Next Article in Journal
Establishment of Canine Oral Mucosal Melanoma Cell Lines and Their Xenogeneic Animal Models
Previous Article in Journal
Mesenchymal Stem Cell-Based Therapies for Temporomandibular Joint Repair: A Systematic Review of Preclinical Studies
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation

1
Department of Pediatric Surgery, University Medical Center Mannheim, University of Heidelberg, 68167 Mannheim, Germany
2
Department of Internal Medicine 3—Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
3
Deutsches Zentrum für Immuntherapie (DZI), Friedrich Alexander University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, 91054 Erlangen, Germany
*
Author to whom correspondence should be addressed.
Cells 2024, 13(11), 991; https://doi.org/10.3390/cells13110991
Submission received: 7 April 2024 / Revised: 27 May 2024 / Accepted: 5 June 2024 / Published: 6 June 2024
(This article belongs to the Special Issue Formation, Aggregation, Persistence, and Maturation of NETs)

Abstract

:
Post-surgical abdominal adhesions, although poorly understood, are highly prevalent. The molecular processes underlying their formation remain elusive. This review aims to assess the relationship between neutrophil extracellular traps (NETs) and the generation of postoperative peritoneal adhesions and to discuss methods for mitigating peritoneal adhesions. A keyword or medical subject heading (MeSH) search for all original articles and reviews was performed in PubMed and Google Scholar. It included studies assessing peritoneal adhesion reformation after abdominal surgery from 2003 to 2023. After assessing for eligibility, the selected articles were evaluated using the Critical Appraisal Skills Programme checklist for qualitative research. The search yielded 127 full-text articles for assessment of eligibility, of which 7 studies met our criteria and were subjected to a detailed quality review using the Critical Appraisal Skills Programme (CASP) checklist. The selected studies offer a comprehensive analysis of adhesion pathogenesis with a special focus on the role of neutrophil extracellular traps (NETs) in the development of peritoneal adhesions. Current interventional strategies are examined, including the use of mechanical barriers, advances in regenerative medicine, and targeted molecular therapies. In particular, this review emphasizes the potential of NET-targeted interventions as promising strategies to mitigate postoperative adhesion development. Evidence suggests that in addition to their role in innate defense against infections and autoimmune diseases, NETs also play a crucial role in the formation of peritoneal adhesions after surgery. Therefore, therapeutic strategies that target NETs are emerging as significant considerations for researchers. Continued research is vital to fully elucidate the relationship between NETs and post-surgical adhesion formation to develop effective treatments.

Graphical Abstract

1. Background

As many as 93–100% of patients develop adhesions after abdominal surgery, which can result in intestinal obstruction, pelvic pain, and infertility complications [1]. These adhesions may respond naturally to tissue injury during surgery, inflammation, or infection [2,3]. The formation of adhesions is a complex process involving multiple cellular and molecular events, including coagulation, fibrinolysis, inflammation, angiogenesis, and extracellular matrix (ECM) remodeling [3,4]. Neutrophil extracellular traps (NETs) are structures composed of deoxyribonucleic acid (DNA), histones, and granule proteins that are released by neutrophils [5]. NET formation is a unique process in immune defense in humans. NETs were initially discovered as a defense mechanism against microbial infections as they can trap and neutralize pathogens [6]. However, recent research has suggested that NETs can also play a role in non-infectious conditions, including inflammation, autoimmune diseases, and tissue repair [7]. The relationship between NETs and adhesions lies in their involvement in the host response to tissue injury and inflammation [8]. When tissue injury occurs, neutrophils are rapidly recruited to the site of the injury or infection [9]. Upon activation, neutrophils can release NETs to neutralize pathogens; however, NETs can also contribute to tissue damage and inflammation due to cytotoxic components [10,11]. Several studies suggest that the recruitment and activation of neutrophils at the site of injury may contribute to adhesion formation through the release of pro-inflammatory mediators and the promotion of fibroblast activation [12,13,14], raising further questions on their particular significance. Duan Z et al.’s work revealed that measuring the neutrophil extracellular trap formation index (NFI) in postoperative drainage fluid offers a more sensitive and specific method for early prediction of deep surgical site infections compared to traditional serum infection indicators like CRP and PCT [15]. Consequently, the objective of this review was to examine research focusing on the role that neutrophils play in abdominal adhesions. Understanding this relationship could lead to novel therapeutic strategies for preventing or treating postoperative adhesions.

2. Review

2.1. Search Strategy

Search strategy and selection criteria: This review was performed according to PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines [16]. Electronic databases, including PubMed and Google Scholar, were systematically searched using the keywords or medical subject headings (MeSHs) “Tissue Adhesion” (or “Surgical Adhesion”, “Surgery-Induced Tissue Adhesions”, “Surgical Adhesion”, “Surgery-Induced Tissue Adhesion”) and “Extracellular Traps” (or “Neutrophil Extracellular Traps”, “Extracellular DNA Traps”, “NETs (Neutrophil Extracellular Traps)”). The literature search was conducted with a timeframe from January 2003 to May 2023 and the search was not restricted to specific languages. The following selection process was conducted, according to the PRISMA 2020 checklist [16] (see also Figure 1): In the first step, duplicates were removed. In the next step, studies were screened based on the abstract only by an independent reviewer and finally included if they contained original data relevant to the topic. Next, a full-text article assessment was performed for the remaining articles, and those with an inadequate methodology and insufficient data and those lacking adhesion markers were eliminated according to the Cochrane Risk of Bias Tool [17]. In the last step, a quality assessment was performed for the remaining articles, adhering to the Critical Appraisal Skills Programme (2023) (CASP) qualitative study checklist [18]; see Table 1.

2.2. Screening and Quality Assessment

Record identification and removal of duplicates: An initial search conducted in the PubMed and Google Scholar databases yielded 912 records. The removal of duplicates reduced the pool to 709 unique articles. Abstract screening: Subsequent screening of the titles and abstracts led to the exclusion of 582 articles. The exclusion criteria were irrelevance to the research question and non-adherence to the predefined inclusion criteria; see Figure 1. Full-text article eligibility assessment: Further evaluation of the full-text articles resulted in the exclusion of 109 records due to inadequate methodological description. Seven studies were excluded for providing insufficient data, which raised concerns about their reproducibility and reliability. Four studies were excluded because they did not utilize specific markers for adhesion; see Figure 1.
Quality Assessment: A detailed quality assessment was conducted on the remaining seven studies using the Critical Appraisal Skills Programme (CASP) checklist [26]. Overall, most studies conformed to the majority of the checklist criteria; see Table 1.
Results: These 7 studies explore various aspects of NET formation and its implications, illustrating a complex interplay of factors contributing to adhesion formation. One study illustrates how mechanical injury to the mesothelial layer during abdominal surgery leads to the recruitment of neutrophils and monocytes, whose formation of NETs is implicated in the pathogenesis of peritoneal adhesions, suggesting that modulating these immune responses may prevent such occurrences [25]. Additionally, research involving gastric cancer patients post-surgery shows that increased NET production by low-density neutrophils facilitates tumor cell attachment and growth, proposing that DNase treatment to disrupt these NETs could prevent peritoneal recurrence [22]. Moreover, investigations reveal that early-recruited neutrophils release NETs that activate the STING-associated inflammatory response, with interventions that disrupt NETs and inhibit STING signaling markedly reducing adhesion burden, thus identifying NET/STING pathways as potential therapeutic targets [24]. Similarly, evidence from murine models indicates that NETs are crucial in forming peritoneal adhesions, with DNase treatments significantly reducing these adhesions, suggesting potential therapeutic approaches pending further clinical validation [23]. Furthermore, another study demonstrates that mesothelial cells in the peritoneal cavity can undergo a mesothelial-to-mesenchymal transition, contributing to the formation of adhesions, with the blockade of the transforming growth factor beta (TGF-β) pathway offering a novel approach to mitigate these effects [21]. In parallel, research on peritoneal fibrosis highlights the role of inflammation and injury from clinical events like surgery in driving fibrosis through complex interactions among myofibroblasts, leukocytes, and other cell types, contributing to the deterioration of the peritoneal membrane [20]. Lastly, a study on the combined treatment with antithrombin and a PAD4 inhibitor in mice suggests that this approach effectively reduces postoperative adhesion formation by mitigating thrombin and NET-related processes, offering promising insights into managing surgical outcomes [19]. Collectively, these studies not only underscore the multifaceted role of NETs in peritoneal adhesion formation but also open avenues for targeted interventions that could ameliorate or prevent the adverse effects of surgical interventions.
Four main parts will be discussed in more detail in the following paragraphs: the pathogenesis of adhesions, the prevention and treatment of NETs, the potential impact of NET inhibition on adhesion formation, and a feasibility analysis of suppressing NETs in post-surgical adhesion.

2.3. Pathogenesis of Adhesions

Adhesions are fibrous bands between internal organs [27]. They form due to events like coagulation, inflammation, and angiogenesis [3,28]. Adhesions can result from surgery, trauma, or inflammatory conditions [28,29]. Upon tissue injury, the body initiates a complex response beginning with the activation of the coagulation cascade. This cascade results in fibrin production, forming a temporary matrix that not only stabilizes the wound but also prevents bleeding [30,31,32]. As a result of injury, inflammatory mediators are released, including cytokines, chemokines, and growth factors, which promote the recruitment of immune cells such as neutrophils and macrophages to the injured area [33,34,35,36]. Another critical element in this process is the mesothelium, a protective layer of cells lining the internal organs and providing a non-adhesive surface [37]. However, when surgical injuries disrupt this layer, there is a significant risk of adhesion formation, highlighting the delicate balance within the body’s internal environment [37]. This disruption leads to a phenomenon known as mesothelial-to-mesenchymal transition (MMT), whereby mesothelial cells acquire a fibroblast-like phenotype. These transformed cells then produce extracellular matrix (ECM) components, further contributing to adhesion formation [37,38,39]. Interestingly, while fibrin deposition acts as a supportive matrix for cell migration and temporary wound stabilization, it also plays a pivotal role in adhesion formation [40,41]. Under normal conditions, this fibrin matrix is gradually degraded by fibrinolytic enzymes like plasmin, which serves to prevent excessive adhesion formation [42,43]. However, an imbalance between fibrin deposition and fibrinolysis can lead to the persistence of fibrin, thereby promoting adhesion [43]. Furthermore, angiogenesis emerges as a critical factor in the development of adhesions. It ensures the supply of oxygen and nutrients to the growing fibrous tissue, underscoring the complexity of adhesion development [44,45]. Angiogenesis is regulated by various pro- and antiangiogenic factors such as vascular endothelial growth factor (VEGF) [46,47] and thrombospondin-1 [48,49,50].
A summary of the pathogenesis, mechanisms, and interventional strategies of adhesion formation can be found in Table 2.

2.4. NET Formation and Degradation

In the realm of innate immunity, neutrophil extracellular traps (NETs) serve as a frontline defense mechanism against invading pathogens. These web-like structures are released by activated neutrophils through a dynamic sequence of events, including nuclear decondensation, cytoplasmic content release, and ultimately, cell rupture—a process collectively referred to as NETosis [93,94,95]. Distinct from traditional apoptosis, NETosis is characterized by the explosive release of cellular components that ensnare and neutralize pathogens [96,97,98,99]. The nomenclature “NETosis” merges “NET” with “apoptosis,” highlighting its deviation from conventional programmed cell death mechanisms. Within the spectrum of NETosis, two primary forms are recognized: suicidal and vital NETosis [100]. Suicidal NETosis, or lytic NETosis, primarily involves a cell death program driven by the production of reactive oxygen species (ROS), either through an NADPH oxidase (NOX)-dependent pathway or via mitochondrial-derived ROS (mROS) from a NOX-independent pathway [101]. Intriguingly, suicidal NETosis can also occur without the activation of NOX2, driven instead by extracellular Ca2+ influx, which can be stimulated by agents like fungal ionophores (e.g., nigericin, ionomycin) and granulocyte macrophage colony-stimulating factor (GM-CSF) [3].
In contrast, vital NETosis does not result in neutrophil death. This pathway allows neutrophils to release NETs while remaining alive and functionally active, which is crucial during acute infections where a rapid immune response is necessary. Both mechanisms have been well documented but serve different roles depending on the inflammatory or infectious context, including aseptic conditions as opposed to responses to microbial infections or in autoimmune diseases [100,102].
Specifically, the initiation of NET formation is triggered by various stimuli, including pathogens and immune mediators such as interleukin-8 (IL-8) and lipopolysaccharide (LPS) [103,104,105]. The process involves protein kinase C (PKC) activation and reactive oxygen species (ROS) production via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase [98,106,107]. Subsequently, increased ROS activate enzymes like neutrophil elastase (NE), leading to chromatin decondensation aided by myeloperoxidase (MPO) [108,109]. This change, along with the formation of pores by gasdermin D (GSDMD), results in the formation of NETs [110,111,112].
Peptidyl arginine deiminase 4 (PAD4) converts arginine residues on histones (proteins that help compact DNA) into citrulline, a process known as citrullination [113]. DNA is unfolded or decondensed as a result of this alteration in the charge distribution of histones [114]. As a result, the transcription machinery can access DNA more easily, which in turn affects gene expression [115]. This chromatin decondensation process is essential for NET formation, as it allows for the re-assembling of nuclear components with cytoplasmic granule proteins. Concurrently, GSDMD, a pore-forming protein, is cleaved by caspases to form pores in the plasma membrane [116]. Through these pores, the decondensed chromatin and proteins are ejected, which then form NETs that trap and neutralize pathogens.
However, the degradation of NETs is crucial for resolving inflammation and preventing pathologies such as tissue adhesion [117,118,119]. The primary mechanism by which NETs are degraded involves the action of nucleases, primarily deoxyribonucleases (DNases), which cleave their DNA backbone of NETs [22,119]. Table 3 summarizes the critical factors involved in NET degradation.

