have been suggested, and two major approaches have been proposed: (a) targeting the CXCL-8/CXCR-1/CXCR-2 axis, thereby blocking TANs or (b) targeting substances produced by polymorpho-nuclear cells that promote tumor growth. Many studies have been accomplished either in vitro and in animal models, whereas clinical studies are restrained, presently, due to the risk of inducing immunosuppression. In this review, we deeply discuss the anti-tumorigenic or pro-tumorigenic activity of TANs. In particular, TANs relevance in tumor prognosis and in vitro therapeutic strategies are widely described. On-going clinical trials, aimed to inhibit neutrophil recruitment into the tumor are also accurately debated.
Collapse
Affiliation(s)
- Maria Teresa Masucci
- Tumor Progression Unit, Department of Experimental Oncology, Istituto Nazionale Tumori Fondazione "G. Pascale" IRCCS, Naples, Italy
| | - Michele Minopoli
- Tumor Progression Unit, Department of Experimental Oncology, Istituto Nazionale Tumori Fondazione "G. Pascale" IRCCS, Naples, Italy
| | - Maria Vincenza Carriero
- Tumor Progression Unit, Department of Experimental Oncology, Istituto Nazionale Tumori Fondazione "G. Pascale" IRCCS, Naples, Italy
| |
Collapse
|
82
|
Clancy DM, Sullivan GP, Moran HBT, Henry CM, Reeves EP, McElvaney NG, Lavelle EC, Martin SJ. Extracellular Neutrophil Proteases Are Efficient Regulators of IL-1, IL-33, and IL-36 Cytokine Activity but Poor Effectors of Microbial Killing. Cell Rep 2019. [PMID: 29539422 DOI: 10.1016/j.celrep.2018.02.062] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Neutrophil granule proteases are thought to function as anti-microbial effectors, cooperatively hydrolyzing microorganisms within phagosomes, or upon deployment into the extracellular space. However, evidence also suggests that neutrophil proteases play an important role in the coordination and escalation of inflammatory reactions, but how this is achieved has been obscure. IL-1 family cytokines are important initiators of inflammation and are typically released via necrosis but require proteolytic processing for activation. Here, we show that proteases liberated from activated neutrophils can positively or negatively regulate the activity of six IL-1 family cytokines (IL-1α, IL-1β, IL-33, IL-36α, IL-36β, and IL-36γ) with exquisite sensitivity. In contrast, extracellular neutrophil proteases displayed very poor bactericidal activity, exhibiting 100-fold greater potency toward cytokine processing than bacterial killing. Thus, in addition to their classical role as phagocytes, neutrophils play an important immunoregulatory role through deployment of their granule proteases into the extracellular space to process multiple IL-1 family cytokines.
Collapse
Affiliation(s)
- Danielle M Clancy
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Graeme P Sullivan
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Hannah B T Moran
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Conor M Henry
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, Department of Medicine, Royal College of Surgeons in Ireland, Beaumont Hospital, Dublin 9, Ireland
| | - Ed C Lavelle
- Adjuvant Research Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College, Dublin 2, Ireland
| | - Seamus J Martin
- Molecular Cell Biology Laboratory, Department of Genetics, The Smurfit Institute, Trinity College, Dublin 2, Ireland.
| |
Collapse
|
83
|
SuFEx-enabled, agnostic discovery of covalent inhibitors of human neutrophil elastase. Proc Natl Acad Sci U S A 2019; 116:18808-18814. [PMID: 31484779 DOI: 10.1073/pnas.1909972116] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Sulfur fluoride exchange (SuFEx) has emerged as the new generation of click chemistry. We report here a SuFEx-enabled, agnostic approach for the discovery and optimization of covalent inhibitors of human neutrophil elastase (hNE). Evaluation of our ever-growing collection of SuFExable compounds toward various biological assays unexpectedly revealed a selective and covalent hNE inhibitor: benzene-1,2-disulfonyl fluoride. Synthetic derivatization of the initial hit led to a more potent agent, 2-(fluorosulfonyl)phenyl fluorosulfate with IC50 0.24 μM and greater than 833-fold selectivity over the homologous neutrophil serine protease, cathepsin G. The optimized, yet simple benzenoid probe only modified active hNE and not its denatured form.
