Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 1502 word(s) 1502 2021-10-09 08:52:09 |
2 Done Meta information modification 1502 2021-10-11 10:14:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Shin, Y.J. Role of Neutrophils on the Ocular Surface. Encyclopedia. Available online: https://encyclopedia.pub/entry/14940 (accessed on 24 April 2024).
Shin YJ. Role of Neutrophils on the Ocular Surface. Encyclopedia. Available at: https://encyclopedia.pub/entry/14940. Accessed April 24, 2024.
Shin, Young Joo. "Role of Neutrophils on the Ocular Surface" Encyclopedia, https://encyclopedia.pub/entry/14940 (accessed April 24, 2024).
Shin, Y.J. (2021, October 11). Role of Neutrophils on the Ocular Surface. In Encyclopedia. https://encyclopedia.pub/entry/14940
Shin, Young Joo. "Role of Neutrophils on the Ocular Surface." Encyclopedia. Web. 11 October, 2021.
Role of Neutrophils on the Ocular Surface
Edit

Neutrophils are considered short-term and terminally differentiated phagocytes with no significant gene expression or regulatory role in adaptive immunity. However, in recent years, opinions on the role of neutrophils have been developing. Neutrophils are primarily short-term polymorphonuclear cells (PMNs) related with the first line combatant to pathogens, which can phagocytose potentially harmful antigens and trigger strong antimicrobial defenses, including the release of reactive oxygen species (ROS) such as superoxide and neutrophil extracellular traps.

ocular surface disease dry eye syndrome neutrophil neutrophil extracellular trap

1. Introduction

The ocular surface of the eyeball is the part of the eye in contact with the outside world, serving as a primary barrier against external substances and pathogens [1]. The cornea is a transparent tissue that refracts light entering the eye, focusing it on the retina and acting as a barrier against the outside [2]. The conjunctiva is a mucous membrane that attaches to the cornea and becomes the surface surrounding the eyeball [3]. It forms a conjunctival sac surrounding the inner eyelid and connecting to the eyelid [2]. In addition, the conjunctiva is connected to the nasal mucosa and supplied with tears from the lacrimal gland through the lacrimal duct [2]. The mucous membrane of the conjunctiva has many blood vessels and produces a large amount of mucus from goblet cells [3]. The subconjunctival tissue contains many lymphoid tissues and the immune system [3]. The ocular surface immune system can be divided into innate and adaptive immune systems [4]. The innate immune system includes basophils, dendritic cells, eosinophils, Langerhans cells, mast cells, monocytes and macrophages, neutrophils, and natural killer cells, whereas the adaptive immune system includes T and B lymphocytes [5].

Neutrophils are members of the innate immune system and are at the forefront against infection, but they are involved in adaptive immunity through interactions with T and B cells [6]. They have been reported to play an essential role in autoimmune diseases such as rheumatoid arthritis, systemic lupus erythematosus, and anti-neutrophil cytoplasmic antibodies-associated vasculitis (AAV) [7]. Although the ocular surface is in contact with external pathogens, both innate and adaptive immunity are involved in the pathogenesis of dry eye syndrome, characterized by tear instability and inflammation of the ocular surface [8]. Therefore, the role of neutrophils in the ocular surface is discussed in this article.

2. Neutrophils in Immunity

Neutrophil extracellular traps have been described as one of the ways in which neutrophils remove microbes [9][10]. Neutrophils release a chromatin network adorned with granular-derived antibacterial peptides and active enzymes, including cathepsin G, MPO, and neutrophil elastase [11]. Neutrophil extracellular trap formation, known as NETosis, was initially reported as an extracellular antibacterial form resisting microbe. However, recently it has been reported that neutrophil extracellular trap formation is involved in autoimmune/rheumatic and auto-inflammatory disease states beyond microbial death [12]. Neutrophil extracellular traps are released after infection with Gram (+) and (-) bacteria, especially by large-sized microbes [13]. Neutrophils generally kill small microbes through phagocytosis, but larger microbes that are not easily digested release cytoplasmic granules and promote nuclear translocation of neutrophil elastases to form neutrophil extracellular traps [9]. In addition, neutrophil extracellular trap formation is induced by nitric oxide, autoantibodies, cytokines such as interleukin (IL)-1β, IL-6, IL-8, and tumor necrosis factor (TNF)-α, hydrogen peroxide, lipopolysaccharides, phorbol-12-myristate-13-acetate, ionophores for calcium ion, and the interaction with activated platelets or vascular endothelial cells [14][15].