2.5. The Role of NETs in Adhesion Development

Neutrophils, especially NETs, may be essential during peritoneal adhesion. Recently, it has been suggested that NETs serve as important scaffolds for adhesion formation [8,138]. NETs may affect peritoneal adhesions at various levels. Post-surgical adhesion formation is a complex process involving multiple cell types and signaling pathways [3]. This complexity originates from the consistent recruitment of immune cells [25,139] coupled with the unchecked proliferation of fibroblasts [140,141] and mesothelial cells [142]. Neutrophils play a pivotal role in the early stages of adhesion formation, acting as key mediators in this complex physiological process. Dominant within the initial adhesive environment, neutrophils regulate the release of vital mediators such as IL-8, interleukin-1 beta (IL-1β), and ROS [143]. An essential pathway of note in this context is the ROS signaling pathway. For instance, transmembrane and immunoglobulin domain-containing 1 (TMIGD1) has been shown to thwart abdominal adhesion formation by mitigating oxidative stress in the mitochondria of peritoneal mesothelial cells [144]. Neutrophil extracellular traps (NETs) play an important role in this landscape. The concentration of NETs peaks during the 1–3 day window following surgery [25], and their persistence, extending beyond the lifespan of neutrophils, imparts a prolonged functional importance that persists even after neutrophil apoptosis [145]. However, it is the complex interplay between NETs and various cellular components that emphasizes their collective importance in the postoperative milieu. Central to this interaction is the concept of pathway interdependence, which accentuates the synergistic relationships between cellular processes and mediators in the development of post-surgical adhesions. For example, the ROS interplay with NET formation is diverse, influencing whether NET formation manifests beneficial or detrimental effects. Excessive ROS, produced during neutrophil activation, are known to trigger NET formation by causing severe DNA damage, like oxidizing guanine to 8-oxo guanine [146]. This eventually initiates a DNA repair pathway, resulting in chromatin decondensation.
In the context of peritoneal adhesions, NET formation predominantly occurs within the peritoneal cavity, particularly proximal to post-capillary venules and at locations affected by surgical interventions or tissue injuries [5]. This spatial distribution is especially pertinent during abdominal surgeries, where neutrophils are stimulated to release NETs in reaction to tissue damage and the presence of foreign materials such as surgical sutures. The formation of NETs at these sites initiates a pro-inflammatory cascade that facilitates critical processes like fibrin deposition and fibroblast activation, which are essential for the development of peritoneal adhesions [147]. Furthermore, the process of histone citrullination, a modification associated with NET formation, serves as a precise biomarker for this phenomenon [5]. What is more, the level of H3cit (citrullinated histone H3) was developed as a biomarker for some diseases associated with NETs, like arterial thromboembolism (ATE) [148], tumors [115], and abdominal aortic aneurysms [149]. The quantification of histone citrullination offers a direct measure of NET activity, thereby linking NET formation to the progression of surgical adhesions. This biomarker provides an invaluable metric for both researching the underlying mechanisms of adhesion formation and evaluating clinical interventions aimed at modulating NET formation to mitigate its pathological effects.
After abdominal surgery, neutrophils are activated to produce NETs through a complex interplay of physiological and biochemical processes. Surgical trauma and tissue damage result in the release of damage-associated molecular patterns (DAMPs), which are recognized by pattern recognition receptors (PRRs) on neutrophils, initiating their activation. The subsequent inflammatory response is characterized by the release of cytokines (e.g., IL-1β, IL-6, TNF-α) and chemokines (e.g., IL-8, CXCL1) [100,150], which prime neutrophils and enhance their sensitivity to stimuli that induce NET formation. Additionally, the potential for contamination or infection during surgery introduces pathogen-associated molecular patterns (PAMPs) that further activate neutrophils through PRRs [151]. Activated platelets, a common occurrence due to tissue injury and bleeding, release mediators such as platelet factor 4 (PF4) and CD40L, and directly interact with neutrophils via surface molecules, thereby promoting NET formation [152]. ROS produced by neutrophils upon activation, alongside intracellular signaling pathways involving enzymes like NADPH oxidase and MPO, contribute to chromatin decondensation and the active release of NETs through NET formation [10]. And many new triggers have been discovered, including those involved in sterile inflammation [153]. Moreover, a recent study by Pandolfi, Laura et al. suggests that neutrophil extracellular traps (NETs), induced by SARS-CoV-2 and combined with factors secreted by alveolar macrophages, can drive the epithelial–mesenchymal transition (EMT) in lung epithelial cells, potentially leading to lung fibrosis in severe COVID-19 patients [154]. Since epithelial cells are also involved in adhesion formation, EMT induced by NET formation might similarly contribute to adhesion development.

2.6. The Potential Impact of NET Inhibition on Adhesion Formation

Understanding the inhibition of NETs is crucial for developing therapeutic strategies to modulate their formation in pathological conditions, where excessive or dysregulated NET formation contributes to disease progression [155]. Various inhibitors and molecular targets that can effectively inhibit NET formation have been identified. In brief, NETs and adhesions are implicated in the inflammatory and healing responses to tissue injury; NETs, released by neutrophils, can induce fibroblast activity and thrombosis [156], thus contributing to adhesion between organs. The dysregulation of NET formation or removal plays a critical role in abnormal tissue repair, potentially exacerbating adhesion development [157]. A summary of the role of NETs in peritoneal adhesion formation is summarized in Table 4.

2.7. Feasibility Analysis of Suppressing NETs in Post-Surgical Adhesion

It is essential to note that the choice of inhibitor and its efficacy may depend on the context of a specific disease or inflammatory condition. NETs are a component of the complex immune response during tissue injury and inflammation. Therefore, the effect of inhibiting NETs on adhesion formation may be context-dependent and varies based on specific surgical or pathological conditions. More research is required to directly investigate the relationship between NET inhibition and adhesion formation and to determine whether targeting NETs could be a viable strategy for adhesion prevention while minimizing the adverse effects.
The role of NETs (neutrophil extracellular traps) in wound healing, especially within the context of bowel anastomosis, remains an underexplored area of research. However, emerging studies underscore a significant interplay between NETs and the wound healing process [157]. A pivotal study published in Nature Medicine in 2015 by Wong et al. illustrated that the presence of diabetes increases the formation of NETs within wounds, prolonging the healing process in diabetic wounds [174]. Concurrently, another investigation revealed that during lung ischemia–reperfusion, the release of mitochondrial DNA (mtDNA) induces the formation of TLR9-mediated NETs, intensifying lung injury [175]. Given that injuries from bowel anastomosis are essentially ischemic reperfusion injuries or arise from tissue hypoxia [176], it is possible that the generation of NETs has a negative impact on the repair of tissues affected by ischemia and hypoxia. Contrarily, in vitro analyses have indicated that under suboptimal concentrations, NETs might bolster the proliferation of keratinocytes, driven by the activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway [177]. This suggests a concentration-dependent regulatory role of NETs in wound healing. This observation aligns with the findings of Saffarzadeh et al. which elucidated that the cytotoxic effects of NETs on epithelial and endothelial cells are predominantly determined by concentration levels [178]. Interestingly, this cytotoxicity is not modulated by DNA fragments but is orchestrated by histones and MPO, while NE does not confer any cytotoxic effects. Histones, as primary constituents of NETs, act as damage-associated molecular patterns (DAMPs) and are hypothesized to be cytotoxic to epithelial and endothelial cells. Additionally, proteins released in conjunction with NETs, such as NE and matrix metalloproteinase (MMP), might impede wound healing by detrimentally affecting the ECM [179]. In summation, while the antimicrobial attributes of NETs offer a positive influence on wound healing, their deleterious effects, especially in conditions like diabetes, are pronounced and frequently correlate with hyperactive NET formation. These findings are summarized in Table 5.
NETS exhibit a bifunctional role in both wound healing and broader physiological responses, illustrating the multifaceted nature of their biological effects. Excessive NET production may precipitate augmented adhesion formation, suggesting that their inhibition could be beneficial. Conversely, it is conceivable that NET suppression might impede wound healing, for instance, in the context of postoperative recovery. Within this complex interplay, the process of anastomosis stands out as a critical determinant of surgical recuperation and sustained patient prognosis. Anastomosis, entailing the surgical juncture of two bodily passages, is essential for re-establishing physiological continuity post-surgery. Anastomotic insufficiencies pose a critical concern in surgical outcomes, indicating the significance of intricate biological processes beyond the well-recognized ischemia–reperfusion injury [196]. The intricate interplay of the gut microbiome in surgical recovery is emerging as a critical factor [197]. This complex network of commensal bacteria, predominantly housed in the colon’s mucus layer, plays a pivotal role in various metabolic and immune processes [198]. Perioperative interventions, such as bowel preparation, antibiotic administration, fasting, and the stress of surgery itself, can severely disrupt this delicate microbial balance [199]. Such disruptions, known as dysbiosis, may lead to nutrient depletion, diminished microbial diversity, and consequently, compromised anastomotic healing due to increased susceptibility to pathogenic bacteria [200]. Moreover, an altered gut microbiome can promote NET formation, a hypothesis supported by current studies [201,202,203]. This is mediated through various mechanisms: microbial metabolites like short-chain fatty acids (SCFAs) modulate immune cell activity [204,205]; dysbiosis increases intestinal permeability, enhancing exposure to pro-inflammatory stimuli like lipopolysaccharides (LPSs) that trigger NET release [206,207,208]; and pathogenic microbes can directly stimulate NET formation via interactions with neutrophil pattern recognition receptors (PRRs) [209].
For these reasons, a cautious approach is warranted. Selective inhibition targeting modulators that minimally affect granule proteins, such as myeloperoxidase (MPO) or neutrophil elastase (NE), may represent a judicious strategy. Previous research highlights the role of neutrophil extracellular traps (NETs) in providing a scaffold for adhesion formation, with DNase administration shown to disrupt this process effectively [23]. Consequently, therapeutic applications of DNases could offer a viable intervention to prevent peritoneal adhesion without hindering the normal healing process.

3. Conclusions

Throughout this review, the involvement of neutrophil extracellular traps (NETs) in the development of postoperative abdominal adhesions has been critically examined. Emerging evidence suggests that interventions targeting NETs could represent a novel therapeutic pathway for the prevention of adhesions. However, the potential effect of NET inhibitors on essential healing processes necessitates further in-depth research. Future studies must delineate NETs’ precise role in the formation of adhesions and rigorously evaluate the therapeutic effectiveness and safety of NET-targeted therapies. Advancing our understanding of NET-mediated adhesion could lead to innovative strategies that reduce surgical adhesion-related complications.

Author Contributions

Conceptualization, Y.L., J.E., M.H., J.K. and M.B.; methodology, Y.L., J.E., M.H., J.K. and M.B.; validation and formal analysis, M.H. and M.B.; investigation, Y.L. and J.E.; resources, M.B.; writing—original draft preparation, Y.L., J.E. and M.H.; writing—review and editing, J.K. and M.B.; visualization, Y.L.; supervision, M.H., J.K. and M.B. All authors have read and agreed to the published version of the manuscript.

