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 -- 1362 2023-08-01 04:15:52 |
2 format correct Meta information modification 1362 2023-08-01 10:25:23 | |
3 format correct Meta information modification 1362 2023-08-01 10:28:54 |

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.
Wu, Y.; Pernet, E.; Touqui, L. Role of sPLA2-IIA in Infectious and Inflammatory Diseases. Encyclopedia. Available online: https://encyclopedia.pub/entry/47462 (accessed on 11 September 2024).
Wu Y, Pernet E, Touqui L. Role of sPLA2-IIA in Infectious and Inflammatory Diseases. Encyclopedia. Available at: https://encyclopedia.pub/entry/47462. Accessed September 11, 2024.
Wu, Yongzheng, Erwan Pernet, Lhousseine Touqui. "Role of sPLA2-IIA in Infectious and Inflammatory Diseases" Encyclopedia, https://encyclopedia.pub/entry/47462 (accessed September 11, 2024).
Wu, Y., Pernet, E., & Touqui, L. (2023, August 01). Role of sPLA2-IIA in Infectious and Inflammatory Diseases. In Encyclopedia. https://encyclopedia.pub/entry/47462
Wu, Yongzheng, et al. "Role of sPLA2-IIA in Infectious and Inflammatory Diseases." Encyclopedia. Web. 01 August, 2023.
Role of sPLA2-IIA in Infectious and Inflammatory Diseases
Edit

Host molecules with antimicrobial properties belong to a large family of mediators including type-IIA secreted phospholipase A2 (sPLA2-IIA). The latter is a potent bactericidal agent with high selectivity against Gram-positive bacteria, but it may also play a role in modulating the host inflammatory response. However, several pathogen-associated molecular patterns (PAMPs) or toxins produced by pathogenic bacteria can modulate the levels of sPLA2-IIA by either inducing or inhibiting its expression in host cells.

bacterial toxins sPLA2 host immunity

1. General Biological Functions of sPLA2-IIA

Phospholipase A2 (PLA2) enzymes hydrolyze the sn-2 position of phospholipids, resulting in the production of free fatty acids and lyso-phospholipids [1][2]. These enzymes are classified into two major families: the low molecular weight-secreted PLA2 (sPLA2) and the high molecular weight intracellular PLA2, such as the cytosolic PLA2 (cPLA2) [1][2]. Based on the number and position of their disulfide bridges, sPLA2 can be classified into several different types, one of which is sPLA2-IIA. PLA2 have been shown to release free arachidonic acid (AA), the precursor of proinflammatory eicosanoids, and to bind to specific receptors present on host surface membranes [3][4]. Initially, sPLA2-IIA was suggested to play a role in the development of various inflammatory diseases [5][6]. For example, this enzyme can hydrolyze pulmonary surfactant phospholipids involved in acute respiratory distress syndrome (ARDS). In addition, sPLA2-IIA has been shown to induce neuronal apoptosis in ischemic stroke [7]. Other studies have also shown the involvement of sPLA2-IIA in atherosclerotic lesions [8][9][10], the hydrolysis of mitochondrial membranes released by platelets [11] and plasma lipoproteins [12]. sPLA2-IIA has also been shown to generate lipid mediators from membrane vesicles of platelets and erythrocytes [13].
Thus, it is clear that sPLA2-IIA can be involved in various pathophysiological processes, but its high bactericidal property (especially against Gram-positive bacteria) is now accepted as its most established biological role [5][6][14]. Therefore, it is important to explore the mechanisms by which bacterial toxins can modulate the expression of this enzyme and the pathophysiological consequences of this modulation.