The three main signaling pathways of neutrophil extracellular trap formation have been discussed. First, after phorbol-12-myristate-13-acetate stimulates neutrophils through the protein kinase C (PKC) and Raf-mitogen-activated protein kinase (MEK) extracellular signal-regulated kinase signaling pathways, it induces the activation of nicotinamide adenine dinucleotide phosphate oxidase 2, triggering the associated signaling cascade and neutrophil extracellular trap formation through the production of ROS [12][16]. ROS formation promotes the migration of two key enzymes, MPO and neutrophil elastase, stored in the neutrophil granules, to the nucleus and induces chromatin decondensation, leading to the release of nuclear neutrophil extracellular traps [12]. Hydrogen peroxide is converted to hypochlorous acid by MPO, which activates neutrophil elastase to break down the cytoskeleton and nuclear membrane, allowing neutrophil extracellular trap excretion [17]. Second, the increase in intracellular calcium levels activates the peptidylarginine deiminase 4 (PAD4) enzyme, which moves to the nucleus, leading to histone citrullination and chromatin decondensation [18]. This mechanism is independent of nicotinamide adenine dinucleotide phosphate oxidase 2 [19]. Third, another form of neutrophil extracellular trap formation is the mitoNET formation [20]. Mitochondria are degraded and release the oxidized mitochondrial DNA into the extracellular space by mitochondrial ROS production or the stimulation of toll-like receptor 4 or complement factor 5a receptor [20][21]. Neutrophil extracellular trap formation induced by nitric oxide and phorbol myristate acetate induces both nuclear and mitochondrial neutrophil extracellular trap formation [15].

Neutrophil extracellular trap is thought to be enrolled in the onset of autoimmune and autoinflammatory diseases [22]. Autoantibodies to neutrophil extracellular trap components, including the citrullinated histones with DNA, MPO-DNA complexes, and neutrophil elastase-DNA complexes, are common in several systemic autoimmune diseases [23]. Defects in the process of neutrophil extracellular trap formation, excessive neutrophil extracellular trap formation, and delayed neutrophil extracellular trap formation clearance are all associated with autoimmunity [23]. Neutrophil extracellular traps have been suggested to play a pivotal role in various autoimmune diseases, including systemic lupus erythematosus, vasculitis, rheumatoid arthritis, and chronic inflammatory bowel diseases such as Crohn’s disease and ulcerative colitis [24][25][26][27]. Circulating and synovial neutrophils in patients with rheumatoid arthritis are more prone to forming neutrophil extracellular traps than in healthy controls [28][29]. Neutrophil extracellular trap formation is a source of autoantibody and stimulates inflammatory responses in rheumatoid arthritis [29]. In rheumatoid arthritis, anti-citrullinated protein antibodies are formed, associated with neutrophil extracellular trap formation and neutrophil count [30]. Neutrophil extracellular trap formation can also be provoked by neutrophil binding of anti-neutrophil cytoplasmic antibodies and anti-ribonuclear protein (RNP) antibodies [31]. As an extracellular bactericidal mechanism used by neutrophils, neutrophil extracellular traps go through steps that include ROS production, PAD4 activation, granule formation, chromatin decondensation, and active release of DNA/histone/cathelicidin antimicrobial peptide cocktail into the extracellular space [19].

Peptidyl-arginine deiminase 2 (PAD2) and PAD4 are the posttranslational modification enzymes converting protein arginine or mono-methylarginine to citrulline [32]. PAD2 and PAD4 are implicated in the pathogenesis of several autoimmune diseases [33]. Histone citrullination by PAD2 and PAD4 is essential for neutrophil extracellular trap formation [23][34][35]. Hypercitrullination in synovial fluid and anti-citrullinated protein antibodies in plasma are found in rheumatoid arthritis [36], suggesting that the hypercitrullinated molecules may serve as autoantigen. PAD2 and PAD4 are potential biomarkers and therapeutic targets of sepsis [37]. PAD4 inhibitor block neutrophil extracellular trap formation [38], reducing bleomycin fibrosis [39][40]. Simultaneous inhibition of PAD2 and PAD4 ameliorates neutrophil extracellular trap formation and reduces inflammatory cytokine production [41].

