Hostname: page-component-7479d7b7d-m9pkr Total loading time: 0 Render date: 2024-07-10T11:38:38.611Z Has data issue: false hasContentIssue false

Immunological mechanisms involved in macrophage activation and polarization in schistosomiasis

Published online by Cambridge University Press:  05 January 2023

Irlla Correia Lima Licá
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Gleycka Cristine Carvalho Gomes Frazão
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Ranielly Araujo Nogueira
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Maria Gabriela Sampaio Lira
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Vitor Augusto Ferreira dos Santos
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
João Gustavo Mendes Rodrigues
Affiliation:
Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
Guilherme Silva Miranda
Affiliation:
Department of Biology, Federal Institute of Education, Science and Technology of Maranhão, São Raimundo das Mangabeiras, Brazil
Rafael Cardoso Carvalho
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Lucilene Amorim Silva
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Rosane Nassar Meireles Guerra
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
Flávia Raquel Fernandes Nascimento*
Affiliation:
Graduate Program in Health Sciences, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Laboratory of Immunophysiology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil Department of Pathology, Center for Biological and Health Sciences, Federal University of Maranhão, São Luís, MA, Brazil
*
Author for correspondence: Flávia Raquel Fernandes Nascimento, E-mail: flavia.nascimento@ufma.br

Abstract

Human schistosomiasis is caused by helminths of the genus Schistosoma. Macrophages play a crucial role in the immune regulation of this disease. These cells acquire different phenotypes depending on the type of stimulus they receive. M1 macrophages can be ‘classically activated’ and can display a proinflammatory phenotype. M2 or ‘alternatively activated’ macrophages are considered anti-inflammatory cells. Despite the relevance of macrophages in controlling infections, the role of the functional types of these cells in schistosomiasis is unclear. This review highlights different molecules and/or macrophage activation and polarization pathways during Schistosoma mansoni and Schistosoma japonicum infection. This review is based on original and review articles obtained through searches in major databases, including Scopus, Google Scholar, ACS, PubMed, Wiley, Scielo, Web of Science, LILACS and ScienceDirect. Our findings emphasize the importance of S. mansoni and S. japonicum antigens in macrophage polarization, as they exert immunomodulatory effects in different stages of the disease and are therefore important as therapeutic targets for schistosomiasis and in vaccine development. A combination of different antigens can provide greater protection, as it possibly stimulates an adequate immune response for an M1 or M2 profile and leads to host resistance; however, this warrants in vitro and in vivo studies.

Type
Review Article
Creative Commons
Creative Common License - CCCreative Common License - BY
This is an Open Access article, distributed under the terms of the Creative Commons Attribution licence (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted re-use, distribution and reproduction, provided the original article is properly cited.
Copyright
Copyright © The Author(s), 2023. Published by Cambridge University Press

Introduction

Human schistosomiasis is a neglected parasitic disease with great relevance to public health. Worldwide, it is estimated that approximately 230–250 people are infected and 700–800 million live in areas that are at risk of infection, mainly in countries located in South America, Asia and Africa (Steinmann et al., Reference Steinmann, Keiser, Bos, Tanner and Utzinger2006; Colley et al., Reference Colley, Bustinduy, Secor and King2014; McManus et al., Reference McManus, Dunne, Sacko, Utzinger, Vennervald and Zhou2018; Wei et al., Reference Wei, Huang, Chen, Chen, Li, Xu and Yang2018; WHO, 2020). In addition, approximately 200 000–280 000 deaths occur each year due to schistosomiasis and its complications (LoVerde, Reference LoVerde2019). The high prevalence of schistosomiasis is mainly related to people living in extreme poverty and poor sanitation, which represent a serious risk to human health (Ismail et al., Reference Ismail, Hong, Babiker, Hassan, Sulaiman, Jeong, Kong, Lee, Cho, Nam, Oh and Lee2014; Bajiro et al., Reference Bajiro, Dana and Levecke2017; Verjee, Reference Verjee2019).

The infection is caused by helminths of the genus Schistosoma (Colley et al., Reference Colley, Bustinduy, Secor and King2014; Stingl and Stingl, Reference Stingl and Stingl2017; WHO, 2020), belonging to the class Trematoda and phylum Platyhelminthes. The main aetiologic agents of this disease, in terms of clinical relevance, are Schistosoma japonicum, Schistosoma mansoni and Schistosoma haematobium (WHO, 2020). In this review, we focus only on S. mansoni and S. japonicum, as they are the main species associated with hepatic and intestinal schistosomiasis (Wilson et al., Reference Wilson, Mentink-Kane, Pesce, Ramalingam, Thompson and Wynn2007; Chen et al., Reference Chen, Peng, Li, Yang, Wang and Chen2013; McManus et al., Reference McManus, Dunne, Sacko, Utzinger, Vennervald and Zhou2018).

There are 2 distinct phases of clinical progression of intestinal schistosomiasis: the acute and the chronic phases (Gobbi et al., Reference Gobbi, Tamarozzi, Buonfrate, van Lieshout, Bisoffi and Bottieau2020). During the early stages of acute phase of schistosomiasis (before parasite oviposition), there is a predominance of the T helper type 1 (Th1) immune response (Pearce et al., Reference Pearce, Caspar, Grzych, Lewis and Sher1991; Hesse et al., Reference Hesse, Modolell, La Flamme, Schito, Fuentes, Cheever, Pearce and Wynn2001; Pearce and MacDonald, Reference Pearce and MacDonald2002; Colley and Secor, Reference Colley and Secor2014). After schistosome oviposition, the immune response becomes strongly polarized to the Th2 profile, which is related to increasing production of interleukin-4 (IL-4), IL-5, IL-9 and IL-13 (Pearce and MacDonald, Reference Pearce and MacDonald2002; Burke et al., Reference Burke, Jones, Gobert, Li, Ellis and McManus2009). This immune environment is responsible for the formation of granulomas in tissues (Grzych et al., Reference Grzych, Pearce, Cheever, Caulada, Caspar, Heiny, Lewis and Sher1991; Brunet et al., Reference Brunet, Finkelman, Cheever, Kopf and Pearce1997; Hoffmann et al., Reference Hoffmann, Cheever and Wynn2000). The granuloma has an important role for the host, because it contains the tissue damage caused by antigens secreted by the schistosome eggs (Hams et al., Reference Hams, Aviello and Fallon2013; Schwartz and Fallon, Reference Schwartz and Fallon2018). In the chronic phase of schistosomiasis, there is an increase in the production of regulatory cells in the granuloma, which can modulate granulomatous inflammation, promoting a minimization of the disease severity (Hesse et al., Reference Hesse, Piccirillo, Belkaid, Prufer, Mentink-Kane, Leusink, Cheever, Shevach and Wynn2004; Lundy and Lukacs, Reference Lundy and Lukacs2013). However, if this inflammatory reaction does not have an adequate modulation, the granulomas progressively may evolve into large areas of fibrosis, responsible for the main pathology of schistosomiasis (Hams et al., Reference Hams, Aviello and Fallon2013; Schwartz and Fallon, Reference Schwartz and Fallon2018), including hepatosplenomegaly (Masi et al., Reference Masi, Perles-Barbacaru, Bernard and Viola2020), portal hypertension (Grieco et al., Reference Grieco, Sulekova, Nardelli, Riggio, Venditti and Taliani2016) and ascites (Fei-Yue et al., Reference Fei-Yue, Hong-Zhuan, Jie, Rui-Hong, Jin-Hua, Xin-Ting and Guang-Hui2017).

Macrophages are cells of the innate immune system that play important roles in controlling infections (Shapouri-Moghaddam et al., Reference Shapouri-Moghaddam, Mohammadian, Vazini, Taghadosi, Esmaeili, Mardani, Seifi, Mohammadi, Afshari and Sahebkar2018), as well as in tissue remodelling processes, both in ontogenesis and wound healing (Kloc et al., Reference Kloc, Ghobrial, Wosik, Lewicka, Lewicki and Kubiak2019). In the course of S. mansoni and S. japonicum infection, either at its initial stage or during the evolution to the chronic phase, macrophages participate in the immune regulation of the disease (Cortes-Selva et al., Reference Cortes-Selva, Elvington, Ready, Rajwa, Pearce, Randolph and Fairfax2018; Ho et al., Reference Ho, Cheng, Huang, Peng, Lee and Cheng2022).

Macrophages can acquire different phenotypes depending on the stimuli to which they are subjected to (Atri et al., Reference Atri, Guerfali and Laouini2018). These cells can be classified into M1 or ‘classically activated’ cells, with pro-inflammatory action, and M2 or ‘alternatively activated’ macrophages, which are considered anti-inflammatory cells (Mills, Reference Mills2015; Ley, Reference Ley2017; Locati et al., Reference Locati, Curtale and Mantovani2020). However, despite the relevance of macrophages in controlling infections, the participation of the functional types of these cells in acute and chronic schistosomiasis is still not well defined. Thus, this review discusses the different molecules and/or pathways of activation and polarization of macrophages during infection by S. mansoni and S. japonicum, leading to a better understanding of the role of these cells in the immunopathology of schistosomiasis. Based on this knowledge, we may help identify potential targets for the development of better treatment strategies to reduce the morbidity of this disease.

Methods and criteria for literature selection

This literature review was performed using recognized databases including Scopus, Google Scholar, ACS, PubMed, Wiley, Scielo, Web of Science, LILACS and ScienceDirect and covered original and review articles published in English from 1966 to 2022. Articles involving in vitro and/or in vivo experiments were included and addressed the main immunological aspects of S. mansoni and S. japonicum infection related to macrophage polarization, activation and effector functions. To search for these articles, combinations of keywords were used, such as ‘Macrophage’, ‘Schistosoma’, ‘macrophage polarization and Schistosoma’, ‘Macrophage and Schistosoma’, ‘Macrophage and Schistosoma mansoni’, ‘Macrophage and Schistosoma japonicum’. Research involving coinfections was not included in this study.

Immunopathology of the definitive host against infection by S. mansoni and S. japonicum

Parasites of the genus Schistosoma have complex life cycles (Fig. 1), with generations of asexual reproducing larvae living in freshwater snails, the intermediate hosts (some species of the genus Biomphalaria for S. mansoni and the genus Oncomelania for S. japonicum) and another stage of sexual reproduction of adult worms in vertebrate hosts (definitive), including humans (McManus et al., Reference McManus, Dunne, Sacko, Utzinger, Vennervald and Zhou2018; Nelwan, Reference Nelwan2019). Each stage of the parasite's life cycle (cercariae, schistosomulae, adult worms and eggs) within the definitive host triggers a series of immune responses, and consequently, clinical signs that can be harmful to humans (Molehin, Reference Molehin2020; Hambrook and Hanington, Reference Hambrook and Hanington2021; Masamba and Kappo, Reference Masamba and Kappo2021). The interactions between the host immune system and the parasite can be divided into 2 phases (Fig. 2): acute phase (after and before parasite oviposition) and chronic phase (Gobbi et al., Reference Gobbi, Tamarozzi, Buonfrate, van Lieshout, Bisoffi and Bottieau2020).

Fig. 1. Life cycle of Schistosoma mansoni and Schistosoma japonicum. (1) The eggs shed in the feces of the definitive host release the miracidia when they come in contact with water (2), which penetrate in soft tissue the intermediate host snail (Biomphalaria spp./Oncomelania spp.). Inside the snail, the miracidia transform into mother sporocysts, which in turn produce daughter sporocysts by asexual reproduction. After around 30 days post-infection, cercariae emerge from the daughter sporocysts and are shedding by the snails in response to the light and heat (4). The cercariae penetrate the skin of the definitive host (5) and later transform into schistosomula. These larvae enter venous blood vessels and are passively carried to the lungs and heart (6). Upon reaching the hepatic portal system, schistosomula mature, become adult worms (male or female) and mate (7). The mated worms migrate to the lower mesenteric veins of the intestine, where the female sheds the eggs. Part of these eggs pass through the intestinal wall and are eliminated in the feces, starting the cycle again. However, some eggs are not eliminated and get trapped in several organs (mainly the liver and intestines), inducing a potent granulomatous inflammatory response, responsible for schistosomiasis pathology. Source: Created with BioRender.com.

Fig. 2. Different immune response profiles during S. mansoni and S. japonicum infection. Source: Created with BioRender.com.

The first clinical manifestations of the acute phase (cercarial dermatitis, oedema and pruritus) begin 48–72 h after cercariae penetrate the host's skin, and occurs mainly in individuals from endemic areas (frequently exposed to infection) (He et al., Reference He, Yu, Yu, Mao and Hu1990, Reference He, Salafsky and Ramaswamy2005; Khammo et al., Reference Khammo, Bartlett, Clothier and Whitfield2002; Ingram et al., Reference Ingram, Bartlett, Brown, Marriott and Whitfield2003; Lambertucci, Reference Lambertucci2010). The first innate immune barrier encountered by cercariae is the skin (Bartlett et al., Reference Bartlett, Brown, Marriott and Whitfield2000; Whitfield et al., Reference Whitfield, Bartlett, Brown and Marriott2003; He et al., Reference He, Salafsky and Ramaswamy2005). This tissue is composed of keratinocytes, whose function is to secrete cytokines with antimicrobial functions (Roupé et al., Reference Roupé, Nybo, Sjöbring, Alberius, Schmidtchen and Sørensen2010; Piipponen et al., Reference Piipponen, Li and Landén2020). Indeed, the keratinocytes are considered the first active cells in response to cercariae infection (Bourke et al., Reference Bourke, Prendergast, Sanin, Oulton, Hall and Mountford2015). These cells rapidly respond to infections by secreting inflammatory cytokines [IL-6, IL-12, tumour necrosis factor-alpha (TNF-α) and IL-1β] to repair damaged tissue (Hogg et al., Reference Hogg, Kumkate, Anderson and Mountford2003a, Reference Hogg, Kumkate and Mountford2003b). When penetrating the host's skin, cercariae also cause an increase in antigen-presenting cells in the innate immune system, such as Langerhans cells and dendritic cells (DCs), as shown in Fig. 2 (Angeli et al., Reference Angeli, Faveeuw, Roye, Fontaine, Teissier, Capron, Wolowezuk, Capron and Trottein2001; Kumkate et al., Reference Kumkate, Jenkins, Paveley, Hogg and Mountford2007; Hambrook and Hanington, Reference Hambrook and Hanington2021), which contribute to a type 1 cellular immune response (He et al., Reference He, Salafsky and Ramaswamy2005; Perona-Wright et al., Reference Perona-Wright, Jenkins and MacDonald2006).

Initial immune responses are activated as a result of excretory/secretory (E/S) products released by the cercariae penetrating glands at the time of penetration into the host's skin (Salter et al., Reference Salter, Lim, Hansell, Hsieh and McKerrow2000; Jenkins et al., Reference Jenkins, Hewitson, Ferret-Bernard and Mountford2005a, Reference Jenkins, Hewitson, Jenkins and Mountford2005b; Curwen et al., Reference Curwen, Ashton, Sundaralingam and Wilson2006; Paveley et al., Reference Paveley, Aynsley, Cook, Turner and Mountford2009). E/S products assist in the immunomodulatory function exerted by cercariae, as well as condition the remodelling of the extracellular matrix, facilitating its penetration into the skin (Janssen et al., Reference Janssen, Silva Santos, Muller, Vieira, De Campos and De Paulo Martins2016; Leontovyč et al., Reference Leontovyč, Ulrychová, Horn and Dvořák2020). Liu et al. (Reference Liu, Ju, Du, Shen, Wang, Li, Zhang, Feng and Hu2015) performed a proteomic analysis of products excreted by S. japonicum cercariae at the time of skin entry and identified a variety of E/S proteins, mainly proteases. Among the enzymes that allow this remodelling, the cercarial elastase of S. mansoni stands out, which is of great importance in the penetration of cercariae into the skin and can degrade a wide variety of macromolecules present in the human integument (Ingram et al., Reference Ingram, Rafi, Eroy-Reveles, Ray, Lambeth, Hsieh, Ruelas, Lim, Sakanari, Craik, Jacobson and McKerrow2012; El-Faham et al., Reference El-Faham, Wheatcroft-Francklow, Price, Sayers and Doenhoff2017).

Parasitic E/S products also promote the activation of prostaglandin E2 (PGE2) and prostaglandin D2-producing keratinocytes (Kaisar et al., Reference Kaisar, Ritter, del Fresno, Jónasdóttir, van der Ham, Pelgrom, Schramm, Layland, Sancho, Prazeres da Costa, Giera, Yazdanbakhsh and Everts2018; Oyesola et al., Reference Oyesola, Shanahan, Kanke, Mooney, Webb, Smita, Matheson, Campioli, Pham, Früh, McGinty, Churchill, Cahoon, Sundaravaradan, Flitter, Mouli, Nadjsombati, Kamynina, Peng, Cubitt, Gronert, Lord, Rauch, von Moltke, Sethupathy and Tait Wojno2021), which are molecules that induce the production of IL-10 via a cyclooxygenase 2-dependent pathway (Ramaswamy et al., Reference Ramaswamy, Kumar and He2000; Harizi et al., Reference Harizi, Juzan, Pitard, Moreau and Gualde2002; Xue et al., Reference Xue, Gyles, Wettey, Gazi, Townsend, Hunter and Pettipher2005). This type of response is responsible for modulating the immune response that favours parasite survival (Angeli et al., Reference Angeli, Faveeuw, Roye, Fontaine, Teissier, Capron, Wolowezuk, Capron and Trottein2001; Hervé et al., Reference Hervé, Angeli, Pinzar, Wintjens, Faveeuw, Narumiya, Capron, Urade, Capron, Riveau and Trottein2003; De Oliveira Fraga et al., Reference De Oliveira Fraga, Torrero, Tocheva, Mitre and Davies2010). Abdel-Ghany et al. (Reference Abdel-Ghany, Rabia, El-Ahwany, Saber, Gamal, Nagy, Mahmoud, Hamad and Barakat2015) suggested that blocking PGE2 might provide partial protection in S. mansoni-infected mice. In addition, during the period when cercariae transform into schistosomules and migrate through the skin, PGE2 acts as a potent vasodilator, helping the passage of these larval forms into circulation (Ruzicka and Printz, Reference Ruzicka and Printz1984).