2. PAMPs, Toxins and Innate Immune Response to Bacterial Infections

Pathogen-associated molecular patterns (PAMPs) are microbial motifs that are highly conserved across a wide range of pathogens. They are essential for the survival of these pathogens and their detection by host cells [15]. They include several virulence factors, such as lipopeptides, lipoteichoic acid (LTA) or peptidoglycans (PGN) of Gram-positive bacteria and lipopolysaccharides (LPS), pili or flagellin of Gram-negative bacteria as well as the double-stranded RNA (dsRNA) of certain viruses [16]. Recognition of PAMPs by specialized host cells is the first step in the host immune response, leading to an inflammatory response and elimination of the invading pathogens. This process involves the interactions of PAMPs with cellular receptors called ‘pattern recognition receptors’, or PRRs.
On the other hand, bacteria also produce a variety of toxins in response to various environmental challenges. These include exotoxins that are actively expressed and secreted into the extracellular media or injected into host cells during the infection process [17][18]. The interaction of pathogens with host cells initiates signaling processes that lead to the production of anti-microbial peptides (AMPs) by these cells. AMPs represent a large family of peptides ranging from 10 to 150 amino acids. In particular, 153 AMPs have been found in humans with net positive charges on the surface of the molecules [19], including defensins, cathelicidin, the type IIA secreted phospholipase A2 (sPLA2-IIA), etc. These antimicrobial molecules interact with bacteria to inhibit the synthesis of bacterial membrane phospholipids, cleave polysaccharides of the bacterial cell wall or increase the permeability of the bacterial membrane [19], which ultimately results in the eradication of the pathogens or the reduction of their proliferation. In particular, sPLA2-IIA has a high net positive charge of +17 [20]. Table 1 shows the reported effects of some PAMPs and toxins on sPLA2-IIA expression by host cells (Table 1).
Table 1. Effects of bacterial PAMPs and toxins on sPLA2-IIA expression by host cells.

3. The sPLA2-IIA, an Endogenous Antibiotic-Like Protein of the Host

The bactericidal activity of sPLA2-IIA is related to its ability to efficiently penetrate the cell wall of Gram-positive bacteria. This particular property is due to the high net positive charge of sPLA2-IIA (+17) [20], whereas Gram-positive bacteria have high net anionic charges due to the presence of the D-alanyl moiety covalently linked to lipoteichoic acid (LTA) [38], which is a major membrane component of Gram-positive bacteria. Thus, the highly efficient and rapid binding of sPLA2-IIA to LTA by electrostatic interaction promotes the penetration of sPLA2-IIA into the peptidoglycan layer, another major wall component of Gram-positive bacteria. This leads to efficient hydrolysis of bacterial membrane lipids and subsequent bacterial killing [14]. One study has reported a classification of mouse and human sPLA2 based on their ability to kill the Gram-positive bacterium Staphylococcus aureus [39] and showed that sPLA2-IIA is the most bactericidal sPLA2 type. Indeed, the concentration of sPLA2-IIA in human tears of healthy subjects exceeds 30 µg/mL, and only 15–80 ng/mL of this protein is sufficient to kill S. aureus [40]. Additionally, concentrations of sPLA2-IIA rapidly increase in host biological fluids as a result of inflammation or bacterial infection [4][41][42], as discussed in Part 4 of this section. These concentrations are virtually sufficient to kill all Gram-positive bacteria that may invade the host. Thus, sPLA2-IIA can be considered a major player in the host’s innate immunity.

4. Role of the sPLA2-IIA in the Gut Microbiota–Lung Axis

Given its potent and selective antibacterial activity against Gram-positive bacteria, it is tempting to speculate that sPLA2-IIA might be involved in shaping the gut and pulmonary microbiota [43][44]. Indeed, two recent studies have investigated the influence of sPLA2-IIA on the gut microbiota [43][44]. Using gain- or loss-function assays, sPLA2-IIA was shown to play a central role in the composition of the gut microbiota by reducing the proportion of Gram-positive strains [43][44]. Most importantly, sPLA2-IIA-driven changes in the gut microbiota contributed to alterations in local and extra-intestinal immune responses, leading to increased susceptibility to cancer and arthritis [43][44]. This evidence suggests that intestinal sPLA2-IIA has profound effects on the host immune response through modulation of the gut microbiota and it may also have an impact on the pulmonary immune response by influencing the airway microbiota. Interestingly, there is a privileged relationship and communication between the lung and the gut (known as the gut–lung axis) that is mediated by the microbiota [45], and the gut microbiota has been associated with immunity to viral and bacterial infections [45][46][47][48]. Therefore, future studies are needed to better address the specific role of sPLA2-IIA in microbiota changes and the associated effects on the gut microbiota–lung axis.

