WO2021209465A1 - Peptide for the treatment of net-associated diseases - Google Patents

Peptide for the treatment of net-associated diseases Download PDF

Info

Publication number
WO2021209465A1
WO2021209465A1 PCT/EP2021/059588 EP2021059588W WO2021209465A1 WO 2021209465 A1 WO2021209465 A1 WO 2021209465A1 EP 2021059588 W EP2021059588 W EP 2021059588W WO 2021209465 A1 WO2021209465 A1 WO 2021209465A1
Authority
WO
WIPO (PCT)
Prior art keywords
nets
peptide
histone
adhesion
monocytes
Prior art date
Application number
PCT/EP2021/059588
Other languages
French (fr)
Inventor
Gerardus Anna Franciscus Nicolaes
Kanin WICHAPONG
Oliver SÖHNLEIN
Original Assignee
Universiteit Maastricht
Academisch Ziekenhuis Maastricht
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Universiteit Maastricht, Academisch Ziekenhuis Maastricht filed Critical Universiteit Maastricht
Publication of WO2021209465A1 publication Critical patent/WO2021209465A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N3/00Spore forming or isolating processes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention is in the field of medical treatments. It provides means and methods for he treatments of diseases related to or caused by neutrophil extracellular traps (NETs).
  • NETs neutrophil extracellular traps
  • LPS Lipopolysaccharide
  • Atherosclerosis is a lipid-driven chronic inflammation of the arterial vessel wall. In its late stages, atherosclerosis is the underlying pathophysiology of myocardial infarction and stroke and hence the leading cause of mortality worldwide. Monocytes and their descendants hold crucial roles throughout all stages of atherosclerosis as they contribute to lipid modification and respond with a pronounced inflammatory response upon uptake of modified lipids [1] With only limited numbers of macrophages residing in large arteries, the vast majority of monocytic cells needs to be de novo recruited in a process known as the leukocyte recruitment cascade, which is orchestrated by fine-tuned interactions of selectins, chemokines, and integrins and their respective receptors [2] In recent years, studies have provided evidence for the contribution of neutrophils to the arterial monocyte recruitment. Herein, neutrophils deposit preformed chemoattractants on endothelial cells or activate endothelial cells directly to stimulate monocyte adhesion [3-6]
  • NETs neutrophil extracellular traps
  • the invention relates to a peptide comprising an amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or equivalents thereof and their use in medical therapy.
  • LPS is known to trigger the release of NETs [20] and hence we investigated whether in our experimental setting NETs may contribute to heightened neutrophil and monocyte recruitment.
  • Intravital microscopy permitted to visualize DAPI+ structures with a NET-like shape in the lumen of mice receiving LPS ( Figure 4).
  • cell-free DNA as well as DNA-MPO complexes, a plasma marker of NETs are strikingly increased in the plasma of mice with endotoxinemia ( Figures 5 - 8).
  • plasma DNA-MPO complexes correlated with plasma endotoxin levels indicating that LPS directly induces NET release ( Figures 5 - 8).
  • mice treated mice with BB Cl-amidine, a potent inhibitor of protein arginine deiminases (PADs) with favorable pharmacokinetics in terms of plasma half-life and EC50.
  • PADs protein arginine deiminases
  • Intravital imaging confirmed that BB Cl-amidine treatment abrogated luminal NET release ( Figure 4).
  • BB Cl-amidine treatment led to a vast reduction of arterial myeloid cell adhesion ( Figure 9, 10) suggesting that luminal NETs promote arterial myeloid cell adhesion during endotoxinemia.
  • NET-resident histone H2a attracts monocytes electrostatically.
  • biophysical interactions such as electrostatic charges could be important in this process.
  • monocytes present an overall negative surface charge (z potential -10.51 ⁇ 0.69 mV), while NETs are decorated with highly cationic proteins.
  • monocytes were incubated with either cholesterol sulfate or oleylamine. Cholesterol sulfate is a negatively-charged steroid lipid, whereas oleylamine is a positively-charged unsaturated fatty acid.
  • a force-distance curve reports on the contact force (I), the force until maximum adhesion (II), the forces needed to fully detach the monocyte from the NET (III) and the distance of this detachment (IV) ( Figure 21).
  • Monocytes with different surface charges were immobilized at the tip of the AFM cantilever and in this position probed on NETs.
  • Manipulation of the monocyte surface charges impacted on the maximum adhesion force, the detachment force and the detachment distance to separate monocytes from NETs, the energy required to separate the monocyte from the NET, and the adhesion frequency.
  • NET-adhesion strength and the ability to adhere to NETs was heightened when monocytes were rendered more negatively charged and opposite effects were found in less negatively charged monocytes ( Figure 22, 23), thus confirming the concept of charge-driven monocyte adhesion.
  • Neutrophil extracellular traps are decorated with a large array of granule- derived, cytoplasmatic, and nuclear proteins.
  • Proteomics of NETs allowed the identification of 73 enriched polypeptides of which two clusters exhibited cationic surface charge as deferred from the peptides isoelectric point. One cluster was centered on nuclear histones of which histone H2a is the most abundant one.
  • a second cluster revealed antimicrobial polypeptides with myeloperoxidase (MPO) and cathepsin G being the two candidates characterized by both cationic charge and high abundance (Figure 24).
  • MPO myeloperoxidase
  • cathepsin G being the two candidates characterized by both cationic charge and high abundance
  • peptide inhibitors to neutralize histone H2a.
  • Peptides contain several advantages such as ease of synthesis and optimization to improve pharmacokinetic and binding properties, large-scale and cost-effective production and applicability to conjugate with specific probes for drug delivery or for using as imaging tools [26]
  • monocyte adhesion can be interrupted by peptide inhibitors [5, 17] and the highly positively charged N-terminus of histone H4 can be neutralized by a peptide [18]
  • Cyclical Histone 2A Interference Peptide (CHIP), with amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC- OH, (SEQ ID NO: 1) bound to the N-terminal domain of histone 2A, and the electrostatic interactions between positively charged residues (Arg and Lys) of histone H2a with negatively charged residues (Asp and Glu) of CHIP and H-bond interactions promoted a stable complex formation ( Figure 29).
  • CHIP Cyclical Histone 2A Interference Peptide
  • NETs-associated diseases include for example autoimmune disorders [40], stroke [41], (transfusion-related) acute lung injury [36,37,38,42], ischaemia/reperfusion injury (IRI) [38,39,43], kidney injury [39,44], liver injury [45,46], trauma [47] and sepsis [48,49]
  • Histones are not only released as a consequence of cellular injury, they also actively contribute towards a self-sustaining cascade of ongoing cell death [50]
  • the invention may also be applied in the treatment of Acute respiratory distress syndrome such as ARDS caused by infection with coronavirus [51 - 53]
  • the peptide according to SEQ ID NO: 1 can therefore also be used to treat inflammation.
  • the invention also relates to a peptide comprising an amino acid sequence H- CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or an equivalent thereof that differs from SEQ ID NO: 1 by not more than 3 amino acids that is circular.
  • Atherosclerosis is the leading cause of mortality in Western society. Excess mortality from cardiovascular disease during influenza epidemics was first recognized early in the 20th century, but the specific association of influenza and other infections with myocardial infarction was not characterized until decades later. In fact, over the last 20 years epidemiological studies have consistently demonstrated the association between acute infection with multiple bacterial and viral pathogens and the short term increase in cardiovascular complications [7] As an example, a self-controlled case series involving U.S.
  • Neutralization of histone H2a by antibodies or by in silico designed peptides permits inhibition of endotoxin-accelerated monocyte adhesion and lesional myeloid cell accumulation, thus providing a potential strategy for improved care of patients at risk of cardiovascular events and experiencing an acute infection.
  • Neutrophils have previously been reported to pave the way for inflammatory monocytes into developing atherosclerotic lesions. Rendering mice neutropenic during initial stages of atherosclerosis diminishes lesion sizes as well as lesion composition, with lowered macrophage accumulation [30] Neutrophils stimulate various mechanisms that promote monocyte recruitment. Among these, secretion of chemotactic proteins stands out as an important mechanism for arterial monocyte recruitment.
  • Cathepsin G, cathelicidin, complexes formed of neutrophil-derived a- defensin and platelet-borne CCL5, and CCL2 released from neutrophils in a circadian fashion were shown to induce firm monocyte adhesion in mouse models of vascular inflammation when immobilized on arterial endothelial cells [3-5, 31]
  • all these ligands bind to specific receptors and increase adhesion through increased integrin affinity and avidity.
  • electrostatic interactions between cationic histone H2a and negatively charged monocyte surfaces as a novel mechanism driving infection-associated monocyte recruitment into atherosclerotic lesions.
  • Histone H2a combines abundance within NETs and surface charge and may hence stand out as an important epitope for monocyte adhesion.
  • histone isotypes may engage similar activities as they are comparable in abundance and charge within NETs.
  • activated smooth muscle cells residing in the fibrous cap of advanced atherosclerotic lesions stimulate neutrophils to release NETs.
  • cytotoxic histone H4 that can puncture human and murine SMCs leading to their death and, when re-occurring, thinning of the fibrous cap [18]
  • therapeutic neutralization of histone epitopes may exert a dual beneficial effect allowing to limit macrophage burden and fibrous cap thinning.
  • the invention therefore relates to a peptide comprising an amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1). More in particular it relates to a peptide as described herein consisting of the amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1). Such a peptide may advantageously be used in the treatment of a disease, more in particular in the treatment of a NETs-related disease.
  • the treatment is related to the treatment of a NETs-related disease selected from the group consisting of atherosclerosis, autoimmune disorders, stroke, transfusion -related acute lung injury, acute lung injury, lung injury, ischaemia/reperfusion injury (IRI), kidney injury, liver injury, trauma, sepsis and inflammation.
  • a NETs-related disease selected from the group consisting of atherosclerosis, autoimmune disorders, stroke, transfusion -related acute lung injury, acute lung injury, lung injury, ischaemia/reperfusion injury (IRI), kidney injury, liver injury, trauma, sepsis and inflammation.
  • Figures 1 - 4 Endotoxinemia accelerates atherosclerosis. Experimental setup. Apoe or Apoe ⁇ Cxacrl ⁇ mice were fed a HFD for 4 weeks and treated with either PBS (Ctrl) or with LPS (1 mg/kg BW). Another LPS-treated group received a BB Cl-amidine (BB Cl-A, 1mg/kg BW) 12 h before and along with LPS injection.
  • Figure 1 Aortic root lesion size analyzed in HE-stained sections. Representative images, scale bar 300 pm.
  • Figure 2 Lesional neutrophils (Ly6G + cells) quantified in root sections.
  • Figure 3 Macrophages (Mac2 + cells) quantified in root sections.