Funding

This study received no external funding.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. ten Broek, R.P.; Strik, C.; Issa, Y.; Bleichrodt, R.P.; van Goor, H. Adhesiolysis-related morbidity in abdominal surgery. Ann. Surg. 2013, 258, 98–106. [Google Scholar] [CrossRef]
  2. Capella-Monsonís, H.; Kearns, S.; Kelly, J.; Zeugolis, D.I. Battling adhesions: From understanding to prevention. BMC Biomed. Eng. 2019, 1, 5. [Google Scholar] [CrossRef]
  3. Fatehi Hassanabad, A.; Zarzycki, A.N.; Jeon, K.; Deniset, J.F.; Fedak, P.W.M. Post-Operative Adhesions: A Comprehensive Review of Mechanisms. Biomedicines 2021, 9, 867. [Google Scholar] [CrossRef]
  4. Moretti, L.; Stalfort, J.; Barker, T.H.; Abebayehu, D. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J. Biol. Chem. 2022, 298, 101530. [Google Scholar] [CrossRef]
  5. Papayannopoulos, V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 2018, 18, 134–147. [Google Scholar] [CrossRef]
  6. Urban, C.F.; Ermert, D.; Schmid, M.; Abu-Abed, U.; Goosmann, C.; Nacken, W.; Brinkmann, V.; Jungblut, P.R.; Zychlinsky, A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009, 5, e1000639. [Google Scholar] [CrossRef]
  7. Fousert, E.; Toes, R.; Desai, J. Neutrophil Extracellular Traps (NETs) Take the Central Stage in Driving Autoimmune Responses. Cells 2020, 9, 915. [Google Scholar] [CrossRef]
  8. Boettcher, M.; Schönfeld, L.; Heuer, A.; Elrod, J.; Stiel, C.; Raluy, L.P.; Trochimiuk, M.; Appl, B.; Saygi, C.; Zlatar, L. Neutrophil extracellular traps orchestrate formation of peritoneal adhesions. J. Sci. 2021, 26, 108289. [Google Scholar]
  9. Wang, J. Neutrophils in tissue injury and repair. Cell Tissue Res. 2018, 371, 531–539. [Google Scholar] [CrossRef]
  10. Delgado-Rizo, V.; Martínez-Guzmán, M.A.; Iñiguez-Gutierrez, L.; García-Orozco, A.; Alvarado-Navarro, A.; Fafutis-Morris, M. Neutrophil Extracellular Traps and Its Implications in Inflammation: An Overview. Front. Immunol. 2017, 8, 81. [Google Scholar] [CrossRef]
  11. Yousefi, S.; Simon, D.; Stojkov, D.; Karsonova, A.; Karaulov, A.; Simon, H.U. In vivo evidence for extracellular DNA trap formation. Cell Death Dis. 2020, 11, 300. [Google Scholar] [CrossRef] [PubMed]
  12. de Oliveira, S.; Rosowski, E.E.; Huttenlocher, A. Neutrophil migration in infection and wound repair: Going forward in reverse. Nat. Rev. Immunol. 2016, 16, 378–391. [Google Scholar] [CrossRef] [PubMed]
  13. Herrero-Cervera, A.; Soehnlein, O.; Kenne, E. Neutrophils in chronic inflammatory diseases. Cell. Mol. Immunol. 2022, 19, 177–191. [Google Scholar] [CrossRef] [PubMed]
  14. Peiseler, M.; Kubes, P. More friend than foe: The emerging role of neutrophils in tissue repair. J. Clin. Investig. 2019, 129, 2629–2639. [Google Scholar] [CrossRef] [PubMed]
  15. Duan, Z.; Sun, S.; Qu, C.; Wang, K.; Chen, F.; Wang, X.; Chu, C.; Liu, B.; Li, J.; Ding, W. Neutrophil extracellular trap formation index predicts occurrences of deep surgical site infection after laparotomy. Ann. Transl. Med. 2021, 9, 1373. [Google Scholar] [CrossRef] [PubMed]
  16. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. Syst. Rev. 2021, 10, 89. [Google Scholar] [CrossRef] [PubMed]
  17. Ma, L.-L.; Wang, Y.-Y.; Yang, Z.-H.; Huang, D.; Weng, H.; Zeng, X.-T. Methodological quality (risk of bias) assessment tools for primary and secondary medical studies: What are they and which is better? Mil. Med. Res. 2020, 7, 7. [Google Scholar] [CrossRef] [PubMed]
  18. The Creativity, Activity and Service Program. CASP Qualitative Studies Checklist; CASP UK: Oxford, UK, 2023. [Google Scholar]
  19. Sudo, M.; Xu, J.; Mitani, K.; Jimbo, M.; Tsutsui, H.; Hatano, E.; Fujimoto, J. Antithrombin Together with NETs Inhibitor Protected Against Postoperative Adhesion Formation in Mice. Cell Physiol. Biochem. 2021, 55, 400–412. [Google Scholar] [CrossRef]
  20. Terri, M.; Trionfetti, F.; Montaldo, C.; Cordani, M.; Tripodi, M.; Lopez-Cabrera, M.; Strippoli, R. Mechanisms of Peritoneal Fibrosis: Focus on Immune Cells-Peritoneal Stroma Interactions. Front. Immunol. 2021, 12, 607204. [Google Scholar] [CrossRef]
  21. Sandoval, P.; Jiménez-Heffernan, J.A.; Guerra-Azcona, G.; Pérez-Lozano, M.L.; Rynne-Vidal, Á.; Albar-Vizcaíno, P.; Gil-Vera, F.; Martín, P.; Coronado, M.J.; Barcena, C.; et al. Mesothelial-to-mesenchymal transition in the pathogenesis of post-surgical peritoneal adhesions. J. Pathol. 2016, 239, 48–59. [Google Scholar] [CrossRef]
  22. Kanamaru, R.; Ohzawa, H.; Miyato, H.; Matsumoto, S.; Haruta, H.; Kurashina, K.; Saito, S.; Hosoya, Y.; Yamaguchi, H.; Yamashita, H.; et al. Low density neutrophils (LDN) in postoperative abdominal cavity assist the peritoneal recurrence through the production of neutrophil extracellular traps (NETs). Sci. Rep. 2018, 8, 632. [Google Scholar] [CrossRef] [PubMed]
  23. Elrod, J.; Heuer, A.; Knopf, J.; Schoen, J.; Schönfeld, L.; Trochimiuk, M.; Stiel, C.; Appl, B.; Raluy, L.P.; Saygi, C.; et al. Neutrophil extracellular traps and DNases orchestrate formation of peritoneal adhesions. iScience 2023, 26, 108289. [Google Scholar] [CrossRef] [PubMed]
  24. Hu, Q.; Shen, X.; Wang, M.; Lu, X.; Liu, S.; Guan, W. NETs Enhance STING To Promote Surgical Adhesion. Res. Squre 2021. preprint. [Google Scholar]
  25. Tsai, J.M.; Shoham, M.; Fernhoff, N.B.; George, B.M.; Marjon, K.D.; McCracken, M.N.; Kao, K.S.; Sinha, R.; Volkmer, A.K.; Miyanishi, M.; et al. Neutrophil and monocyte kinetics play critical roles in mouse peritoneal adhesion formation. Blood Adv. 2019, 3, 2713–2721. [Google Scholar] [CrossRef] [PubMed]
  26. Greenhalgh, T.; Taylor, R. Papers that go beyond numbers (qualitative research). BMJ 1997, 315, 740–743. [Google Scholar] [CrossRef] [PubMed]
  27. Stapleton, L.M.; Lucian, H.J.; Grosskopf, A.K.; Smith, A.A.; Totherow, K.P.; Woo, Y.J.; Appel, E.A. Dynamic hydrogels for prevention of post-operative peritoneal adhesions. Adv. Ther. 2021, 4, 2000242. [Google Scholar] [CrossRef]
  28. Bianchi, E.; Boekelheide, K.; Sigman, M.; Lamb, D.J.; Hall, S.J.; Hwang, K. Ghrelin inhibits post-operative adhesions via blockage of the TGF-β signaling pathway. PLoS ONE 2016, 11, e0153968. [Google Scholar] [CrossRef]
  29. Zwicky, S.N.; Stroka, D.; Zindel, J. Sterile injury repair and adhesion formation at serosal surfaces. Front. Immunol. 2021, 12, 684967. [Google Scholar] [CrossRef]
  30. Palta, S.; Saroa, R.; Palta, A. Overview of the coagulation system. Indian J. Anaesth. 2014, 58, 515–523. [Google Scholar] [CrossRef]
  31. Periayah, M.H.; Halim, A.S.; Mat Saad, A.Z. Mechanism Action of Platelets and Crucial Blood Coagulation Pathways in Hemostasis. Int. J. Hematol. Oncol. Stem. Cell Res. 2017, 11, 319–327. [Google Scholar]
  32. Laurens, N.; Koolwijk, P.; de Maat, M.P. Fibrin structure and wound healing. J. Thromb. Haemost. 2006, 4, 932–939. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, J.M.; An, J. Cytokines, inflammation, and pain. Int. Anesthesiol. Clin. 2007, 45, 27–37. [Google Scholar] [CrossRef] [PubMed]
  35. Su, Y.; Gao, J.; Kaur, P.; Wang, Z. Neutrophils and Macrophages as Targets for Development of Nanotherapeutics in Inflammatory Diseases. Pharmaceutics 2020, 12, 1222. [Google Scholar] [CrossRef] [PubMed]
  36. Hartl, J.; Serpas, L.; Wang, Y.; Rashidfarrokhi, A.; Perez, O.A.; Sally, B.; Sisirak, V.; Soni, C.; Khodadadi-Jamayran, A.; Tsirigos, A.; et al. Autoantibody-mediated impairment of DNASE1L3 activity in sporadic systemic lupus erythematosus. J. Exp. Med. 2021, 218, e20201138. [Google Scholar] [CrossRef] [PubMed]
  37. Mutsaers, S.E. The mesothelial cell. Int. J. Biochem. Cell Biol. 2004, 36, 9–16. [Google Scholar] [CrossRef] [PubMed]
  38. Namvar, S.; Woolf, A.S.; Zeef, L.A.; Wilm, T.; Wilm, B.; Herrick, S.E. Functional molecules in mesothelial-to-mesenchymal transition revealed by transcriptome analyses. J. Pathol. 2018, 245, 491–501. [Google Scholar] [CrossRef] [PubMed]
  39. Mao, X.; Xu, J.; Wang, W.; Liang, C.; Hua, J.; Liu, J.; Zhang, B.; Meng, Q.; Yu, X.; Shi, S. Crosstalk between cancer-associated fibroblasts and immune cells in the tumor microenvironment: New findings and future perspectives. Mol. Cancer 2021, 20, 131. [Google Scholar] [CrossRef] [PubMed]
  40. Barker, T.H.; Engler, A.J. The provisional matrix: Setting the stage for tissue repair outcomes. Matrix Biol. 2017, 60–61, 1–4. [Google Scholar] [CrossRef]
  41. Jara, C.P.; Wang, O.; Paulino do Prado, T.; Ismail, A.; Fabian, F.M.; Li, H.; Velloso, L.A.; Carlson, M.A.; Burgess, W.; Lei, Y.; et al. Novel fibrin-fibronectin matrix accelerates mice skin wound healing. Bioact. Mater. 2020, 5, 949–962. [Google Scholar] [CrossRef]
  42. Altaf, F.; Wu, S.; Kasim, V. Role of Fibrinolytic Enzymes in Anti-Thrombosis Therapy. Front. Mol. Biosci. 2021, 8, 680397. [Google Scholar] [CrossRef]
  43. Vilar, R.; Fish, R.J.; Casini, A.; Neerman-Arbez, M. Fibrin(ogen) in human disease: Both friend and foe. Haematologica 2020, 105, 284–296. [Google Scholar] [CrossRef]
  44. Saman, H.; Raza, S.S.; Uddin, S.; Rasul, K. Inducing Angiogenesis, a Key Step in Cancer Vascularization, and Treatment Approaches. Cancers 2020, 12, 1172. [Google Scholar] [CrossRef]
  45. Velazquez, O.C. Angiogenesis and vasculogenesis: Inducing the growth of new blood vessels and wound healing by stimulation of bone marrow-derived progenitor cell mobilization and homing. J. Vasc. Surg. 2007, 45 (Suppl. A), A39–A47. [Google Scholar] [CrossRef]
  46. Shibuya, M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer 2011, 2, 1097–1105. [Google Scholar] [CrossRef]
  47. Niu, G.; Chen, X. Vascular endothelial growth factor as an anti-angiogenic target for cancer therapy. Curr. Drug Targets 2010, 11, 1000–1017. [Google Scholar] [CrossRef]
  48. Lawler, P.R.; Lawler, J. Molecular basis for the regulation of angiogenesis by thrombospondin-1 and -2. Cold Spring Harb. Perspect. Med. 2012, 2, a006627. [Google Scholar] [CrossRef]
  49. Lawler, J. Counter regulation of tumor angiogenesis by vascular endothelial growth factor and thrombospondin-1. Semin. Cancer Biol. 2022, 86, 126–135. [Google Scholar] [CrossRef]
  50. Rohrs, J.A.; Sulistio, C.D.; Finley, S.D. Predictive model of thrombospondin-1 and vascular endothelial growth factor in breast tumor tissue. NPJ Syst. Biol. Appl. 2016, 2, 16030. [Google Scholar] [CrossRef]