5. sPLA2-IIA Levels in Biological Fluids of Infectious and Inflammatory Diseases

sPLA2-IIA was originally identified in synovial fluid from patients with rheumatoid arthritis [49], suggesting its involvement in excessive inflammatory conditions, such as autoimmunity. Subsequent studies have reported elevated levels of sPLA2-IIA in biological fluids from inflammatory diseases, including ARDS [50], pancreatitis, sepsis, cardiovascular disease [4][41][42] and in nasal fluids from patients with allergic rhinitis [51]. ARDS is defined as a life-threatening lung injury characterized by non-cardiogenic pulmonary edema and arterial hypoxemia [50]. The alteration of pulmonary surfactant is a hallmark of ARDS, accounting for increased surface tension at the air–liquid interface, resulting in impaired gas exchange and alveolar collapse.

References

  1. Dennis, E.A. The growing phospholipase A2 superfamily of signal transduction enzymes. Trends Biochem. Sci. 1997, 22, 1–2.
  2. Ghosh, M.; Tucker, D.E.; Burchett, S.A.; Leslie, C.C. Properties of the Group IV phospholipase A2 family. Prog. Lipid Res. 2006, 45, 487–510.
  3. Murakami, M.; Lambeau, G. Emerging roles of secreted phospholipase A(2) enzymes: An update. Biochimie 2013, 95, 43–50.
  4. Touqui, L.; Arbibe, L. A role for phospholipase A2 in ARDS pathogenesis. Mol. Med. Today 1999, 5, 244–249.
  5. Touqui, L.; Alaoui-El-Azher, M. Mammalian secreted phospholipases A2 and their pathophysiological significance in inflammatory diseases. Curr. Mol. Med. 2001, 1, 739–754.
  6. Nevalainen, T.J.; Graham, G.G.; Scott, K.F. Antibacterial actions of secreted phospholipases A2. Review. Biochim. Biophys. Acta 2008, 1781, 1–9.
  7. Yagami, T.; Ueda, K.; Asakura, K.; Hata, S.; Kuroda, T.; Sakaeda, T.; Takasu, N.; Tanaka, K.; Gemba, T.; Hori, Y. Human group IIA secretory phospholipase A2 induces neuronal cell death via apoptosis. Mol. Pharm. 2002, 61, 114–126.
  8. Kugiyama, K.; Ota, Y.; Takazoe, K.; Moriyama, Y.; Kawano, H.; Miyao, Y.; Sakamoto, T.; Soejima, H.; Ogawa, H.; Doi, H.; et al. Circulating levels of secretory type II phospholipase A(2) predict coronary events in patients with coronary artery disease. Circulation 1999, 100, 1280–1284.
  9. Mattsson, N.; Magnussen, C.G.; Ronnemaa, T.; Mallat, Z.; Benessiano, J.; Jula, A.; Taittonen, L.; Kahonen, M.; Juonala, M.; Viikari, J.S.; et al. Metabolic syndrome and carotid intima-media thickness in young adults: Roles of apolipoprotein B, apolipoprotein A-I, C-reactive protein, and secretory phospholipase A2: The cardiovascular risk in young Finns study. Arter. Thromb. Vasc. Biol. 2010, 30, 1861–1866.
  10. Rosenson, R.S.; Gelb, M.H. Secretory phospholipase A2: A multifaceted family of proatherogenic enzymes. Curr. Cardiol. Rep. 2009, 11, 445–451.
  11. Boudreau, L.H.; Duchez, A.C.; Cloutier, N.; Soulet, D.; Martin, N.; Bollinger, J.; Pare, A.; Rousseau, M.; Naika, G.S.; Levesque, T.; et al. Platelets release mitochondria serving as substrate for bactericidal group IIA-secreted phospholipase A2 to promote inflammation. Blood 2014, 124, 2173–2183.