  • Figure 4 Count of luminal NET-like structures in left common carotid artery. Data are analyzed by one- way ANOVA with Dunnett's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001 , ****p£0.0001. All data are presented as meaniSEM.
  • FIGs 5 - 8 Endotoxinemia-triggered NET release stimulates arterial myeloid cell recruitment.
  • Apoe mice were fed a HFD for 4 weeks and treated with either PBS (Ctrl) or with LPS (1 mg/kg BW).
  • Another LPS-treated group received a BB Cl-amidine (BB Cl-A, 1 mg/kg BW) 12 h before and along with LPS injection.
  • Quantification of cell free DNA in plasma Figure 5
  • plasma endotoxin units Figure 6
  • circulating MPO-DNA complexes Figure 7 as measure of NET release. Pearson correlation of NETs (MPO-DNA complexes) with plasma endotoxin units ( Figure 8).
  • FIGs 11 - 17 Monocyte adhesion to NETs is receptor independent. In vitro monocyte adhesion to expelled NETs under static or flow conditions.
  • Figure 11 Monocytes were added to neutrophils (Ctrl) or NETting neutrophils (induced by A23187) and adhesion was quantified by a fluorescence plate reader. NETs were also degraded by DNase I.
  • Figure 12 Monocytes were perfused at 0.5 dyne/cm 2 over neutrophils, NETs, or degraded NETs and their adhesion was quantified manually.
  • Monocytes where pre-treated with antagonists or antibodies to chemokine receptors (Figure 13), Toll-like receptors ( Figure 14), or integrins (Figure 15) prior to incubation with NETs.
  • Monocytes fixed with PFA or treated with pertussis toxin (PTx) were used in static ( Figure 16) or flow (Figure 17) adhesion assays.
  • Data are analyzed by one way ANOVA with Dunnett's multiple comparisons test or Kruskal-Wallis test with Dunn's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001 , **** p£0.0001. All data are presented as meaniSEM.
  • Figures 18 - 25 Monocyte adhesion to NETs in regulated by cationic histone H2a. z-potential analysis of isolated monocytes treated with Ch-sulfate (Figure 18) or oleylamine (Figure 19).
  • Figure 20 Pearson correlation of monocyte adhesion to NETs versus monocyte z-potential.
  • Figure 21 Scheme of single cell atomic force microscopy (AFM) force spectroscopy. Monocytes were probed on expelled NETs at 200 pN contact force.
  • Figures 22 and 23 Quantification of the area under the curve reflecting the energy required to rupture the monocyte-NET interaction.
  • Figure 24 Proteome analysis of NET resident proteins.
  • Circle size reflects protein abundance, while color codes for charge (red cationic, green anionic).
  • Figure 25 Monocyte adhesion to NETs pre-incubated with antibodies to indicated NET-associated proteins. Data are analyzed by unpaired t-test, Kruskal-Wallis test with Dunn's multiple comparisons test or one-way ANOVA with Dunnett's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001. All data are presented as mean ⁇ SEM. HNP, human neutrophil peptides; MPO, myeloperoxidase; PR3, proteinase 3; NE, neutrophil elastase; CatG, cathepsin G.
  • Figure 26 - 40 Neutralization of histone H2a inhibits endotoxin-induced arterial myeloid cell recruitment and atheroprogression.
  • Experimental outline Apoe Cx 3 cr1 GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with isotype respective control (IgG), or a histone H2a-targeting antibody (anti-H2a).
  • IgG isotype respective control
  • anti-H2a anti-H2a
  • Figure 29 The structure of histone H2a (magenta), CHIP (orange) and the histone H2a-CHIP complex which was derived from protein-protein docking and molecular dynamic simulation. CHIP bound and interacted with the N- terminal part of histone H2a. The electrostatic interactions between Arg or Lys of Histone H2a (green sticks) with Glu or Asp of CHIP (cyan sticks) as well as hydrogen bonds (displayed as dash lines) help to stabilize the complex formation between histone H2a and CHIP.
  • Figures 30 - 33 Pharmacological interruption of histone H2a monocyte-binding was validated in static adhesion assays ( Figure 30) as well as by single cell force spectroscopy ( Figures 31 - 33).
  • Figures 34 - 40 In vivo validation of therapeutic effect of CHIP in endotoxin-accelerated atherosclerosis. Experimental outline. Apoe ⁇ / ⁇ Cx 3 cr1 GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with vehicle or CHIP (5 mg/kg).
  • Figures 35 - 37 Intravital microscopy was used to quantify luminal NET-like structures (Figure 35) in the left carotid artery as well as adherent neutrophils (Figure 37) and monocytes (Figure 36).
  • Figure 38 Aortic root lesion size analyzed after HE staining.
  • Figures 41 and 42 Blocking histone H2a does not affect plasma lipids or cell counts in blood but rather diminishes aortic myeloid cell accumulation.
  • Apoe Cx 3 cr1 GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with isotype respective control (IgG), or a histone H2a-targeting antibody (anti-H2a).
  • Figure 41- 42 Apoe Cx 3 cr1 GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with vehicle or CHIP (5 mg/kg).
  • Flow cytometry was used to assess neutrophils (G) and classical monocytes (H) in aortic homogenates. Data were analyzed with Mann-Whitney test (A-C) or Unpaired t test (D-H); **p£0.01. All data are presented as mean ⁇ SEM.
  • DAPI 4',6-Diamidino- 2-phenylindol
  • mice received antibodies to histone H2a (20 pg/mouse, Biorbyt), its respective IgG isotype control (Jackson ImmunoResearch), or a cyclical histone 2A interference peptide (CHIP, 5 mg/kg BW). All animal experiments were approved by the local ethics committee and performed in accordance with institutional guidelines.
  • Leukocyte counts were quantified by flow cytometry. Staining of single cell suspensions of blood and bone marrow were conducted using combinations of antibodies specific for CD45 (A20, eBioscience), CD11b (M1/70, eBioscience), CD115 (AFS98, eBioscience), Gr1 (RB6-8C5, BioLegend). Before cell staining, red blood cell lysis was performed.
  • aortas were digested by liberase (1.25mg/ml, Roche) and single cells were labeled with antibodies to CD11b (M1/70, bioLegend), Ly6G (1A8, BioLegend), MHC II (M5/114.15.2, BD Bioscience), Gr1 (RB6-8C5, BioLegend), F4/80 (BM8, BioLegend), CD45 (A20, eBioscience).
  • Cells were washed with Hanks Balanced Salt Solution (HBSS) and directly analyzed by flow cytometry using a FACSCanto II (BD). Absolute cell numbers were assessed by use of CountBrightTM absolute counting beads (Invitrogen). Data were analyzed with FlowJo Software (10.1 Flowjo LLC).
  • Neutrophil extracellular traps were measured in plasma by detecting myeloperoxidase and cell free DNA as described elsewhere [15] Briefly, MPO-DNA complexes were quantified by modified MPO ELISA (Hycult Biotech) combined with cell death detection kit (Roche).
  • Plasma samples were diluted 1 :50 in endotoxin free water and heat inactivated for 15 min at 70°C. Amebocyte lysate reagent was added to each well, incubated for 30 min followed by chromogenic substrate solution incubation. After 6 min of incubation, the stop solution was added, and the absorption was measured with Tecan infinite F200 pro (OD 405 nm).
  • the size of atherosclerotic plaques was measured in aortic roots after staining with Hematoxylin (Merck) and Eosin (Roth) followed by computerized image analysis and quantification (ImageJ, 1.48v). Aortic roots were stained with antibodies to Ly6G (1A8, BD Biosciences) and Mac2 (M3/38, biozol). Nuclei were counter-stained with DAPI (Thermo Fischer). A Leica DM4000 microscope with a 20x objective (Leica Microsystems) and a Leica DFC 365FX camera were used to capture images.
  • Monocytes were added (0.5x10 5 cell/well) to flat bottom 96-well plates (Falcon corning) or flow chambers (m-slide VI 0.1 ibiTreat (Ibidi)) containing either neutrophils (2x10 5 cells/well, labeled with cell tracker red CMTPX (Thermo fisher scientific)) or NETing neutrophils (25 mM calcium ionophore A23187 (Sigma Aldrich)) for 15 min or 3 min under applied flow (0.5 dyne/ cm 2 ; PHD ULTRA syringe pump, Harvard Apparatus). Non-adherent monocytes were washed off.
  • NETs were visualized by co-staining DNA with DAPI and citrullinated H3 or Sytox green nucleic acid stain (Thermo Fisher Scientific).
  • traps were treated with DNAse (10U, Sigma Aldrich), antibodies to Histone H2a (10 pg/ml; Cell Signaling), Myeloperoxidase (10 pg/ml; AB1224, Merck), Proteinase 3 (10 pg/ml; MAB6134, R&D systems), Neutrophil elastase (10 pg/ml; Biorbyt), Cathepsin G (10 pg/ml; Biorbyt), LL-37 (10 pg/ml; Santa Cruz Biotechnology), a-defensin (10 pg/ml; Hycult Biotech), CHIP (200 pg/ml, Pepscan).
  • monocytes were pre-incubated with antagonists to CCR1 (1 pM; BX471 , Tocris), CCR2 (1 mM; RS504393, Tocris), CCR5 (1 pM; SB32437, Tocris), CCR5 (0.1 mM; DAPTA, Tocris), CXCR2 (1 mM; SB225002, Tocris), CXCR4 (1 mM; AMD3100, Sigma Aldrich), FPR1 (10 mM: Cyclosporin H, Tocris), FPR2 (10 mM; WRW4, Tocris), TLR1-2 (100 mM; CU CPT 22, Tocris), TLR4 (100 mM; C34, Tocris), TLR9 (100 mM; Hydroxychloroquine sulfate, Tocris), VLA-4 (10 mM; BI01211 , Tocris), P2x7 (0.1 mM; A 74 0003, Tocris), or P
  • Binding affinity of the designed peptides were prioritized according to their predicted binding free energy and the peptide with the lowest binding free energy, which is CHIP (H-CEPLSEVEDYLDSSLKYNAKDTINYC- OH, SEQ ID NO: 1), containing an S-S bond between the cysteine residues at the N- and C-terminus, was selected for synthesis and further experimental testing.
  • CHIP H-CEPLSEVEDYLDSSLKYNAKDTINYC- OH, SEQ ID NO: 1
  • Neutrophils (0.5 x 10 6 ) were seeded on a flouro dish (WPI Inc.) with a glass bottom and left for adherence and NET-formation as described above.
  • a single monocyte was captured on a cantilever (MLCT-010, Bruker) coated with 0.1 mg/ml concavalin A (Merck).
  • MLCT-010 Molecular Cell Type 1
  • HBSS HBSS
  • Cantilever was routinely calibrated on a clean area in the probing dish.
  • Monocyte viability was controlled by propidiumiodid (5 mI/sample, eBioscience) staining.
  • AFM force spectroscopy was only performed with living cells (at least 8 cells per experimental setup).
  • the single monocyte on the cantilever tip was brought above a single NET-structure and probed with an approach and retraction speed of 10 pm/s, the pulling range was set to 25 pm, and the maximal contact force applied to the monocyte was 200 pN.
  • the monocytes were probed on an area of 10 pm 2 resulting in ten acquired force curves. Data were analyzed by using JPK Data Pocessing software (Version spm-5.0.96).
  • Isolated human neutrophils were stimulated to produce NETs. Alu I was added to each well to cut the NETs DNA into smaller soluble pieces. Samples were centrifuged to eliminate cell debris and keep the supernatant containing the DNA and associated NET proteins in solution. A commercially-available mass spectrometry sample preparation kit was utilized to ensure minimal sample loss and reproducibility. The supernatant containing the DNA and associated NET proteins in solution was prepared with the iST Kit for proteomic sample preparation (PreOmics GmbH). In brief, this involved denaturation, alkylation, digestion and peptide purification. Reduction of disulfide bridges, cysteine alkylation and protein denaturation was performed at 95°C for 10 min.
  • proteins were propionylated, trypsin and LysC were added to the mixture at a ratio of 1 :100 micrograms of enzyme to micrograms of protein. Digestion was performed at 37°C for 1 h. The cartridges were washed twice and eluted with 200 ul of 80%ACN + 0.25% TFA solution. The elution was speed vacuumed until dryness and stored until MS analysis.
  • Musher DM Abers MS, Corrales-Medina VF. Acute infection and myocardial infarction.
  • the New England journal of medicine. 2019;380:171-176 8.
  • Smeeth L Thomas SL, Hall AJ, Hubbard R, Farrington P, Vallance P. Risk of myocardial infarction and stroke after acute infection or vaccination.
  • Clark SR Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet tlr4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nature medicine. 2007;13:463-469
  • Circulating histones are major mediators of systemic inflammation and cellular injury in patients with acute liver failure. Cell Death Dis. 2016;7.