  51. Ząbczyk, M.; Natorska, J.; Undas, A. Fibrin clot properties in atherosclerotic vascular disease: From pathophysiology to clinical outcomes. J. Clin. Med. 2021, 10, 2999. [Google Scholar] [CrossRef] [PubMed]
  52. Tracy, L.E.; Minasian, R.A.; Caterson, E.J. Extracellular Matrix and Dermal Fibroblast Function in the Healing Wound. Adv. Wound Care 2016, 5, 119–136. [Google Scholar] [CrossRef] [PubMed]
  53. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef] [PubMed]
  54. Gomes, R.N.; Manuel, F.; Nascimento, D.S. The bright side of fibroblasts: Molecular signature and regenerative cues in major organs. NPJ Regen. Med. 2021, 6, 43. [Google Scholar] [CrossRef] [PubMed]
  55. Theocharis, A.D.; Manou, D.; Karamanos, N.K. The extracellular matrix as a multitasking player in disease. FEBS J. 2019, 286, 2830–2869. [Google Scholar] [CrossRef] [PubMed]
  56. Pompili, S.; Latella, G.; Gaudio, E.; Sferra, R.; Vetuschi, A. The charming world of the extracellular matrix: A dynamic and protective network of the intestinal wall. Front. Med. 2021, 8, 610189. [Google Scholar] [CrossRef] [PubMed]
  57. Mutsaers, S.E.; Prêle, C.M.-A.; Pengelly, S.; Herrick, S.E. Mesothelial cells and peritoneal homeostasis. Fertil. Steril. 2016, 106, 1018–1024. [Google Scholar] [CrossRef] [PubMed]
  58. Mutsaers, S.E.; Birnie, K.; Lansley, S.; Herrick, S.E.; Lim, C.B.; Prêle, C.M. Mesothelial cells in tissue repair and fibrosis. Front. Pharmacol. 2015, 6, 113. [Google Scholar] [CrossRef] [PubMed]
  59. Niederstätter, I.M.; Schiefer, J.L.; Fuchs, P.C. Surgical Strategies to Promote Cutaneous Healing. Med. Sci. 2021, 9, 45. [Google Scholar] [CrossRef]
  60. Bedder, M.D.; Bedder, H.F. Spinal cord stimulation surgical technique for the nonsurgically trained. Neuromodul. Technol. Neural Interface 2009, 12, 1–19. [Google Scholar] [CrossRef]
  61. Ghobrial, S.; Ott, J.; Parry, J.P. An Overview of Postoperative Intraabdominal Adhesions and Their Role on Female Infertility: A Narrative Review. J. Clin. Med. 2023, 12, 2263. [Google Scholar] [CrossRef]
  62. Rocca, A.; Aprea, G.; Surfaro, G.; Amato, M.; Giuliani, A.; Paccone, M.; Salzano, A.; Russo, A.; Tafuri, D.; Amato, B. Prevention and treatment of peritoneal adhesions in patients affected by vascular diseases following surgery: A review of the literature. Open Med. 2016, 11, 106–114. [Google Scholar] [CrossRef] [PubMed]
  63. Aref-Adib, M.; Phan, T.; Ades, A. Preventing adhesions in laparoscopic surgery: The role of anti-adhesion agents. Obstet. Gynaecol. 2019, 21, 185–192. [Google Scholar] [CrossRef]
  64. Wei, C.-Z.; Hou, C.-L.; Gu, Q.-S.; Jiang, L.-X.; Zhu, B.; Sheng, A.-L. A thermosensitive chitosan-based hydrogel barrier for post-operative adhesions’ prevention. Biomaterials 2009, 30, 5534–5540. [Google Scholar] [CrossRef] [PubMed]
  65. Park, H.; Baek, S.; Kang, H.; Lee, D. Biomaterials to Prevent Post-Operative Adhesion. Materials 2020, 13, 3056. [Google Scholar] [CrossRef] [PubMed]
  66. Kang, S.; Park, S.; Baek, I.; Song, Y.; Kim, S.; Choi, D.; Kim, J.; Lee, Y. Development of poly (D, L-lactic-co-glycolic acid) films coated with biomembrane-mimicking polymers for anti-adhesion activity. Mater. Sci. Eng. C 2021, 120, 111780. [Google Scholar] [CrossRef] [PubMed]
  67. Ahmad, G.; Kim, K.; Thompson, M.; Agarwal, P.; O’Flynn, H.; Hindocha, A.; Watson, A. Barrier agents for adhesion prevention after gynaecological surgery. Cochrane Database Syst. Rev. 2020, 3, Cd000475. [Google Scholar] [CrossRef] [PubMed]
  68. Fatehi Hassanabad, A.; Zarzycki, A.N.; Jeon, K.; Dundas, J.A.; Vasanthan, V.; Deniset, J.F.; Fedak, P.W. Prevention of post-operative adhesions: A comprehensive review of present and emerging strategies. Biomolecules 2021, 11, 1027. [Google Scholar] [CrossRef] [PubMed]
  69. Naficy, S.; Le, T.Y.L.; Oveissi, F.; Lee, A.; Hung, J.C.; Wise, S.G.; Winlaw, D.S.; Dehghani, F. Highly porous, biocompatible tough hydrogels, processable via gel fiber spinning and 3D gel printing. Adv. Mater. Interfaces 2020, 7, 1901770. [Google Scholar] [CrossRef]
  70. Tang, J.; Xiang, Z.; Bernards, M.T.; Chen, S. Peritoneal adhesions: Occurrence, prevention and experimental models. Acta Biomater. 2020, 116, 84–104. [Google Scholar] [CrossRef]
  71. Arung, W.; Meurisse, M.; Detry, O. Pathophysiology and prevention of postoperative peritoneal adhesions. World J. Gastroenterol. WJG 2011, 17, 4545. [Google Scholar] [CrossRef]
  72. Díaz-González, F.; Sánchez-Madrid, F. NSAIDs: Learning new tricks from old drugs. Eur. J. Immunol. 2015, 45, 679–686. [Google Scholar] [CrossRef]
  73. Chapman, S.N.; Mehndiratta, P.; Johansen, M.C.; McMurry, T.L.; Johnston, K.C.; Southerland, A.M. Current perspectives on the use of intravenous recombinant tissue plasminogen activator (tPA) for treatment of acute ischemic stroke. Vasc. Health Risk Manag. 2014, 10, 75–87. [Google Scholar]
  74. Huinen, Z.R.; Huijbers, E.J.; van Beijnum, J.R.; Nowak-Sliwinska, P.; Griffioen, A.W. Anti-angiogenic agents—Overcoming tumour endothelial cell anergy and improving immunotherapy outcomes. Nat. Rev. Clin. Oncol. 2021, 18, 527–540. [Google Scholar] [CrossRef]
  75. Fitzsimmons, R.E.B.; Mazurek, M.S.; Soos, A.; Simmons, C.A. Mesenchymal Stromal/Stem Cells in Regenerative Medicine and Tissue Engineering. Stem. Cells Int. 2018, 2018, 8031718. [Google Scholar] [CrossRef]
  76. Merimi, M.; El-Majzoub, R.; Lagneaux, L.; Moussa Agha, D.; Bouhtit, F.; Meuleman, N.; Fahmi, H.; Lewalle, P.; Fayyad-Kazan, M.; Najar, M. The Therapeutic Potential of Mesenchymal Stromal Cells for Regenerative Medicine: Current Knowledge and Future Understandings. Front. Cell Dev. Biol. 2021, 9, 661532. [Google Scholar] [CrossRef]
  77. Lu, Y.; Yang, L.; Chen, X.; Liu, J.; Nie, A.; Chen, X. Bone marrow mesenchymal stem cell-derived exosomes improve renal fibrosis by reducing the polarisation of M1 and M2 macrophages through the activation of EP2 receptors. IET Nanobiotechnol 2022, 16, 14–24. [Google Scholar] [CrossRef]
  78. El-Sherbiny, I.M.; Yacoub, M.H. Hydrogel scaffolds for tissue engineering: Progress and challenges. Glob. Cardiol. Sci. Pract. 2013, 2013, 316–342. [Google Scholar] [CrossRef]
  79. Zhao, W.; Zhu, J.; Hang, J.; Zeng, W. Biomaterials to promote vascularization in tissue engineering organs and ischemic fibrotic diseases. MedComm–Biomater. Appl. 2022, 1, e16. [Google Scholar] [CrossRef]
  80. Mazzoni, E.; Iaquinta, M.R.; Lanzillotti, C.; Mazziotta, C.; Maritati, M.; Montesi, M.; Sprio, S.; Tampieri, A.; Tognon, M.; Martini, F. Bioactive materials for soft tissue repair. Front. Bioeng. Biotechnol. 2021, 9, 613787. [Google Scholar] [CrossRef]
  81. Awonuga, A.O.; Belotte, J.; Abuanzeh, S.; Fletcher, N.M.; Diamond, M.P.; Saed, G.M. Advances in the Pathogenesis of Adhesion Development: The Role of Oxidative Stress. Reprod. Sci. 2014, 21, 823–836. [Google Scholar] [CrossRef]
  82. Bhullar, K.S.; Lagarón, N.O.; McGowan, E.M.; Parmar, I.; Jha, A.; Hubbard, B.P.; Rupasinghe, H.V. Kinase-targeted cancer therapies: Progress, challenges and future directions. Mol. Cancer 2018, 17, 48. [Google Scholar] [CrossRef]
  83. Liu, S.; Wu, F.; Gu, S.; Wu, T.; Chen, S.; Chen, S.; Wang, C.; Huang, G.; Jin, T.; Cui, W. Gene silencing via PDA/ERK2-siRNA-mediated electrospun fibers for peritendinous antiadhesion. Adv. Sci. 2019, 6, 1801217. [Google Scholar] [CrossRef]
  84. Moore, C.B.; Guthrie, E.H.; Huang, M.T.; Taxman, D.J. Short hairpin RNA (shRNA): Design, delivery, and assessment of gene knockdown. Methods Mol. Biol. 2010, 629, 141–158. [Google Scholar] [CrossRef]
  85. Chen, X.; Mangala, L.S.; Rodriguez-Aguayo, C.; Kong, X.; Lopez-Berestein, G.; Sood, A.K. RNA interference-based therapy and its delivery systems. Cancer Metastasis Rev. 2018, 37, 107–124. [Google Scholar] [CrossRef]
  86. Che, M.; Shi, T.; Feng, S.; Li, H.; Zhang, X.; Feng, N.; Lou, W.; Dou, J.; Tang, G.; Huang, C. The microRNA-199a/214 cluster targets E-cadherin and claudin-2 and promotes high glucose-induced peritoneal fibrosis. J. Am. Soc. Nephrol. 2017, 28, 2459–2471. [Google Scholar] [CrossRef]
  87. Deng, L.; Li, Q.; Lin, G.; Huang, D.; Zeng, X.; Wang, X.; Li, P.; Jin, X.; Zhang, H.; Li, C.; et al. P-glycoprotein Mediates Postoperative Peritoneal Adhesion Formation by Enhancing Phosphorylation of the Chloride Channel-3. Theranostics 2016, 6, 204–218. [Google Scholar] [CrossRef]
  88. Diamond, M.P.; Burns, E.L.; Accomando, B.; Mian, S.; Holmdahl, L. Seprafilm(®) adhesion barrier: (2) a review of the clinical literature on intraabdominal use. Gynecol. Surg. 2012, 9, 247–257. [Google Scholar] [CrossRef]
  89. Naito, M.; Ogura, N.; Yamanashi, T.; Sato, T.; Nakamura, T.; Miura, H.; Tsutsui, A.; Sakamoto, Y.; Tanaka, R.; Kumagai, Y.; et al. Prospective randomized controlled study on the validity and safety of an absorbable adhesion barrier (Interceed®) made of oxidized regenerated cellulose for laparoscopic colorectal surgery. Asian J. Endosc. Surg. 2017, 10, 7–11. [Google Scholar] [CrossRef]
  90. Zhao, J.; Huang, C.; Zhu, J.; Zhu, J.; Yuan, R.; Zhu, Z. Efficacy and safety of Seprafilm for preventing intestinal obstruction after gastrointestinal neoplasms surgery: A systematic review and meta-analysis. Acta Chir. Belg. 2021, 121, 1–15. [Google Scholar] [CrossRef]
  91. Maciver, A.H.; McCall, M.; Shapiro, A.J. Intra-abdominal adhesions: Cellular mechanisms and strategies for prevention. Int. J. Surg. 2011, 9, 589–594. [Google Scholar] [CrossRef]
  92. Fortin, C.N.; Saed, G.M.; Diamond, M.P. Predisposing factors to post-operative adhesion development. Hum. Reprod. Update 2015, 21, 536–551. [Google Scholar] [CrossRef]
  93. Rosazza, T.; Warner, J.; Sollberger, G. NET formation—mechanisms and how they relate to other cell death pathways. Febs. J. 2021, 288, 3334–3350. [Google Scholar] [CrossRef]
  94. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Al-Hussaniy, H.A.; Al-Harcan, N.A.H.; Alexiou, A.; Batiha, G.E. Neutrophil Extracellular Traps (NETs) and Covid-19: A new frontiers for therapeutic modality. Int. Immunopharmacol. 2022, 104, 108516. [Google Scholar] [CrossRef]
