Acessibilidade / Reportar erro

FVB/NJ strain as a mouse model for cutaneous leishmaniasis by Leishmania (L.) amazonensis

Abstract

BACKGROUND

Leishmaniases encompass a spectrum of neglected diseases caused by parasites of the genus Leishmania, grouped in two forms: tegumentary and visceral leishmaniasis.

OBJECTIVES

In this study, we propose Friend Virus B NIH Jackson (FVB/NJ) mouse strain as a new experimental model of infection with Leishmania (Leishmania) amazonensis, the second most prevalent agent of tegumentary leishmaniasis in Brazil.

METHODS AND FINDINGS

We performed in vitro infections of FVB/NJ macrophages and compared them with BALB/c macrophages, showing that BALB/c cells have higher infection percentages and a higher number of amastigotes/cell. Phagocytosis assays indicated that BALB/c and FVB/NJ macrophages have similar capacity to uptake parasites after 5 min incubations. We also investigated promastigotes’ resistance to sera from FVB/NJ and BALB/c and observed no difference between the two sera, even though FVB/NJ has a deficiency in complement components. Finally, we subcutaneously infected FVB/NJ and BALB/c mice with 2 × 106 parasites expressing luciferase. Analysis of lesion development for 12 weeks showed that FVB/NJ and BALB/c mice have similar lesion profiles and parasite burdens.

MAIN CONCLUSIONS

This work characterises for the first time the FVB/NJ mouse as a new model for tegumentary leishmaniasis caused by Leishmania (L.) amazonensis.

Key words:
FVB/NJ mouse strain; infection; Leishmania (L.) amazonensis ; tegumentary leishmaniasis


Leishmaniases constitute a spectrum of diseases caused by protozoan parasites of the genus Leishmania, transmitted by the bite of infected female phlebotomine sandflies.11. Steverding D. The history of leishmaniasis. Parasit Vectors. 2017; 10(1): 1-10. According to the World Health Organization (WHO), leishmaniases are endemic in 99 countries and territories, with more than 1 billion people at risk of infection.22. WHO - World Health Organization. Leishmaniasis. [cited 2023 Sep 4]. Available from: https://www.who.int/data/gho/data/themes/topics/gho-ntd-leishmaniasis.
https://www.who.int/data/gho/data/themes...
The estimated number of annual cases of the diseases varies from 700,000 to 1 million.22. WHO - World Health Organization. Leishmaniasis. [cited 2023 Sep 4]. Available from: https://www.who.int/data/gho/data/themes/topics/gho-ntd-leishmaniasis.
https://www.who.int/data/gho/data/themes...

The main clinical forms of leishmaniases are visceral leishmaniasis (VL) and tegumentary leishmaniasis (TL). TL is the most frequent form, and is further subdivided into localised cutaneous, mucosal/mucocutaneous, diffuse cutaneous and disseminated cutaneous.33. Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infect Dis. 2007; 7(9): 581-96.

Infection in humans is caused by more than 20 species of Leishmania, most of which belong to subgenera Leishmania and Viannia and are transmitted by over 90 species of phlebotomine sandflies.22. WHO - World Health Organization. Leishmaniasis. [cited 2023 Sep 4]. Available from: https://www.who.int/data/gho/data/themes/topics/gho-ntd-leishmaniasis.
https://www.who.int/data/gho/data/themes...
,44. Akhoundi M, Kuhls K, Cannet A, Votýpka J, Marty P, Delaunay P, et al. A historical overview of the classification, evolution, and dispersion of Leishmania parasites and sandflies. PLoS Negl Trop Dis. 2016; 10(3): e0004349. The most common vectors of Leishmania species involved in human disease are species of Phlebotomus in the Old World and Lutzomyia in the New World.55. Torres-Guerrero E, Quintanilla-Cedillo MR, Ruiz-Esmenjaud J, Arenas R. Leishmaniasis: a review. F1000Res. 2017; 6: 750.

Leishmania species have a digenetic life cycle, with an extracellular phase in the vector’s intestine and an intracellular phase in the vertebrate host.66. Coughlan S, Mulhair P, Sanders M, Schonian G, Cotton JA, Downing T. The genome of Leishmania adleri from a mammalian host highlights chromosome fission in Sauroleishmania. Sci Rep. 2017; 7(1): 43747. Transmission to mammals occurs when the female phlebotomine regurgitates metacyclic promastigotes into the skin during blood feeding.77. Doehl JSP, Bright Z, Dey S, Davies H, Magson J, Brown N, et al. Skin parasite landscape determines host infectiousness in visceral leishmaniasis. Nat Commun. 2017; 8(1): 1-10.,88. Rougeron V, de Meeûs T, Ouraga SK, Hide M, Bañuls AL. "Everything you always wanted to know about sex (but were afraid to ask)" in Leishmania after two decades of laboratory and field analyses. Rall GF, editor. PLoS Pathog. 2010; 6(8): 7-8. The promastigotes are phagocytosed by macrophages and other mononuclear phagocytic cells and differentiate into amastigotes, which survive and multiply through simple division within a phagolysosome. Macrophages usually rupture, releasing amastigotes that are phagocytosed by other cells.33. Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infect Dis. 2007; 7(9): 581-96.,99. Scott P, Novais FO. Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat Rev Immunol. 2016; 16(9): 581-92. Contamination of the vector occurs when female sandflies ingest infected phagocytes during blood feeding. In the vector’s intestine, amastigotes differentiate into procyclic promastigotes, which undergo metacyclogenesis generating metacyclic promastigotes, which migrate to the proboscis, being regurgitated during a new blood meal.88. Rougeron V, de Meeûs T, Ouraga SK, Hide M, Bañuls AL. "Everything you always wanted to know about sex (but were afraid to ask)" in Leishmania after two decades of laboratory and field analyses. Rall GF, editor. PLoS Pathog. 2010; 6(8): 7-8.,1010. Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev. 2013; 26(1): 58-85.,1111. Giraud E, Svobodova M, Muller I, Volf P, Rogers ME. Promastigote secretory gel from natural and unnatural sand fly vectors exacerbate Leishmania major and Leishmania tropica cutaneous leishmaniasis in mice. Parasitology. 2019; 146(14): 1796-802.

Depending on the host and on the Leishmania species, the parasite multiplies in epithelial macrophages or in macrophages in secondary lymphoid organs, potentially causing TL or VL, respectively.1010. Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev. 2013; 26(1): 58-85.,1212. Lauletta-Lindoso JA, Cunha MA, Queiroz I, Moreira CHV. Leishmaniasis-HIV coinfection: current challenges. HIV/AIDS - research and palliative care. 2016; 8: 147-56.

