Acessibilidade / Reportar erro

CCR6 expression reduces mouse survival upon malarial challenge with Plasmodium berghei NK65 strain

Abstract

BACKGROUND

It has been demonstrated that proteins expressed by liver-stage Plasmodium parasites can inhibit the translocation of transcription factors to the nucleus of different cells. This process would hinder the expression of immune genes, such as the CCL20 chemokine.

OBJECTIVE

Since CCR6 is the only cognate receptor for CCL20, we investigated the importance of this chemokine-receptor axis against rodent malaria.

METHODS

CCR6-deficient (KO) and wild-type (WT) C57BL/6 mice were challenged with Plasmodium berghei (Pb) NK65 sporozoites or infected red blood cells (iRBCs). Liver parasitic cDNA, parasitemia and serum cytokine concentrations were respectively evaluated through reverse transcription-polymerase chain reaction (RT-PCR), staining thin-blood smears with Giemsa solution, and enzyme-linked immunosorbent assay (ELISA).

FINDINGS

Although the sporozoite challenges yielded similar liver parasitic cDNA and parasitemia, KO mice presented a prolonged survival than WT mice. After iRBC challenges, KO mice kept displaying higher survival rates as well as a decreased IL-12 p70 concentration in the serum than WT mice.

CONCLUSION

Our data suggest that malaria triggered by PbNK65 liver- or blood-stage forms elicit a pro-inflammatory environment that culminates with a decreased survival of infected C57BL/6 mice.

Key words:
malaria; sporozoites; iRBCs; chemokine receptor; Th1 cytokines


Malaria is a parasitic disease caused by multiple Plasmodium species that show distinct distributions worldwide and represents one of the largest global public health issues. In 2019, nearly 229 million cases and 409 thousand malaria-derived deaths were reported.11. WHO - World Health Organization. World malaria report 2020. 2020. Available from: https://www.who.int/publications/i/item/9789240015791.
https://www.who.int/publications/i/item/...
Despite the efficiency of the current therapies, these high numbers of cases and mortality have been associated with the selection of drug-resistant parasites.22. Uwimana A, Legrand E, Stokes BH, Ndikumana J-LM, Warsame M, Umulisa N, et al. Author correction: emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat Med. 2021; 27: 1113-15. Since the first malaria vaccine has been recently approved,33. Maxmen A. Scientists hail historic malaria vaccine approval - but point to challenges ahead. Nature [Internet]. 2021 Oct 8 [cited 2021 Oct 15]. Available from: https://www.nature.com/articles/d41586-021-02755-5.
https://www.nature.com/articles/d41586-0...
there is a belief that a wide vaccination coverage may control this massive epidemiology impact in a midterm period.

Plasmodium infective forms are mosquito-transmitted sporozoites that majorly express the circumsporozoite protein (CSP) on their surface. After invading the host skin, these parasites migrate through the circulation until reaching the liver. Inside hepatocytes, they proliferate and differentiate into exo-erythrocytic forms (EEFs), finalizing the pre-erythrocytic stage of the infection. Of note, the CSP presence in the hepatocyte cytoplasm inhibits the functionality of the transcriptional factor NF-κB, reducing the gene expression related to immunity. Indeed, the transcriptome data from EEF-infected HepG2 cell cultures suggest that the CSP down-regulates in almost 65-fold the expression of the CCL20 (also named MIP-3α) chemokine gene.44. Singh AP, Buscaglia CA, Wang Q, Levay A, Nussenzweig DR, Walker JR, et al. Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell. 2007; 131(3): 492-504.

The only receptor known as capable of binding CCL20 is CCR6.55. Wasilko DJ, Johnson ZL, Ammirati M, Che Y, Griffor MC, Han S, et al. Structural basis for chemokine receptor CCR6 activation by the endogenous protein ligand CCL20. Nat Commun. 2020; 11(1): 3031. The blood RNA consensus dataset from the human protein atlas (https://www.proteinatlas.org/ENSG00000112486-CCR6/blood) indicates that CCR6-expressing cells are mainly composed of T cells (regulatory, MAIT, and memory), B cells (naive and memory), NK cells, and dendritic cells (myeloid and plasmacytoid). Also, a DNA vaccination study performed with a CSP sequence fused with the CCL20 counterpart provided protection against a malarial challenge in mice66. Luo K, Zhang H, Zavala F, Biragyn A, Espinosa DA, Markham RB. Fusion of antigen to a dendritic cell targeting chemokine combined with adjuvant yields a malaria DNA vaccine with enhanced protective capabilities. PLoS One. 2014; 9(3): e90413. and infant macaques.77. Luo K, Gordy JT, Zavala F, Markham RB. A chemokine-fusion vaccine targeting immature dendritic cells elicits elevated antibody responses to malaria sporozoites in infant macaques. Sci Rep. 2021; 11(1): 1220. Considering the importance of all these cell CCR6+ subsets in the malaria immunity88. Waki S, Uehara S, Kanbe K, Ono K, Suzuki M, Nariuchi H. The role of T cells in pathogenesis and protective immunity to murine malaria. Immunology. 1992; 75(4): 646-51.,99. Niikura M, Kamiya S, Kita K, Kobayashi F. Coinfection with nonlethal murine malaria parasites suppresses pathogenesis caused by Plasmodium berghei NK65. J Immunol. 2008; 180(10): 6877-84.,1010. Silveira ELV, Dominguez MR, Soares IS. To B or not to B: understanding B cell responses in the development of malaria infection. Front Immunol. 2018; 9: 2961.,1111. Galvão-Filho B, de Castro JT, Figueiredo MM, Rosmaninho CG, Antonelli LRDV, Gazzinelli RT. The emergence of pathogenic TNF/iNOS producing dendritic cells (Tip-DCs) in a malaria model of acute respiratory distress syndrome (ARDS) is dependent on CCR4. Mucosal Immunol. 2019; 12(2): 312-22. and the early lethality presented by C57BL/6 wild-type (WT) mice challenged with Plasmodium berghei NK65 (PbNK65),99. Niikura M, Kamiya S, Kita K, Kobayashi F. Coinfection with nonlethal murine malaria parasites suppresses pathogenesis caused by Plasmodium berghei NK65. J Immunol. 2008; 180(10): 6877-84.,1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.,1414. Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98.,1515. Findlay EG, Greig R, Stumhofer JS, Hafalla JCR, de Souza JB, Saris CJ, et al. Essential role for IL-27 receptor signaling in prevention of Th1-mediated immunopathology during malaria infection. J Immunol. 2010; 185(4): 2482-92.,1616. Ishida H, Imai T, Suzue K, Hirai M, Taniguchi T, Yoshimura A, et al. IL-23 protection against Plasmodium berghei infection in mice is partially dependent on IL-17 from macrophages. Eur J Immunol. 2013; 43(10): 2696-706.,1717. Oliveira-Lima OC, Bernardes D, Xavier-Pinto MC, Esteves-Arantes RM, Carvalho-Tavares J. Mice lacking inducible nitric oxide synthase develop exacerbated hepatic inflammatory responses induced by Plasmodium berghei NK65 infection. Microbes Infect. 2013; 15(13): 903-10.,1818. Vieira-Santos F, Leal-Silva T, Padrão LLS, Ruas ACL, Nogueira DS, Kraemer L, et al. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J. 2021; 20(1): 296. we hypothesized that CCR6 KO mice could be even lesser resistant than WT mice to this parasite.