3. Endoplasmic Reticulum Stress in Neutrophils

Endoplasmic reticulum stress is involved in the pathogenesis of many diseases such as dry eye, rheumatoid arthritis, diabetes, dementia, and cancers [42][43][44][45]. It is linked to cellular dysfunction, inflammation, oxidative stress, apoptosis, and autophagy. Mitochondrial activity and endoplasmic reticulum stress are required for neutrophil differentiation [46]. Endoplasmic reticulum stress reduces during both neutrophil and macrophage differentiations, and the activities of protein kinase R-like endoplasmic reticulum kinase and activating transcription factor 6 were decreased, and that of inositol-requiring enzyme 1-α is enhanced during neutrophil differentiation [46]. The role of endoplasmic reticulum stress of neutrophils was investigated in acute lung injury [47]. Elevated endoplasmic reticulum stress levels were observed in infiltrated neutrophils in the acute lung injury mice model [47]. Sensors for endoplasmic reticulum stress, including protein kinase R-like endoplasmic reticulum kinase, activating transcription factor 6, and inositol-requiring kinase 1, were enhanced in neutrophil in acute lung injury [47]. Suppression of endoplasmic reticulum stress inhibited the inflammation [47]. Inositol-requiring enzyme 1-α is a crucial regulator of neutrophil extracellular traps through ROS generation and caspase-2 activation [48]. Endoplasmic reticulum calcium level is increased in the neutrophils in cystic fibrosis in response to endoplasmic reticulum stress response, which exaggerates the inflammation [49]. Tunicamycin-induced endoplasmic reticulum stress signaling (protein kinase R-like endoplasmic reticulum kinase/activating transcription factor 4 /CCAAT-enhancer-binding protein homologous protein signaling) aggravates airway inflammation via elevation of inflammatory cytokines (IL-6, IL-8, and TNF-α) in a murine model of neutrophilic asthma [50]. Endoplasmic reticulum stress/X-box-binding protein 1 enhances mucin secretion through the influence of neutrophil elastase [51]. Neutrophil induces apoptosis in cancer cells through an endoplasmic reticulum stress pathway [52].

4. Neutrophils in Aging

Neutrophil extracellular traps remove the old vessels to promote remodeling [53]. However, aging drives neutrophils to be pathogenic, contributing to vascular diseases [54][55]. The intestinal microtome regulates neutrophil aging by enhancing C-X-C motif chemokine receptor 4 and reducing L-selectin [56]. Interaction between neutrophils and the microbiome contributes to the maturation of the immune system and the pathogenesis of immune-mediated diseases and cardiovascular diseases [57][58]. Aged neutrophils are characterized by altered expression of surface molecules such as lymphocyte function-associated antigen-1, macrophage-1 antigen, toll-like receptor-4, platelet endothelial cell adhesion molecule-1, and higher oxidative stress levels [59]. In addition, neutrophil extracellular traps are more prone to be formed in aged neutrophils [59]. Neutrophil extracellular traps accumulation compromises organ functions and impairs revascularization and vascular repair after ischemic injuries [60]. Delayed clearance of neutrophil extracellular traps facilitates autoimmune reactivity [61].

Neutrophil aging induces chronic inflammation of vessels, affecting lacrimal glands and ocular surfaces. Since neutrophil extracellular trap formation is also easily activated on the ocular surface in the elderly, it may be one of the pathogenic mechanisms of dry eyes in elderly patients.