After penetrating the host's skin, cercariae undergo morphological and biochemical changes, transforming into juvenile forms, known as schistosomula, that reach blood vessels (Brink et al., Reference Brink, McLaren and Smithers1977; Wilson, Reference Wilson1987; Curwen and Wilson, Reference Curwen and Wilson2003). In the bloodstream, the schistosomula is passively transported to the lungs and heart until they finally reach the hepatic portal system, where they develop into adult male or female worms (Miller and Wilson, Reference Miller and Wilson1978; Wheater and Wilson, Reference Wheater and Wilson1979; Nation et al., Reference Nation, Da'dara, Marchant and Skelly2020) (Fig. 1). In this phase before the parasite's oviposition (early stages of acute phase), the host produces a predominantly type 1 immune response, which reaches greater activation between the 3rd and 5th weeks after exposure to cercariae (Dunne and Cooke, Reference Dunne and Cooke2005; Gryseels et al., Reference Gryseels, Polman, Clerinx and Kestens2006). This response is characterized by high production of pro-inflammatory cytokines, such as IL-1, IL-2, IL-6, IL-12, interferon-gamma (IFN-γ) and TNF-α (Fig. 2) (Grzych et al., Reference Grzych, Pearce, Cheever, Caulada, Caspar, Heiny, Lewis and Sher1991; Pearce et al., Reference Pearce, Caspar, Grzych, Lewis and Sher1991; Egesa et al., Reference Egesa, Lubyayi, Tukahebwa, Bagaya, Chalmers, Wilson, Hokke, Hoffmann, Dunne, Yazdanbakhsh, Labuda and Cose2018; Zheng et al., Reference Zheng, Zhang, Chen, Nie, Miller, Gong and Liu2020). Coinciding with the migration and sexual maturation of adult worms, a systemic hypersensitivity reaction occurs in the host, called Katayama syndrome (for S. japonicum) or the toxaemic form (for S. mansoni), which is associated with an intense Th1 response (Neves, 1992; Ross et al., Reference Ross, Vickers, Olds, Shah and McManus2007; Caldas et al., Reference Caldas, Campi-Azevedo, Oliveira, Silveira, Oliveira and Gazzinelli2008; Langenberg et al., Reference Langenberg, Hoogerwerf, Janse, Van Lieshout, Corstjens, Roestenberg, Van Dam, Van Diepen, De Dood, Feijt, Ganesh, Gerritsma, Hardeman, Hokke, Koopman, Kos-Van Oosterhoud, Kruize, Meij, Ozir-Fazalalikhan, Van Schuijlenburg, Smits, Verbeek-Menken, Visser, De Vries, Winkel and Yazdanbakhsh2019). During primary infections in non-immune individuals, the main symptoms related to this systemic inflammation include a high fever accompanied by chills, profuse sweating, asthenia, myalgia, headache and a non-productive cough (Schwartz et al., Reference Schwartz, Rozenman and Perelman2000; Bottieau et al., Reference Bottieau, Clerinx, de Vega, Van den Enden, Colebunders, Van Esbroeck, Vervoort, Van Gompel and Van den Ende2006).

After parasite oviposition (between 5th and 6th weeks post-infection), there is a change in the profile of immune mediators produced by the host, and the immune response becomes predominantly Th2, which is associated with increasing production of IL-4, IL-5, IL-9 and IL-13 (MacDonald et al., Reference MacDonald, Straw, Dalton and Pearce2002; Pearce et al., Reference Pearce, Kane, Sun, Taylor, McKee and Cervi2004; Bartley et al., Reference Bartley, Ramm, Jones, Ruddell, Li and McManus2006; Burke et al., Reference Burke, Jones, Gobert, Li, Ellis and McManus2009). Such changes are responses to soluble egg antigens (SEAs) (Hams et al., Reference Hams, Aviello and Fallon2013), that is composed of a complex mixture of immunostimulatory antigens that are known for their ability to condition DCs to initiate the induction of a Th2 profile (Mouser et al., Reference Mouser, Pollakis, Smits, Thomas, Yazdanbakhsh, De Jong and Paxton2019).

DCs detect, capture and process antigens derived from eggs of S. mansoni (Cervi et al., Reference Cervi, MacDonald, Kane, Dzierszinski and Pearce2004; van Liempt et al., Reference van Liempt, van Vliet, Engering, García Vallejo, Bank, Sanchez-Hernandez, van Kooyk and van Die2007), resulting in their ability to lead to Th2 polarization both in vitro and in vivo (de Jong et al., Reference de Jong, Vieira, Kalinski, Schuitemaker, Tanaka, Wierenga, Yazdanbakhsh and Kapsenberg2002; MacDonald et al., Reference MacDonald, Straw, Dalton and Pearce2002; Perona-Wright et al., Reference Perona-Wright, Jenkins and MacDonald2006). The main antigens responsible for this potent induction of a Th2 response are glycoproteins omega 1 (ω-1) and IPSE (IL-4-inducing principle of S. mansoni eggs)/alpha 1 (α-1) (Schramm et al., Reference Schramm, Gronow, Knobloch, Wippersteg, Grevelding, Galle, Fuller, Stanley, Chiodini, Haas and Doenhoff2006; Meevissen et al., Reference Meevissen, Wuhrer, Doenhoff, Schramm, Haas, Deelder and Hokke2010). Glycoprotein ω-1 is present in both SEAs (Dunne et al., Reference Dunne, Jones and Doenhoff1991) and E/S products from live eggs (Cass et al., Reference Cass, Johnson, Califf, Xu, Hernandez, Stadecker, Yates and Williams2007), and activates DCs (via C-type and Toll-type lectin receptors), which in turn promotes Th2 differentiation, the main source of type 2 cytokines such as IL-4, IL-5 and IL-13 (Everts et al., Reference Everts, Perona-Wright, Smits, Hokke, van der Ham, Fitzsimmons, Doenhoff, van der Bosch, Mohrs, Haas, Mohrs, Yazdanbakhsh and Schramm2009). On the other hand, a previous study (Schramm et al., Reference Schramm, Gronow, Knobloch, Wippersteg, Grevelding, Galle, Fuller, Stanley, Chiodini, Haas and Doenhoff2006) showed that the glycoprotein IPSE/α-1 is exclusively released from mature eggs, but likely possesses the same potential to initiate a Th2 response during S. mansoni infection. IPSE/α-1 binds to immunoglobulin and activates basophils, leading to the release of histamine and facilitating the production of Th2-type cytokines, mainly IL-4 and IL-13 (Schramm et al., Reference Schramm, Mohrs, Wodrich, Doenhoff, Pearce, Haas and Mohrs2007; Meyer et al., Reference Meyer, Mayerhofer, Tripsianes, Blindow, Barths, Mewes, Weimar, Köhli, Bade, Madl, Frey, Haas, Mueller-Dieckmann, Sattler and Schramm2015; Knuhr et al., Reference Knuhr, Langhans, Nyenhuis, Viertmann, Overgaard Kildemoes, Doenhoff, Haas and Schramm2018). Thus, the Th2 response (Fig. 2) is related to low production of IFN-γ and high concentrations of anti-inflammatory cytokines (IL-4, IL-5, IL-10 and IL-13) (Grzych et al., Reference Grzych, Pearce, Cheever, Caulada, Caspar, Heiny, Lewis and Sher1991; Pearce et al., Reference Pearce, Kane, Sun, Taylor, McKee and Cervi2004; Zheng et al., Reference Zheng, Zhang, Chen, Nie, Miller, Gong and Liu2020).

Mechanisms associated with macrophage polarization

Macrophages are cells of the innate immune system that have phagocytic capacity and are involved in the elimination of foreign particles from the body (Gordon and Martinez-Pomares, Reference Gordon and Martinez-Pomares2017; Uribe-Querol and Rosales, Reference Uribe-Querol and Rosales2020) and in the presentation of antigens, constituting an important link between innate and adaptive immunity. These cells are part of the mononuclear phagocytic system and are implicated in tissue homoeostasis and various infectious and inflammatory processes (Rahman et al., Reference Rahman, Vengrenyuk, Ramsey, Vila, Girgis, Liu, Gusarova, Gromada, Weinstock, Moore, Loke and Fisher2017; Shapouri-Moghaddam et al., Reference Shapouri-Moghaddam, Mohammadian, Vazini, Taghadosi, Esmaeili, Mardani, Seifi, Mohammadi, Afshari and Sahebkar2018).

Macrophages are activated during phagocytosis or by contact with molecular patterns associated with pathogenic microorganisms. This activation results in inflammatory responses and increased production of cytokines and/or physicochemical factors and, consequently, can differentiate into various phenotypes depending on the state and changes in the microenvironment (Schmall et al., Reference Schmall, Al-Tamari, Herold, Kampschulte, Weigert, Wietelmann, Vipotnik, Grimminger, Seeger, Pullamsetti and Savai2015; Murray, Reference Murray2017). There are 2 main subtypes of macrophages classified according to the expression of their cell surface markers, production of specific factors and biological activities: classically activated or inflammatory M1 macrophages and alternatively activated or anti-inflammatory M2 macrophages (Parisi et al., Reference Parisi, Gini, Baci, Tremolati, Fanuli, Bassani, Farronato, Bruno and Mortara2018) (Fig. 3). Macrophage subtypes play a role in the initiation and/or progression of many diseases. The M1/M2 paradigm emerged as homologous with the one previously described for Th response profiles, which also presents 2 subtypes: Th cell type 1 (Th1) and type 2 (Th2) (Mills, Reference Mills2015).

Fig. 3. Different macrophage phenotypes, specific stimuli and markers. Source: Created with BioRender.com.

M1 macrophage subtypes polarize in the presence of Th1 cytokines such as IFN-γ and TNF-α or when exposed to inflammatory molecules such as lipopolysaccharides (LPS) (Yunna et al., Reference Yunna, Mengru, Lei and Weidong2020), through the following mechanisms: (1) JAK/STAT (Janus kinase/signal transducer and transcriptional activator) signalling pathway. IFN-γ activates JAK-inducing phosphorylation of STAT1, which in turn leads to macrophage polarization to M1 (Wang et al., Reference Wang, Shang, Qi, Ba, Lv, Zhou, Wang, Shaxika, Wang and Ma2020); (2) Toll-like receptor (TLR) 4/nuclear factor κB (NF-κB) signalling pathway. LPS binds to TLR4 to activate NF-κB and activator protein 1 (AP-1), promoting the expression of inflammatory factors (Chen et al., Reference Chen, Tang, Fu, Wang, Han and Meng2017b; Ciesielska et al., Reference Ciesielska, Matyjek and Kwiatkowska2021) and (3) cytokine signalling through specific receptors that activate AP-1 (Liu et al., Reference Liu, Zou, Chai and Yao2014).

M1 macrophages are recruited soon after lesion formation and are mainly involved in the initial response to infectious processes (Vannella and Wynn, Reference Vannella and Wynn2017). These increase local inflammation, producing large amounts of pro-inflammatory cytokines, including IL-1β, IL-6, IL-12, IL-18, IL-23, TNF-α and IFN type 1 (Shapouri-Moghaddam et al., Reference Shapouri-Moghaddam, Mohammadian, Vazini, Taghadosi, Esmaeili, Mardani, Seifi, Mohammadi, Afshari and Sahebkar2018), as shown in Fig. 3. The M1 macrophage phenotype expresses high levels of inducible nitric oxide synthase (iNOS), major histocompatibility complex class II (MHC II), CD16/32, CD80 and CD86, as well as chemokines that attract Th1 cells, including CXCL9 and CXCL12 (Orecchioni et al., Reference Orecchioni, Ghosheh, Pramod and Ley2019). Functionally, M1 macrophages are characterized by antimicrobial and antitumour activities and participate in the elimination of infectious agents through the activation of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase and, consequently, the generation of reactive oxygen species (ROS) (Murray et al., Reference Murray, Allen, Biswas, Fisher, Gilroy, Goerdt, Gordon, Hamilton, Ivashkiv, Lawrence, Locati, Mantovani, Martinez, Mege, Mosser, Natoli, Saeij, Schultze, Shirey, Sica, Suttles, Udalova, van, Vogel and Wynn2014).

On the other hand, M2 macrophages are induced by Th2 cytokines IL-4 and IL-13 (Fig. 3), mainly via STAT6 activation (Sica and Mantovani, Reference Sica and Mantovani2012; Enderlin et al., Reference Enderlin Vaz Da Silva, Lehr and Velin2014). This pathway is extremely important, as IL-4 inhibits M1 and induces M2 polarization (He et al., Reference He, Gao, Zhang, Zhou, Cao and Yao2020). Gao et al. (Reference Gao, Zhou, Liu, Wang, Gao, Wu, Zhao, Liu, Wang, Liu, Guo, Shen, Wu and Yuan2015) demonstrated that the expression of STAT6 was positively regulated by curcumin and by the secretion of IL-4 and IL-13, capable of inducing M0 and M1 macrophages to polarize into M2. IL-4 type I and type II receptors also activate STAT6 (Gong et al., Reference Gong, Zhuo and Ma2017), which in turn induces the transcription of typical M2 polarization genes, such as mannose receptor 1, type α resistin (Retnla) and chitinase 3-like 3 (Chi3l3, Ym1) (Martinez and Gordon, Reference Martinez and Gordon2014). M2 polarization can also be induced by IL-10 through STAT3 activation (Yin et al., Reference Yin, Ma, Lin, Lu, Zhang, Chen and Jian2018). However, the STAT6 pathway is considered to activate M2 macrophages (Murray, Reference Murray2017).

The M2 macrophage phenotype has a profile of anti-inflammatory cytokines, characterized by low production of IL-1, IL-6 and TNF-α, and high production of IL-10 and transforming growth factor-beta (TGF-β) (Fig. 3), as well as chemokines CCL1, CCL17, CCL18, CCL22 and CCL24 (Yunna et al., Reference Yunna, Mengru, Lei and Weidong2020). Additionally, this phenotype can be characterized by the expression of arginase 1 (Arg-1), CD163, CD209 and CD206. CD206 interacts with glycoproteins and glycolipids found on the surfaces of pathogens (Suzuki et al., Reference Suzuki, Shirai, Asada, Yasui, Karayama, Hozumi, Furuhashi, Enomoto, Fujisawa, Nakamura, Inui, Shirai, Hayakawa and Suda2018; Xu et al., Reference Xu, Gu, Wang, Jin, Wen, Ma, Tang, Mao, Qian and Lin2019). Thus, CD206 plays a role in immunological recognition of pathogens after antigen internalization and presentation (Hussell and Bell, Reference Hussell and Bell2014). Functionally, M2 macrophages can inhibit inflammation, promote tissue repair and wound healing, prevent parasitic infection and have proangiogenic and profibrotic properties (Jetten et al., Reference Jetten, Verbruggen, Gijbels, Post, De Winther and Donners2014; Braga et al., Reference Braga, Agudelo and Camara2015). Furthermore, because M2 macrophages produce complex cytokines and are characterized by the functional expression of alternative activation markers, they can be divided into 4 subtypes: M2a, M2b, M2c and M2d (Yao et al., Reference Yao, Xu and Jin2019). These subtypes differ from each other based on their cell surface markers, secreted cytokines and biological functions, as is demonstrated in Fig. 3.

M2a macrophages are induced by the cytokine IL-4 or IL-13 and express high levels of CD86, CD200R and MHC II and low levels of CD14 and TLR4 (Yao et al., Reference Yao, Xu and Jin2019). In addition to being major producers of CCL24, CCL17 and CCL22, they use CCR3 and CCR4 receptors, resulting in the recruitment of eosinophils, basophils and Th2 cells, promoting the upregulation of a type 2 immune response (Fraternale et al., Reference Fraternale, Brundu and Magnani2015). M2b-type macrophages are induced by immune complexes, LPS or IL-1 receptor antagonist and are characterized by increased expression of CD206 and CD86 (Viola et al., Reference Viola, Munari, Sánchez-Rodríguez, Scolaro and Castegna2019). Upon activation, this subtype secretes pro- and anti-inflammatory cytokines TNF-α, IL-1β, IL-6 and IL-10 and functions in regulating the immune response and inflammation (Wang et al., Reference Wang, Ye, Zeng and Qiao2019). M2c macrophages are induced by IL-10, TGF-β or glucocorticoids, and express CD206 and CD163, in addition to secreting IL-10, TGF-β, CCL16 and CCL18, which play crucial roles in the phagocytosis of apoptotic cells (Ross et al., Reference Ross, Devitt and Johnson2021). Finally, induced by TLR antagonists, M2d macrophages express high levels of CD206, IL-10 and iNOS, secrete CCL5, CXCL10 and CXCL6 and express low levels of IL-12 and TNF-α (Viola et al., Reference Viola, Munari, Sánchez-Rodríguez, Scolaro and Castegna2019). This subtype also secretes the vascular endothelial growth factor and promotes angiogenesis and tumour progression (Ferrante et al., Reference Ferrante, Pinhal-Enfield, Elson, Cronstein, Hasko, Outram and Leibovich2013). Notably, all subtypes of M2 macrophages express IL-10.

M1 and M2 macrophages can also be differentiated by the way they metabolize arginine, as shown in Fig. 3. M1 macrophages metabolize arginine by the enzyme iNOS to produce nitric oxide (NO) and citrulline; on the contrary, M2 macrophages metabolize arginine by Arg-1 to produce l-ornithine and urea, a precursor molecule of polyamines involved in tissue repair and cell proliferation (Rath et al., Reference Rath, Müller, Kropf, Closs and Munder2014; Yang and Ming, Reference Yang and Ming2014).

