Next Article in Journal
Therapeutic Prospects of Mesenchymal Stem Cell and Their Derived Exosomes in the Regulation of the Gut Microbiota in Inflammatory Bowel Disease
Previous Article in Journal
Influence of Gut Microbiota-Mediated Immune Regulation on Response to Chemotherapy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Neutrophil Extracellular Trap Formation: Exploring Promising Pharmacological Strategies for the Treatment of Preeclampsia

by
Leticia Lorena Hernández González
1,2,†,
Laura Pérez-Campos Mayoral
3,†,
María Teresa Hernández-Huerta
4,
Gabriel Mayoral Andrade
3,
Margarito Martínez Cruz
1,
Edgar Ramos-Martínez
5,
Eduardo Pérez-Campos Mayoral
3,
Víctor Cruz Hernández
6,
Ismael Antonio García
6,
Carlos Alberto Matias-Cervantes
4,
Miriam Emily Avendaño Villegas
1,
Carlos Mauricio Lastre Domínguez
1,
Carlos Romero Díaz
1,3,
Juan de Dios Ruiz-Rosado
7,8,*,‡ and
Eduardo Pérez-Campos
1,9,*,‡
1
National Technology of Mexico/IT Oaxaca, Oaxaca de Juárez, Oaxaca 68030, Mexico
2
Faculty of Biological Systems and Technological Innovation, Autonomous University “Benito Juárez” of Oaxaca, Oaxaca 68125, Mexico
3
Research Center, Faculty of Medicine UNAM-UABJO, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico
4
CONAHCyT, Faculty of Medicine and Surgery, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico
5
School of Sciences, Autonomous University “Benito Juárez” of Oaxaca (UABJO), Oaxaca 68020, Mexico
6
“Dr Aurelio Valdivieso” Hospital General. SS., Oaxaca 68040, Mexico
7
Kidney and Urinary Tract Research Center, Abigail Wexner Research Institute, Nationwide Children’s Hospital, Columbus, OH 43215, USA
8
Division of Nephrology and Hypertension, Nationwide Children’s Hospital, Columbus, OH 43205, USA
9
Clinical Pathology Laboratory, “Eduardo Pérez Ortega”, Oaxaca 68000, Mexico
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work and shared first authorship.
These authors contributed equally to this work and share senior authorship.
Pharmaceuticals 2024, 17(5), 605; https://doi.org/10.3390/ph17050605
Submission received: 23 April 2024 / Accepted: 4 May 2024 / Published: 9 May 2024
(This article belongs to the Section Pharmacology)

Abstract

:
Neutrophils, which constitute the most abundant leukocytes in human blood, emerge as crucial players in the induction of endothelial cell death and the modulation of endothelial cell responses under both physiological and pathological conditions. The hallmark of preeclampsia is endothelial dysfunction induced by systemic inflammation, in which neutrophils, particularly through the formation of neutrophil extracellular traps (NETs), play a pivotal role in the development and perpetuation of endothelial dysfunction and the hypertensive state. Considering the potential of numerous pharmaceutical agents to attenuate NET formation (NETosis) in preeclampsia, a comprehensive assessment of the extensively studied candidates becomes imperative. This review aims to identify mechanisms associated with the induction and negative regulation of NETs in the context of preeclampsia. We discuss potential drugs to modulate NETosis, such as NF-κβ inhibitors, vitamin D, and aspirin, and their association with mutagenicity and genotoxicity. Strong evidence supports the notion that molecules involved in the activation of NETs could serve as promising targets for the treatment of preeclampsia.

1. Introduction

Preeclampsia (PE) is a multifactorial and multisystem syndrome specific to human pregnancy [1]. It manifests as hypertension that emerges at 20 weeks of gestation in previously normotensive women [2,3].
PE is categorized into subtypes, including early and late-onset, as well as severe, mild, and mild to severe [4]. From a haemodynamic perspective, based on the value of cardiac output (CO) at rest, PE is divided into two types, hypo- or hypervolemic. The hypovolemic type also denoted as “classic” or placental early onset, is characterized by placental hypertension induced by vasoconstriction [5]. It presents inadequate placental perfusion, endothelial damage, placental insufficiency, and elevated levels of anti-angiogenic molecules such as soluble fms-like tyrosine kinase 1 (sFlt-1) [6] and soluble endoglin (sEng). The hypervolemic, maternal, or term type is more prevalent. This variant is associated with elevated CO, increased oedema due to water retention, augmented placental perfusion, higher foetal birth weight, relaxed vasculature, and maternal obesity [7].
Severe preeclampsia can lead to the development of HELLP syndrome (Haemolysis, Elevated Liver enzyme, and Low Platelets). This syndrome is characterised by microangiopathic haemolytic anaemia, hypertension, proteinuria, oedema, liver dysfunction, thrombocytopenia [8], increased CO, and inotropy [9].
Furthermore, endothelial cells, platelets, and neutrophils play pivotal roles in the inflammatory, hypertensive, and prothrombotic processes observed in preeclampsia [10,11,12]. Although distinct neutrophil subpopulations have been characterized in different human pathologies and experimental models [13,14,15,16,17,18], their specific roles in preeclampsia remain under investigation. Numerous groups of molecules orchestrate the relationships between neutrophils, platelets, and endothelial cells, such as the P-selectin-PSGL-1 axis [19] via phosphodiesterase type-4 or Src family kinases and the vWF-glycoprotein Ibα axis [20].
Studies have demonstrated alterations in the quantity and functionality of phagocytosis, degranulation, extracellular vesicle release, and the release of neutrophil extracellular traps (NETs) in preeclampsia. These changes are intricately linked to the systemic inflammatory and immune state. Furthermore, there is compelling evidence indicating increased elastase [21] and nucleosomes [22] release, particularly in early-onset PE. These findings suggest that alterations in neutrophil functions, such as NETosis, may contribute to inflammation and endothelial dysfunction, thereby promoting the development of PE.
In this review, we discuss the role of neutrophil activation and NETosis in the context of preeclampsia. Additionally, we examined pharmacological compounds that exhibit a negative regulatory influence on NETs, which could potentially be employed in the treatment of preeclampsia. Additionally, we explored the mutagenicity and genotoxicity associated with the use of these compounds.
To complete this review, we conducted a focused search and discussed the selection of drugs for analysis with a panel of experts using a Delphi exercise. We searched on PubMed, Google Scholar, and the Cochrane Library from 1 January 2023 to 31 January 2024, using the search terms “preeclampsia”, “neutrophils”, “neutrophil extracellular traps”, “NETs”, “NETosis”, “preeclampsia and oxidative stress”, and “preeclampsia and NF-κβ inhibitors”; we also cross-referenced these terms with the following: “aspirin”, “acetylsalicylic acid”, “dexamethasone”, “glucocorticoids”, “resveratrol”, “cyclosporine A”, “azithromycin”, “chloramphenicol”, “metformin”, “hydroxychloroquine”, “heparin”, “vitamin D”, “disulfiram”, “curcumin”, “roflumilast, “apremilast”, “rolipram, “Glycyrrhiza glabra”, “activated protein C”, and “recombinant human DNase I”.

2. Activation of Neutrophils in PE

Neutrophil activation in preeclampsia is characterized by distinct molecular and functional changes. An analysis of the expression of 40 inflammation-related genes in leukocytes has revealed a significant increase in the mRNA expression of the nuclear factor of Kappa light chain (NFKβ 1A), Cyclin-dependent kinase inhibitor 1A (CDKN-1A), Interleukin-8 (IL)-8, and IL-1b genes in leukocytes of pregnant women with PE compared to their healthy counterparts. This heightened expression favours the polarization of neutrophils towards an inflammatory profile [23]. Additionally, peripheral blood neutrophils in PE show lower expression of apoptosis markers compared to neutrophils from healthy pregnant women. Higher expression of these markers of apoptosis was observed in neutrophils from non-pregnant women, suggesting that delayed neutrophil apoptosis contributes to complications during pregnancy, such as PE [24,25].
In PE, neutrophil granule release and ROS production cause direct damage to the cell membrane through peroxidation of membrane lipids, leading to cell death [26]. This phenomenon triggers endothelial activation characterized by an increased expression of vascular cell adhesion molecule-1 (VCAM-1) and inactivation of factors such as endothelium-derived relaxing and prostacyclin (PGI2). In addition, there is an increase in the number of activated neutrophils, which is associated with a programmed neutrophil extracellular traps formation (NETosis) [27,28,29].
Neutrophils from pregnant women with PE exhibit increased expression of the adhesion molecule CD11b, which serves as a marker of cell activation [30]. Furthermore, there is a notable reduction in the surface expression of L-selectin (CD62L) on CD15+ neutrophils, which correlates with immune activation in PE [31]. Studies have shown that NETs can induce platelet activation and a prothrombotic state during PE [28,32]. This body of evidence suggests that neutrophils are activated and potentially primed for NET release in PE, both in the microcirculation and periphery, contributing to a feedback loop involving inflammation and endothelial dysfunction.

3. Neutrophils in the Uteroplacental Microvasculature in PE

In the uteroplacental microvasculature of pregnant women with PE, there is a marked infiltration of neutrophils associated with increased expression of the cell adhesion molecule ICAM-1 and a positive gradient of IL-8. In addition, expression of the β-2 integrins CD11a, CD11b, and CD11c is increased in neutrophils from women with PE compared to their healthy counterparts [33]. This heightened presence of neutrophils and associated molecules in the maternal tissue vasculature can cause vasoconstriction, ischemia, oxidative stress, inflammation, and subsequent endothelial dysfunction [34].
Analyses of biopsies obtained by caesarean section in pregnant women with PE have shown that the number of neutrophils attached to the vasculature is elevated [35,36]. A higher concentration of neutrophils in the vasculature potentially contributes to endothelial dysfunction [37].
The syncytiotrophoblast, the membrane layer that covers the placenta and is in direct contact with maternal blood, releases microvesicles (placental syncytiotrophoblast microvesicles, STBMs) [38] that can stimulate ROS production in neutrophils [39]. Syncytiotrophoblast cells and serum from pregnant women with PE contain higher levels of IL-32β compared to healthy pregnant women. IL-32 exerts multiple functions such as NET induction [40].

4. NETs

NETs are structures consisting of nuclear or mitochondrial DNA with proteins such as lactoferrin, actin, histones, neutrophil elastase (NE), and myeloperoxidase (MPO) [41]. The main function of NETs is the containment and elimination of pathogens [42,43]; however, their participation in thrombosis, autoimmune diseases, cancer, diabetes [44], and cardiovascular diseases such as hypertension [45] has also been reported.
NETosis can be triggered by various stimuli, including microbial pathogens, inflammatory cytokines, and chemical agents. The formation of NETs relies on histone citrullination, catalysed by elevated levels of neutrophil peptidyl-arginine deaminase-4 (PAD4). Additionally, NET formation depends on the production of ROS and activation of MPO and NE [46], leading to nucleosome histone digestion, chromatin decondensation, and release of DNA and antimicrobial molecules [47].
There are several signalling pathways involved in NET formation [48]; one pathway involves the generation of ROS through the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. Activation of the NADPH oxidase complex 2 (NOX2) triggers the Raf/MEK/ERK pathway and augments cytosolic ROS levels [49], resulting in citrullinated histone H3-dependent (Cit-Histone H3) lytic or suicidal NETosis, Figure 1. These lytic NETs are released by the presence of extracellular microbes, fungi, viruses, interferon, phorbol myristate acetate (PMA), IL-8, antibody–antigen complexes, autoantibodies, and concanavalin A.
NOX2-independent pathways can lead to the formation of non-lytic NETs, also known as vital NETs [50]. These NETs are released in the presence of Staphylococcus aureus (S. aureus), Escherichia coli (E. coli), damage-associated molecular patterns (DAMPs), platelets, Toll-like receptors 2/4 (TLR2/4), and lymphocytes associated with a function neutrophil antigen 1 (LFA1).
Other less-studied pathways that induce NETs include those involving signal inhibitory receptor 1 (SIRL1) [51] and extracellular cold-inducible RNA-binding protein (eCIRP) through PD4 [52]. In addition, the formation of mitochondria-dependent vital NETs by silent information regulator 1 (SIRT1) that has been reported in both tumour-associated aged neutrophils (Naged, CXCR4+ CD62L low) in breast cancer lung metastasis [53] and non-tumour pathologies [54] has not been studied in preeclampsia.
NETs can be identified and quantified by distinct visualisation methods or through markers of their released products. Visualisation methods use molecules that are intercalated into DNA, such as propidium iodide and SYTOX Orange. On the other hand, the markers include those that detect DNA-histone complexes (nucleosomes), MPO [55], circulating MPO complexes (MPO)-DNA [56], surface citrullinated histone (H3cit), double-stranded DNA (ds), myeloid-related protein (MRP), DNase, and elastase [57,58].