Collapse
|
84
|
Dias BT, Dias-Teixeira KL, Godinho JP, Faria MS, Calegari-Silva T, Mukhtar MM, Lopes U, Mottram JC, Lima APCA. Neutrophil elastase promotes Leishmania donovani infection via interferon-β. FASEB J 2019; 33:10794-10807. [PMID: 31284755 PMCID: PMC6766642 DOI: 10.1096/fj.201900524r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Visceral leishmaniasis is a deadly illness caused by Leishmania donovani that provokes liver and spleen inflammation and tissue destruction. In cutaneous leishmaniasis, the protein of L. major, named inhibitor of serine peptidases (ISP) 2, inactivates neutrophil elastase (NE) present at the macrophage surface, resulting in blockade of TLR4 activation, prevention of TNF-α and IFN-β production, and parasite survival. We report poor intracellular growth of L. donovani in macrophages from knockout mice for NE (ela-/-), TLR4, or TLR2. NE and TLR4 colocalized with the parasite in the parasitophorous vacuole. Parasite load in the liver and spleen of ela-/- mice were reduced and accompanied by increased NO and decreased TGF-β production. Expression of ISP2 was not detected in L. donovani, and a transgenic line constitutively expressing ISP2, displayed poor intracellular growth in macrophages and decreased burden in mice. Infected ela-/- macrophages displayed significantly lower IFN-β mRNA than background mice macrophages, and the intracellular growth was fully restored by exogenous IFN-β. We propose that L. donovani utilizes the host NE-TLR machinery to induce IFN-β necessary for parasite survival and growth during early infection. Low or absent expression of parasite ISP2 in L. donovani is necessary to preserve the activation of the NE-TLR pathway.-Dias, B. T., Dias-Teixeira, K. L., Godinho, J. P., Faria, M. S., Calegari-Silva, T., Mukhtar, M. M., Lopes, U. G., Mottram, J. C., Lima, A. P. C. A. Neutrophil elastase promotes Leishmania donovani infection via interferon-β.
Collapse
Affiliation(s)
- Bruna T Dias
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karina Luisa Dias-Teixeira
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Joseane P Godinho
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marilia S Faria
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Teresa Calegari-Silva
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maowia M Mukhtar
- Bioscience Research Institute, Ibn Sina University, Khartoum, Sudan
| | - Ulisses Lopes
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jeremy C Mottram
- Department of Biology, York Biomedical Research Institute, University of York, York, United Kingdom
| | - Ana Paula C A Lima
- Instituto de Biofisica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
85
|
Bonaventura A, Montecucco F, Dallegri F, Carbone F, Lüscher TF, Camici GG, Liberale L. Novel findings in neutrophil biology and their impact on cardiovascular disease. Cardiovasc Res 2019; 115:1266-1285. [PMID: 30918936 DOI: 10.1093/cvr/cvz084] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 03/04/2019] [Accepted: 03/26/2019] [Indexed: 08/30/2023] Open
Abstract
Neutrophils are the most abundant circulating leucocytes in healthy humans. These cells are central players during acute inflammatory responses, although a growing body of evidence supports a crucial role in chronic inflammation and chemokines and cytokines related to it as well. Thus, both humoral and cellular components are involved in the development of plaque formation and atherosclerosis. Accordingly, CANTOS trial using an interleukin-1 beta antibody confirmed that inflammatory cytokines contribute to the occurrence of myocardial infarction and cardiac death independent of changes in lipids. Recent data revealed that neutrophils are a heterogeneous population with different subsets and functional characteristics (i.e. CD177+ cells, OLFM4+ neutrophils, proangiogenic neutrophils, neutrophils undergoing reverse migration, and aged neutrophils). Importantly, neutrophils are able to synthesize de novo proteins. Neutrophil extracellular trap generation and NETosis have been considered as very important weapons in sterile inflammation. Neutrophil-derived microvesicles represent another mechanism by which neutrophils amplify inflammatory processes, being found at high levels both at the site of injury and in the bloodstream. Finally, neutrophil aging can influence their functions also in relation with host age. These recent acquisitions in the field of neutrophil biology might pave the way for new therapeutic targets to prevent or even treat patients experiencing cardiovascular (CV) diseases. Here, we discuss novel findings in neutrophil biology, their impact on CV and cerebrovascular diseases, and the potential implementation of these notions into daily clinical practice.
Collapse
Affiliation(s)
- Aldo Bonaventura
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Division of Cardiology, Department of Internal Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, USA
| | - Fabrizio Montecucco
- First Clinic of Internal Medicine, Department of Internal Medicine and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Franco Dallegri
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 10 Largo Benzi, Genoa, Italy
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals and Imperial College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
- University Heart Center, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
- Department of Research and Education, University Hospital Zürich, Rämistrasse 100, Zürich, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 6 viale Benedetto XV, Genoa, Italy
- Center for Molecular Cardiology, University of Zürich, Wagistrasse 12, Schlieren, Switzerland
| |
Collapse
|
86
|
Crocetti L, Quinn MT, Schepetkin IA, Giovannoni MP. A patenting perspective on human neutrophil elastase (HNE) inhibitors (2014-2018) and their therapeutic applications. Expert Opin Ther Pat 2019; 29:555-578. [PMID: 31204543 PMCID: PMC9642779 DOI: 10.1080/13543776.2019.1630379] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 06/07/2019] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Human neutrophil elastase (HNE) is involved in a variety of serious chronic diseases, especially cardiopulmonary pathologies. For this reason, the regulation of HNE activity represents a promising therapeutic approach, which is evident by the development of a number of new and selective HNE inhibitors, both in the academic and pharmaceutical environments. AREAS COVERED The present review analyzes and summarizes the patent literature regarding human neutrophil elastase inhibitors for the treatment of cardiopulmonary diseases over 2014-2018. EXPERT OPINION HNE is an interesting and defined target to treat various inflammatory diseases, including a number of cardiopulmonary pathologies. The research in this field is quite active, and a number of HNE inhibitors are currently in various stages of clinical development. In addition, new opportunities for HNE inhibitor development stem from recent studies demonstrating the involvement of HNE in many other inflammatory pathologies, including rheumatoid arthritis, inflammatory bowel disease, skin diseases, and cancer. Furthermore, the development of dual HNE/proteinase 3 inhibitors is being pursued as an innovative approach for the treatment of neutrophilic inflammatory diseases. Thus, these new developments will likely stimulate new and increased interest in this important therapeutic target and for the development of novel and selective HNE inhibitors.