The 2 macrophage populations must be balanced to maintain homoeostasis and to protect the organism. Once an imbalance occurs, the exacerbated activity of M1 or M2 macrophages can lead to the development of inflammatory diseases or host immunosuppression (Sica et al., Reference Sica, Erreni, Allavena and Porta2015). However, the remarkable plasticity of macrophages confers significant benefits to the host, especially in the course of chronic helminth infections (Lechner et al., Reference Lechner, Bohnacker and Esser-von Bieren2021) since it limits excessive tissue damage when it is unable to overcome the initial injury. This feature has been well-documented in schistosomiasis.

Participation of M1 and M2 macrophages in the response to Schistosoma infection

Initially, blood monocytes differentiate into macrophages at inflammatory sites (Rückerl and Cook, Reference Rückerl and Cook2019) and exhibit high plasticity as a result of exposure to various stimuli, signalling molecules, nutrients and metabolites in the context of schistosomiasis (Cortes-Selva and Fairfax, Reference Cortes-Selva and Fairfax2021). These phagocytes can exert pro-inflammatory or anti-inflammatory functions (Zhu et al., Reference Zhu, Xu, Chen, Zhou, Zhang, Chi, Li, Song, Liu and Su2014) in different clinical forms of schistosomiasis (acute and chronic phases) (Fig. 2). In the acute phase, macrophages secrete pro-inflammatory cytokines and consequently increase inflammation, recruit more immune cells and promote the formation of the initial granuloma. In the chronic phase, macrophages have an immunoregulatory activity to decrease the damage caused by granulomas (Wolde et al., Reference Wolde, Laan, Medhin, Gadissa, Berhe and Tsegaye2020).

During the life cycle of S. mansoni and S. japonicum, several antigens are excreted by their different evolutive forms (Curwen et al., Reference Curwen, Ashton, Sundaralingam and Wilson2006; Jang-Lee et al., Reference Jang-Lee, Curwen, Ashton, Tissot, Mathieson, Panico, Dell, Wilson and Haslam2007; Acharya et al., Reference Acharya, Da'dara and Skelly2021). For example, Sm16 – a low molecular weight protein that is secreted by S. mansoni cercariae, helps the parasite to enter the host's skin (Brännström et al., Reference Brännström, Sellin, Holmfeldt, Brattsand and Gullberg2009; Sanin and Mountford, Reference Sanin and Mountford2015). Sm29, present in the tegument of schistosomula and adult S. mansoni worms, can induce the maturation and activation of human monocyte-derived DCs (Cardoso et al., Reference Cardoso, Macedo, Gava, Kitten, Mati, de Melo, Caliari, Almeida, Venancio, Verjovski-Almeida and Oliveira2008; Lopes et al., Reference Lopes, Oliveira, Page, Carvalho, Carvalho and Cardoso2019). Sj-C is an example of a protein secreted from the tegument of S. japonicum, which may suppress the presentation of exogenous antigens by DCs (He et al., Reference He, Cai, Ni, Li, Zong and He2011; Chen et al., Reference Chen, He, Hou and He2017a). IPSE/α-1 and ω-1 are examples of proteins secreted by S. mansoni eggs, which help direct a Th2 response (Everts et al., Reference Everts, Perona-Wright, Smits, Hokke, van der Ham, Fitzsimmons, Doenhoff, van der Bosch, Mohrs, Haas, Mohrs, Yazdanbakhsh and Schramm2009; Knuhr et al., Reference Knuhr, Langhans, Nyenhuis, Viertmann, Overgaard Kildemoes, Doenhoff, Haas and Schramm2018). These molecules can induce the activation and modulation of innate and adaptive immune responses and facilitate the evasion of the parasite from the host-defense mechanisms (Jenkins et al., Reference Jenkins, Hewitson, Ferret-Bernard and Mountford2005a, Reference Jenkins, Hewitson, Jenkins and Mountford2005b; Hai et al., Reference Hai, Edwards, Van Zandt, Hoffmann and Christianson2014; Hambrook and Hanington, Reference Hambrook and Hanington2021). Schistosoma antigens can be proteins (such enzymes), polysaccharides and the most commonly used are crude extracts prepared by breaking up worms, larvae or eggs (Doenhoff et al., Reference Doenhoff, Butterworth, Hayes, Sturrock, Ouma, Koech, Prentice and Bain1993; Doenhoff, Reference Doenhoff1998). Thus, it is clear that antigen changes in the microenvironment during schistosomiasis are important for the polarization of macrophages to the M1 or M2 profile (Xu et al., Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014; Sanin and Mountford, Reference Sanin and Mountford2015; Assunção et al., Reference Assunção, Magalhães, Carneiro, Molinaro, Almeida, Atella, Castro-Faria-Neto and Bozza2017).

According to Tables 1 and 2, we highlight some in vitro and in vivo studies that demonstrate the relationship between the stimulation of S. japonicum and S. mansoni antigens and macrophage polarization. In addition, we also highlight other molecules involved in macrophage polarization in schistosomiasis, providing molecular evidence of great relevance in the process of differentiation of these cells, which will be discussed in this article.

Table 1. Molecules and/or antigens involved in macrophage polarization in Schistosoma mansoni infection

Table 2. Molecules and/or antigens involved in macrophage polarization in Schistosoma japonicum infection

Cercariae and schistosomula antigens can induce an M1 profile

During the penetrating of the human skin, cercariae of S. mansoni and S. japonicum release E/S products, which have remodelling and immunoregulatory functions (Liu et al., Reference Liu, Ju, Du, Shen, Wang, Li, Zhang, Feng and Hu2015; Sanin and Mountford, Reference Sanin and Mountford2015), that facilitate their penetration and subsequent establishment in the host's body, in the form of schistosomula (Janssen et al., Reference Janssen, Silva Santos, Muller, Vieira, De Campos and De Paulo Martins2016). This phase represents the first contact with innate immune responses in the skin, especially Langerhans cells, which are considered tissue-resident macrophages (West and Bennett, Reference West and Bennett2018). These cells phagocytize E/S and secrete pro-inflammatory (IL-6 and IL-12p40) and anti-inflammatory (IL-10) cytokines in a TLR-dependent manner (Jenkins et al., Reference Jenkins, Hewitson, Ferret-Bernard and Mountford2005a, Reference Jenkins, Hewitson, Jenkins and Mountford2005b).

One of the ways in which macrophages are activated is through the action of TLRs. These receptors are a family of pattern recognition receptors that are important for innate immune response (El-Zayat et al., Reference El-Zayat, Sibaii and Mannaa2019). These receptors recognize invading pathogens, trigger innate immune responses and subsequently initiate adaptive immunity against infections, including Gram-positive and Gram-negative bacteria, fungi, viruses and parasites (Lu et al., Reference Lu, Li, Liu, Zhang and Zhang2018). These receptors mediate macrophage recognition by microbial ligands, inducing the expression of microbicidal molecules and cytokines via the adapter protein MyD88 (Jin et al., Reference Jin, Yuan, Chen, Wu, Gong, Yuan, Zeng, Pei and Chen2019). Xu et al. (Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014) showed that normal cercariae antigen (NCA) and attenuated cercariae antigen (ACA) from S. japonicum induced polarization to the M1 profile, with increased levels of IL-12, CD136/32 and iNOS (Table 2). However, these values decreased when the TLR4 pathway blockers were used. Thus, the authors suggested that the polarization of the M1 profile is dependent on the TLR4 pathway and this may play a protective role in S. japonicum infection (Tang et al., Reference Tang, Shen, Hong, Zhang, Zhai, Fu, Li, Lu and Lin2021).

In fact, the TLR4 pathway is extremely important for the polarization of macrophages to the M1 phenotype, as demonstrated in some studies (Freitas et al., Reference Freitas, Oliveira, Da Silva, Fernandes, Gonçales, Almeida and Roque-Barreira2016; Shi et al., Reference Shi, Luo, Wang, Horst, Bläsius, Relja, Xu, Hildebrand and Greven2020). Sanin and Mountford (Reference Sanin and Mountford2015) demonstrated that Sm16 (a molecule produced by S. mansoni cercariae) is able to block TLR4 and TLR3 pathways in human monocyte, which negatively affect the classic activation of macrophages (M1) in response to IFN-γ (Table 1). This is considered an important mechanism of immune evasion promoted by S. mansoni because it limits the production of NO, which is toxic to the parasite (Shiels et al., Reference Shiels, Cwiklinskiid, Alvarado, Thivierge, Cotton, Santana, Toid, Donnelly, Taggart, Weldonid and Dalton2020).

After complete transformation from a cercariae into a schistosomula, the larva migrates into the bloodstream, travelling through the lungs until reaching maturation in the mesenteric veins. This stage of the cycle is also characterized as a key target for the elimination of infection through innate host immune responses (Houlder et al., Reference Houlder, Costain, Cook and MacDonald2021). Some histological studies conducted in the lungs of mice infected with S. mansoni and S. japonicum showed inflammatory foci consisting of neutrophils, eosinophils and macrophages (Crabtree and Wilson, Reference Crabtree and Wilson1986; Burke et al., Reference Burke, McGarvey, McSorley, Bielefeldt-Ohmann, McManus and Gobert2011).

Macrophages function as cytotoxic cells, mainly in schistosomula (James and Glaven, Reference James and Glaven1989). Oswald et al. (Reference Oswald, Wynn, Sher and James1994) demonstrated that macrophages could produce NO, leading to schistosomula death in animal models independent of the production of pro-inflammatory cytokines. In contrast, Cardoso et al. (Reference Cardoso, Macedo, Gava, Kitten, Mati, de Melo, Caliari, Almeida, Venancio, Verjovski-Almeida and Oliveira2008) determined that the antigen Sm29, present in the integument of S. mansoni schistosomula, induced a Th1-type immune response, with an increase in pro-inflammatory cytokines (IFN-γ, TNF and IL-12) in mice, leading to a reduction in worm burden and liver pathology. James et al. (Reference James, Cheever, Caspar and Wynn1998) demonstrated that IFN-γ was a cytokine of great importance in the activation of macrophages in the lungs for the immunological killing of S. mansoni larvae and played a critical role in protective immunity.

In the early stages of schistosomiasis, lung macrophages may have an M1 phenotypic trait. Menson and Wilson (Reference Menson and Wilson1990) characterized the expression of surface markers in alveolar macrophages associated with the immune response to S. mansoni. The authors demonstrated an increase in IFN-γ expression in the lungs of C57BL/6 mice and suggested that activated macrophages might be responsible for initiating and maintaining focal inflammation that blocks parasite migration (Table 1).

Worm antigens can induce an M1 or M2 profile

During the acute phase of Schistosoma infection, before parasite oviposition (approximately 5–7 weeks post-infection), immune responses are largely of the CD4+ Th1 type, associated with increased numbers of M1 macrophages that produce IL-12, IL-6, TNF-α and NO (Pearce et al., Reference Pearce, Caspar, Grzych, Lewis and Sher1991; Coulson et al., Reference Coulson, Smythies, Betts, Mabbott, Sternberg, Wei, Liew and Wilson1998; Gordon, Reference Gordon2003). These early pro-inflammatory responses are mainly related to the antigens from immature worms (schistosomula) during their migration (Wilson, Reference Wilson1998; Egesa et al., Reference Egesa, Lubyayi, Tukahebwa, Bagaya, Chalmers, Wilson, Hokke, Hoffmann, Dunne, Yazdanbakhsh, Labuda and Cose2018). Activation of these responses may be through binding to TLR and C-type lectin receptors on macrophages; however, further studies are needed to clarify this mechanism of macrophage activation via schistosomula antigens.

In contrast to the schistosomula antigens, the adult worm antigen preparations [soluble worm antigen (SWAP or SWA)] were better explored in experimental studies. Although the antigenic composition is not the same as the live worm, the use of SWAP or SWA constitutes a valuable experimental tool to evaluate many aspects of immune responses promoted by different host cells (Xu et al., Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014; Zhu et al., Reference Zhu, Xu, Chen, Zhou, Zhang, Chi, Li, Song, Liu and Su2014). This antigen is the easiest to obtain and is essentially an extract based on Tris-HCl or phosphate-buffered saline from mixed male and female worms and prepared in various ways, either by homogenization, sonication or freeze/thaw (or a combination of these) (Grenfell et al., Reference Grenfell, Martins, Silva-Moraes, Barata, Ribeiro, Oliveira and Coelho2012; Neves et al., Reference Neves, Sanson, Wilson and Castro-Borges2015). Some studies have demonstrated that SWAP could induce an M1-like profile (Xu et al., Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014; Zhu et al., Reference Zhu, Xu, Chen, Zhou, Zhang, Chi, Li, Song, Liu and Su2014). Thus, Zhu et al. (Reference Zhu, Xu, Chen, Zhou, Zhang, Chi, Li, Song, Liu and Su2014), when performing a co-culture of peritoneal macrophages obtained from mice with S. japonicum SWA (Table 2), observed that there was an increase in the expression of specific markers related to M1 (TNF-α, IL-12, CXCL9, CXCL10, CXCL11 and iNOS).

Aiming to understand which mechanisms lead SWAP to induce polarization of the M1 profile, Shen et al. (Reference Shen, Wang, Huang, Zhu, Sun and Duan2021) demonstrated that this antigen promoted the expression of a protein called lipocalin 2 (LCN2) and, consequently, induced the M1 profile of macrophages (Table 2) through the upregulation of the NF-κB signalling pathway. It has already been reported that this protein is increased in macrophages and can potentiate the M1 phenotype of microglia in the central nervous system (Jang et al., Reference Jang, Lee, Kim, Kim, Seo, Lee, Mori, Nakao and Suk2013). The NF-κB signalling pathway can activate macrophages to produce M1 polarization upon LPS induction (Liu et al., Reference Liu, Zhang, Tan, Xu, Zhou, Luo, Li, Huang and Zeng2017). In addition, some studies have shown that this pathway could regulate the expression of LCN2, thereby stimulating the inflammatory response in infectious processes (Zhao and Stephens, Reference Zhao, Elks and Stephens2014; Ghosh et al., Reference Ghosh, Shang, Yazdankhah, Bhutto, Hose, Montezuma, Luo, Chattopadhyay, Qian, Lutty, Ferrington, Zigler and Sinha2017).

In addition to the antigens of adult worms that induce an M1 profile, studies have shown that adult worms of S. mansoni and S. japonicum also release extracellular vesicles (EVs), known as exosomes, which modulate the host immune response (Nowacki et al., Reference Nowacki, Swain, Klychnikov, Niazi, Ivens, Quintana, Hensbergen, Hokke, Buck and Hoffmann2015; Wang et al., Reference Wang, Altinoglu, Takeda and Xu2015; Sotillo et al., Reference Sotillo, Pearson, Potriquet, Becker, Pickering, Mulvenna and Loukas2016; Zhu et al., Reference Zhu, Wang, Lin, Jiang, Cui, Wang, Zhang and Pan2016). Exosomes are membrane-bound vesicles secreted by various types of mammalian cells in normal and diseased states (Avni and Avni, Reference Avni and Avni2021). Exosomes play an important role in cell–cell communication and have been implicated in the regulation of cell development, immune regulation, angiogenesis and cell migration (Raposo and Stoorvogel, Reference Raposo and Stoorvogel2013; Zhu et al., Reference Zhu, Wang, Lin, Jiang, Cui, Wang, Zhang and Pan2016). Wang et al. (Reference Wang, Altinoglu, Takeda and Xu2015) observed that RAW264.7 macrophages, when cultured with exosome-like vesicles isolated from S. japonicum, exhibited an M1 profile (Table 2), due to the increase in the surface markers CD16/32, iNOS and TNF-α. Liu et al. (Reference Liu, Zhu, Wang, Qiu, Chen, Davis and Cheng2019) investigated miRNAs from S. japonicum EVs and found that they increased macrophage proliferation in vitro (RAW264.7) and in vivo (mice and rabbits) as well as TNF-α expression. miRNAs are involved in the regulation of the development, differentiation and activation of immune cells, including macrophages (Montagner et al., Reference Montagner, Dehó and Monticelli2014; Mehta and Baltimore, Reference Mehta and Baltimore2016). Thus, the polarization of M1 induced by schistosome EVs may represent an important mechanism for parasite survival in vertebrate hosts, via modulation of the immune response. However, there are still controversies about the possible role of the schistosome tegument as a source of EVs, because, to date, no study has been performed to prove the exact origin of these vesicles (Wilson and Jones, Reference Wilson and Jones2021).

On the other hand, adult worm products can also bias the M2 profile (Smith et al., Reference Smith, Forman, Mair and Else2018). Indeed, Xu et al. (Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014) showed that adult S. japonicum worms could induce an M2 macrophage profile. The authors, when stimulating RAW264.7 macrophages with SWAP from S. japonicum, observed an increase in the expression of surface markers (CD16/32 and CD206) and in the production of cytokines (IL-12 and IL-10), suggesting that this antigen could induce both M1 and M2 macrophage profiles. The potential explanation for this could be related to how the antigens of adult worms were obtained since some adult female worms possess eggs in the process of maturation into their uterus/ootype, and consequently, this antigen could have been contaminated with SEAs. However, further studies are needed to understand macrophage polarization by SWAP and its relationship with SEA contamination.