5. NETs in PE

In PE, there is an increased trafficking of foetal cells into the maternal circulation. The elevated concentration of DNA in blood from foetal-derived cells could activate the immune response and induce cytokine production, potentially leading to pregnancy complications and an increased risk of foetal rejection.
Higher concentrations of NET components (DNA, histones, and MPO) have been reported in the plasma of pregnant women with preeclampsia compared to healthy pregnant women [55]. The first molecule associated with neutrophil activation in the blood of women with PE was neutrophil elastase [59].
Concordantly, in vitro experiments have shown that neutrophils from pregnant women with PE are more likely to form NETs compared to healthy pregnant and non-pregnant women [28]. Also, women with PE and obesity have a higher presence of MPO in their systemic vasculature. MPO catalyses the formation of ROS and RNS in PE by consuming circulating nitric oxide [60,61].
Marder et al. analysed placental samples from women with PE, healthy pregnant women, and abdominal tissue from non-pregnant women, revealing that MPO expression is elevated in tissues from pregnant women compared to non-pregnant women. Notably, NETs were found in the intervillous space in PE [62].
Using anti-NE, Gupta et al. demonstrated that NETs were present in the vicinity of the syncytiotrophoblastic layer in healthy pregnant and preeclamptic women. However, the intervillous space was frequently infiltrated by numerous NETs, and their presence increased in PE [63]. Anti-histone 2A (H2A) staining showed that H2A was significantly increased in the intervillous space of women with PE [64]. Gupta et al. showed that inflammatory syncytiotrophoblast microparticles (STBM) and IL-8-induced neutrophil extracellular DNA extrusion increased CD11b expression and ROS generation in PE [65].
PE is associated with markers of autoimmunity, including anti-β2 glycoprotein-I (ab2GPI); anticardiolipin antibodies (aCL); lupus anticoagulant (LA); β1, β2, and α1 adrenoreceptors; prothrombin; endothelin-1 receptor type A (ETA-AA); and Angiotensin II receptor type 1(AT1-AA) antibodies [66,67] as well as antibodies with different specificity than AT1-AA [68]. Furthermore, the level of antineutrophil cytoplasmic autoantibodies (ANCA) in PE patients was significantly elevated compared with a healthy pregnancy [69]. Although these autoantibodies may induce NETosis during PE, this hypothesis remains to be addressed.
Altogether, the studies mentioned above provide a compelling association between preeclampsia and the increased presence of NETs in pregnant women, Figure 2. Strategies targeting NETs in preeclampsia may hold promise for mitigating immune responses and complications during pregnancy.

6. Pharmacological Modulation of NETs

NETs can be targeted through different drugs with diverse mechanisms of action, Table 1.
Several pharmacological agents are currently being studied for the treatment of moderate and severe preeclampsia (Figure 3), including aspirin, sildenafil citrate, hydroxychloroquine, pravastatin, metformin, magnesium sulphate, nifedipine, labetalol, and nitro-glycerine. However, only one trial has been reported, on clinicaltrials.gov, that was designed to inhibit NET formation in PE using Toll-like receptor (TLR) blocking antibodies. Some agents mitigate NETs by inhibiting ROS, including flavonoids like epicatechin and rutin, along with vitamin C, 5-aminosalicylic acid (5-ASA), N-acetyl-L-cysteine (NAC), dexamethasone, azithromycin, and Glycyrrhiza glabra [70,71]. Others inhibit the JAK/STAT pathway such as tofacitinib, ruxolitinib, and baricitinib [72]. Furthermore, interleukin antagonists, such as tocilizumab, canakinumab, anakinra, and rilonacept, also modulate NETs [73].
Amid the COVID-19 pandemic, there has been a surge in accelerated investigations on drugs and molecules repurposed for their potential to inhibit NETs, as summarized in Table 1. Some of them have garnered attention for their well-established anti-inflammatory effects. Some have not only been utilized in inflammation management but also in aiding lung maturation during preterm births and mitigating preeclampsia [74,75,76,77,78,79,80,81]. Aspirin, vitamin D, and recombinant human DNase I have emerged as candidates worthy of study due to their reported lower risk or absence of mutagenicity and genotoxicity.
The Raf-MEK-ERK signalling pathway is involved in NET formation through the activation of NADPH oxidase. Drugs that reduce NET formation through this pathway include curcumin, GW5074 [82], and Celastrol [83]. While drugs that inhibit NETs by inhibiting the phosphorylation of the p65 subunit of NF-κβ are anti-inflammatory drugs such as acetylsalicylic acid (ASA), BAY 11-7082, and Ro 106-9920 [84].
On the other hand, cyclosporine A uses the calcineurin pathway to reduce NETs [85], while the antioxidants N-acetylcysteine, ethotrexate, trolox, tempol [86], epigallocatechin-3-gallate [87], and diphenyleneiodonium chloride (DPI) [88] decrease NETs by reducing mitochondrial ROS formation [85]. DPI also reduces NET formation through the PKC-βII pathway [89] like metformin. Other drugs that have been shown to significantly inhibit ROS-dependent NET production are propofol and lipid emulsion [90].
Furthermore, drugs such as Hydroxychloroquine (HCQ) [91], PF3758309, and IPA-3 inhibit NETs through Rac2 [92]. Chloroquine (CQ) and HCQ can prevent NET formation by inhibiting PAD4 [93] or TLR signalling as with HCQ, CQ, and enpatoran [94]. In addition, PDE4 inhibitors such as apremilast, rolipram, and crisaborole can reduce the formation of NETs [95].
Table 1. Molecules that downregulate NETosis.
Table 1. Molecules that downregulate NETosis.
Drugs SubstanceMechanism of ActionEvaluated for NOX-Dependent (NOX-D) and ROS-Independent (ROX-I) NET FormationClinical Trials/Models/ExamplesMutagenicity and Genotoxicity In Vitro
Drugs
Aspirin (ASA)Human neutrophils were stimulated with Phorbol 12-myristate 13-acetate (PMA) or TNF-α. In addition, ASA, BAY 11-7082, and Ro 106-9920 prevented the formation of NETs by reducing the phosphorylation of the p65 subunit of NF-κβ [84].NOX-DIn isolated neutrophils stimulated with sodium hydroxide, ASA can enhance the migration of corneal epithelial cells (HCEs) and reduce the formation of NETs [96].ASA can protect against genotoxicity [97].
DexamethasoneInhibits neutrophil functions such as intracellular ROS, degranulation, and NETosis.NOX-DNeutrophils cultured with dexamethasone showed reduced NET formation, after stimulation with PMA [98].It can induce significant DNA damage in human cells; however, it passes the Ames/Salmonella assay [99,100].
ResveratrolDecreases the release of free DNA from neutrophils and NETosisNOX-DDuring in vitro tests in the presence of PMA, it was shown that resveratrol decreases the formation of NETs and cytokine production in healthy controls and with COVID-19 [101].Despite its genotoxic effects, it does not cause mutagenesis and is used for its genotoxic activity against gastric cell adenocarcinoma [102,103].
Cyclosporine AInhibits IL-8-induced NETosis by inhibiting the calcineurin pathway.NOX-DIn isolated neutrophils, stimulated with PMA or ionomycin, treated with Cyclosporine A or ascomycin, the formation of NETs decreases [104].It is not genotoxic in humans. It inhibits the protein phosphatase calcineurin and can induce lymphoma in Xpa/p53 mice [105].
AzithromycinDecreases the production of ROS NOX-DPre-treatment with Azithromycin decreases NETosis in neutrophils isolated from PMA-stimulated healthy subjects. This effect is observed at low doses [106].It does not induce mutations or chromosomal aberrations in microbial or mammalian cells [107].
ChloramphenicolReduces the formation of NETs, possibly by inhibiting myeloperoxidase (MPO) NOX-DPre-treatment with chloramphenicol reduces PMA-induced NET release [94].In rodents and human cells, it is a pro-mutagenic compound [108].
MetforminAffects nuclear dynamics (delobulation and decondensation) as well as PKC-βII membrane translocation and NADPH oxidase activation in neutrophils [109].NOX-DMetformin decreases NETosis and its components such as elastase, proteinase-3, histones, and double-strand DNA in PMA-stimulated neutrophils in in vitro and clinical trial samples [109].There is conflicting evidence about the effects of metformin. Micronucleus assay suggests it may be genotoxic; however, analyses using chromosomal aberration (CA) and cytokinesis-block micronucleus (CBMN) assay report that it has a radioprotective effect on DNA damage and apoptosis in human lymphocytes [110,111].
HydroxychloroquineInhibits the expression of PAC4, Rac2, and the formation of NETsNOX-DHydroxychloroquine alleviates hepatic ischemia/reperfusion (IR) injury in severe combined immunodeficiency (SCID) mice and C57BL/6 mice by inhibiting NETosis [91].It induces both oxidative DNA damage detected by 8-oxodG and the induction of mutants in mouse embryonic fibroblasts [112].
HeparinNET molecules, such as neutrophil elastase, interact with heparin and heparin oligomers to form molecular complexes that can regulate NETosis [113].
Low molecular weight heparin (LMWH) can repair nucleosome/histone 3-mediated damage in trophoblasts [69].
--In vitro studies have shown that heparinized adsorbents such as heparin sepharose deplete PF4, histones/nucleosomes, and HMGB1 [114].
Heparin pre-treatment decreased serum and lung NETs in a C57BL/6J mice model [115].
Circulating histones bound to H3 and H4 nucleosomes are increased in patients with preeclampsia and intrauterine growth restriction. H3 affects extravillous trophoblast migration, invasion, and survival. This effect can be reversed in vitro by LMWH, but not with ASA [69].
LMWH does not show any mutagenic activity [116,117].
Vitamin DVitamin D supplementation has been shown to reduce the risk of preeclampsia [118], as well as decrease the generation of NETs, particularly when combined with omega-3 PUFAs [119].NOX-DPMA-stimulated neutrophils from patients with systemic lupus erythematosus (SLE) and hypovitaminosis D were treated with calcitriol/1,25(OH)2D3. The authors reported a dose-independent decrease in externalised neutrophil elastase (NE) during NETosis [120].In cancer rodents treated with cyclophosphamide, it reduced the frequency of chromosomal aberrations in Chinese hamster lung cells and reduced micronuclei and lymphocyte damage in mice [121].
DisulfiramInhibits NETs [122].NOX-DIt reduced NETs and perivascular fibrosis and downregulated innate immune and complement/coagulation pathways [111].--
CurcuminInhibits the generation of NETs by suppressing the MEK/ERK pathway [123].NOX-DIn a mouse model, curcumin was shown to reduce hepatic ischemia-reperfusion injury by inhibiting NET formation [91].High concentrations are cytotoxic and increase the frequency of micronuclei in PC12 cells; at low doses, it reduces the number of micronuclei induced by cisplatin [124].
Phosphodiesterase Type-4 (PDE4) inhibitors such as
Roflumilast (Daliresp), Apremilast (Otezla), and Rolipram
Roflumilast blocks PDE4 and reduces in vitro and in vivo NETosis in animal models [125].
Inhibition of PDE4 by rolipram prevents the adhesion of platelets and neutrophils, which involves members of the Src family kinase (SFK) [126,127].
ROX-IClinical trials have been conducted with Roflumilast for severe chronic obstructive pulmonary disease (COPD) and with Otezla for psoriasis [128].--
Glycyrrhiza glabraInhibits ROS, mitochondrial ROS (mtROS), NET generation, and cytokine release.--In an animal model, Glycyrrhiza glabra was proven to decrease COVID-19 pathology by reducing NETosis [129]. --
Protein molecules
Activated protein C (APC)Cleaves and detoxifies extracellular histones and its effect on reducing NETs dependent on endothelial protein C receptor (EPCR), protease-activated receptor 3 (PAR3), and macrophage antigen-1 (Mac-1) [130].--A clinical trial was conducted evaluating the safety and efficacy of recombinant human-activated protein C (rhAPC; drotrecogin alfa [activated]) in preeclampsia, but further studies are needed [131].
APC variants have been designed to have a greater ability to destroy histone H3 with fewer anticoagulant properties [132].
Drotrecogin alfa (activated) has not been studied for carcinogenicity [133].
Recombinant human DNase I (rhDNase I, rhDNase, Pulmozyme®, dornase alfa)Hydrolyses extracellular DNA released by neutrophils [134].--Intravenous administration of rhDNase to mice degraded NETs and attenuated coagulopathy in the acute respiratory distress syndrome (ARDS) model [135].It does not show cytotoxicity in human peripheral blood mononuclear cells [136].
Other molecules such as PGE2, which has been used in labour induction, decrease NET formation through cAMP production. However, there are also no reports of trials with PGE2 to reduce NETs in preeclampsia [137,138,139].
Others, although they reduce NET formation, are not considered due to the risk of mutagenicity or genotoxicity during pregnancy. A mutagenic agent causes damage to DNA and results in mutation, while a genotoxic agent can cause damage to DNA or chromosomes but without necessarily resulting in mutation [140], such as 5-fluorouracil (5FU) [141].