Collapse
Affiliation(s)
- L Crocetti
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - MT Quinn
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - IA Schepetkin
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT, USA
| | - MP Giovannoni
- Department of NEUROFARBA, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| |
Collapse
|
87
|
Nasri M, Ritter M, Mir P, Dannenmann B, Aghaallaei N, Amend D, Makaryan V, Xu Y, Fletcher B, Bernhard R, Steiert I, Hahnel K, Berger J, Koch I, Sailer B, Hipp K, Zeidler C, Klimiankou M, Bajoghli B, Dale DC, Welte K, Skokowa J. CRISPR/Cas9-mediated ELANE knockout enables neutrophilic maturation of primary hematopoietic stem and progenitor cells and induced pluripotent stem cells of severe congenital neutropenia patients. Haematologica 2019; 105:598-609. [PMID: 31248972 PMCID: PMC7049355 DOI: 10.3324/haematol.2019.221804] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 06/21/2019] [Indexed: 11/09/2022] Open
Abstract
A Autosomal-dominant ELANE mutations are the most common cause of severe congenital neutropenia. Although the majority of congenital neutropenia patients respond to daily granulocyte colony stimulating factor, approximately 15 % do not respond to this cytokine at doses up to 50 μg/kg/day and approximately 15 % of patients will develop myelodysplasia or acute myeloid leukemia. “Maturation arrest,” the failure of the marrow myeloid progenitors to form mature neutrophils, is a consistent feature of ELANE associated congenital neutropenia. As mutant neutrophil elastase is the cause of this abnormality, we hypothesized that ELANE associated neutropenia could be treated and “maturation arrest” corrected by a CRISPR/Cas9-sgRNA ribonucleoprotein mediated ELANE knockout. To examine this hypothesis, we used induced pluripotent stem cells from two congenital neutropenia patients and primary hematopoietic stem and progenitor cells from four congenital neutropenia patients harboring ELANE mutations as well as HL60 cells expressing mutant ELANE. We observed that granulocytic differentiation of ELANE knockout induced pluripotent stem cells and primary hematopoietic stem and progenitor cells were comparable to healthy individuals. Phagocytic functions, ROS production, and chemotaxis of the ELANE KO (knockout) neutrophils were also normal. Knockdown of ELANE in the mutant ELANE expressing HL60 cells also allowed full maturation and formation of abundant neutrophils. These observations suggest that ex vivo CRISPR/Cas9 RNP based ELANE knockout of patients’ primary hematopoietic stem and progenitor cells followed by autologous transplantation may be an alternative therapy for congenital neutropenia.
Collapse
Affiliation(s)
- Masoud Nasri
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Malte Ritter
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Perihan Mir
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Benjamin Dannenmann
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Narges Aghaallaei
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Diana Amend
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Vahagn Makaryan
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Yun Xu
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Breanna Fletcher
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Regine Bernhard
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Ingeborg Steiert
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Karin Hahnel
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Jürgen Berger
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Iris Koch
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Brigitte Sailer
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Katharina Hipp
- Max Planck Institute for Developmental Biology, Tübingen, Germany
| | - Cornelia Zeidler
- Department of Oncology, Hematology, Immunology and Bone Marrow Transplantation, Hannover Medical School, Hannover, Germany
| | - Maksim Klimiankou
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - Baubak Bajoghli
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| | - David C Dale
- Department of Medicine, University of Washington, Seattle, WA, USA
| | - Karl Welte
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany.,University Children's Hospital Tübingen, Tübingen, Germany
| | - Julia Skokowa
- Department of Oncology, Hematology, Immunology, Rheumatology and Clinical Immunology, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
88
|
Hiroyasu S, Turner CT, Richardson KC, Granville DJ. Proteases in Pemphigoid Diseases. Front Immunol 2019; 10:1454. [PMID: 31297118 PMCID: PMC6607946 DOI: 10.3389/fimmu.2019.01454] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 06/10/2019] [Indexed: 12/28/2022] Open
Abstract
Pemphigoid diseases are a subgroup of autoimmune skin diseases characterized by widespread tense blisters. Standard of care typically involves immunosuppressive treatments, which may be insufficient and are often associated with significant adverse events. As such, a deeper understanding of the pathomechanism(s) of pemphigoid diseases is necessary in order to identify improved therapeutic approaches. A major initiator of pemphigoid diseases is the accumulation of autoantibodies against proteins at the dermal-epidermal junction (DEJ), followed by protease activation at the lesion. The contribution of proteases to pemphigoid disease pathogenesis has been investigated using a combination of in vitro and in vivo models. These studies suggest proteolytic degradation of anchoring proteins proximal to the DEJ is crucial for dermal-epidermal separation and blister formation. In addition, proteases can also augment inflammation, expose autoantigenic cryptic epitopes, and/or provoke autoantigen spreading, which are all important in pemphigoid disease pathology. The present review summarizes and critically evaluates the current understanding with respect to the role of proteases in pemphigoid diseases.