Over the past 50 years, various murine strains have been employed as models for TL and VL. These models have been used for studies about cell types, cytokines, anti-Leishmania effector mechanisms and drugs, as well as for evaluating clinical disease resolution, resistance to secondary infection, and vaccine development.1313. Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol. 2002; 2(11): 845-58.,1414. Loría-Cervera EN, Andrade-Narváez FJ. Animal models for the study of leishmaniasis immunology. Rev Inst Med Trop São Paulo. 2014; 56(1): 1-11. The pattern and severity of infection in mice depend on the Leishmania species and on the mouse strain. These models reproduce many aspects of human diseases, with a spectrum of susceptibility depending on the mouse strain.1515. Handman E. Leishmaniasis: current status of vaccine development. Clin Microbiol Rev. 2001; 14(2): 229-43. Mouse models are most used for studying TL pathogenesis owing to the high availability of cellular markers and inbred, congenic, and transgenic strains.1616. Mears ER, Modabber F, Don R, Johnson GE. A Review: The current in vivo models for the discovery and utility of new anti-leishmanial drugs targeting cutaneous leishmaniasis. PLoS Negl Trop Dis. 2015; 9(9): e0003889. Mice of different genotypes inherently display diverse susceptibility to a wide variety of Leishmania strains and species, as observed in non-healing and self-healing models.1717. de Oliveira CI, Teixeira MJ, Gomes R, Barral A, Brodskyn C. Animal models for infectious diseases caused by parasites: Leishmaniasis. Drug Discov Today Dis Models. 2004; 1(1): 81-6. Infection with L. (L.) major stands out as the most extensively investigated experimental model for TL,1313. Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol. 2002; 2(11): 845-58. in which polarised Th2 x Th1 responses are observed in BALB/c and C57BL/6 strains, respectively.1818. Heinzel FP, Sadick MD, Holaday BJ, Coffman RL, Locksley RM. Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets. J Exp Med. 1989; 169(1): 59-72. Other species, such as L. (L.) amazonensis, induce different responses according to the mouse strain, but do not display Th1 x Th2 polarization.1919. Ji J, Masterson J, Sun J, Soong L. CD4+CD25+ Regulatory T cells restrain pathogenic responses during Leishmania amazonensis infection. J Immunol. 2005; 174(11): 7147-53.,2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703.

The Friend Virus B NIH Jackson (FVB/NJ) mouse strain, derived from Swiss N:GP (NIH, 1935), is known for its sensitivity to the B strain of Friend murine leukaemia virus.2121. Taketo M, Schroeder AC, Mobraaten LE, Gunning KB, Hanten G, Fox RR, et al. FVB/N: An inbred mouse strain preferable for transgenic analyses. Proc Natl Acad Sci USA. 1991; 88(6): 2065-9. This strain is commonly used for the development of transgenic lineages due to its docile behaviour, well-defined inbred background, large litters and the development of numerous pronuclei, which facilitate microinjection of DNA.2121. Taketo M, Schroeder AC, Mobraaten LE, Gunning KB, Hanten G, Fox RR, et al. FVB/N: An inbred mouse strain preferable for transgenic analyses. Proc Natl Acad Sci USA. 1991; 88(6): 2065-9. These mice exhibit deficiency in the C5 factor of the complement system.2222. Wetsel RA, Fleischer DT, Haviland DL. Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5'-exon. J Biol Chem. 1990; 265(5): 2435-40. Indeed, the Swiss strain (SWR/J) has a deletion of two base pairs near the 5’ terminal region of the C5 gene, resulting in a truncated translation of the C5 protein, which is not functional.2222. Wetsel RA, Fleischer DT, Haviland DL. Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5'-exon. J Biol Chem. 1990; 265(5): 2435-40. The membrane attack complex (MAC) is an effector of the complement system that generates pores on the pathogen’s surface, leading to their death. MAC assembly initiates by the cleavage of C5-by-C5 convertase, generating C5a and C5b. C5a is an anaphylatoxin with pro-inflammatory and chemotactic effects, while C5b initiates MAC assembly on the pathogen’s surface. A deficiency in C5 thus results in failure in MAC assembly.2323. Bayly-Jones C, Bubeck D, Dunstone MA. The mystery behind membrane insertion: a review of the complement membrane attack complex. Philos Trans R Soc Lond B Biol Sci. 2017; 372(1726): 20160221. On the other hand, opsonisation of Leishmania by C3bi, generated after cleavage of C3b, is important for parasite binding to macrophage CR3, and consequently for phagocytosis and survival.2424. Mosser DM, Edelson PJ. The mouse macrophage receptor for C3bi (CR3) is a major mechanism in the phagocytosis of Leishmania promastigotes. J Immunol. 1985; 135(4): 2785-9.,2525. Mosser DM, Edelson PJ. The third component of complement (C3) is responsible for the intracellular survival of Leishmania major. Nature. 1987; 327(6120): 329-31. Indeed, parasites incubated with C5-deficient mouse serum were more efficiently internalised than non-opsonised parasites.2525. Mosser DM, Edelson PJ. The third component of complement (C3) is responsible for the intracellular survival of Leishmania major. Nature. 1987; 327(6120): 329-31. Parasite factors such as ISP2 (inhibitor of serine peptidase 2) were shown to inhibit the formation of C3 and C5 convertase, thus reducing the formation of MAC and the lysis of L. (L.) donovani promastigotes.2626. Verma S, Mandal A, Ansari MdY, Kumar A, Abhishek K, Ghosh AK, et al. Leishmania donovani inhibitor of serine peptidases 2 mediated inhibition of lectin pathway and upregulation of C5aR signaling promote parasite survival inside host. Front Immunol. 2018; 9: 63. It was also previously reported that C5a is crucial for the recruitment of neutrophils to draining lymph nodes following L. mexicana infection.2727. Prat-Luri B, Neal C, Passelli K, Ganga E, Amore J, Firmino-Cruz L, et al. The C5a-C5aR1 complement axis is essential for neutrophil recruitment to draining lymph nodes via high endothelial venules in cutaneous leishmaniasis. Cell Rep. 2022; 39(5): 110777. These data show the importance of the complement system in Leishmania infection.

Regarding the regulatory immune cells, it was observed that the phenotype and function of regulatory T cells (Tregs) varies among BALB/c, C57BL/6 and FVB/NJ.2828. Tanner SM, Lorenz RG. FVB/N mouse strain regulatory T cells differ in phenotype and function from the C57BL/6 and BALB/C strains. FASEB Bioadv. 2022; 4(10): 648-61. Indeed, FVB/NJ splenic Tregs secrete IL-10 at higher levels than C57BL/6 Treg cells. Besides, FVB/NJ Tregs perform an optimal suppression in a cell-cell contact, while C57BL/6 cells do not. FVB/NJ CD4+ splenocytes have lower expression of the master Treg transcription factor Foxp3 than C57BL/6 and BALB/c, which have similar levels.2828. Tanner SM, Lorenz RG. FVB/N mouse strain regulatory T cells differ in phenotype and function from the C57BL/6 and BALB/C strains. FASEB Bioadv. 2022; 4(10): 648-61.