References

  1. Agrahari, V.; Mandal, A.; Agrahari, V.; Trinh, H.M.; Joseph, M.; Ray, A.; Hadji, H.; Mitra, R.; Pal, D.; Mitra, A.K. A comprehensive insight on ocular pharmacokinetics. Drug Deliv. Transl. Res. 2016, 6, 735–754.
  2. Sridhar, M.S. Anatomy of cornea and ocular surface. Indian J. Ophthalmol. 2018, 66, 190–194.
  3. Hodges, R.R.; Dartt, D.A. Tear film mucins: Front line defenders of the ocular surface; comparison with airway and gastrointestinal tract mucins. Exp. Eye Res. 2013, 117, 62–78.
  4. Galletti, J.G.; de Paiva, C.S. The ocular surface immune system through the eyes of aging. Ocul. Surf. 2021, 20, 139–162.
  5. Sokol, C.L.; Luster, A.D. The chemokine system in innate immunity. Cold Spring Harb. Perspect. Biol. 2015, 7, a016303.
  6. Mocsai, A. Diverse novel functions of neutrophils in immunity, inflammation, and beyond. J. Exp. Med. 2013, 210, 1283–1299.
  7. Navegantes, K.C.; de Souza Gomes, R.; Pereira, P.A.T.; Czaikoski, P.G.; Azevedo, C.H.M.; Monteiro, M.C. Immune modulation of some Autoimmune Dis.eases: The critical role of macrophages and neutrophils in the innate and adaptive immunity. J. Transl. Med. 2017, 15, 36.
  8. Reyes, J.L.; Vannan, D.T.; Eksteen, B.; Avelar, I.J.; Rodriguez, T.; Gonzalez, M.I.; Mendoza, A.V. Innate and Adaptive Cell Populations Driving Inflammation in Dry Eye Disease. Mediators Inflamm. 2018, 2018, 2532314.
  9. Brinkmann, V.; Zychlinsky, A. Neutrophil extracellular traps: Is immunity the second function of chromatin? J. Cell Biol. 2012, 198, 773–783.
  10. Brinkmann, V.; Reichard, U.; Goosmann, C.; Fauler, B.; Uhlemann, Y.; Weiss, D.S.; Weinrauch, Y.; Zychlinsky, A. Neutrophil extracellular traps kill bacteria. Science 2004, 303, 1532–1535.
  11. Kaplan, M.J.; Radic, M. Neutrophil extracellular traps: Double-edged swords of innate immunity. J. Immunol. 2012, 189, 2689–2695.
  12. Delgado-Rizo, V.; Martinez-Guzman, M.A.; Iniguez-Gutierrez, L.; Garcia-Orozco, A.; Alvarado-Navarro, A.; Fafutis-Morris, M. Neutrophil Extracellular Traps and Its Implications in Inflammation: An Overview. Front. Immunol. 2017, 8, 81.
  13. Manda, A.; Pruchniak, M.P.; Arazna, M.; Demkow, U.A. Neutrophil extracellular traps in physiology and pathology. Cent. Eur. J. Immunol. 2014, 39, 116–121.
  14. Keshari, R.S.; Jyoti, A.; Dubey, M.; Kothari, N.; Kohli, M.; Bogra, J.; Barthwal, M.K.; Dikshit, M. Cytokines induced neutrophil extracellular traps formation: Implication for the inflammatory disease condition. PLoS ONE 2012, 7, e48111.
  15. Hoppenbrouwers, T.; Autar, A.S.A.; Sultan, A.R.; Abraham, T.E.; van Cappellen, W.A.; Houtsmuller, A.B.; van Wamel, W.J.B.; van Beusekom, H.M.M.; van Neck, J.W.; de Maat, M.P.M. In vitro induction of NETosis: Comprehensive live imaging comparison and systematic review. PLoS ONE 2017, 12, e0176472.
  16. Fonseca, Z.; Diaz-Godinez, C.; Mora, N.; Aleman, O.R.; Uribe-Querol, E.; Carrero, J.C.; Rosales, C. Entamoeba histolytica Induce Signaling via Raf/MEK/ERK for Neutrophil Extracellular Trap (NET) Formation. Front. Cell Infect. Microbiol. 2018, 8, 226.
  17. Palmer, L.J.; Cooper, P.R.; Ling, M.R.; Wright, H.J.; Huissoon, A.; Chapple, I.L. Hypochlorous acid regulates neutrophil extracellular trap release in humans. Clin. Exp. Immunol. 2012, 167, 261–268.