Collapse
Affiliation(s)
- Sho Hiroyasu
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Christopher T. Turner
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - Katlyn C. Richardson
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| | - David J. Granville
- International Collaboration On Repair Discoveries (ICORD), Vancouver Coastal Health Research Institute (VCHRI), Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia (UBC), Vancouver, BC, Canada
- BC Professional Firefighters' Burn and Wound Healing Group, Vancouver Coastal Health Research Institute (VCHRI), University of British Columbia (UBC), Vancouver, BC, Canada
| |
Collapse
|
89
|
Warheit-Niemi HI, Hult EM, Moore BB. A pathologic two-way street: how innate immunity impacts lung fibrosis and fibrosis impacts lung immunity. Clin Transl Immunology 2019; 8:e1065. [PMID: 31293783 PMCID: PMC6593479 DOI: 10.1002/cti2.1065] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 05/29/2019] [Accepted: 06/06/2019] [Indexed: 12/11/2022] Open
Abstract
Lung fibrosis is characterised by the accumulation of extracellular matrix within the lung and is secondary to both known and unknown aetiologies. This accumulation of scar tissue limits gas exchange causing respiratory insufficiency. The pathogenesis of lung fibrosis is poorly understood, but immunologic‐based treatments have been largely ineffective. Despite this, accumulating evidence suggests that innate immune cells and receptors play important modulatory roles in the initiation and propagation of the disease. Paradoxically, while innate immune signalling may be important for the pathogenesis of fibrosis, there is also evidence to suggest that innate immune function against pathogens may be impaired, leading to dysregulated and/or impaired host defence. This review summarises the evidence for this pathologic two‐way street, highlights new concepts of pathogenesis and recommends future directions for research emphasis.
Collapse
Affiliation(s)
| | - Elissa M Hult
- Department of Molecular and Integrative Physiology University of Michigan Ann Arbor MI USA
| | - Bethany B Moore
- Department of Microbiology and Immunology University of Michigan Ann Arbor MI USA.,Department of Internal Medicine Division of Pulmonary and Critical Care Medicine University of Michigan Ann Arbor MI USA
| |
Collapse
|
90
|
Neutrophil Elastase Activity Imaging: Recent Approaches in the Design and Applications of Activity-Based Probes and Substrate-Based Probes. CONTRAST MEDIA & MOLECULAR IMAGING 2019; 2019:7417192. [PMID: 31281234 PMCID: PMC6594253 DOI: 10.1155/2019/7417192] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 05/19/2019] [Indexed: 02/07/2023]
Abstract
The last few decades of protease research has confirmed that a number of important biological processes are strictly dependent on proteolysis. Neutrophil elastase (NE) is a critical protease in immune response and host defense mechanisms in both physiological and disease-associated conditions. Particularly, NE has been identified as a promising biomarker for early diagnosis of lung inflammation. Recent studies have shown an increasing interest in developing methods for NE activity imaging both in vitro and in vivo. Unlike anatomical imaging modalities, functional molecular imaging, including enzymatic activities, enables disease detection at a very early stage and thus constitutes a much more accurate approach. When combined with advanced imaging technologies, opportunities arise for measuring imbalanced proteolytic activities with unprecedented details. Such technologies consist in building the highest resolved and sensitive instruments as well as the most specific probes based either on peptide substrates or on covalent inhibitors. This review outlines strengths and weaknesses of these technologies and discuss their applications to investigate NE activity as biomarker of pulmonary inflammatory diseases by imaging.
Collapse
|
91
|
Suzuki K, Okada H, Takemura G, Takada C, Kuroda A, Yano H, Zaikokuji R, Morishita K, Tomita H, Oda K, Matsuo S, Uchida A, Fukuta T, Sampei S, Miyazaki N, Kawaguchi T, Watanabe T, Yoshida T, Ushikoshi H, Yoshida S, Maekawa Y, Ogura S. Neutrophil Elastase Damages the Pulmonary Endothelial Glycocalyx in Lipopolysaccharide-Induced Experimental Endotoxemia. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1526-1535. [PMID: 31108101 DOI: 10.1016/j.ajpath.2019.05.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/28/2019] [Accepted: 05/01/2019] [Indexed: 11/25/2022]
Abstract
Neutrophil elastase (NE) is necessary for effective sterilization of phagocytosed bacterial and fungal pathogens; however, NE increases alveolocapillary permeability and induces proinflammatory cytokine production in sepsis-induced acute respiratory distress syndrome. Under septic conditions, the pulmonary endothelial glycocalyx covering on the healthy endothelium surface is injured, but the contribution of NE to this injury remains unknown. Our aim was to examine whether NE-induced pulmonary endothelial injury is associated with endotoxemia. Lipopolysaccharide (LPS; 20 mg/kg) was injected intraperitoneally into 9- to 12-week-old granulocyte colony-stimulating factor knockout (G-CSFKO) mice, which harbor few neutrophils, and littermate control mice; in a second assay, mice were injected with the NE-inhibitor sivelestat (0.2 mg/kg) at 3, 6, 9, and 12 hours after LPS administration. Subsequently, vascular endothelial injury was evaluated through ultrastructural analysis. At 48 hours after LPS injection, survival rate was more than threefold higher among G-CSFKO than control mice, and degradation of both thrombomodulin and syndecan-1 was markedly attenuated in G-CSFKO compared with control mice. Ultrastructural analysis revealed attenuated vascular endothelial injury and clear preservation of the endothelial glycocalyx in G-CSFKO mice. Moreover, after LPS exposure, survival rate was approximately ninefold higher among sivelestat-injected mice than control mice, and sivelestat treatment potently preserved vascular endothelial structures and the endothelial glycocalyx. In conclusion, NE is associated with pulmonary endothelial injury under LPS-induced endotoxemic conditions.