Abstract

The invention is in the field of medical treatments. It provides means and methods for the treatments of diseases related to or caused by neutrophil extracellular traps (NETs). More in particular, it relates to a peptide comprising an amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or an equivalent thereof that differs from SEQ ID NO: 1 by not more than 3 amino acids.

Description

Title: PEPTIDE FOR THE TREATMENT OF NET-ASSOCIATED DISEASES
Field of the invention
The invention is in the field of medical treatments. It provides means and methods for he treatments of diseases related to or caused by neutrophil extracellular traps (NETs).
Background of the invention
Acute infection is a well-established risk factor of cardiovascular inflammation increasing the risk for a cardiovascular complication within the first three weeks after infection. However, the nature of the processes underlying such aggravation remains unclear. Lipopolysaccharide (LPS) derived from Gram-negative bacteria is a potent activator of circulating immune cells including neutrophils, which foster inflammation through discharge of neutrophil extracellular traps (NETs).
Atherosclerosis is a lipid-driven chronic inflammation of the arterial vessel wall. In its late stages, atherosclerosis is the underlying pathophysiology of myocardial infarction and stroke and hence the leading cause of mortality worldwide. Monocytes and their descendants hold crucial roles throughout all stages of atherosclerosis as they contribute to lipid modification and respond with a pronounced inflammatory response upon uptake of modified lipids [1] With only limited numbers of macrophages residing in large arteries, the vast majority of monocytic cells needs to be de novo recruited in a process known as the leukocyte recruitment cascade, which is orchestrated by fine-tuned interactions of selectins, chemokines, and integrins and their respective receptors [2] In recent years, studies have provided evidence for the contribution of neutrophils to the arterial monocyte recruitment. Herein, neutrophils deposit preformed chemoattractants on endothelial cells or activate endothelial cells directly to stimulate monocyte adhesion [3-6]
Multiple bacterial and viral pathogens have been associated with atherosclerosis by seroepidemiologial studies and identification of the infectious agent in human atherosclerotic tissue. Moreover, there is strong clinical evidence for the acceleration of arterial inflammation by acute infection [7] As an example, urinary tract infection and bacteremia associate with an increase in the short-term risk of myocardial infarction [8, 9] Most striking data, however are available from patients with pneumonia, with the risk of myocardial infarction peaking at the onset of infection and is proportional to the severity of illness [10-12] Division of Gram-negative bacteria or their elimination leads to release of lipopolysaccharide (LPS) into the blood stream, i. e. endotoxinemia. Neutrophils are rapidly activated by LPS leading to the release of neutrophil extracellular traps (NETs), a complex structure composed of nuclear chromatin and proteins of nuclear cytoplasmatic and granule origin [13] Proteins reported to attract monocytes are localized within NETs. Moreover, NETs have been detected at the luminal side of large arteries [14]
The role of NETs in inflammation has still to be elucidated in more detail so that therapies for NET-related diseases can be developed and improved.
Summary of the invention
The invention relates to a peptide comprising an amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or equivalents thereof and their use in medical therapy.
Detailed description of the invention
Here we utilize a model of endotoxinemia to link acute infection and subsequent neutrophil activation with acceleration of vascular inflammation.
To investigate the effect of endotoxinemia on myeloid cell recruitment during atheroprogression, Apoe-/- mice were fed a high fed diet for 4 weeks and injected with LPS. Treatment in this way resulted in the striking expansion of atherosclerotic lesion sizes (Figure 1). While plasma cholesterol and triglyceride levels were not affected by treatment in this way, counts of circulating neutrophils and monocytes were depleted, likely due to margination and extravasation of activated myeloid cells. Indeed, the number of neutrophils, monocytes and macrophages recruited to atherosclerotic lesions as well as to inflamed aorta was drastically increased in LPS-treated mice (Figures 2 and 3). This notion was confirmed by use of intravital microscopy of carotid arteries performed in Apoe-/-Cx3cr1eGFP mice. There, LPS enhanced adhesion of both GFP+ monocytes as well as of antibody-labeled neutrophils.
LPS is known to trigger the release of NETs [20] and hence we investigated whether in our experimental setting NETs may contribute to heightened neutrophil and monocyte recruitment. Intravital microscopy permitted to visualize DAPI+ structures with a NET-like shape in the lumen of mice receiving LPS (Figure 4). Additionally, cell-free DNA as well as DNA-MPO complexes, a plasma marker of NETs, are strikingly increased in the plasma of mice with endotoxinemia (Figures 5 - 8). Importantly, plasma DNA-MPO complexes correlated with plasma endotoxin levels indicating that LPS directly induces NET release (Figures 5 - 8).
To test if NETs along the arterial lumen would contribute to the atherosclerosis phenotype observed in endotoxinemic mice, we treated mice with BB Cl-amidine, a potent inhibitor of protein arginine deiminases (PADs) with favorable pharmacokinetics in terms of plasma half-life and EC50. Intravital imaging confirmed that BB Cl-amidine treatment abrogated luminal NET release (Figure 4). In addition, heightened lesion formation as well as lesional accumulation of myeloid cells in response to LPS was completely abolished (Figure 1 - 3). In agreement herewith, BB Cl-amidine treatment led to a vast reduction of arterial myeloid cell adhesion (Figure 9, 10) suggesting that luminal NETs promote arterial myeloid cell adhesion during endotoxinemia.
We next investigated how NETs promote monocyte adhesion independent of receptor signaling. Therefore, we allowed classical monocytes to sediment on NETs in vitro. Indeed, we witnessed a significant adhesion of monocytes to NETting neutrophils (Figure 11). Fluorescence imaging revealed that these monocytes predominantly adhered directly to the NET scaffold and inhibition of monocyte adhesion after NET degradation with DNase I confirmed the importance of NETs in static monocyte adhesion (Figure 11). To transfer these findings to a physiologically relevant setting, we initiated NET release in adherent neutrophils and perfused classical monocytes in a flow chamber assay. As for the static adhesion assay, we found that monocytes primarily bound to DNA fibers and that degradation of NETs abolished adhesion evoked by activated neutrophils (Figure 12).
These observations are reminiscent of earlier work showing that proteins typically found in NETs or NETs themselves can increase cell adhesion by engaging chemokine receptor signaling and leukocyte integrin activation [2, 21-24] Thus, we neutralized chemokine receptors (Figure 13), receptors of alarmins (Figure 14), or integrins (Figure 15) prior to incubation with NETs.
Much to our surprise, none of these treatments impacted on adhesion evoked by NETs. Thus we concluded that the adhesion evoked by NETs may be signaling independent. Indeed, depletion of calcium by use of a chelator did not affect NET-mediated adhesion. In addition, abrogation of signaling of G-protein coupled receptors by pertussis toxin or even fixation of monocytes with 4% paraformaldehyde (PFA) did not impair adhesion of monocytes to NETs (Figure 16, 17). Taken together, these data show that monocyte adhesion to NETs is independent of receptor signaling.
We went on to show that NET-resident histone H2a attracts monocytes electrostatically. Given the signaling-independent adhesion of monocytes to NETs, we assumed that biophysical interactions such as electrostatic charges could be important in this process. In fact, monocytes present an overall negative surface charge (z potential -10.51±0.69 mV), while NETs are decorated with highly cationic proteins. To test the importance of charge interaction in monocyte adhesion to NETs, monocytes were incubated with either cholesterol sulfate or oleylamine. Cholesterol sulfate is a negatively-charged steroid lipid, whereas oleylamine is a positively-charged unsaturated fatty acid. Both lipids integrate with their lipophilic part into the phospholipid bilayer of the cell membrane and hence allow manipulating cell surface charge. In our hands, incubation with cholesterol sulfate or oleyelamine rendered monocyte surface charges more negative or more positive, respectively (Figure 18, 19).
To assess the relevance of the monocyte surface charge during adhesion to NETs we allowed monocytes of different surface charges to adhere to NETs. In these experiments we were able to generate a stringent correlation with more negative surface charges resulting in higher monocyte adhesion and vice versa (Figure 20). To assess the physical properties of charge-dependent adhesion in more depth, we performed atomic force spectroscopy with a monocyte immobilized at the tip of the cantilever. Atomic force microscopy (AFM) is a scanning probe microscope with piezoelectric elements to move the spring like cantilever. The deflecting cantilever is used to directly measure forces acting on the monocyte. A force-distance curve reports on the contact force (I), the force until maximum adhesion (II), the forces needed to fully detach the monocyte from the NET (III) and the distance of this detachment (IV) (Figure 21). Monocytes with different surface charges were immobilized at the tip of the AFM cantilever and in this position probed on NETs. Manipulation of the monocyte surface charges impacted on the maximum adhesion force, the detachment force and the detachment distance to separate monocytes from NETs, the energy required to separate the monocyte from the NET, and the adhesion frequency. Overall, NET-adhesion strength and the ability to adhere to NETs was heightened when monocytes were rendered more negatively charged and opposite effects were found in less negatively charged monocytes (Figure 22, 23), thus confirming the concept of charge-driven monocyte adhesion.
Neutrophil extracellular traps are decorated with a large array of granule- derived, cytoplasmatic, and nuclear proteins. Proteomics of NETs allowed the identification of 73 enriched polypeptides of which two clusters exhibited cationic surface charge as deferred from the peptides isoelectric point. One cluster was centered on nuclear histones of which histone H2a is the most abundant one. A second cluster revealed antimicrobial polypeptides with myeloperoxidase (MPO) and cathepsin G being the two candidates characterized by both cationic charge and high abundance (Figure 24). To assess the contribution of these NET-resident proteins towards monocyte adhesion, we treated NETs with antibodies towards these polypeptides and recorded monocyte adhesion. In these studies, neutralization of antimicrobial polypeptides abundantly found in NETs (MPO, cathepsin G) and of such previously found to contribute to monocyte adhesion (LL37, elastase, proteinase 3, HNP1-3) yielded no effect (Figure 25) [4, 5, 25] However, neutralization of histone H2a, the most abundant nuclear protein found in NETs, fully abrogated NET-driven monocyte adhesion (Figure 25). High resolution confocal microscopy revealed that monocytes indeed bound to NET-resident histone H2a. Finally, we found that histone H2a binds to monocytes in a charge dependent fashion, thus confirming our understanding of charge-mediated monocyte adhesion.
Then we set out to investigate whether therapeutic neutralization of histone H2a diminishes arterial monocyte adhesion and atheroprogression during endotoxinemia. Thus far our data suggest that histone H2a presented in NETs released during endotoxinemia causes myeloid cell adhesion, a process dramatically accelerating atheroprogression. Hence, we devised a protocol aimed at halting histone H2a-induced monocyte adhesion in vivo. Herein, a histone H2a neutralizing antibody or an isotype control IgG were administered in hypercholesterolemic, LPS-treated mice and luminal adhesive events were studied by intravital microscopy. Whereas treatment with histone H2a targeting antibodies did not impact on the presence of luminal NET-like structures and the number of circulating leukocytes, adhesion of myeloid cells was significantly reduced (Figures 26 - 28).
We also set out to design peptide inhibitors to neutralize histone H2a. Peptides contain several advantages such as ease of synthesis and optimization to improve pharmacokinetic and binding properties, large-scale and cost-effective production and applicability to conjugate with specific probes for drug delivery or for using as imaging tools [26] Moreover, we have previously demonstrated and proven that monocyte adhesion can be interrupted by peptide inhibitors [5, 17] and the highly positively charged N-terminus of histone H4 can be neutralized by a peptide [18] We in silico designed and developed peptides targeting histone H2a as described in the Examples section. The most potent peptide called Cyclical Histone 2A Interference Peptide (CHIP), with amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC- OH, (SEQ ID NO: 1) bound to the N-terminal domain of histone 2A, and the electrostatic interactions between positively charged residues (Arg and Lys) of histone H2a with negatively charged residues (Asp and Glu) of CHIP and H-bond interactions promoted a stable complex formation (Figure 29).
To test the functionality of this peptide we first performed in vitro experiments. Herein, pretreatment of NETs with CHIP reduced adhesion significantly (Figure 30). Biophysically, CHIP reduced the interaction strength of monocytes and NETs as shown by atomic force microscopy (Figures 31 - 33).
