GET THE APP

Macrophage Polarization in Infectious Diseases
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2015) Volume 0, Issue 0

Macrophage Polarization in Infectious Diseases

Debora Decote-Ricardo1, Leonardo Freire-de-Lima2, Alexandre Morrot3* and Célio Geraldo Freire-de-Lima2*
1Instituto de Veterinária, Universidade Federal Rural do Rio de Janeiro, Rio de Janeiro, Brazil
2Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
3Departamento de Microbiologia Geral, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
*Corresponding Author(s): Dr. Alexandre Morrot, Instituto de Microbiologia, Universidade Federal do Rio de Janeiro (UFRJ), CCS - Sala D1-035, Av. Carlos Chagas Filho, 373 - Cidade Universitária, CEP 21.941-902, Ilha Do Fundão, Rio De Janeiro, RJ, Brazil Email:
Célio Geraldo Freire-de-Lima, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro (UFRJ), CCS - Sala C1-015, Av. Carlos Chagas Filho, 373 - Cidade Universitária, CEP 21.941-902, Ilha Do Fundão, Rio De Janeiro, RJ, Brazil, Tel: 55-21-2562-652 Email:

Abstract

Macrophages are present in almost all tissues of the body and are endowed with alternative differentiation programs resulting in a variety of terminal differentiated cells. They have role in the innate responses as well as in development and maintenance of adaptive immunity against invading pathogens. These cells have phagocytic activity and can sense the microenvironmental stimuli including microbial components that result in differentiation of distinct marker expression patterns and functions that clearly define macrophage subsets. Here we review the functional plasticity of macrophages in response to infections and their integration into adaptive immunity.

Keywords: Macrophages; Infections; Adaptive immunity; Homeostasis; Polarization

Introduction

One of the first descriptions of the immune system function was evidenced by the characterization of macrophages as cells with phagocytic activity by Metchnikoff [1]. These cells are present in different tissues from the organism as resident macrophages, encompassing the mononuclear phagocytic system. As these cells have a wide tissue distribution, they play a role not only in the immune responses but also during development, homeostasis and repair of the different tissues [2,3].

Recent studies are beginning to uncover the transcriptional regulation of the tissue-specific macrophages [4-6]. The cellular heterogeneity of these cells has also raised questions regarding their origin. A long-held dogma in the field has been assumed that all tissue-resident macrophages could be derived from local differentiation of circulating monocytes [7]. In this line of thinking, the tissue resident macrophages were thought to be exclusively derived from circulating monocytes found in the blood. These precursor cells are differentiated from bone marrow progenitors and have such plasticity to differentiate in a wide range of macrophage subsets with distinct phenotype and function profiles.

However, recent studies have provided conclusive evidences for the existence of a monocyte-independent differentiation pathway of resident macrophages, leading to a shift a in the paradigm of this model [8,9]. Although the term macrophage refers to multiple differentiation states in the ontogeny of these cells, when stimulated in polarizing conditions the macrophages can present dichotomous profiles between two states of the classical inflammatory responses [8,9]. The polarized categories are referred to as M1 and M2, which are both, defined in the context of cytokines and innate receptors present during the activation of macrophages, such as Toll-like receptors (TLRs), and the cytokines interferon-γ (IFN-γ), interleukin-4 (IL-4) and IL-13. In extreme polarizing conditions, the M1 phenotype is induced by Th1-derived interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α) or toll-like receptor ligands (TLR), showing a cytokine expression profile for inflammatory mediators (IL-12, TNF-α and IL-23) [10,11]. M1 macrophages produce high amounts of microbicide products such as nitric oxide (NO) and/or reactive oxygen intermediators (ROS) and mediate cell-mediated immunity in intracellular infection responses [10]. Beside its role on innate responses, the M1 cytokines are critical to promote increased phagocytic activity of macrophages, augmented MHC class II expression and co-stimulatory receptors that collectively have a role on the activation of the antigen processing and presentation to T cells [10,11].