MATERIALS AND METHODS

Mice - Eight to ten-week-old female C57BL/6 mice were purchased from Taconic Farms Inc (USA). CCR6-deficient (CCR6 KO) mice from the same genetic background as C57BL/6 mice were kindly provided by Dr. Sergio Lira (Icahn School of Medicine at Mount Sinai, New York, USA). All animals were kept under specific pathogen-free conditions at the New York University LARC animal facility. The experiments were performed in accordance with the guidelines approved by the ethics committee of the New York University.

Parasitic infection - Mice were infected with either sporozoites or blood-stage PbNK65 strain parasites derived from the Department of Parasitology at the New York University (USA).1919. Vandermosten L, Pham TT, Possemiers H, Knoops S, Van Herck E, Deckers J, et al. Experimental malaria-associated acute respiratory distress syndrome is dependent on the parasite-host combination and coincides with normocyte invasion. Malar J. 2018; 17: 102.,2020. Akkaya M, Bansal A, Sheehan PW, Pena M, Molina-Cruz A, Orchard LM, et al. A single-nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor alters the development of host immunity. Sci Adv. 2020; 6(6): eaaw6957.,2121. Akkaya M, Bansal A, Sheehan PW, Pena M, Cimperman CK, Qi CF, et al. Testing the impact of a single nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor on experimental cerebral malaria in mice. Sci Rep. 2020; 10(1): 13630. For the liver-stage infections, sporozoite challenges were performed through two distinct methods: (a) intravenous (iv) injection of parasites obtained from salivary glands of infected Anopheles stephensi mosquitoes; (b) natural infection by mosquito bites. Eighteen days before the mouse infection, reared mosquitoes were infected with Plasmodium berghei NK65 upon a C57BL/6 mouse blood meal in the insectary from the Department of Parasitology at the New York University, USA) as previously described.2222. Vanderberg J. The transmission by mosquitoes of Plasmodia in the laboratory. In: Kreier J, editor. Malaria: pathology, vector studies and culture. Academic Press; 1980. p. 154-218. On day 0, mice were anesthetized with ketamine and xylazine and exposed to the bites of PbNK65 strain-infected mosquitoes for 15 minutes. To ensure the infectivity of the mosquito bites, sporozoites harvested from the salivary glands of other mosquitoes from the same buckets were quantified before the challenge. Blood-stage challenges were performed with infected RBCs (iRBCs) harvested from a donor PbNK65-infected C57BL/6 mouse. More specifically, donor blood samples were collected at any time from day 4 to 10 post-infection, which corresponds to ascending periods of parasitemia and eventually stored at -80ºC until usage. Once thawed, parasites could be cycling at different stages, and iRBCs were resuspended in 1X RPMI 1640 medium and washed twice with the same culture medium solution through 600 x g centrifugation at 4ºC for 5 min. To measure the iRBC percentage in the donor mouse, a total of 40 fields were analyzed from each thin blood smear stained with Giemsa solution. Then, another batch of animals was intravenously injected with either 2,000 or 20,000 iRBCs.

Enzyme-linked immunosorbent assay (ELISA) - IL-12p70 and IFN-γ serum levels were measured through ELISA kits (Thermo-fisher 88-7121 and 88-7314, respectively), kindly provided by Dr. Moriya Tsuji (The Aaron Diamond AIDS Research Center - affiliated with The Rockefeller University, New York, USA). Serum samples from naïve and infected mice were collected on days 2, 4, and 6 post iRBC challenge and diluted 1:10 for these quantifications.