FVB/NJ mice have been employed in studies involving cancer, neurodegenerative diseases, metabolic disorders, autoimmune conditions, and infectious diseases. Among the microorganisms and parasites analysed in this mouse strain we can cite Helicobacter pylori,2929. Mähler M, Janke C, Wagner S, Hedrich HJ. Differential susceptibility of inbred mouse strains to Helicobacter pylori infection. Scand J Gastroenterol. 2002; 37(3): 267-78. Influenza,3030. Toth LA, Verhulst SJ. Strain differences in sleep patterns of healthy and influenza-infected inbred mice. Behav Genet. 2003; 33(3): 325-36.Echinostoma Hortense,3131. Lee KJ, Park SK, Im JA, Kim SK, Kim GH, Kim GY, et al. Susceptibility of several strains of mice to Echinostoma hortense infection. Korean J Parasitol. 2004; 42(2): 51-6.Heligmosomoides polygyrus,3232. Cywinska A, Czuminska K, Schollenberger A. Granulomatous inflammation during Heligmosomoides polygyrus primary infections in FVB mice. J Helminthol. 2004; 78(1): 17-24.Staphylococcus aureus,3333. Robertson CM, Perrone EE, McConnell KW, Dunne WM, Boody B, Brahmbhatt T, et al. Neutrophil depletion causes a fatal defect in murine pulmonary Staphylococcus aureus clearance. J Surg Res. 2008; 150(2): 278-85.Nippostrongylus brasiliensis,3434. Knott ML, Hogan SP, Wang H, Matthaei KI, Dent LA. FVB/N mice are highly resistant to primary infection with Nippostrongylus brasiliensis. Parasitology. 2009; 136(1): 93-106.Litomosoides sigmodontis,3535. Specht S, Taylor MD, Hoeve MA, Allen JE, Lang R, Hoerauf A. Over expression of IL-10 by macrophages overcomes resistance to murine filariasis. Exp Parasitol. 2012; 132(1): 90-6.Trypanosoma cruzi,3636. Machado MPR, Rocha AM, de Oliveira LF, de Cuba MB, Loss IO, Castellano LR, et al. Autonomic nervous system modulation affects the inflammatory immune response in mice with acute Chagas disease. Exp Physiol. 2012; 97(11): 1186-202.Clonorchis sinensis,3737. Kim EM, Bae YM, Choi MH, Hong ST. Cyst formation, increased anti-inflammatory cytokines and expression of chemokines support for Clonorchis sinensis infection in FVB mice. Parasitol Int. 2012; 61(1): 124-9.,3838. Kim EM, Yu HS, Jin Y, Choi MH, Bae YM, Hong ST. Local immune response to primary infection and re-infection by Clonorchis sinensis in FVB mice. Parasitol Int. 2017; 66(4): 436-42.Mycobacterium ulcerans,3939. Marion E, Jarry U, Cano C, Savary C, Beauvillain C, Robbe-Saule M, et al. FVB/N mice spontaneously heal ulcerative lesions induced by Mycobacterium ulcerans and switch M. ulcerans into a low mycolactone producer. J Immunol. 2016; 196(6): 2690-8.Pneumocystis,4040. Bhagwat SP, Gigliotti F, Wang J, Wang Z, Notter RH, Murphy PS, et al. Intrinsic programming of alveolar macrophages for protective antifungal innate immunity against Pneumocystis infection. Front Immunol. 2018; 9: 2131. and Zika virus.4141. Khaiboullina SF, Lopes P, De Carvalho TG, Real ALCV, Souza DG, Costa VV, et al. Host immune response to ZIKV in an immunocompetent embryonic mouse model of intravaginal infection. Viruses. 2019; 11(6): 558. These studies indicate that FVB/NJ mice exhibit varying susceptibility depending on the pathogen used, generally triggering a strong inflammatory response. In infections with T. cruzi, diffuse inflammatory infiltration of mononuclear cells was observed in cardiac tissues, with development of acute myocarditis.3636. Machado MPR, Rocha AM, de Oliveira LF, de Cuba MB, Loss IO, Castellano LR, et al. Autonomic nervous system modulation affects the inflammatory immune response in mice with acute Chagas disease. Exp Physiol. 2012; 97(11): 1186-202. In viral infections with Influenza and Zika, FVB/NJ mice showed reduced body temperature, locomotor activity, and neuroinflammatory changes in foetuses.3030. Toth LA, Verhulst SJ. Strain differences in sleep patterns of healthy and influenza-infected inbred mice. Behav Genet. 2003; 33(3): 325-36.,4141. Khaiboullina SF, Lopes P, De Carvalho TG, Real ALCV, Souza DG, Costa VV, et al. Host immune response to ZIKV in an immunocompetent embryonic mouse model of intravaginal infection. Viruses. 2019; 11(6): 558. Upon bacterial infections, FVB/NJ mice were permissive to S. aureus after neutrophil depletion.3333. Robertson CM, Perrone EE, McConnell KW, Dunne WM, Boody B, Brahmbhatt T, et al. Neutrophil depletion causes a fatal defect in murine pulmonary Staphylococcus aureus clearance. J Surg Res. 2008; 150(2): 278-85.H. pylori and M. ulcerans triggered a strong innate immune response, without clinical signs and with spontaneous healing of lesions after the ulcerative phase, respectively.2929. Mähler M, Janke C, Wagner S, Hedrich HJ. Differential susceptibility of inbred mouse strains to Helicobacter pylori infection. Scand J Gastroenterol. 2002; 37(3): 267-78.,3939. Marion E, Jarry U, Cano C, Savary C, Beauvillain C, Robbe-Saule M, et al. FVB/N mice spontaneously heal ulcerative lesions induced by Mycobacterium ulcerans and switch M. ulcerans into a low mycolactone producer. J Immunol. 2016; 196(6): 2690-8. For M. ulcerans, there was reduced inflammation and infiltration of myeloid cells in draining lymph nodes, without involvement of humoral response in pathogen clearance and protection.3939. Marion E, Jarry U, Cano C, Savary C, Beauvillain C, Robbe-Saule M, et al. FVB/N mice spontaneously heal ulcerative lesions induced by Mycobacterium ulcerans and switch M. ulcerans into a low mycolactone producer. J Immunol. 2016; 196(6): 2690-8. Considering worm infections, FVB/NJ showed resistance to E. hortense, N. brasiliensis, and C. sinensis.3131. Lee KJ, Park SK, Im JA, Kim SK, Kim GH, Kim GY, et al. Susceptibility of several strains of mice to Echinostoma hortense infection. Korean J Parasitol. 2004; 42(2): 51-6.,3434. Knott ML, Hogan SP, Wang H, Matthaei KI, Dent LA. FVB/N mice are highly resistant to primary infection with Nippostrongylus brasiliensis. Parasitology. 2009; 136(1): 93-106.,3737. Kim EM, Bae YM, Choi MH, Hong ST. Cyst formation, increased anti-inflammatory cytokines and expression of chemokines support for Clonorchis sinensis infection in FVB mice. Parasitol Int. 2012; 61(1): 124-9.,3838. Kim EM, Yu HS, Jin Y, Choi MH, Bae YM, Hong ST. Local immune response to primary infection and re-infection by Clonorchis sinensis in FVB mice. Parasitol Int. 2017; 66(4): 436-42. On the other hand, this mouse strain is susceptible to H. polygyrus and L. sigmodontis.3232. Cywinska A, Czuminska K, Schollenberger A. Granulomatous inflammation during Heligmosomoides polygyrus primary infections in FVB mice. J Helminthol. 2004; 78(1): 17-24.,3535. Specht S, Taylor MD, Hoeve MA, Allen JE, Lang R, Hoerauf A. Over expression of IL-10 by macrophages overcomes resistance to murine filariasis. Exp Parasitol. 2012; 132(1): 90-6. Infection with the fungus Pneumocystis led to a strong innate immune response and did not trigger the activation of a protective humoral response.4040. Bhagwat SP, Gigliotti F, Wang J, Wang Z, Notter RH, Murphy PS, et al. Intrinsic programming of alveolar macrophages for protective antifungal innate immunity against Pneumocystis infection. Front Immunol. 2018; 9: 2131.