Besides the classical macrophage polarization (M1 and M2), products excreted by schistosomes, such as haemozoin, are also able to induce immunomodulation. Adult worms of S. mansoni acquire nutrients by haematophagy of the host's blood, and this process can form toxic haem for the parasite (Zussman et al., Reference Zussman, Bauman and Petruska1970). However, the schistosomes are able to neutralize the free haem in their intestine through crystallization in haemozoin (Oliveira et al., Reference Oliveira, D'Avila, Torres, Oliveira, Tempone, Rumjanek, Braga, Silva, Dansa-Petretski, Oliveira, De Souza and Ferreira2000). This haemozoin is regurgitated by the worms into the host bloodstream and can be accumulated in the liver (Kloetzel and Lewert, Reference Kloetzel and Lewert1966), which may activate the immune response of the host. From this perspective, a previous study (Truscott et al., Reference Truscott, Evans, Gunn and Hoffmann2013) highlighted that haemozoin formed from S. mansoni is able to maintain the M2 macrophage profile previously activated by IL-4 stimulation, but also exerts specific modulatory effects on these cells (Table 1). These authors showed that haemozoin mediated the suppression of Retnla (resistin-like molecule-α or Fizz1) expression and Retnla protein secretion in the M2 macrophages. The role of Retnla during experimental schistosomiasis is associated with the limitation of Th2 inflammatory response (Pesce et al., Reference Pesce, Ramalingam, Wilson, Mentink-Kane, Thompson, Cheever, Urban, Wynn and Mansfield2009). However, further studies are necessary to better explain the possible impact of haemozoin in the immunopathology of schistosomiasis.

SEAs can induce an M2 profile

After maturation of the adult worms and subsequent oviposition, the activation of a type 2 profile begins in response to the soluble antigens secreted by the eggs of S. mansoni and S. japonicum (Tables 1 and 2) (Cheever et al., Reference Cheever, Hoffmann and Wynn2000; Pearce and MacDonald, Reference Pearce and MacDonald2002; Pearce et al., Reference Pearce, Kane, Sun, Taylor, McKee and Cervi2004; Burke et al., Reference Burke, Jones, Gobert, Li, Ellis and McManus2009; Costain et al., Reference Costain, MacDonald and Smits2018). The type 2 profile of schistosomiasis is characterized by the expansion of Th2 cells, eosinophils and basophils, and increased production of IL-4, IL-5 and IL-13 (Hams et al., Reference Hams, Aviello and Fallon2013; Schwartz et al., Reference Schwartz, Oeser, Prazeres da Costa, Layland and Voehringer2014), as previously described. IL-4 and IL-13 protect hosts against various helminth parasites by signalling through the IL-4Rα chain (Barron and Wynn, Reference Barron and Wynn2011; Jenkins et al., Reference Jenkins, Ruckerl, Cook, Jones, Finkelman, Van Rooijen, MacDonald and Allen2011). The production of these cytokines reduces the inflammation levels produced by the type 1 profile of the initial stage of acute phase, preventing acute pathology, such as intestinal haemorrhage and liver damage; however, the Th2 immune response is responsible for the formation of hepatic and intestinal granulomas (Brunet et al., Reference Brunet, Finkelman, Cheever, Kopf and Pearce1997; Hams et al., Reference Hams, Aviello and Fallon2013; Zheng et al., Reference Zheng, Zhang, Chen, Nie, Miller, Gong and Liu2020).

Granulomas are essential for sequestering toxic antigens produced by eggs and preventing further tissue damage. However, if unregulated by the immune response of the host, granulomas grow excessively and progress to fibrotic stages, which are responsible for severe forms of the disease, such as cirrhosis, portal hypertension, liver failure and even host death (Lenzi et al., Reference Lenzi, Kimmel, Schechtman, Pelajo-Machado, Romanha, Pacheco, Mariano and Lenzi1998; Cheever et al., Reference Cheever, Hoffmann and Wynn2000; Takaki et al., Reference Takaki, Rinaldi, Berriman, Pagán and Ramakrishnan2021). Macrophages are one of the main cellular components of hepatic granulomas (Beljaars et al., Reference Beljaars, Schippers, Reker-Smit, Martinez, Helming, Poelstra and Melgert2014; Schwartz and Fallon, Reference Schwartz and Fallon2018). Recent studies have demonstrated that M2 macrophages play a direct and critical role in fibrosis, granuloma maintenance, tissue repair and host survival (Cortes-Selva et al., Reference Cortes-Selva, Elvington, Ready, Rajwa, Pearce, Randolph and Fairfax2018; Song et al., Reference Song, Yin, Mu, Li, Gao, Zhang, Dong, Mei and Hua2020). Ye et al. (Reference Ye, Huang, Zhang, Mei, Zheng, Li, Chen and Lu2020) showed that M2 macrophage markers (CD200R, Arg-1 and Ym1) were increased in the liver, spleen, large intestine and peritoneal macrophages of S. japonicum-infected mice. Jenkins et al. (Reference Jenkins, Ruckerl, Cook, Jones, Finkelman, Van Rooijen, MacDonald and Allen2011) observed that IL-4/IL-13 signalling via IL-4Rα induces an alternative phenotype in resident macrophages. In this sense, a study performed with macrophages derived from the bone marrow of mice infected with S. mansoni demonstrated that the tumour suppressor gene p16 INK4a was an excellent modulator of the activation and polarization of macrophages induced by IL-4 through the JAK2–STAT1 pathway (Cudejko et al., Reference Cudejko, Wouters, Fuentes, Hannou, Paquet, Bantubungi, Bouchaert, Vanhoutte, Fleury, Remy, Tailleux, Chinetti-Gbaguidi, Dombrowicz, Staels and Paumelle2011).

Egg antigens induce granulomas, consisting mainly of M2 macrophages (Yu et al., Reference Yu, Wang, Wang, Gu, Lei, Tang, Wei, Xu, Wang, Chen, Pu, Qi, Yu, Chen, Zhu, Li, Zhang, Zhou and Su2021). Zhu et al. (Reference Zhu, Xu, Chen, Zhou, Zhang, Chi, Li, Song, Liu and Su2014) showed that peritoneal macrophages obtained from healthy mice, when stimulated with S. japonicum SEAs, expressed high levels of chemokines (CCL2, CCL17 and CCL22), IL-10 and Arg-1. Similarly, Xu et al. (Reference Xu, Zhang, Chen, Zhang, Ji, Wu and Wu2014), after stimulating RAW264.7 macrophages with S. japonicum SEAs, also observed higher levels of IL-10. In chronic schistosomiasis infection, the main function of IL-10 is to control liver damage and regulate antifibrotic processes (Dewals et al., Reference Dewals, Marillier, Hoving, Leeto, Schwegmann and Brombacher2010; Kamdem et al., Reference Kamdem, Moyou-Somo, Brombacher and Nono2018). Previous studies have shown that low levels of IL-10 expression are related to liver fibrosis in S. mansoni-infected patients (Mutengo et al., Reference Mutengo, Mduluza, Kelly, Mwansa, Kwenda, Musonda and Chipeta2018). On the other hand, little is known about the mechanisms by which an SEA preferentially induces M2 macrophage differentiation. Previous studies have demonstrated that an SEA from S. mansoni could induce the expression of the notch Jagged1 ligand in mice and human macrophages, suggesting that Jagged1 might have a specific role in the M2 polarization process of macrophages (Goh et al., Reference Goh, Irvine, Lovelace, Donnelly, Jones, Brion, Hume, Kotze, Dalton, Ingham and Sweet2009) (Table 1). Macrophages found in liver tissues exhibit functional M2 polarization, which is dependent on the activation of notch1/Jagged1 signalling (Zheng et al., Reference Zheng, Zhang, Chen, Zheng, Zheng and Weng2016).

During S. mansoni infection, basophils detect egg IPSE/α-1 glycoprotein and stimulate the production of IL-4 and IL-13, which trigger the alternative activation of human monocytes (Table 1), as observed by the increased expression of CD206 and CD209 (Knuhr et al., Reference Knuhr, Langhans, Nyenhuis, Viertmann, Overgaard Kildemoes, Doenhoff, Haas and Schramm2018). IL-13 is a key cytokine that induces M2 macrophage polarization via the IL-13α1 signalling pathway (Chiaramonte et al., Reference Chiaramonte, Schopf, Neben, Cheever, Donaldson and Wynn1999; Liu et al., Reference Liu, Munker, Müllenbach and Weng2012). Li et al. (Reference Li, Chen, Dang, Wang, Shang, Ma, Wang, Zhang, Luo, Li and Zhao2017) performed a study with corilagin, an active component of many medicinal plants, and found that this component could suppress Schistosoma egg-induced liver fibrosis by inhibiting M2 macrophage polarization (Table 1) in the IL-13Rα1 signalling pathway. Corilagin has great potential to reduce liver fibrosis caused by egg antigens in S. japonicum infection by decreasing the expression of molecules associated with the IL-13/STAT6 signalling pathway in liver M2 macrophages (Du et al., Reference Du, Ma, Zhu, Li, Wang, Li, Chen, Shang, Zhang and Zhao2016).

Signaling via TLR2 may be another way egg antigens polarize M2 macrophages during schistosomiasis. Gong et al. (Reference Gong, Huang, Sun, Yu, Zhang, Xu, Shen and Cao2018) showed that antigens derived from S. japonicum eggs could activate macrophages, which exhibit M2b polarization dependent on NF-κB signalling, mediated by the MyD88/mitogen-activated protein kinase (MAPK) pathway in a TLR2-dependent manner (Table 1). In contrast, Tundup et al. (Reference Tundup, Srivastava, Nagy and Harn2014) showed that the CD14 TLR co-receptor was upregulated in hepatic macrophages after S. mansoni infection and acted as a crucial negative regulator of M2 polarization, possibly as part of a parasitic defense mechanism against granuloma formation (Table 1). Gao et al. (Reference Gao, Chen, Hou, Chen, Ji, Wu and Wu2013) observed that an SEA of S. japonicum, known as SjEA, upregulated programmed death ligand 2 (PD-L2) expression in mouse bone marrow-derived macrophages (BMDCs) via TLR2, which binds PD-1 primarily on CD4+ T cells. This mechanism can help inhibit the T cell response during S. japonicum infection.

Lysophosphatidylcholine (LPC) from S. mansoni eggs can also induce macrophage differentiation into the M2 phenotype (Assunção et al., Reference Assunção, Magalhães, Carneiro, Molinaro, Almeida, Atella, Castro-Faria-Neto and Bozza2017), as shown in Table 1. The authors observed that LPC from S. mansoni activates peroxisome proliferator-activated receptor gamma (PPAR-γ), a transcription factor necessary for M2 polarization, leading to higher expression of Arg-1 and CD206, while increasing the production of IL-10, TGF-β and PGE2 in peritoneal macrophages in vitro. Schistosoma mansoni eggs induced a 7-fold increase in PPAR-γ expression in human liver cell cultures (Anthony et al., Reference Anthony, Mathieson, de Castro-Borges and Allen2010). PPAR-γ, in addition to being of great importance in M2 polarization, can regulate lipid uptake and metabolism (Ahmadian et al., Reference Ahmadian, Suh, Hah, Liddle, Atkins, Downes and Evans2013; Abdalla et al., Reference Abdalla, Napimoga, Lopes, de Macedo Maganin, Cunha, Van Dyke and Clemente Napimoga2020).

Fang et al. (Reference Fang, Wu, Chen, Wu, Yang, Guo, Chen and Wang2015) showed that BMDCs from C57BL/6 mice, when stimulated with a specific S. japonicum egg protein known as SjE16.7, promoted the production of pro- (IL-12, IL-6 and TNF-α) and anti-inflammatory (IL-10) cytokines through the phosphorylation of MAPKs and increased the expression of MHC II on the surface of macrophages. Previous studies have shown that S. mansoni and S. japonicum egg antigens could stimulate the MAPK pathway in macrophages (Wang et al., Reference Wang, Yang, Li, Yu, Brindley, McManus, Wei, Han, Feng, Li and Hu2006; de Andrade et al., Reference de Andrade, de Mourão, Geraldo, Coelho, Silva, Neves, Volpini, Machado-Silva, Araujo, Nacif-Pimenta, Caffrey and Oliveira2014). MAPKs are essential transmitters of extracellular signals that can mediate key cellular processes, including cell differentiation, division and death (Yang et al., Reference Yang, Sharrocks and Whitmarsh2003). Thus, SjE16.7 is a potent macrophage activator. However, in another study, Shen et al. (Reference Shen, Wang, Peng, Liu, Zhang, Zhou, Sun and Wu2019), when using the Sj16 antigen, noticed that it decreased hepatic granulomas in mice infected with S. japonicum and associated this improvement with the suppression of cytokine production, such as IFN-γ, TNF-α, IL-4 and IL- 6. The authors reported that the mechanisms of Sj16 attenuation of hepatic granulomatous inflammation and fibrosis in these infected mice might be related to the induction of macrophages for M2 polarization (Table 2). These authors also demonstrated, by flow cytometry, the increase in the expression of CD206 after stimulation of Sj16 in peritoneal macrophages and leucocytes from the livers of mice. Corroborating these results, Hu et al. (Reference Hu, Wu, Yang and Fung2009) showed that Sj16 decreased the levels of pro-inflammatory cytokines, such as IL-6 and TNF-α, and increased the levels of IL-10 in RAW264.7 macrophages. Vannella et al. (Reference Vannella, Barron, Borthwick, Kindrachuk, Narasimhan, Hart, Thompson, White, Cheever, Ramalingam and Wynn2014) observed that mice infected with S. mansoni showed an increase in M2 macrophages that expressed Arg-1, which attenuated the progression of inflammation and fibrosis (Table 1). Stimulation of RAW264.7 macrophages with another S. japonicum egg protein (SjCP1412) also increased the expression of CD206, Arg-1 and IL-10, which are related to M2-type macrophage differentiation (Ke et al., Reference Ke, Shen, Song, Yu, Kikuchi, Hirayama, Gao, Wang, Yin, Yao, Liu and Zhou2017). Overall, these findings emphasize that M2 macrophages are important in reducing the lesions caused by schistosomiasis through downregulation of the Th1 response and inflammation promoted by egg antigens. Additionally, the role of these cells was previously investigated in a mouse model of liver injury induced by acetaminophen (paracetamol) (Starkey Lewis et al., Reference Starkey Lewis, Campana, Aleksieva, Cartwright, Mackinnon, O'Duibhir, Kendall, Vermeren, Thomson, Gadd, Dwyer, Aird, Man, Rossi, Forrester, Park and Forbes2020). The authors demonstrated that the injection of M2 macrophages in this experimental model was able to rapidly reduce liver damage and inflammation. These data indicate that M2 macrophages may constitute a new potential cell-based therapy for this disease. Based on this, it seems promissory also to apply these cells in the immunotherapy of schistosomiasis.

Interestingly, despite being produced by M1 macrophages, a recent study demonstrated that the production of ROS by egg antigens may be a potential mechanism for M2 macrophage differentiation (Table 2). ROS have several biological activities, such as participation in innate and adaptive immune responses, and can be cytotoxic against pathogens (Canton et al., Reference Canton, Sánchez-Rodríguez, Spera, Venegas, Favia, Viola and Castegna2021). Yu et al. (Reference Yu, Wang, Wang, Gu, Lei, Tang, Wei, Xu, Wang, Chen, Pu, Qi, Yu, Chen, Zhu, Li, Zhang, Zhou and Su2021) observed that a significant increase in ROS in the liver of mice infected with S. japonicum was related to fibrosis and the differentiation of M2 macrophages. The authors hypothesized that their findings were due to NADPH oxidase (NOX2) inhibiting SEA-stimulated ROS production in macrophages, suggesting that NOX might act as the main source of ROS production in SEA-stimulated macrophages. NADPH oxidase is the first source of ROS identified in macrophages (Nathan et al., Reference Nathan and Cunningham-Bussel2013). Macrophages produce large amounts of ROS, primarily through NOX2 activation (Paik et al., Reference Paik, Kim, Aoyama, De Minicis, Bataller and Brenner2014). Thus, the production of ROS induced by schistosome eggs may be a target for the treatment of schistosomiasis.

Future perspectives and final considerations

Findings about the mechanisms behind macrophage activation during different metabolic profiles in human diseases present an exciting prospect, as there are pathologies that have been associated with a particular macrophage phenotype. In this context, the polarization of macrophages in schistosomiasis and their consequent ability to promote an effective immune response seem to be an attractive therapeutic approach associated with conventional chemotherapy treatments.

Overall, the findings highlighted in this review demonstrate the relevance and complexity of understanding the mechanisms involved in macrophage polarization (M1/M2) in schistosomiasis. The S. japonicum and S. mansoni antigens in macrophage polarization are particularly important in this process. These products have been shown to have immunomodulatory effects in different phases of schistosomiasis and are seen as potential therapeutic targets for this disease, especially in the chronic phase. Among the potential therapeutics, the combination of different schistosome antigens can result in higher levels of host protection, stimulating an adequate immune response for either an M1 or M2 profile; however, this can only be achieved after many in vitro and in vivo experiments.

Acknowledgements

We acknowledge the Graduate Program in Health Sciences at the Federal University of Maranhão for providing the support. We would like to acknowledge Editage (www.editage.com) for English-language editing.

Author's contributions

I. C. L. L.: investigation, conceptualization and writing of the original draft; I. C. L. L., G. C. C. G. F., R. A. N., M. G. S. L., V. A. F. S., J. G. M. R., G. S. M., R. C. C., L. A. S., R. N. M. G. and F. R. F. N.: review and editing of original draft. All the authors reviewed and approved the final version of this manuscript.

Financial support

I. C. L. L., G. C. C. G. F., R. A. N. and V. A. F. S. were supported by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior doctoral fellowship. R. N. M. G. and F. R. F. N. were supported by CNPq researcher fellowship. The research was supported by FAPEMA (Universal 01001/19) and by CAPES (Financial code 001).

Conflict of interest

The authors declare no conflict of interest.