Quantitative polymerase chain reaction (qPCR) - Forty-two hours following a sporozoite challenge (iv injection or exposure to infected mosquito bites), mouse livers were excised and processed for the isolation of total RNA. Four micrograms of total RNA were used in the reverse transcription as previously described.2323. Bruna-Romero O, Hafalla JCR, González-Aseguinolaza G, Sano G, Tsuji M, Zavala F. Detection of malaria liver-stages in mice infected through the bite of a single Anopheles mosquito using a highly sensitive real-time PCR. Int J Parasitol. 2001; 31(13): 1499-502. The resulting cDNA was used as a template for a quantitative real-time PCR for Pb 18S rRNA sequences combined with the Pb 18S forward (5’ - AAGCATTAAATAAAGCGAATACATCCTTAC - 3’) and reverse (5’ - GGAGATTGGTTTTGACGTTTATGTG - 3’) primers. The amplification was performed with the iCycler iQ Real-Time PCR Detection System (Bio-Rad, Hercules, CA). The quantification of Pb18S normalized gene expression was based on the cycle threshold (Ct) values as described earlier.2323. Bruna-Romero O, Hafalla JCR, González-Aseguinolaza G, Sano G, Tsuji M, Zavala F. Detection of malaria liver-stages in mice infected through the bite of a single Anopheles mosquito using a highly sensitive real-time PCR. Int J Parasitol. 2001; 31(13): 1499-502. A double-stranded-DNA-specific iQ SYBR Green supermix (Biorad Laboratories) was used to detect the PCR products. As a positive control, a defined number of copies of a Pb18S rRNA plasmid standard was utilized.

Statistical analysis - Survival curves (Kaplan-Meier) were assessed using the log-rank Mantel-Cox test (Prisma Software). Two-independent sample student’s t-test (http://faculty.vassar.edu/lowry/VassarStats.htm) was used to compare the cytokine production between WT and CCR6 KO mice. A p-value < 0.05 indicated significant differences between the mouse strains.

RESULTS

CCR6 expression diminishes malaria survival in PbNK65 sporozoite-infected mice - To address the impact of the CCR6 expression in the immunity against PbNK65 malaria, our first approach was to challenge WT and CCR6 KO mice with different sporozoite numbers via iv injection. Thus, mice were infected with lethal inoculums ranging from 250 to 250,000 live parasites and parasitemia levels and survival were measured. In all sporozoite challenges, we found similar parasitemia levels among these mouse strains [Fig. 1A-B, Supplementary data (Fig. 1A-B)]. Surprisingly, CCR6 KO mice presented a significant delay in the mortality relative to WT mice when challenged with the smaller parasitic inoculums (250 sporozoites - p = 0.0015; 2,500 sporozoites - p = 0.0027) (Fig. 1C-D). However, this survival pattern observed in CCR6 KO mice disappeared with the challenges with 25,000 and 250,000 sporozoites (p = 0.0644 and p = 0.3173, respectively) [Supplementary data (Fig. 1C-D)].

Fig. 1:
CCR6 KO mice displayed an extended survival upon a Plasmodium berghei NK65 sporozoite challenge in comparison to wild-type (WT) mice. Mice were infected via intravenous injection with an inoculum size similar to the parasite amount naturally transmitted by mosquitoes ((A) 250 or (B) 2,500 PbNK65 strain live sporozoites). A-B) Parasitemia levels (mean ± standard deviation) were quantified from three days after the challenge onward in Giemsa stained-blood smears slides. † Number of mice found dead. C-D) Kaplan-Meier curves represent the daily survival of challenged mice (C) 250 or (D) 2,500 PbNK65 strain live sporozoites). Log-rank test was performed on this analysis. Circles - WT mice; Squares - CCR6 KO mice. This experiment was performed only once.

Another approach to evaluate the resistance of these animals against liver-stage PbNK65 parasites was to expose them to the bites of PbNK65-infected mosquitoes. Thus, a total of three, seven, or 15 infectious mosquitoes were allowed to bite WT and CCR6 KO mice for 15 min. To estimate how many sporozoites those mosquitoes had in their salivary glands, we determined the parasitic loads derived from the salivary glands of other mosquitoes not used in the challenges, but derived from the same buckets. They ranged from 5,333 to 14,000 sporozoites per mosquito. Regardless of the mosquito numbers used in this challenge, parasitemia levels remained equivalent among these mouse strains [Fig. 2A, Supplementary data (Fig. 2A-B)]. Similar to the previous sporozoite challenge via iv injection, CCR6 KO mice still displayed a prolonged survival when exposed to the bites of three infectious mosquitoes (p = 0.0246) (Fig. 2B). In contrast, the sporozoite challenges through the biting of seven or 15 infectious mosquitoes induced similar survival rates between WT and CCR6 mice (p = 0.0849 and p = 0.0639, respectively) [Supplementary data (Fig. 2C-D)].

To ensure that the higher survival pattern found in CCR6 KO mice was related to the immunity against the malaria liver stage, we quantified the liver parasitic loads in the challenged mice through qPCR. After the exposure to either 250 live sporozoites via the intravenous route (A) or the biting of three infected mosquitoes (B), equivalent liver parasitic loads were detected in both mouse strains (p > 0.05 - Fig. 3A-B). Therefore, our data indicate that the global murine CCR6 expression is associated with a decreased malarial survival elicited by PbNK65 liver-stage parasites.

Fig. 2:
CCR6 KO mice displayed an extended survival against malaria transmitted by the bites of Plasmodium berghei NK65-infected mosquitoes relative to wild-type (WT) mice. Each mouse was anesthetized and exposed to the bites of three infected mosquitoes for 15 minutes. About 5,333 sporozoites were found in the salivary glands of other mosquitoes not used in the challenge but from the same bucket. (A) Parasitemia levels (mean ± standard deviation) were quantified from five days after the challenge onward in Giemsa stained-blood smears slides. (B) Kaplan-Meier curves represent the daily survival of challenged mice. Log-rank test was performed on this analysis. Circles - WT mice; Squares - CCR6 KO mice. This experiment was performed only once.

Fig. 3:
extended survival of CCR6 KO mice against Plasmodium berghei malaria induced by a live sporozoite injection or the exposure to infected-mosquito bites is independent of the pre-erythrocytic stage of the infection. Mice were infected via intravenous injection with 250 live PbNK65 sporozoites (A) or exposed to the bites of three infected mosquitoes (B). Forty-two hours after the challenge, mice were euthanized and livers were harvested for RNA extraction. To estimate the parasitic loads in the liver, the ensuing cDNA was analyzed for the normalized expression of Pb rRNA 18S through quantitative real-time PCR. GAPDH gene expression was utilized as a positive control. Data represent the mean ± standard deviation. Blue columns - wild-type (WT) mice; Red columns - CCR6 KO mice. This experiment was performed only once.