Up to the present moment, no studies have used FVB/NJ mouse for Leishmania infections. Therefore, the aim of the current study was to characterise the FVB/NJ mouse as another model for cutaneous infection by Leishmania (L.) amazonensis, one of the main causative agents of TL in Brazil. For this purpose, in vitro experiments were conducted to assess parasite phagocytosis and infection kinetics in macrophages, and in vivo infections were performed to evaluate the development of footpad lesions and parasite burden. Our data demonstrate that the FVB/NJ strain displays infection patterns similar to BALB/c mice both in vitro and in vivo. These results, along with the docile behaviour and vigorous reproductive performance, with generation of large litters (between 9 and 13 animals), render the FVB/NJ mouse a promising and suitable infection model for tegumentary leishmaniasis.

MATERIALS AND METHODS

Ethical statement - Experiments with FVB/NJ and BALB/c mice were performed according to the Brazilian College of Animal Experimentation (CONEP) guidelines and with the approval of the Institutional Animal Care and Use Committee (CEUA) of the University of São Paulo (protocol number 2314160519).

Leishmania (L.) amazonensis culture - Promastigotes of L. (L) amazonensis from the strain LV79 (MPRO/72/M1841) and M2269 (MHOM/BR/1973/M2269) expressing luciferase (La-LUC) were cultured at 24ºC in complete medium 199 (pH 7.2) (Gibco, Invitrogen) containing 40 mM HEPES (pH 7.4), 0.1 mM adenine, 0.0005% hemin, 20 μg/mL gentamicin, and 10% foetal bovine serum (FBS). For M2269 La-LUC the culture was supplemented with 32 µg/mL of geneticin (G418). The cultures were passaged weekly to a density of 2 x 106 parasites/mL and used in the stationary phase.

Bone marrow-derived macrophages (BMDM) - BALB/c and FVB/NJ mice were euthanised in a CO2 chamber. Cells were collected from the bone marrow and differentiated as previously described.4242. Peña MS, Tang FHF, Franco FAL, Rodrigues AT, Carrara GMP, Araujo TLS, et al. Leishmania (L.) amazonensis LaLRR17 increases parasite entry in macrophage by a mechanism dependent on GRP78. Parasitology. 2023; 150(10): 922-33. A total of 2 x 105 or 4 x 105 macrophages/well were plated in RPMI 1640 (pH 7.2) with 10% serum in 48- or 24-well plates on 9 mm or 13 mm glass coverslips, respectively, in a 37ºC incubator with 5% CO2 until the next day.

Peritoneal macrophages - Resident peritoneal macrophages were obtained from BALB/c and FVB/NJ animals as previously described.2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703. Cells were suspended in RPMI and transferred to 48- or 24-well plates with glass coverslips. After 2 h of incubation in a 37ºC incubator with 5% CO2, the medium was changed to RPMI supplemented with 10% FBS and 20 μg/mL gentamicin, and cells were incubated overnight at 37ºC with 5% CO2.

Infection of murine macrophages - Murine macrophages derived from BM precursors or recovered from the peritoneal cavity were plated and infected as previously described.2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703. At the end of the experiment, coverslips were fixed with methanol for 10 min, stained with the Instant Prov Kit (Newprov) and mounted on slides using Entellan (Merck). One hundred macrophages per coverslip (in triplicate) were analysed, and the percentage of infected macrophages and the mean number of amastigotes per infected macrophage were determined.

Phagocytosis of promastigotes by murine macrophages - Murine BMDM from BALB/c and FVB/NJ were plated on 13 mm coverslips in 100 mm culture dishes. Stationary-phase promastigotes were added at a multiplicity of infection (MOI) of 1:10 and plates were kept at 4ºC on ice for 2 h. Subsequently, plates were transferred to a 34ºC incubator with 5% CO2 for 5 min. Cells were thoroughly washed with sterile PBS and fixed with 2% paraformaldehyde (PFA) for 1 h. They were then quenched with 50 mM ammonium chloride for 10 min and washed with PBS for 10 min. Coverslips were blocked with 2% bovine serum albumin (BSA) in PBS for 30 min and incubated overnight with anti-Leishmania mouse serum at a dilution of 1:300 in 0.2% BSA-PBS. After washing with PBS, they were incubated for 1 h with anti-mouse IgG (H+L) 488 Alexa Fluor (1:200) and DAPI (1:1000) in sterile PBS. After washing, coverslips were mounted with ProLong (Molecular Probes) for analysis under fluorescence microscopy. Images were captured using an inverted fluorescence microscope model AxioVert.A1 (Carl Zeiss) and analysed using the ZEISS ZEN 3.7 software. A total of 100 macrophages were analysed in terms of numbers of adhered (green) and internalised parasites (blue, labelled with DAPI).