The differentiation of M2 macrophages, however, is dependent of Th2-derived IL-4 responses found in the context of extracellular parasitic infection, allergies or healing-type circumstance without infections. The M2 phenotype can also be amplified in a feedback loop mechanism by IL-4, IL-10 and/or IL-13. These anti-inflammatory macrophage subsets are well characterized by the up-regulation of Dectin-1, DC-SIGN, mannose receptor, scavenger receptor A, scavenger receptor B-1, CD163, CCR2, CXCR1, CXCR2 and LIGHT [11]. Alternatively activated macrophages are permissive to parasites due to its inhibitory effect on the IL-12 expression, which is determinant for the induction of pro-inflammatory IFN-γ dependent responses [11].

The range between these two categories of macrophage is in fact represented by a wide variation in the transition state of the cellular differentiation program as a result of a complex sort of numerous other cytokines and innate receptor stimulation present in the inflammatory sites of infection whose influence are determinant for the final activation state of these cells. In fact, tissue-resident macrophages show high transcriptional diversity with slightest overlap that reinforce their different categories of cells acting as sentinels and promptly responding to disturbances in the physiological homeostasis of the tissues as well as to threats from invading microorganisms. This review will focus on macrophage responses to pathogens taking into account the diversity of their different lineages and its functional adaptation to different battle scenario requested in every type of immune response in infectious diseases.

Signaling Pathways Governing the Macrophage Polarization

Recent studies have characterized several signaling pathways implicated in the regulation of macrophage polarization. The JAK-STAT signaling pathway consist an important via mediating responses to the cytokine-induced macrophage polarization. The signaling pathway mediated by IFN-γ during M1 polarization activates the receptor-associated STAT1 necessary to induce the transcription of pro-inflammatory cytokines [12-14]. The Interferon-regulatory factor 5 (IRF5) is one of the proteins induced by IFN-γ that endorse the development of Th1 responses by promoting the transcription of IL-12-encoded genes responsible for induction of Th1 responses while inhibiting those that promote development of Th2 cells [15,16].

The polarization towards the M1 phenotype is also accompanied by the expression of Th1-attracting chemokines such as CXCL9 and CXCL10 [17,18]. It has been shown that the granulocyte-macrophage colony-stimulating factor (GM-CSF) controls the polarization of macrophages by leading to downstream activation of IRF5 during M1 development [16-19] . Moreover, the activation of IFN-γ-induced genes increases production of reactive oxygen species that culminates in the intracellular pathogen elimination by M1 macrophages [20]. In contrast, the polarization of M2 phenotype depends on the IL-4 and IL-13-induced STAT6 pathway and is repressed by the effects of SHIP (SH2-containingInositol 5′-Phosphatase), a component of growth factor receptor signaling that is shown to inhibit the IL-4 production from basophils [21].

Unlike to the M1 pathway, the peroxisome proliferator-activated receptors (PPARs) are activated to promote the polarization of differentiating macrophages toward the anti-inflammatory M2 phenotype [22,23]. PPARs are nuclear receptors that induce signaling and transcription of different pathways [24]. Overall, they participate in the regulation of lipid metabolism and glucose homeostasis, and are also activated by specific ligands [24,25]. The family of PPARs is mostly composed of three known isoforms: PPARα, PPARβ/δ, and PPARγ. These receptors share a structural homology that consists of four functional units (A,B,C, and D) [24-26].

The unit A/B of PPARs are located in N-terminal region of the receptor and controls the activation domain by AF-1 ligand. The units C and D represent a DNA binding domain that includes two zinc fingers motives and a docking domain [24-26]. The C-terminal region contains a specific binding domain and a transactivation domain for AF-2 [25]. This region is very important for nuclear localization of PPARs and other interactions with activator factors in the signaling pathway of these receptors [24-26]. The binding of specific-PPAR receptor agonists leads to association between PPARs and retinoic acid receptor (RXR). This receptor-associated heterodimers bind to specific PPRE regions of DNA to activate different target genes [27]. In addition, these receptor heterodimers can interact with other co-activator proteins such as CBP/p300, SRC1, PBP, and PGC-1α to induce a specific gene expression [26,27].