7. Therapeutic Approaches to Inhibit NET Formation in PE

Low-dose aspirin (75 mg) is used in the clinic to prevent preeclampsia; its best-known action is the inhibition of prostaglandin and thromboxane A2 synthesis by cyclooxygenases COX-1 and COX-2.
Preeclampsia is characterized by elevation of TXA2 and decreased prostacyclin. TXA2 is a procoagulant molecule related to thrombosis observed in preeclampsia; prostacyclin PG1 is a vasodilator molecule. In PE, there is an increase in TXA and a decrease in PGI; aspirin acts on both, although platelets are the main producers of TXA2 [142].
PE is related to an inflammatory and prothrombotic state, and thus the effect of aspirin in inhibiting inflammation and platelet aggregation is considered to prevent and treat it; however, beyond its antithrombotic action, the effect of aspirin on the activation and modulation of the inflammatory response of neutrophils has recently been studied.
During pregnancy, neutrophils express PAR-1 and F2R, which are thrombin receptors. Aspirin prevented p65 translocation of NF-kβ to the nucleus and TXA2 production in neutrophils from pregnant women via PAR-1. In addition, aspirin inhibits lipid peroxidation by COX-2 in pregnant neutrophils [143]. These PAR-1 and F2R receptors are not expressed in neutrophils from pregnant women.
Both aspirin and glucocorticoids reduce NF-κβ activity. It is a family of transcription factors, phosphorylated p65 (RelA), RelB, c-Rel, p105/p50 (NF-κβ1) and p100/52 (NF-κβ2), that regulate the immune response, inflammatory response, apoptosis and proliferation [144].
In PE, neutrophil adhesion to the vasculature increases. This adhesion could be reduced in the presence of an intermediate in aspirin metabolism and aspirin-activated lipoxin A4 (ATL, 15-epi-LXA4) [145]. Aspirin generally inhibits NETosis, but more studies are needed to analyse its usefulness in preeclampsia.
Some meta-analyses and individual cases have shown that low-dose aspirin can prevent preeclampsia in people with a risk factor, but aspirin crosses the placental barrier and can inhibit foetal platelet aggregation, increasing the risk of spontaneous abortion; thus, its use is under medical supervision. The use of aspirin has been empirical, but more data are needed to implement it as a treatment [146].
Furthermore, the therapeutic use of glucocorticoids in PE is primarily indicated in cases of foetal growth restriction. It has been proposed that single doses after week 24 (2 doses of 12 mg betamethasone 24 h apart or 6 mg dexamethasone 12 h apart before delivery) help foetal lung maturation in PE [147,148]. Dexamethasone has been shown to transiently restore absent end-diastolic flow in the umbilical artery in early-onset preeclampsia [149]. However, dexamethasone treatment for PE should be carefully considered, as the use of high doses and repeated doses should be avoided for fear of possible long-term adverse effects on the foetal brain [150].
On the other hand, glucocorticoids such as dexamethasone, prednisone, and rimexolone interact through glucocorticoid receptor antagonism in PE [151]. Glucocorticoid receptors are expressed in the trophoblast, and their inhibition favours the expression of angiotensin 2 receptor antagonist (ART2), which has vasodilatory effects. In normal pregnancy, ART2 predominates, reducing hypertension, while ART1, which has a vasoconstrictor effect, increases in PE [152]. Numerous studies show that dexamethasone in early pregnancy has harmful effects such as deficient trophoblast development, increased trophoblast invasion inhibitor SERPINE1, and increased systolic blood pressure [153,154], even causing DNA damage (Table 1). Dexamethasone caused abnormal mitochondrial morphology and mitochondrial dysfunction in the placentas of pregnant rats, in addition to altering placental signalling pathways such as oxidative phosphorylation, inflammation, and the insulin-like growth factor system [155].
PE is characterized by a state of systemic inflammation where platelet abnormalities occur. It has been shown that dexamethasone significantly inhibited degranulation, intracellular ROS production, CXCL8 release, and neutrophil NETosis in patients with severe COVID-19 pneumonia [98]. Dexamethasone has been shown to inhibit Staphylococcus aureus-induced NET formation via upregulation of TLR2 and TLR4 receptors [156], this could mean that Dexamethasone induces vital NETs. In PE, dexamethasone has been shown to delay neutrophil apoptosis in early-onset preeclampsia; however, it does not affect the rate of neutrophil apoptosis between late-onset preeclampsia and normal pregnancy [157].
Another drug with a potential therapeutic effect, particularly in preeclampsia associated with severe obesity [158], is metformin. This is a hypoglycemic agent that at therapeutic doses has a reduced impact on trophoblast differentiation [159]. Metformin activates AMPK signalling pathways involved in the regulation of NF-κβ/sFlt-1, and Nrf2/HO-1 signalling pathways, thus inhibiting the inflammatory response and oxidative stress [160].
Finally, several systematic reviews have reported that 25-hydroxyvitamin D supplementation before 20 weeks of gestation could reduce NETosis (Table 1) and the risk of PE [161,162], through inhibition of lysosome-associated membrane glycoprotein 3 [163].

8. Conclusions

The pathophysiology of preeclampsia is significantly aligned with the activation of neutrophils and the release of mediators, especially those associated with neutrophil extracellular traps (NETs). Consequently, understanding the role of neutrophils and NETs becomes imperative in the context of preeclampsia treatment. Currently, certain drugs are already under review in clinical trials; these include azithromycin, hydroxychloroquine, aspirin, and metformin. Additionally, promising results have already been observed in the use of vitamin D for preeclampsia. This underscores the importance of exploring and considering neutrophil and NET-targeted treatments in the comprehensive approach to managing preeclampsia.

Author Contributions

Conceptualization, J.d.D.R.-R. and E.P.-C.; investigation, L.L.H.G., L.P.-C.M., M.T.H.-H. and G.M.A.; writing—original draft preparation, L.L.H.G., L.P.-C.M., M.T.H.-H., G.M.A., M.M.C. and E.P.-C.M.; writing—review and editing, L.L.H.G., L.P.-C.M., M.T.H.-H., G.M.A., V.C.H., I.A.G., C.A.M.-C., E.R.-M., C.R.D., C.M.L.D. and M.E.A.V.; supervision, C.A.M.-C., M.E.A.V., J.d.D.R.-R. and E.P.-C.; project administration, M.M.C. and E.P.-C.M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health (NIDDK) K01 DK128379-01 (J.d.D.R.R.).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Data sharing is not applicable.