Phagocytosis of zymosan by murine macrophages - Zymosan particles (Molecular Probes) were suspended at 1 mg/mL (1x107 particles/mL) in RPMI medium pH 7.2 supplemented with 10% FBS. The zymosan phagocytosis experiments were conducted similarly to those with Leishmania. Cells were incubated at a ratio of two zymosan particles per cell for 1 h, fixed with absolute methanol for 5 min, stained with Giemsa and mounted as described in infection assays.

In vivo infection of FVB/NJ and BALB/c mice for analysis of lesion size and parasite burden through in vivo imaging - The FVB/NJ strain was kindly provided by Prof Francisco Laurindo from INCOR, School of Medicine, University of São Paulo (FM-USP). Mice from FVB/NJ (10) and BALB/c (10) strains, aged four to eight weeks, were infected with 2 × 106 promastigotes of L. (L.) amazonensis MHOM/BR/1973/M2269 expressing luciferase (La-LUC), as previously described by our group.2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703. The progression of lesions and imaging were monitored weekly for 12 weeks and analysed as described.2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703.

The comparison between the expected and observed number of infected animals in each group was calculated using the Fisher’s Exact Test. Animals that did not show infection after 12 weeks were not submitted to quantification of parasite burden.

Complement-mediated lysis assay - Blood samples from BALB/c and FVB/NJ mice were collected and incubated at 4ºC for 15 min. Tubes were centrifuged at 1500 ×g at 4ºC for 10 min, the sera were collected and pooled, and stored at -80ºC. Sera from three different pools of each mouse strain were read at 595 nm and those with the optical densities (ODs) between 0.03 and 0.04 were chosen. Lysis and MTT (5 mg/mL) assays were conducted following the published protocol,4343. Tano FT, Telleria EL, Rêgo FD, Coelho FS, de Rezende E, Soares RP, et al. Phenotypical differences between Leishmania (Leishmania) amazonensis PH8 and LV79 strains may impact survival in mammal host and in phlebotomine sand flies. Pathogens. 2023; 12(2): 173. except that parasite incubations with serum were carried out at 34ºC.

RESULTS

In vitro infection of BMDM and peritoneal macrophages from FVB/NJ and BALB/c with L. (L.) amazonensis - BMDM and peritoneal macrophages from FVB/NJ and BALB/c were infected with L. (L.) amazonensis for 24, 48, 72, and 96 h to characterise the infection over time in FVB/NJ macrophages. BMDM of BALB/c showed a higher percentage of infection starting from 48 h (Fig. 1A) compared with those from FVB/NJ. Regarding parasites per macrophage, those from BALB/c exhibited significantly higher numbers of amastigotes per cell at all time points (24 to 96 h) (Fig. 1B), indicating that FVB/NJ macrophages might either phagocytise fewer promastigotes or control intracellular parasites, preventing their survival or differentiation into amastigotes. In peritoneal macrophages, there was no statistically significant difference between the strains in terms of infection percentage, although BALB/c macrophages were slightly more infected than FVB/NJ macrophages [Supplementary data (Fig. 1A)]. In terms of parasites per macrophage, a significant difference was only observed at the 48-h time point, when BALB/c peritoneal macrophages exhibited a higher number of parasites per cell. In other time points, BALB/c peritoneal macrophages had slightly more intracellular parasites [Supplementary data (Fig. 1B)].

Fig. 1:
infection of bone marrow-derived macrophages (BMDM) from Friend Virus B NIH Jackson (FVB/NJ) and BALB/c with Leishmania (Leishmania) amazonensis promastigotes (MOI 1:10) for 24, 48, 72, and 96 h. The percentage of infected macrophages (A) and the number of parasites per infected macrophage (B) were determined. Results from a representative experiment out of three with similar profiles. Statistical analysis: Student’s t-test between FVB/NJ and BALB/c at each time point. *p ≤ 0.05. **p ≤ 0.01. ***p ≤ 0.001. ****p ≤ 0.0001.

Phagocytosis of promastigotes by FVB/NJ and BALB/c macrophages - We demonstrated that BALB/c BMDMs had significantly higher numbers of amastigotes per cell compared with FVB/NJ at 24-, 48-, 72-, and 96-h post-infection. This suggests that BALB/c macrophages either phagocytise more promastigotes or that FVB/NJ macrophages kill more parasites after phagocytosis. To address this question, we compared the adhesion and phagocytosis of promastigotes by BMDM from FVB/NJ and BALB/c strains. As shown in Fig. 2, BALB/c macrophages exhibited statistically similar number of internalised promastigotes compared with FVB/NJ, indicating that differences in phagocytosis do not explain the higher infection observed in BALB/c macrophages.

Fig. 2:
phagocytosis of Leishmania (Leishmania) amazonensis promastigotes by bone marrow-derived macrophages (BMDM) from Friend Virus B NIH Jackson (FVB/NJ) and BALB/c. (A) Representative images of the phagocytosis experiment. White arrows indicate internalised promastigotes, labelled with DAPI (blue), and yellow arrows indicate adhered promastigotes, labelled with Alexa 488 (green) (upper images). Parasite-unlabelled images represent the control without anti-Leish serum (lower images). Macrophage nuclei are stained with DAPI. Images were processed by deconvolution. (B) Number of bound and phagocytosed promastigotes per 100 macrophages after 5 min. Results from a representative experiment out of three with similar profiles. Student’s t-test between bound to FVB/NJ and BALB/c and between phagocytosed by FVB/NJ and BALB/c.

Phagocytosis of zymosan by FVB/NJ and BALB/c macrophages - With the aim of evaluating the process of phagocytosis by FVB/NJ and BALB/c macrophages, cells were incubated with zymosan particles for 1 h. Fig. 3 shows that BALB/c macrophages phagocytic capacity (Fig. 3A-C).

Fig. 3:
phagocytosis of zymosan particles (yellow arrows) by bone marrow-derived macrophages (BMDM) from Friend Virus B NIH Jackson (FVB/NJ) and BALB/c (A). Percentage of macrophages with zymosan (left) and number of particles phagocytosed by macrophage (right) after 1 h (B). Results from a representative experiment out of three with similar profiles. Student’s t-test.

Resistance of promastigotes to FVB/NJ and BALB/c complement - As already mentioned, FVB/NJ strain has a deficiency in the complement factor C5 and in the expression of C3 convertase. This prompted us to compare the resistance of L. (L.) amazonensis promastigotes to FVB/NJ and BALB/c sera. Strikingly, the results shown in Fig. 4 indicate that sera from the two mouse strains have similar lytic capacity, despite the known complement deficiency of FVB/NJ mice.