We then set out to testing CHIP in vivo. Delivery of CHIP to hypercholesterolemic, LPS-treated mice, did not impact on the presence of NET-like structures in the arterial lumen, but significantly diminished arterial adhesion of neutrophils and monocytes (Figures 34 - 37). Beyond impacting on luminal events, CHIP allowed to drastically reduce atherosclerotic lesion sizes (Figure 38). These changes in lesion size were not associated with differences in blood counts or plasma lipid levels. The lesions of mice treated with CHIP were characterized by the accumulation of less neutrophils and macrophages (Figures 39, 40). In line herewith, aortic neutrophil and monocyte numbers were strikingly reduced by CHIP (Figures 41, 42). Taken together, neutralization of histone H2a by a peptide according to SEQ ID NO: 1 results in the reduction of arterial myeloid cell recruitment and accelerated atherosclerosis evoked by endotoxinemia.
It will be clear from the above results that other NETs-associated diseases can be treated equally well with the peptide according to SEQ ID NO: 1. Such NETs-associated diseases include for example autoimmune disorders [40], stroke [41], (transfusion-related) acute lung injury [36,37,38,42], ischaemia/reperfusion injury (IRI) [38,39,43], kidney injury [39,44], liver injury [45,46], trauma [47] and sepsis [48,49] Histones are not only released as a consequence of cellular injury, they also actively contribute towards a self-sustaining cascade of ongoing cell death [50] The invention may also be applied in the treatment of Acute respiratory distress syndrome such as ARDS caused by infection with coronavirus [51 - 53]
The peptide according to SEQ ID NO: 1 can therefore also be used to treat inflammation.
The teaching as provided herein should not be interpreted so narrowly that it relates only to a peptide comprising the sequence according to SEQ ID NO: 1. An alanine scan of the peptide according to SEQ ID NO: 1 can be easily performed by the skilled person and will reveal that at least 1 , such as 2 and 3 amino acids in SEQ ID NO: 1 can be replaced by another amino acid without affecting the function of the peptide.
Moreover, when we tested the peptide according to SEQ ID NO: 1 as a circular peptide wherein the N-terminal cysteine residue and the C-terminal cysteine residue were used to circularise the peptide, results were even improved. Hence, the invention also relates to a peptide comprising an amino acid sequence H- CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or an equivalent thereof that differs from SEQ ID NO: 1 by not more than 3 amino acids that is circular.
In summary, the following may be noted from this study. Atherosclerosis is the leading cause of mortality in Western society. Excess mortality from cardiovascular disease during influenza epidemics was first recognized early in the 20th century, but the specific association of influenza and other infections with myocardial infarction was not characterized until decades later. In fact, over the last 20 years epidemiological studies have consistently demonstrated the association between acute infection with multiple bacterial and viral pathogens and the short term increase in cardiovascular complications [7] As an example, a self-controlled case series involving U.S. veterans showed a remarkable increase in the risk of myocardial infarction during the first 15 days after hospitalization for acute bacterial pneumonia, to a risk that was 48 times higher than that in any 15-day period during the year before or after the onset of infection [27] An increase in the short-term risk of myocardial infarction has also been described in association with urinary tract infection and bacteremia [8, 9] The strength and temporal pattern of the association between acute infections and an increased risk of myocardial infarction suggest a causal relationship.
Because the association has been shown with a variety of pathogens, sites of infection, and the association is stronger and lasts longer when the infection is more severe, it is likely that the infection and the host response to infection are major determinants in this relationship. Here we used a model of endotoxinemia to mimic acute, severe infection. In these experiments, administration of endotoxin induces a rapid expansion of atherosclerotic lesions characterized by extension of the lesional myeloid cell compartment. This increase was fully abrogated when release of NETs was pharmacologically inhibited. Our studies identify a mechanism that centers on monocyte adhesion to luminal NETs in general and to NET-resident histone H2a in particular. Neutralization of histone H2a by antibodies or by in silico designed peptides permits inhibition of endotoxin-accelerated monocyte adhesion and lesional myeloid cell accumulation, thus providing a potential strategy for improved care of patients at risk of cardiovascular events and experiencing an acute infection.
Since our study was performed with endotoxin stimulation only, this may be perceived as limitation in the applicability of our study to clinical scenarios. However, an increased risk for cardiovascular complications after an acute infections has been observed for numerous pathogens, including viruses such as influenza virus, corona virus, or respiratory syncytial virus [7], Gram-positive bacteria including Streptococcus pneumonia and Staphylococcus aureus, as well as Gram-negative bacteria like Escherichia coli and Haemophilus influenza [9, 10, 12, 28] Interestingly, NET release is triggered by a variety of stimuli including all of the pathogens listed above [29] and hence the NET-centered mechanism identified herein may in part be applicable to a large variety of pathogen-associated cardiovascular complications although confirmation in additional studies is required.
Neutrophils have previously been reported to pave the way for inflammatory monocytes into developing atherosclerotic lesions. Rendering mice neutropenic during initial stages of atherosclerosis diminishes lesion sizes as well as lesion composition, with lowered macrophage accumulation [30] Neutrophils stimulate various mechanisms that promote monocyte recruitment. Among these, secretion of chemotactic proteins stands out as an important mechanism for arterial monocyte recruitment. Cathepsin G, cathelicidin, complexes formed of neutrophil-derived a- defensin and platelet-borne CCL5, and CCL2 released from neutrophils in a circadian fashion were shown to induce firm monocyte adhesion in mouse models of vascular inflammation when immobilized on arterial endothelial cells [3-5, 31] However, all these ligands bind to specific receptors and increase adhesion through increased integrin affinity and avidity. Herein we identify electrostatic interactions between cationic histone H2a and negatively charged monocyte surfaces as a novel mechanism driving infection-associated monocyte recruitment into atherosclerotic lesions. Histone H2a combines abundance within NETs and surface charge and may hence stand out as an important epitope for monocyte adhesion. However, other histone isotypes may engage similar activities as they are comparable in abundance and charge within NETs. Of note, a recent study has shown that activated smooth muscle cells residing in the fibrous cap of advanced atherosclerotic lesions stimulate neutrophils to release NETs. These are rich in cytotoxic histone H4 that can puncture human and murine SMCs leading to their death and, when re-occurring, thinning of the fibrous cap [18] Thus, therapeutic neutralization of histone epitopes may exert a dual beneficial effect allowing to limit macrophage burden and fibrous cap thinning.
The mechanism of NET-driven monocyte recruitment is receptor independent and hence lacks specificity. In fact, surgical removal of tumors (especially colorectal cancer) is frequently associated with bacteremia and NETs triggered in such settings have been suggested to promote immobilization of tumor cells in vascular beds thus promoting formation of metastases [32-34] Mechanistically, some studies reported betal integrins on tumor cells to mediate adhesion to NETs while other studies found unspecific binding to NETs underlying the process of NET-driven metastasis [2, 21 , 22] Consequently, degradation of NETs using DNase I has been found to limit metastases formation in animal models combining infection and tumor development [2, 21] Observations made in the here presented study suggest that histone H2a can also promote adhesion of tumor cells and promote metastases and perioperative treatment with histone-neutralizing therapies may provide an efficient way to reduce cancer spreading.
Infections in cardiovascular risk patients increase the chance of cardiovascular complications within the first three weeks after an infection several fold. We here identify a mechanism centered on extracellular histone H2a which induces adhesion and recruitment of monocytes. While data from the CANTOS trial reveal the overall positive effects of anti-inflammation therapy in the context of atherosclerosis, neutralization of ILIbeta was also associated to heightened risk of bacterial infections35. Therapeutic neutralization of histone H2a will likely not be linked to such adverse side effects as the antimicrobial actions of histone H2a can be compensated by the abundance of other antimicrobial polypeptides residing within NETs. In addition, the strict focus on infection-associated cardiovascular complications will be relevant to a very short time window of not more than three weeks after infection and can be combined with standard antibiotic treatment. Thus, targeting histone H2a may stand out as an innovative way to lower arterial monocyte recruitment accelerated by infection while coming with limited intrinsic side effects.
The invention therefore relates to a peptide comprising an amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1). More in particular it relates to a peptide as described herein consisting of the amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1). Such a peptide may advantageously be used in the treatment of a disease, more in particular in the treatment of a NETs-related disease. In one embodiment of the invention, the treatment is related to the treatment of a NETs-related disease selected from the group consisting of atherosclerosis, autoimmune disorders, stroke, transfusion -related acute lung injury, acute lung injury, lung injury, ischaemia/reperfusion injury (IRI), kidney injury, liver injury, trauma, sepsis and inflammation.
The teaching as provided herein should not be so narrowly interpreted as that only peptides comprising the exact sequences of SEQ ID NO: 1 would work in a therapy as described in the claims. Equivalents of such a peptide will work as well and can be found by the skilled person with only little experimental effort. So is it well within range of the skills of that person to produce and test peptides that differ only one, two or even three amino acids from the sequence as provided in SEQ ID NO: 1 and still work. Such peptides are termed herein “equivalents” or “equivalent peptides”.
Legend to the figures
Figures 1 - 4: Endotoxinemia accelerates atherosclerosis. Experimental setup. Apoe or Apoe^Cxacrl^ mice were fed a HFD for 4 weeks and treated with either PBS (Ctrl) or with LPS (1 mg/kg BW). Another LPS-treated group received a BB Cl-amidine (BB Cl-A, 1mg/kg BW) 12 h before and along with LPS injection. Figure 1 : Aortic root lesion size analyzed in HE-stained sections. Representative images, scale bar 300 pm. Figure 2: Lesional neutrophils (Ly6G+ cells) quantified in root sections. Figure 3: Macrophages (Mac2+ cells) quantified in root sections. Figure 4: Count of luminal NET-like structures in left common carotid artery. Data are analyzed by one- way ANOVA with Dunnett's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001 , ****p£0.0001. All data are presented as meaniSEM.
Figures 5 - 8: Endotoxinemia-triggered NET release stimulates arterial myeloid cell recruitment. Apoe mice were fed a HFD for 4 weeks and treated with either PBS (Ctrl) or with LPS (1 mg/kg BW). Another LPS-treated group received a BB Cl-amidine (BB Cl-A, 1 mg/kg BW) 12 h before and along with LPS injection. Quantification of cell free DNA in plasma (Figure 5), plasma endotoxin units (Figure 6), and circulating MPO-DNA complexes (Figure 7) as measure of NET release. Pearson correlation of NETs (MPO-DNA complexes) with plasma endotoxin units (Figure 8). Data are analyzed by one-way ANOVA with Dunnett's multiple comparisons test, Kurskal-Wallis test with Dunn’s post test or unpaired t-test; *p£0.05, **p£0.01 , ***p£0.001, ****p£0.0001. All data are presented as mean ±SEM.
Figures 9 - 10: Endotoxinemia accelerates atherosclerosis.
Experimental setup. Apoe or Apoe^Cxacrl^ mice were fed a HFD for 4 weeks and treated with either PBS (Ctrl) or with LPS (1 mg/kg BW). Another LPS-treated group received a BB Cl-amidine (BB Cl-A, 1 mg/kg BW) 12 h before and along with LPS injection. Quantification of luminally adherent neutrophils (Figure 9) and monocytes (Figure 10).
Figures 11 - 17: Monocyte adhesion to NETs is receptor independent. In vitro monocyte adhesion to expelled NETs under static or flow conditions. Figure 11 : Monocytes were added to neutrophils (Ctrl) or NETting neutrophils (induced by A23187) and adhesion was quantified by a fluorescence plate reader. NETs were also degraded by DNase I. Figure 12: Monocytes were perfused at 0.5 dyne/cm2 over neutrophils, NETs, or degraded NETs and their adhesion was quantified manually. Monocytes where pre-treated with antagonists or antibodies to chemokine receptors (Figure 13), Toll-like receptors (Figure 14), or integrins (Figure 15) prior to incubation with NETs. Monocytes fixed with PFA or treated with pertussis toxin (PTx) were used in static (Figure 16) or flow (Figure 17) adhesion assays. Data are analyzed by one way ANOVA with Dunnett's multiple comparisons test or Kruskal-Wallis test with Dunn's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001 , **** p£0.0001. All data are presented as meaniSEM.
Figures 18 - 25: Monocyte adhesion to NETs in regulated by cationic histone H2a. z-potential analysis of isolated monocytes treated with Ch-sulfate (Figure 18) or oleylamine (Figure 19). Figure 20: Pearson correlation of monocyte adhesion to NETs versus monocyte z-potential. Figure 21 : Scheme of single cell atomic force microscopy (AFM) force spectroscopy. Monocytes were probed on expelled NETs at 200 pN contact force. Figures 22 and 23: Quantification of the area under the curve reflecting the energy required to rupture the monocyte-NET interaction. Figure 24: Proteome analysis of NET resident proteins. Circle size reflects protein abundance, while color codes for charge (red cationic, green anionic). Figure 25: Monocyte adhesion to NETs pre-incubated with antibodies to indicated NET-associated proteins. Data are analyzed by unpaired t-test, Kruskal-Wallis test with Dunn's multiple comparisons test or one-way ANOVA with Dunnett's multiple comparisons test; *p£0.05, **p£0.01 , ***p£0.001. All data are presented as mean ±SEM. HNP, human neutrophil peptides; MPO, myeloperoxidase; PR3, proteinase 3; NE, neutrophil elastase; CatG, cathepsin G.