CCR6 expression also decreases malaria survival in mice infected with PbNK65 blood-stage parasites - Although liver parasite loads and parasitemia levels were undistinguishable between WT and CCR6 mice after sporozoite challenges, the latter animals still displayed enhanced survival against PbNK65 malaria (Figs 1-3). Thus, we investigated whether the CCR6 expression could disturb specifically the immunity against blood-stage parasites. For this, these mouse strains were intravenously (iv) infected with 2,000 or 20,000 PbNK65-infected red blood cells (iRBCs). In these challenges, both mouse strains showed similar parasitemia levels (Fig. 4A-B). Similar to the exposure to liver-stage parasites, CCR6 KO mice remained presenting a prolonged survival relative to WT mice, regardless the amount of iRBCs used (2,000 iRBCs - p = 0.0089; 20,000 iRBCs - p = 0.0007) (Fig. 4C-D). In conclusion, our data suggest that the global CCR6 expression can be detrimental to the murine survival against PbNK65 blood-stage parasites.

Fig. 4:
CCR6 KO mice presented a longer survival upon malaria induced by the intravenous injection of Plasmodium berghei NK65-infected red blood cells than wild-type (WT) mice. Mice were challenged with 2,000 (A) or 20,000 infected RBCs. (A-B) Parasitemia levels (mean ± standard deviation) were quantified from three days after the challenge onward in Giemsa stained-blood smears slides. † Number of mice found dead. (C-D) Kaplan-Meier curves represent the daily survival of challenged mice. Log-rank tests were performed to these analyses. (C) 2,000 or (D) 20,000 iRBCs). Circles - WT mice; Squares - CCR6 KO mice. Two independent experiments were performed for the 2,000 iRBC challenge, while the 20,000 iRBC challenge was run only once.

CCR6 KO mice have a reduced IL-12 p70 secretion in the serum upon the PbNK65-iRBC challenge in comparison to WT mice - Pro-inflammatory cytokines, such as IL-12 p70 and IFN-γ, have been described as harmful factors for the malaria resistance induced by PbNK65-iRBCs in mice. Indeed, treatments with anti-IL-12 or anti-IFN-γ monoclonal antibodies strengthen the malaria survival as much as observed in PbNK65-iRBC challenged IL-12 KO or IFN-γ KO mice.1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34. Based on our previous data (Figs 1-4), we hypothesized that the enhanced malaria survival detected in CCR6 KO mice could be related to lower secretion levels of pro-inflammatory cytokines after the parasite exposure. Hence, IFN-γ and IL-12 p70 levels were quantified in serum samples of these mouse strains pre and post-challenge with PbNK65-iRBCs (days 2, 4, and 6) by ELISA. A comparable IFN-γ production was observed between WT and CCR6 KO mice throughout the time points evaluated (Fig. 5A). In contrast, the IL-12p70 production was significantly diminished to an undetectable level in CCR6 KO mice 6 days after infection when compared to WT mice (p < 0.05 - Fig. 5B). Therefore, our data propose that the prolonged survival of CCR6 KO mice after PbNK65 malaria is associated with a decreased serum IL-12 p70 production.

Fig. 5:
CCR6 KO mice secreted a lower production of IL-12 p70 after a malarial challenge in the serum than wild-type (WT) mice. Sera samples derived from infected WT (n = 7) and CCR6 KO (n = 9 for all time points, except day 6 that had n = 6) mice were collected at various time points for the measurement of IFN-γ (A) and IL-12 p70 (B) concentrations through enzyme-linked immunosorbent assay (ELISA). Naïve WT and CCR6 KO mice were utilized as negative controls (n = 6). Data represent the mean ± standard error of mean. Cytokine concentrations were compared through a two-independent sample student’s t-test in all-time points between both mouse strains. Blue columns - WT mice; Red columns - CCR6 KO mice. Dashed line - limit of detection; *p < 0.05. This experiment was performed only once.

DISCUSSION

In this study, we confirmed hallmarking aspects of the PbNK65-iRBC infection in C57BL/6 mice, such as high parasitemia and early mortality.1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34. In this model, the malarial pathogenesis seems to be more severe when pro-inflammatory cytokines, such as IL-12 and IFN-γ, are produced.1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.,1414. Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98. Of note, the PbNK65 virulence was recently associated with a single polymorphism identified in the DNA binding domain of the parasitic APiAP2 transcription factor. The presence of a nucleotide (cytosine) at the position 5468 of the APiAP2 sequence would trigger a lethal iRBC infection with high parasitemia,2121. Akkaya M, Bansal A, Sheehan PW, Pena M, Cimperman CK, Qi CF, et al. Testing the impact of a single nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor on experimental cerebral malaria in mice. Sci Rep. 2020; 10(1): 13630. as observed in our data (Fig. 4A-B). Some PbNK65 lines can also induce malaria-associated acute respiratory distress syndrome (MA-ARDS) in C57BL/6 mice.2424. Van den Steen PE, Geurts N, Deroost K, Van Aelst I, Verhenne S, Heremans H, et al. Immunopathology and dexamethasone therapy in a new model for malaria-associated acute respiratory distress syndrome. Am J Respir Crit Care Med. 2010; 181(9): 957-68. However, our iRBC stock is composed of the New York line that can accumulate in the lungs and fat tissues without developing MA-ARDS in these animals.2525. Vandermosten L, Pham TT, Knoops S, De Geest C, Lays N, Van der Molen K, et al. Adrenal hormones mediate disease tolerance in malaria. Nat Commun. 2018; 9(2018).