Fig. 4:
resistance of Leishmania (Leishmania) amazonensis promastigotes (Day 4) to Friend Virus B NIH Jackson (FVB/NJ) and BALB/c complements. The percentage of viable cells relative to the control (without serum) after 30 min incubation at 34ºC in the presence of 2.5%, 5%, 10%, and 20% serum pooled from mice of the two strains. Assays performed in technical triplicates. Statistical analysis: Student’s t-test. Results are from a representative experiment out of three with similar profiles.

In vivo infection of FVB/NJ and BALB/c strains with L. (L.) amazonensis - To characterise cutaneous lesion development and parasite burden over 12 weeks, FVB/NJ and BALB/c mice were infected in the footpad with L. (L.) amazonensis expressing luciferase (La-LUC). Lesion size was measured weekly, and parasite burden was estimated weekly by luminescence.

The number of animals that developed or not active infection 12 weeks after inoculation was analysed, and only luminescence from animals with active infection was quantified. As shown in Fig. 5A, six animals from FVB/NJ and eight from BALB/c showed active infection. Fisher’s exact test indicated no statistical difference between the two strains.

Fig. 5:
lesion development and parasite load of Friend Virus B NIH Jackson (FVB/NJ) and BALB/c mice (10 animals in each group) infected with Leishmania (Leishmania) amazonensis MHOM/BR/1973/M2269 La-LUC. (A) Number of animals with (infection) and without (no infection) active infection 12 weeks after inoculation. Statistical analysis by Fisher’s exact test. (B) Footpad lesion thickness of FVB/NJ and BALB/c mice over 12 weeks. Lesion development (left) and area under curve (AUC) (right). Statistical analysis: Student’s t-test for AUC. (C) Parasite burden calculated by radiance from the region of interest (ROI) (luminescence) in FVB/NJ and BALB/c mice over 12 weeks. The difference between photon emissions (lesion - uninfected footpad) was calculated and expressed in photons/s/cm2/sr. The same ROI was applied to all animals. Radiance (left) and AUC (right). Statistical analysis: Student’s t-test for AUC.

As shown in Fig. 5B-C, there was also no statistical difference in lesion size and in parasite burden between the two strains over the course of 12 weeks, although BALB/c developed slightly larger lesions after the seventh week of infection.

DISCUSSION

No study up to now employed FVB mice in Leishmania infections. SWR/J mice, from which FVB/NJ was derived, have rarely been used as model for Leishmania infection.4444. Kiige SG, Mutiso JM, Laban LT, Khayeka-Wandabwa C, Anjili CO, Ingonga J, et al. F1 cross-breed between susceptible BALB/c and resistant Swiss mice infected with Leishmania major exhibit an intermediate phenotype for lesion sizes and type 1 cytokines but show low level of total IgG antibodies. Scand J Immunol. 2014; 79(5): 283-91.In vitro infection of BMDM and peritoneal macrophages with L. (L.) infantum (syn L. (L.) chagasi) showed that BALB/c macrophages are more susceptible to infection and more permissive to amastigote replication than SWR/J macrophages, with no difference in cytokine production.4545. Van den Kerkhof M, Van Bockstal L, Gielis JF, Delputte P, Cos P, Maes L, et al. Impact of primary mouse macrophage cell types on Leishmania infection and in vitro drug susceptibility. Parasitol Res. 2018; 117(11): 3601-12. Our observations that FVB/NJ BMDM are less infected by L. (L.) amazonensis than BALB/c seem similar to the results obtained for SWR/J macrophages.

Our in vivo infections indicated that FVB/NJ is permissive to infection by L. (L.) amazonensis. In vivo infection in the footpad of BALB/c and SWR/J with the Isabel-NIH strain of L. (L.) amazonensis, diffuse leishmaniasis, resulted in lesions that became necrotic in both strains, and the loss of the affected footpad was frequently observed after 14 weeks.4646. Shaw BA, Grayson JB, Petersen EA. Experimental murine infection with the diffuse cutaneous leishmaniasis strain, Isabel. Ann Trop Med Parasitol. 1987; 81(1): 9-14. In the present study, lesions did not lead to footpad loss neither in FVB/NJ nor in BALB/c strains after 12 weeks. Indeed, we have previously shown that infection with L. (L.) amazonensis LV79 strain for 13 weeks did not lead to footpad loss.2020. Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703.,4747. de Rezende E, Kawahara R, Peña MS, Palmisano G, Stolf BS. Quantitative proteomic analysis of amastigotes from Leishmania (L.) amazonensis LV79 and PH8 strains reveals molecular traits associated with the virulence phenotype. PLoS Negl Trop Dis. 2017; 11(11): e0006090.

SWR/J mice were more effective in controlling in vivo infection by L. (L.) major compared to BALB/c mice and to animals resulting from a cross between the two strains.4444. Kiige SG, Mutiso JM, Laban LT, Khayeka-Wandabwa C, Anjili CO, Ingonga J, et al. F1 cross-breed between susceptible BALB/c and resistant Swiss mice infected with Leishmania major exhibit an intermediate phenotype for lesion sizes and type 1 cytokines but show low level of total IgG antibodies. Scand J Immunol. 2014; 79(5): 283-91. Infection of SWR/J and C57BL/6 mice with L. (L.) major exhibited a similar pattern of lesion development, which regressed after the fourth week, and these two strains have a similar cytokine production pattern, with high levels of IFN-γ and low levels of IL-4.4848. De Oliveira MR, Tafuri WL, Nicoli JR, Vieira EC, Melo MN, Vieira LQ. Influence of microbiota in experimental cutaneous leishmaniasis in Swiss MICE. Rev Inst Med Trop Sao Paulo. 1999; 41(2): 87-94. Another study reported that in vivo infection with L. (L.) major may show differences between BALB/c and SWR/J mice depending on the site of inoculation. SWR/J mice exhibited higher ability to control infections in the footpad than BALB/c mice, and ulcerated lesions were observed after the eighth week of infection in BALB/c footpads. From the eleventh week on lesions in SWR/J mice started to resolve, with the presence of small, ulcerated lesions, while BALB/c lesions did not regress. BALB/c mice had significantly higher numbers of parasites in the footpad, spleen, and draining lymph nodes compared with SWR/J mice. The parasite burden followed the disease progression profile for both mice strains.4949. Nabors GS, Farrell JP. Site-specific immunity to Leishmania major in SWR mice: the site of infection influences susceptibility and expression of the antileishmanial immune response. Infect Immun. 1994; 62(9): 3655-62.

In the present study, BALB/c mice also developed slightly larger lesions in the footpad compared with FVB/NJ mice from the seventh week of infection on, although with no statistical differences. The parasite burden followed the disease progression profile (lesion thickness) until the tenth week. Curiously, the known C5-deficiency of FVB/NJ mice had no impact in parasite survival in in vivo infections. Accordingly, and different from our expectations, we observed no differences in promastigote survival after incubations with BALB/c and FVB/NJ serum.