PPARγ play an important role in modulating macrophage M2 polarization induced by IL-4 or IL-13 [28]. Studies using PPARγ-deficient macrophages have shown the role of this nuclear receptor in promoting M2 activation to protect mice from insulin resistance [22]. A similar role was also found for PPARδ in determination of macrophage polarization [29]. Using the myeloid specific transcription factor (KLF-4) knockout mice [30], demonstrated the role of KLF-4 during M2 polarization in a protection model from obesity-induced insulin resistance. Similarly, IRF4 is also involved in regulating the expression of genes associated with M2 polarization [30].

Another signaling pathway involved in M2 differentiation relies on the activation of the phosphoinositol-3-kinase (PI3K) signaling pathway. PI3K activates multiple cascades through phosphorylation of the hydroxyl group of the inositol ring of phosphatidylinositol (PtdIns) to generate the second messenger phosphatidylinositol (3,4,5)-trisphosphate (PIP3) [11,31]. This pathway controls the activation of mTOR pathway, which is determinant for differentiation of M2 macrophages expressing anti-inflammatory cytokines. Importantly, the suppressor of cytokine signaling 1 (SOCS1), a member of the STAT-induced STAT inhibitor (SSI), is upregulated by IL-4 and mediates inhibition of IFN-gamma-induced STAT1 and is essential in sustaining the enhanced PI3K signaling pathway activity that promotes the M2 polarization responses [21].

Role of Macrophage Polarization in Infectious Diseases

The macrophages have an important role in both innate and adaptive immune responses as these cells acquire different ways to sense the presence of pathogens in every tissue of the organism [32]. The Toll-like receptors (TLRs) and other pattern recognition receptors (PRRs) are determinant to discriminate the presence of Pathogen-associated molecular patterns (PAMPs), which are molecules associated with groups of pathogens [33]. These components can be referred to as molecular motifs conserved within a class of microbes and recognized by the innate TLR and PRR receptors present in the macrophage and other cells of the immune system as well. Once engaged by their ligands, the innate receptors promote the acquisition of macrophage`s microbicidal activity against the pathogens [34-36].

Given the critical role of macrophages in the host defense, several pathogens evolved strategies to subvert the macrophage differentiation program by altering the M1 and M2 phenotype commitment in their favor. Bacteria that infect the host intracellular compartment such as Salmonella typhimurium and Mycobacterium turbeculosis are adapted to avoid the classically activated M1 cells by subverting the pro-inflammatory differentiation program of macrophages in order to enhance their own survival [37,38]. The outcome of this interference in the host phagocytic system has been well studied during murine pulmonary infection with Staphylococcus aureus. These pathogenic bacteria activate the PI3K pathway to promote SOCS1 signaling thus avoiding the differentiation of an anti-inflammatory M1 phenotype [39].

The M1 macrophage differentiation program is overall correlated with protection against intracellular pathogens. This is the case of typhoid fever which is caused by infection with the bacteria Salmonella typhi. This microbe induces M1 polarization during protective-mediated response against the infection. The production of reactive nitrogen species such as nitric oxide (NO) in M1 cells is also known to play a critical role in the intracellular killing mediated-responses against Salmonella infection [40]. Further studies have also demonstrated a critical role of IFNγ depended-M1 polarization responses on the host protective immune responses against mycobacteria and chlamydial infections [41-44].

Other pathogens such as viruses can employ different strategies exerted by bacteria species to increase the disease severity by promoting the inflammatory activity of M1 macrophages. Chronic viruses such as Hepatitis C virus stablish persistent infections with sustained inflammatory responses along the disease. The mechanism underlined in this event partially depends on the expression of the viral protein NS3 along with recombinant GP96 that increases IL-12 and TNF-α secretion profile of differentiating M1 macrophage [45]. In addition, this polarization effect can be seen in avian H5N1 influenza virus infection in which augmented levels of the pro-inflammatory cytokines IL-1β, IL-6, TNFα and IFNγ are implicated in M1 polarization and exacerbation of the infection [46-48].