Collapse
Affiliation(s)
- Kodai Suzuki
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hideshi Okada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan.
| | - Genzou Takemura
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Chihiro Takada
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ayumi Kuroda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hirohisa Yano
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Ryogen Zaikokuji
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan; Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, Gifu, Japan
| | - Kentaro Morishita
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroyuki Tomita
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Kazumasa Oda
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Saori Matsuo
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Akihiro Uchida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Tetsuya Fukuta
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - So Sampei
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Nagisa Miyazaki
- Department of Internal Medicine, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| | - Tomonori Kawaguchi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Takatomo Watanabe
- Department of Clinical Laboratory, Gifu University Hospital, Gifu, Japan
| | - Takahiro Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hiroaki Ushikoshi
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Shozo Yoshida
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Yoichi Maekawa
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu, Japan; Domain of Integrated Life Systems, Center for Highly Advanced Integration of Nano and Life Sciences (G-CHAIN), Gifu University, Gifu, Japan
| | - Shinji Ogura
- Department of Emergency and Disaster Medicine, Gifu University Graduate School of Medicine, Gifu, Japan
| |
Collapse
|
92
|
Voisin MB, Leoni G, Woodfin A, Loumagne L, Patel NS, Di Paola R, Cuzzocrea S, Thiemermann C, Perretti M, Nourshargh S. Neutrophil elastase plays a non-redundant role in remodeling the venular basement membrane and neutrophil diapedesis post-ischemia/reperfusion injury. J Pathol 2019; 248:88-102. [PMID: 30632166 PMCID: PMC6850085 DOI: 10.1002/path.5234] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 11/09/2018] [Accepted: 12/23/2018] [Indexed: 12/20/2022]
Abstract
Ischemia/reperfusion (I/R) injury is a severe inflammatory insult associated with numerous pathologies, such as myocardial infarction, stroke and acute kidney injury. I/R injury is characterized by a rapid influx of activated neutrophils secreting toxic free radical species and degrading enzymes that can irreversibly damage the tissue, thus impairing organ functions. Significant efforts have been invested in identifying therapeutic targets to suppress neutrophil recruitment and activation post‐I/R injury. In this context, pharmacological targeting of neutrophil elastase (NE) has shown promising anti‐inflammatory efficacy in a number of experimental and clinical settings of I/R injury and is considered a plausible clinical strategy for organ care. However, the mechanisms of action of NE, and hence its inhibitors, in this process are not fully understood. Here we conducted a comprehensive analysis of the impact of NE genetic deletion on neutrophil infiltration in four murine models of I/R injury as induced in the heart, kidneys, intestine and cremaster muscle. In all models, neutrophil migration into ischemic regions was significantly suppressed in NE−/− mice as compared with wild‐type controls. Analysis of inflamed cremaster muscle and mesenteric microvessels by intravital and confocal microscopy revealed a selective entrapment of neutrophils within venular walls, most notably at the level of the venular basement membrane (BM) following NE deletion/pharmacological blockade. This effect was associated with the suppression of NE‐mediated remodeling of the low matrix protein expressing regions within the venular BM used by transmigrating neutrophils as exit portals. Furthermore, whilst NE deficiency led to reduced neutrophil activation and vascular leakage, levels of monocytes and prohealing M2 macrophages were reduced in tissues of NE−/− mice subjected to I/R. Collectively our results identify a vital and non‐redundant role for NE in supporting neutrophil breaching of the venular BM post‐I/R injury but also suggest a protective role for NE in promoting tissue repair. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Mathieu-Benoit Voisin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Giovanna Leoni
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK.,Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilian University (LMU), Munich, Germany
| | - Abigail Woodfin
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Laure Loumagne
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nimesh Sa Patel
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Rosanna Di Paola
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Christoph Thiemermann
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Sussan Nourshargh
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
93
|
Qian X, Zhao H, Chen X, Li J. Disruption of transient receptor potential melastatin 2 decreases elastase release and bacterial clearance in neutrophils. Innate Immun 2019; 24:122-130. [PMID: 29495939 PMCID: PMC6830898 DOI: 10.1177/1753425918759181] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Elastase released by neutrophils is critical for eliminating Gram-negative
bacteria. Ca2+ influx plays a key role in elastase release and
bacterial clearance in neutrophils. Transient receptor potential melastatin 2
(TRPM2) is a Ca2+-permeable cation channel highly expressed in
neutrophils. Here, we explore the role and possible mechanism of TRPM2 in
bacterial clearance in TRPM2 knockout (TRPM2-KO) mice neutrophils. After
exposure to Escherichia coli, TRPM2–KO bone marrow
neutrophils (BMNs) had increased bacterial burden and decreased elastase
release. The same was observed for septic TRPM2-KO mice which also had decreased
survival rate. After stimulation with chemotactic peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP), elastase release
was lower in TRPM2-KO BMNs than in wild type (WT) BMNs. Pre-treatment of WT BMNs
with p38 MAPK inhibitor reduced fMLP-induced elastase release. Compared with WT
BMNs, TRPM2-KO BMNs had decreased p38 MAPK phosphorylation after fMLP
stimulation. Removal of extracellular Ca2+ reduced fMLP-induced p38
MAPK phosphorylation and elastase release. The concentration of intracellular
Ca2+ decreased in TRPM2-KO BMNs compared with WT BMNs after fMLP
treatment. Hence, TRPM2 plays an important role in bacterial clearance in
neutrophils, possibly by regulating elastase release. TRPM2-mediated
Ca2+ influx regulates elastase release partially via p38 MAPK
phosphorylation in neutrophils.
