Differentiation of macrophages in the presence of anaphylatoxin C3a: effect on efferocytosis

Cover Page


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

BACKGROUND: Macrophages are unique professional phagocytes involved in both innate and adaptive immune responses. This is made possible by their functional plasticity—the ability to acquire different phenotypes. Switching between subtypes during interactions with other immune system components is referred to as polarization. Anaphylatoxin C3a has been shown to influence macrophage polarization. Depending on the polarization state, macrophages exhibit different phagocytic activities. Given the contradictory evidence regarding C3a’s effects on macrophages, determining its impact on phagocytosis in general and on efferocytosis in particular is of special interest.

AIM: The work aimed to investigate the effect of anaphylatoxin C3a on efferocytosis by M0 and M2 macrophages.

METHODS: The experiments were conducted using human peripheral blood mononuclear cells. Macrophages were differentiated in the presence of C3a, and M2 polarization was induced with interleukin-4. Efferocytosis was assessed using confocal microscopy, whereas phagocytosis was evaluated via flow cytometry. Real-time reverse transcription polymerase chain reaction was used to measure expression of the efferocytosis receptor genes mertk, axl, and tyro3.

RESULTS: M2 macrophages demonstrated greater efferocytic capacity compared to M0 macrophages. Anaphylatoxin C3a did not affect the phagocytosis of dead Escherichia coli bacteria but inhibited the phagocytosis of apoptotic cells in both unpolarized M0 and alternatively activated M2 macrophages. It was also shown that C3a reduces the expression of the Tyro3 receptor gene in both M0 and M2 macrophages.

CONCLUSION: Anaphylatoxin C3a exerts a suppressive effect on efferocytosis, likely through modulation of apoptotic cell recognition processes.

Full Text

Restricted Access

About the authors

Aleksandra A. Dmitrieva

Institute of Experimental Medicine

Author for correspondence.
Email: aleksandra-2001@mail.ru
ORCID iD: 0000-0003-2680-4069
SPIN-code: 3009-2698
Russian Federation, Saint Petersburg

Anna A. Ivanova

Institute of Experimental Medicine

Email: anna.ivantcova@gmail.com
ORCID iD: 0000-0002-8673-9628
SPIN-code: 5306-1995
Russian Federation, Saint Petersburg

Alexandra V. Burnusuz

Institute of Experimental Medicine

Email: alexandraburnusuz@gmail.com
ORCID iD: 0000-0002-2281-5548
Russian Federation, Saint Petersburg

Sergey V. Orlov

Institute of Experimental Medicine

Email: serge@iem.spb.ru
ORCID iD: 0000-0002-3134-1989
SPIN-code: 1690-8110

Cand. Sci. (Biology)

Russian Federation, Saint Petersburg

References

  1. Green DR, Oguin TH, Martinez J. The clearance of dying cells: table for two. Cell Death Differ. 2016;23:915–926. doi: 10.1038/cdd.2015.172
  2. Guan X, Wang Y, Li W, et al. The role of macrophage efferocytosis in the pathogenesis of apical periodontitis. Int J Mol Sci. 2024;25(7):3854. doi: 10.3390/ ijms25073854
  3. Yunna C, Mengru H, Lei W, Weidong C. Macrophage M1/M2 polarization. Eur J Pharmacol. 2020;877:173090. doi: 10.1016/j.ejphar.2020.173090
  4. Perez S, Rius-Perez S. Macrophage polarization and reprogramming in acute inflammation: a redox perspective. Antioxidants (Basel). 2022;11(7):1394. doi: 10.3390/antiox11071394
  5. Hanna J, Ah-Pine F, Boina C, et al. Deciphering the role of the anaphylatoxin C3a: a key function in modulating the tumor microenvironment. Cancers (Basel). 2023;15(11):2986. doi: 10.3390/cancers15112986
  6. Reid RC, Yau M-K, Singh R, et al. Downsizing a human inflammatory protein to a small molecule with equal potency and functionality. Nat Commun. 2013;4:1–9. doi: 10.1038/ncomms3802
  7. Strainic MG, Liu J, Huang D, et al. Locally produced complement fragments C5a and C3a provide both costimulatory and survival signals to naive CD4+ T cells. Immunity. 2008;28(3):425–435. doi: 10.1016/j.immuni.2008.02.001
  8. Coulthard LG, Woodruff TM. Is the complement activation product C3a a proinflammatory molecule? Re-evaluating the evidence and the myth. J Immunol. 2013;194(8):3542–3548. doi: 10.4049/jimmunol.1403068
  9. Mogilenko DA, Danko K, Larionova EE, et al. Differentiation of human macrophages with anaphylatoxin C3a impairs alternative M2 polarization and decreases lipopolysaccharide-induced cytokine secretion. Immunol Cell Biol. 2022;100(3):186–204. doi: 10.1111/imcb.12534
  10. Evans AL, Blackburn JW, Yin C, Heit B. Quantitative efferocytosis assays. Methods Mol Biol. 2017;1519:25–41. doi: 10.1007/978-1-4939-6581-6_3
  11. Taruc K, Yin C, Wootton DG, Heit B. Quantification of efferocytosis by single-cell fluorescence microscopy. J Vis Exp. 2018;(138):58149. doi: 10.3791/58149
  12. Mogilenko DA, Kudriavtsev IV, Shavva VS, et al. Peroxisome proliferator-activated receptor alpha positively regulates complement C3 expression but inhibits tumor necrosis factor α mediated activation of C3 gene in mammalian hepatic derived cells. J Biol Chem. 2013;288(3):1726–1738. doi: 10.1074/jbc.M112.437525
  13. Toobian D, Ghosh P, Katkar GD. Parsing the role of PPARs in macrophage processes. Front Immunol. 2021;12:783780. doi: 10.3389/fimmu.2021.783780
  14. Roszer T, Menendez-Gutierrez MP, Lefterova MI, et al. Autoimmune kidney disease and impaired engulfment of apoptotic cells in mice with macrophage peroxisome proliferator-activated receptor γ or retinoid X receptor α deficiency. J Immunol. 2011;186(1):621–631. doi: 10.4049/jimmunol.1002230
  15. Mukundan L, Odegaard JI, Morel CR, et al. PPAR-δ senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat Med. 2009;15(11):1266–1272. doi: 10.1038/nm.2048
  16. A-Gonzalez N, Bensinger SJ, Hong C, et al. Apoptotic cells promote their own clearance and immune tolerance through activation of LXR. Immunity. 2009;31(2):245–258. doi: 10.1016/j.immuni.2009.06.018
  17. Pastore M, Grimaudo S, Pipitone RM, et al. Role of myeloid-epithelial-reproductive tyrosine kinase and macrophage polarization in the progression of atherosclerotic lesions associated with nonalcoholic fatty liver disease. Front Pharmacol. 2019;10:604. doi: 10.3389/fphar.2019.00604
  18. Chinetti-Gbaguidi G, Baron M, Bouhlel MA, et al. Human atherosclerotic plaque alternative macrophages display low cholesterol handling but high phagocytosis because of distinct activities of the PPARγ and LXRα pathways. Circ Res. 2011;108(8):985–995. doi: 10.1161/CIRCRESAHA.110.233775
  19. Myers KV, Amend SR, Pienta KJ. Targeting Tyro3, Axl and MerTK (TAM receptors): implications for macrophages in the tumor microenvironment. Mol Cancer. 2019;18(1):94. doi: 10.1186/s12943-019-1022-2