Notably, viruses such as HIV-1 and Human cytomegalovirus (HCMV) take advantage from the M2 macrophage subset. These cells consist of an important reservoir of replication for both viruses as they have weak microbial activity thus enhancing the viral burden. Moreover, the HCMV infection is able to polarize the macrophage differentiation towards the M2 phenotype through activation of mTOR pathway [49]. The use of M2 cells as a reservoir is an evasion strategy also shared by the intracellular protozoan parasites. It has been shown that the uptake of apoptotic cells by T. cruzi infected macrophages promotes an anti-inflammatory state of these host phagocyte cells that are permissive to parasite replication [50-54].

The modulation of the mononuclear phagocytic system by T. cruzi parasites depends on the induction of prostaglandins, transforming growth factor-beta (TGF-β), arginase and polyamine biosynthesis to reduce the inflammation and microbicidal functions of macrophages [51,53,54]. It is possible that these mechanisms exert a regulatory role in the primary site of infection by jeopardizing the phagocytic responses of tissue resident macrophages thus increasing the parasite spread through host tissues. This adaptation is also seen in infection with the trypanosomatids protozoan Leishmania in which efferocytosis of apoptotic neutrophils modulate the macrophage activation and microbicidal activity therefore favoring the parasite growth inside the phagocytic cells [55-59]. Macrophage responses to microbial and immunological stimuli lead to discrete, stereotyped phenotypes [57]. Classically activated, or M1, macrophages are microbicidal, while alternatively activated (M2) macrophages are permissive to parasites [57-59]. These polarized states of activation represent a conceptual model for understanding the extremes of the cellular differentiation program capabilities of macrophages. It is therefore possible to conceive a range of potential intermediate phenotypes although their phenotypic characterizations are not well demonstrated. Importantly, the stereotypic M1/M2 macrophage profiles provide insights into the role of these cells in the physiologic and pathologic responses of the immune system.

Conclusions

The immunity is equalized between the strength of inflammatory responses and the regulatory counterparts that limit the side effects of the host defense system. The M1 and M2 activities are the essence of this balance, as these cells are able to participate in all the instances of immune responses. The macrophages are endowed with the capacity of sensing microbial components and host-derived factors during the first steps of pathogen–host cell invasion interactions that play a determinant role in the phagocyte differentiation pathways. The different macrophage commitments might be important to the role of these cells in reshaping the subsequent responses to microbial encounters during acquisition of adaptive immunity and its homeostasis. Understanding the paradigm of alternative activation pathways of macrophage differentiation will help us to clarify the role of these cells in the disease pathogenesis making them ideal for therapeutic targets.