References

Abdalla, HB, Napimoga, MH, Lopes, AH, de Macedo Maganin, AG, Cunha, TM, Van Dyke, TE and Clemente Napimoga, JT (2020) Activation of PPAR-γ induces macrophage polarization and reduces neutrophil migration mediated by heme oxygenase 1. International Immunopharmacology 84, 106565.CrossRefGoogle ScholarPubMed
Abdel-Ghany, R, Rabia, I, El-Ahwany, E, Saber, S, Gamal, R, Nagy, F, Mahmoud, O, Hamad, RS and Barakat, W (2015) Blockade of PGE2, PGD2 receptors confers protection against prepatent Schistosomiasis mansoni in mice. Journal of the Egyptian Society of Parasitology 45, 511520.Google ScholarPubMed
Acharya, S, Da'dara, AA and Skelly, PJ (2021) Schistosome immunomodulators. PLoS Pathogens 17, e1010064.CrossRefGoogle ScholarPubMed
Ahmadian, M, Suh, JM, Hah, N, Liddle, C, Atkins, AR, Downes, M and Evans, RM (2013) PPARγ signaling and metabolism: the good, the bad and the future. Nature Medicine 19, 557566.CrossRefGoogle Scholar
Angeli, V, Faveeuw, C, Roye, O, Fontaine, J, Teissier, E, Capron, A, Wolowezuk, I, Capron, M and Trottein, F (2001) Role of the parasite-derived prostaglandin D2 in the inhibition of epidermal Langerhans cell migration during schistosomiasis infection. The Journal of Experimental Medicine 193, 11351147.CrossRefGoogle ScholarPubMed
Anthony, B, Mathieson, W, de Castro-Borges, W and Allen, J (2010) Schistosoma mansoni: egg-induced downregulation of hepatic stellate cell activation and fibrogenesis. Experimental Parasitology 124, 409420.CrossRefGoogle ScholarPubMed
Assunção, LS, Magalhães, KG, Carneiro, AB, Molinaro, R, Almeida, PE, Atella, GC, Castro-Faria-Neto, HC and Bozza, PT (2017) Schistosomal-derived lysophosphatidylcholine triggers M2 polarization of macrophages through PPARγ dependent mechanisms. Biochimica et Biophysica Acta. Molecular and Cell Biology of Lipids 1862, 246254.CrossRefGoogle ScholarPubMed
Atri, C, Guerfali, FZ and Laouini, D (2018) Role of human macrophage polarization in inflammation during infectious diseases. International Journal of Molecular Sciences 19, 1801.CrossRefGoogle ScholarPubMed
Avni, D and Avni, O (2021) Extracellular vesicles: schistosomal long-range precise weapon to manipulate the immune response. Frontiers in Cellular and Infection Microbiology 11, 196.CrossRefGoogle ScholarPubMed
Bajiro, M, Dana, D and Levecke, B (2017) Prevalence and intensity of Schistosoma mansoni infections among schoolchildren attending primary schools in an urban setting in southwest, ethiopia. BMC Research Notes 10, 16.CrossRefGoogle Scholar
Barron, L and Wynn, TA (2011) Macrophage activation governs schistosomiasis-induced inflammation and fibrosis. European Journal of Immunology 41, 2509.CrossRefGoogle ScholarPubMed
Bartlett, A, Brown, M, Marriott, C and Whitfield, PJ (2000) The infection of human skin by schistosome cercariae: studies using Franz cells. Parasitology 121, 4954.CrossRefGoogle ScholarPubMed
Bartley, PB, Ramm, GA, Jones, MK, Ruddell, RG, Li, Y and McManus, DP (2006) A contributory role for activated hepatic stellate cells in the dynamics of Schistosoma japonicum egg-induced fibrosis. International Journal for Parasitology 36, 9931001.CrossRefGoogle ScholarPubMed
Beljaars, L, Schippers, M, Reker-Smit, C, Martinez, FO, Helming, L, Poelstra, K and Melgert, BN (2014) Hepatic localization of macrophage phenotypes during fibrogenesis and resolution of fibrosis in mice and humans. Frontiers in Immunology 5, 430.CrossRefGoogle ScholarPubMed
Bottieau, E, Clerinx, J, de Vega, MR, Van den Enden, E, Colebunders, R, Van Esbroeck, M, Vervoort, T, Van Gompel, A and Van den Ende, J (2006) Imported Katayama fever: clinical and biological features at presentation and during treatment. The Journal of Infection 52, 339345.CrossRefGoogle ScholarPubMed
Bourke, CD, Prendergast, CT, Sanin, DE, Oulton, TE, Hall, RJ and Mountford, AP (2015) Epidermal keratinocytes initiate wound healing and pro-inflammatory immune responses following percutaneous schistosome infection. International Journal for Parasitology 45, 215224.CrossRefGoogle ScholarPubMed
Braga, TT, Agudelo, JSH and Camara, NOS (2015) Macrophages during the fibrotic process: M2 as friend and foe. Frontiers in Immunology 6, 602.CrossRefGoogle ScholarPubMed
Brännström, K, Sellin, ME, Holmfeldt, P, Brattsand, M and Gullberg, M (2009) The Schistosoma mansoni protein Sm16/SmSLP/SmSPO-1 assembles into a nine-subunit oligomer with potential to inhibit Toll-like receptor signaling. Infection and Immunity 77, 1144.CrossRefGoogle ScholarPubMed
Brink, LH, McLaren, DJ and Smithers, SR (1977) Schistosoma mansoni: a comparative study of artificially transformed schistosomula and schistomula recovered after cercarial penetration of isolated skin. Parasitology 74, 7386.CrossRefGoogle Scholar
Brunet, LR, Finkelman, FD, Cheever, AW, Kopf, MA and Pearce, EJ (1997) IL-4 protects against TNF-alpha-mediated cachexia and death during acute schistosomiasis. The Journal of Immunology 159, 777785.CrossRefGoogle ScholarPubMed
Burke, ML, Jones, MK, Gobert, GN, Li, YS, Ellis, MK and McManus, DP (2009) Immunopathogenesis of human schistosomiasis. Parasite Immunology 31, 163176.CrossRefGoogle ScholarPubMed
Burke, ML, McGarvey, L, McSorley, HJ, Bielefeldt-Ohmann, H, McManus, DP and Gobert, GN (2011) Migrating Schistosoma japonicum schistosomula induce an innate immune response and wound healing in the murine lung. Molecular Immunology 49, 191200.CrossRefGoogle ScholarPubMed
Caldas, IR, Campi-Azevedo, AC, Oliveira, LFA, Silveira, AMS, Oliveira, RC and Gazzinelli, G (2008) Human Schistosomiasis mansoni: immune responses during acute and chronic phases of the infection. Acta Tropica 108, 109117.CrossRefGoogle ScholarPubMed
Canton, M, Sánchez-Rodríguez, R, Spera, I, Venegas, FC, Favia, M, Viola, A and Castegna, A (2021) Reactive oxygen species in macrophages: sources and targets. Frontiers in Immunology 12, 734229.CrossRefGoogle ScholarPubMed
Cardoso, FC, Macedo, GC, Gava, E, Kitten, GT, Mati, VL, de Melo, AL, Caliari, MV, Almeida, GT, Venancio, TM, Verjovski-Almeida, S and Oliveira, SC (2008) Schistosoma mansoni tegument protein Sm29 is able to induce a Th1-type of immune response and protection against parasite infection. PLoS Neglected Tropical Diseases 2, e-308.CrossRefGoogle ScholarPubMed
Cass, CL, Johnson, JR, Califf, LL, Xu, T, Hernandez, HJ, Stadecker, MJ, Yates, JR and Williams, DL (2007) Proteomic analysis of Schistosoma mansoni egg secretions. Molecular and Biochemical Parasitology 155, 84.CrossRefGoogle ScholarPubMed
Cervi, L, MacDonald, AS, Kane, C, Dzierszinski, F and Pearce, EJ (2004) Cutting edge: dendritic cells copulsed with microbial and helminth antigens undergo modified maturation, segregate the antigens to distinct intracellular compartments, and concurrently induce microbe-specific Th1 and helminth-specific Th2 responses. The Journal of Immunology 172, 20162020.CrossRefGoogle Scholar
Cheever, AW, Hoffmann, KF and Wynn, TA (2000) Immunopathology of Schistosomiasis mansoni in mice and men. Immunology Today 21, 465466.CrossRefGoogle ScholarPubMed
Chen, BL, Peng, J, Li, QF, Yang, M, Wang, Y and Chen, W (2013) Exogenous bone morphogenetic protein-7 reduces hepatic fibrosis in Schistosoma japonicum-infected mice via transforming growth factor-β/Smad signaling. World Journal of Gastroenterology 19, 1405.CrossRefGoogle ScholarPubMed
Chen, L, He, B, Hou, W and He, L (2017 a) Cysteine protease inhibitor of Schistosoma japonicum – a parasite-derived negative immunoregulatory factor. Parasitology Research 116, 901908.CrossRefGoogle ScholarPubMed
Chen, XX, Tang, L, Fu, YM, Wang, Y, Han, ZH and Meng, JG (2017 b) Paralemmin-3 contributes to lipopolysaccharide-induced inflammatory response and is involved in lipopolysaccharide-Toll-like receptor-4 signaling in alveolar macrophages. International Journal of Molecular Medicine 40, 19211931.Google ScholarPubMed
Chiaramonte, MG, Schopf, LR, Neben, TY, Cheever, AW, Donaldson, DD and Wynn, TA (1999) IL-13 is a key regulatory cytokine for Th2 cell-mediated pulmonary granuloma formation and IgE responses induced by Schistosoma mansoni eggs. Journal of Immunology 162, 920930.CrossRefGoogle ScholarPubMed
Ciesielska, A, Matyjek, M and Kwiatkowska, K (2021) TLR4 and CD14 trafficking and its influence on LPS-induced pro-inflammatory signaling. Cellular and Molecular Life Sciences 78, 12331261.CrossRefGoogle ScholarPubMed
Colley, DG and Secor, WE (2014) Immunology of human schistosomiasis. Parasite Immunology 36, 347357.CrossRefGoogle ScholarPubMed
Colley, DG, Bustinduy, AL, Secor, WE and King, CH (2014) Human schistosomiasis. The Lancet 383, 22532264.CrossRefGoogle ScholarPubMed
Cortes-Selva, D and Fairfax, K (2021) Schistosome and intestinal helminth modulation of macrophage immunometabolism. Immunology 162, 123134.CrossRefGoogle ScholarPubMed
Cortes-Selva, D, Elvington, AF, Ready, A, Rajwa, B, Pearce, EJ, Randolph, GJ and Fairfax, KC (2018) Schistosoma mansoni infection-induced transcriptional changes in hepatic macrophage metabolism correlate with an athero-protective phenotype. Frontiers in Immunology 9, 2580.CrossRefGoogle ScholarPubMed
Costain, AH, MacDonald, AS and Smits, HH (2018) Schistosome egg migration: mechanisms, pathogenesis and host immune responses. Frontiers in Immunology 9, 3042.CrossRefGoogle ScholarPubMed
Coulson, PS, Smythies, LE, Betts, C, Mabbott, NA, Sternberg, JM, Wei, XG, Liew, FY and Wilson, RA (1998) Nitric oxide produced in the lungs of mice immunized with the radiation-attenuated schistosome vaccine is not the major agent causing challenge parasite elimination. Immunology 93, 5563.CrossRefGoogle Scholar
Crabtree, JE and Wilson, RA (1986) The role of pulmonary cellular reactions in the resistance of vaccinated mice to Schistosoma mansoni. Parasite Immunology 8, 265285.CrossRefGoogle ScholarPubMed
Cudejko, C, Wouters, K, Fuentes, L, Hannou, SA, Paquet, C, Bantubungi, K, Bouchaert, E, Vanhoutte, J, Fleury, S, Remy, P, Tailleux, A, Chinetti-Gbaguidi, G, Dombrowicz, D, Staels, B and Paumelle, R (2011) p16INK4a deficiency promotes IL-4-induced polarization and inhibits proinflammatory signaling in macrophages. Blood 118, 25562566.CrossRefGoogle ScholarPubMed
Curwen, RS and Wilson, RA (2003) Invasion of skin by schistosome cercariae: some neglected facts. Trends in Parasitology 19, 6366.CrossRefGoogle ScholarPubMed
Curwen, RS, Ashton, PD, Sundaralingam, S and Wilson, RA (2006) Identification of novel proteases and immunomodulators in the secretions of schistosome cercariae that facilitate host entry. Molecular and Cellular Proteomics 5, 835844.CrossRefGoogle ScholarPubMed
de Andrade, LF, de Mourão, MM, Geraldo, JA, Coelho, FS, Silva, LL, Neves, RH, Volpini, A, Machado-Silva, JR, Araujo, N, Nacif-Pimenta, R, Caffrey, CR and Oliveira, G (2014) Regulation of Schistosoma mansoni development and reproduction by the mitogen-activated protein kinase signaling pathway. PLoS Neglected Tropical Diseases 8, e2949.CrossRefGoogle Scholar
de Jong, EC, Vieira, PL, Kalinski, P, Schuitemaker, JHN, Tanaka, Y, Wierenga, EA, Yazdanbakhsh, M and Kapsenberg, ML (2002) Microbial compounds selectively induce Th1 cell-promoting or Th2 cell-promoting dendritic cells in vitro with diverse Th cell-polarizing signals. The Journal of Immunology 168, 17041709.CrossRefGoogle ScholarPubMed
De Oliveira Fraga, LA, Torrero, MN, Tocheva, AS, Mitre, E and Davies, SJ (2010) Induction of type 2 responses by schistosome worms during prepatent infection. The Journal of Infectious Diseases 201, 464.CrossRefGoogle ScholarPubMed
Dewals, BG, Marillier, RG, Hoving, JC, Leeto, M, Schwegmann, A and Brombacher, F (2010) IL-4Rα-independent expression of mannose receptor and Ym1 by macrophages depends on their IL-10 responsiveness. PLoS Neglected Tropical Diseases 4, e689.CrossRefGoogle ScholarPubMed
Doenhoff, MJ (1998) A vaccine for schistosomiasis: alternative approaches. Parasitology Today 14, 105109.CrossRefGoogle ScholarPubMed
Doenhoff, MJ, Butterworth, AE, Hayes, RJ, Sturrock, RF, Ouma, JH, Koech, D, Prentice, M and Bain, J (1993) Seroepidemiology and serodiagnosis of schistosomiasis in Kenya using crude and purified egg antigens of Schistosoma mansoni in ELISA. Transactions of the Royal Society of Tropical Medicine and Hygiene 87, 4248.CrossRefGoogle ScholarPubMed
Du, P, Ma, Q, Zhu, Z, Li, G, Wang, Y, Li, QQ, Chen, YF, Shang, ZZ, Zhang, J and Zhao, L (2016) Mechanism of corilagin interference with IL-13/STAT6 signaling pathways in hepatic alternative activation macrophages in schistosomiasis-induced liver fibrosis in mouse model. European Journal of Pharmacology 793, 119126.CrossRefGoogle ScholarPubMed
Dunne, DW and Cooke, A (2005) A worm's eye view of the immune system: consequences for evolution of human autoimmune disease. Nature Reviews. Immunology 5, 420426.CrossRefGoogle ScholarPubMed
Dunne, DW, Jones, FM and Doenhoff, MJ (1991) The purification, characterization, serological activity and hepatotoxic properties of two cationic glycoproteins (alpha 1 and omega 1) from Schistosoma mansoni eggs. Parasitology 103(Pt 2), 225236.CrossRefGoogle ScholarPubMed
Egesa, M, Lubyayi, L, Tukahebwa, EM, Bagaya, BS, Chalmers, IW, Wilson, S, Hokke, CH, Hoffmann, KF, Dunne, DW, Yazdanbakhsh, M, Labuda, LA and Cose, S (2018) Schistosoma mansoni schistosomula antigens induce Th1/pro-inflammatory cytokine responses. Parasite Immunology 40, e12592.CrossRefGoogle ScholarPubMed
El-Faham, MH, Wheatcroft-Francklow, KJ, Price, HP, Sayers, JR and Doenhoff, MJ (2017) Schistosoma mansoni cercarial elastase (SmCE): differences in immunogenic properties of native and recombinant forms. Parasitology 144, 13561364.CrossRefGoogle ScholarPubMed
El-Zayat, SR, Sibaii, H and Mannaa, FA (2019) Toll-like receptors activation, signaling, and targeting: an overview. Bulletin of the National Research Centre 43, 112.CrossRefGoogle Scholar
Enderlin Vaz Da Silva, Z, Lehr, HA and Velin, D (2014) In vitro and in vivo repair activities of undifferentiated and classically and alternatively activated macrophages. Pathobiology: Journal of Immunopathology, Molecular and Cellular Biology 81, 8693.CrossRefGoogle ScholarPubMed
Everts, B, Perona-Wright, G, Smits, HH, Hokke, CH, van der Ham, AJ, Fitzsimmons, CM, Doenhoff, MJ, van der Bosch, J, Mohrs, K, Haas, H, Mohrs, M, Yazdanbakhsh, M and Schramm, G (2009) Omega-1, a glycoprotein secreted by Schistosoma mansoni eggs, drives Th2 responses. The Journal of Experimental Medicine 206, 16731680.CrossRefGoogle ScholarPubMed
Fang, Y, Wu, C, Chen, Q, Wu, J, Yang, Y, Guo, X, Chen, G and Wang, Z (2015) SjE16.7 activates macrophages and promotes Schistosoma japonicum egg-induced granuloma development. Acta Tropica 149, 4958.CrossRefGoogle ScholarPubMed
Fei-Yue, L, Hong-Zhuan, T, Jie, Z, Rui-Hong, Z, Jin-Hua, Z, Xin-Ting, C and Guang-Hui, R (2017) Analysis of characteristics of medical assistance to advanced schistosomiasis patients in Hunan province, 2015. Zhongguo xue xi chong bing fang zhi za zhi = Chinese Journal of Schistosomiasis Control 29, 281285.Google ScholarPubMed