  95. Rada, B. Neutrophil Extracellular Traps. Methods Mol. Biol. 2019, 1982, 517–528. [Google Scholar] [CrossRef]
  96. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry 2020, 85, 1178–1190. [Google Scholar] [CrossRef]
  97. Remijsen, Q.; Vanden Berghe, T.; Wirawan, E.; Asselbergh, B.; Parthoens, E.; De Rycke, R.; Noppen, S.; Delforge, M.; Willems, J.; Vandenabeele, P. Neutrophil extracellular trap cell death requires both autophagy and superoxide generation. Cell Res. 2011, 21, 290–304. [Google Scholar] [CrossRef]
  98. Thiam, H.R.; Wong, S.L.; Qiu, R.; Kittisopikul, M.; Vahabikashi, A.; Goldman, A.E.; Goldman, R.D.; Wagner, D.D.; Waterman, C.M. NETosis proceeds by cytoskeleton and endomembrane disassembly and PAD4-mediated chromatin decondensation and nuclear envelope rupture. Proc. Natl. Acad. Sci. USA 2020, 117, 7326–7337. [Google Scholar] [CrossRef]
  99. Mesa, M.A.; Vasquez, G. NETosis. Autoimmune Dis. 2013, 2013, 651497. [Google Scholar] [CrossRef]
  100. Hidalgo, A.; Libby, P.; Soehnlein, O.; Aramburu, I.V.; Papayannopoulos, V.; Silvestre-Roig, C. Neutrophil extracellular traps: From physiology to pathology. Cardiovasc. Res. 2022, 118, 2737–2753. [Google Scholar] [CrossRef]
  101. Ronchetti, L.; Boubaker, N.S.; Barba, M.; Vici, P.; Gurtner, A.; Piaggio, G. Neutrophil extracellular traps in cancer: Not only catching microbes. J. Exp. Clin. Cancer Res. 2021, 40, 231. [Google Scholar] [CrossRef]
  102. 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]
  103. Huang, J.; Hong, W.; Wan, M.; Zheng, L. Molecular mechanisms and therapeutic target of NETosis in diseases. MedComm 2022, 3, e162. [Google Scholar] [CrossRef]
  104. Chen, T.; Li, Y.; Sun, R.; Hu, H.; Liu, Y.; Herrmann, M.; Zhao, Y.; Muñoz, L.E. Receptor-Mediated NETosis on Neutrophils. Front. Immunol. 2021, 12, 775267. [Google Scholar] [CrossRef]
  105. Kiwit, A.; Lu, Y.; Lenz, M.; Knopf, J.; Mohr, C.; Ledermann, Y.; Klinke-Petrowsky, M.; Raluy, L.P.; Reinshagen, K.; Herrmann, M.; et al. The Dual Role of Neutrophil Extracellular Traps (NETs) in Sepsis and Ischemia-Reperfusion Injury: Comparative Analysis across Murine Models. Int. J. Mol. Sci. 2024, 25, 3787. [Google Scholar] [CrossRef]
  106. Cosentino-Gomes, D.; Rocco-Machado, N.; Meyer-Fernandes, J.R. Cell signaling through protein kinase C oxidation and activation. Int. J. Mol. Sci. 2012, 13, 10697–10721. [Google Scholar] [CrossRef]
  107. Nguyen, G.T.; Green, E.R.; Mecsas, J. Neutrophils to the ROScue: Mechanisms of NADPH Oxidase Activation and Bacterial Resistance. Front. Cell Infect. Microbiol. 2017, 7, 373. [Google Scholar] [CrossRef]
  108. Metzler, K.D.; Goosmann, C.; Lubojemska, A.; Zychlinsky, A.; Papayannopoulos, V. A myeloperoxidase-containing complex regulates neutrophil elastase release and actin dynamics during NETosis. Cell Rep. 2014, 8, 883–896. [Google Scholar] [CrossRef]
  109. Klopf, J.; Brostjan, C.; Eilenberg, W.; Neumayer, C. Neutrophil Extracellular Traps and Their Implications in Cardiovascular and Inflammatory Disease. Int. J. Mol. Sci. 2021, 22, 559. [Google Scholar] [CrossRef]
  110. Vorobjeva, N.V. Neutrophil Extracellular Traps: New Aspects. Mosc. Univ. Biol. Sci. Bull. 2020, 75, 173–188. [Google Scholar] [CrossRef]
  111. Sollberger, G.; Tilley, D.O.; Zychlinsky, A. Neutrophil Extracellular Traps: The Biology of Chromatin Externalization. Dev. Cell 2018, 44, 542–553. [Google Scholar] [CrossRef]
  112. Thiam, H.R.; Wong, S.L.; Wagner, D.D.; Waterman, C.M. Cellular Mechanisms of NETosis. Annu. Rev. Cell Dev. Biol. 2020, 36, 191–218. [Google Scholar] [CrossRef]
  113. Jones, J.E.; Causey, C.P.; Knuckley, B.; Slack-Noyes, J.L.; Thompson, P.R. Protein arginine deiminase 4 (PAD4): Current understanding and future therapeutic potential. Curr. Opin. Drug Discov. Dev. 2009, 12, 616–627. [Google Scholar]
  114. Zheng, Q.; Osunsade, A.; David, Y. Protein arginine deiminase 4 antagonizes methylglyoxal-induced histone glycation. Nat. Commun. 2020, 11, 3241. [Google Scholar] [CrossRef]
  115. Zhu, D.; Zhang, Y.; Wang, S. Histone citrullination: A new target for tumors. Mol. Cancer 2021, 20, 90. [Google Scholar] [CrossRef]
  116. Rogers, C.; Erkes, D.A.; Nardone, A.; Aplin, A.E.; Fernandes-Alnemri, T.; Alnemri, E.S. Gasdermin pores permeabilize mitochondria to augment caspase-3 activation during apoptosis and inflammasome activation. Nat. Commun. 2019, 10, 1689. [Google Scholar] [CrossRef]
  117. Demkow, U. Molecular Mechanisms of Neutrophil Extracellular Trap (NETs) Degradation. Int. J. Mol. Sci. 2023, 24, 4896. [Google Scholar] [CrossRef]
  118. Gupta, S.; Kaplan, M.J. The role of neutrophils and NETosis in autoimmune and renal diseases. Nat. Rev. Nephrol. 2016, 12, 402–413. [Google Scholar] [CrossRef]
  119. 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]
  120. Sorvillo, N.; Cherpokova, D.; Martinod, K.; Wagner, D.D. Extracellular DNA NET-Works with Dire Consequences for Health. Circ. Res. 2019, 125, 470–488. [Google Scholar] [CrossRef]
  121. Yu, Y.; Su, K. Neutrophil Extracellular Traps and Systemic Lupus Erythematosus. J. Clin. Cell. Immunol. 2013, 4, 2. [Google Scholar] [CrossRef]
  122. Hakkim, A.; Fürnrohr, B.G.; Amann, K.; Laube, B.; Abed, U.A.; Brinkmann, V.; Herrmann, M.; Voll, R.E.; Zychlinsky, A. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA 2010, 107, 9813–9818. [Google Scholar] [CrossRef]
  123. Keyel, P.A. Dnases in health and disease. Dev. Biol. 2017, 429, 1–11. [Google Scholar] [CrossRef]
  124. Ozçakar, Z.B.; Foster, J., 2nd; Diaz-Horta, O.; Kasapcopur, O.; Fan, Y.S.; Yalçınkaya, F.; Tekin, M. DNASE1L3 mutations in hypocomplementemic urticarial vasculitis syndrome. Arthritis Rheum. 2013, 65, 2183–2189. [Google Scholar] [CrossRef]
  125. Wu, X.A.; Choi, C.H.; Zhang, C.; Hao, L.; Mirkin, C.A. Intracellular fate of spherical nucleic acid nanoparticle conjugates. J. Am. Chem. Soc. 2014, 136, 7726–7733. [Google Scholar] [CrossRef]
  126. Monticolo, F.; Palomba, E.; Termolino, P.; Chiaiese, P.; de Alteriis, E.; Mazzoleni, S.; Chiusano, M.L. The Role of DNA in the Extracellular Environment: A Focus on NETs, RETs and Biofilms. Front. Plant Sci. 2020, 11, 589837. [Google Scholar] [CrossRef]
  127. Wilton, M.; Halverson, T.W.R.; Charron-Mazenod, L.; Parkins, M.D.; Lewenza, S. Secreted Phosphatase and Deoxyribonuclease Are Required by Pseudomonas aeruginosa To Defend against Neutrophil Extracellular Traps. Infect. Immun. 2018, 86, 10–1128. [Google Scholar] [CrossRef]
  128. Lind, N.A.; Rael, V.E.; Pestal, K.; Liu, B.; Barton, G.M. Regulation of the nucleic acid-sensing Toll-like receptors. Nat. Rev. Immunol. 2022, 22, 224–235. [Google Scholar] [CrossRef]
  129. Farrera, C.; Fadeel, B. Macrophage clearance of neutrophil extracellular traps is a silent process. J. Immunol. 2013, 191, 2647–2656. [Google Scholar] [CrossRef]
  130. Manfredi, A.A.; Ramirez, G.A.; Rovere-Querini, P.; Maugeri, N. The Neutrophil’s Choice: Phagocytose vs Make Neutrophil Extracellular Traps. Front. Immunol. 2018, 9, 288. [Google Scholar] [CrossRef]
  131. Song, C.; Li, H.; Li, Y.; Dai, M.; Zhang, L.; Liu, S.; Tan, H.; Deng, P.; Liu, J.; Mao, Z.; et al. NETs promote ALI/ARDS inflammation by regulating alveolar macrophage polarization. Exp. Cell Res. 2019, 382, 111486. [Google Scholar] [CrossRef]
  132. Lazzaretto, B.; Fadeel, B. Intra-and extracellular degradation of neutrophil extracellular traps by macrophages and dendritic cells. J. Immunol. 2019, 203, 2276–2290. [Google Scholar] [CrossRef]
  133. Haider, P.; Kral-Pointner, J.B.; Mayer, J.; Richter, M.; Kaun, C.; Brostjan, C.; Eilenberg, W.; Fischer, M.B.; Speidl, W.S.; Hengstenberg, C.; et al. Neutrophil Extracellular Trap Degradation by Differently Polarized Macrophage Subsets. Arter. Thromb. Vasc. Biol. 2020, 40, 2265–2278. [Google Scholar] [CrossRef]
  134. Jiménez-Alcázar, M.; Rangaswamy, C.; Panda, R.; Bitterling, J.; Simsek, Y.J.; Long, A.T.; Bilyy, R.; Krenn, V.; Renné, C.; Renné, T. Host DNases prevent vascular occlusion by neutrophil extracellular traps. Science 2017, 358, 1202–1206. [Google Scholar] [CrossRef]
  135. de Bont, C.M.; Boelens, W.C.; Pruijn, G.J.M. NETosis, complement, and coagulation: A triangular relationship. Cell. Mol. Immunol. 2019, 16, 19–27. [Google Scholar] [CrossRef]
  136. Dunkelberger, J.R.; Song, W.-C. Complement and its role in innate and adaptive immune responses. Cell Res. 2010, 20, 34–50. [Google Scholar] [CrossRef]
  137. Leffler, J.; Martin, M.; Gullstrand, B.; Tydén, H.; Lood, C.; Truedsson, L.; Bengtsson, A.A.; Blom, A.M. Neutrophil extracellular traps that are not degraded in systemic lupus erythematosus activate complement exacerbating the disease. J. Immunol. 2012, 188, 3522–3531. [Google Scholar] [CrossRef]
  138. Dölling, M.; Herrmann, M.; Boettcher, M. NETworking for Health and in Disease: Neutrophil Extracellular Traps in Pediatric Surgical Care. Children 2024, 11, 295. [Google Scholar] [CrossRef]
  139. Ambler, D.R.; Fletcher, N.M.; Diamond, M.P.; Saed, G.M. Effects of hypoxia on the expression of inflammatory markers IL-6 and TNF-a in human normal peritoneal and adhesion fibroblasts. Syst. Biol. Reprod. Med. 2012, 58, 324–329. [Google Scholar] [CrossRef]
  140. Yáñez-Mó, M.; Lara-Pezzi, E.; Selgas, R.; Ramírez-Huesca, M.; Domínguez-Jiménez, C.; Jiménez-Heffernan, J.A.; Aguilera, A.; Sánchez-Tomero, J.A.; Bajo, M.A.; Alvarez, V.; et al. Peritoneal dialysis and epithelial-to-mesenchymal transition of mesothelial cells. N. Engl. J. Med. 2003, 348, 403–413. [Google Scholar] [CrossRef]
  141. Chen, Y.T.; Chang, Y.T.; Pan, S.Y.; Chou, Y.H.; Chang, F.C.; Yeh, P.Y.; Liu, Y.H.; Chiang, W.C.; Chen, Y.M.; Wu, K.D.; et al. Lineage tracing reveals distinctive fates for mesothelial cells and submesothelial fibroblasts during peritoneal injury. J. Am. Soc. Nephrol. 2014, 25, 2847–2858. [Google Scholar] [CrossRef]
  142. Christodoulidis, G.; Tsilioni, I.; Spyridakis, M.E.; Kiropoulos, T.; Oikonomidi, S.; Koukoulis, G.; Tepetes, K. Matrix metaloproteinase-2 and -9 serum levels as potential markers of intraperitoneal adhesions. J. Investig. Surg. 2013, 26, 134–140. [Google Scholar] [CrossRef]