References

  1. Tauber AI (2003) Metchnikoff and the phagocytosis theory. Nat Rev Mol Cell Biol 4: 897-901.
  2. Davies LC, Rosas M, Jenkins SJ, Liao CT, Scurr MJ, et al. (2013) Distinct bone marrow-derived and tissue-resident macrophage lineages proliferate at key stages during inflammation. Nat Commun 4: 1886.
  3. Gautier EL, Chow A, Spanbroek R, Marcelin G, Greter M, et al. (2012) Systemic analysis of PPARgamma in mouse macrophage populations reveals marked diversity in expression with critical roles in resolution of inflammation and airway immunity. J Immunol 189: 2614-2624.
  4. Haldar M, Murphy KM (2014) Origin, development, and homeostasis of tissue-resident macrophages. Immunol Rev 262: 25-35.
  5. Okabe Y, Medzhitov R (2014) Tissue-specific signals control reversible program of localization and functional polarization of macrophages. Cell 157: 832-844.
  6. A-Gonzalez N, Guillen JA, Gallardo G, Diaz M, de la Rosa JV, et al. (2013) The nuclear receptor LXRalpha controls the functional specialization of splenic macrophages. Nat Immunol 14: 831-839.
  7. van Furth R, Cohn ZA, Hirsch JG, Humphrey JH, Spector WG, et al. (1972) The mononuclear phagocyte system: a new classification of macrophages, monocytes, and their precursor cells. Bull World Health Organ 46: 845-852.
  8. Ginhoux F, Jung S (2014) Monocytes and macrophages: developmental pathways and tissue homeostasis. Nat Rev Immunol 14: 392-404.
  9. Schulz C, Gomez Perdiguero E, Chorro L, Szabo-Rogers H, Cagnard N, et al. (2012) A lineage of myeloid cells independent of Myb and hematopoietic stem cells. Science 336: 86-90.
  10. Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M, et al. (2004) Human IL-23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages subvert immunity to (myco)bacteria. Proc Natl Acad Sci U S A 101: 4560-4565.
  11. Martinez FO, Helming L, Gordon S (2009) Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 27: 451-483.
  12. Lawrence T, Natoli G (2011) Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat Rev Immunol 11: 750-761.
  13. Qin H, Yeh WI, De Sarno P, Holdbrooks AT, Liu Y, et al. (2012) Signal transducer and activator of transcription-3/suppressor of cytokine signaling-3 (STAT3/SOCS3) axis in myeloid cells regulates neuroinflammation. Proc Natl Acad Sci U S A 109: 5004-5009.
  14. Qin H, Holdbrooks AT, Liu Y, Reynolds SL, Yanagisawa LL, et al. (2012) SOCS3 deficiency promotes M1 macrophage polarization and inflammation. J Immunol 189: 3439-3448.
  15. Krausgruber T1, Blazek K, Smallie T, Alzabin S, Lockstone H, et al. (2011) IRF5 promotes inflammatory macrophage polarization and TH1-TH17 responses. Nat Immunol 12: 231-238.
  16. Weiss M, Blazek K, Byrne AJ, Perocheau DP, Udalova IA (2013) IRF5 is a specific marker of inflammatory macrophages in vivo. Mediators Inflamm 2013: 245804.
  17. Martinez FO, Gordon S, Locati M, Mantovani A (2006) Transcriptional profiling of the human monocyte-to-macrophage differentiation and polarization: new molecules and patterns of gene expression. J Immunol 177: 7303-7311.
  18. Biswas SK, Mantovani A (2010) Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm. Nat Immunol 11: 889-896.
  19. Gazzinelli RT, Oswald IP, Hieny S, James SL, Sher A (1992) The microbicidal activity of interferon-gamma-treated macrophages against Trypanosoma cruzi involves an L-arginine-dependent, nitrogen oxide-mediated mechanism inhibitable by interleukin-10 and transforming growth factor-beta. Eur J Immunol 22: 2501-2506.
  20. Murray HW, Rubin BY, Rothermel CD (1983) Killing of intracellular Leishmania donovani by lymphokine-stimulated human mononuclear phagocytes. Evidence that interferon-gamma is the activating lymphokine. J Clin Invest 72: 1506-1510.