Ferrante, CJ, Pinhal-Enfield, G, Elson, G, Cronstein, BN, Hasko, G, Outram, S and Leibovich, SJ (2013) The adenosine-dependent angiogenic switch of macrophages to an M2-like phenotype is independent of interleukin-4 receptor alpha (IL4Rα) signaling. Inflammation 36, 921.CrossRefGoogle Scholar
Fraternale, A, Brundu, S and Magnani, M (2015) Polarization and repolarization of macrophages. Journal of Clinical and Cellular Immunology 6, 1000319.Google Scholar
Freitas, MS, Oliveira, AF, Da Silva, TA, Fernandes, FF, Gonçales, RA, Almeida, F and Roque-Barreira, MC (2016) Paracoccin induces M1 polarization of macrophages via interaction with TLR4. Frontiers in Microbiology 7, 1003.CrossRefGoogle ScholarPubMed
Gao, Y, Chen, L, Hou, M, Chen, Y, Ji, M, Wu, H and Wu, G (2013) TLR2 directing PD-L2 expression inhibit T cells response in Schistosoma japonicum infection. PLoS ONE 8, e82480.CrossRefGoogle ScholarPubMed
Gao, S, Zhou, J, Liu, N, Wang, L, Gao, Q, Wu, Y, Zhao, Q, Liu, P, Wang, S, Liu, Y, Guo, N, Shen, Y, Wu, Y and Yuan, Z (2015) Curcumin induces M2 macrophage polarization by secretion IL-4 and/or IL-13. Journal of Molecular and Cellular Cardiology 85, 131139.CrossRefGoogle ScholarPubMed
Ghosh, S, Shang, P, Yazdankhah, M, Bhutto, I, Hose, S, Montezuma, SR, Luo, T, Chattopadhyay, S, Qian, J, Lutty, GA, Ferrington, DA, Zigler, JS and Sinha, D (2017) Activating the AKT2-nuclear factor-κB–lipocalin-2 axis elicits an inflammatory response in age-related macular degeneration. The Journal of Pathology 241, 583588.CrossRefGoogle ScholarPubMed
Gobbi, F, Tamarozzi, F, Buonfrate, D, van Lieshout, L, Bisoffi, Z and Bottieau, E (2020) New insights on acute and chronic schistosomiasis: do we need a redefinition? Trends in Parasitology 36, 660667.CrossRefGoogle Scholar
Goh, F, Irvine, KM, Lovelace, E, Donnelly, S, Jones, MK, Brion, K, Hume, DA, Kotze, AC, Dalton, JP, Ingham, A and Sweet, MJ (2009) Selective induction of the notch ligand Jagged-1 in macrophages by soluble egg antigen from Schistosoma mansoni involves ERK signalling. Immunology 127, 326.CrossRefGoogle ScholarPubMed
Gong, M, Zhuo, X and Ma, A (2017) STAT6 upregulation promotes M2 macrophage polarization to suppress atherosclerosis. Medical Science Monitor Basic Research 23, 240249.CrossRefGoogle ScholarPubMed
Gong, W, Huang, F, Sun, L, Yu, A, Zhang, X, Xu, Y, Shen, Y and Cao, J (2018) Toll-like receptor-2 regulates macrophage polarization induced by excretory–secretory antigens from Schistosoma japonicum eggs and promotes liver pathology in murine schistosomiasis. PLoS Neglected Tropical Diseases 12, e0007000.CrossRefGoogle ScholarPubMed
Gordon, S (2003) Alternative activation of macrophages. Nature Reviews Immunology 3, 2335.CrossRefGoogle ScholarPubMed
Gordon, S and Martinez-Pomares, L (2017) Physiological roles of macrophages. Pflugers Archiv: European Journal of Physiology 469, 365374.CrossRefGoogle ScholarPubMed
Grenfell, RFQ, Martins, WH, Silva-Moraes, V, Barata, SVB, Ribeiro, EG, Oliveira, E and Coelho, PMZ (2012) Antigens of worms and eggs showed a differentiated detection of specific IgG according to the time of Schistosoma mansoni infection in mice. Revista da Sociedade Brasileira de Medicina Tropical 45, 505509.CrossRefGoogle Scholar
Grieco, S, Sulekova, LF, Nardelli, S, Riggio, O, Venditti, M and Taliani, G (2016) Portal hypertension related to schistosomiasis treated with a transjugular intrahepatic portosystemic shunt. Journal of Clinical Gastroenterology 50, 608610.CrossRefGoogle ScholarPubMed
Gryseels, B, Polman, K, Clerinx, J and Kestens, L (2006) Human schistosomiasis. The Lancet 368, 11061118.CrossRefGoogle ScholarPubMed
Grzych, JM, Pearce, E, Cheever, A, Caulada, ZA, Caspar, P, Heiny, S, Lewis, F and Sher, A (1991) Egg deposition is the major stimulus for the production of Th2 cytokines in murine Schistosomiasis mansoni. The Journal of Immunology 146, 132213227.CrossRefGoogle ScholarPubMed
Hai, Y, Edwards, JE, Van Zandt, MC, Hoffmann, KF and Christianson, DW (2014) Crystal structure of Schistosoma mansoni arginase, a potential drug target for the treatment of schistosomiasis. Biochemistry 53, 46714684.CrossRefGoogle ScholarPubMed
Hambrook, JR and Hanington, PC (2021) Immune evasion strategies of schistosomes. Frontiers in Immunology 11, 624178.CrossRefGoogle ScholarPubMed
Hams, E, Aviello, G and Fallon, PG (2013) The schistosoma granuloma: friend or foe? Frontiers in Immunology 4, 89.CrossRefGoogle ScholarPubMed
Harizi, H, Juzan, M, Pitard, V, Moreau, JF and Gualde, N (2002) Cyclooxygenase-2-issued prostaglandin e(2) enhances the production of endogenous IL-10, which down-regulates dendritic cell functions. The Journal of Immunology 168, 22552263.CrossRefGoogle ScholarPubMed
He, YX, Yu, QF, Yu, P, Mao, CS and Hu, YQ (1990) Penetration of Schistosoma japonicum cercaria into host skin. Chinese Medical Journal 103, 3444.Google ScholarPubMed
He, YX, Salafsky, B and Ramaswamy, K (2005) Comparison of skin invasion among three major species of Schistosoma. Trends in Parasitology 21, 201203.CrossRefGoogle ScholarPubMed
He, B, Cai, G, Ni, Y, Li, Y, Zong, H and He, L (2011) Characterization and expression of a novel cystatin gene from Schistosoma japonicum. Molecular and Cellular Probes 25, 186193.CrossRefGoogle ScholarPubMed
He, Y, Gao, Y, Zhang, Q, Zhou, G, Cao, F and Yao, S (2020) IL-4 switches microglia/macrophage M1/M2 polarization and alleviates neurological damage by modulating the JAK1/STAT6 pathway following ICH. Neuroscience 437, 161171.CrossRefGoogle ScholarPubMed
Hervé, M, Angeli, V, Pinzar, E, Wintjens, R, Faveeuw, C, Narumiya, S, Capron, A, Urade, Y, Capron, M, Riveau, G and Trottein, F (2003) Pivotal roles of the parasite PGD2 synthase and of the host D prostanoid receptor 1 in schistosome immune evasion. European Journal of Immunology 33, 27642772.CrossRefGoogle Scholar
Hesse, M, Modolell, M, La Flamme, AC, Schito, M, Fuentes, JM, Cheever, AW, Pearce, EJ and Wynn, TA (2001) Differential regulation of nitric oxide synthase-2 and arginase-1 by type 1/type 2 cytokines in vivo: granulomatous pathology is shaped by the pattern of l-arginine metabolism. The Journal of Immunology 167, 65336544.CrossRefGoogle ScholarPubMed
Hesse, M, Piccirillo, CA, Belkaid, Y, Prufer, J, Mentink-Kane, M, Leusink, M, Cheever, AW, Shevach, EM and Wynn, TA (2004) The pathogenesis of schistosomiasis is controlled by cooperating IL-10-producing innate effector and regulatory T cells. The Journal of Immunology 172, 31573166.CrossRefGoogle ScholarPubMed
Ho, CH, Cheng, CH, Huang, TW, Peng, SY, Lee, KM and Cheng, PC (2022) Switched phenotypes of macrophages during the different stages of Schistosoma japonicum infection influenced the subsequent trends of immune responses. Journal of Microbiology, Immunology and Infection 55, 503526.CrossRefGoogle ScholarPubMed
Hoffmann, KF, Cheever, AW and Wynn, TA (2000) IL-10 and the dangers of immune polarization: excessive type 1 and type 2 cytokine responses induce distinct forms of lethal immunopathology in murine schistosomiasis. The Journal of Immunology 164, 64066416.CrossRefGoogle ScholarPubMed
Hogg, KG, Kumkate, S, Anderson, S and Mountford, AP (2003 a) Interleukin-12 p40 secretion by cutaneous CD11c+ and F4/80+ cells is a major feature of the innate immune response in mice that develop Th1-mediated protective immunity to Schistosoma mansoni. Infection and Immunity 71, 35633571.CrossRefGoogle Scholar
Hogg, KG, Kumkate, S and Mountford, AP (2003 b) IL-10 regulates early IL-12-mediated immune responses induced by the radiation-attenuated schistosome vaccine. International Immunology 15, 14511459.CrossRefGoogle ScholarPubMed
Houlder, EL, Costain, AH, Cook, PC and MacDonald, AS (2021) Schistosomes in the lung: immunobiology and opportunity. Frontiers in Immunology 12, 1330.CrossRefGoogle ScholarPubMed
Hu, S, Wu, Z, Yang, L and Fung, MC (2009) Molecular cloning and expression of a functional anti-inflammatory protein, Sj16, of Schistosoma japonicum. International Journal for Parasitology 39, 191200.CrossRefGoogle ScholarPubMed
Hussell, T and Bell, TJ (2014) Alveolar macrophages: plasticity in a tissue-specific context. Nature Reviews. Immunology 14, 8193.CrossRefGoogle Scholar
Ingram, RJ, Bartlett, A, Brown, MB, Marriott, C and Whitfield, PJ (2003) Penetration of human skin by the cercariae of Schistosoma mansoni: an investigation of the effect of multiple cercarial applications. Journal of Helminthology 77, 2731.CrossRefGoogle ScholarPubMed
Ingram, JR, Rafi, SB, Eroy-Reveles, AA, Ray, M, Lambeth, L, Hsieh, I, Ruelas, D, Lim, KC, Sakanari, J, Craik, CS, Jacobson, MP and McKerrow, JH (2012) Investigation of the proteolytic functions of an expanded cercarial elastase gene family in Schistosoma mansoni. PLoS Neglected Tropical Diseases 6, e1589.CrossRefGoogle ScholarPubMed
Ismail, HAHA, Hong, ST, Babiker, ATEB, Hassan, RMAE, Sulaiman, MAZ, Jeong, HG, Kong, WH, Lee, SH, Cho, HI, Nam, HS, Oh, CH and Lee, YH (2014) Prevalence, risk factors, and clinical manifestations of schistosomiasis among school children in the White Nile River basin, Sudan. Parasites & Vectors 7, 478.CrossRefGoogle ScholarPubMed
James, SL and Glaven, J (1989) Macrophage cytotoxicity against schistosomula of Schistosoma mansoni involves arginine-dependent production of reactive nitrogen intermediates. The Journal of Immunology 143, 42084212.CrossRefGoogle ScholarPubMed
James, SL, Cheever, AW, Caspar, P and Wynn, TA (1998) Inducible nitric oxide synthase-deficient mice develop enhanced type 1 cytokine-associated cellular and humoral immune responses after vaccination with attenuated Schistosoma mansoni cercariae but display partially reduced resistance. Infection and Immunity 66, 3510.CrossRefGoogle ScholarPubMed
Jang-Lee, J, Curwen, RS, Ashton, PD, Tissot, B, Mathieson, W, Panico, M, Dell, A, Wilson, RA and Haslam, SM (2007) Glycomics analysis of Schistosoma mansoni egg and cercarial secretions. Molecular & Cellular Proteomics 6, 14851499.CrossRefGoogle ScholarPubMed
Jang, E, Lee, S, Kim, JH, Kim, JH, Seo, JW, Lee, WH, Mori, K, Nakao, K and Suk, K (2013) Secreted protein lipocalin-2 promotes microglial M1 polarization. FASEB Journal 27, 11761190.CrossRefGoogle ScholarPubMed
Janssen, L, Silva Santos, GL, Muller, HS, Vieira, ARA, De Campos, TA and De Paulo Martins, V (2016) Schistosome-derived molecules as modulating actors of the immune system and promising candidates to treat autoimmune and inflammatory diseases. Journal of Immunology Research 2016, 5267485.CrossRefGoogle ScholarPubMed
Jenkins, SJ, Hewitson, JP, Ferret-Bernard, S and Mountford, AP (2005 a) Schistosome larvae stimulate macrophage cytokine production through TLR4-dependent and -independent pathways. International Immunology 17, 14091418.CrossRefGoogle ScholarPubMed
Jenkins, SJ, Hewitson, JP, Jenkins, GR and Mountford, AP (2005 b) Modulation of the host's immune response by schistosome larvae. Parasite Immunology 27, 385393.CrossRefGoogle ScholarPubMed
Jenkins, SJ, Ruckerl, D, Cook, PC, Jones, LH, Finkelman, FD, Van Rooijen, N, MacDonald, AS and Allen, JE (2011) Local macrophage proliferation, rather than recruitment from the blood, is a signature of Th2 inflammation. Science (New York, N.Y.) 332, 12841288.CrossRefGoogle ScholarPubMed
Jetten, N, Verbruggen, S, Gijbels, MJ, Post, MJ, De Winther, MPJ and Donners, MMPC (2014) Anti-inflammatory M2, but not pro-inflammatory M1 macrophages promote angiogenesis in vivo. Angiogenesis 17, 109118.CrossRefGoogle Scholar
Jin, L, Yuan, F, Chen, C, Wu, J, Gong, R, Yuan, G, Zeng, H, Pei, J and Chen, T (2019) Degradation products of polydopamine restrained inflammatory response of LPS-stimulated macrophages through mediation TLR-4-MyD88 dependent signaling pathways by antioxidant. Inflammation 42, 658671.CrossRefGoogle ScholarPubMed
Kaisar, MMM, Ritter, M, del Fresno, C, Jónasdóttir, HS, van der Ham, AJ, Pelgrom, LR, Schramm, G, Layland, LE, Sancho, D, Prazeres da Costa, C, Giera, M, Yazdanbakhsh, M and Everts, B (2018) Dectin-1/2-induced autocrine PGE2 signaling licenses dendritic cells to prime Th2 responses. PLoS Biology 16, e2005504.CrossRefGoogle ScholarPubMed
Kamdem, SD, Moyou-Somo, R, Brombacher, F and Nono, JK (2018) Host regulators of liver fibrosis during human schistosomiasis. Frontiers in Immunology 9, 2781.CrossRefGoogle ScholarPubMed
Ke, XD, Shen, S, Song, LJ, Yu, CX, Kikuchi, M, Hirayama, K, Gao, H, Wang, J, Yin, X, Yao, Y, Liu, Q and Zhou, W (2017) Characterization of Schistosoma japonicum CP1412 protein as a novel member of the ribonuclease T2 molecule family with immune regulatory function. Parasites & Vectors 10, 119.CrossRefGoogle ScholarPubMed
Khammo, N, Bartlett, A, Clothier, RH and Whitfield, PJ (2002) The attachment of Schistosoma mansoni cercariae to human skin cells. Parasitology 124, 2530.CrossRefGoogle ScholarPubMed
Kloc, M, Ghobrial, RM, Wosik, J, Lewicka, A, Lewicki, S and Kubiak, JZ (2019) Macrophage functions in wound healing. Journal of Tissue Engineering and Regenerative Medicine 13, 99109.Google ScholarPubMed
Kloetzel, K and Lewert, RM (1966) Pigment formation in Schistosoma mansoni infections in the white mouse. The American Journal of Tropical Medicine and Hygiene 15, 2831.CrossRefGoogle ScholarPubMed
Knuhr, K, Langhans, K, Nyenhuis, S, Viertmann, K, Overgaard Kildemoes, AM, Doenhoff, MJ, Haas, H and Schramm, G (2018) Schistosoma mansoni egg-released IPSE/alpha-1 dampens inflammatory cytokine responses via basophil interleukin (IL)-4 and IL-13. Frontiers in Immunology 9, 2293.CrossRefGoogle ScholarPubMed
Kumkate, S, Jenkins, GR, Paveley, RA, Hogg, KG and Mountford, AP (2007) CD207+ Langerhans cells constitute a minor population of skin-derived antigen-presenting cells in the draining lymph node following exposure to Schistosoma mansoni. International Journal for Parasitology 37, 209220.CrossRefGoogle ScholarPubMed
Lambertucci, JR (2010) Acute Schistosomiasis mansoni: revisited and reconsidered. Memórias do Instituto Oswaldo Cruz 105, 422435.CrossRefGoogle ScholarPubMed
Langenberg, MCC, Hoogerwerf, MA, Janse, JJ, Van Lieshout, L, Corstjens, PLAM, Roestenberg, M, Van Dam, GJ, Van Diepen, A, De Dood, CJ, Feijt, C, Ganesh, MS, Gerritsma, H, Hardeman, G, Hokke, CH, Koopman, JPR, Kos-Van Oosterhoud, J, Kruize, YCM, Meij, P, Ozir-Fazalalikhan, A, Van Schuijlenburg, R, Smits, HH, Verbeek-Menken, PH, Visser, LG, De Vries, JJC, Winkel, BMF and Yazdanbakhsh, M (2019) Katayama syndrome without Schistosoma mansoni eggs. Annals of Internal Medicine 170, 732733.CrossRefGoogle ScholarPubMed
Lechner, A, Bohnacker, S and Esser-von Bieren, J (2021) Macrophage regulation & function in helminth infection. Seminars in Immunology 53, 101526.CrossRefGoogle ScholarPubMed