  18. Rohrbach, A.S.; Slade, D.J.; Thompson, P.R.; Mowen, K.A. Activation of PAD4 in NET formation. Front. Immunol. 2012, 3, 360.
  19. Vorobjeva, N.V.; Chernyak, B.V. NETosis: Molecular Mechanisms, Role in Physiology and Pathology. Biochemistry 2020, 85, 1178–1190.
  20. 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.
  21. Suliman, H.B.; Welty-Wolf, K.E.; Carraway, M.S.; Schwartz, D.A.; Hollingsworth, J.W.; Piantadosi, C.A. Toll-like receptor 4 mediates mitochondrial DNA damage and biogenic responses after heat-inactivated E. coli. FASEB J. 2005, 19, 1531–1533.
  22. Frizinsky, S.; Haj-Yahia, S.; Machnes Maayan, D.; Lifshitz, Y.; Maoz-Segal, R.; Offengenden, I.; Kidon, M.; Agmon-Levin, N. The innate immune perspective of autoimmune and autoinflammatory conditions. Rheumatology 2019, 58, vi1–vi8.
  23. Tsourouktsoglou, T.D.; Warnatsch, A.; Ioannou, M.; Hoving, D.; Wang, Q.; Papayannopoulos, V. Histones, DNA, and Citrullination Promote Neutrophil Extracellular Trap Inflammation by Regulating the Localization and Activation of TLR4. Cell Rep. 2020, 31, 107602.
  24. Fatemi, A.; Alipour, R.; Khanahmad, H.; Alsahebfosul, F.; Andalib, A.; Pourazar, A. The impact of neutrophil extracellular trap from patients with systemic lupus erythematosus on the viability, CD11b expression and oxidative burst of healthy neutrophils. BMC Immunol. 2021, 22, 12.
  25. Wang, H.; Li, T.; Chen, S.; Gu, Y.; Ye, S. Neutrophil Extracellular Trap Mitochondrial DNA and Its Autoantibody in Systemic Lupus Erythematosus and a Proof-of-Concept Trial of Metformin. Arthritis Rheumatol. 2015, 67, 3190–3200.
  26. Bach, M.; Moon, J.; Moore, R.; Pan, T.; Nelson, J.L.; Lood, C. A Neutrophil Activation Biomarker Panel in Prognosis and Monitoring of Patients with Rheumatoid Arthritis. Arthritis Rheumatol. 2020, 72, 47–56.
  27. Angelidou, I.; Chrysanthopoulou, A.; Mitsios, A.; Arelaki, S.; Arampatzioglou, A.; Kambas, K.; Ritis, D.; Tsironidou, V.; Moschos, I.; Dalla, V.; et al. REDD1/Autophagy Pathway Is Associated with Neutrophil-Driven IL-1beta Inflammatory Response in Active Ulcerative Colitis. J. Immunol. 2018, 200, 3950–3961.
  28. Zhang, L.; Yuan, Y.; Xu, Q.; Jiang, Z.; Chu, C.Q. Contribution of neutrophils in the pathogenesis of rheumatoid arthritis. J. Biomed. Res. 2019, 34, 86–93.
  29. Khandpur, R.; Carmona-Rivera, C.; Vivekanandan-Giri, A.; Gizinski, A.; Yalavarthi, S.; Knight, J.S.; Friday, S.; Li, S.; Patel, R.M.; Subramanian, V.; et al. NETs are a source of citrullinated autoantigens and stimulate inflammatory responses in rheumatoid arthritis. Sci. Transl. Med. 2013, 5, 178ra140.
  30. Demoruelle, M.K.; Harrall, K.K.; Ho, L.; Purmalek, M.M.; Seto, N.L.; Rothfuss, H.M.; Weisman, M.H.; Solomon, J.J.; Fischer, A.; Okamoto, Y.; et al. Anti-Citrullinated Protein Antibodies Are Associated With Neutrophil Extracellular Traps in the Sputum in Relatives of Rheumatoid Arthritis Patients. Arthritis Rheumatol. 2017, 69, 1165–1175.
  31. Gestermann, N.; Di Domizio, J.; Lande, R.; Demaria, O.; Frasca, L.; Feldmeyer, L.; Di Lucca, J.; Gilliet, M. Netting Neutrophils Activate Autoreactive B Cells in Lupus. J. Immunol. 2018, 200, 3364–3371.
  32. Shi, L.; Yao, H.; Liu, Z.; Xu, M.; Tsung, A.; Wang, Y. Endogenous PAD4 in Breast Cancer Cells Mediates Cancer Extracellular Chromatin Network Formation and Promotes Lung Metastasis. Mol. Cancer Res. 2020, 18, 735–747.