  21. Kuroda E, Ho V, Ruschmann J, Antignano F, Hamilton M, et al. (2009) SHIP represses the generation of IL-3-induced M2 macrophages by inhibiting IL-4 production from basophils. J Immunol 183: 3652-3660.
  22. Odegaard JI, Ricardo-Gonzalez RR, Goforth MH, Morel CR, Subramanian V, et al. (2007) Macrophage-specific PPARgamma controls alternative activation and improves insulin resistance. Nature 447: 1116-1120.
  23. Bouhlel MA, Derudas B, Rigamonti E, Dièvart R, Brozek J, et al. (2007) PPARgamma activation primes human monocytes into alternative M2 macrophages with anti-inflammatory properties. Cell Metab 6: 137-143.
  24. Rosen ED, Spiegelman BM (2001) PPARgamma : a nuclear regulator of metabolism, differentiation, and cell growth. J Biol Chem 276: 37731-37734.
  25. Kota BP, Huang TH, Roufogalis BD (2005) An overview on biological mechanisms of PPARs. Pharmacol Res 51: 85-94.
  26. Ahmadian M, Suh JM, Hah N, Liddle C, Atkins AR, et al. (2013) PPARγ signaling and metabolism: the good, the bad and the future. Nat Med 19: 557-566.
  27. Forman BM, Chen J, Evans RM (1996) The peroxisome proliferator-activated receptors: ligands and activators. Ann N Y Acad Sci 804: 266-275.
  28. Luzina IG, Keegan AD, Heller NM, Rook GA, Shea-Donohue T, et al. (2012) Regulation of inflammation by interleukin-4: a review of "alternatives". J Leukoc Biol 92: 753-764.
  29. Mukundan L, Odegaard JI, Morel CR, Heredia JE, Mwangi JW, et al. (2009) PPAR-delta senses and orchestrates clearance of apoptotic cells to promote tolerance. Nat Med 15: 1266-1272.
  30. Eguchi J, Wang X, Yu S, Kershaw EE, Chiu PC, et al. (2011) Transcriptional control of adipose lipid handling by IRF4. Cell Metab 13: 249-259.
  31. Gordon S, Martinez FO (2010) Alternative activation of macrophages: mechanism and functions. Immunity 32: 593-604.
  32. Arango Duque G, Descoteaux A (2014) Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol 5: 491.
  33. Janeway CA Jr, Medzhitov R (2002) Innate immune recognition. Annu Rev Immunol 20: 197-216.
  34. Kaisho T, Akira S (2000) Critical roles of Toll-like receptors in host defense. Crit Rev Immunol 20: 393-405.
  35. Underhill DM (2003) Macrophage recognition of zymosan particles. J Endotoxin Res 9: 176-180.
  36. Yáñez A, Goodridge HS, Gozalbo D, Gil ML (2013) TLRs control hematopoiesis during infection. Eur J Immunol 43: 2526-2533.
  37. Lee KS, Jeong ES, Heo SH, Seo JH, Jeong DG, et al. (2011) IL-10 suppresses bactericidal response of macrophages against Salmonella Typhimurium. J Microbiol 49: 1050-1053.
  38. Lopes RL, Borges TJ, Araújo JF, Pinho NG, Bergamin LS, et al. (2014) Extracellular mycobacterial DnaK polarizes macrophages to the M2-like phenotype. PLoS One 9: e113441.
  39. Xu F, Kang Y, Zhang H, Piao Z, Yin H, et al. (2013) Akt1-mediated regulation of macrophage polarization in a murine model of Staphylococcus aureus pulmonary infection. J Infect Dis 208: 528-538.
  40. Kyrova K, Stepanova H, Rychlik I, Faldyna M, Volf J (2012) SPI-1 encoded genes of Salmonella Typhimurium influence differential polarization of porcine alveolar macrophages in vitro. BMC Vet Res 8: 115.
  41. Rottenberg ME, Gigliotti-Rothfuchs A, Wigzell H (2002) The role of IFN-gamma in the outcome of chlamydial infection. Curr Opin Immunol 14: 444-451.
  42. Ehrt S, Schnappinger D, Bekiranov S, Drenkow J, Shi S, et al. (2001) Reprogramming of the macrophage transcriptome in response to interferon-gamma and Mycobacterium tuberculosis: signaling roles of nitric oxide synthase-2 and phagocyte oxidase. J Exp Med 194: 1123-1140.
  43. Chacón-Salinas R, Serafín-López J, Ramos-Payán R, Méndez-Aragón P, Hernández-Pando R (2005) Differential pattern of cytokine expression by macrophages infected in vitro with different Mycobacterium tuberculosis genotypes. Clin Exp Immunol 140: 443-449.