  12. Pruzanski, W.; Lambeau, G.; Lazdunski, M.; Cho, W.; Kopilov, J.; Kuksis, A. Hydrolysis of minor glycerophospholipids of plasma lipoproteins by human group IIA, V and X secretory phospholipases A2. Biochim. Biophys. Acta 2007, 1771, 5–19.
  13. Fourcade, O.; Simon, M.F.; Viode, C.; Rugani, N.; Leballe, F.; Ragab, A.; Fournie, B.; Sarda, L.; Chap, H. Secretory phospholipase A2 generates the novel lipid mediator lysophosphatidic acid in membrane microvesicles shed from activated cells. Cell 1995, 80, 919–927.
  14. van Hensbergen, V.P.; Wu, Y.; van Sorge, N.M.; Touqui, L. Type IIA Secreted Phospholipase A2 in Host Defense against Bacterial Infections. Trends Immunol. 2020, 41, 313–326.
  15. Janeway, C.A., Jr. Approaching the asymptote? Evolution and revolution in immunology. Cold. Spring Harb. Symp. Quant. Biol. 1989, 54 Pt 1, 1–13.
  16. Kumar, V. Toll-like receptors in sepsis-associated cytokine storm and their endogenous negative regulators as future immunomodulatory targets. Int. Immunopharmacol. 2020, 89, 107087.
  17. Hauser, A.R. The type III secretion system of Pseudomonas aeruginosa: Infection by injection. Nat. Rev. Microbiol. 2009, 7, 654–665.
  18. Shenoy, A.R.; Furniss, R.C.D.; Goddard, P.J.; Clements, A. Modulation of Host Cell Processes by T3SS Effectors. Curr. Top Microbiol. Immunol. 2018, 416, 73–115.
  19. Wang, G. Human antimicrobial peptides and proteins. Pharmaceuticals 2014, 7, 545–594.
  20. Baker, S.F.; Othman, R.; Wilton, D.C. Tryptophan-containing mutant of human (group IIa) secreted phospholipase A2 has a dramatically increased ability to hydrolyze phosphatidylcholine vesicles and cell membranes. Biochemistry 1998, 37, 13203–13211.
  21. Poltorak, A.; He, X.; Smirnova, I.; Liu, M.Y.; Van Huffel, C.; Du, X.; Birdwell, D.; Alejos, E.; Silva, M.; Galanos, C.; et al. Defective LPS signaling in C3H/HeJ and C57BL/10ScCr mice: Mutations in Tlr4 gene. Science 1998, 282, 2085–2088.
  22. Arbibe, L.; Vial, D.; Rosinski-Chupin, I.; Havet, N.; Huerre, M.; Vargaftig, B.B.; Touqui, L. Endotoxin induces expression of type II phospholipase A2 in macrophages during acute lung injury in guinea pigs: Involvement of TNF-alpha in lipopolysaccharide-induced type II phospholipase A2 synthesis. J. Immunol. 1997, 159, 391–400.
  23. Pernet, E.; Guillemot, L.; Burgel, P.R.; Martin, C.; Lambeau, G.; Sermet-Gaudelus, I.; Sands, D.; Leduc, D.; Morand, P.C.; Jeammet, L.; et al. Pseudomonas aeruginosa eradicates Staphylococcus aureus by manipulating the host immunity. Nat. Commun. 2014, 5, 5105.
  24. Chaput, C.; Boneca, I.G. Peptidoglycan detection by mammals and flies. Microbes Infect. 2007, 9, 637–647.
  25. Inohara, N.; Ogura, Y.; Fontalba, A.; Gutierrez, O.; Pons, F.; Crespo, J.; Fukase, K.; Inamura, S.; Kusumoto, S.; Hashimoto, M.; et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J. Biol. Chem. 2003, 278, 5509–5512.