Figure 26 - 40: Neutralization of histone H2a inhibits endotoxin-induced arterial myeloid cell recruitment and atheroprogression. Experimental outline: Apoe Cx3cr1GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with isotype respective control (IgG), or a histone H2a-targeting antibody (anti-H2a). (Figures 26 - 28) Intravital microscopy was used to quantify luminal NET-like structures (Figure 26) in the left carotid artery as well as adherent neutrophils (Figure 27) and monocytes (Figure 28). Figure 29: The structure of histone H2a (magenta), CHIP (orange) and the histone H2a-CHIP complex which was derived from protein-protein docking and molecular dynamic simulation. CHIP bound and interacted with the N- terminal part of histone H2a. The electrostatic interactions between Arg or Lys of Histone H2a (green sticks) with Glu or Asp of CHIP (cyan sticks) as well as hydrogen bonds (displayed as dash lines) help to stabilize the complex formation between histone H2a and CHIP. Figures 30 - 33: Pharmacological interruption of histone H2a monocyte-binding was validated in static adhesion assays (Figure 30) as well as by single cell force spectroscopy (Figures 31 - 33). Figures 34 - 40: In vivo validation of therapeutic effect of CHIP in endotoxin-accelerated atherosclerosis. Experimental outline. Apoe~/~Cx3cr1GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with vehicle or CHIP (5 mg/kg). Figures 35 - 37: Intravital microscopy was used to quantify luminal NET-like structures (Figure 35) in the left carotid artery as well as adherent neutrophils (Figure 37) and monocytes (Figure 36). Figure 38: Aortic root lesion size analyzed after HE staining. Quantification of Lesional neutrophils (Ly6G+ cells) (Figure 39), and macrophages (Mac2+ cells) (Figure 39). Figure 40: Representative immunofluorescence images showing lesional Mac2+ cells (grey) and nuclei (DAPI, blue), scale bar 50 pm. Data are analyzed by Mann-Whitney test; *p£0.05, **p£0.01 , ***p£0.001. All data are presented as mean ±SEM.
Figures 41 and 42: Blocking histone H2a does not affect plasma lipids or cell counts in blood but rather diminishes aortic myeloid cell accumulation. Apoe Cx3cr1GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with isotype respective control (IgG), or a histone H2a-targeting antibody (anti-H2a). Figure 41- 42: Apoe Cx3cr1GFP were fed a HFD for 4 weeks, treated with LPS (1 mg/kg, 4h) and injected with vehicle or CHIP (5 mg/kg). Flow cytometry was used to assess neutrophils (G) and classical monocytes (H) in aortic homogenates. Data were analyzed with Mann-Whitney test (A-C) or Unpaired t test (D-H); **p£0.01. All data are presented as mean ±SEM.
Examples
Example 1 : Animal experiments
We surveyed atheroprogression in Apoe or Apoe^Cxscrl^ reporter mice on C57BI/6J background after 4 weeks of high-fat diet (HFD) (21% fat, Ssniff). Endotoxinemia was induced by intraperitoneal (i.p.) injection of lipopolysaccharide (LPS, Escherichia coii, 0111 :B4, Sigma Aldrich, 1 mg/kg BW, i.p.). Control mice received vehicle (PBS, 100 pi, i.p.). Thereafter, atherosclerotic lesions were analyzed in aortic root sections or cell adhesion was studied by intravital microscopy of the left carotid artery [4] To visualize neutrophils and NET-like structures, mice were injected i.v. with a PE-conjugated antibody to Ly6G (1A8, 1 pg, Biolegend) and 4',6-Diamidino- 2-phenylindol (DAPI, Thermo Fischer). To assess the effect of NETs we blocked NET- formation with BB Cl-amidine (1 mg/kg BW, Cayman Chemical Company). In additional experiments, mice received antibodies to histone H2a (20 pg/mouse, Biorbyt), its respective IgG isotype control (Jackson ImmunoResearch), or a cyclical histone 2A interference peptide (CHIP, 5 mg/kg BW). All animal experiments were approved by the local ethics committee and performed in accordance with institutional guidelines.
Example 2: Flow cytometry
Leukocyte counts were quantified by flow cytometry. Staining of single cell suspensions of blood and bone marrow were conducted using combinations of antibodies specific for CD45 (A20, eBioscience), CD11b (M1/70, eBioscience), CD115 (AFS98, eBioscience), Gr1 (RB6-8C5, BioLegend). Before cell staining, red blood cell lysis was performed. Furthermore, aortas were digested by liberase (1.25mg/ml, Roche) and single cells were labeled with antibodies to CD11b (M1/70, bioLegend), Ly6G (1A8, BioLegend), MHC II (M5/114.15.2, BD Bioscience), Gr1 (RB6-8C5, BioLegend), F4/80 (BM8, BioLegend), CD45 (A20, eBioscience). Cells were washed with Hanks Balanced Salt Solution (HBSS) and directly analyzed by flow cytometry using a FACSCanto II (BD). Absolute cell numbers were assessed by use of CountBright™ absolute counting beads (Invitrogen). Data were analyzed with FlowJo Software (10.1 Flowjo LLC).
Example 3: Cell free DNA measurement
Cell free DNA was measured in the plasma with Quant-iT PicoGreen dsDNA assay (Life Technologies) the mean fluorescence intensity was quantified and according to the standard curve, results are expressed as ng/ml . Samples were measured with a plate reader (Tecan infinite F200 pro).
Example 4: NET-ELISA
Neutrophil extracellular traps were measured in plasma by detecting myeloperoxidase and cell free DNA as described elsewhere [15] Briefly, MPO-DNA complexes were quantified by modified MPO ELISA (Hycult Biotech) combined with cell death detection kit (Roche).
Example 5: Endotoxin measurement
LPS plasma levels were assessed with Pierce Chromogenic Endotoxin Quant kit (Thermo Fisher Scientific). Plasma samples were diluted 1 :50 in endotoxin free water and heat inactivated for 15 min at 70°C. Amebocyte lysate reagent was added to each well, incubated for 30 min followed by chromogenic substrate solution incubation. After 6 min of incubation, the stop solution was added, and the absorption was measured with Tecan infinite F200 pro (OD 405 nm).
Example 6: Histology and Immunohistochemistry
The size of atherosclerotic plaques was measured in aortic roots after staining with Hematoxylin (Merck) and Eosin (Roth) followed by computerized image analysis and quantification (ImageJ, 1.48v). Aortic roots were stained with antibodies to Ly6G (1A8, BD Biosciences) and Mac2 (M3/38, biozol). Nuclei were counter-stained with DAPI (Thermo Fischer). A Leica DM4000 microscope with a 20x objective (Leica Microsystems) and a Leica DFC 365FX camera were used to capture images.
Example 7: In vitro adhesion assay
Blood was drawn from healthy volunteers. Neutrophils were isolated by polymorphprep according to the manufacturer’s instructions. Monocytes were isolated via negative selection using monocyte isolation kit II (MACS Miltenyi biotec). Adhesion of monocytes to NETs was studied under static and flow conditions. Isolated monocytes were labeled with CellTrace™ calcein violet AM (Thermo fisher scientific). Monocytes were added (0.5x105 cell/well) to flat bottom 96-well plates (Falcon corning) or flow chambers (m-slide VI 0.1 ibiTreat (Ibidi)) containing either neutrophils (2x105 cells/well, labeled with cell tracker red CMTPX (Thermo fisher scientific)) or NETing neutrophils (25 mM calcium ionophore A23187 (Sigma Aldrich)) for 15 min or 3 min under applied flow (0.5 dyne/ cm2; PHD ULTRA syringe pump, Harvard Apparatus). Non-adherent monocytes were washed off. Statically adherent monocytes were quantified in a microplate reader (Tecan infinite™ 200 pro). Monocytes adherent under flow conditions were counted manually. For each flow channel three fields were quantified and averaged. NETs were visualized by co-staining DNA with DAPI and citrullinated H3 or Sytox green nucleic acid stain (Thermo Fisher Scientific). To study monocyte adhesion to NETs, traps were treated with DNAse (10U, Sigma Aldrich), antibodies to Histone H2a (10 pg/ml; Cell Signaling), Myeloperoxidase (10 pg/ml; AB1224, Merck), Proteinase 3 (10 pg/ml; MAB6134, R&D systems), Neutrophil elastase (10 pg/ml; Biorbyt), Cathepsin G (10 pg/ml; Biorbyt), LL-37 (10 pg/ml; Santa Cruz Biotechnology), a-defensin (10 pg/ml; Hycult Biotech), CHIP (200 pg/ml, Pepscan). In another experimental setting monocytes were pre-incubated with antagonists to CCR1 (1 pM; BX471 , Tocris), CCR2 (1 mM; RS504393, Tocris), CCR5 (1 pM; SB32437, Tocris), CCR5 (0.1 mM; DAPTA, Tocris), CXCR2 (1 mM; SB225002, Tocris), CXCR4 (1 mM; AMD3100, Sigma Aldrich), FPR1 (10 mM: Cyclosporin H, Tocris), FPR2 (10 mM; WRW4, Tocris), TLR1-2 (100 mM; CU CPT 22, Tocris), TLR4 (100 mM; C34, Tocris), TLR9 (100 mM; Hydroxychloroquine sulfate, Tocris), VLA-4 (10 mM; BI01211 , Tocris), P2x7 (0.1 mM; A 74 0003, Tocris), or P2y2 (10 mM; AR- C118925xx, Tocris). To block chemokine receptor signaling, cells were pretreated with Pertussis toxin (0.8 pg/ml, Sigma-Aldrich); Ca2+ mobilization was abrogated using the calcium chelator BAPTA AM (2 mM; Thermo Fisher Scientific). Finally, antibodies to Mac1 (M1/70, 10 mg, Biolegend) or LFA1 (10 mg, R7.1 , Biolegend) were used to test the relevance of integrin activation.
Example 8: Molecular Dynamics (MD) Simulations and Peptide Design
Three-dimensional (3D) structures of protein-protein complexes were used as a starting point for rationally design and develop bioactive compounds to modulate protein-protein interactions [16-18] Thus, the complex between human BRPF1 bromodomain and histone H2a peptide (PDB code: 4QYL) was selected and utilized to design potential histone H2a peptide inhibitors. The loop regions (residue 31-43 and 78-91) of bromodomain binding with the N-terminal tail of histone H2a were extracted and used as a starting peptide which were then docked onto the N-termini histone H2a (chain C of the nucleosome structure, PDB code: 1 KX5). Based on the derived complex between the template peptide and histone H2a several peptides were designed and then docked onto the N-terminal tail of histone H2a as well by application of protein-protein docking program (HADDOCK2.2 Webserver [19]). The derived histone H2a -peptide complexes were subsequently subjected to structural optimization and binding free energy calculations to predict binding strength of peptides with histone H2a. Similar protocols and standard parameters as in our previous work were applied for these purposes [17, 18] Briefly, energy minimization was carried out for 10,000 steps (5,000 steps of steepest descent followed by 5,000 steps of conjugate gradient algorithm) and subsequently a short MD simulation (500 ps) was performed by using TIP3P water model and AMBER14SB force field for peptides and protein (histone H2a) and setting essential parameters at the standard values (e.g. temperature at 300 K, pressure at 1 bar, time step at 2 fs with SHAKE constraint). Binding free energy was calculated by using molecular mechanics/generalized Born surface area (MM/GBSA) approach (generalized Born model 8 with default parameters). MD simulations and binding calculations were carried out by using AMBER16 program. Binding affinity of the designed peptides were prioritized according to their predicted binding free energy and the peptide with the lowest binding free energy, which is CHIP (H-CEPLSEVEDYLDSSLKYNAKDTINYC- OH, SEQ ID NO: 1), containing an S-S bond between the cysteine residues at the N- and C-terminus, was selected for synthesis and further experimental testing. Example 9: Zeta (Ci potential
Cell surface charge of either untreated monocytes, monocytes incubated with sodium cholesterol sulfate (200 mM, Sigma-Aldrich) and sulfatase inhibitor STX64 (1 mM, Sigma-Aldrich), or monocytes incubated with oleylamine (200 mM, Sigma- Aldrich) was measured using a Malvern Zetasizer Nano. For z potential measurements, 10 mI cell suspension (1x106 cells/ml) was with mixed with 90 mI 300 mM sucrose (Sigma-Aldrich) and the sample was loaded at the bottom of a DTS1070 cuvette (Malvern Instruments) that was prefilled with 10 mM sodium chloride (Sigma- Aldrich). Measurements 30 runs per measurement, monomodal mode) were recorded at 37 °C as technical triplicates.
Example 10: Atomic force microscopy
Neutrophils (0.5 x 106) were seeded on a flouro dish (WPI Inc.) with a glass bottom and left for adherence and NET-formation as described above. A single monocyte was captured on a cantilever (MLCT-010, Bruker) coated with 0.1 mg/ml concavalin A (Merck). Before capturing monocytes, the dish and cantilever were washed 3x (HBSS, Gibco) and the cantilever was routinely calibrated on a clean area in the probing dish. Monocyte viability was controlled by propidiumiodid (5 mI/sample, eBioscience) staining. AFM force spectroscopy was only performed with living cells (at least 8 cells per experimental setup). The single monocyte on the cantilever tip was brought above a single NET-structure and probed with an approach and retraction speed of 10 pm/s, the pulling range was set to 25 pm, and the maximal contact force applied to the monocyte was 200 pN. The monocytes were probed on an area of 10 pm2 resulting in ten acquired force curves. Data were analyzed by using JPK Data Pocessing software (Version spm-5.0.96).
Example 11 : Sample preparation for MS
Isolated human neutrophils were stimulated to produce NETs. Alu I was added to each well to cut the NETs DNA into smaller soluble pieces. Samples were centrifuged to eliminate cell debris and keep the supernatant containing the DNA and associated NET proteins in solution. A commercially-available mass spectrometry sample preparation kit was utilized to ensure minimal sample loss and reproducibility. The supernatant containing the DNA and associated NET proteins in solution was prepared with the iST Kit for proteomic sample preparation (PreOmics GmbH). In brief, this involved denaturation, alkylation, digestion and peptide purification. Reduction of disulfide bridges, cysteine alkylation and protein denaturation was performed at 95°C for 10 min. After a 5 min cooling step at room temperature, proteins were propionylated, trypsin and LysC were added to the mixture at a ratio of 1 :100 micrograms of enzyme to micrograms of protein. Digestion was performed at 37°C for 1 h. The cartridges were washed twice and eluted with 200 ul of 80%ACN + 0.25% TFA solution. The elution was speed vacuumed until dryness and stored until MS analysis.