  143. Stoiber, W.; Obermayer, A.; Steinbacher, P.; Krautgartner, W.D. The Role of Reactive Oxygen Species (ROS) in the Formation of Extracellular Traps (ETs) in Humans. Biomolecules 2015, 5, 702–723. [Google Scholar] [CrossRef]
  144. Wu, Y.; Li, E.; Wang, Z.; Shen, T.; Shen, C.; Liu, D.; Gao, Q.; Li, X.; Wei, G. TMIGD1 Inhibited Abdominal Adhesion Formation by Alleviating Oxidative Stress in the Mitochondria of Peritoneal Mesothelial Cells. Oxidative Med. Cell. Longev. 2021, 2021, 9993704. [Google Scholar] [CrossRef]
  145. Ackermann, M.; Anders, H.-J.; Bilyy, R.; Bowlin, G.L.; Daniel, C.; De Lorenzo, R.; Egeblad, M.; Henneck, T.; Hidalgo, A.; Hoffmann, M.; et al. Patients with COVID-19: In the dark-NETs of neutrophils. Cell Death Differ. 2021, 28, 3125–3139. [Google Scholar] [CrossRef]
  146. Azzouz, D.; Khan, M.A.; Palaniyar, N. ROS induces NETosis by oxidizing DNA and initiating DNA repair. Cell Death Discov. 2021, 7, 113. [Google Scholar] [CrossRef]
  147. Hu, Q.; Xia, X.; Kang, X.; Song, P.; Liu, Z.; Wang, M.; Lu, X.; Guan, W.; Liu, S. A review of physiological and cellular mechanisms underlying fibrotic postoperative adhesion. Int. J. Biol. Sci. 2021, 17, 298–306. [Google Scholar] [CrossRef]
  148. Grilz, E.; Mauracher, L.-M.; Königsbrügge, O.; Posch, F.; Lang, I.; Pabinger, I.; Ay, C. The Role of Neutrophil Extracellular Traps in Cancer-Associated Arterial Thrombosis. Blood 2018, 132, 2508. [Google Scholar] [CrossRef]
  149. Eilenberg, W.; Zagrapan, B.; Bleichert, S.; Ibrahim, N.; Knöbl, V.; Brandau, A.; Martelanz, L.; Grasl, M.T.; Hayden, H.; Nawrozi, P.; et al. Histone citrullination as a novel biomarker and target to inhibit progression of abdominal aortic aneurysms. Transl. Res. 2021, 233, 32–46. [Google Scholar] [CrossRef]
  150. Schoen, J.; Euler, M.; Schauer, C.; Schett, G.; Herrmann, M.; Knopf, J.; Yaykasli, K.O. Neutrophils′ Extracellular Trap Mechanisms: From Physiology to Pathology. Int. J. Mol. Sci. 2022, 23, 12855. [Google Scholar] [CrossRef]
  151. Denning, N.L.; Aziz, M.; Gurien, S.D.; Wang, P. DAMPs and NETs in Sepsis. Front. Immunol. 2019, 10, 2536. [Google Scholar] [CrossRef]
  152. Kwak, S.-B.; Kim, S.J.; Kim, J.; Kang, Y.-L.; Ko, C.W.; Kim, I.; Park, J.-W. Tumor regionalization after surgery: Roles of the tumor microenvironment and neutrophil extracellular traps. Exp. Mol. Med. 2022, 54, 720–729. [Google Scholar] [CrossRef] [PubMed]
  153. Eustache, J.H.; Tohme, S.; Milette, S.; Rayes, R.F.; Tsung, A.; Spicer, J.D. Casting a wide net on surgery: The central role of neutrophil extracellular traps. Ann. Surg. 2020, 272, 277–283. [Google Scholar] [CrossRef]
  154. Pandolfi, L.; Bozzini, S.; Frangipane, V.; Percivalle, E.; De Luigi, A.; Violatto, M.B.; Lopez, G.; Gabanti, E.; Carsana, L.; D′Amato, M.; et al. Neutrophil Extracellular Traps Induce the Epithelial-Mesenchymal Transition: Implications in Post-COVID-19 Fibrosis. Front. Immunol. 2021, 12, 663303. [Google Scholar] [CrossRef]
  155. Chamardani, T.M.; Amiritavassoli, S. Inhibition of NETosis for treatment purposes: Friend or foe? Mol. Cell. Biochem. 2022, 477, 673–688. [Google Scholar] [CrossRef]
  156. Sharma, S.; Hofbauer, T.M.; Ondracek, A.S.; Chausheva, S.; Alimohammadi, A.; Artner, T.; Panzenboeck, A.; Rinderer, J.; Shafran, I.; Mangold, A.; et al. Neutrophil extracellular traps promote fibrous vascular occlusions in chronic thrombosis. Blood 2021, 137, 1104–1116. [Google Scholar] [CrossRef]
  157. Zhu, S.; Yu, Y.; Ren, Y.; Xu, L.; Wang, H.; Ling, X.; Jin, L.; Hu, Y.; Zhang, H.; Miao, C.; et al. The emerging roles of neutrophil extracellular traps in wound healing. Cell Death Dis. 2021, 12, 984. [Google Scholar] [CrossRef] [PubMed]
  158. Abaricia, J.O.; Shah, A.H.; Olivares-Navarrete, R. Substrate stiffness induces neutrophil extracellular trap (NET) formation through focal adhesion kinase activation. Biomaterials 2021, 271, 120715. [Google Scholar] [CrossRef]
  159. Erpenbeck, L.; Gruhn, A.L.; Kudryasheva, G.; Günay, G.; Meyer, D.; Busse, J.; Neubert, E.; Schön, M.P.; Rehfeldt, F.; Kruss, S. Effect of Adhesion and Substrate Elasticity on Neutrophil Extracellular Trap Formation. Front. Immunol. 2019, 10, 2320. [Google Scholar] [CrossRef]
  160. Filep, J.G. Targeting Neutrophils for Promoting the Resolution of Inflammation. Front. Immunol. 2022, 13, 866747. [Google Scholar] [CrossRef]
  161. Akbar, M.; McLean, M.; Garcia-Melchor, E.; Crowe, L.A.; McMillan, P.; Fazzi, U.G.; Martin, D.; Arthur, A.; Reilly, J.H.; McInnes, I.B.; et al. Fibroblast activation and inflammation in frozen shoulder. PLoS ONE 2019, 14, e0215301. [Google Scholar] [CrossRef]
  162. Kendall, R.T.; Feghali-Bostwick, C.A. Fibroblasts in fibrosis: Novel roles and mediators. Front. Pharmacol. 2014, 5, 123. [Google Scholar] [CrossRef]
  163. Winkler, J.; Abisoye-Ogunniyan, A.; Metcalf, K.J.; Werb, Z. Concepts of extracellular matrix remodelling in tumour progression and metastasis. Nat. Commun. 2020, 11, 5120. [Google Scholar] [CrossRef] [PubMed]
  164. Pfisterer, K.; Shaw, L.E.; Symmank, D.; Weninger, W. The Extracellular Matrix in Skin Inflammation and Infection. Front. Cell Dev. Biol. 2021, 9, 682414. [Google Scholar] [CrossRef]
  165. Aguilar-Cazares, D.; Chavez-Dominguez, R.; Carlos-Reyes, A.; Lopez-Camarillo, C.; Hernadez de la Cruz, O.N.; Lopez-Gonzalez, J.S. Contribution of Angiogenesis to Inflammation and Cancer. Front. Oncol. 2019, 9, 1399. [Google Scholar] [CrossRef] [PubMed]
  166. Jaipersad, A.S.; Lip, G.Y.; Silverman, S.; Shantsila, E. The role of monocytes in angiogenesis and atherosclerosis. J. Am. Coll. Cardiol. 2014, 63, 1–11. [Google Scholar] [CrossRef]
  167. Stapleton, L.M.; Steele, A.N.; Wang, H.; Lopez Hernandez, H.; Yu, A.C.; Paulsen, M.J.; Smith, A.A.A.; Roth, G.A.; Thakore, A.D.; Lucian, H.J.; et al. Use of a supramolecular polymeric hydrogel as an effective post-operative pericardial adhesion barrier. Nat. Biomed. Eng. 2019, 3, 611–620. [Google Scholar] [CrossRef]
  168. Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef] [PubMed]
  169. Selders, G.S.; Fetz, A.E.; Radic, M.Z.; Bowlin, G.L. An overview of the role of neutrophils in innate immunity, inflammation and host-biomaterial integration. Regen. Biomater. 2017, 4, 55–68. [Google Scholar] [CrossRef]
  170. D′Urso, M.; Kurniawan, N.A. Mechanical and Physical Regulation of Fibroblast-Myofibroblast Transition: From Cellular Mechanoresponse to Tissue Pathology. Front. Bioeng. Biotechnol. 2020, 8, 609653. [Google Scholar] [CrossRef]
  171. Zhang, S.; Jia, X.; Zhang, Q.; Zhang, L.; Yang, J.; Hu, C.; Shi, J.; Jiang, X.; Lu, J.; Shen, H. Neutrophil extracellular traps activate lung fibroblast to induce polymyositis-related interstitial lung diseases via TLR9-miR-7-Smad2 pathway. J. Cell. Mol. Med. 2020, 24, 1658–1669. [Google Scholar] [CrossRef]
  172. Heuer, A.; Stiel, C.; Elrod, J.; Königs, I.; Vincent, D.; Schlegel, P.; Trochimiuk, M.; Appl, B.; Reinshagen, K.; Raluy, L.P.; et al. Therapeutic Targeting of Neutrophil Extracellular Traps Improves Primary and Secondary Intention Wound Healing in Mice. Front. Immunol. 2021, 12, 614347. [Google Scholar] [CrossRef] [PubMed]
  173. Schilrreff, P.; Alexiev, U. Chronic Inflammation in Non-Healing Skin Wounds and Promising Natural Bioactive Compounds Treatment. Int. J. Mol. Sci. 2022, 23, 4928. [Google Scholar] [CrossRef]
  174. Wong, S.L.; Demers, M.; Martinod, K.; Gallant, M.; Wang, Y.; Goldfine, A.B.; Kahn, C.R.; Wagner, D.D. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 2015, 21, 815–819. [Google Scholar] [CrossRef] [PubMed]
  175. Mallavia, B.; Liu, F.; Lefrançais, E.; Cleary, S.J.; Kwaan, N.; Tian, J.J.; Magnen, M.; Sayah, D.M.; Soong, A.; Chen, J. Mitochondrial DNA stimulates TLR9-dependent neutrophil extracellular trap formation in primary graft dysfunction. Am. J. Respir. Cell Mol. Biol. 2020, 62, 364–372. [Google Scholar] [CrossRef] [PubMed]
  176. Fontoura-Andrade, J.L.; Pinto, L.M.; Carneiro, F.P.; Sousa, J.B. Effect of preconditioning and postoperative hyperbaric oxygen therapy on colonic anastomosis healing with and without ischemia in rats. Acta Cir. Bras. 2020, 35, e202000503. [Google Scholar] [CrossRef] [PubMed]
  177. Tonello, S.; Rizzi, M.; Migliario, M.; Rocchetti, V.; Renò, F. Low concentrations of neutrophil extracellular traps induce proliferation in human keratinocytes via NF-kB activation. J. Dermatol. Sci. 2017, 88, 110–116. [Google Scholar] [CrossRef] [PubMed]
  178. Saffarzadeh, M.; Juenemann, C.; Queisser, M.A.; Lochnit, G.; Barreto, G.; Galuska, S.P.; Lohmeyer, J.; Preissner, K.T. Neutrophil extracellular traps directly induce epithelial and endothelial cell death: A predominant role of histones. PLoS ONE 2012, 7, e32366. [Google Scholar] [CrossRef] [PubMed]
  179. Shafqat, A.; Abdul Rab, S.; Ammar, O.; Al Salameh, S.; Alkhudairi, A.; Kashir, J.; Alkattan, K.; Yaqinuddin, A. Emerging role of neutrophil extracellular traps in the complications of diabetes mellitus. Front. Med. 2022, 9, 995993. [Google Scholar] [CrossRef] [PubMed]
  180. Amara, N.; Cooper, M.P.; Voronkova, M.A.; Webb, B.A.; Lynch, E.M.; Kollman, J.M.; Ma, T.; Yu, K.; Lai, Z.; Sangaraju, D.; et al. Selective activation of PFKL suppresses the phagocytic oxidative burst. Cell 2021, 184, 4480–4494.e4415. [Google Scholar] [CrossRef]
  181. Yu, E.; Zhang, E.; Lv, X.; Yan, L.; Lin, Z.; Siaw-Debrah, F.; Zhang, Y.; Yang, S.; Ruan, L.; ZhuGe, Q.; et al. LDC7559 Exerts Neuroprotective Effects by Inhibiting GSDMD-Dependent Pyroptosis of Microglia in Mice with Traumatic Brain Injury. J. Neurotrauma 2023, 40, 742–757. [Google Scholar] [CrossRef]
  182. Mayati, A.; Moreau, A.; Le Vée, M.; Stieger, B.; Denizot, C.; Parmentier, Y.; Fardel, O. Protein Kinases C-Mediated Regulations of Drug Transporter Activity, Localization and Expression. Int. J. Mol. Sci. 2017, 18, 764. [Google Scholar] [CrossRef] [PubMed]
  183. Li, M.; Lin, C.; Deng, H.; Strnad, J.; Bernabei, L.; Vogl, D.T.; Burke, J.J.; Nefedova, Y. A Novel Peptidylarginine Deiminase 4 (PAD4) Inhibitor BMS-P5 Blocks Formation of Neutrophil Extracellular Traps and Delays Progression of Multiple Myeloma. Mol. Cancer Ther. 2020, 19, 1530–1538. [Google Scholar] [CrossRef]