This work shows that FVB/NJ mouse strain is promising as a model for studying tegumentary leishmaniasis by Leishmania (L.) amazonensis. Indeed, its similarity with the BALB/c strain (the most frequently used murine model for Leishmania infection) in terms of phagocytosis and in vivo infections suggests that it might be another murine strain susceptible to this Leishmania species, useful for analysing different aspects of cutaneous localised leishmaniasis. The use of this novel model also paves the way for further studies aimed at understanding the pathogenesis of lesions caused by L. (L.) amazonensis and other dermotropic species of Leishmania. This is especially pertinent given the widespread use of FVB/NJ as a transgenic model in several disease pathogenesis studies.

ACKNOWLEDGEMENTS

To Prof Francisco Laurindo and Victor Debbas for providing FVB/NJ mice. We also acknowledge CEFAP-USP facility for making available the bioimaging equipment (IVIS).

REFERENCES

  • 1
    Steverding D. The history of leishmaniasis. Parasit Vectors. 2017; 10(1): 1-10.
  • 2
    WHO - World Health Organization. Leishmaniasis. [cited 2023 Sep 4]. Available from: https://www.who.int/data/gho/data/themes/topics/gho-ntd-leishmaniasis
    » https://www.who.int/data/gho/data/themes/topics/gho-ntd-leishmaniasis
  • 3
    Reithinger R, Dujardin JC, Louzir H, Pirmez C, Alexander B, Brooker S. Cutaneous leishmaniasis. Lancet Infect Dis. 2007; 7(9): 581-96.
  • 4
    Akhoundi M, Kuhls K, Cannet A, Votýpka J, Marty P, Delaunay P, et al. A historical overview of the classification, evolution, and dispersion of Leishmania parasites and sandflies. PLoS Negl Trop Dis. 2016; 10(3): e0004349.
  • 5
    Torres-Guerrero E, Quintanilla-Cedillo MR, Ruiz-Esmenjaud J, Arenas R. Leishmaniasis: a review. F1000Res. 2017; 6: 750.
  • 6
    Coughlan S, Mulhair P, Sanders M, Schonian G, Cotton JA, Downing T. The genome of Leishmania adleri from a mammalian host highlights chromosome fission in Sauroleishmania. Sci Rep. 2017; 7(1): 43747.
  • 7
    Doehl JSP, Bright Z, Dey S, Davies H, Magson J, Brown N, et al. Skin parasite landscape determines host infectiousness in visceral leishmaniasis. Nat Commun. 2017; 8(1): 1-10.
  • 8
    Rougeron V, de Meeûs T, Ouraga SK, Hide M, Bañuls AL. "Everything you always wanted to know about sex (but were afraid to ask)" in Leishmania after two decades of laboratory and field analyses. Rall GF, editor. PLoS Pathog. 2010; 6(8): 7-8.
  • 9
    Scott P, Novais FO. Cutaneous leishmaniasis: immune responses in protection and pathogenesis. Nat Rev Immunol. 2016; 16(9): 581-92.
  • 10
    Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev. 2013; 26(1): 58-85.
  • 11
    Giraud E, Svobodova M, Muller I, Volf P, Rogers ME. Promastigote secretory gel from natural and unnatural sand fly vectors exacerbate Leishmania major and Leishmania tropica cutaneous leishmaniasis in mice. Parasitology. 2019; 146(14): 1796-802.
  • 12
    Lauletta-Lindoso JA, Cunha MA, Queiroz I, Moreira CHV. Leishmaniasis-HIV coinfection: current challenges. HIV/AIDS - research and palliative care. 2016; 8: 147-56.
  • 13
    Sacks D, Noben-Trauth N. The immunology of susceptibility and resistance to Leishmania major in mice. Nat Rev Immunol. 2002; 2(11): 845-58.
  • 14
    Loría-Cervera EN, Andrade-Narváez FJ. Animal models for the study of leishmaniasis immunology. Rev Inst Med Trop São Paulo. 2014; 56(1): 1-11.
  • 15
    Handman E. Leishmaniasis: current status of vaccine development. Clin Microbiol Rev. 2001; 14(2): 229-43.
  • 16
    Mears ER, Modabber F, Don R, Johnson GE. A Review: The current in vivo models for the discovery and utility of new anti-leishmanial drugs targeting cutaneous leishmaniasis. PLoS Negl Trop Dis. 2015; 9(9): e0003889.
  • 17
    de Oliveira CI, Teixeira MJ, Gomes R, Barral A, Brodskyn C. Animal models for infectious diseases caused by parasites: Leishmaniasis. Drug Discov Today Dis Models. 2004; 1(1): 81-6.
  • 18
    Heinzel FP, Sadick MD, Holaday BJ, Coffman RL, Locksley RM. Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets. J Exp Med. 1989; 169(1): 59-72.
  • 19
    Ji J, Masterson J, Sun J, Soong L. CD4+CD25+ Regulatory T cells restrain pathogenic responses during Leishmania amazonensis infection. J Immunol. 2005; 174(11): 7147-53.
  • 20
    Velasquez LG, Galuppo MK, De Rezende E, Brandão WN, Peron JP, Uliana SRB, et al. Distinct courses of infection with Leishmania (L.) amazonensis are observed in BALB/c, BALB/c nude and C57BL/6 mice. Parasitology. 2016; 143(6): 692-703.
  • 21
    Taketo M, Schroeder AC, Mobraaten LE, Gunning KB, Hanten G, Fox RR, et al. FVB/N: An inbred mouse strain preferable for transgenic analyses. Proc Natl Acad Sci USA. 1991; 88(6): 2065-9.
  • 22
    Wetsel RA, Fleischer DT, Haviland DL. Deficiency of the murine fifth complement component (C5). A 2-base pair gene deletion in a 5'-exon. J Biol Chem. 1990; 265(5): 2435-40.
  • 23
    Bayly-Jones C, Bubeck D, Dunstone MA. The mystery behind membrane insertion: a review of the complement membrane attack complex. Philos Trans R Soc Lond B Biol Sci. 2017; 372(1726): 20160221.
  • 24
    Mosser DM, Edelson PJ. The mouse macrophage receptor for C3bi (CR3) is a major mechanism in the phagocytosis of Leishmania promastigotes. J Immunol. 1985; 135(4): 2785-9.
  • 25
    Mosser DM, Edelson PJ. The third component of complement (C3) is responsible for the intracellular survival of Leishmania major. Nature. 1987; 327(6120): 329-31.
  • 26