Example 12: LC-MS/MS analysis
Peptides were re-suspended in 17 pi of 0.1 % TFA. A total of 5.0 mI were injected into a nano-HPLC device (Thermo Fisher Scientific) using a gradient from 4% B to 90% B (solvent A 0.1% FA in water, solvent B 80% ACN, 0.1% FA in water) over 90 min at a flow rate of 300 nl/min. Data was acquired in DDA positive mode using a Q Exactive HF spectrometer (Thermo Fisher Scientific). MS1 spectra were acquired at 60K resolution and AGC target value of 3e6 within a m/z range of 250 to 1600. The top 10 precursor ions were isolated with a window of 2.0 m/z and fragmented with NCE of 27 eV. Dynamic exclusion windows of of 12.0 ppm and 20 s were used. MS2 spectra were obtained at 15K resolution and AGC target value of 1e5 within a m/z range of 200 to 2000.
Example 13: Statistics
All statistics analyses were performed by using GraphPad Prism 8 software. Outliers have been determined by Grubbs' test with a=0.05. To test normal distribution, the D’Agostino-Pearson omnibus test was used. If normality was passed, data were tested by two-tailed unpaired t-test or one-way ANOVA with Dunnet’s correction. The Mann-Whitney test or Kurskal-Wallis test with Dunn’s correction was performed when data were not normally distributed. In all used tests a 95% confidence interval was utilized with p<0.05 was assumed as significant difference. All data are represented as mean ±SEM. References
1. Fayad ZA, Swirski FK, Calcagno C, Robbins CS, Mulder W, Kovacic JC. Monocyte and macrophage dynamics in the cardiovascular system: Jacc macrophage in cvd series (part 3). Journal of the American College of Cardiology. 2018;72:2198- 2212
2. Park J, Wysocki RW, Amoozgar Z, Maiorino L, Fein MR, Jorns J, Schott AF, Kinugasa-Katayama Y, Lee Y, Won NH, Nakasone ES, Hearn SA, Kuttner V, Qiu J, Almeida AS, Perurena N, Kessenbrock K, Goldberg MS, Egeblad M. Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps. Science translational medicine. 2016;8:361ra138
3. Winter C, Silvestre-Roig C, Ortega-Gomez A, Lemnitzer P, Poelman H, Schumski A, Winter J, Drechsler M, de Jong R, Immler R, Sperandio M, Hristov M, Zeller T, Nicolaes GAF, Weber C, Viola JR, Hidalgo A, Scheiermann C, Soehnlein O. Chrono-pharmacological targeting of the ccl2-ccr2 axis ameliorates atherosclerosis. Cell metabolism. 2018;28:175-182 e175
4. Ortega-Gomez A, Salvermoser M, Rossaint J, Pick R, Brauner J, Lemnitzer P, Tilgner J, de Jong RJ, Megens RT, Jamasbi J, Doring Y, Pham CT, Scheiermann C, Siess W, Drechsler M, Weber C, Grommes J, Zarbock A, Walzog B, Soehnlein O. Cathepsin g controls arterial but not venular myeloid cell recruitment. Circulation. 2016;134:1176-1188
5. Alard JE, Ortega-Gomez A, Wichapong K, Bongiovanni D, Horckmans M, Megens RT, Leoni G, Ferraro B, Rossaint J, Paulin N, Ng J, Ippel H, Suylen D, Hinkel R, Blanchet X, Gaillard F, D'Amico M, von Hundelshausen P, Zarbock A, Scheiermann C, Hackeng TM, Steffens S, Kupatt C, Nicolaes GA, Weber C, Soehnlein O. Recruitment of classical monocytes can be inhibited by disturbing heteromers of neutrophil hnp1 and platelet ccl5. Science translational medicine. 2015;7:317ra196
6. Zhang J, Alcaide P, Liu L, Sun J, He A, Luscinskas FW, Shi GP. Regulation of endothelial cell adhesion molecule expression by mast cells, macrophages, and neutrophils. PloS one. 2011 ;6:e14525
7. Musher DM, Abers MS, Corrales-Medina VF. Acute infection and myocardial infarction. The New England journal of medicine. 2019;380:171-176 8. Smeeth L, Thomas SL, Hall AJ, Hubbard R, Farrington P, Vallance P. Risk of myocardial infarction and stroke after acute infection or vaccination. The New England journal of medicine. 2004;351:2611-2618
9. Dalager-Pedersen M, Sogaard M, Schonheyder HC, Nielsen H, Thomsen RW. Risk for myocardial infarction and stroke after community-acquired bacteremia: A 20-year population-based cohort study. Circulation. 2014;129:1387- 1396
10. Corrales-Medina VF, Musher DM, Wells GA, Chirinos JA, Chen L, Fine MJ. Cardiac complications in patients with community-acquired pneumonia: Incidence, timing, risk factors, and association with short-term mortality. Circulation. 2012;125:773-781
11. Ramirez J, Aliberti S, Mirsaeidi M, Peyrani P, Filardo G, Amir A, Moffett B, Gordon J, Blasi F, Bordon J. Acute myocardial infarction in hospitalized patients with community-acquired pneumonia. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2008;47:182-187
12. Corrales-Medina VF, Alvarez KN, Weissfeld LA, Angus DC, Chirinos JA, Chang CC, Newman A, Loehr L, Folsom AR, Elkind MS, Lyles MF, Kronmal RA, Yende S. Association between hospitalization for pneumonia and subsequent risk of cardiovascular disease. Jama. 2015;313:264-274
13. Pieterse E, Rother N, Yanginlar C, Hilbrands LB, van der Vlag J. Neutrophils discriminate between lipopolysaccharides of different bacterial sources and selectively release neutrophil extracellular traps. Frontiers in immunology. 2016;7:484
14. Megens RT, Vijayan S, Lievens D, Doring Y, van Zandvoort MA, Grommes J, Weber C, Soehnlein O. Presence of luminal neutrophil extracellular traps in atherosclerosis. Thrombosis and haemostasis. 2012;107:597-598
15. Maruchi Y, Tsuda M, Mori H, Takenaka N, Gocho T, Huq MA, Takeyama N. Plasma myeloperoxidase-conjugated DNA level predicts outcomes and organ dysfunction in patients with septic shock. Critical care. 2018;22:176
16. Wichapong K, Poelman H, Ercig B, Hrdinova J, Liu X, Lutgens E, Nicolaes GA. Rational modulator design by exploitation of protein-protein complex structures. Future medicinal chemistry. 2019;11 :1015-1033
17. Wichapong K, Alard JE, Ortega-Gomez A, Weber C, Hackeng TM, Soehnlein O, Nicolaes GA. Structure-based design of peptidic inhibitors of the interaction between cc chemokine ligand 5 (ccl5) and human neutrophil peptides 1 (hnp1). Journal of medicinal chemistry. 2016;59:4289-4301
18. Silvestre-Roig C, Braster Q, Wichapong K, Lee EY, Teulon JM, Berrebeh N, Winter J, Adrover JM, Santos GS, Froese A, Lemnitzer P, Ortega-Gomez A, Chevre R, Marschner J, Schumski A, Winter C, Perez-Olivares L, Pan C, Paulin N, Schoufour T, Hartwig H, Gonzalez-Ramos S, Kamp F, Megens RTA, Mowen KA, Gunzer M, Maegdefessel L, Hackeng T, Lutgens E, Daemen M, von Blume J, Anders HJ, Nikolaev VO, Pellequer JL, Weber C, Hidalgo A, Nicolaes GAF, Wong GCL, Soehnlein O. Externalized histone h4 orchestrates chronic inflammation by inducing lytic cell death. Nature. 2019;569:236-240
19. van Zundert GCP, Rodrigues J, Trellet M, Schmitz C, Kastritis PL, Karaca E, Melquiond ASJ, van Dijk M, de Vries SJ, Bonvin A. The haddock2.2 web server: User-friendly integrative modeling of biomolecular complexes. Journal of molecular biology. 2016;428:720-725
20. Clark SR, Ma AC, Tavener SA, McDonald B, Goodarzi Z, Kelly MM, Patel KD, Chakrabarti S, McAvoy E, Sinclair GD, Keys EM, Allen-Vercoe E, Devinney R, Doig CJ, Green FH, Kubes P. Platelet tlr4 activates neutrophil extracellular traps to ensnare bacteria in septic blood. Nature medicine. 2007;13:463-469
21. Cools-Lartigue J, Spicer J, McDonald B, Gowing S, Chow S, Giannias B, Bourdeau F, Kubes P, Ferri L. Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis. The Journal of clinical investigation. 2013
22. Najmeh S, Cools-Lartigue J, Rayes RF, Gowing S, Vourtzoumis P, Bourdeau F, Giannias B, Berube J, Rousseau S, Ferri LE, Spicer JD. Neutrophil extracellular traps sequester circulating tumor cells via betal-integrin mediated interactions. International journal of cancer. 2017;140:2321-2330
23. Monti M, lommelli F, De Rosa V, Carriero MV, Miceli R, Camerlingo R, Di Minno G, Del Vecchio S. Integrin-dependent cell adhesion to neutrophil extracellular traps through engagement of fibronectin in neutrophil-like cells. PloS one. 2017;12:e0171362
24. Monti M, De Rosa V, lommelli F, Carriero MV, Terlizzi C, Camerlingo R, Belli S, Fonti R, Di Minno G, Del Vecchio S. Neutrophil extracellular traps as an adhesion substrate for different tumor cells expressing rgd-binding integrins. International journal of molecular sciences. 2018;19 25. Wantha S, Alard JE, Megens RT, van der Does AM, Doring Y, Drechsler M, Pham CT, Wang MW, Wang JM, Gallo RL, von Hundelshausen P, Lindbom L, Hackeng T, Weber C, Soehnlein O. Neutrophil-derived cathelicidin promotes adhesion of classical monocytes. Circulation research. 2013;112:792-801
26. Henninot A, Collins JC, Nuss JM. The current state of peptide drug discovery: Back to the future? Journal of medicinal chemistry. 2018;61 :1382-1414
27. Corrales-Medina VF, Serpa J, Rueda AM, Giordano TP, Bozkurt B, Madjid M, Tweardy D, Musher DM. Acute bacterial pneumonia is associated with the occurrence of acute coronary syndromes. Medicine. 2009;88:154-159
28. Violi F, Cangemi R, Falcone M, Taliani G, Pieralli F, Vannucchi V, Nozzoli C, Venditti M, Chirinos JA, Corrales-Medina VF, Group SS. Cardiovascular complications and short-term mortality risk in community-acquired pneumonia. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2017;64:1486-1493
29. Delgado-Rizo V, Martinez-Guzman MA, Iniguez-Gutierrez L, Garcia- Orozco A, Alvarado-Navarro A, Fafutis-Morris M. Neutrophil extracellular traps and its implications in inflammation: An overview. Frontiers in immunology. 2017;8:81
30. Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation. 2010;122:1837-1845
31. Doring Y, Drechsler M, Wantha S, Kemmerich K, Lievens D, Vijayan S, Gallo RL, Weber C, Soehnlein O. Lack of neutrophil-derived cramp reduces atherosclerosis in mice. Circulation research. 2012;110:1052-1056
32. Yang LY, Luo Q, Lu L, Zhu WW, Sun HT, Wei R, Lin ZF, Wang XY, Wang CQ, Lu M, Jia HL, Chen JH, Zhang JB, Qin LX. Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response. Journal of hematology & oncology. 2020; 13:3
33. Tohme S, Yazdani HO, Al-Khafaji AB, Chidi AP, Loughran P, Mowen K, Wang Y, Simmons RL, Huang H, Tsung A. Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress. Cancer research. 2016;76:1367-1380
34. Sawabata N, Okumura M, Utsumi T, Inoue M, Shiono H, Minami M, Nishida T, Sawa Y. Circulating tumor cells in peripheral blood caused by surgical manipulation of non-small-cell lung cancer: Pilot study using an immunocytology method. General thoracic and cardiovascular surgery. 2007;55:189-192
35. Ridker PM, Everett BM, Thuren T, MacFadyen JG, Chang WH, Ballantyne C, Fonseca F, Nicolau J, Koenig W, Anker SD, Kastelein JJP, Cornel JH, Pais P, Pella D, Genest J, Cifkova R, Lorenzatti A, Forster T, Kobalava Z, Vida-Simiti L, Flather M, Shimokawa H, Ogawa H, Dellborg M, Rossi PRF, Troquay RPT, Libby P, Glynn RJ, Group CT. Antiinflammatory therapy with canakinumab for atherosclerotic disease. The New England journal of medicine. 2017;377: 1119-1131
36. Bosmann M, Grailer J J, Ruemmler R, Russkamp NF, Zetoune FS, Sarma JV, et al. Extracellular histones are essential effectors of C5aR-and C5L2-mediated tissue damage and inflammation in acute lung injury. FASEB J. 2013;27:5010-21.
37. Lv X, Wen T, Song J, Xie D, Wu L, Jiang X, et al. Extracellular histones are clinically relevant mediators in the pathogenesis of acute respiratory distress syndrome. Respir Res. 2017;18:1-9.
38. Huang H, Evankovich J, Yan W, Nace G, Zhang L, Ross M, et al. Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll like receptor 9 in mice. Hepatology. 2011 ;54:999-1008.
39. Allam R, Scherbaum CR, Darisipudi MN, Mulay SR, Hagele H, Lichtnekert J, et al. Histones from Dying Renal Cells Aggravate Kidney Injury via TLR2 and TLR4. J. Am. Soc. Nephrol. 2012. p. 1375-88.
40. Kessenbrock K, Krumbholz M, Schonermarck U, Back W, Wolfgang L, Werb Z, et al. Netting neutrophils in autoimmune small-vessel vasculitis. Nat Med. 2009; 15:623- 5.
41. De Meyer SF, Suidan GL, Fuchs TA, Monestier M, Wagner DD. Extracellular chromatin is an important mediator of ischemic stroke in mice. Arterioscler Thromb Vase Biol. 2012;32:1884-91.
42. Zhang Y, Wen Z, Guan L, Jiang P, Gu T, Zhao J, Lv X WT. Extracellular Histones Play an Inflammatory Role in Acid Aspiration-induced Acute Respiratory Distress Syndrome. Anesthesiology. 2015;122:127-39.
43. Hai Huang, Hui-Wei Chen, John Evankovich, Wei Yan, Brian R. Rosborough, Gary W. Nace, Qing Ding, Patricia Loughran, Donna Beer-Stolz, Timothy R. Billiar, Charles T. Esmon and AT. Histones activate the NLRP3 Inflammasome in Kupffer Cells during Sterile Inflammatory Liver Injury. J Immunol. 2013;191 :2655-79. 44. Kumar SV, Kulkarni OP, Mulay SR, Darisipudi MN, Romoli S, Thomasova D, et al. Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN. J Am Soc Nephrol. 2015;26:2399-413.
45. Huang H, Tohme S, Al-Khafaji AB, Tai S, Loughran P, Chen L, et al. Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury. Hepatology. 2015;62:600-14.
46. Wen Z, Lei Z, Yao L, Jiang P, Gu T, Ren F, et al. Circulating histones are major mediators of systemic inflammation and cellular injury in patients with acute liver failure. Cell Death Dis. 2016;7.
47. Kutcher M, Xu J, Vilardi R, Ho C, Esmon CM, Cohen MJ. Extracellular histone release in response to traumatic injury: implications for a compensatory role of activated Protein C. J Trauma Acute Care Surg. 2012;73:1-14.
48. Wildhagen KC, Wiewel MA, Schultz MJ, Horn J, Schrijver R, Reutelingsperger CP, et al. Extracellular histone H3 levels are inversely correlated with antithrombin levels and platelet counts and are associated with mortality in sepsis patients. Thromb Res. 2015;136:542-7.
49. Ekaney ML, Otto GP, Sossdorf M, Sponholz C, Boehringer M, Loesche W, et al. Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation. Crit Care. 2014;18:1-9.
50. Kawai C, Kotani H, Miyao M, Ishida T, Jemail L, Abiru H, et al. Circulating extracellular histones are clinically relevant mediators of multiple organ injury. Am J Pathol. 2016;186:829-43.
51. Jin, Y. et.al. Extracellular histones aggravate acute respiratory distress syndrome by inducing peripheral blood mononuclear cells pyroptosis, (2019);31 (11 ): 1357-1362
52. Ward, P.A., et.al., Acute lung injury and the role of histones, Translational respiratory medicine (2014) ;2: 1
53. Lafrancais, E. et.al., Neutralizing extracellular histones in acute respiratory distress syndrome, American journal of respiratory and critical care medicine (2017); 196(2): 122-123.