Acknowledgments

Thanks to the Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCyT) for the scholarship granted to L.L.H.G., CVU number (792544). Also, to the National Technology of Mexico (TecNM)/IT Oaxaca 18501.23-P, and to Eli Cruz Parada and Charlotte Grundy for their technical advice.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Robillard, P.Y.; Dekker, G.; Iacobelli, S.; Chaouat, G. An essay of reflection: Why does preeclampsia exist in humans, and why are there such huge geographical differences in epidemiology? J. Reprod. Immunol. 2016, 114, 44–47. [Google Scholar] [CrossRef]
  2. Elgari, M.M.; Khabour, O.F.; Alhag, S.M. Correlations between changes in hematological indices of mothers with preeclampsia and umbilical cord blood of newborns. Clin. Exp. Hypertens. 2019, 41, 58–61. [Google Scholar] [CrossRef]
  3. Schindler, A.E. New data about preeclampsia: Some possibilities of prevention. Gynecol. Endocrinol. 2018, 34, 636–637. [Google Scholar] [CrossRef]
  4. Roberts, J.M.; Rich-Edwards, J.W.; McElrath, T.F.; Garmire, L.; Myatt, L.; Global Pregnancy Collaboration. Subtypes of Preeclampsia: Recognition and Determining Clinical Usefulness. Hypertension 2021, 77, 1430–1441. [Google Scholar] [CrossRef]
  5. Gyselaers, W. Preeclampsia Is a Syndrome with a Cascade of Pathophysiologic Events. J. Clin. Med. 2020, 9, 2245. [Google Scholar] [CrossRef]
  6. Shokry, M.; Bedaiwy, M.A.; Fathalla, M.M.; Alsemary, A.; Elwakil, S.; Murphy, A. Maternal serum placental growth factor and soluble fms-like tyrosine kinase 1 as early predictors of preeclampsia. Acta Obstet. Gynecol. Scand. 2010, 89, 143–146. [Google Scholar] [CrossRef] [PubMed]
  7. Tamás, P.; Kovács, K.; Várnagy, Á.; Farkas, B.; Alemu Wami, G.; Bódis, J. Preeclampsia subtypes: Clinical aspects regarding pathogenesis, signs, and management with special attention to diuretic administration. Eur. J. Obstet. Gynecol. Reprod. Biol. 2022, 274, 175–181. [Google Scholar] [CrossRef]
  8. Celik, C.; Gezginç, K.; Altintepe, L.; Tonbul, H.Z.; Yaman, S.T.; Akyürek, C.; Türk, S. Results of the pregnancies with HELLP syndrome. Ren. Fail. 2003, 25, 613–618. [Google Scholar] [CrossRef]
  9. Dennis, A.T. Cardiac Function in Women with Preeclampsia; University of Melbourne, Department of Pharmacology: Parkville, Australia, 2010. [Google Scholar]
  10. Wang, Y.; Gu, Y.; Lucas, M.J. Expression of thrombin receptors in endothelial cells and neutrophils from normal and preeclamptic pregnancies. J. Clin. Endocrinol. Metab. 2002, 87, 3728–3734. [Google Scholar] [CrossRef] [PubMed]
  11. Wang, Y.; Gu, Y.; Philibert, L.; Lucas, M.J. Neutrophil activation induced by placental factors in normal and pre-eclamptic pregnancies in vitro. Placenta 2001, 22, 560–565. [Google Scholar] [CrossRef] [PubMed]
  12. Crocker, I.P.; Wellings, R.P.; Fletcher, J.; Baker, P.N. Neutrophil function in women with pre-eclampsia. Br. J. Obstet. Gynaecol. 1999, 106, 822–828. [Google Scholar] [CrossRef]
  13. Fridlender, Z.G.; Sun, J.; Kim, S.; Kapoor, V.; Cheng, G.; Ling, L.; Worthen, G.S.; Albelda, S.M. Polarization of tumor-associated neutrophil phenotype by TGF-beta: “N1” versus “N2” TAN. Cancer Cell 2009, 16, 183–194. [Google Scholar] [CrossRef]
  14. Grieshaber-Bouyer, R.; Nigrovic, P.A. Neutrophil Heterogeneity as Therapeutic Opportunity in Immune-Mediated Disease. Front. Immunol. 2019, 10, 346. [Google Scholar] [CrossRef]
  15. McKenna, E.; Mhaonaigh, A.U.; Wubben, R.; Dwivedi, A.; Hurley, T.; Kelly, L.A.; Stevenson, N.J.; Little, M.A.; Molloy, E.J. Neutrophils: Need for Standardized Nomenclature. Front. Immunol. 2021, 12, 602963. [Google Scholar] [CrossRef]
  16. Mihaila, A.C.; Ciortan, L.; Macarie, R.D.; Vadana, M.; Cecoltan, S.; Preda, M.B.; Hudita, A.; Gan, A.M.; Jakobsson, G.; Tucureanu, M.M.; et al. Transcriptional Profiling and Functional Analysis of N1/N2 Neutrophils Reveal an Immunomodulatory Effect of S100A9-Blockade on the Pro-Inflammatory N1 Subpopulation. Front. Immunol. 2021, 10, 708770. [Google Scholar] [CrossRef]
  17. Vono, M.; Lin, A.; Norrby-Teglund, A.; Koup, R.A.; Liang, F.; Loré, K. Neutrophils acquire the capacity for antigen presentation to memory CD4+ T cells in vitro and ex vivo. Blood 2017, 129, 1991–2001. [Google Scholar] [CrossRef]
  18. Dolislager, C.G.; Callahan, S.M.; Donohoe, D.R.; Johnson, J.G. Campylobacter jejuni induces differentiation of human neutrophils to the CD16hi /CD62Llo subtype. J. Leukoc. Biol. 2022, 112, 1457–1470. [Google Scholar] [CrossRef]
  19. Granai, M.; Warm, V.; Vogelsberg, A.; Milla, J.; Greif, K.; Vogel, U.; Bakchoul, T.; Rosenberger, P.; Quintanilla-Martinez, L.; Schürch, C.M.; et al. Impact of P-selectin-PSGL-1 Axis on Platelet-Endothelium-Leukocyte Interactions in Fatal COVID-19. Lab. Investig. 2023, 103, 100179. [Google Scholar] [CrossRef]
  20. Dehghani, T.; Panitch, A. Endothelial cells, neutrophils and platelets: Getting to the bottom of an inflammatory tri-angle. Open Biol. 2020, 10, 200161. [Google Scholar] [CrossRef]
  21. Gupta, A.K.; Gebhardt, S.; Hillermann, R.; Holzgreve, W.; Hahn, S. Analysis of plasma elastase levels in early and late onset preeclampsia. Arch. Gynecol. Obstet. 2006, 273, 239–242. [Google Scholar] [CrossRef]
  22. Zhong, X.Y.; Gebhardt, S.; Hillermann, R.; Tofa, K.C.; Holzgreve, W.; Hahn, S. Circulatory nucleosome levels are significantly increased in early and late-onset preeclampsia. Prenat. Diagn. 2005, 25, 700–703. [Google Scholar] [CrossRef] [PubMed]
  23. Lok, C.A.; Jebbink, J.; Nieuwland, R.; Faas, M.M.; Boer, K.; Sturk, A.; Van Der Post, J.A. Leukocyte activation and circulating leukocyte-derived microparticles in preeclampsia. Am. J. Reprod. Immunol. 2009, 61, 346–359. [Google Scholar] [CrossRef]
  24. von Dadelszen, P.; Watson, R.W.; Noorwali, F.; Marshall, J.C.; Parodo, J.; Farine, D.; Lye, S.J.; Ritchie, J.W.; Rotstein, O.D. Maternal neutrophil apoptosis in normal pregnancy, preeclampsia, and normotensive intrauterine growth restriction. Am. J. Obstet. Gynecol. 1999, 181, 408–414. [Google Scholar] [CrossRef]
  25. Xin, H.; Wang, H.L. Expression changes and clinical significance of annexin V in maternal blood and placenta in patients with preeclampsia. Zhonghua Fu Chan Ke Za Zhi 2011, 46, 88–93. (In Chinese) [Google Scholar] [PubMed]
  26. Barden, A.; Ritchie, J.; Walters, B.; Michael, C.; Rivera, J.; Mori, T.; Croft, K.; Beilin, L. Study of plasma factors associated with neutrophil activation and lipid peroxidation in preeclampsia. Hypertension 2001, 38, 803–808. [Google Scholar] [CrossRef] [PubMed]
  27. Sabatier, F.; Bretelle, F.; D’ercole, C.; Boubli, L.; Sampol, J.; Dignat-George, F. Neutrophil activation in preeclampsia and isolated intrauterine growth restriction. Am. J. Obstet. Gynecol. 2000, 183, 1558–1563. [Google Scholar] [CrossRef]
  28. Hu, Y.; Li, H.; Yan, R.; Wang, C.; Wang, Y.; Zhang, C.; Liu, M.; Zhou, T.; Zhu, W.; Zhang, H.; et al. Increased Neutrophil Activation and Plasma DNA Levels in Patients with Pre-Eclampsia. Thromb. Haemost. 2018, 118, 2064–2073. [Google Scholar] [CrossRef]
  29. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry 2020, 85, 1178–1190. [Google Scholar] [CrossRef]
  30. Barden, A.; Graham, D.; Beilin, L.J.; Ritchie, J.; Baker, R.; Walters, B.N.; Michael, C.A. Neutrophil CD11B expression and neutrophil activation in pre-eclampsia. Clin. Sci. 1997, 92, 37–44. [Google Scholar] [CrossRef]
  31. Luppi, P.; Tse, H.; Lain, K.Y.; Markovic, N.; Piganelli, J.D.; DeLoia, J.A. Preeclampsia activates circulating immune cells with engagement of the NF-kappaβ pathway. Am. J. Reprod. Immunol. 2006, 56, 135–144. [Google Scholar] [CrossRef]
  32. Han, C.; Chen, Y.Y.; Dong, J.F. Prothrombotic state associated with preeclampsia. Curr. Opin. Hematol. 2021, 28, 323–330. [Google Scholar] [CrossRef] [PubMed]
  33. Mellembakken, J.R.; Aukrust, P.; Olafsen, M.K.; Ueland, T.; Hestdal, K.; Videm, V. Activation of leukocytes during the uteroplacental passage in preeclampsia. Hypertension 2002, 39, 155–160. [Google Scholar] [CrossRef] [PubMed]
  34. Leik, C.E.; Walsh, S.W. Neutrophils infiltrate resistance-sized vessels of subcutaneous fat in women with preeclampsia. Hypertension 2004, 44, 72–77. [Google Scholar] [CrossRef]
  35. Shah, T.J.; Walsh, S.W. Activation of NF-kappaβ and expression of COX-2 in association with neutrophil infiltration in systemic vascular tissue of women with preeclampsia. Am. J. Obstet. Gynecol. 2007, 196, 48.e1–48.e18. [Google Scholar] [CrossRef]
  36. Shukla, J.; Walsh, S.W. Neutrophil release of myeloperoxidase in systemic vasculature of obese women may put them at risk for preeclampsia. Reprod. Sci. 2015, 22, 300–307. [Google Scholar] [CrossRef] [PubMed]
  37. Cadden, K.A.; Walsh, S.W. Neutrophils, but not lymphocytes or monocytes, infiltrate maternal systemic vasculature in women with preeclampsia. Hypertens. Pregnancy 2008, 27, 396–405. [Google Scholar] [CrossRef]
  38. Mincheva-Nilsson, L.; Baranov, V. Placenta-derived exosomes and syncytiotrophoblast microparticles and their role in human reproduction: Immune modulation for pregnancy success. Am. J. Reprod. Immunol. 2014, 72, 440–457. [Google Scholar] [CrossRef]
  39. Aly, A.S.; Khandelwal, M.; Zhao, J.; Mehmet, A.H.; Sammel, M.D.; Parry, S. Neutrophils are stimulated by syncytiotrophoblast microvillous membranes to generate superoxide radicals in women with preeclampsia. Am. J. Obstet. Gynecol. 2004, 190, 252–258. [Google Scholar] [CrossRef] [PubMed]