Collapse
Affiliation(s)
- XiaoWei Qian
- 1 Department of Anesthesiology, Women's Hospital, School of Medicine, Zhejiang University, China.,2 Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children Hospital of Wenzhou Medical University, China
| | - Hang Zhao
- 3 Department of Anesthesiology, Yancheng Third People's Hospital, China
| | - XinZhong Chen
- 1 Department of Anesthesiology, Women's Hospital, School of Medicine, Zhejiang University, China
| | - Jun Li
- 2 Department of Anesthesiology, Critical Care and Pain Medicine, The Second Affiliated Hospital and Yuying Children Hospital of Wenzhou Medical University, China
| |
Collapse
|
94
|
Potey PM, Rossi AG, Lucas CD, Dorward DA. Neutrophils in the initiation and resolution of acute pulmonary inflammation: understanding biological function and therapeutic potential. J Pathol 2019; 247:672-685. [PMID: 30570146 PMCID: PMC6492013 DOI: 10.1002/path.5221] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/12/2018] [Accepted: 12/17/2018] [Indexed: 12/13/2022]
Abstract
Acute respiratory distress syndrome (ARDS) is the often fatal sequelae of a broad range of precipitating conditions. Despite decades of intensive research and clinical trials there remain no therapies in routine clinical practice that target the dysregulated and overwhelming inflammatory response that characterises ARDS. Neutrophils play a central role in the initiation, propagation and resolution of this complex inflammatory environment by migrating into the lung and executing a variety of pro-inflammatory functions. These include degranulation with liberation of bactericidal proteins, release of cytokines and reactive oxygen species as well as production of neutrophil extracellular traps. Although these functions are advantageous in clearing bacterial infection, the consequence of associated tissue damage, the contribution to worsening acute inflammation and prolonged neutrophil lifespan at sites of inflammation are deleterious. In this review, the importance of the neutrophil will be considered, together with discussion of recent advances in understanding neutrophil function and the factors that influence them throughout the phases of inflammation in ARDS. From a better understanding of neutrophils in this context, potential therapeutic targets are identified and discussed. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Philippe Md Potey
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Adriano G Rossi
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Christopher D Lucas
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - David A Dorward
- The University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
95
|
Liu S, Kang X, Catacchio CR, Liu M, Fang L, Schroeder SG, Li W, Rosen BD, Iamartino D, Iannuzzi L, Sonstegard TS, Van Tassell CP, Ventura M, Low WY, Williams JL, Bickhart DM, Liu GE. Computational detection and experimental validation of segmental duplications and associated copy number variations in water buffalo ( Bubalus bubalis ). Funct Integr Genomics 2019; 19:409-419. [PMID: 30734132 DOI: 10.1007/s10142-019-00657-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 12/13/2018] [Accepted: 01/09/2019] [Indexed: 01/25/2023]
Abstract
Duplicated sequences are an important source of gene evolution and structural variation within mammalian genomes. Using a read depth approach based on next-generation sequencing, we performed a genome-wide analysis of segmental duplications (SDs) and associated copy number variations (CNVs) in the water buffalo (Bubalus bubalis). By aligning short reads of Olimpia (the reference water buffalo) to the UMD3.1 cattle genome, we identified 1,038 segmental duplications comprising 44.6 Mb (equivalent to ~1.73% of the cattle genome) of the autosomal and X chromosomal sequence in the buffalo genome. We experimentally validated 70.3% (71/101) of these duplications using fluorescent in situ hybridization. We also detected a total of 1,344 CNV regions across 14 additional water buffaloes, amounting to 59.8 Mb of variable sequence or the equivalent of 2.2% of the cattle genome. The CNV regions overlap 1,245 genes that are significantly enriched for specific biological functions including immune response, oxygen transport, sensory system and signal transduction. Additionally, we performed array Comparative Genomic Hybridization (aCGH) experiments using the 14 water buffaloes as test samples and Olimpia as the reference. Using a linear regression model, a high Pearson correlation (r = 0.781) was observed between the log2 ratios between copy number estimates and the log2 ratios of aCGH probes. We further designed Quantitative PCR assays to confirm CNV regions within or near annotated genes and found 74.2% agreement with our CNV predictions. These results confirm sub-chromosome-scale structural rearrangements present in the cattle and water buffalo. The information on genome variation that will be of value for evolutionary and phenotypic studies, and may be useful for selective breeding of both species.