  26. Raymond, B.; Leduc, D.; Ravaux, L.; Le Goffic, R.; Candela, T.; Raymondjean, M.; Goossens, P.L.; Touqui, L. Edema toxin impairs anthracidal phospholipase A2 expression by alveolar macrophages. PLoS Pathog. 2007, 3, e187.
  27. Schwandner, R.; Dziarski, R.; Wesche, H.; Rothe, M.; Kirschning, C.J. Peptidoglycan- and lipoteichoic acid-induced cell activation is mediated by toll-like receptor 2. J. Biol. Chem. 1999, 274, 17406–17409.
  28. Pernet, E.; Brunet, J.; Guillemot, L.; Chignard, M.; Touqui, L.; Wu, Y. Staphylococcus aureus Adenosine Inhibits sPLA2-IIA-Mediated Host Killing in the Airways. J. Immunol. 2015, 194, 5312–5319.
  29. Hayashi, F.; Smith, K.D.; Ozinsky, A.; Hawn, T.R.; Yi, E.C.; Goodlett, D.R.; Eng, J.K.; Akira, S.; Underhill, D.M.; Aderem, A. The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5. Nature 2001, 410, 1099–1103.
  30. Proft, T.; Baker, E.N. Pili in Gram-negative and Gram-positive bacteria—Structure, assembly and their role in disease. Cell Mol. Life Sci. 2009, 66, 613–635.
  31. Touqui, L.; Paya, M.; Thouron, F.; Guiyoule, A.; Zarantonelli, M.L.; Leduc, D.; Wu, Y.; Taha, M.K.; Alonso, J.M. Neisseria meningitidis pili induce type-IIA phospholipase A2 expression in alveolar macrophages. FEBS Lett. 2005, 579, 4923–4927.
  32. Vabulas, R.M.; Ahmad-Nejad, P.; da Costa, C.; Miethke, T.; Kirschning, C.J.; Hacker, H.; Wagner, H. Endocytosed HSP60s use toll-like receptor 2 (TLR2) and TLR4 to activate the toll/interleukin-1 receptor signaling pathway in innate immune cells. J. Biol. Chem. 2001, 276, 31332–31339.
  33. Bauer, S.; Kirschning, C.J.; Hacker, H.; Redecke, V.; Hausmann, S.; Akira, S.; Wagner, H.; Lipford, G.B. Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG motif recognition. Proc. Natl. Acad. Sci. USA 2001, 98, 9237–9242.
  34. Hasko, G.; Linden, J.; Cronstein, B.; Pacher, P. Adenosine receptors: Therapeutic aspects for inflammatory and immune diseases. Nat. Rev. Drug. Discov. 2008, 7, 759–770.
  35. Guermonprez, P.; Khelef, N.; Blouin, E.; Rieu, P.; Ricciardi-Castagnoli, P.; Guiso, N.; Ladant, D.; Leclerc, C. The adenylate cyclase toxin of Bordetella pertussis binds to target cells via the alpha(M)beta(2) integrin (CD11b/CD18). J. Exp. Med. 2001, 193, 1035–1044.
  36. Bradley, K.A.; Mogridge, J.; Mourez, M.; Collier, R.J.; Young, J.A. Identification of the cellular receptor for anthrax toxin. Nature 2001, 414, 225–229.
  37. Scobie, H.M.; Rainey, G.J.; Bradley, K.A.; Young, J.A. Human capillary morphogenesis protein 2 functions as an anthrax toxin receptor. Proc. Natl. Acad. Sci. USA 2003, 100, 5170–5174.
  38. Neuhaus, F.C.; Baddiley, J. A continuum of anionic charge: Structures and functions of D-alanyl-teichoic acids in gram-positive bacteria. Microbiol. Mol. Biol. Rev. 2003, 67, 686–723.
  39. Koduri, R.S.; Gronroos, J.O.; Laine, V.J.; Le Calvez, C.; Lambeau, G.; Nevalainen, T.J.; Gelb, M.H. Bactericidal properties of human and murine groups I, II, V, X, and XII secreted phospholipases A(2). J. Biol. Chem. 2002, 277, 5849–5857.