Lenzi, HL, Kimmel, E, Schechtman, H, Pelajo-Machado, M, Romanha, WS, Pacheco, RG, Mariano, M and Lenzi, (1998) Histoarchitecture of schistosomal granuloma development and involution: morphogenetic and biomechanical approaches. Memórias do Instituto Oswaldo Cruz 93, 141151.CrossRefGoogle ScholarPubMed
Leontovyč, A, Ulrychová, L, Horn, M and Dvořák, J (2020) Collection of excretory/secretory products from individual developmental stages of the blood fluke Schistosoma mansoni. Methods in Molecular Biology 2151, 5563.CrossRefGoogle ScholarPubMed
Ley, K (2017) M1 means kill; M2 means heal. The Journal of Immunology 199, 21912193.CrossRefGoogle ScholarPubMed
Li, YQ, Chen, YF, Dang, YP, Wang, Y, Shang, ZZ, Ma, Q, Wang, YJ, Zhang, J, Luo, L, Li, QQ and Zhao, L (2017) Corilagin counteracts IL-13Rα1 signaling pathway in macrophages to mitigate schistosome egg-induced hepatic fibrosis. Frontiers in Cellular and Infection Microbiology 7, 443.CrossRefGoogle ScholarPubMed
Liu, Y, Munker, S, Müllenbach, R and Weng, HL (2012) IL-13 signaling in liver fibrogenesis. Frontiers in Immunology 3, 116.CrossRefGoogle ScholarPubMed
Liu, YC, Zou, XB, Chai, YF and Yao, YM (2014) Macrophage polarization in inflammatory diseases. International Journal of Biological Sciences 10, 520529.CrossRefGoogle ScholarPubMed
Liu, M, Ju, C, Du, XF, Shen, HM, Wang, JP, Li, J, Zhang, XM, Feng, Z and Hu, W (2015) Proteomic analysis on cercariae and schistosomula in reference to potential proteases involved in host invasion of Schistosoma japonicum larvae. Journal of Proteome Research 14, 46234634.CrossRefGoogle ScholarPubMed
Liu, CP, Zhang, X, Tan, QL, Xu, WX, Zhou, CY, Luo, M, Li, X, Huang, RY and Zeng, X (2017) NF-κB pathways are involved in M1 polarization of RAW 264.7 macrophage by polyporus polysaccharide in the tumor microenvironment. PLoS ONE 12, e0188317.CrossRefGoogle ScholarPubMed
Liu, J, Zhu, L, Wang, J, Qiu, L, Chen, Y, Davis, RE and Cheng, G (2019) Schistosoma japonicum extracellular vesicle miRNA cargo regulates host macrophage functions facilitating parasitism. PLoS Pathogens 15, e1007817.CrossRefGoogle ScholarPubMed
Locati, M, Curtale, G and Mantovani, A (2020) Diversity, mechanisms, and significance of macrophage plasticity. Annual Review of Pathology 15, 123147.CrossRefGoogle ScholarPubMed
Lopes, DM, Oliveira, SC, Page, B, Carvalho, LP, Carvalho, EM and Cardoso, LS (2019) Schistosoma mansoni rSm29 antigen induces a regulatory phenotype on dendritic cells and lymphocytes from patients with cutaneous leishmaniasis. Frontiers in Immunology 9, 3122.CrossRefGoogle ScholarPubMed
LoVerde, PT (2019) Schistosomiasis. Advances in Experimental Medicine and Biology 1154, 4570.CrossRefGoogle ScholarPubMed
Lu, Y, Li, X, Liu, S, Zhang, Y and Zhang, D (2018) Toll-like receptors and inflammatory bowel disease. Frontiers in Immunology 9, 72.CrossRefGoogle ScholarPubMed
Lundy, SK and Lukacs, NW (2013) Chronic schistosome infection leads to modulation of granuloma formation and systemic immune suppression. Frontiers in Immunology 4, 39.CrossRefGoogle ScholarPubMed
MacDonald, AS, Straw, AD, Dalton, NM and Pearce, EJ (2002) Cutting edge: Th2 response induction by dendritic cells: a role for CD40. The Journal of Immunology 168, 537540.CrossRefGoogle ScholarPubMed
Martinez, FO and Gordon, S (2014) The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000Prime Reports 6, 13.CrossRefGoogle ScholarPubMed
Masamba, P and Kappo, AP (2021) Immunological and biochemical interplay between cytokines, oxidative stress and schistosomiasis. International Journal of Molecular Sciences 22, 7216.CrossRefGoogle ScholarPubMed
Masi, B, Perles-Barbacaru, TA, Bernard, M and Viola, A (2020) Clinical and preclinical imaging of hepatosplenic schistosomiasis. Trends in Parasitology 36, 206226.CrossRefGoogle ScholarPubMed
McManus, DP, Dunne, DW, Sacko, M, Utzinger, J, Vennervald, BJ and Zhou, XN (2018) Schistosomiasis. Nature Reviews. Disease Primers 4, 13.CrossRefGoogle ScholarPubMed
Meevissen, MHJ, Wuhrer, M, Doenhoff, MJ, Schramm, G, Haas, H, Deelder, AM and Hokke, CH (2010) Structural characterization of glycans on omega-1, a major Schistosoma mansoni egg glycoprotein that drives Th2 responses. Journal of Proteome Research 9, 26302642.CrossRefGoogle Scholar
Mehta, A and Baltimore, D (2016) MicroRNAs as regulatory elements in immune system logic. Nature Reviews. Immunology 16, 279294.CrossRefGoogle ScholarPubMed
Menson, EN and Wilson, RA (1990) Lung-phase immunity to Schistosoma mansoni: definition of alveolar macrophage phenotypes after vaccination and challenge of mice. Parasite Immunology 12, 353366.CrossRefGoogle ScholarPubMed
Meyer, NH, Mayerhofer, H, Tripsianes, K, Blindow, S, Barths, D, Mewes, A, Weimar, T, Köhli, T, Bade, S, Madl, T, Frey, A, Haas, H, Mueller-Dieckmann, J, Sattler, M and Schramm, G (2015) A crystallin fold in the interleukin-4-inducing principle of Schistosoma mansoni eggs (IPSE/α-1) mediates IgE binding for antigen-independent basophil activation. The Journal of Biological Chemistry 290, 2211122126.CrossRefGoogle ScholarPubMed
Miller, P and Wilson, RA (1978) Migration of the schistosomula of Schistosoma mansoni from skin to lungs. Parasitology 77, 281302.CrossRefGoogle ScholarPubMed
Mills, CD (2015) Anatomy of a discovery: M1 and M2 macrophages. Frontiers in Immunology 6, 212.CrossRefGoogle ScholarPubMed
Molehin, AJ (2020) Current understanding of immunity against schistosomiasis: impact on vaccine and drug development. Research and Reports in Tropical Medicine 11, 119128.CrossRefGoogle ScholarPubMed
Montagner, S, Dehó, L and Monticelli, S (2014) MicroRNAs in hematopoietic development. BMC Immunology 15, 14.CrossRefGoogle ScholarPubMed
Mouser, EEIM, Pollakis, G, Smits, HH, Thomas, J, Yazdanbakhsh, M, De Jong, EC and Paxton, WA (2019) Schistosoma mansoni soluble egg antigen (SEA) and recombinant omega-1 modulate induced CD4+ T-lymphocyte responses and HIV-1 infection in vitro. PLoS Pathogens 15, e1007924.CrossRefGoogle ScholarPubMed
Murray, PJ (2017) Macrophage polarization. Annual Review of Physiology 79, 541566.CrossRefGoogle ScholarPubMed
Murray, PJ, Allen, JE, Biswas, SK, Fisher, EA, Gilroy, DW, Goerdt, S, Gordon, S, Hamilton, JA, Ivashkiv, LB, Lawrence, T, Locati, M, Mantovani, A, Martinez, FO, Mege, JL, Mosser, DM, Natoli, G, Saeij, JP, Schultze, JL, Shirey, KA, Sica, A, Suttles, J, Udalova, I, van, Ginderachter, Vogel, SN and Wynn, TA (2014) Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 1420.CrossRefGoogle ScholarPubMed
Mutengo, MM, Mduluza, T, Kelly, P, Mwansa, JCL, Kwenda, G, Musonda, P and Chipeta, J (2018) Low IL-6, IL-10, and TNF-α and high IL-13 cytokine levels are associated with severe hepatic fibrosis in Schistosoma mansoni chronically exposed individuals. Journal of Parasitology Research 73, 349360.Google Scholar
Nathan, C and Cunningham-Bussel, A (2013) Beyond oxidative stress: an immunologist's guide to reactive oxygen species. Nature Reviews. Immunology 13, 349361.CrossRefGoogle ScholarPubMed
Nation, CS, Da'dara, AA, Marchant, JK and Skelly, PJ (2020) Schistosome migration in the definitive host. PLoS Neglected Tropical Diseases 14, 112.CrossRefGoogle ScholarPubMed
Nelwan, ML (2019) Schistosomiasis: life cycle, diagnosis, and control. Current Therapeutic Research, Clinical and Experimental 91, 59.CrossRefGoogle ScholarPubMed
Neves, LX, Sanson, AL, Wilson, RA and Castro-Borges, W (2015) What's in SWAP? Abundance of the principal constituents in a soluble extract of Schistosoma mansoni revealed by shotgun proteomics. Parasites & Vectors 8, 19.CrossRefGoogle Scholar
Nowacki, FC, Swain, MT, Klychnikov, OI, Niazi, U, Ivens, A, Quintana, JF, Hensbergen, PJ, Hokke, CH, Buck, AH and Hoffmann, KF (2015) Protein and small non-coding RNA-enriched extracellular vesicles are released by the pathogenic blood fluke Schistosoma mansoni. Journal of Extracellular Vesicles 4, 28665.CrossRefGoogle ScholarPubMed
Oliveira, MF, D'Avila, JCP, Torres, CR, Oliveira, PL, Tempone, AJ, Rumjanek, FD, Braga, CMS, Silva, JR, Dansa-Petretski, M, Oliveira, MA, De Souza, W and Ferreira, ST (2000) Haemozoin in Schistosoma mansoni. Molecular and Biochemical Parasitology 111, 217221.CrossRefGoogle ScholarPubMed
Orecchioni, M, Ghosheh, Y, Pramod, AB and Ley, K (2019) Macrophage polarization: different gene signatures in M1(LPS+) vs classically and M2(LPS−) vs alternatively activated macrophages. Frontiers in Immunology 10, 1084.CrossRefGoogle ScholarPubMed
Oswald, IP, Wynn, TA, Sher, A and James, SL (1994) NO as an affector molecule of parasite killing: modulation of its synthesis by cytokines. Comparative Biochemistry and Physiology Part C: Pharmacology, Toxicology and Endocrinology 108, 1118.Google Scholar
Oyesola, OO, Shanahan, MT, Kanke, M, Mooney, BM, Webb, LM, Smita, S, Matheson, MK, Campioli, P, Pham, D, Früh, SP, McGinty, JW, Churchill, MJ, Cahoon, JL, Sundaravaradan, P, Flitter, BA, Mouli, K, Nadjsombati, MS, Kamynina, E, Peng, SA, Cubitt, RL, Gronert, K, Lord, JD, Rauch, I, von Moltke, J, Sethupathy, P and Tait Wojno, ED (2021) PGD2 and CRTH2 counteract type 2 cytokine-elicited intestinal epithelial responses during helminth infection. The Journal of Experimental Medicine 218, e20202178.CrossRefGoogle ScholarPubMed
Paik, YH, Kim, J, Aoyama, T, De Minicis, S, Bataller, R and Brenner, DA (2014) Role of NADPH oxidases in liver fibrosis. Antioxidants & Redox Signaling 20, 2854.CrossRefGoogle ScholarPubMed
Parisi, L, Gini, E, Baci, D, Tremolati, M, Fanuli, M, Bassani, B, Farronato, G, Bruno, A and Mortara, L (2018) Macrophage polarization in chronic inflammatory diseases: killers or builders? Journal of Immunology Research 2018, 8917804.CrossRefGoogle ScholarPubMed
Paveley, RA, Aynsley, SA, Cook, PC, Turner, JD and Mountford, AP (2009) Fluorescent imaging of antigen released by a skin-invading helminth reveals differential uptake and activation profiles by antigen presenting cells. PLoS Neglected Tropical Diseases 3, e528.CrossRefGoogle ScholarPubMed
Pearce, EJ and MacDonald, AS (2002) The immunobiology of schistosomiasis. Nature Reviews Immunology 2, 499511.CrossRefGoogle ScholarPubMed
Pearce, EJ, Caspar, P, Grzych, JM, Lewis, FA and Sher, A (1991) Downregulation of Th1 cytokine production accompanies induction of Th2 responses by a parasitic helminth, Schistosoma mansoni. The Journal of Experimental Medicine 173, 159166.CrossRefGoogle ScholarPubMed
Pearce, EJ, Kane, CM, Sun, J, Taylor, JJ, McKee, AS and Cervi, L (2004) Th2 response polarization during infection with the helminth parasite Schistosoma mansoni. Immunological Reviews 201, 117126.CrossRefGoogle ScholarPubMed
Perona-Wright, G, Jenkins, SJ and MacDonald, AS (2006) Dendritic cell activation and function in response to Schistosoma mansoni. International Journal for Parasitology 36, 711721.CrossRefGoogle ScholarPubMed
Pesce, JT, Ramalingam, TR, Wilson, MS, Mentink-Kane, MM, Thompson, RW, Cheever, AW, Urban, JF, Wynn, TA and Mansfield, JM (2009) Retnla (relmalpha/fizz1) suppresses helminth-induced Th2-type immunity. PLoS Pathogens 5, e1000393.CrossRefGoogle ScholarPubMed
Piipponen, M, Li, D and Landén, NX (2020) The immune functions of keratinocytes in skin wound healing. International Journal of Molecular Sciences 21, 126.CrossRefGoogle ScholarPubMed
Rahman, K, Vengrenyuk, Y, Ramsey, SA, Vila, NR, Girgis, NM, Liu, J, Gusarova, V, Gromada, J, Weinstock, A, Moore, KJ, Loke, P and Fisher, EA (2017) Inflammatory Ly6Chi monocytes and their conversion to M2 macrophages drive atherosclerosis regression. The Journal of Clinical Investigation 127, 29042915.CrossRefGoogle ScholarPubMed
Ramaswamy, K, Kumar, P and He, YX (2000) A role for parasite-induced PGE2 in IL-10-mediated host immunoregulation by skin stage schistosomula of Schistosoma mansoni. The Journal of Immunology 165, 45674574.CrossRefGoogle ScholarPubMed
Raposo, G and Stoorvogel, W (2013) Extracellular vesicles: exosomes, microvesicles, and friends. The Journal of Cell Biology 200, 373383.CrossRefGoogle ScholarPubMed
Rath, M, Müller, I, Kropf, P, Closs, EI and Munder, M (2014) Metabolism via arginase or nitric oxide synthase: two competing arginine pathways in macrophages. Frontiers in Immunology 5, 532.CrossRefGoogle ScholarPubMed
Ross, AG, Vickers, D, Olds, GR, Shah, SM and McManus, DP (2007) Katayama syndrome. The Lancet. Infectious Diseases 7, 218224.CrossRefGoogle ScholarPubMed
Ross, EA, Devitt, A and Johnson, JR (2021) Macrophages: the good, the bad, and the gluttony. Frontiers in Immunology 12, 708186.CrossRefGoogle ScholarPubMed
Roupé, KM, Nybo, M, Sjöbring, U, Alberius, P, Schmidtchen, A and Sørensen, OE (2010) Injury is a major inducer of epidermal innate immune responses during wound healing. The Journal of Investigative Dermatology 130, 11671177.CrossRefGoogle Scholar
Rückerl, D and Cook, PC (2019) Macrophages assemble! But do they need IL-4R during schistosomiasis? European Journal of Immunology 49, 9961000.CrossRefGoogle ScholarPubMed
Ruzicka, T and Printz, MP (1984) Arachidonic acid metabolism in skin: a review. Reviews of Physiology, Biochemistry and Pharmacology 100, 121160.CrossRefGoogle ScholarPubMed
Salter, JP, Lim, KC, Hansell, E, Hsieh, I and McKerrow, JH (2000) Schistosome invasion of human skin and degradation of dermal elastin are mediated by a single serine protease. Journal of Biological Chemistry 275, 3866738673.CrossRefGoogle ScholarPubMed
Sanin, DE and Mountford, AP (2015) Sm16, a major component of Schistosoma mansoni cercarial excretory/secretory products, prevents macrophage classical activation and delays antigen processing. Parasites & Vectors 8, 112.CrossRefGoogle Scholar
Schmall, A, Al-Tamari, HM, Herold, S, Kampschulte, M, Weigert, A, Wietelmann, A, Vipotnik, N, Grimminger, F, Seeger, W, Pullamsetti, SS and Savai, R (2015) Macrophage and cancer cell cross-talk via CCR2 and CX3CR1 is a fundamental mechanism driving lung cancer. American Journal of Respiratory and Critical Care Medicine 191, 437447.CrossRefGoogle ScholarPubMed
Schramm, G, Gronow, A, Knobloch, J, Wippersteg, V, Grevelding, CG, Galle, J, Fuller, H, Stanley, RG, Chiodini, PL, Haas, H and Doenhoff, MJ (2006) IPSE/alpha-1: a major immunogenic component secreted from Schistosoma mansoni eggs. Molecular and Biochemical Parasitology 147, 919.CrossRefGoogle Scholar
Schramm, G, Mohrs, K, Wodrich, M, Doenhoff, MJ, Pearce, EJ, Haas, H and Mohrs, M (2007) Cutting edge: IPSE/alpha-1, a glycoprotein from Schistosoma mansoni eggs, induces IgE-dependent, antigen-independent IL-4 production by murine basophils in vivo. The Journal of Immunology 178, 60236027.CrossRefGoogle ScholarPubMed
Schwartz, C and Fallon, PG (2018) Schistosoma ‘eggs-iting’ the host: granuloma formation and egg excretion. Frontiers in Immunology 9, 2492.CrossRefGoogle ScholarPubMed
Schwartz, E, Rozenman, J and Perelman, M (2000) Pulmonary manifestations of early schistosome infection among nonimmune travelers. The American Journal of Medicine 109, 718722.CrossRefGoogle ScholarPubMed