  33. Liu, Y.; Lightfoot, Y.L.; Seto, N.; Carmona-Rivera, C.; Moore, E.; Goel, R.; O’Neil, L.; Mistry, P.; Hoffmann, V.; Mondal, S.; et al. Peptidylarginine deiminases 2 and 4 modulate innate and adaptive immune responses in TLR-7-dependent lupus. JCI Insight 2018, 3, e124729.
  34. Leshner, M.; Wang, S.; Lewis, C.; Zheng, H.; Chen, X.A.; Santy, L.; Wang, Y. PAD4 mediated histone hypercitrullination induces heterochromatin decondensation and chromatin unfolding to form neutrophil extracellular trap-like structures. Front. Immunol. 2012, 3, 307.
  35. Li, P.; Li, M.; Lindberg, M.R.; Kennett, M.J.; Xiong, N.; Wang, Y. PAD4 is essential for antibacterial innate immunity mediated by neutrophil extracellular traps. J. Exp. Med. 2010, 207, 1853–1862.
  36. Romero, V.; Fert-Bober, J.; Nigrovic, P.A.; Darrah, E.; Haque, U.J.; Lee, D.M.; van Eyk, J.; Rosen, A.; Andrade, F. Immune-mediated pore-forming pathways induce cellular hypercitrullination and generate citrullinated autoantigens in rheumatoid arthritis. Sci. Transl. Med. 2013, 5, 209ra150.
  37. Tian, Y.; Qu, S.; Alam, H.B.; Williams, A.M.; Wu, Z.; Deng, Q.; Pan, B.; Zhou, J.; Liu, B.; Duan, X.; et al. Peptidylarginine deiminase 2 has potential as both a biomarker and therapeutic target of sepsis. JCI Insight 2020, 5, e138873.
  38. 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.
  39. Suzuki, M.; Ikari, J.; Anazawa, R.; Tanaka, N.; Katsumata, Y.; Shimada, A.; Suzuki, E.; Tatsumi, K. PAD4 Deficiency Improves Bleomycin-induced Neutrophil Extracellular Traps and Fibrosis in Mouse Lung. Am. J. Respir. Cell Mol. Biol. 2020, 63, 806–818.
  40. Lewis, H.D.; Liddle, J.; Coote, J.E.; Atkinson, S.J.; Barker, M.D.; Bax, B.D.; Bicker, K.L.; Bingham, R.P.; Campbell, M.; Chen, Y.H.; et al. Inhibition of PAD4 activity is sufficient to disrupt mouse and human NET formation. Nat. Chem. Biol. 2015, 11, 189–191.
  41. Wu, Z.; Deng, Q.; Pan, B.; Alam, H.B.; Tian, Y.; Bhatti, U.F.; Liu, B.; Mondal, S.; Thompson, P.R.; Li, Y. Inhibition of PAD2 Improves Survival in a Mouse Model of Lethal LPS-Induced Endotoxic Shock. Inflammation 2020, 43, 1436–1445.
  42. Park, Y.J.; Yoo, S.A.; Kim, W.U. Role of endoplasmic reticulum stress in rheumatoid arthritis pathogenesis. J. Korean Med. Sci. 2014, 29, 2–11.
  43. Eizirik, D.L.; Cardozo, A.K.; Cnop, M. The role for endoplasmic reticulum stress in diabetes mellitus. Endocr. Rev. 2008, 29, 42–61.
  44. Ghosh, R.; Colon-Negron, K.; Papa, F.R. Endoplasmic reticulum stress, degeneration of pancreatic islet beta-cells, and therapeutic modulation of the unfolded protein response in diabetes. Mol. Metab. 2019, 27S, S60–S68.
  45. Santos, L.E.; Ferreira, S.T. Crosstalk between endoplasmic reticulum stress and brain inflammation in Alzheimer’s disease. Neuropharmacology 2018, 136, 350–360.
  46. Tanimura, A.; Miyoshi, K.; Horiguchi, T.; Hagita, H.; Fujisawa, K.; Noma, T. Mitochondrial Activity and Unfolded Protein Response are Required for Neutrophil Differentiation. Cell. Physiol. Biochem. 2018, 47, 1936–1950.
  47. Hu, R.; Chen, Z.F.; Yan, J.; Li, Q.F.; Huang, Y.; Xu, H.; Zhang, X.P.; Jiang, H. Endoplasmic Reticulum Stress of Neutrophils Is Required for Ischemia/Reperfusion-Induced Acute Lung Injury. J. Immunol. 2015, 195, 4802–4809.