  40. Qu, X.D.; Lehrer, R.I. Secretory phospholipase A2 is the principal bactericide for staphylococci and other gram-positive bacteria in human tears. Infect. Immun. 1998, 66, 2791–2797.
  41. Murakami, M.; Nakatani, Y.; Atsumi, G.; Inoue, K.; Kudo, I. Regulatory functions of phospholipase A2. Crit. Rev. Immunol. 1997, 17, 225–283.
  42. Weinrauch, Y.; Elsbach, P.; Madsen, L.M.; Foreman, A.; Weiss, J. The potent anti-Staphylococcus aureus activity of a sterile rabbit inflammatory fluid is due to a 14-kD phospholipase A2. J. Clin. Investig. 1996, 97, 250–257.
  43. Miki, Y.; Taketomi, Y.; Kidoguchi, Y.; Yamamoto, K.; Muramatsu, K.; Nishito, Y.; Park, J.; Hosomi, K.; Mizuguchi, K.; Kunisawa, J.; et al. Group IIA secreted phospholipase A2 controls skin carcinogenesis and psoriasis by shaping the gut microbiota. JCI Insight 2022, 7, e152611.
  44. Dore, E.; Joly-Beauparlant, C.; Morozumi, S.; Mathieu, A.; Levesque, T.; Allaeys, I.; Duchez, A.C.; Cloutier, N.; Leclercq, M.; Bodein, A.; et al. The interaction of secreted phospholipase A2-IIA with the microbiota alters its lipidome and promotes inflammation. JCI Insight 2022, 7, e152638.
  45. Enaud, R.; Prevel, R.; Ciarlo, E.; Beaufils, F.; Wieers, G.; Guery, B.; Delhaes, L. The Gut-Lung Axis in Health and Respiratory Diseases: A Place for Inter-Organ and Inter-Kingdom Crosstalks. Front. Cell. Infect. Microbiol. 2020, 10, 9.
  46. Ichinohe, T.; Pang, I.K.; Kumamoto, Y.; Peaper, D.R.; Ho, J.H.; Murray, T.S.; Iwasaki, A. Microbiota regulates immune defense against respiratory tract influenza A virus infection. Proc. Natl. Acad. Sci. USA 2011, 108, 5354–5359.
  47. Khan, N.; Mendonca, L.; Dhariwal, A.; Fontes, G.; Menzies, D.; Xia, J.; Divangahi, M.; King, I.L. Intestinal dysbiosis compromises alveolar macrophage immunity to Mycobacterium tuberculosis. Mucosal Immunol. 2019, 12, 772–783.
  48. Brown, R.L.; Sequeira, R.P.; Clarke, T.B. The microbiota protects against respiratory infection via GM-CSF signaling. Nat. Commun. 2017, 8, 1512.
  49. Pruzanski, W.; Vadas, P. Secretory synovial fluid phospholipase A2 and its role in the pathogenesis of inflammation in arthritis. J. Rheumatol. 1988, 15, 1601–1603.
  50. Nakos, G.; Kitsiouli, E.; Hatzidaki, E.; Koulouras, V.; Touqui, L.; Lekka, M.E. Phospholipases A2 and platelet-activating-factor acetylhydrolase in patients with acute respiratory distress syndrome. Crit. Care Med. 2005, 33, 772–779.
  51. Touqui, L.; Herpin-Richard, N.; Gene, R.M.; Jullian, E.; Aljabi, D.; Hamberger, C.; Vargaftig, B.B.; Dessange, J.F. Excretion of platelet activating factor-acetylhydrolase and phospholipase A2 into nasal fluids after allergenic challenge: Possible role in the regulation of platelet activating factor release. J. Allergy Clin. Immunol. 1994, 94, 109–119.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 254
Revisions: 3 times (View History)
Update Date: 01 Aug 2023
1000/1000
ScholarVision Creations