  44. Murphy JT, Sommer S, Kabara EA, Verman N, Kuelbs MA, et al. (2006) Gene expression profiling of monocyte-derived macrophages following infection with Mycobacterium avium subspecies avium and Mycobacterium avium subspecies paratuberculosis. Physiol Genomics 28: 67-75.
  45. Hajizadeh MR, Mokarram P, sarvestani EK , Bolhassani A, Pour ZM (2013) Recombinant Nonstructural 3 Protein, rNS3, of Hepatitis C Virus Along With Recombinant GP96 Induce IL-12, TNFalpha and alpha5integrin Expression in Antigen Presenting Cells. Hepat Mon 13: e8104.
  46. Szretter KJ, Balish AL, Katz JM (2006) Influenza: propagation, quantification, and storage. Curr Protoc Microbiol Chapter 15: Unit 15G 11.
  47. Perrone LA, Szretter KJ, Katz JM, Mizgerd JP, Tumpey TM (2010) Mice lacking both TNF and IL-1 receptors exhibit reduced lung inflammation and delay in onset of death following infection with a highly virulent H5N1 virus. J Infect Dis 202: 1161-1170.
  48. Lemon K, de Vries RD, Mesman AW, McQuaid S, van Amerongen G, et al. (2011) Early target cells of measles virus after aerosol infection of non-human primates. PLoS Pathog 7: e1001263.
  49. Poglitsch M, Weichhart T, Hecking M, Werzowa J, Katholnig K, et al. (2012) CMV late phase-induced mTOR activation is essential for efficient virus replication in polarized human macrophages. Am J Transplant 12: 1458-1468.
  50. Luna-Gomes T, Filardy AA, Rocha JD, Decote-Ricardo D, LaRocque-de-Freitas IF, et al. (2014) Neutrophils increase or reduce parasite burden in Trypanosoma cruzi-infected macrophages, depending on host strain: role of neutrophil elastase. PLoS One 9: e90582.
  51. D'Avila H, Freire-de-Lima CG, Roque NR, Teixeira L, Barja-Fidalgo C, et al. (2011) Host cell lipid bodies triggered by Trypanosoma cruzi infection and enhanced by the uptake of apoptotic cells are associated with prostaglandin E(2) generation and increased parasite growth. J Infect Dis 204: 951-961.
  52. Freire-de-Lima CG, Xiao YQ, Gardai SJ, Bratton DL, Schiemann WP, et al. (2006) Apoptotic cells, through transforming growth factor-beta, coordinately induce anti-inflammatory and suppress pro-inflammatory eicosanoid and NO synthesis in murine macrophages. J Biol Chem 281:38376-38384.
  53. Freire-de-Lima CG, Nascimento DO, Soares MB, Bozza PT, Castro-Faria-Neto HC, et al. (2000) Uptake of apoptotic cells drives the growth of a pathogenic trypanosome in macrophages. Nature 403: 199-203.
  54. Lopes MF, Freire-de-Lima CG, DosReis GA (2000) The macrophage haunted by cell ghosts: a pathogen grows. Immunol Today 21: 489-494.
  55. Lopes MF, Costa-da-Silva AC, DosReis GA (2014) Innate immunity to Leishmania infection: within phagocytes. Mediators Inflamm 2014: 754965.
  56. Filardy AA, Pires DR, Nunes MP, Takiya CM, Freire-de-Lima CG, et al. (2010) Proinflammatory clearance of apoptotic neutrophils induces an IL-12(low)IL-10(high) regulatory phenotype in macrophages. J Immunol 185: 2044-2050.
  57. Filardy AA, Costa-da-Silva AC, Koeller CM, Guimarães-Pinto K, Ribeiro-Gomes FL, et al. (2014) Infection with Leishmania major induces a cellular stress response in macrophages. PLoS One 9: e85715.
  58. Afonso L, Borges VM, Cruz H, Ribeiro-Gomes FL, DosReis GA, et al. (2008) Interactions with apoptotic but not with necrotic neutrophils increase parasite burden in human macrophages infected with Leishmania amazonensis. J Leukoc Biol 84: 389-396.
  59. Ribeiro-Gomes FL, Otero AC, Gomes NA, Moniz-De-Souza MC, Cysne-Finkelstein L, et al. (2004) Macrophage interactions with neutrophils regulate Leishmania major infection. J Immunol 172: 4454-4462.
Citation: Decote-Ricardo D, Freire-de-Lima L, Morrot A, Freire-de-Lima CG (2015) Macrophage Polarization in Infectious Diseases. J Clin Cell Immunol 6:294.

Copyright: © 2015 Decote-Ricardo D, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top