  184. Stojkov, D.; Claus, M.J.; Kozlowski, E.; Oberson, K.; Schären, O.P.; Benarafa, C.; Yousefi, S.; Simon, H.U. NET formation is independent of gasdermin D and pyroptotic cell death. Sci. Signal. 2023, 16, eabm0517. [Google Scholar] [CrossRef]
  185. Sollberger, G.; Choidas, A.; Burn, G.L.; Habenberger, P.; Di Lucrezia, R.; Kordes, S.; Menninger, S.; Eickhoff, J.; Nussbaumer, P.; Klebl, B.; et al. Gasdermin D plays a vital role in the generation of neutrophil extracellular traps. Sci. Immunol. 2018, 3, eaar6689. [Google Scholar] [CrossRef]
  186. Liu, X.; Xia, S.; Zhang, Z.; Wu, H.; Lieberman, J. Channelling inflammation: Gasdermins in physiology and disease. Nat. Rev. Drug Discov. 2021, 20, 384–405. [Google Scholar] [CrossRef] [PubMed]
  187. Wang, Y.; Li, Y.; Chen, Z.; Yuan, Y.; Su, Q.; Ye, K.; Chen, C.; Li, G.; Song, Y.; Chen, H.; et al. GSDMD-dependent neutrophil extracellular traps promote macrophage-to-myofibroblast transition and renal fibrosis in obstructive nephropathy. Cell Death Dis. 2022, 13, 693. [Google Scholar] [CrossRef]
  188. Hu, J.J.; Liu, X.; Xia, S.; Zhang, Z.; Zhang, Y.; Zhao, J.; Ruan, J.; Luo, X.; Lou, X.; Bai, Y.; et al. FDA-approved disulfiram inhibits pyroptosis by blocking gasdermin D pore formation. Nat. Immunol. 2020, 21, 736–745. [Google Scholar] [CrossRef]
  189. Keir, H.R.; Chalmers, J.D. Neutrophil extracellular traps in chronic lung disease: Implications for pathogenesis and therapy. Eur. Respir. Rev. 2022, 31, 210241. [Google Scholar] [CrossRef]
  190. Wong, R.P.; Davis, T.M. Statins as potential antimalarial drugs: Low relative potency and lack of synergy with conventional antimalarial drugs. Antimicrob. Agents Chemother. 2009, 53, 2212–2214. [Google Scholar] [CrossRef] [PubMed]
  191. 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]
  192. Boettcher, M.; Meier, D.; Jiménez-Alcázar, M.; Eschenburg, G.; Mietzsch, S.; Vincent, D.; Klinke, M.; Trochimiuk, M.; Appl, B.; Tiemann, B.; et al. Degradation of Extracellular DNA by DNase1 Significantly Reduces Testicular Damage After Testicular Torsion in Rats. Urology 2017, 109, e221–e223. [Google Scholar] [CrossRef] [PubMed]
  193. Albadawi, H.; Oklu, R.; Raacke Malley, R.E.; O′Keefe, R.M.; Uong, T.P.; Cormier, N.R.; Watkins, M.T. Effect of DNase I treatment and neutrophil depletion on acute limb ischemia-reperfusion injury in mice. J. Vasc. Surg. 2016, 64, 484–493. [Google Scholar] [CrossRef]
  194. Boettcher, M.; Eschenburg, G.; Mietzsch, S.; Jiménez-Alcázar, M.; Klinke, M.; Vincent, D.; Tiemann, B.; Bergholz, R.; Reinshagen, K.; Fuchs, T.A. Therapeutic targeting of extracellular DNA improves the outcome of intestinal ischemic reperfusion injury in neonatal rats. Sci. Rep. 2017, 7, 15377. [Google Scholar] [CrossRef] [PubMed]
  195. Alekseeva, L.; Sen′kova, A.; Savin, I.; Zenkova, M.; Mironova, N. Human Recombinant DNase I (Pulmozyme(®)) Inhibits Lung Metastases in Murine Metastatic B16 Melanoma Model That Correlates with Restoration of the DNase Activity and the Decrease SINE/LINE and c-Myc Fragments in Blood Cell-Free DNA. Int. J. Mol. Sci. 2021, 22, 12074. [Google Scholar] [CrossRef]
  196. Jones, C.E.; Watson, T.J. Anastomotic Leakage Following Esophagectomy. Thorac. Surg. Clin. 2015, 25, 449–459. [Google Scholar] [CrossRef] [PubMed]
  197. Shakhsheer, B.A.; Lec, B.; Zaborin, A.; Guyton, K.; Defnet, A.M.; Bagrodia, N.; Kandel, J.J.; Zaborina, O.; Hernandez, S.L.; Alverdy, J. Lack of evidence for tissue hypoxia as a contributing factor in anastomotic leak following colon anastomosis and segmental devascularization in rats. Int. J. Colorectal. Dis. 2017, 32, 539–547. [Google Scholar] [CrossRef] [PubMed]
  198. Paone, P.; Cani, P.D. Mucus barrier, mucins and gut microbiota: The expected slimy partners? Gut 2020, 69, 2232–2243. [Google Scholar] [CrossRef] [PubMed]
  199. Lee, J.A.; Chico, T.J.A.; Renshaw, S.A. The triune of intestinal microbiome, genetics and inflammatory status and its impact on the healing of lower gastrointestinal anastomoses. FEBS J. 2018, 285, 1212–1225. [Google Scholar] [CrossRef] [PubMed]
  200. Zamorano, D.; Ivulic, D.; Viver, T.; Morales, F.; López-Kostner, F.; Vidal, R.M. Microbiota Phenotype Promotes Anastomotic Leakage in a Model of Rats with Ischemic Colon Resection. Microorganisms 2023, 11, 680. [Google Scholar] [CrossRef]
  201. Tian, Z.; Zhang, Y.; Zheng, Z.; Zhang, M.; Zhang, T.; Jin, J.; Zhang, X.; Yao, G.; Kong, D.; Zhang, C. Gut microbiome dysbiosis contributes to abdominal aortic aneurysm by promoting neutrophil extracellular trap formation. Cell Host Microbe 2022, 30, 1450–1463.e1458. [Google Scholar] [CrossRef]
  202. Liang, Y.; Wang, X.; He, D.; You, Q.; Zhang, T.; Dong, W.; Fei, J.; Xing, Y.; Wu, J. Ameliorating gut microenvironment through staphylococcal nuclease-mediated intestinal NETs degradation for prevention of type 1 diabetes in NOD mice. Life Sci. 2019, 221, 301–310. [Google Scholar] [CrossRef] [PubMed]
  203. dos Santos Ramos, A.; Viana, G.C.S.; de Macedo Brigido, M.; Almeida, J.F. Neutrophil extracellular traps in inflammatory bowel diseases: Implications in pathogenesis and therapeutic targets. Pharmacol. Res. 2021, 171, 105779. [Google Scholar] [CrossRef]
  204. Íñiguez-Gutiérrez, L.; Godínez-Méndez, L.A.; Fafutis-Morris, M.; Padilla-Arellano, J.R.; Corona-Rivera, A.; Bueno-Topete, M.R.; Rojas-Rejón, Ó.A.; Delgado-Rizo, V. Physiological concentrations of short-chain fatty acids induce the formation of neutrophil extracellular traps in vitro. Int. J. Immunopathol. Pharmacol. 2020, 34, 2058738420958949. [Google Scholar] [CrossRef] [PubMed]
  205. Yan, Q.; Jia, S.; Li, D.; Yang, J. The role and mechanism of action of microbiota-derived short-chain fatty acids in neutrophils: From the activation to becoming potential biomarkers. Biomed. Pharmacother. 2023, 169, 115821. [Google Scholar] [CrossRef] [PubMed]
  206. Chen, K.; Shao, L.H.; Wang, F.; Shen, X.F.; Xia, X.F.; Kang, X.; Song, P.; Wang, M.; Lu, X.F.; Wang, C.; et al. Netting Gut Disease: Neutrophil Extracellular Trap in Intestinal Pathology. Oxidative Med. Cell. Longev. 2021, 2021, 5541222. [Google Scholar] [CrossRef] [PubMed]
  207. Drury, B.; Hardisty, G.; Gray, R.D.; Ho, G.-T. Neutrophil Extracellular Traps in Inflammatory Bowel Disease: Pathogenic Mechanisms and Clinical Translation. Cell. Mol. Gastroenterol. Hepatol. 2021, 12, 321–333. [Google Scholar] [CrossRef]
  208. Violi, F.; Cammisotto, V.; Bartimoccia, S.; Pignatelli, P.; Carnevale, R.; Nocella, C. Gut-derived low-grade endotoxaemia, atherothrombosis and cardiovascular disease. Nat. Rev. Cardiol. 2023, 20, 24–37. [Google Scholar] [CrossRef] [PubMed]
  209. Vong, L.; Yeung, C.W.; Pinnell, L.J.; Sherman, P.M. Adherent-invasive Escherichia coli Exacerbates Antibiotic-associated Intestinal Dysbiosis and Neutrophil Extracellular Trap Activation. Inflamm. Bowel Dis. 2015, 22, 42–54. [Google Scholar] [CrossRef]
Figure 1. A PRISMA [16] flow diagram showing the search and selection process. The initial search across PubMed and Google Scholar databases yielded 912 records. After the removal of duplicates, this was refined to 709 unique articles. A review of titles and abstracts led to the exclusion of 582 articles due to irrelevance or those not meeting the inclusion criteria. A total of 127 records underwent full-text article assessment for eligibility and a quality assessment, finally yielding 7 articles for inclusion in our final review.
Figure 1. A PRISMA [16] flow diagram showing the search and selection process. The initial search across PubMed and Google Scholar databases yielded 912 records. After the removal of duplicates, this was refined to 709 unique articles. A review of titles and abstracts led to the exclusion of 582 articles due to irrelevance or those not meeting the inclusion criteria. A total of 127 records underwent full-text article assessment for eligibility and a quality assessment, finally yielding 7 articles for inclusion in our final review.
Cells 13 00991 g001
Table 1. Quality assessment using Critical Appraisal Skills Programme (2023) (CASP) qualitative study checklist.
Table 1. Quality assessment using Critical Appraisal Skills Programme (2023) (CASP) qualitative study checklist.
abcdefghij
Sudo, Makoto et al. [19]YYYYYYYYYY
Terri, Michela et al. [20]YYYYYYYYYY
Sandoval, Pilar et al. [21]YNYYNYYYYY
Kanamaru, Rihito et al. [22]YYYYYYYYYY
Elrod, Julia et al. [23]YYYYYCannot tellYYYY
Hu, Qiongyu et al. [24]YYYYYNYYYY
Jonathan M. Tsai et al. [25]YYYYYYYYYY
Note: (a) Was there a clear statement of the aims of the research? (b) Is a qualitative methodology appropriate? (c) Was the research design appropriate to address the aims of the research? (d) Was the recruitment strategy appropriate to the aims of the research? (e) Was the data collected in a way that addressed the research issue? (f) Has the relationship between researcher and participants been adequately considered? (g) Have ethical issues been taken into consideration? (h) Was the data analysis efficiently rigorous? (i) Is there a clear statement of findings? (j) How valuable is the research? Y = yes; N = no.
Table 2. Overview of adhesion pathogenesis, mechanisms, and interventional strategies. ECM—extracellular matrix, VEGF—vascular endothelial growth factor, siRNA—small interfering RNA, and shRNA—short hairpin RNA.
Table 2. Overview of adhesion pathogenesis, mechanisms, and interventional strategies. ECM—extracellular matrix, VEGF—vascular endothelial growth factor, siRNA—small interfering RNA, and shRNA—short hairpin RNA.
Overview of Adhesion Pathogenesis, Mechanisms, and Interventional Strategies
AspectDescriptionReferences
Etiology and PathogenesisAdhesions, fibrous bands between organs, result from coagulation, inflammation, and angiogenesis initiated by tissue injury from surgery, trauma, or inflammation, leading to fibrin deposition and cytokine-mediated immune activation.[3,25,27,28,29,30,31,32,33,35]
Fibrin Deposition and FibrinolysisFibrin forms a matrix for cell adhesion; imbalance due to impaired fibrinolysis promotes adhesion, as seen with reduced plasmin activity.[40,41,42,43,51]
ECM Remodeling and Fibroblast ProliferationFibroblasts migrate to the injury site, expanding and remodeling ECM with collagen, fibronectin, elastin, proteoglycans, and glycoproteins, essential for adhesion maturation.[52,53,54,55,56]