Supplementary files

Supplementary Files
Action
1. JATS XML
2. Fig. 1. Verification of macrophage polarization: a, measurement of CD206 expression levels by flow cytometry. Number of replicates: n = 3. The gray peak corresponds to cells stained with nonspecific fluorescently labeled antibodies (isotype control). The background fluorescence is indicated by a cut-off line. Dashed line, M0 macrophages (median 9.5; first and third quartiles 7.3 and 12.4, respectively); solid line, M2 macrophages (median 13.9; first and third quartiles 9.4 and 19.9, respectively); *p < 0.05 (Mann–Whitney test); b, IL-1Ra levels in the culture supernatants of M0 and M2 macrophages measured by enzyme-linked immunosorbent assay. Box plots represent medians and interquartile ranges; *p < 0.05 (Mann–Whitney test); c, mRNA levels of cd206, il1ra, and pparγ in macrophages. Box plots represent medians and interquartile ranges; *p < 0.05 (Mann–Whitney test).

Download (162KB)
3. Fig. 2. Efferocytosis of cells by macrophages. Macrophages (M0 and M2) were differentiated and polarized in the presence of C3a (10 nM): a, confocal microscopy; macrophages were stained with DAPI, and apoptotic Jurkat cells were labeled with CFDA-SE. The image was obtained by merging signals from three channels (DIC, CFSE, DAPI). Arrows indicate macrophages that had engulfed apoptotic cells, whereas crossed-out arrows indicate events that could not be identified as efferocytosis; b, quantitative assessment of efferocytosis, with the y-axis showing the efferocytic index: the ratio of the number of internalized apoptotic cells to the total number of macrophages in the field of view (100 fields of view for each data point), multiplied by 100%. Box plots represent the medians and interquartile ranges of the analyzed data. *p < 0.05 (Kruskal–Wallis test).

Download (306KB)
4. Fig. 3. Phagocytosis of E. coli by macrophages. Number of replicates: n = 3. The vertical line indicates the autofluorescence threshold of macrophages. Blue: macrophages that phagocytosed E. coli–FITC (median 235; first and third quartiles 229 and 243, respectively); black line: macrophages differentiated in the presence of C3a (10 nM) and that phagocytosed E. coli–FITC (median 271; first and third quartiles 262 and 275, respectively). *p < 0.05 (Mann–Whitney test).

Download (83KB)
5. Fig. 4. mRNA levels of TAM receptor genes mertk, axl, and tyro3 in macrophages. Box plots represent the medians and interquartile ranges. *p < 0.05 (Kruskal–Wallis test).

Download (97KB)

Copyright (c) 2025 Eco-Vector



СМИ зарегистрировано Федеральной службой по надзору в сфере связи, информационных технологий и массовых коммуникаций (Роскомнадзор).
Регистрационный номер и дата принятия решения о регистрации СМИ: серия ПИ № ФС 77 - 74760 от 29.12.2018 г.