Claims

1. Peptide comprising an amino acid sequence
H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or an equivalent thereof that differs from SEQ ID NO: 1 by not more than 3 amino acids.
2. Peptide according to claim 1 consisting of the amino acid sequence H-CEPLSEVEDYLDSSLKYNAKDTINYC-OH (SEQ ID NO: 1) or an equivalent thereof that differs from SEQ ID NO: 1 by not more than 3 amino acids.
3. Peptide according to claim 1 that is circular.
4. Peptide according to any one of claims 1 - 3 for use in the treatment of a disease.
5. Peptide for use according to claim 4 wherein the disease is a NETs- related disease.
6. Peptide for use according to claim 5 wherein the NETs-related disease is selected from the group consisting of atherosclerosis, autoimmune disorders, stroke, transfusion-related)acute lung injury, lung injury, ischaemia/reperfusion injury (IRI), kidney injury, liver injury, trauma, sepsis and inflammation.
PCT/EP2021/059588 2020-04-14 2021-04-13 Peptide for the treatment of net-associated diseases WO2021209465A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP20169440 2020-04-14
EP20169440.3 2020-04-14

Publications (1)

Publication Number Publication Date
WO2021209465A1 true WO2021209465A1 (en) 2021-10-21

Family

ID=70289579

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2021/059588 WO2021209465A1 (en) 2020-04-14 2021-04-13 Peptide for the treatment of net-associated diseases

Country Status (1)

Country Link
WO (1) WO2021209465A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3205347A1 (en) * 2014-10-08 2017-08-16 Keio University White blood cell extracellular trap formation inhibitor
WO2020038963A1 (en) * 2018-08-21 2020-02-27 Modiquest B.V. Antibodies binding to citrullinated histone 2a and/or 4

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3205347A1 (en) * 2014-10-08 2017-08-16 Keio University White blood cell extracellular trap formation inhibitor
WO2020038963A1 (en) * 2018-08-21 2020-02-27 Modiquest B.V. Antibodies binding to citrullinated histone 2a and/or 4

Non-Patent Citations (57)