In disagreement with our hypothesis that the global recruitment of CCR6-expressing cells was critical for the immunity against the malarial liver-stage, some in vitro approaches suggested that the sporozoite infection can activate, at least, a transient inflammatory response in hepatocyte cell lineages (Hepa 1-6, and HepG2 cells)2626. Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng X, Kappe SHI, et al. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics. 2009; 10(1): 270. and primary cells.2727. Torgler R, Bongfen SE, Romero JC, Tardivel A, Thome M, Corradin G. Sporozoite-mediated hepatocyte wounding limits plasmodium parasite development via MyD88-mediated NF-?B activation and inducible NO synthase expression. J Immunol. 2008; 180(6): 3990-99. More specifically, the CCL20 expression can be up-regulated in these hepatocytes few hours upon the sporozoite invasion.2626. Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng X, Kappe SHI, et al. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics. 2009; 10(1): 270. This process elicits the rupture of the hepatocyte membrane, releasing inflammation-mediating cytosolic factors.2727. Torgler R, Bongfen SE, Romero JC, Tardivel A, Thome M, Corradin G. Sporozoite-mediated hepatocyte wounding limits plasmodium parasite development via MyD88-mediated NF-?B activation and inducible NO synthase expression. J Immunol. 2008; 180(6): 3990-99.

In our study, it is important to highlight that the sporozoite numbers chosen for the mouse challenges followed the same potency scale usually injected by mosquitoes in the host skin.2828. Jin Y, Kebaier C, Vanderberg J. Direct microscopic quantification of dynamics of Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect Immun. 2007; 75(11): 5532-39.,2929. Wells MB, Andrew DJ. Anopheles salivary gland architecture shapes Plasmodium sporozoite availability for transmission. MBio. 2019; 10(4): e01238-19. Despite the lack of a linear correlation between the sporozoite loads and infection, mosquitoes harboring over 10,000 sporozoites in their salivary glands facilitate the malaria establishment.3030. Aleshnick M, Ganusov VV, Nasir G, Yenokyan G, Sinnis P. Experimental determination of the force of malaria infection reveals a non-linear relationship to mosquito sporozoite loads. PLoS Pathog. 2020; 16(5): e1008181. Indeed, an injection surpassing 25,000 sporozoites [Supplementary data (Fig. 1)] or the utilization of mosquitoes exceeding 10,000 sporozoites in their salivary glands [Supplementary data (Fig. 2A-B)] may have overloaded the immune system of CCR6 KO mice, hampering their ability to survive for a longer period than WT mice. The mechanisms related to the prolonged survival of CCR6 KO mice might be associated with distinct types of malarial pathogenesis. More specifically, several studies showed that animal deaths seen nearly day 25 post PbNK65-iRBC infection strongly correlate with an exacerbated liver inflammation.99. Niikura M, Kamiya S, Kita K, Kobayashi F. Coinfection with nonlethal murine malaria parasites suppresses pathogenesis caused by Plasmodium berghei NK65. J Immunol. 2008; 180(10): 6877-84.,1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.,1414. Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98. Although mouse deaths detected at day 10 remain elusive, this animal model of malaria was also described to induce inflammation of the cerebral microvasculature with cerebral edema, congestion, death of endothelial cells, parenchymal hemorrhage, proliferation of glia, accumulation of erythrocytes and leukocyte adhesion, of which can culminate with an earlier death.3131. Lacerda-Queiroz N, Lima OC, Carneiro CM, Vilela MC, Teixeira AL, Teixeira-Carvalho A, et al. Plasmodium berghei NK65 induces cerebral leukocyte recruitment in vivo: an intravital microscopic study. Acta Trop. 2011; 120(1-2): 31-9.

The infusion of a specific antibody to systemically neutralize chemokines or cytokines is an elegant approach to investigate their roles in a determined condition. To evaluate the relevance of the CCR6-CCL20 axis in the malarial immunity, we also treated WT mice with anti-CCL20 monoclonal antibodies before and during the blood-stage infection. However, the treatment did not have any effect on parasitemia and survival (data not shown). Initially, we suspected that the mAb amount used per mouse was not enough to neutralize the circulating chemokine. Some studies have demonstrated that similar mAb doses could neutralize the respective target in vivo.1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,3232. Teramoto K, Miura S, Tsuzuki Y, Hokari R, Watanabe C, Inamura T, et al. Increased lymphocyte trafficking to colonic microvessels is dependent on MAdCAM-1 and C-C chemokine mLARC/CCL20 in DSS-induced mice colitis. Clin Exp Immunol. 2005; 139(3): 421-8.,3333. Katchar K, Kelly CP, Keates S, O'Brien MJ, Keates AC. MIP-3a neutralizing monoclonal antibody protects against TNBS-induced colonic injury and inflammation in mice. Am J Physiol Gastrointest Liver Physiol. 2007; 292(5): G1263-71. Although most of the chemokine or cytokine neutralization attempts were not effective against this malaria model of infection, the use of transgenic mice confirmed the importance of some molecules in the immunity against this disease though. More specifically, IL-12 KO, iNOS KO, IL-27R KO, IL-23 (p19)-KO, IL-17A KO, Perforin KO, and MyD88 KO mice displayed a lower malarial pathogenesis than WT mice after an iRBC challenge.1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.,1515. Findlay EG, Greig R, Stumhofer JS, Hafalla JCR, de Souza JB, Saris CJ, et al. Essential role for IL-27 receptor signaling in prevention of Th1-mediated immunopathology during malaria infection. J Immunol. 2010; 185(4): 2482-92.,1616. Ishida H, Imai T, Suzue K, Hirai M, Taniguchi T, Yoshimura A, et al. IL-23 protection against Plasmodium berghei infection in mice is partially dependent on IL-17 from macrophages. Eur J Immunol. 2013; 43(10): 2696-706.,1717. Oliveira-Lima OC, Bernardes D, Xavier-Pinto MC, Esteves-Arantes RM, Carvalho-Tavares J. Mice lacking inducible nitric oxide synthase develop exacerbated hepatic inflammatory responses induced by Plasmodium berghei NK65 infection. Microbes Infect. 2013; 15(13): 903-10.

