25
Views
0
CrossRef citations to date
0
Altmetric
Short Communication

Proteomic Alterations Associated with the Formation of Monocyte Extracellular Trap Induced by Candida Albicans Hyphae

ORCID Icon, , , , &
Pages 307-316 | Received 12 Feb 2023, Accepted 03 Aug 2023, Published online: 15 Feb 2024

References

  • Mora Carpio AL, Climaco A. Fungemia candidiasis. In: StatPearls [Internet]. StatPearls Publishing, FL, USA (2023). https://www.ncbi.nlm.nih.gov/books/NBK436012/
  • Noble SM, Gianetti BA, Witchley JN. Candida albicans cell-type switching and functional plasticity in the mammalian host. Nat. Rev. Microbiol. 15(2), 96–108 (2017).
  • Urban CF, Reichard U, Brinkmann V, Zychlinsky A. Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol. 8(4), 668–676 (2006).
  • Olivier FaB, Hilsenstein V, Weerasinghe H et al. The escape of Candida albicans from macrophages is enabled by the fungal toxin candidalysin and two host cell death pathways. Cell Rep. 40(12), 111374 (2022).
  • Ginhoux F, Jung S. Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat. Rev. Immunol. 14(6), 392–404 (2014).
  • Camilli G, Blagojevic M, Naglik JR, Richardson JP. Programmed cell death: central player in fungal infections. Trends Cell Biol. 31(3), 179–196 (2021).
  • Halder LD, Abdelfatah MA, Jo EA et al. Factor H binds to extracellular DNA traps released from human blood monocytes in response to Candida albicans . Front. Immunol. 7, 671 (2016).
  • Granger V, Faille D, Marani V et al. Human blood monocytes are able to form extracellular traps. J. Leukoc. Biol. 102(3), 775–781 (2017).
  • Conceição-Silva F, Reis CSM, De Luca PM et al. The immune system throws its traps: cells and their extracellular traps in disease and protection. Cells 10(8), 1891 (2021).
  • Wartha F, Henriques-Normark B. ETosis: a novel cell death pathway. Sci. Signal 1(21), pe25 (2008).
  • Papayannopoulos V. Neutrophil extracellular traps in immunity and disease. Nat. Rev. Immunol. 18(2), 134–147 (2018).
  • Proust A, Lévesque JC, Barat C, Sato S, Tremblay MJ. A new tool for detection of extracellular traps. Methods Appl. Fluoresc. 6(3), 037002 (2018).
  • Kenno S, Perito S, Mosci P, Vecchiarelli A, Monari C. Autophagy and reactive oxygen species are involved in neutrophil extracellular traps release induced by C. albicans morphotypes. Front. Microbiol. 7, 879 (2016).
  • Shankar M, Lo TL, Traven A. Natural variation in clinical isolates of Candida albicans modulates neutrophil responses. mSphere 5(4), e00501-20 (2020).
  • Ermert D, Urban CF, Laube B, Goosmann C, Zychlinsky A, Brinkmann V. Mouse neutrophil extracellular traps in microbial infections. J. Innate Immun. 1(3), 181–193 (2009).
  • Wu X, Zhang G, Yang WH et al. GLUT3 as an intersection of glycerophospholipid metabolism and the innate immune response to Candida albicans . Front. Cell Infect. Microbiol. 11, 648988 (2021).
  • Loureiro A, Pais C, Sampaio P. Relevance of macrophage extracellular traps in C. albicans killing. Front. Immunol. 10, 2767 (2019).
  • Domínguez-Andrés J, Arts RJW, Ter Horst R et al. Rewiring monocyte glucose metabolism via C-type lectin signaling protects against disseminated candidiasis. PLoS Pathog. 13(9), e1006632 (2017).
  • De Vries DH, Matzaraki V, Bakker OB et al. Integrating GWAS with bulk and single-cell RNA-sequencing reveals a role for LY86 in the anti-Candida host response. PLoS Pathog. 16(4), e1008408 (2020).
  • Shi J, Li Q, Sheng M, Zheng M, Yu M, Zhang L. The role of TLR4 in M1 macrophage-induced epithelial–mesenchymal transition of peritoneal mesothelial cells. Cell Physiol. Biochem. 40(6), 1538–1548 (2016).
  • Williams DW, Engle EL, Shirk EN et al. Splenic damage during SIV infection: role of T-cell depletion and macrophage polarization and infection. Am. J. Pathol. 186(8), 2068–2087 (2016).
  • Buckner CM, Calderon TM, Willams DW, Belbin TJ, Berman JW. Characterization of monocyte maturation/differentiation that facilitates their transmigration across the blood–brain barrier and infection by HIV: implications for NeuroAIDS. Cell Immunol. 267(2), 109–123 (2011).
  • Ferwerda G, Meyer-Wentrup F, Kullberg BJ, Netea MG, Adema GJ. Dectin-1 synergizes with TLR2 and TLR4 for cytokine production in human primary monocytes and macrophages. Cell. Microbiol. 10(10), 2058–2066 (2008).
  • Nija RJ, Sanju S, Sidharthan N, Mony U. Extracellular trap by blood cells: clinical implications. Tissue Eng. Regen. Med. 17(2), 141–153 (2020).
  • Vorobjeva NV, Chernyak BV. NETosis: molecular mechanisms, role in physiology and pathology. Biochemistry (Mosc.) 85(10), 1178–1190 (2020).
  • He Y, Liu J, Chen Y, Yan L, Wu J. Neutrophil extracellular traps in Candida albicans infection. Front. Immunol. 13, 913028 (2022).
  • Martchenko M, Alarco AM, Harcus D, Whiteway M. Superoxide dismutases in Candida albicans: transcriptional regulation and functional characterization of the hyphal-induced SOD5 gene. Mol. Biol. Cell 15(2), 456–467 (2004).
  • Rossi DCP, Gleason JE, Sanchez H et al. Candida albicans FRE8 encodes a member of the NADPH oxidase family that produces a burst of ROS during fungal morphogenesis. PLoS Pathog. 13(12), e1006763 (2017).
  • Karkowska-Kuleta J, Smolarz M, Seweryn-Ozog K et al. Proteinous components of neutrophil extracellular traps are arrested by the cell wall proteins of Candida albicans during fungal infection, and can be used in the host invasion. Cells 10(10), 2736 (2021).
  • Lopez CM, Wallich R, Riesbeck K, Skerka C, Zipfel PF. Candida albicans uses the surface protein Gpm1 to attach to human endothelial cells and to keratinocytes via the adhesive protein vitronectin. PLOS ONE 9(3), e90796 (2014).
  • Byrd AS, O’brien XM, Johnson CM, Lavigne LM, Reichner JS. An extracellular matrix-based mechanism of rapid neutrophil extracellular trap formation in response to Candida albicans . J. Immunol. 190(8), 4136–4148 (2013).
  • Wu SY, Wu-Hsieh BA. Neutrophil extracellular trap killing assay of Candida albicans . Bio. Protoc. 10(16), e3716 (2020).
  • Wu SY, Weng CL, Jheng MJ et al. Candida albicans triggers NADPH oxidase-independent neutrophil extracellular traps through dectin-2. PLoS Pathog. 15(11), e1008096 (2019).
  • Sumagin R, Prizant H, Lomakina E, Waugh RE, Sarelius IH. LFA-1 and Mac-1 define characteristically different intralumenal crawling and emigration patterns for monocytes and neutrophils in situ. J. Immunol. 185(11), 7057–7066 (2010).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.