Collapse
Affiliation(s)
- Shuli Liu
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
- College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiaolong Kang
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
- College of Agriculture, Ningxia University, Yinchuan, 750021, China
| | | | - Mei Liu
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
- College of Animal Science and Technology, Shaanxi Key Laboratory of Agricultural Molecular Biology, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Lingzhao Fang
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
- Department of Animal and Avian Sciences, University of Maryland, College Park, Maryland, 20742, USA
| | - Steven G Schroeder
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
| | - Wenli Li
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA, ARS, Madison, WI 53706, USA
| | - Benjamin D Rosen
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
| | - Daniela Iamartino
- AIA-LGS, Associazione Italiana Allevatori - Laboratorio Genetica e Servizi, Via Bergamo 292, 26100 (CR), Cremona, Italy
- Parco Tecnologico Padano, Via Einstein, Polo Universitario, 26900, Lodi, Italy
| | - Leopoldo Iannuzzi
- Laboratory of Animal Cytogenetics and Gene Mapping, Nationa Research Council (CNR), ISPAAM, Via Argine 1085, 80147, Naples, Italy
| | | | - Curtis P Van Tassell
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA
| | - Mario Ventura
- Department of Biology, University of Bari, 70126, Bari, Italy
| | - Wai Yee Low
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, 5371, Australia
| | - John L Williams
- Davies Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy, SA, 5371, Australia
| | - Derek M Bickhart
- The Cell Wall Utilization and Biology Laboratory, US Dairy Forage Research Center, USDA, ARS, Madison, WI 53706, USA.
| | - George E Liu
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, Maryland, 20705, USA.
| |
Collapse
|
96
|
Yildiz K, Gokpinar S, Sursal N, Babur C, Ozen D, Azkur AK. Extracellular Trap Formation by Donkey Polymorphonuclear Neutrophils Against Toxoplasma gondii. J Equine Vet Sci 2019. [DOI: 10.1016/j.jevs.2018.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
97
|
PepN is a non-essential, cell wall-localized protein that contributes to neutrophil elastase-mediated killing of Streptococcus pneumoniae. PLoS One 2019; 14:e0211632. [PMID: 30707714 PMCID: PMC6358159 DOI: 10.1371/journal.pone.0211632] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/17/2019] [Indexed: 12/23/2022] Open
Abstract
Streptococcus pneumoniae (Spn) is an asymptomatic colonizer of the human nasopharynx but can also cause disease in the inner ear, meninges, lung and blood. Although various mechanisms contribute to the effective clearance of Spn, opsonophagocytosis by neutrophils is perhaps most critical. Upon phagocytosis, Spn is exposed to various degradative molecules, including a family of neutrophil serine proteases (NSPs) that are stored within intracellular granules. Despite the critical importance of NSPs in killing Spn, the bacterial proteins that are degraded by NSPs leading to Spn death are still unknown. In this report, we identify a 90kDa protein in a purified cell wall (CW) preparation, aminopeptidase N (PepN) that is degraded by the NSP neutrophil elastase (NE). Since PepN lacked a canonical signal sequence or LPxTG motif, we created a mutant expressing a FLAG tagged version of the protein and confirmed its localization to the CW compartment. We determined that not only is PepN a CW-localized protein, but also is a substrate of NE in the context of intact Spn cells. Furthermore, in comparison to wild-type TIGR4 Spn, a mutant strain lacking PepN demonstrated a significant hyper-resistance phenotype in vitro in the presence of purified NE as well as in opsonophagocytic assays with purified human neutrophils ex vivo. Taken together, this is the first study to demonstrate that PepN is a CW-localized protein and a substrate of NE that contributes to the effective killing of Spn by NSPs and human neutrophils.
Collapse
|
98
|
Frydrych LM, Bian G, Fattahi F, Morris SB, O'Rourke RW, Lumeng CN, Kunkel SL, Ward PA, Delano MJ. GM-CSF Administration Improves Defects in Innate Immunity and Sepsis Survival in Obese Diabetic Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2019; 202:931-942. [PMID: 30578307 DOI: 10.4049/jimmunol.1800713] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 11/29/2018] [Indexed: 12/19/2022]
Abstract
Sepsis is the leading cause of death in the intensive care unit with an overall mortality rate of 20%. Individuals who are obese and have type 2 diabetes have increased recurrent, chronic, nosocomial infections that worsen the long-term morbidity and mortality from sepsis. Additionally, animal models of sepsis have shown that obese, diabetic mice have lower survival rates compared with nondiabetic mice. Neutrophils are essential for eradication of bacteria, prevention of infectious complications, and sepsis survival. In diabetic states, there is a reduction in neutrophil chemotaxis, phagocytosis, and reactive oxygen species (ROS) generation; however, few studies have investigated the extent to which these deficits compromise infection eradication and mortality. Using a cecal ligation and puncture model of sepsis in lean and in diet-induced obese mice, we demonstrate that obese diabetic mice have decreased "emergency hematopoiesis" after an acute infection. Additionally, both neutrophils and monocytes in obese, diabetic mice have functional defects, with decreased phagocytic ability and a decreased capacity to generate ROS. Neutrophils isolated from obese diabetic mice have decreased transcripts of Axl and Mertk, which partially explains the phagocytic dysfunction. Furthermore, we found that exogenous GM-CSF administration improves sepsis survival through enhanced neutrophil and monocytes phagocytosis and ROS generation abilities in obese, diabetic mice with sepsis.