Schwartz, C, Oeser, K, Prazeres da Costa, C, Layland, LE and Voehringer, D (2014) T cell-derived IL-4/IL-13 protects mice against fatal Schistosoma mansoni infection independently of basophils. The Journal of Immunology 193, 35903599.CrossRefGoogle ScholarPubMed
Shapouri-Moghaddam, A, Mohammadian, S, Vazini, H, Taghadosi, M, Esmaeili, SA, Mardani, F, Seifi, B, Mohammadi, A, Afshari, JT and Sahebkar, A (2018) Macrophage plasticity, polarization, and function in health and disease. Journal of Cellular Physiology 233, 64256440.CrossRefGoogle ScholarPubMed
Shen, J, Wang, L, Peng, M, Liu, Z, Zhang, B, Zhou, T, Sun, X and Wu, Z (2019) Recombinant Sj16 protein with novel activity alleviates hepatic granulomatous inflammation and fibrosis induced by Schistosoma japonicum associated with M2 macrophages in a mouse model. Parasites & Vectors 12, 457.CrossRefGoogle ScholarPubMed
Shen, H, Wang, Z, Huang, A, Zhu, D, Sun, P and Duan, Y (2021) Lipocalin 2 is a regulator during macrophage polarization induced by soluble worm antigens. Frontiers in Cellular and Infection Microbiology 11, 1.CrossRefGoogle ScholarPubMed
Shi, Y, Luo, P, Wang, W, Horst, K, Bläsius, F, Relja, B, Xu, D, Hildebrand, F and Greven, J (2020) M1 but not M0 extracellular vesicles induce polarization of RAW264.7 macrophages via the TLR4-NFκB pathway in vitro. Inflammation 43, 16111619.CrossRefGoogle Scholar
Shiels, J, Cwiklinskiid, K, Alvarado, R, Thivierge, K, Cotton, S, Santana, BG, Toid, J, Donnelly, S, Taggart, CC, Weldonid, S and Dalton, JP (2020) Schistosoma mansoni immunomodulatory molecule Sm16/SPO-1/SmSLP is a member of the trematode-specific helminth defence molecules (HDMs). PLoS Neglected Tropical Diseases 14, 125.CrossRefGoogle ScholarPubMed
Sica, A and Mantovani, A (2012) Macrophage plasticity and polarization: in vivo veritas. The Journal of Clinical Investigation 122, 787795.CrossRefGoogle ScholarPubMed
Sica, A, Erreni, M, Allavena, P and Porta, C (2015) Macrophage polarization in pathology. Cellular and Molecular Life Sciences 72, 41114126.CrossRefGoogle Scholar
Smith, H, Forman, R, Mair, I and Else, KJ (2018) Interactions of helminths with macrophages: therapeutic potential for inflammatory intestinal disease. Expert Review of Gastroenterology & Hepatology 12, 9971006.CrossRefGoogle ScholarPubMed
Song, LJ, Yin, XR, Mu, SS, Li, JH, Gao, H, Zhang, Y, Dong, PP, Mei, CJ and Hua, ZC (2020) The differential and dynamic progression of hepatic inflammation and immune responses during liver fibrosis induced by Schistosoma japonicum or carbon tetrachloride in mice. Frontiers in Immunology 11, 2633.CrossRefGoogle ScholarPubMed
Sotillo, J, Pearson, M, Potriquet, J, Becker, L, Pickering, D, Mulvenna, J and Loukas, A (2016) Extracellular vesicles secreted by Schistosoma mansoni contain protein vaccine candidates. International Journal for Parasitology 46, 15.CrossRefGoogle ScholarPubMed
Starkey Lewis, P, Campana, L, Aleksieva, N, Cartwright, JA, Mackinnon, A, O'Duibhir, E, Kendall, T, Vermeren, M, Thomson, A, Gadd, V, Dwyer, B, Aird, R, Man, TY, Rossi, AG, Forrester, L, Park, BK and Forbes, SJ (2020) Alternatively activated macrophages promote resolution of necrosis following acute liver injury. Journal of Hepatology 73, 349360.CrossRefGoogle ScholarPubMed
Steinmann, P, Keiser, J, Bos, R, Tanner, M and Utzinger, J (2006) Schistosomiasis and water resources development: systematic review, meta-analysis, and estimates of people at risk. Lancet Infectious Diseases 6, 411425.CrossRefGoogle ScholarPubMed
Stingl, P and Stingl, T (2017) Schistosomiasis. MMW Fortschritte der Medizin 159, 5154.CrossRefGoogle ScholarPubMed
Suzuki, Y, Shirai, M, Asada, K, Yasui, H, Karayama, M, Hozumi, H, Furuhashi, K, Enomoto, N, Fujisawa, T, Nakamura, Y, Inui, N, Shirai, T, Hayakawa, H and Suda, T (2018) Macrophage mannose receptor, CD206, predict prognosis in patients with pulmonary tuberculosis. Scientific Reports 8, 19.CrossRefGoogle ScholarPubMed
Takaki, KK, Rinaldi, G, Berriman, M, Pagán, AJ and Ramakrishnan, L (2021) Schistosoma mansoni eggs modulate the timing of granuloma formation to promote transmission. Cell Host & Microbe 29, 58.CrossRefGoogle ScholarPubMed
Tang, Y, Shen, Y, Hong, Y, Zhang, Z, Zhai, Q, Fu, Z, Li, H, Lu, K and Lin, J (2021) miR-181a regulates the host immune response against Schistosoma japonicum infection through the TLR4 receptor pathway. Parasites & Vectors 14, 548.CrossRefGoogle ScholarPubMed
Truscott, M, Evans, DA, Gunn, M and Hoffmann, KF (2013) Schistosoma mansoni hemozoin modulates alternative activation of macrophages via specific suppression of Retnla expression and secretion. Infection and Immunity 81, 133142.CrossRefGoogle ScholarPubMed
Tundup, S, Srivastava, L, Nagy, T and Harn, D (2014) CD14 influences host immune responses and alternative activation of macrophages during Schistosoma mansoni infection. Infection and Immunity 82, 3240.CrossRefGoogle ScholarPubMed
Uribe-Querol, E and Rosales, C (2020) Phagocytosis: our current understanding of a universal biological process. Frontiers in Immunology 11, 1066.CrossRefGoogle ScholarPubMed
van Liempt, E, van Vliet, SJ, Engering, A, García Vallejo, JJ, Bank, CMC, Sanchez-Hernandez, M, van Kooyk, Y and van Die, I (2007) Schistosoma mansoni soluble egg antigens are internalized by human dendritic cells through multiple C-type lectins and suppress TLR-induced dendritic cell activation. Molecular Immunology 44, 26052615.CrossRefGoogle ScholarPubMed
Vannella, KM and Wynn, TA (2017) Mechanisms of organ injury and repair by macrophages. Annual Review of Physiology 79, 593617.CrossRefGoogle ScholarPubMed
Vannella, KM, Barron, L, Borthwick, LA, Kindrachuk, KN, Narasimhan, PB, Hart, KM, Thompson, RW, White, S, Cheever, AW, Ramalingam, TR and Wynn, T (2014) Incomplete deletion of IL-4Rα by LysMCre reveals distinct subsets of M2 macrophages controlling inflammation and fibrosis in chronic schistosomiasis. PLoS Pathogens 10, e1004372.CrossRefGoogle ScholarPubMed
Verjee, MA (2019) Schistosomiasis: still a cause of significant morbidity and mortality. Research and Reports in Tropical Medicine 10, 153163.CrossRefGoogle Scholar
Viola, A, Munari, F, Sánchez-Rodríguez, R, Scolaro, T and Castegna, A (2019) The metabolic signature of macrophage responses. Frontiers in Immunology 10, 1462.CrossRefGoogle ScholarPubMed
Wang, L, Yang, Z, Li, Y, Yu, F, Brindley, PJ, McManus, DP, Wei, D, Han, Z, Feng, Z, Li, Y and Hu, W (2006) Reconstruction and in silico analysis of the MAPK signaling pathways in the human blood fluke, Schistosoma japonicum. FEBS Letters 580, 36773686.CrossRefGoogle ScholarPubMed
Wang, M, Altinoglu, S, Takeda, YS and Xu, Q (2015) Integrating protein engineering and bioorthogonal click conjugation for extracellular vesicle modulation and intracellular delivery. PLoS ONE 10, e0141860.CrossRefGoogle ScholarPubMed
Wang, S, Ye, Q, Zeng, X and Qiao, S (2019) Functions of macrophages in the maintenance of intestinal homeostasis. Journal of Immunology Research 2019, 1512969.CrossRefGoogle ScholarPubMed
Wang, L, Shang, X, Qi, X, Ba, D, Lv, J, Zhou, X, Wang, H, Shaxika, N, Wang, J and Ma, X (2020) Clinical significance of M1/M2 macrophages and related cytokines in patients with spinal tuberculosis. Disease Markers 2020, 2509454.CrossRefGoogle ScholarPubMed
Wei, Y, Huang, N, Chen, S, Chen, D, Li, X, Xu, J and Yang, Z (2018) The diagnosis and treatment introspection of the first imported case of atypical cerebral schistosomiasis in Guangzhou city. PLoS Neglected Tropical Diseases 12, e0006171.CrossRefGoogle ScholarPubMed
West, HC and Bennett, CL (2018) Redefining the role of Langerhans cells as immune regulators within the skin. Frontiers in Immunology 8, 1941.CrossRefGoogle ScholarPubMed
Wheater, PR and Wilson, RA (1979) Schistosoma mansoni: a histological study of migration in the laboratory mouse. Parasitology 79, 4962.CrossRefGoogle ScholarPubMed
Whitfield, PJ, Bartlett, A, Brown, MB and Marriott, C (2003) Invasion by schistosome cercariae: studies with human skin explants. Trends in Parasitology 19, 339340.CrossRefGoogle ScholarPubMed
Wilson, RA (1987) Cercariae to liver worms: development and migration in the mammalian host. In D Rollinson, AJG Simpson (eds), The Biology of Schistosomes. New York: From Genes to Latrines, pp. 115146.Google Scholar
Wilson, RA (1998) Interferon gamma is a key cytokine in lung phase immunity to schistosomes but what is its precise role? Brazilian Journal of Medical and Biological Research 31, 157161.CrossRefGoogle ScholarPubMed
Wilson, RA and Jones, MK (2021) Fifty years of the schistosome tegument: discoveries, controversies, and outstanding questions. International Journal for Parasitology 51, 12131232.CrossRefGoogle ScholarPubMed
Wilson, MS, Mentink-Kane, MM, Pesce, JT, Ramalingam, TR, Thompson, R and Wynn, TA (2007) Immunopathology of schistosomiasis. Immunology and Cell Biology 85, 148.CrossRefGoogle ScholarPubMed
Wolde, M, Laan, LC, Medhin, G, Gadissa, E, Berhe, N and Tsegaye, A (2020) Human monocytes/macrophage inflammatory cytokine changes following in vivo and in vitro Schistomam mansoni infection. Journal of Inflammation Research 13, 3543.CrossRefGoogle Scholar
World Health Organization (2020) Status of Schistosomiasis Endemic Countries: 2018. Available at https://apps.who.int/neglected_diseases/ntddata/sch/sch.html.Google Scholar
Xu, J, Zhang, H, Chen, L, Zhang, D, Ji, M, Wu, H and Wu, G (2014) Schistosoma japonicum infection induces macrophage polarization. Journal of Biomedical Research 28, 299308.Google ScholarPubMed
Xu, ZJ, Gu, Y, Wang, CZ, Jin, Y, Wen, XM, Ma, JC, Tang, LJ, Mao, ZW, Qian, J and Lin, J (2019) The M2 macrophage marker CD206: a novel prognostic indicator for acute myeloid leukemia. Oncoimmunology 9, 1683347.CrossRefGoogle ScholarPubMed
Xue, L, Gyles, SL, Wettey, FR, Gazi, L, Townsend, E, Hunter, MG and Pettipher, R (2005) Prostaglandin D2 causes preferential induction of proinflammatory Th2 cytokine production through an action on chemoattractant receptor-like molecule expressed on Th2 cells. The Journal of Immunology 175, 65316536.CrossRefGoogle ScholarPubMed
Yang, Z and Ming, XF (2014) Functions of arginase isoforms in macrophage inflammatory responses: impact on cardiovascular diseases and metabolic disorders. Frontiers in Immunology 5, 533.CrossRefGoogle ScholarPubMed
Yang, SH, Sharrocks, AD and Whitmarsh, AJ (2003) Transcriptional regulation by the MAP kinase signaling cascades. Gene 320, 321.CrossRefGoogle ScholarPubMed
Yao, Y, Xu, XH and Jin, L (2019) Macrophage polarization in physiological and pathological pregnancy. Frontiers in Immunology 10, 792.CrossRefGoogle ScholarPubMed
Ye, Z, Huang, S, Zhang, Y, Mei, X, Zheng, H, Li, M, Chen, J and Lu, F (2020) Galectins, eosinophiles, and macrophages may contribute to Schistosoma japonicum egg-induced immunopathology in a mouse model. Frontiers in Immunology 11, 146.CrossRefGoogle ScholarPubMed
Yin, Z, Ma, T, Lin, Y, Lu, X, Zhang, C, Chen, S and Jian, Z (2018) IL-6/STAT3 pathway intermediates M1/M2 macrophage polarization during the development of hepatocellular carcinoma. Journal of Cellular Biochemistry 119, 94199432.CrossRefGoogle ScholarPubMed
Yu, Y, Wang, J, Wang, X, Gu, P, Lei, Z, Tang, R, Wei, C, Xu, L, Wang, C, Chen, Y, Pu, Y, Qi, X, Yu, B, Chen, X, Zhu, J, Li, Y, Zhang, Z, Zhou, S and Su, C (2021) Schistosome eggs stimulate reactive oxygen species production to enhance M2 macrophage differentiation and promote hepatic pathology in schistosomiasis. PLoS Neglected Tropical Diseases 15, e0009696.CrossRefGoogle ScholarPubMed
Yunna, C, Mengru, H, Lei, W and Weidong, C (2020) Macrophage M1/M2 polarization. European Journal of Pharmacology 877, 173090.CrossRefGoogle ScholarPubMed
Zhao, P, Elks, CM and Stephens, JM (2014) The induction of lipocalin-2 protein expression in vivo and in vitro. The Journal of Biological Chemistry 289, 59605969.CrossRefGoogle Scholar
Zheng, S, Zhang, P, Chen, Y, Zheng, S, Zheng, L and Weng, Z (2016) Inhibition of notch signaling attenuates schistosomiasis hepatic fibrosis via blocking macrophage M2 polarization. PLoS ONE 11, e0166808.CrossRefGoogle ScholarPubMed
Zheng, B, Zhang, J, Chen, H, Nie, H, Miller, H, Gong, Q and Liu, C (2020) T lymphocyte-mediated liver immunopathology of schistosomiasis. Frontiers in Immunology 11, 61.CrossRefGoogle ScholarPubMed
Zhu, J, Xu, Z, Chen, X, Zhou, S, Zhang, W, Chi, Y, Li, W, Song, X, Liu, F and Su, C (2014) Parasitic antigens alter macrophage polarization during Schistosoma japonicum infection in mice. Parasites & Vectors 7, 19.CrossRefGoogle ScholarPubMed
Zhu, S, Wang, S, Lin, Y, Jiang, P, Cui, X, Wang, X, Zhang, Y and Pan, W (2016) Release of extracellular vesicles containing small RNAs from the eggs of Schistosoma japonicum. Parasites & Vectors 9, 19.CrossRefGoogle ScholarPubMed
Zussman, RA, Bauman, PM and Petruska, JC (1970) The role of ingested hemoglobin in the nutrition of Schistosoma mansoni. The Journal of Parasitology 56, 7579.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. Life cycle of Schistosoma mansoni and Schistosoma japonicum. (1) The eggs shed in the feces of the definitive host release the miracidia when they come in contact with water (2), which penetrate in soft tissue the intermediate host snail (Biomphalaria spp./Oncomelania spp.). Inside the snail, the miracidia transform into mother sporocysts, which in turn produce daughter sporocysts by asexual reproduction. After around 30 days post-infection, cercariae emerge from the daughter sporocysts and are shedding by the snails in response to the light and heat (4). The cercariae penetrate the skin of the definitive host (5) and later transform into schistosomula. These larvae enter venous blood vessels and are passively carried to the lungs and heart (6). Upon reaching the hepatic portal system, schistosomula mature, become adult worms (male or female) and mate (7). The mated worms migrate to the lower mesenteric veins of the intestine, where the female sheds the eggs. Part of these eggs pass through the intestinal wall and are eliminated in the feces, starting the cycle again. However, some eggs are not eliminated and get trapped in several organs (mainly the liver and intestines), inducing a potent granulomatous inflammatory response, responsible for schistosomiasis pathology. Source: Created with BioRender.com.

Figure 1

Fig. 2. Different immune response profiles during S. mansoni and S. japonicum infection. Source: Created with BioRender.com.

Figure 2

Fig. 3. Different macrophage phenotypes, specific stimuli and markers. Source: Created with BioRender.com.

Figure 3

Table 1. Molecules and/or antigens involved in macrophage polarization in Schistosoma mansoni infection

Figure 4

Table 2. Molecules and/or antigens involved in macrophage polarization in Schistosoma japonicum infection