Regarding the capacity of CCR6 KO mice in producing pro-inflammatory cytokines, it has been already described that their peritoneal macrophages secrete a reduced IL-12 p70 concentration after LPS stimulation relative to the WT mouse counterparts. This issue was further associated with the higher resistance of CCR6 KO mice to cecal ligation and puncture-derived sepsis3434. Wen H, Hogaboam CM, Lukacs NW, Cook DN, Lira SA, Kunkel SL. The chemokine receptor CCR6 is an important component of the innate immune response. Eur J Immunol. 2007; 37(9): 2487-98. and Yersinia oral infection.3535. Westphal S, Lügering A, Von Wedel J, Von Eiff C, Maaser C, Spahn T, et al. Resistance of chemokine receptor 6-deficient mice to Yersinia enterocolitica infection: evidence of defective M-cell formation in vivo. Am J Pathol. 2008; 172(3): 671-80. Also, IL-12 p70 was demonstrated to activate some lymphocytes, such as NK and NKT cells (DX5-expressing cells) that can kill hepatocytes.1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.,1414. Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98. Similar numbers of NK1.1+ cells (NK or NKT cells) were enumerated in the peritoneum of naïve WT and CCR6 KO mice.3434. Wen H, Hogaboam CM, Lukacs NW, Cook DN, Lira SA, Kunkel SL. The chemokine receptor CCR6 is an important component of the innate immune response. Eur J Immunol. 2007; 37(9): 2487-98. However, malaria can significantly increase these cell subsets in the murine liver.1414. Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98. If CCR6 KO mice present a diminished frequency of these cell subsets and an ensuing milder malaria than WT mice, it needs to be confirmed.

Although the CCR6 receptor binds to CCL20 with high affinity, it also possesses a low affinity to some antimicrobial peptides known as beta-defensins.3636. Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, et al. ß-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science. 1999; 286(5439): 525-8. These molecules are usually expressed in several types of epithelia, which can be colonized during an infection.3737. Stolzenberg ED, Anderson GM, Ackermann MR, Whitlock RH, Zasloff M. Epithelial antibiotic induced in states of disease. Proc Natl Acad Sci USA. 1997; 94(16): 8686-90. Despite all uncertainty about their effective mechanisms, they represent an important innate component of the immune system against bacteria,3838. Wu M, McClellan SA, Barrett RP, Zhang Y, Hazlett LD. ß-defensins 2 and 3 together promote resistance to Pseudomonas aeruginosa keratitis. J Immunol. 2009; 183(12): 8054-60. fungi,3939. Schofield DA, Westwater C, Balish E. ß-defensin expression in immunocompetent and immunodeficient germ-free and Candida albicans-monoassociated mice. J Infect Dis. 2004; 190(7): 1327-34. viral,4040. Weinberg A, Quiñones-Mateu ME, Lederman MM. Role of human ß-defensins in HIV infection. Adv Dental Res. 2006; 19(1): 42-8. and parasite infections, such as Cryptosporidium parvum.4141. Zaalouk TK, Bajaj-Elliott M, George JT, McDonald V. Differential regulation of ß-defensin gene expression during Cryptosporidium parvum infection. Infect Immun. 2004; 72(5): 2772-9. These peptides can also attract cells, such as monocytes,4242. Wu Z, Hoover DM, Yang D, Boulègue C, Santamaria F, Oppenheim JJ, et al. Engineering disulfide bridges to dissect antimicrobial and chemotactic activities of human ß-defensin 3. Proc Natl Acad Sci USA. 2003; 100(15): 8880-85. macrophages,4343. Soruri A, Grigat J, Forssmann U, Riggert J, Zwirner J. ß-defensins chemoattract macrophages and mast cells but not lymphocytes and dendritic cells: CCR6 is not involved. Eur J Immunol. 2007; 37(9): 2474-86. dendritic cells, and T cells,3636. Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, et al. ß-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science. 1999; 286(5439): 525-8. and act as an endogenous ligand for TLR4.4444. Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A, Chertov O, et al. Toll-like receptor 4-dependent activation of dendritic cells by ß-dDefensin 2. Science. 2002; 298(5595): 1025-9. However, the chemotaxis to dendritic cells and T cells seems controversial about the CCR6 dependence.4343. Soruri A, Grigat J, Forssmann U, Riggert J, Zwirner J. ß-defensins chemoattract macrophages and mast cells but not lymphocytes and dendritic cells: CCR6 is not involved. Eur J Immunol. 2007; 37(9): 2474-86. Regarding the malarial infection, any correlation between beta-defensins and protection has never been outlined.

In conclusion - We found evidence that the CCR6 expression can influence the survival against the PbNK65 liver- and blood-stage malaria in C57Bl/6 mice. Moreover, this study reinforced the idea that the survival of these animals seems to be associated with a lower Th1 pattern of immune response upon the parasitic exposure as previously shown.1212. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.,1313. Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.