  48. Sule, G.; Abuaita, B.H.; Steffes, P.A.; Fernandes, A.T.; Estes, S.K.; Dobry, C.; Pandian, D.; Gudjonsson, J.E.; Kahlenberg, J.M.; O’Riordan, M.X.; et al. Endoplasmic reticulum stress sensor IRE1alpha propels neutrophil hyperactivity in lupus. J. Clin. Investig. 2021, 131, e137866.
  49. White, M.M.; Geraghty, P.; Hayes, E.; Cox, S.; Leitch, W.; Alfawaz, B.; Lavelle, G.M.; McElvaney, O.J.; Flannery, R.; Keenan, J.; et al. Neutrophil Membrane Cholesterol Content is a Key Factor in Cystic Fibrosis Lung Disease. EBioMedicine 2017, 23, 173–184.
  50. Guo, Q.; Li, H.; Liu, J.; Xu, L.; Yang, L.; Sun, Z.; Zhou, B. Tunicamycin aggravates endoplasmic reticulum stress and airway inflammation via PERK-ATF4-CHOP signaling in a murine model of neutrophilic asthma. J. Asthma 2017, 54, 125–133.
  51. Xu, X.; Li, Q.; Li, L.; Zeng, M.; Zhou, X.; Cheng, Z. Endoplasmic reticulum stress/XBP1 promotes airway mucin secretion under the influence of neutrophil elastase. Int. J. Mol. Med. 2021, 47, 1–9.
  52. Garcia-Navas, R.; Gajate, C.; Mollinedo, F. Neutrophils drive endoplasmic reticulum stress-mediated apoptosis in cancer cells through arginase-1 release. Sci. Rep. 2021, 11, 12574.
  53. Binet, F.; Cagnone, G.; Crespo-Garcia, S.; Hata, M.; Neault, M.; Dejda, A.; Wilson, A.M.; Buscarlet, M.; Mawambo, G.T.; Howard, J.P.; et al. Neutrophil extracellular traps target senescent vasculature for tissue remodeling in retinopathy. Science 2020, 369, eaay5356.
  54. Roy-O’Reilly, M.A.; Ahnstedt, H.; Spychala, M.S.; Munshi, Y.; Aronowski, J.; Sansing, L.H.; McCullough, L.D. Aging exacerbates neutrophil pathogenicity in ischemic stroke. Aging 2020, 12, 436–461.
  55. Weisenburger-Lile, D.; Dong, Y.; Yger, M.; Weisenburger, G.; Polara, G.F.; Chaigneau, T.; Ochoa, R.Z.; Marro, B.; Lapergue, B.; Alamowitch, S.; et al. Harmful neutrophil subsets in patients with ischemic stroke: Association with disease severity. Neurol. Neuroimmunol. Neuroinflamm. 2019, 6, e571.
  56. Zhang, D.; Chen, G.; Manwani, D.; Mortha, A.; Xu, C.; Faith, J.J.; Burk, R.D.; Kunisaki, Y.; Jang, J.E.; Scheiermann, C.; et al. Neutrophil ageing is regulated by the microbiome. Nature 2015, 525, 528–532.
  57. Zhang, D.; Frenette, P.S. Cross talk between neutrophils and the microbiota. Blood 2019, 133, 2168–2177.
  58. Mangold, A.; Alias, S.; Scherz, T.; Hofbauer, M.; Jakowitsch, J.; Panzenbock, A.; Simon, D.; Laimer, D.; Bangert, C.; Kammerlander, A.; et al. Coronary neutrophil extracellular trap burden and deoxyribonuclease activity in ST-elevation acute coronary syndrome are predictors of ST-segment resolution and infarct size. Circ. Res. 2015, 116, 1182–1192.
  59. Uhl, B.; Vadlau, Y.; Zuchtriegel, G.; Nekolla, K.; Sharaf, K.; Gaertner, F.; Massberg, S.; Krombach, F.; Reichel, C.A. Aged neutrophils contribute to the first line of defense in the acute inflammatory response. Blood 2016, 128, 2327–2337.
  60. Kang, L.; Yu, H.; Yang, X.; Zhu, Y.; Bai, X.; Wang, R.; Cao, Y.; Xu, H.; Luo, H.; Lu, L.; et al. Neutrophil extracellular traps released by neutrophils impair revascularization and vascular remodeling after stroke. Nat. Commun. 2020, 11, 2488.
  61. Hakkim, A.; Furnrohr, 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.
More
Information
Subjects: Ophthalmology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 502
Revisions: 2 times (View History)
Update Date: 11 Oct 2021
1000/1000