    Verma S, Mandal A, Ansari MdY, Kumar A, Abhishek K, Ghosh AK, et al. Leishmania donovani inhibitor of serine peptidases 2 mediated inhibition of lectin pathway and upregulation of C5aR signaling promote parasite survival inside host. Front Immunol. 2018; 9: 63.
  • 27
    Prat-Luri B, Neal C, Passelli K, Ganga E, Amore J, Firmino-Cruz L, et al. The C5a-C5aR1 complement axis is essential for neutrophil recruitment to draining lymph nodes via high endothelial venules in cutaneous leishmaniasis. Cell Rep. 2022; 39(5): 110777.
  • 28
    Tanner SM, Lorenz RG. FVB/N mouse strain regulatory T cells differ in phenotype and function from the C57BL/6 and BALB/C strains. FASEB Bioadv. 2022; 4(10): 648-61.
  • 29
    Mähler M, Janke C, Wagner S, Hedrich HJ. Differential susceptibility of inbred mouse strains to Helicobacter pylori infection. Scand J Gastroenterol. 2002; 37(3): 267-78.
  • 30
    Toth LA, Verhulst SJ. Strain differences in sleep patterns of healthy and influenza-infected inbred mice. Behav Genet. 2003; 33(3): 325-36.
  • 31
    Lee KJ, Park SK, Im JA, Kim SK, Kim GH, Kim GY, et al. Susceptibility of several strains of mice to Echinostoma hortense infection. Korean J Parasitol. 2004; 42(2): 51-6.
  • 32
    Cywinska A, Czuminska K, Schollenberger A. Granulomatous inflammation during Heligmosomoides polygyrus primary infections in FVB mice. J Helminthol. 2004; 78(1): 17-24.
  • 33
    Robertson CM, Perrone EE, McConnell KW, Dunne WM, Boody B, Brahmbhatt T, et al. Neutrophil depletion causes a fatal defect in murine pulmonary Staphylococcus aureus clearance. J Surg Res. 2008; 150(2): 278-85.
  • 34
    Knott ML, Hogan SP, Wang H, Matthaei KI, Dent LA. FVB/N mice are highly resistant to primary infection with Nippostrongylus brasiliensis. Parasitology. 2009; 136(1): 93-106.
  • 35
    Specht S, Taylor MD, Hoeve MA, Allen JE, Lang R, Hoerauf A. Over expression of IL-10 by macrophages overcomes resistance to murine filariasis. Exp Parasitol. 2012; 132(1): 90-6.
  • 36
    Machado MPR, Rocha AM, de Oliveira LF, de Cuba MB, Loss IO, Castellano LR, et al. Autonomic nervous system modulation affects the inflammatory immune response in mice with acute Chagas disease. Exp Physiol. 2012; 97(11): 1186-202.
  • 37
    Kim EM, Bae YM, Choi MH, Hong ST. Cyst formation, increased anti-inflammatory cytokines and expression of chemokines support for Clonorchis sinensis infection in FVB mice. Parasitol Int. 2012; 61(1): 124-9.
  • 38
    Kim EM, Yu HS, Jin Y, Choi MH, Bae YM, Hong ST. Local immune response to primary infection and re-infection by Clonorchis sinensis in FVB mice. Parasitol Int. 2017; 66(4): 436-42.
  • 39
    Marion E, Jarry U, Cano C, Savary C, Beauvillain C, Robbe-Saule M, et al. FVB/N mice spontaneously heal ulcerative lesions induced by Mycobacterium ulcerans and switch M. ulcerans into a low mycolactone producer. J Immunol. 2016; 196(6): 2690-8.
  • 40
    Bhagwat SP, Gigliotti F, Wang J, Wang Z, Notter RH, Murphy PS, et al. Intrinsic programming of alveolar macrophages for protective antifungal innate immunity against Pneumocystis infection. Front Immunol. 2018; 9: 2131.
  • 41
    Khaiboullina SF, Lopes P, De Carvalho TG, Real ALCV, Souza DG, Costa VV, et al. Host immune response to ZIKV in an immunocompetent embryonic mouse model of intravaginal infection. Viruses. 2019; 11(6): 558.
  • 42
    Peña MS, Tang FHF, Franco FAL, Rodrigues AT, Carrara GMP, Araujo TLS, et al. Leishmania (L.) amazonensis LaLRR17 increases parasite entry in macrophage by a mechanism dependent on GRP78. Parasitology. 2023; 150(10): 922-33.
  • 43
    Tano FT, Telleria EL, Rêgo FD, Coelho FS, de Rezende E, Soares RP, et al. Phenotypical differences between Leishmania (Leishmania) amazonensis PH8 and LV79 strains may impact survival in mammal host and in phlebotomine sand flies. Pathogens. 2023; 12(2): 173.
  • 44
    Kiige SG, Mutiso JM, Laban LT, Khayeka-Wandabwa C, Anjili CO, Ingonga J, et al. F1 cross-breed between susceptible BALB/c and resistant Swiss mice infected with Leishmania major exhibit an intermediate phenotype for lesion sizes and type 1 cytokines but show low level of total IgG antibodies. Scand J Immunol. 2014; 79(5): 283-91.
  • 45
    Van den Kerkhof M, Van Bockstal L, Gielis JF, Delputte P, Cos P, Maes L, et al. Impact of primary mouse macrophage cell types on Leishmania infection and in vitro drug susceptibility. Parasitol Res. 2018; 117(11): 3601-12.
  • 46
    Shaw BA, Grayson JB, Petersen EA. Experimental murine infection with the diffuse cutaneous leishmaniasis strain, Isabel. Ann Trop Med Parasitol. 1987; 81(1): 9-14.
  • 47
    de Rezende E, Kawahara R, Peña MS, Palmisano G, Stolf BS. Quantitative proteomic analysis of amastigotes from Leishmania (L.) amazonensis LV79 and PH8 strains reveals molecular traits associated with the virulence phenotype. PLoS Negl Trop Dis. 2017; 11(11): e0006090.
  • 48
    De Oliveira MR, Tafuri WL, Nicoli JR, Vieira EC, Melo MN, Vieira LQ. Influence of microbiota in experimental cutaneous leishmaniasis in Swiss MICE. Rev Inst Med Trop Sao Paulo. 1999; 41(2): 87-94.
  • 49
    Nabors GS, Farrell JP. Site-specific immunity to Leishmania major in SWR mice: the site of infection influences susceptibility and expression of the antileishmanial immune response. Infect Immun. 1994; 62(9): 3655-62.
  • Financial support: FAPESP (grant numbers 2018/14972-8 and 2021/08915-4 to BSS), CAPES (88887.360800/2019-00).

Publication Dates

  • Publication in this collection
    15 Mar 2024
  • Date of issue
    2024

History

  • Received
    05 Oct 2023
  • Accepted
    19 Feb 2024
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