REFERENCES

  • 1
    WHO - World Health Organization. World malaria report 2020. 2020. Available from: https://www.who.int/publications/i/item/9789240015791
    » https://www.who.int/publications/i/item/9789240015791
  • 2
    Uwimana A, Legrand E, Stokes BH, Ndikumana J-LM, Warsame M, Umulisa N, et al. Author correction: emergence and clonal expansion of in vitro artemisinin-resistant Plasmodium falciparum kelch13 R561H mutant parasites in Rwanda. Nat Med. 2021; 27: 1113-15.
  • 3
    Maxmen A. Scientists hail historic malaria vaccine approval - but point to challenges ahead. Nature [Internet]. 2021 Oct 8 [cited 2021 Oct 15]. Available from: https://www.nature.com/articles/d41586-021-02755-5
    » https://www.nature.com/articles/d41586-021-02755-5
  • 4
    Singh AP, Buscaglia CA, Wang Q, Levay A, Nussenzweig DR, Walker JR, et al. Plasmodium circumsporozoite protein promotes the development of the liver stages of the parasite. Cell. 2007; 131(3): 492-504.
  • 5
    Wasilko DJ, Johnson ZL, Ammirati M, Che Y, Griffor MC, Han S, et al. Structural basis for chemokine receptor CCR6 activation by the endogenous protein ligand CCL20. Nat Commun. 2020; 11(1): 3031.
  • 6
    Luo K, Zhang H, Zavala F, Biragyn A, Espinosa DA, Markham RB. Fusion of antigen to a dendritic cell targeting chemokine combined with adjuvant yields a malaria DNA vaccine with enhanced protective capabilities. PLoS One. 2014; 9(3): e90413.
  • 7
    Luo K, Gordy JT, Zavala F, Markham RB. A chemokine-fusion vaccine targeting immature dendritic cells elicits elevated antibody responses to malaria sporozoites in infant macaques. Sci Rep. 2021; 11(1): 1220.
  • 8
    Waki S, Uehara S, Kanbe K, Ono K, Suzuki M, Nariuchi H. The role of T cells in pathogenesis and protective immunity to murine malaria. Immunology. 1992; 75(4): 646-51.
  • 9
    Niikura M, Kamiya S, Kita K, Kobayashi F. Coinfection with nonlethal murine malaria parasites suppresses pathogenesis caused by Plasmodium berghei NK65. J Immunol. 2008; 180(10): 6877-84.
  • 10
    Silveira ELV, Dominguez MR, Soares IS. To B or not to B: understanding B cell responses in the development of malaria infection. Front Immunol. 2018; 9: 2961.
  • 11
    Galvão-Filho B, de Castro JT, Figueiredo MM, Rosmaninho CG, Antonelli LRDV, Gazzinelli RT. The emergence of pathogenic TNF/iNOS producing dendritic cells (Tip-DCs) in a malaria model of acute respiratory distress syndrome (ARDS) is dependent on CCR4. Mucosal Immunol. 2019; 12(2): 312-22.
  • 12
    Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, Nariuchi H. A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol. 1998; 160(11): 5500-05.
  • 13
    Adachi K, Tsutsui H, Kashiwamura SI, Seki E, Nakano H, Takeuchi O, et al. Plasmodium berghei infection in mice induces liver injury by an IL-12- and toll-like receptor/myeloid differentiation factor 88-dependent mechanism. J Immunol. 2001; 167(10): 5928-34.
  • 14
    Adachi K, Tsutsui H, Seki E, Nakano H, Takeda K, Okumura K, et al. Contribution of CD1d-unrestricted hepatic DX5+ NKT cells to liver injury in Plasmodium berghei-parasitized erythrocyte-injected mice. Int Immunol. 2004; 16(6): 787-98.
  • 15
    Findlay EG, Greig R, Stumhofer JS, Hafalla JCR, de Souza JB, Saris CJ, et al. Essential role for IL-27 receptor signaling in prevention of Th1-mediated immunopathology during malaria infection. J Immunol. 2010; 185(4): 2482-92.
  • 16
    Ishida H, Imai T, Suzue K, Hirai M, Taniguchi T, Yoshimura A, et al. IL-23 protection against Plasmodium berghei infection in mice is partially dependent on IL-17 from macrophages. Eur J Immunol. 2013; 43(10): 2696-706.
  • 17
    Oliveira-Lima OC, Bernardes D, Xavier-Pinto MC, Esteves-Arantes RM, Carvalho-Tavares J. Mice lacking inducible nitric oxide synthase develop exacerbated hepatic inflammatory responses induced by Plasmodium berghei NK65 infection. Microbes Infect. 2013; 15(13): 903-10.
  • 18
    Vieira-Santos F, Leal-Silva T, Padrão LLS, Ruas ACL, Nogueira DS, Kraemer L, et al. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J. 2021; 20(1): 296.
  • 19
    Vandermosten L, Pham TT, Possemiers H, Knoops S, Van Herck E, Deckers J, et al. Experimental malaria-associated acute respiratory distress syndrome is dependent on the parasite-host combination and coincides with normocyte invasion. Malar J. 2018; 17: 102.
  • 20
    Akkaya M, Bansal A, Sheehan PW, Pena M, Molina-Cruz A, Orchard LM, et al. A single-nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor alters the development of host immunity. Sci Adv. 2020; 6(6): eaaw6957.
  • 21
    Akkaya M, Bansal A, Sheehan PW, Pena M, Cimperman CK, Qi CF, et al. Testing the impact of a single nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor on experimental cerebral malaria in mice. Sci Rep. 2020; 10(1): 13630.
  • 22
    Vanderberg J. The transmission by mosquitoes of Plasmodia in the laboratory. In: Kreier J, editor. Malaria: pathology, vector studies and culture. Academic Press; 1980. p. 154-218.
  • 23
    Bruna-Romero O, Hafalla JCR, González-Aseguinolaza G, Sano G, Tsuji M, Zavala F. Detection of malaria liver-stages in mice infected through the bite of a single Anopheles mosquito using a highly sensitive real-time PCR. Int J Parasitol. 2001; 31(13): 1499-502.