  40. Liu, D.; Li, Q.; Ding, H.; Zhao, G.; Wang, Z.; Cao, C.; Dai, Y.; Zheng, M.; Zhu, X.; Wu, Q.; et al. Placenta-derived IL-32β activates neutrophils to promote preeclampsia development. Cell. Mol. Immunol. 2021, 18, 979–991. [Google Scholar] [CrossRef]
  41. Kenny, E.F.; Herzig, A.; Krüger, R.; Muth, A.; Mondal, S.; Thompson, P.R.; Brinkmann, V.; Bernuth, H.V.; Zychlinsky, A. Diverse stimuli engage different neutrophil extracellular trap pathways. Elife 2017, 6, e24437. [Google Scholar] [CrossRef]
  42. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535. [Google Scholar] [CrossRef] [PubMed]
  43. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  44. Van der Zwan, L.P.; Scheffer, P.G.; Dekker, J.M.; Stehouwer, C.D.; Heine, R.J.; Teerlink, T. Hyperglycemia and oxidative stress strengthen the association between myeloperoxidase and blood pressure. Hypertension 2010, 55, 1366–1372. [Google Scholar] [CrossRef] [PubMed]
  45. Mahat, R.K.; Singh, N.; Rathore, V. Association of myeloperoxidase with cardiovascular disease risk factors in prediabetic subjects. Diabetes Metab. Syndr. 2019, 13, 396–400. [Google Scholar] [CrossRef]
  46. Parker, H.; Winterbourn, C.C. Reactive oxidants and myeloperoxidase and their involvement in neutrophil extracellular traps. Front. Immunol. 2013, 3, 424. [Google Scholar] [CrossRef] [PubMed]
  47. Demkow, U. Molecular Mechanisms of Neutrophil Extracellular Trap (NETs) Degradation. Int. J. Mol. Sci. 2023, 24, 4896. [Google Scholar] [CrossRef]
  48. Zhang, Y.; Song, J.; Zhang, Y.; Li, T.; Peng, J.; Zhou, H.; Zong, Z. Emerging Role of Neutrophil Extracellular Traps in Gastrointestinal Tumors: A Narrative Review. Int. J. Mol. Sci. 2022, 24, 334. [Google Scholar] [CrossRef]
  49. Chen, W.A.; Boskovic, D.S. Neutrophil Extracellular DNA Traps in Response to Infection or Inflammation, and the Roles of Platelet Interactions. Int. J. Mol. Sci. 2024, 25, 3025. [Google Scholar] [CrossRef]
  50. Tan, C.; Aziz, M.; Wang, P. The vitals of NETs. J. Leukoc. Biol. 2021, 110, 797–808. [Google Scholar] [CrossRef]
  51. Besteman, S.B.; Callaghan, A.; Hennus, M.P.; Westerlaken, G.H.A.; Meyaard, L.; Bont, L.L. Signal inhibitory receptor on leukocytes (SIRL)-1 and leukocyte- associated immunoglobulin-like receptor (LAIR)-1 regulate neutrophil function in infants. Clin. Immunol. 2020, 211, 108324. [Google Scholar] [CrossRef]
  52. Ode, Y.; Aziz, M.; Jin, H.; Arif, A.; Nicastro, J.G.; Wang, P. Cold-inducible RNA-binding Protein Induces Neutrophil Extracellular Traps in the Lungs during Sepsis. Sci. Rep. 2019, 9, 6252. [Google Scholar] [CrossRef]
  53. Yang, C.; Wang, Z.; Li, L.; Zhang, Z.; Jin, X.; Wu, P.; Sun, S.; Pan, J.; Su, K.; Jia, F.; et al. Aged neutrophils form mitochondria-dependent vital NETs to promote breast cancer lung metasta-sis. J. Immunother. Cancer 2021, 9, e002875. [Google Scholar] [CrossRef] [PubMed]
  54. Kawabata, S.; Takagaki, M.; Nakamura, H.; Nishida, T.; Terada, E.; Kadono, Y.; Izutsu, N.; Takenaka, T.; Matsui, Y.; Yamada, S.; et al. Association of Gut Microbiome with Early Brain Injury After Subarachnoid Hemorrhage: An Experimental Study. Transl. Stroke Res. 2024, 15, 87–100. [Google Scholar] [CrossRef]
  55. de Buhr, N.; von Köckritz-Blickwede, M. Detection, Visualization, and Quantification of Neutrophil Extracellular Traps (NETs) and NET Markers. Methods Mol. Biol. 2020, 2087, 425–442. [Google Scholar] [CrossRef] [PubMed]
  56. Donkel, S.J.; Wolters, F.J.; Ikram, M.A.; de Maat, M.P.M. Circulating Myeloperoxidase (MPO)-DNA complexes as marker for Neutrophil Extracellular Traps (NETs) levels and the association with cardiovascular risk factors in the general population. PLoS ONE 2021, 16, e0253698. [Google Scholar] [CrossRef]
  57. Lee, K.H.; Cavanaugh, L.; Leung, H.; Yan, F.; Ahmadi, Z.; Chong, B.H.; Passam, F. Quantification of NETs-associated markers by flow cytometry and serum assays in patients with thrombosis and sepsis. Int. J. Lab. Hematol. 2018, 40, 392–399. [Google Scholar] [CrossRef] [PubMed]
  58. Grilz, E.; Mauracher, L.M.; Posch, F.; Königsbrügge, O.; Zöchbauer-Müller, S.; Marosi, C.; Lang, I.; Pabinger, I.; Ay, C. Citrullinated histone H3, a biomarker for neutrophil extracellular trap formation, predicts the risk of mortality in patients with cancer. Br. J. Haematol. 2019, 186, 311–320. [Google Scholar] [CrossRef] [PubMed]
  59. Halim, A.; Kanayama, N.; El Maradny, E.; Maehara, K.; Bhuiyan, A.B.; Terao, T. Correlated plasma elastase and sera cytotoxicity in eclampsia. A possible role of endothelin-1 induced neutrophil activation in preeclampsia-eclampsia. Am. J. Hypertens. 1996, 9, 33–38. [Google Scholar] [CrossRef] [PubMed]
  60. von Leitner, E.C.; Klinke, A.; Atzler, D.; Slocum, J.L.; Lund, N.; Kielstein, J.T.; Maas, R.; Schmidt-Haupt, R.; Pekarova, M.; Hellwinkel, O.; et al. Pathogenic cycle between the endogenous nitric oxide synthase inhibitor asymmetrical dimethylarginine and the leukocyte-derived hemoprotein myeloperoxidase. Circulation 2011, 124, 2735–2745. [Google Scholar] [CrossRef]
  61. Rocha-Penha, L.; Caldeira-Dias, M.; Tanus-Santos, J.E.; de Carvalho Cavalli, R.; Sandrim, V.C. Myeloperoxidase in Hypertensive Disorders of Pregnancy and Its Relation With Nitric Oxide. Hypertension 2017, 69, 1173–1180. [Google Scholar] [CrossRef]
  62. Marder, W.; Knight, J.S.; Kaplan, M.J.; Somers, E.C.; Zhang, X.; O’Dell, A.A.; Padmanabhan, V.; Lieberman, R.W. Placental histology and neutrophil extracellular traps in lupus and pre-eclampsia pregnancies. Lupus Sci. Med. 2016, 3, e000134. [Google Scholar] [CrossRef] [PubMed]
  63. Gupta, A.; Hasler, P.; Gebhardt, S.; Holzgreve, W.; Hahn, S. Occurrence of neutrophil extracellular DNA traps (NETs) in pre-eclampsia: A link with elevated levels of cell-free DNA? Ann. N. Y. Acad. Sci. 2006, 1075, 118–122. [Google Scholar] [CrossRef] [PubMed]
  64. Moodley, M.; Moodley, J.; Naicker, T. Neutrophil extracellular traps: The synergy source in the placentae of HIV infected women with pre-eclampsia. Pregnancy Hypertens. 2020, 20, 69–74. [Google Scholar] [CrossRef] [PubMed]
  65. Gupta, A.K.; Hasler, P.; Holzgreve, W.; Gebhardt, S.; Hahn, S. Induction of neutrophil extracellular DNA lattices by placental microparticles and IL-8 and their presence in preeclampsia. Hum. Immunol. 2005, 66, 1146–1154. [Google Scholar] [CrossRef] [PubMed]
  66. Jung, E.; Romero, R.; Yeo, L.; Gomez-Lopez, N.; Chaemsaithong, P.; Jaovisidha, A.; Gotsch, F.; Erez, O. The etiology of preeclampsia. Am. J. Obstet. Gynecol. 2022, 226 (Suppl. S2), S844–S866. [Google Scholar] [CrossRef] [PubMed]
  67. Buttrup Larsen, S.; Wallukat, G.; Schimke, I.; Sandager, A.; Tvilum Christensen, T.; Uldbjerg, N.; Tørring, N. Functional autoantibodies against Endothelin-1 receptor type A and Angiotensin II receptor type 1 in patients with preeclampsia. Pregnancy Hypertens. 2018, 14, 189–194. [Google Scholar] [CrossRef]
  68. Elliott, S.E.; Parchim, N.F.; Liu, C.; Xia, Y.; Kellems, R.E.; Soffici, A.R.; Daugherty, P.S. Characterization of antibody specificities associated with preeclampsia. Hypertension 2014, 63, 1086–1093. [Google Scholar] [CrossRef]
  69. Shaarawy, M.; el-Mallah, S.Y.; el-Yamani, A.M. The prevalence of serum antineutrophil cytoplasmic autoantibodies in preeclampsia and eclampsia. J. Soc. Gynecol. Investig. 1997, 4, 34–39. [Google Scholar] [CrossRef] [PubMed]
  70. Mutua, V.; Gershwin, L.J. A Review of Neutrophil Extracellular Traps (NETs) in Disease: Potential Anti-NETs Therapeutics. Clin. Rev. Allergy Immunol. 2021, 61, 194–211. [Google Scholar] [CrossRef]
  71. Kirchner, T.; Hermann, E.; Möller, S.; Klinger, M.; Solbach, W.; Laskay, T.; Behnen, M. Flavonoids and 5-aminosalicylic acid inhibit the formation of neutrophil extracellular traps. Mediat. Inflamm. 2013, 2013, 710239. [Google Scholar] [CrossRef]
  72. Palmroth, M.; Kuuliala, K.; Peltomaa, R.; Virtanen, A.; Kuuliala, A.; Kurttila, A.; Kinnunen, A.; Leirisalo-Repo, M.; Silvennoinen, O.; Isomäki, P. Tofacitinib Suppresses Several JAK-STAT Pathways in Rheumatoid Arthritis In Vivo and Baseline Signaling Profile Associates With Treatment Response. Front. Immunol. 2021, 12, 738481. [Google Scholar] [CrossRef] [PubMed]
  73. Malcova, H.; Milota, T.; Strizova, Z.; Cebecauerova, D.; Striz, I.; Sediva, A.; Horvath, R. Interleukin-1 Blockade in Polygenic Autoinflammatory Disorders: Where Are We now? Front. Pharmacol. 2021, 11, 619273. [Google Scholar] [CrossRef]
  74. Basyreva, L.Y.; Brodsky, I.B.; Gusev, A.A.; Zhapparova, O.N.; Mikhalchik, E.V.; Gusev, S.A.; Shor, D.B.; Dahan, S.; Blank, M.; Shoenfeld, Y. The effect of Intravenous Immunoglobulin (IVIG) on \textit{ex vivo} activation of human leukocytes. Hum. Antibodies 2016, 24, 39–44. [Google Scholar] [CrossRef]
  75. Lood, C.; Hughes, G.C. Neutrophil extracellular traps as a potential source of autoantigen in cocaine-associated autoimmunity. Rheumatology 2017, 56, 638–643. [Google Scholar] [CrossRef]
  76. Hair, P.S.; Enos, A.I.; Krishna, N.K.; Cunnion, K.M. Inhibition of Immune Complex Complement Activation and Neutrophil Extracellular Trap Formation by Peptide Inhibitor of Complement C1. Front. Immunol. 2018, 9, 558. [Google Scholar] [CrossRef] [PubMed]