AngiogenesisNeovascularization, essential for supplying nutrients to fibrotic tissue, is driven by factors like VEGF and inhibited by thrombospondin-1.[44,45,46,47,48,49,50]
Mesothelial Cell DysfunctionSurgical trauma induces mesothelial-to-mesenchymal transition, with mesothelial cells producing ECM, facilitated by disruptions to the protective mesothelial layer.[37,38,39,57,58]
Prophylactic Surgical TechniquesPrecision techniques, including minimally invasive surgery like laparoscopy, aim to minimize tissue trauma and reduce foreign material use to prevent adhesions.[59,60,61,62]
Mechanical Barrier InterventionsBioresorbable films and gels, such as Seprafilm (sodium hyaluronate and carboxymethylcellulose), and hydrogels, such as polyethylene glycol-based products, are used to prevent tissue adhesion.[63,64,65,66,67,68,69]
Pharmacological InterventionsAnti-inflammatory drugs, corticosteroids, tissue plasminogen activators, and antiangiogenic agents are investigated to mitigate adhesion formation.[70,71,72,73,74]
Regenerative Medical ApproachesStem cell therapy, specifically mesenchymal stem cells, and tissue engineering with bioactive scaffolds or hydrogels are explored for their anti-adhesive effects.[75,76,77,78,79,80]
Targeted Molecular TherapiesMolecular interventions including siRNA or shRNA for gene silencing and monoclonal antibodies targeting specific growth factors or signaling pathways are developed to inhibit adhesion pathways.[81,82,83,84,85,86,87]
Barrier Implementation and Pharmacological StrategiesMechanical barriers, such as Seprafilm and pharmacological agents, are utilized during surgery to reduce adhesion incidence, with an increasing interest in their combined roles.[88,89,90]
Integrated Strategy and Future ResearchAppropriate surgical techniques are currently the most effective prevention, with ongoing research into optimizing barrier methods, pharmacological agents, and novel molecular and regenerative therapies.[59,60,61,62,68,91,92]
Table 3. Main factors of NET degradation. SLE—systemic lupus erythematosus, DNase1—deoxyribonuclease-1, DNase1L3—deoxyribonuclease-1-like 3, DNase2—Deoxyribonuclease-2, HUVS—hypocomplementemic urticarial vasculitis syndrome, ALI—acute lung injury, ARDS—acute respiratory distress syndrome, DCs—dendritic cells, and MerTK—Mer tyrosine kinase.
Table 3. Main factors of NET degradation. SLE—systemic lupus erythematosus, DNase1—deoxyribonuclease-1, DNase1L3—deoxyribonuclease-1-like 3, DNase2—Deoxyribonuclease-2, HUVS—hypocomplementemic urticarial vasculitis syndrome, ALI—acute lung injury, ARDS—acute respiratory distress syndrome, DCs—dendritic cells, and MerTK—Mer tyrosine kinase.
Enzyme/Cell TypeDetailed Function and LocationDisease/Condition AssociationNET Degradation and Clearance MechanismReferences
DNase1Cleaves extracellular DNA within NETs; crucial for NET disassembly. Normally active in the circulation. SLE: Patients experience NET accumulation due to absent or impaired DNase1 activity.Central to the degradation process of NETs; absence leads to severe accumulation of NETs, highlighting its importance for immune homeostasis.[120,121,122]
DNase1L3Shares structural and functional properties with DNase1; crucial for extracellular NET degradation.Linked to autoimmune diseases due to DNase1L3 deficiency. Specific mutations related to HUVS and SLE.Similar to DNase1 in preventing an autoimmune response through degradation of NETs[36,123,124]
DNase2Functions at an acidic pH within lysosomes; not a primary actor in NET degradation.Cystic fibrosis: NETs contribute to airway obstruction; DNase2 may degrade DNA in slightly acidic environments.Less effective than DNase1/L3 at degrading NETs, but may act if airway surface pH is sufficiently acidic, indicating a secondary role in NET clearance.[125,126,127,128]
Macrophages, phagocytes, and DCsPhagocytes that engulf and digest NETs through their endosomal–lysosomal system. Express receptors like MerTK for NET recognition. DCs also remove and degrade NETs.ALI, ARDS, and autoimmune responses: NETs exacerbate inflammation.Both cell types play crucial roles in physical engulfment and biochemical degradation of NETs, including the production of DNases.[129,130,131,132,133,134]
Complement-mediated degradationPart of the innate immune system that can recognize and clear NETs once complement proteins have been deposited.SLE: NETs act as potent activators of the complement system, impacting SLE pathology.The complement system marks NETs for phagocytic destruction, aiding in clearance and potentially affecting autoimmune disease progression.[10,135,136,137]
Table 4. The role of neutrophil extracellular traps in peritoneal adhesion formation. ECM—extracellular matrix, ROS—reactive oxygen species, PM—polymyositis, ILD—interstitial lung disease, TLR9—Toll-like receptor 9, LF—lung fibroblast, MF—myofibroblast, and TGF-β—transforming growth factor beta.
Table 4. The role of neutrophil extracellular traps in peritoneal adhesion formation. ECM—extracellular matrix, ROS—reactive oxygen species, PM—polymyositis, ILD—interstitial lung disease, TLR9—Toll-like receptor 9, LF—lung fibroblast, MF—myofibroblast, and TGF-β—transforming growth factor beta.
The Role of Neutrophil Extracellular Traps in Peritoneal Adhesion Formation
AspectDescriptionReferences
Inflammatory ResponseInflammation activates cells like neutrophils to release NETs, promoting fibroblast activation, ECM remodeling, and angiogenesis, which are involved in adhesion formation. NETs and adhesions are interconnected through inflammation and tissue injury. Excessive ROS trigger NET formation through severe DNA damage, initiating DNA repair pathways. [146,158,159,160,161,162,163,164,165,166]
Tissue Injury ResponseAdhesions and NETs play significant roles in responding to tissue injury and inflammation. Adhesions form fibrous bands between organs post-injury, while NETs are immune response structures released by activated neutrophils.[94,167,168,169]
Fibroblast InteractionNETs interact with fibroblasts, promoting their activation and ECM production, contributing to adhesion formation. They also influence polymyositis (PM)-associated interstitial lung disease (ILD) and the TLR9-miR-7-Smad2 signaling pathway in lung fibroblast (LF) proliferation and myofibroblast (MF) differentiation.[170,171]
Tissue RemodelingNETs modulate tissue remodeling and repair, essential for wound healing. Dysregulated NET formation may result in abnormal tissue repair and adhesion formation.[157,172,173]
CoagulationNETs interact with the coagulation system, enhancing thrombosis, which contributes to adhesion development by promoting fibrin deposition as a scaffold. Neutrophil-mediated inflammation enhances TGF-β signaling, leading to fibrotic thrombus remodeling.[156]
Table 5. The potential therapeutic targets of NET inhibition in the context of adhesion formation. NETs—neutrophil extracellular traps; ROS—reactive oxygen species; NADPH—nicotinamide adenine dinucleotide phosphate; PKC—protein kinase C; NE—neutrophil elastase; MPO—myeloperoxidase; PAD4—peptidylarginine deiminase 4; NSA—necrosulfonamide; GSDMD—gasdermin D; DPI—diphenyleneiodonium; NOX—NADPH oxidase.
Table 5. The potential therapeutic targets of NET inhibition in the context of adhesion formation. NETs—neutrophil extracellular traps; ROS—reactive oxygen species; NADPH—nicotinamide adenine dinucleotide phosphate; PKC—protein kinase C; NE—neutrophil elastase; MPO—myeloperoxidase; PAD4—peptidylarginine deiminase 4; NSA—necrosulfonamide; GSDMD—gasdermin D; DPI—diphenyleneiodonium; NOX—NADPH oxidase.
The Potential Therapeutic Targets of NET Inhibition in the Context of Adhesion Formation
Therapeutic TargetsSpecific Agents or ApproachesDescription and ImpactExamplesReferences
Inhibition of NET formationROS inhibitorsAgents such as DPI and apocynin target the NADPH oxidase complex to reduce oxidative stress and subsequent NET formation.DPI, apocynin, LDC7559, and NA-11[111,112,180,181]
PKC inhibitorsGö6976 and Ro-31-8220 suppress the protein kinase C pathway crucial in the signaling cascade that leads to NET formation.Gö6976 and Ro-31-8220[113,182]
NE and MPO inhibitorsSivelestat and ABAH inhibit NE and MPO, respectively, preventing chromatin decondensation critical for NET release.sivelestat/ONO 5046 and ABAH[114,115]
PAD4 inhibitorsCompounds such as Cl-amidine and GSK484 target PAD4, thus blocking histone citrullination, a key step in NET assembly.Cl-amidine, GSK484, and BMS-P5[116,117,183]
GSDMD inhibitorsBlocking of the pore-forming protein gasdermin D hinders the expansion of chromatin and granular proteins during NET formation.NSA and disulfiram[118,119,184,185,186,187,188]
Pharmacological agentsNET suppressionExhibition of anti-inflammatory effects that indirectly reduce NET formation, with chloroquine specifically inhibiting NADPH oxidase and metformin, affecting PKC-βII in neutrophils.chloroquine, simvastatin, and metformin[120,189,190,191]
Endogenous moleculesAcceleration of NET degradationDegradation of extracellular NET structures by endogenous enzymes, potentially reducing NET-mediated thrombosis and improving ischemia–reperfusion outcomes; Pulmozyme® (recombinant human DNase1) has shown efficacy in disintegrating NETs in clinical settings, suggesting its utility in managing NET-related complications.DNase1 and DNase1L3[121,122,123,124,192,193,194,195]
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

Lu, Y.; Elrod, J.; Herrmann, M.; Knopf, J.; Boettcher, M. Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation. Cells 2024, 13, 991. https://doi.org/10.3390/cells13110991

AMA Style

Lu Y, Elrod J, Herrmann M, Knopf J, Boettcher M. Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation. Cells. 2024; 13(11):991. https://doi.org/10.3390/cells13110991

Chicago/Turabian Style

Lu, Yuqing, Julia Elrod, Martin Herrmann, Jasmin Knopf, and Michael Boettcher. 2024. "Neutrophil Extracellular Traps: A Crucial Factor in Post-Surgical Abdominal Adhesion Formation" Cells 13, no. 11: 991. https://doi.org/10.3390/cells13110991

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