  • 24
    Van den Steen PE, Geurts N, Deroost K, Van Aelst I, Verhenne S, Heremans H, et al. Immunopathology and dexamethasone therapy in a new model for malaria-associated acute respiratory distress syndrome. Am J Respir Crit Care Med. 2010; 181(9): 957-68.
  • 25
    Vandermosten L, Pham TT, Knoops S, De Geest C, Lays N, Van der Molen K, et al. Adrenal hormones mediate disease tolerance in malaria. Nat Commun. 2018; 9(2018).
  • 26
    Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng X, Kappe SHI, et al. Host cell transcriptional profiling during malaria liver stage infection reveals a coordinated and sequential set of biological events. BMC Genomics. 2009; 10(1): 270.
  • 27
    Torgler R, Bongfen SE, Romero JC, Tardivel A, Thome M, Corradin G. Sporozoite-mediated hepatocyte wounding limits plasmodium parasite development via MyD88-mediated NF-?B activation and inducible NO synthase expression. J Immunol. 2008; 180(6): 3990-99.
  • 28
    Jin Y, Kebaier C, Vanderberg J. Direct microscopic quantification of dynamics of Plasmodium berghei sporozoite transmission from mosquitoes to mice. Infect Immun. 2007; 75(11): 5532-39.
  • 29
    Wells MB, Andrew DJ. Anopheles salivary gland architecture shapes Plasmodium sporozoite availability for transmission. MBio. 2019; 10(4): e01238-19.
  • 30
    Aleshnick M, Ganusov VV, Nasir G, Yenokyan G, Sinnis P. Experimental determination of the force of malaria infection reveals a non-linear relationship to mosquito sporozoite loads. PLoS Pathog. 2020; 16(5): e1008181.
  • 31
    Lacerda-Queiroz N, Lima OC, Carneiro CM, Vilela MC, Teixeira AL, Teixeira-Carvalho A, et al. Plasmodium berghei NK65 induces cerebral leukocyte recruitment in vivo: an intravital microscopic study. Acta Trop. 2011; 120(1-2): 31-9.
  • 32
    Teramoto K, Miura S, Tsuzuki Y, Hokari R, Watanabe C, Inamura T, et al. Increased lymphocyte trafficking to colonic microvessels is dependent on MAdCAM-1 and C-C chemokine mLARC/CCL20 in DSS-induced mice colitis. Clin Exp Immunol. 2005; 139(3): 421-8.
  • 33
    Katchar K, Kelly CP, Keates S, O'Brien MJ, Keates AC. MIP-3a neutralizing monoclonal antibody protects against TNBS-induced colonic injury and inflammation in mice. Am J Physiol Gastrointest Liver Physiol. 2007; 292(5): G1263-71.
  • 34
    Wen H, Hogaboam CM, Lukacs NW, Cook DN, Lira SA, Kunkel SL. The chemokine receptor CCR6 is an important component of the innate immune response. Eur J Immunol. 2007; 37(9): 2487-98.
  • 35
    Westphal S, Lügering A, Von Wedel J, Von Eiff C, Maaser C, Spahn T, et al. Resistance of chemokine receptor 6-deficient mice to Yersinia enterocolitica infection: evidence of defective M-cell formation in vivo. Am J Pathol. 2008; 172(3): 671-80.
  • 36
    Yang D, Chertov O, Bykovskaia SN, Chen Q, Buffo MJ, Shogan J, et al. ß-defensins: linking innate and adaptive immunity through dendritic and T cell CCR6. Science. 1999; 286(5439): 525-8.
  • 37
    Stolzenberg ED, Anderson GM, Ackermann MR, Whitlock RH, Zasloff M. Epithelial antibiotic induced in states of disease. Proc Natl Acad Sci USA. 1997; 94(16): 8686-90.
  • 38
    Wu M, McClellan SA, Barrett RP, Zhang Y, Hazlett LD. ß-defensins 2 and 3 together promote resistance to Pseudomonas aeruginosa keratitis. J Immunol. 2009; 183(12): 8054-60.
  • 39
    Schofield DA, Westwater C, Balish E. ß-defensin expression in immunocompetent and immunodeficient germ-free and Candida albicans-monoassociated mice. J Infect Dis. 2004; 190(7): 1327-34.
  • 40
    Weinberg A, Quiñones-Mateu ME, Lederman MM. Role of human ß-defensins in HIV infection. Adv Dental Res. 2006; 19(1): 42-8.
  • 41
    Zaalouk TK, Bajaj-Elliott M, George JT, McDonald V. Differential regulation of ß-defensin gene expression during Cryptosporidium parvum infection. Infect Immun. 2004; 72(5): 2772-9.
  • 42
    Wu Z, Hoover DM, Yang D, Boulègue C, Santamaria F, Oppenheim JJ, et al. Engineering disulfide bridges to dissect antimicrobial and chemotactic activities of human ß-defensin 3. Proc Natl Acad Sci USA. 2003; 100(15): 8880-85.
  • 43
    Soruri A, Grigat J, Forssmann U, Riggert J, Zwirner J. ß-defensins chemoattract macrophages and mast cells but not lymphocytes and dendritic cells: CCR6 is not involved. Eur J Immunol. 2007; 37(9): 2474-86.
  • 44
    Biragyn A, Ruffini PA, Leifer CA, Klyushnenkova E, Shakhov A, Chertov O, et al. Toll-like receptor 4-dependent activation of dendritic cells by ß-dDefensin 2. Science. 2002; 298(5595): 1025-9.
  • Financial support: Dr Victor Nussenzweig (NYU Langone Medical Center, New York, NY).
  • 2
    Current address: aUniversidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, Laboratório de Imunologia de células B, São Paulo, SP, Brasil; bSanaria Inc., Rockville, MD, USA; cIndeov, São Paulo, SP, Brasil.

Publication Dates

  • Publication in this collection
    17 June 2022
  • Date of issue
    2022

History

  • Received
    23 Aug 2021
  • Accepted
    26 Apr 2022
Instituto Oswaldo Cruz, Ministério da Saúde Av. Brasil, 4365 - Pavilhão Mourisco, Manguinhos, 21040-900 Rio de Janeiro RJ Brazil, Tel.: (55 21) 2562-1222, Fax: (55 21) 2562 1220 - Rio de Janeiro - RJ - Brazil
E-mail: memorias@fiocruz.br