  77. Hair, P.S.; Enos, A.I.; Krishna, N.K.; Cunnion, K.M. Inhibition of complement activation, myeloperoxidase, NET formation and oxidant activity by PIC1 peptide variants. PLoS ONE 2019, 14, e0226875. [Google Scholar] [CrossRef] [PubMed]
  78. Kajioka, H.; Kagawa, S.; Ito, A.; Yoshimoto, M.; Sakamoto, S.; Kikuchi, S.; Kuroda, S.; Yoshida, R.; Umeda, Y.; Noma, K.; et al. Targeting neutrophil extracellular traps with thrombomodulin prevents pancreatic cancer metastasis. Cancer Lett. 2021, 497, 1–13. [Google Scholar] [CrossRef]
  79. Tadie, J.M.; Bae, H.B.; Jiang, S.; Park, D.W.; Bell, C.P.; Yang, H.; Pittet, J.F.; Tracey, K.; Thannickal, V.J.; Abraham, E.; et al. HMGB1 promotes neutrophil extracellular trap formation through interactions with Toll-like receptor 4. Am. J. Physiol. Lung Cell Mol. Physiol. 2013, 304, L342–L349. [Google Scholar] [CrossRef]
  80. Wygrecka, M.; Kosanovic, D.; Wujak, L.; Reppe, K.; Henneke, I.; Frey, H.; Didiasova, M.; Kwapiszewska, G.; Marsh, L.M.; Baal, N.; et al. Antihistone Properties of C1 Esterase Inhibitor Protect against Lung Injury. Am. J. Respir. Crit. Care Med. 2017, 196, 186–199. [Google Scholar] [CrossRef]
  81. Kakigano, A.; Tomimatsu, T.; Mimura, K.; Kanayama, T.; Fujita, S.; Minato, K.; Kumasawa, K.; Taniguchi, Y.; Kanagawa, T.; Endo, M.; et al. Drug Repositioning for Preeclampsia Therapeutics by In Vitro Screening: Phosphodiesterase-5 Inhibitor Vardenafil Restores Endothelial Dysfunction via Induction of Placental Growth Factor. Reprod. Sci. 2015, 22, 1272–1280. [Google Scholar] [CrossRef]
  82. Hakkim, A.; Fuchs, T.A.; Martinez, N.E.; Hess, S.; Prinz, H.; Zychlinsky, A.; Waldmann, H. Activation of the Raf-MEK-ERK pathway is required for neutrophil extracellular trap formation. Nat. Chem. Biol. 2011, 7, 75–77. [Google Scholar] [CrossRef] [PubMed]
  83. Yu, Y.; Koehn, C.D.; Yue, Y.; Li, S.; Thiele, G.M.; Hearth-Holmes, M.P.; Mikuls, T.R.; O’Dell, J.R.; Klassen, L.W.; Zhang, Z.; et al. Celastrol inhibits inflammatory stimuli-induced neutrophil extracellular trap formation. Curr. Mol. Med. 2015, 15, 401–410. [Google Scholar] [CrossRef] [PubMed]
  84. Lapponi, M.J.; Carestia, A.; Landoni, V.I.; Rivadeneyra, L.; Etulain, J.; Negrotto, S.; Pozner, R.G.; Schattner, M. Regulation of neutrophil extracellular trap formation by anti-inflammatory drugs. J. Pharmacol. Exp. Ther. 2013, 345, 430–437. [Google Scholar] [CrossRef]
  85. Zafarani, A.; Razizadeh, M.H.; Haghi, A. Neutrophil extracellular traps in influenza infection. Heliyon 2023, 9, e23306. [Google Scholar] [CrossRef] [PubMed]
  86. Vorobjeva, N.V.; Pinegin, B.V. Effects of the antioxidants Trolox, Tiron and Tempol on neutrophil extracellular trap formation. Immunobiology 2016, 221, 208–219. [Google Scholar] [CrossRef] [PubMed]
  87. Li, H.; Qiao, C.; Zhao, L.; Jing, Q.; Xue, D.; Zhang, Y. Epigallocatechin-3-gallate reduces neutrophil extracellular trap formation and tissue injury in severe acute pancreatitis. J. Leukoc. Biol. 2022, 112, 1427–1443. [Google Scholar] [CrossRef]
  88. Ostafin, M.; Pruchniak, M.P.; Ciepiela, O.; Reznick, A.Z.; Demkow, U. Different procedures of diphenyleneiodonium chloride addition affect neutrophil extracellular trap formation. Anal. Biochem. 2016, 509, 60–66. [Google Scholar] [CrossRef] [PubMed]
  89. Zhu, W.; Yang, S.; Meng, D.; Wang, Q.; Ji, J. Targeting NADPH Oxidase and Integrin α5β1 to Inhibit Neutrophil Extracellular Traps-Mediated Metastasis in Colorectal Cancer. Int. J. Mol. Sci. 2023, 24, 16001. [Google Scholar] [CrossRef]
  90. Meier, A.; Chien, J.; Hobohm, L.; Patras, K.A.; Nizet, V.; Corriden, R. Inhibition of Human Neutrophil Extracellular Trap (NET) Production by Propofol and Lipid Emulsion. Front. Pharmacol. 2019, 10, 323. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, S.; Zhang, Q.; Wang, F.; Guo, X.; Liu, T.; Zhao, Y.; Gu, B.; Chen, H.; Li, Y. Hydroxychloroquine inhibiting neutrophil extracellular trap formation alleviates hepatic ischemia/reperfusion injury by blocking TLR9 in mice. Clin. Immunol. 2020, 216, 108461, Erratum in Clin. Immunol. 2021, 225, 108681. [Google Scholar] [CrossRef]
  92. Gavillet, M.; Martinod, K.; Renella, R.; Wagner, D.D.; Williams, D.A. A key role for Rac and Pak signaling in neutrophil extracellular traps (NETs) formation defines a new potential therapeutic target. Am. J. Hematol. 2018, 93, 269–276. [Google Scholar] [CrossRef] [PubMed]
  93. Ivey, A.D.; Matthew Fagan, B.; Murthy, P.; Lotze, M.T.; Zeh, H.J.; Hazlehurst, L.A.; Geldenhuys, W.J.; Boone, B.A. Chloroquine reduces neutrophil extracellular trap (NET) formation through inhibition of peptidyl arginine deiminase 4 (PAD4). Clin. Exp. Immunol. 2023, 211, 239–247. [Google Scholar] [CrossRef] [PubMed]
  94. Juha, M.; Molnár, A.; Jakus, Z.; Ledó, N. NETosis: An emerging therapeutic target in renal diseases. Front. Immunol. 2023, 14, 1253667. [Google Scholar] [CrossRef] [PubMed]
  95. Crocetti, L.; Floresta, G.; Cilibrizzi, A.; Giovannoni, M.P. An Overview of PDE4 Inhibitors in Clinical Trials: 2010 to Early 2022. Molecules 2022, 27, 4964. [Google Scholar] [CrossRef] [PubMed]
  96. Wan, T.; Zhang, Y.; Yuan, K.; Min, J.; Mou, Y.; Jin, X. Acetylsalicylic Acid Promotes Corneal Epithelium Migration by Regulating Neutrophil Extracellular Traps in Alkali Burn. Front. Immunol. 2020, 11, 551057. [Google Scholar] [CrossRef] [PubMed]
  97. Jiang, H.; Swacha, P.; Aung, K.M.; Gekara, N.O. Aspirin protects against genotoxicity by promoting genome repair. Cell Res. 2023, 33, 325–327. [Google Scholar] [CrossRef] [PubMed]
  98. Hafkamp, F.M.J.; Mol, S.; Waqué, I.; De Jong, E.C. Dexamethasone, but Not Vitamin D or A, Dampens the Inflammatory Neutrophil Response to Protect At-risk COVID-19 Patients. Immune Netw. 2022, 22, e36. [Google Scholar] [CrossRef] [PubMed]
  99. Singh, H.; Singh, J.R.; Dhillon, V.S.; Bali, D.; Paul, H. In vitro and in vivo genotoxicity evaluation of hormonal drugs. II. Dexamethasone. Mutat. Res. 1994, 308, 89–97. [Google Scholar] [CrossRef] [PubMed]
  100. Ghosh, M.; Biswas, D.; Mukherjee, A. High-altitude medicines: A short-term genotoxicity study. Toxicol. Ind. Health 2010, 26, 417–424. [Google Scholar] [CrossRef]
  101. de Souza Andrade, M.M.; Leal, V.N.C.; Fernandes, I.G.; Gozzi-Silva, S.C.; Beserra, D.R.; Oliveira, E.A.; Teixeira, F.M.E.; Yendo, T.M.; Sousa, M.D.G.T.; Teodoro, W.R.; et al. Resveratrol Downmodulates Neutrophil Extracellular Trap (NET) Generation by Neutrophils in Patients with Severe COVID-19. Antioxidants 2022, 11, 1690. [Google Scholar] [CrossRef]
  102. Fox, J.T.; Sakamuru, S.; Huang, R.; Teneva, N.; Simmons, S.O.; Xia, M.; Tice, R.R.; Austin, C.P.; Myung, K. High-throughput genotoxicity assay identifies antioxidants as inducers of DNA damage response and cell death. Proc. Natl. Acad. Sci. USA 2012, 109, 5423–5428. [Google Scholar] [CrossRef] [PubMed]
  103. Mitruţ, P.; Burada, F.; Enescu, A.; Scorei, R.; Badea, D.; Genunche-Dumitrescu, A.; Rogoz, I.; Manea, M. The genotoxicity study of resveratrol in primary gastric adenocarcinoma cell cultures. Rom. J. Morphol. Embryol. 2009, 50, 429–433. [Google Scholar] [PubMed]
  104. Gupta, A.K.; Giaglis, S.; Hasler, P.; Hahn, S. Efficient neutrophil extracellular trap induction requires mobilization of both intracellular and extracellular calcium pools and is modulated by cyclosporine A. PLoS ONE 2014, 9, e97088. [Google Scholar] [CrossRef] [PubMed]
  105. van Kesteren, P.C.; Beems, R.B.; Luijten, M.; Robinson, J.; de Vries, A.; van Steeg, H. DNA repair-deficient Xpa/p53 knockout mice are sensitive to the non-genotoxic carcinogen cyclosporine A: Escape of initiated cells from immunosurveillance? Carcinogenesis 2009, 30, 538–543. [Google Scholar] [CrossRef] [PubMed]
  106. Bystrzycka, W.; Manda-Handzlik, A.; Sieczkowska, S.; Moskalik, A.; Demkow, U.; Ciepiela, O. Azithromycin and Chloramphenicol Diminish Neutrophil Extracellular Traps (NETs) Release. Int. J. Mol. Sci. 2017, 18, 2666. [Google Scholar] [CrossRef] [PubMed]
  107. Amacher, D.E.; Ellis, J.H., Jr.; Joyce, A.J.; Muehlbauer, P.A.; Turner, G.N.; Wahrenburg, M.G.; Holden, H.E.; Ray, V.A. Preclinical toxicology studies with azithromycin: Genetic toxicology evaluation. Mutat. Res. 1993, 300, 79–90. [Google Scholar] [CrossRef]
  108. Martelli, A.; Mattioli, F.; Pastorino, G.; Robbiano, L.; Allavena, A.; Brambilla, G. Genotoxicity testing of chloramphenicol in rodent and human cells. Mutat. Res. 1991, 260, 65–72. [Google Scholar] [CrossRef] [PubMed]
  109. Menegazzo, L.; Scattolini, V.; Cappellari, R.; Bonora, B.M.; Albiero, M.; Bortolozzi, M.; Romanato, F.; Ceolotto, G.; Vigili de Kreutzeberg, S.; Avogaro, A.; et al. The antidiabetic drug metformin blunts NETosis in vitro and reduces circulating NETosis biomarkers in vivo. Acta Diabetol. 2018, 55, 593–601. [Google Scholar] [CrossRef]
  110. Harishankar, M.K.; Logeshwaran, S.; Sujeevan, S.; Aruljothi, K.N.; Dannie, M.A.; Devi, A. Genotoxicity evaluation of metformin and glimepiride by micronucleus assay in exfoliated urothelial cells of type 2 diabetes mellitus patients. Food Chem. Toxicol. 2015, 83, 146–150. [Google Scholar] [CrossRef]
  111. Cheki, M.; Shirazi, A.; Mahmoudzadeh, A.; Bazzaz, J.T.; Hosseinimehr, S.J. The radioprotective effect of metformin against cytotoxicity and genotoxicity induced by ionizing radiation in cultured human blood lymphocytes. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2016, 809, 24–32. [Google Scholar] [CrossRef]