* Cited by examiner, † Cited by third party
Title
ALARD JEORTEGA-GOMEZ AWICHAPONG KBONGIOVANNI DHORCKMANS MMEGENS RTLEONI GFERRARO BROSSAINT JPAULIN N: "Recruitment of classical monocytes can be inhibited by disturbing heteromers of neutrophil hnp1 and platelet ccl5", SCIENCE TRANSLATIONAL MEDICINE, vol. 7, 2015, pages 317ra196, XP055247245, DOI: 10.1126/scitranslmed.aad5330
ALLAM RSCHERBAUM CRDARISIPUDI MNMULAY SRHAGELE HLICHTNEKERT J ET AL.: "Histones from Dying Renal Cells Aggravate Kidney Injury via TLR2 and TLR4", J. AM. SOC. NEPHROL., 2012, pages 1375 - 88, XP055314668, DOI: 10.1681/ASN.2011111077
BOSMANN MGRAILER JJRUEMMLER RRUSSKAMP NFZETOUNE FSSARMA JV ET AL.: "Extracellular histones are essential effectors of C5aR-and C5L2-mediated tissue damage and inflammation in acute lung injury", FASEB J, vol. 27, 2013, pages 5010 - 21, XP055288681, DOI: 10.1096/fj.13-236380
CLARK SRMA ACTAVENER SAMCDONALD BGOODARZI ZKELLY MMPATEL KDCHAKRABARTI SMCAVOY ESINCLAIR GD: "Platelet tlr4 activates neutrophil extracellular traps to ensnare bacteria in septic blood", NATURE MEDICINE, vol. 13, 2007, pages 463 - 469, XP009111444, DOI: 10.1038/nm1565
COOLS-LARTIGUE JSPICER JMCDONALD BGOWING SCHOW SGIANNIAS BBOURDEAU FKUBES PFERRI L: "Neutrophil extracellular traps sequester circulating tumor cells and promote metastasis", THE JOURNAL OF CLINICAL INVESTIGATION, 2013
CORRALES-MEDINA VFALVAREZ KNWEISSFELD LAANGUS DCCHIRINOS JACHANG CCNEWMAN ALOEHR LFOLSOM ARELKIND MS: "Association between hospitalization for pneumonia and subsequent risk of cardiovascular disease", JAMA, vol. 313, 2015, pages 264 - 274
CORRALES-MEDINA VFMUSHER DMWELLS GACHIRINOS JACHEN LFINE MJ: "Cardiac complications in patients with community-acquired pneumonia: Incidence, timing, risk factors, and association with short-term mortality", CIRCULATION, vol. 125, 2012, pages 773 - 781
CORRALES-MEDINA VFSERPA JRUEDA AMGIORDANO TPBOZKURT BMADJID MTWEARDY DMUSHER DM: "Acute bacterial pneumonia is associated with the occurrence of acute coronary syndromes", MEDICINE, vol. 88, 2009, pages 154 - 159
DALAGER-PEDERSEN MSOGAARD MSCHONHEYDER HCNIELSEN HTHOMSEN RW: "Risk for myocardial infarction and stroke after community-acquired bacteremia: A 20-year population-based cohort study", CIRCULATION, vol. 129, 2014, pages 1387 - 1396
DE MEYER SFSUIDAN GLFUCHS TAMONESTIER MWAGNER DD: "Extracellular chromatin is an important mediator of ischemic stroke in mice", ARTERIOSCLER THROMB VASC BIOL, vol. 32, 2012, pages 1884 - 91
DELGADO-RIZO VMARTINEZ-GUZMAN MAINIGUEZ-GUTIERREZ LGARCIA-OROZCO AALVARADO-NAVARRO AFAFUTIS-MORRIS M: "Neutrophil extracellular traps and its implications in inflammation: An overview", FRONTIERS IN IMMUNOLOGY, vol. 8, 2017, pages 81
DORING YDRECHSLER MWANTHA SKEMMERICH KLIEVENS DVIJAYAN SGALLO RLWEBER CSOEHNLEIN O: "Lack of neutrophil-derived cramp reduces atherosclerosis in mice", CIRCULATION RESEARCH, vol. 110, 2012, pages 1052 - 1056
DRECHSLER MMEGENS RTVAN ZANDVOORT MWEBER CSOEHNLEIN O: "Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis", CIRCULATION, vol. 122, 2010, pages 1837 - 1845
EKANEY MLOTTO GPSOSSDORF MSPONHOLZ CBOEHRINGER MLOESCHE W ET AL.: "Impact of plasma histones in human sepsis and their contribution to cellular injury and inflammation", CRIT CARE, vol. 18, 2014, pages 1 - 9
FAYAD ZASWIRSKI FKCALCAGNO CROBBINS CSMULDER WKOVACIC JC: "Monocyte and macrophage dynamics in the cardiovascular system: Jacc macrophage in cvd series (part 3", JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY, vol. 72, 2018, pages 2198 - 2212, XP085510675, DOI: 10.1016/j.jacc.2018.08.2150
HAI HUANGHUI-WEI CHENJOHN EVANKOVICHWEI YANBRIAN RROSBOROUGH, GARY WNACE, QING DINGPATRICIA LOUGHRANDONNA BEER-STOLZTIMOTHY R. BIL: "activate the NLRP3 Inflammasome in Kupffer Cells during Sterile Inflammatory Liver Injury", J IMMUNOL, vol. 191, 2013, pages 2655 - 79
HENNINOT ACOLLINS JCNUSS JM: "The current state of peptide drug discovery: Back to the future?", JOURNAL OF MEDICINAL CHEMISTRY, vol. 61, 2018, pages 1382 - 1414
HUANG HEVANKOVICH JYAN WNACE GZHANG LROSS M ET AL.: "Endogenous histones function as alarmins in sterile inflammatory liver injury through Toll-like receptor 9 in mice", HEPATOLOGY, vol. 54, 2011, pages 999 - 1008, XP055194650, DOI: 10.1002/hep.24501
HUANG HTOHME SAI-KHAFAJI ABTAI SLOUGHRAN PCHEN L ET AL.: "Damage-associated molecular pattern-activated neutrophil extracellular trap exacerbates sterile inflammatory liver injury", HEPATOLOGY, vol. 62, 2015, pages 600 - 14
JIN, Y, EXTRACELLULAR HISTONES AGGRAVATE ACUTE RESPIRATORY DISTRESS SYNDROME BY INDUCING PERIPHERAL BLOOD MONONUCLEAR CELLS PYROPTOSIS, vol. 31, no. 11, 2019, pages 1357 - 1362
KAWAI CKOTANI HMIYAO MISHIDA TJEMAIL LABIRU H ET AL.: "Circulating extracellular histones are clinically relevant mediators of multiple organ injury", AM J PATHOL, vol. 186, 2016, pages 829 - 43
KESSENBROCK KKRUMBHOLZ MSCHONERMARCK UBACK WWOLFGANG LWERB Z ET AL.: "Netting neutrophils in autoimmune small-vessel vasculitis", NAT MED, vol. 15, 2009, pages 623 - 5, XP055466319, DOI: 10.1038/nm.1959
KUMAR SVKULKARNI OPMULAY SRDARISIPUDI MNROMOLI STHOMASOVA D ET AL.: "Neutrophil Extracellular Trap-Related Extracellular Histones Cause Vascular Necrosis in Severe GN", J AM SOC NEPHROL, vol. 26, 2015, pages 2399 - 413
KUTCHER MXU JVILARDI RHO CESMON CMCOHEN MJ: "Extracellular histone release in response to traumatic injury: implications for a compensatory role of activated Protein C", J TRAUMA ACUTE CARE SURG, vol. 73, 2012, pages 1 - 14
LAFRANCAIS, E.: "Neutralizing extracellular histones in acute respiratory distress syndrome", AMERICAN JOURNAL OF RESPIRATORY AND CRITICAL CARE MEDICINE, vol. 196, no. 2, 2017, pages 122 - 123
LV XWEN TSONG JXIE DWU LJIANG X ET AL.: "Extracellular histones are clinically relevant mediators in the pathogenesis of acute respiratory distress syndrome", RESPIR RES, vol. 18, 2017, pages 1 - 9, XP055797959, DOI: 10.1186/s12931-017-0651-5
MARUCHI YTSUDA MMORI HTAKENAKA NGOCHO THUQ MATAKEYAMA N: "Plasma myeloperoxidase-conjugated DNA level predicts outcomes and organ dysfunction in patients with septic shock", CRITICAL CARE, vol. 22, 2018, pages 176
MEGENS RTVIJAYAN SLIEVENS DDORING YVAN ZANDVOORT MAGROMMES JWEBER CSOEHNLEIN O: "Presence of luminal neutrophil extracellular traps in atherosclerosis", THROMBOSIS AND HAEMOSTASIS, vol. 2012, no. 107, pages 597 - 598
MONTI MDE ROSA VLOMMELLI FCARRIERO MVTERLIZZI CCAMERLINGO RBELLI SFONTI RDI MINNO GDEL VECCHIO S: "Neutrophil extracellular traps as an adhesion substrate for different tumor cells expressing rgd-binding integrins", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 19, 2018
MONTI MLOMMELLI FDE ROSA VCARRIERO MVMICELI RCAMERLINGO RDI MINNO GDEL VECCHIO S: "Integrin-dependent cell adhesion to neutrophil extracellular traps through engagement of fibronectin in neutrophil-like cells", PLOS ONE, vol. 12, 2017, pages e0171362
MUSHER DMABERS MSCORRALES-MEDINA VF: "Acute infection and myocardial infarction", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 380, 2019, pages 171 - 176
NAJMEH SCOOLS-LARTIGUE JRAYES RFGOWING SVOURTZOUMIS PBOURDEAU FGIANNIAS BBERUBE JROUSSEAU SFERRI LE: "Neutrophil extracellular traps sequester circulating tumor cells via beta1-integrin mediated interactions", INTERNATIONAL JOURNAL OF CANCER, vol. 140, 2017, pages 2321 - 2330
ORTEGA-GOMEZ ASALVERMOSER MROSSAINT JPICK RBRAUNER JLEMNITZER PTILGNER JDE JONG RJMEGENS RTJAMASBI J: "Cathepsin g controls arterial but not venular myeloid cell recruitment", CIRCULATION, vol. 134, 2016, pages 1176 - 1188
PARK JWYSOCKI RWAMOOZGAR ZMAIORINO LFEIN MRJORNS JSCHOTT AFKINUGASA-KATAYAMA YLEE YWON NH: "Cancer cells induce metastasis-supporting neutrophil extracellular DNA traps", SCIENCE TRANSLATIONAL MEDICINE, vol. 8, 2016, pages 361 - 138
PIETERSE EROTHER NYANGINLAR CHILBRANDS LBVAN DER VLAG J: "Neutrophils discriminate between lipopolysaccharides of different bacterial sources and selectively release neutrophil extracellular traps", FRONTIERS IN IMMUNOLOGY, vol. 2016, no. 7, pages 484
RAMIREZ JALIBERTI SMIRSAEIDI MPEYRANI PFILARDO GAMIR AMOFFETT BGORDON JBLASI FBORDON J: "Acute myocardial infarction in hospitalized patients with community-acquired pneumonia", CLINICAL INFECTIOUS DISEASES : AN OFFICIAL PUBLICATION OF THE INFECTIOUS DISEASES SOCIETY OF AMERICA, vol. 47, 2008, pages 182 - 187, XP009174237, DOI: 10.1086/589246
RIDKER PMEVERETT BMTHUREN TMACFADYEN JGCHANG WHBALLANTYNE CFONSECA FNICOLAU JKOENIG WANKER SD: "Antiinflammatory therapy with canakinumab for atherosclerotic disease", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 377, 2017, pages 1119 - 1131, XP055524405, DOI: 10.1056/NEJMoa1707914
SAWABATA NOKUMURA MUTSUMI TINOUE MSHIONO HMINAMI MNISHIDA TSAWA Y: "Circulating tumor cells in peripheral blood caused by surgical manipulation of non-small-cell lung cancer: Pilot study using an immunocytology method", GENERAL THORACIC AND CARDIOVASCULAR SURGERY, vol. 55, 2007, pages 189 - 192
SCHUMSKI ARIANE ET AL: "Endotoxinemia Accelerates Atherosclerosis Through Electrostatic Charge-Mediated Monocyte Adhesion", CIRCULATION, vol. 143, no. 3, 10 November 2020 (2020-11-10), US, pages 254 - 266, XP055815015, ISSN: 0009-7322, Retrieved from the Internet <URL:http://dx.doi.org/10.1161/CIRCULATIONAHA.120.046677> DOI: 10.1161/CIRCULATIONAHA.120.046677 *
SILVESTRE-ROIG CARLOS ET AL: "Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death", NATURE, MACMILLAN JOURNALS LTD, LONDON, vol. 569, no. 7755, 1 May 2019 (2019-05-01), pages 236 - 240, XP036900459, ISSN: 0028-0836, [retrieved on 20190501], DOI: 10.1038/S41586-019-1167-6 *
SILVESTRE-ROIG CBRASTER QWICHAPONG KLEE EYTEULON JMBERREBEH NWINTER JADROVER JMSANTOS GSFROESE A: "Externalized histone h4 orchestrates chronic inflammation by inducing lytic cell death", NATURE, vol. 569, 2019, pages 236 - 240, XP036900459, DOI: 10.1038/s41586-019-1167-6
SMEETH LTHOMAS SLHALL AJHUBBARD RFARRINGTON PVALLANCE P: "Risk of myocardial infarction and stroke after acute infection or vaccination", THE NEW ENGLAND JOURNAL OF MEDICINE, vol. 351, 2004, pages 2611 - 2618
TOHME SYAZDANI HOAL-KHAFAJI ABCHIDI APLOUGHRAN PMOWEN KWANG YSIMMONS RLHUANG HTSUNG A: "Neutrophil extracellular traps promote the development and progression of liver metastases after surgical stress", CANCER RESEARCH, vol. 76, 2016, pages 1367 - 1380
VAN ZUNDERT GCPRODRIGUES JTRELLET MSCHMITZ CKASTRITIS PLKARACA EMELQUIOND ASJVAN DIJK MDE VRIES SJBONVIN A: "The haddock2.2 web server: User-friendly integrative modeling of biomolecular complexes", JOURNAL OF MOLECULAR BIOLOGY, vol. 428, 2016, pages 720 - 725, XP029452202, DOI: 10.1016/j.jmb.2015.09.014
VIOLI FCANGEMI RFALCONE MTALIANI GPIERALLI FVANNUCCHI VNOZZOLI CVENDITTI MCHIRINOS JACORRALES-MEDINA VF: "Cardiovascular complications and short-term mortality risk in community-acquired pneumonia", CLINICAL INFECTIOUS DISEASES : AN OFFICIAL PUBLICATION OF THE INFECTIOUS DISEASES SOCIETY OF AMERICA, vol. 64, 2017, pages 1486 - 1493
VOLKER BRINKMANN AND ARTURO ZYCHLINSKY: "Beneficial suicide: why neutrophils die to make NETs", PROGRESS, 1 January 2007 (2007-01-01), XP055701064, Retrieved from the Internet <URL:https://www.nature.com/articles/nrmicro1710.pdf> [retrieved on 20200604] *
WANTHA SALARD JEMEGENS RTVAN DER DOES AMDORING YDRECHSLER MPHAM CTWANG MWWANG JMGALLO RL: "Neutrophil-derived cathelicidin promotes adhesion of classical monocytes", CIRCULATION RESEARCH, vol. 112, 2013, pages 792 - 801
WARD, P.A.: "Acute lung injury and the role of histones", TRANSLATIONAL RESPIRATORY MEDICINE, vol. 2, 2014, pages 1
WEN ZLEI ZYAO LJIANG PGU TREN F ET AL.: "Circulating histones are major mediators of systemic inflammation and cellular injury in patients with acute liver failure", CELL DEATH DIS, vol. 7, 2016, XP055682669, DOI: 10.1038/cddis.2016.303
WICHAPONG KALARD JEORTEGA-GOMEZ AWEBER CHACKENG TMSOEHNLEIN ONICOLAES GA: "Structure-based design of peptidic inhibitors of the interaction between cc chemokine ligand 5 (ccl5) and human neutrophil peptides 1 (hnp1", JOURNAL OF MEDICINAL CHEMISTRY, vol. 59, 2016, pages 4289 - 4301, XP055348304, DOI: 10.1021/acs.jmedchem.5b01952
WICHAPONG KANIN ET AL: "Structure-based peptide design targeting intrinsically disordered proteins: Novel histone H4 and H2A peptidic inhibitors", COMPUTATIONAL AND STRUCTURAL BIOTECHNOLOGY JOURNAL, vol. 19, 21 January 2021 (2021-01-21), Sweden, pages 934 - 948, XP055815009, ISSN: 2001-0370, DOI: 10.1016/j.csbj.2021.01.026 *
WICHAPONG KPOELMAN HERCIG BHRDINOVA JLIU XLUTGENS ENICOLAES GA: "Rational modulator design by exploitation of protein-protein complex structures", FUTURE MEDICINAL CHEMISTRY, vol. 11, 2019, pages 1015 - 1033
WILDHAGEN KCWIEWEL MASCHULTZ MJHORN JSCHRIJVER RREUTELINGSPERGER CP ET AL.: "Extracellular histone H3 levels are inversely correlated with antithrombin levels and platelet counts and are associated with mortality in sepsis patients", THROMB RES, vol. 136, 2015, pages 542 - 7, XP055312885, DOI: 10.1016/j.thromres.2015.06.035
WINTER CSILVESTRE-ROIG CORTEGA-GOMEZ ALEMNITZER PPOELMAN HSCHUMSKI AWINTER JDRECHSLER MDE JONG RIMMLER R: "Chrono-pharmacological targeting of the ccl2-ccr2 axis ameliorates atherosclerosis", CELL METABOLISM, vol. 28, 2018, pages 175 - 182,e175
YANG LYLUO QLU LZHU WWSUN HTWEI RLIN ZFWANG XYWANG CQLU M: "Increased neutrophil extracellular traps promote metastasis potential of hepatocellular carcinoma via provoking tumorous inflammatory response", JOURNAL OF HEMATOLOGY & ONCOLOGY, vol. 13, 2020, pages 3
ZHANG JALCAIDE PLIU LSUN JHE A, LUSCINSKAS FWSHI GP.: "Regulation of endothelial cell adhesion molecule expression by mast cells, macrophages, and neutrophils", PLOS ONE, vol. 6, 2011, pages e14525
ZHANG YWEN ZGUAN LJIANG PGU TZHAO JLV X WT: "Extracellular Histones Play an Inflammatory Role in Acid Aspiration-induced Acute Respiratory Distress Syndrome", ANESTHESIOLOGY, vol. 122, 2015, pages 127 - 39

Similar Documents

Publication Publication Date Title
Kanki et al. Identification of targeting peptides for ischemic myocardium by in vivo phage display
EP2162462B1 (en) A cxc chemokine receptor 4 (cxcr4) antagonistic polypeptide
EP1768677B1 (en) Nucleic acids for the treatment of hmgb1-related pathologies
Janssens et al. Truncation of CXCL12 by CD26 reduces its CXC chemokine receptor 4-and atypical chemokine receptor 3-dependent activity on endothelial cells and lymphocytes
Wahid et al. High-mobility group box 1 serves as an inflammation driver of cardiovascular disease
Quagliariello et al. Interleukin-1 blocking agents as promising strategy for prevention of anticancer drug-induced cardiotoxicities: possible implications in cancer patients with COVID-19.
US20150010552A1 (en) Compositions and methods for treating inflammation and fibrosis
CA2793838A1 (en) Integrin interaction inhibitors for the treatment of cancer
Pesapane et al. Recent advances in the function of the 67 kDa laminin receptor and its targeting for personalized therapy in cancer
Rastelli et al. A KDR-binding peptide (ST100, 059) can block angiogenesis, melanoma tumor growth and metastasis in vitro and in vivo
Noda et al. Neuropathic pain inhibitor, RAP-103, is a potent inhibitor of microglial CCL1/CCR8
US20170096478A1 (en) Therapeutic agent for inflammatory disease
WO2021209465A1 (en) Peptide for the treatment of net-associated diseases
EP3328426B1 (en) Treatment of patients diagnosed with pancreatic ductal adenocarcinoma using using monoclonal antibodies against the epidermal growth factor receptor (egfr)
WO2021011480A1 (en) Compositions and methods to block and bind cxcr4 to modulate cellular function
Shen et al. C‑X‑C motif chemokine ligand 8 promotes endothelial cell homing via the Akt‑signal transducer and activator of transcription pathway to accelerate healing of ischemic and hypoxic skin ulcers
EP2866832A2 (en) Uses of cxcl17, a novel chemokine marker of human lung and gastrointestinal disease
JP6566947B2 (en) MAP kinase P38 binding compound
Dai et al. TfR binding peptide screened by phage display technology-characterization to target cancer cells
Yang et al. Novel TLR4-antagonizing peptides inhibit LPS-induced release of inflammatory mediators by monocytes
CN110357946B (en) Polypeptide for inhibiting tumor metastasis and application thereof
EP3836951A1 (en) Peptides and compositions for targeted treatment and imaging
Yu et al. Mouse thymus targeted peptide isolated by in vivo phage display can inhibit bioactivity of thymus output in vivo
Liu Cardiomyocytes Deficiency of SENP1 Promotes Myocardial Fibrosis via Paracrine Action
Satala et al. Candida parapsilosis cell wall proteins—CPAR2_404800 and CPAR2_404780—Are adhesins that bind to human epithelial and endothelial cells and extracellular matrix proteins

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21717463

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21717463

Country of ref document: EP

Kind code of ref document: A1