Collapse
Affiliation(s)
- Lynn M Frydrych
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Guowu Bian
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109
| | - Fatemeh Fattahi
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Robert W O'Rourke
- Department of Surgery, University of Michigan Medical School, Michigan Medicine, and Ann Arbor Veterans Administration Hospital, Ann Arbor, MI 48105; and
| | - Carey N Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI 48109
| | - Steven L Kunkel
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Peter A Ward
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109
| | - Matthew J Delano
- Division of Acute Care Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI 48109;
| |
Collapse
|
99
|
Agraz-Cibrian JM, Giraldo DM, Urcuqui-Inchima S. 1,25-Dihydroxyvitamin D 3 induces formation of neutrophil extracellular trap-like structures and modulates the transcription of genes whose products are neutrophil extracellular trap-associated proteins: A pilot study. Steroids 2019; 141:14-22. [PMID: 30414422 DOI: 10.1016/j.steroids.2018.11.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/09/2018] [Accepted: 11/05/2018] [Indexed: 01/05/2023]
Abstract
Neutrophils are components of the innate immune system that participate in controlling infectious diseases through microbicidal mechanisms such as phagocytosis, degranulation and the release of neutrophil extracellular traps (NETs). NETs are DNA structures that are released through the decondensation and spreading of chromatin and the adherence of various proteins, including neutrophil elastase (NE), myeloperoxidase (MPO) and peptidyl arginine deiminase 4 (PDA4). Since NETs recovered after treatment of activated polymorphonuclear neutrophils can enhance IL-1β and IFN-α production by LPS-activated macrophages, they are thought to be keys to the host's defenses and inflammation. 1,25(OH)2D3 has been shown to play an important role in modulating neutrophils activation and in preventing infections. Therefore, the aim of this study was to assess the effect of 1,25(OH)2D3 in modulating induction of the release of NETs and in regulating the transcription of genes whose products in human neutrophils are NETs-associated proteins, TLRs and interferon. We observed that 1,25(OH)2D3 induced production of NETs-like structures while also upregulating NE/PAD4/COX-3/GAPDH mRNA levels. Additionally, we found an increase in TLR7 and type I interferon (IFN) mRNA levels as a result of neutrophil activation by 1,25(OH)2D3. Since the transcription of genes whose products constitute NETs-associated proteins are differentially-regulated by 1,25(OH)2D3, we proposed that this might restrict the spread of pathogens, such as virus, by inducing NETs, the expression of TLR7 and secretion of IFN-α. Our results suggest the potential importance of this hormone in preventing infections by inducing NETs formation.
Collapse
Affiliation(s)
- Juan Manuel Agraz-Cibrian
- Unidad Académica de Ciencias Químico Biológicas y Farmacéuticas, Universidad Autónoma de Nayarit, Tepic, Nayarit, Mexico
| | - Diana M Giraldo
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia
| | - Silvio Urcuqui-Inchima
- Grupo Inmunovirología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| |
Collapse
|
100
|
Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: A Cellular Perspective. Physiol Rev 2019; 99:665-706. [PMID: 30475656 PMCID: PMC6442927 DOI: 10.1152/physrev.00067.2017] [Citation(s) in RCA: 1531] [Impact Index Per Article: 255.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 06/25/2018] [Accepted: 06/28/2018] [Indexed: 02/08/2023] Open
Abstract
Wound healing is one of the most complex processes in the human body. It involves the spatial and temporal synchronization of a variety of cell types with distinct roles in the phases of hemostasis, inflammation, growth, re-epithelialization, and remodeling. With the evolution of single cell technologies, it has been possible to uncover phenotypic and functional heterogeneity within several of these cell types. There have also been discoveries of rare, stem cell subsets within the skin, which are unipotent in the uninjured state, but become multipotent following skin injury. Unraveling the roles of each of these cell types and their interactions with each other is important in understanding the mechanisms of normal wound closure. Changes in the microenvironment including alterations in mechanical forces, oxygen levels, chemokines, extracellular matrix and growth factor synthesis directly impact cellular recruitment and activation, leading to impaired states of wound healing. Single cell technologies can be used to decipher these cellular alterations in diseased states such as in chronic wounds and hypertrophic scarring so that effective therapeutic solutions for healing wounds can be developed.
Collapse
Affiliation(s)
- Melanie Rodrigues
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Nina Kosaric
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Clark A Bonham
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| | - Geoffrey C Gurtner
- Department of Surgery, Stanford University School of Medicine , Stanford, California
| |
Collapse
|