  112. Besaratinia, A.; Caliri, A.W.; Tommasi, S. Hydroxychloroquine induces oxidative DNA damage and mutation in mammalian cells. DNA Repair 2021, 106, 103180. [Google Scholar] [CrossRef] [PubMed]
  113. Niu, C.; Du, Y.; Kaltashov, I.A. Towards better understanding of the heparin role in NETosis: Feasibility of using native mass spectrometry to monitor interactions of neutrophil elastase with heparin oligomers. Int. J. Mass. Spectrom. 2021, 463, 116550. [Google Scholar] [CrossRef]
  114. Ebeyer-Masotta, M.; Eichhorn, T.; Weiss, R.; Semak, V.; Lauková, L.; Fischer, M.B.; Weber, V. Heparin-Functionalized Adsorbents Eliminate Central Effectors of Immunothrombosis, including Platelet Factor 4, High-Mobility Group Box 1 Protein and Histones. Int. J. Mol. Sci. 2022, 23, 1823. [Google Scholar] [CrossRef] [PubMed]
  115. Jiang, J.; Mu, S.; Zhang, F.; Qiao, Y.; Wu, Y.; Zhang, Z.; Ma, X. Effect of heparin pretreatment on the level of neutrophil extracellular traps of serum and lung tissue in septic mice. Zhonghua Wei Zhong Bing Ji Jiu Yi Xue 2017, 29, 337–341. (In Chinese) [Google Scholar] [CrossRef]
  116. Borelli, G.; Bertoli, D. Toxic and mutagenic effects of a low molecular weight heparin in rats. Arzneimittelforschung 1986, 36, 1256–1260. [Google Scholar]
  117. Bertoli, D.; Borelli, G. Peri- and postnatal, teratology and reproductive studies of a low molecular weight heparin in rats. Arzneimittelforschung 1986, 36, 1260–1263. [Google Scholar]
  118. Irwinda, R.; Hiksas, R.; Lokeswara, A.W.; Wibowo, N. Vitamin D supplementation higher than 2000 IU/day compared to lower dose on maternal-fetal outcome: Systematic review and meta-analysis. Womens Health 2022, 18, 17455057221111066. [Google Scholar] [CrossRef]
  119. Basyreva, L.Y.; Vakhrusheva, T.V.; Letkeman, Z.V.; Maximov, D.I.; Fedorova, E.A.; Panasenko, O.M.; Ostrovsky, E.M.; Gusev, S.A. Effect of Vitamin D3 in combination with Omega-3 Polyunsaturated Fatty Acids on NETosis in Type 2 Diabetes Mellitus Patients. Oxid. Med. Cell Longev. 2021, 2021, 8089696. [Google Scholar] [CrossRef]
  120. Handono, K.; Sidarta, Y.O.; Pradana, B.A.; Nugroho, R.A.; Hartono, I.A.; Kalim, H.; Endharti, A.T. Vitamin D prevents endothelial damage induced by increased neutrophil extracellular traps formation in patients with systemic lupus erythematosus. Acta Med. Indones. 2014, 46, 189–198. [Google Scholar]
  121. Liu, H.; Feng, X.; Wu, S.; Zong, T.; Li, B.; Zhang, Z. Vitamin D Resists Cyclophosphamide-Induced Genomic and DNA Damage in CHL Cells In Vitro and in Mice In Vivo. Nutr. Cancer 2019, 71, 1030–1039. [Google Scholar] [CrossRef]
  122. Adrover, J.M.; Carrau, L.; Daßler-Plenker, J.; Bram, Y.; Chandar, V.; Houghton, S.; Redmond, D.; Merrill, J.R.; Shevik, M.; tenOever, B.R.; et al. Disulfiram inhibits neutrophil extracellular trap formation and protects rodents from acute lung injury and SARS-CoV-2 infection. JCI Insight 2022, 7, e157342. [Google Scholar] [CrossRef]
  123. Zhu, C.; Shi, S.; Jiang, P.; Huang, X.; Zhao, J.; Jin, Y.; Shen, Y.; Zhou, X.; Liu, H.; Cai, J. Curcumin Alleviates Hepatic Ischemia-Reperfusion Injury by Inhibiting Neutrophil Extracellular Traps Formation. J. Investig. Surg. 2023, 36, 2164813. [Google Scholar] [CrossRef]
  124. Mendonça, L.M.; Dos Santos, G.C.; Antonucci, G.A.; Dos Santos, A.C.; Bianchi Mde, L.; Antunes, L.M. Evaluation of the cytotoxicity and genotoxicity of curcumin in PC12 cells. Mutat. Res. 2009, 675, 29–34. [Google Scholar] [CrossRef]
  125. Totani, L.; Amore, C.; Piccoli, A.; Dell’Elba, G.; Di Santo, A.; Plebani, R.; Pecce, R.; Martelli, N.; Rossi, A.; Ranucci, S.; et al. Type-4 Phosphodiesterase (PDE4) Blockade Reduces NETosis in Cystic Fibrosis. Front. Pharmacol. 2021, 12, 702677. [Google Scholar] [CrossRef]
  126. Totani, L.; Piccoli, A.; Dell’Elba, G.; Concetta, A.; Di Santo, A.; Martelli, N.; Federico, L.; Pamuklar, Z.; Smyth, S.S.; Evangelista, V. Phosphodiesterase type 4 blockade prevents platelet-mediated neutrophil recruitment at the site of vascular injury. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 1689–1696. [Google Scholar] [CrossRef]
  127. Dastgheib, M.; Shetab-Boushehri, S.V.; Baeeri, M.; Gholami, M.; Karimi, M.Y.; Hosseini, A. Rolipram and pentoxifylline combination ameliorates experimental diabetic neuropathy through inhibition of oxidative stress and inflammatory pathways in the dorsal root ganglion neurons. Metab. Brain Dis. 2022, 37, 2615–2627. [Google Scholar] [CrossRef]
  128. Silverberg, J.I.; French, L.E.; Warren, R.B.; Strober, B.; Kjøller, K.; Sommer, M.O.A.; Andres, P.; Felding, J.; Weiss, A.; Tutkunkardas, D.; et al. Pharmacology of orismilast, a potent and selective PDE4 inhibitor. J. Eur. Acad. Dermatol. Venereol. 2023, 37, 721–729. [Google Scholar] [CrossRef]
  129. Rizvi, Z.A.; Babele, P.; Sadhu, S.; Madan, U.; Tripathy, M.R.; Goswami, S.; Mani, S.; Kumar, S.; Awasthi, A.; Dikshit, M. Prophylactic treatment of Glycyrrhiza glabra mitigates COVID-19 pathology through inhibition of pro-inflammatory cytokines in the hamster model and NETosis. Front. Immunol. 2022, 13, 945583. [Google Scholar] [CrossRef]
  130. Healy, L.D.; Puy, C.; Fernández, J.A.; Mitrugno, A.; Keshari, R.S.; Taku, N.A.; Chu, T.T.; Xu, X.; Gruber, A.; Lupu, F.; et al. Activated protein C inhibits neutrophil extracellular trap formation in vitro and activation in vivo. J. Biol. Chem. 2017, 92, 8616–8629. [Google Scholar] [CrossRef]
  131. von Dadelszen, P.; Magee, L.A.; Benton, S.J.; Hu, Y.; Ansermino, J.M.; Carleton, B.; Carter, C.; Douglas, M.J.; Janssen, P.A.; Lee, S.K.; et al. Activated protein C as disease-modifying therapy in antenatal preeclampsia: An open-label, single arm safety and efficacy trial. Pregnancy Hypertens. 2018, 13, 121–126. [Google Scholar] [CrossRef]
  132. Huckriede, J.B.; Beurskens, D.M.H.; Wildhagen, K.C.C.A.; Reutelingsperger, C.P.M.; Wichapong, K.; Nicolaes, G.A.F. Design and characterization of novel activated protein C variants for the proteolysis of cytotoxic extracellular histone H3. J. Thromb. Haemost. 2023, 21, 3557–3567. [Google Scholar] [CrossRef] [PubMed]
  133. Xigris 2 Summary of Product Characteristics. Available online: https://www.ema.europa.eu/en/documents/product-information/xigris-epar-product-information_en.pdf (accessed on 16 January 2024).
  134. Baldo, B.A. Enzymes approved for human therapy: Indications, mechanisms and adverse effects. BioDrugs 2015, 29, 31–55. [Google Scholar] [CrossRef]
  135. Jarrahi, A.; Khodadadi, H.; Moore, N.S.; Lu, Y.; Awad, M.E.; Salles, E.L.; Vaibhav, K.; Baban, B.; Dhandapani, K.M. Recombinant human DNase-I improves acute respiratory distress syndrome via neutrophil extracellular trap degradation. J. Thromb. Haemostasis 2023, 21, 2473–2484. [Google Scholar] [CrossRef]
  136. Okur, H.K.; Yalcin, K.; Tastan, C.; Demir, S.; Yurtsever, B.; Karakus, G.S.; Kancagi, D.D.; Abanuz, S.; Seyis, U.; Zengin, R.; et al. Preliminary report of in vitro and in vivo effectiveness of dornase alfa on SARS-CoV-2 infection. New Microbes New Infect. 2020, 37, 100756. [Google Scholar] [CrossRef]
  137. Vural, P.; Akgül, C.; Canbaz, M. Urinary PGE2 and PGF2alpha levels and renal functions in preeclampsia. Gynecol. Obs. Investig. 1998, 45, 237–241. [Google Scholar] [CrossRef]
  138. Shishikura, K.; Horiuchi, T.; Sakata, N.; Trinh, D.A.; Shirakawa, R.; Kimura, T.; Asada, Y.; Horiuchi, H. Prostaglandin E2 inhibits neutrophil extracellular trap formation through production of cyclic AMP. Br. J. Pharmacol. 2016, 173, 319–331. [Google Scholar] [CrossRef]
  139. Domingo-Gonzalez, R.; Martínez-Colón, G.J.; Smith, A.J.; Smith, C.K.; Ballinger, M.N.; Xia, M.; Murray, S.; Kaplan, M.J.; Yanik, G.A.; Moore, B.B. Inhibition of Neutrophil Extracellular Trap Formation after Stem Cell Transplant by Prostaglandin E2. Am. J. Respir. Crit. Care Med. 2016, 193, 186–197. [Google Scholar] [CrossRef]
  140. Baan, R.A.; Stewart, B.W.; Straif, K. (Eds.) Tumour Site Concordance and Mechanisms of Carcinogenesis; International Agency for Research on Cancer: Lyon, France, 2019. [Google Scholar]
  141. Basyreva, L.Y.; Voinova, E.V.; Gusev, A.A.; Mikhalchik, E.V.; Kuskov, A.N.; Goryachaya, A.V.; Gusev, S.A.; Shtilman, M.I.; Velonia, K.; Tsatsakis, A.M. Fluorouracil neutrophil extracellular traps formation inhibited by polymer nanoparticle shielding. Mater. Sci. Eng. C Mater. Biol. Appl. 2020, 108, 110382. [Google Scholar] [CrossRef]
  142. Walsh, S.W.; Strauss, J.F. The Road to Low-Dose Aspirin Therapy for the Prevention of Preeclampsia Began with the Placenta. Int. J. Mol. Sci. 2021, 22, 6985. [Google Scholar] [CrossRef]
  143. Walsh, S.W.; Al Dulaimi, M.; Strauss, J.F. Aspirin Inhibits the Inflammatory Response of Protease-Activated Receptor 1 in Pregnancy Neutrophils: Implications for Treating Women with Preeclampsia. Int. J. Mol. Sci. 2022, 23, 13218. [Google Scholar] [CrossRef] [PubMed]
  144. Christian, F.; Smith, E.L.; Carmody, R.J. The Regulation of NF-κβ Subunits by Phosphorylation. Cells 2016, 5, 12. [Google Scholar] [CrossRef]
  145. Gil-Villa, A.M.; Norling, L.V.; Serhan, C.N.; Cordero, D.; Rojas, M.; Cadavid, A. Aspirin triggered-lipoxin A4 reduces the adhesion of human polymorphonuclear neutrophils to endothelial cells initiated by preeclamptic plasma. Prostaglandins Leukot. Essent. Fat. Acids 2012, 87, 127–134. [Google Scholar] [CrossRef] [PubMed]
  146. Atallah, A.; Lecarpentier, E.; Goffinet, F.; Doret-Dion, M.; Gaucherand, P.; Tsatsaris, V. Aspirin for Prevention of Preeclampsia. Drugs 2017, 77, 1819–1831. [Google Scholar] [CrossRef]
  147. Scott, S.M.; Rose, S.R. Use of Glucocorticoids for the Fetus and Preterm Infant. Clin. Perinatol. 2018, 45, 93–102. [Google Scholar] [CrossRef]
  148. Haram, K.; Svendsen, E.; Abildgaard, U. The HELLP syndrome: Clinical issues and management. A Review. BMC Pregnancy Childbirth 2009, 9, 8. [Google Scholar] [CrossRef]
  149. Tica, O.S.; Tica, A.A.; Cojocaru, D.; Tica, I.; Petcu, C.L.; Cojocaru, V.; Alexandru, D.O.; Tica, V.I. Maternal Steroids on Fetal Doppler Indices, in Growth-Restricted Fetuses with Abnormal Umbilical Flow from Pregnancies Complicated with Early-Onset Severe Preeclampsia. Diagnostics 2023, 13, 428. [Google Scholar] [CrossRef]
  150. Whitelaw, A.; Thoresen, M. Antenatal steroids and the developing brain. Arch. Dis. Child.-Fetal Neonatal Ed. 2000, 83, F154–F157. [Google Scholar] [CrossRef]
  151. Peng, Y.; Hong, H.; Gao, N.; Wan, A.; Ma, Y. Bioinformatics methods in biomarkers of preeclampsia and associated potential drug applications. BMC Genom. 2022, 23, 711. [Google Scholar] [CrossRef] [PubMed]
  152. Causevic, M.; Mohaupt, M. 11beta-Hydroxysteroid dehydrogenase type 2 in pregnancy and preeclampsia. Mol. Asp. Med. 2007, 28, 220–226. [Google Scholar] [CrossRef]
  153. Kisanga, E.P.; Tang, Z.; Guller, S.; Whirledge, S. Glucocorticoid signaling regulates cell invasion and migration in the human first-trimester trophoblast cell line Sw.71. Am. J. Reprod. Immunol. 2018, 80, e12974. [Google Scholar] [CrossRef]
  154. Zhang, D.; Liu, H.; Zeng, J.; Miao, X.; Huang, W.; Chen, H.; Huang, Y.; Li, Y.; Ye, D. Glucocorticoid exposure in early placentation induces preeclampsia in rats via interfering trophoblast development. Gen. Comp. Endocrinol. 2016, 225, 61–70. [Google Scholar] [CrossRef]
  155. Long, J.; Huang, Y.; Wang, G.; Tang, Z.; Shan, Y.; Shen, S.; Ni, X. Mitochondrial ROS Accumulation Contributes to Maternal Hypertension and Impaired Remodeling of Spiral Artery but Not IUGR in a Rat PE Model Caused by Maternal Glucocorticoid Exposure. Antioxidants 2023, 12, 987. [Google Scholar] [CrossRef]
  156. Wan, T.; Zhao, Y.; Fan, F.; Hu, R.; Jin, X. Dexamethasone Inhibits S. aureus-Induced Neutrophil Extracellular Pathogen-Killing Mechanism, Possibly through Toll-Like Receptor Regulation. Front. Immunol. 2017, 8, 60. [Google Scholar] [CrossRef]
  157. Fuchisawa, A.; van Eeden, S.; Magee, L.A.; Whalen, B.; Leung, P.C.; Russell, J.A.; Walley, K.R.; von Dadelszen, P. Neutrophil apoptosis in preeclampsia, do steroids confound the relationship? J. Obstet. Gynaecol. Res. 2004, 30, 342–348. [Google Scholar] [CrossRef]
  158. Tosti, G.; Barberio, A.; Tartaglione, L.; Rizzi, A.; Di Leo, M.; Viti, L.; Sirico, A.; De Carolis, S.; Pontecorvi, A.; Lanzone, A.; et al. Lights and shadows on the use of metformin in pregnancy: From the preconception phase to breastfeeding and beyond. Front. Endocrinol. 2023, 14, 1176623. [Google Scholar] [CrossRef]
  159. Nashif, S.K.; Mahr, R.M.; Jena, S.; Jo, S.; Nelson, A.B.; Sadowski, D.; Crawford, P.A.; Puchalska, P.; Alejandro, E.U.; Gearhart, M.D.; et al. Metformin impairs trophoblast metabolism and differentiation in a dose-dependent manner. Front. Cell Dev. Biol. 2023, 11, 1167097. [Google Scholar] [CrossRef]
  160. He, L.; Wu, X.; Zhan, F.; Li, X.; Wu, J. Protective role of metformin in preeclampsia via the regulation of NF-κβ/sFlt-1 and Nrf2/HO-1 signaling pathways by activating AMPK. Placenta 2023, 143, 91–99. [Google Scholar] [CrossRef] [PubMed]
  161. Dahma, G.; Reddy, G.; Craina, M.; Dumitru, C.; Popescu, A.; Stelea, L.; Neamtu, R.; Gluhovschi, A.; Nitu, R.; Maghiari, A.L.; et al. The Effects of Vitamin D Supplementation before 20 Weeks of Gestation on Preeclampsia: A Systematic Review. J. Pers. Med. 2023, 13, 996. [Google Scholar] [CrossRef]
  162. AlSubai, A.; Baqai, M.H.; Agha, H.; Shankarlal, N.; Javaid, S.S.; Jesrani, E.K.; Golani, S.; Akram, A.; Qureshi, F.; Ahmed, S.; et al. Vitamin D and preeclampsia: A systematic review and meta-analysis. SAGE Open Med. 2023, 11, 20503121231212093. [Google Scholar] [CrossRef]
  163. Tian, X.; Zheng, L.; Ma, J.; Xu, Y.; Zhang, Y.; Pi, Y. Inhibition of LAMP3 mediates the protective effect of vitamin D against hypoxia/reoxygenation in trophoblast cells. Braz. J. Med. Biol. Res. 2023, 56, e12816. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of NET formation. Neutrophils generate NETs in response to various stimuli. Four types of NETs have been described based on structural changes during their formation. These changes involve the activation of different signalling pathways. In the case of mitochondrial NETosis, nuclear DNA remains inside the cell and has been observed in specific cases.
Figure 1. Mechanisms of NET formation. Neutrophils generate NETs in response to various stimuli. Four types of NETs have been described based on structural changes during their formation. These changes involve the activation of different signalling pathways. In the case of mitochondrial NETosis, nuclear DNA remains inside the cell and has been observed in specific cases.
Pharmaceuticals 17 00605 g001
Figure 2. NETs in preeclampsia. In preeclampsia, neutrophils can be activated in two different environments. On the one hand, systemic inflammation with a consequent release of cytokines, DAMPs, and activation of platelets and macrophages stimulates neutrophils, promoting the expression of selectins and their migration to the site of damage, such as the uteroplacental microvasculature where NETs will be released. In addition, neutrophil activation in the peripheral vasculature can lead to the formation of NETs. These networks and their components promote thrombosis, inflammation, damage, and endothelial dysregulation. Likewise, inflammation and ischemia activate neutrophils resident in the uteroplacental tissue, promoting the formation of NETs. These NETs perpetuate and increase inflammation, damaging local tissues and promoting the formation of inflammatory mediators that reach the circulation, attracting more neutrophils.
Figure 2. NETs in preeclampsia. In preeclampsia, neutrophils can be activated in two different environments. On the one hand, systemic inflammation with a consequent release of cytokines, DAMPs, and activation of platelets and macrophages stimulates neutrophils, promoting the expression of selectins and their migration to the site of damage, such as the uteroplacental microvasculature where NETs will be released. In addition, neutrophil activation in the peripheral vasculature can lead to the formation of NETs. These networks and their components promote thrombosis, inflammation, damage, and endothelial dysregulation. Likewise, inflammation and ischemia activate neutrophils resident in the uteroplacental tissue, promoting the formation of NETs. These NETs perpetuate and increase inflammation, damaging local tissues and promoting the formation of inflammatory mediators that reach the circulation, attracting more neutrophils.
Pharmaceuticals 17 00605 g002
Figure 3. Drugs and molecules that reduce neutrophil extracellular traps (NETs). Signalling pathways involved in reducing NETs include Raf-MEK-ERK signalling, calcineurin signalling, phosphorylation of the p65 subunit of NF-κβ via Rac2, PAD4, and block receptors with monoclonal antibodies and DNAases.
Figure 3. Drugs and molecules that reduce neutrophil extracellular traps (NETs). Signalling pathways involved in reducing NETs include Raf-MEK-ERK signalling, calcineurin signalling, phosphorylation of the p65 subunit of NF-κβ via Rac2, PAD4, and block receptors with monoclonal antibodies and DNAases.
Pharmaceuticals 17 00605 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Hernández González, L.L.; Pérez-Campos Mayoral, L.; Hernández-Huerta, M.T.; Mayoral Andrade, G.; Martínez Cruz, M.; Ramos-Martínez, E.; Pérez-Campos Mayoral, E.; Cruz Hernández, V.; Antonio García, I.; Matias-Cervantes, C.A.; et al. Targeting Neutrophil Extracellular Trap Formation: Exploring Promising Pharmacological Strategies for the Treatment of Preeclampsia. Pharmaceuticals 2024, 17, 605. https://doi.org/10.3390/ph17050605

AMA Style

Hernández González LL, Pérez-Campos Mayoral L, Hernández-Huerta MT, Mayoral Andrade G, Martínez Cruz M, Ramos-Martínez E, Pérez-Campos Mayoral E, Cruz Hernández V, Antonio García I, Matias-Cervantes CA, et al. Targeting Neutrophil Extracellular Trap Formation: Exploring Promising Pharmacological Strategies for the Treatment of Preeclampsia. Pharmaceuticals. 2024; 17(5):605. https://doi.org/10.3390/ph17050605

Chicago/Turabian Style

Hernández González, Leticia Lorena, Laura Pérez-Campos Mayoral, María Teresa Hernández-Huerta, Gabriel Mayoral Andrade, Margarito Martínez Cruz, Edgar Ramos-Martínez, Eduardo Pérez-Campos Mayoral, Víctor Cruz Hernández, Ismael Antonio García, Carlos Alberto Matias-Cervantes, and et al. 2024. "Targeting Neutrophil Extracellular Trap Formation: Exploring Promising Pharmacological Strategies for the Treatment of Preeclampsia" Pharmaceuticals 17, no. 5: 605. https://doi.org/10.3390/ph17050605

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop