Cellular Immunology 289 (2014) 162–166

Contents lists available at ScienceDirect

Cellular Immunology journal homepage: www.elsevier.com/locate/ycimm

Reactivity of autoantibodies against not only erythrocytes but also hepatocytes in sera of mice with malaria Yasuhiro Kanda a, Toshihiko Kawamura a,⇑, Takahiro Kobayashi a,b, Hiroki Kawamura a,c, Hisami Watanabe d, Toru Abo a a

Division of Immunology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan Department of Clinical Engineering and Medical Technology, Niigata University of Health and Welfare, Niigata 950-3198, Japan d Division of Cellular and Molecular Immunology, Center of Molecular Biosciences, University of Ryukyus, Okinawa 903-0213, Japan b c

a r t i c l e

i n f o

Article history: Received 18 September 2013 Accepted 15 April 2014 Available online 24 April 2014 Keywords: Autoantibody Plasmodium yoelii Malaria Hepatocytes Erythrocytes

a b s t r a c t In order to further examine the reactivity of autoantibodies, mice were infected with a non-lethal strain of Plasmodium yoelii. Parasitemia appeared between days 10 and 21. During this period, hyperglycemia and hypothermia were serially obeserved and this phenomenon resembled stress-associated responses. In parallel with parasitemia, autoantibodies appeared against nucleus and double-stranded DNA in the sera. To examine further the reactivity of autoantibodies against tissues, immunohistochemical staining using sera from mice with or without malaria was conducted. Autoantibodies contained reactivity to erythrocytes in the spleen, bone marrow and peripheral blood, especially against tissues obtained from mice with malaria. In the liver and intestine, autoantibodies reacted with hepatocytes and intestinal epithelial cells, respectively. These results suggested that the reactivity of autoantibodies against erythrocytes and hepatocytes might be associated with the modulation of the disease course in malaria. Ó 2014 Elsevier Inc. All rights reserved.

1. Introduction Protection against malaria was considered an event of the conventional immune system against malaria protozoa acting as foreign antigens [1–7]. Thymus-derived (T) lymphocytes and bone marrow-derived (B) lymphocytes play important roles in adoptive immune responses. This concept is the basis for malaria vaccination trials using antigens from the malaria protozoa [8–11]. It is considered that conventional B cells (B-2 cells) and helper T cells recognize protozoa antigens and produce protective antibodies [12–14]. However, malaria also induces innate immune responses and results in the activation of autoreactive extrathymic T cells [15– 19] and the production of autoantibodies by B-1 cells during infection [20–23]. Clinical studies have reported the detection of high titers of autoantibodies against human nucleus or double-stranded (ds) DNA in the sera of patients with malaria [24–29]. Thus, we have intensively characterized the properties of autoreactive extrathymic T cells and autoantibody-producing B-1 cells in mice with malaria. Simultaneous activation of extrathymic T

⇑ Corresponding author. Fax: +81 25 227 0766. E-mail address: [email protected] (T. Kawamura). http://dx.doi.org/10.1016/j.cellimm.2014.04.008 0008-8749/Ó 2014 Elsevier Inc. All rights reserved.

cells and B-1 cells is a common response in mice with malaria [30–33]. In the present study, we investigated immune responses during blood-stage malaria in mice and examined the reactivity of the autoantibodies in sera. The unique reactivity of autoantibodies to self-tissues during malaria (autoreactivity to erythrocytes and hepatocytes) suggested the beneficial function of autoantibodies for protection against malaria.

2. Materials and methods 2.1. Mice and parasites C57BL/6 (B6) mice at age 8–14 weeks were used. These mice were maintained at the animal facility of Niigata University (Niigata, Japan) under specific pathogen-free conditions. Plasmodium yoelii (P. yoelii) 17XNL (non-lethal strain), a generous gift from Dr. S. Waki (Gunma Prefectural College of Health Science, Maebashi, Japan) was used. Parasites were maintained by routine in vivo passage in mice [21]. Mice were infected by intraperitoneal injection of 104 parasitized erythrocytes per mouse. Parasitemia in the blood was observed by Giemsa staining every 2 or 3 days and mice were sacrificed at the indicated days after infection.

Y. Kanda et al. / Cellular Immunology 289 (2014) 162–166

163

2.2. Measurement of blood glucose and rectal temperature Blood glucose was measured from 3.5 ll blood from mice by venesection at the tail tip with Precision Xceed (Abbott Japan Co., Chiba, Japan). The rectal temperature of mice was measured every 2 or 3 days with a digital thermometer (TD-300, Shibaura Electronics Co. Ltd., Tokyo, Japan). 2.3. Identification of autoantibodies Autoantibodies were detected using a HEp-2 cell line in conjunction with an immunofluorescence test. Sera obtained from P. yoelii 17XNL infected or uninfected control mice were used after a 1/10 dilution. FITC-conjugated anti-mouse Ig (Dako, Glostrup, Denmark) was used as a secondary antibody for detection. The activity of such anti-ds DNA antibodies was also detected in the sera and supernatant using an anti-ds DNA mouse enzyme-linked immunosorbent assay (ELISA) kit (Shibayagi, Gunma, Japan). 2.4. Immnohistochemistry Tissues were removed from euthanized mice and fixed in formalin, dehydrated, embedded in paraffin and sectioned at 3 lm as previously described [34]. Blood smears were fixed with methanol. Endogenous peroxidase in the sections was blocked with 3% hydrogen peroxide in water for 5 min, and the Avidin/Biotin Blocking Kit (Vector, Burlingame, CA, USA) was used to block endogenous avidin and biotin. Mouse on Mouse (M.O.M.) Immunodetection Kit (Vector, Burlingame, CA, USA) was used for blocking non-specific mouse immunoglobulin (Ig) and reducing the background staining. The sections were incubated with Mouse Ig Blocking reagent for 1 h and M.O.M. Protein Concentrate solution. Sera of infected or uninfected mice at a 1/5 dilution were applied to the section for 30 min as primary autoantibodies. Subsequently, M.O.M. biotinylated anti-mouse IgG reagent was incubated for 10 min, followed by incubation with Vectastain Elite ABC Kit. Finally, the slides were stained with diaminobenzidine and counterstained with hematoxylin.

Fig. 1. Time kinetic study on parasitemia, blood glucose and rectal temperature. Four mice were used to determine the mean ± SD.

3. Results 3.1. Parasitemia and stress-associated responses A time-kinetic study on parasitemia was conducted after P. yoelii injection (Fig. 1). From day 7, erythrocytes infected with the protozoa were observed in the blood. Parasitemia gradually became prominent and continued up to day 21. In parallel, the levels of blood glucose and rectal temperature were examined. The level of blood glucose increased at days 10–17 and the rectal temperature decreased approximately at day 21. The presence of hyperglycemia and hypothermia resembled stress-associated responses. 3.2. Reactivity of autoantibodies Using HEp-2 cells, the reactivity of autoantibodies in sera from mice with malaria was examined (Fig. 2A). Sera of autoimmune prone MRL-lpr/lpr mice was used as a positive control (top of the figure). The low reactivity of sera (total Ig) was observed on day 14 but the highest reactivity of sera was present at day 28. This reactivity was confirmed by IgG and IgM isotypes. IgG autoantibodies had reactivity to the nucleus while IgM autoantibodies were reactive to a perinuclear area of cells. In the case of sera from MRLlpr/lpr mice, only IgG autoantibodies had specific reactivity to the nucleus. There was no reactivity of the IgM isotype in this case.

By ELISA, the titer of anti-ds DNA antibodies was examined (Fig. 2B). Reactivity appeared from day 14 after infection and increased up to day 28. Positive control sera from MRL-lpr/lpr mice had the highest titer of anti-ds DNA antibodies. 3.3. Reactivity of autoantibodies to the cell surface of erythrocytes and others Immunofluorescence and ELISA analysis suggested a wide reactivity of autoantibodies to the nucleus and cytoplasm (perinuclear area). We then determined whether the sera from mice with malaria had reactivity to tissues and erythrocytes (Fig. 3). First, the reactivity of sera against the spleen was examined. Sera from mice with malaria showed a positive reaction against the normal spleen, especially in the splenic medullary region where denatured erythrocytes were abundant. The reactivity of sera with follicular area was lower. In contrast, control sera did not have reactivity against the spleen. A similar reactivity was also observed using malaria-infected splenic tissue (right column). In this picture, dark spots in the spleen represent malarial pigments. Second, the reactivity of sera against bone marrow (BM) was examined. Some reactivity against BM cells (mainly erythroid cells) was observed even in normal BM.

164

Y. Kanda et al. / Cellular Immunology 289 (2014) 162–166

Fig. 2. Autoantibodies against intracellular components. (A) Immunofluorescence analysis using HEp-2 cells, (B) ELISA assay to detect the titers of anti-ds DNA antibodies. In experiment b, four mice were used to determine the mean ± SD.

This reactivity was confirmed using malaria-infected BM (right column). The reactivity of sera was examined in the smear of peripheral blood and was slightly positive against normal erythrocytes. Higher reactivity was seen against erythrocytes obtained from mice with malaria (day 28). The surface of erythrocytes was highly positive. The reactivity of sera against the liver (especially hepatocytes) was then examined using liver tissue. Control sera did not show reactivity against normal liver. In sharp contrast, sera from mice with malaria showed a positive reaction against normal liver and malaria-infected liver. In the right column, dark spots seen in the liver represent malarial pigments. Finally, the large intestine (Li) was used to examine the reactivity of sera. Considerable sera reactivity was observed in the epithelial region of the Li. This was apparent, irrespective of infection. 4. Discussion In the present study, we demonstrated that sera from mice with malaria contained autoantibodies with reactivity against erythrocytes and hepatocytes in tissue sections by immunohistochemical analysis. Although the reactivity was higher against infected cells compared with control cells, some antigens on normal erythro-

cytes and hepatocytes were targets of the autoantibodies in malaria. Therefore, the reactivity was not against malaria parasite-specific antigens but against erythrocytes and hepatocytes, irrespective of infection. Thus, malaria may increase the antigenicity of erythrocytes and hepatocytes. Immunofluorescence and ELISA assays demonstrated that autoantibodies were reactive with many cell components; not only nucleus and ds DNA, but also surface antigens of erythrocytes and hepatocytes. In humans and mice, malaria consistently induces serum autoantibodies [20–29]. In contrast, there is little evidence that conventional T cells (TCRhigh cells) and B cells (B-2 cells) are activated during malaria, especially in mice. Even if conventional immunity is activated, memory immune responses against malaria protozoa tend to disappear within 1 year [35,36]. However, extrathymic T cells (TCRint cells) are prominently activated in mice with malaria [15,16]. Previously, we demonstrated that autoantibody-producing B-1 cells were activated in parallel with the activation of extrathymic T cells [20,21]. In light of these findings, protection from malaria may be due to innate immunity mediated by unconventional extrathymic T cells and B-1 cells and is supported by the observation of reactive autoantibodies identified in this study. Malaria consists of a hepatic stage and blood stage during infection [37]. During the hepatic stage, intracellular malaria parasites invade hepatocytes whereas during the blood stage they invade erythrocytes. Therefore, conventional T cells and conventional antibodies against parasites may not react to parasitic antigens. Under this situation, autoantibodies against surface antigens on hepatocytes and erythrocytes may be important for protection from malaria. Previous studies demonstrated that autoantibodies against erythrocytes or their surface molecules were induced in malaria [38–43] (Table 1). In addition, the autoantibodies were found to react with intracellular components and many cell surface components, including those on neutrophils [44,45] and thrombocytes [46,47]. The immune status observed in malaria is summarized in Table 1. Since the antigenicity of such surface antigens (especially on erythrocytes) is increased by malaria, the autoreactivity of activated extrathymic T cells and the autoantibodies of activated B-1 cells might efficiently react with infected erythrocytes and hepatocytes. Finally, such denatured hepatocytes and erythrocytes are processed by macrophages in the spleen and Kupffer cells in the liver. Hepatosplenomegaly with malaria pigments is consistently observed in humans and mice in malaria [33]. Results from immunohistochemistry of tissue sections revealed that the most reactive sites of autoantibodies in mice with malaria were the splenic medulla, bone marrow and peripheral erythrocytes, especially in infected tissues. This reactivity supports the idea that autoantibodies preferentially attack infected erythrocytes. In addition to hepatocytes, epithelial cells in the intestine were reactive to the autoantibodies. Since hepatocytes were phylogenetically generated from intestinal epithelial cells [48], this suggests cross-reactivity between hepatocytes and intestinal epithelial cells. An immune status of activated extrathymic T cells and autoantibody-producing B-1 cells is common to aging [49,50], autoimmune diseases [51,52], chronic graft-vs-host disease [53,54] and malaria. During these immune responses, the thymus often becomes atrophied or involuted. Moreover, these immune states also resemble stress-associated responses [55,56]. This study confirmed that stress-associated responses such as hypothermia and hyperglycemia were observed in malaria. Taken together, this suggests that malaria induces an innate immune response rather than an adaptive immune response and that the reactivity of autoantibodies against the surface of erythrocytes and hepatocytes should be further studied.

Y. Kanda et al. / Cellular Immunology 289 (2014) 162–166

165

Fig. 3. Immunohistochemical staining of sera using various tissues. The spleen, bone marrow (BM), peripheral blood (PB), liver and large intestine (Li) were examined. Normal and malaria-infected tissues were used. Sections were stained with hematoxylin (blue) and diaminobenzidine (brown).

Table 1 Immunologic state seen in malarial infection. T cells

B cells (antibody production)

Thymic atrophy and suppression of conventional T cells [15,16]

Dysregulation of conventional B cells (B-2) and paucity of antibodies against Plasmodium [20,21] Activation of B-1 cells [20–23] and production of autoantibodies against:

Activation of unconventinal T cells, including NKT cells and extrathymic T cells in mice [15– 19] and in humans [32]

Intracellular components [24–29] e.g., nucleus, ds DNA, actin, etc. Cell surface components e.g., erythrocytes [38–43], neutrophils [44,45], thrombocytes [46,47], hepatocytesa, etc. [ ] References a This paper.

Acknowledgments We wish to thank Mrs. Yuko Kaneko for manuscript preparation. This work was supported by a grant-in-aid for scientific

research from the Ministry of Education, Science, and Culture, Japan. References [1] S. Dick, M. Waterfall, J. Currie, A. Maddy, E. Riley, Naive human ab T cells respond to membrane-associated components of malaria-infected erythrocytes by proliferation and production of interferon-c, Immunology 88 (1996) 412–420. [2] Q. Chen, A. Heddini, A. Barragan, V. Fernandez, S.F.A. Pearce, M. Wahlgren, The semiconserved head structure of Plasmodium falciparum erythrocyte membrane protein 1 mediates binding to multiple independent host receptors, J. Exp. Med. 192 (2000) 1–10. [3] J. Langhorne, The role of CD4+ T-cells in the immune response to Plasmodium chabaudi, Parasitol. Today 5 (1989) 362–364. [4] R.S. Phillips, K.E. Mathers, A.W. Taylor-Robinson, T cells in immunity to Plasmodium chabaudi chabaudi: operation and regulation of different pathways of protection, Res. Immunol. 145 (1994) 406–412. [5] A.W. Taylor-robinson, Regulation of immunity to malaria: valuable lessons learned from murine models, Parasitol. Today 11 (1995) 334–342. [6] J. Currier, H.P. Beck, B. Currie, M.F. Good, Antigens released at schizont burst stimulate Plasmodium falciparum-specific CD4+ T cells from non-exposed donors: Potential for cross-reactive memory T cells to cause disease, Int. Immunol. 7 (1995) 821–833.

166

Y. Kanda et al. / Cellular Immunology 289 (2014) 162–166

[7] D.L. Doolan, S.L. Hoffman, IL-12 and NK cells are required for antigen-specific adaptive immunity against malaria initiated by CD8+ T cells in the Plasmodium yoelii model, J. Immunol. 163 (1999) 884–892. [8] J.H. Playfair, J.B. De Souza, R.R. Freeman, A.A. Holder, Vaccination with a purified blood-stage malaria antigen in mice. correlation of protection with T cell mediated immunity, Clin. Exp. Immunol. 62 (1985) 19–23. [9] J.B. De Souza, J.H. Playfair, A novel adjuvant for use with a blood-stage malaria vaccine, Vaccine 13 (1995) 1316–1319. [10] Y. Miyahira, A. Garcia-Sastre, D. Rodriguez, J.R. Rodriguez, K. Murata, M. Tsuji, P. Palese, M. Esteban, F. Zavala, R.S. Nussenzweig, Recombinant viruses expressing a human malaria antigen can elicit potentially protective immune CD8+ responses in mice, Proc. Natl. Acad. Sci. USA 95 (1998) 3954–3959. [11] O. Bruna-Romero, G. Gonzalez-Aseguinolaza, J.C.R. Hafalla, M. Tsuji, R.S. Nussenzweig, Complete, long-lasting protection against malaria of mice primed and boosted with two distinct viral vectors expressing the same plasmodial antigen, Proc. Natl. Acad. Sci. USA 98 (2001) 11491–11496. [12] W.R. Weiss, M. Sedegah, J.A. Berzofsky, S.L. Hoffman, The role of CD4+ T cells in immunity to malaria sporozoites, J. Immunol. 151 (1993) 2690–2698. [13] Y. Charoenvit, V.F. Majam, G. Corradin, J.B.Jr. Sacci, R. Wang, D.L. Doolan, T.R. Jones, E. Abot, M.E. Patarroyo, F. Guzman, S.L. Hoffman, CD4+ T-cell- and gamma interferon-dependent protection against murine malaria by immunization with linear synthetic peptides from a Plasmodium yoelii 17kilodalton hepatocyte erythrocyte protein, Infect. Immun. 67 (1999) 5604– 5614. [14] T. Schlotmann, I. Waase, C. Jülch, U. Klauenberg, B. Müller-Myhsok, M. Dietrich, B. Fleischer, B.M. Bröker, CD4 ab T lymphocytes express high levels of the T lymphocyte antigen CTLA-4 (CD152) in acute malaria, J. Infect. Dis. 182 (2000) 367–370. [15] A. Weerasinghe, H. Sekikawa, H. Watanabe, M.D.K. Mannoor, S.R.M. Morshed, R.C. Halder, T. Kawamura, T. Kosaka, C. Miyaji, H. Kawamura, S. Seki, T. Abo, Association of intermediate T cell receptor cells, mainly their NK1.1- subset, with protection from malaria, Cell. Immunol. 207 (2001) 28–35. [16] M.K. Mannoor, A. Weerasinghe, R.C. Halder, S.R.M. Morshed, A. Ariyasinghe, H. Watanabe, H. Sekikawa, T. Abo, Resistance to malarial infection is achieved by the cooperation of NK1.1+ and NK1.1- subsets of intermediate TCR cells which are constituents of innate immunity, Cell. Immunol. 211 (2001) 96–104. [17] S. Pied, J. Roland, A. Louise, D. Voegtle, V. Soulard, D. Mazier, P.A. Cazenave, Liver CD4-CD8- NK1.1+ TCRab intermediate cells increase during experimental malaria infection and are able to exhibit inhibitory activity against the parasite liver stage in vitro, J. Immunol. 164 (2000) 1463–1469. [18] A. Molano, S.H. Park, Y.H. Chiu, S. Nosseir, A. Bendelac, M. Tsuji, The IgG response to the circumsporozoite protein is MHC class II-dependent and CD1d-independent: exploring the role of GPIs in NK T cell activation and antimalarial responses, J. Immunol. 164 (2000) 5005–5009. [19] G. Gonzalez-Aseguinolaza, C. De Oliveira, M. Tomaska, S. Hong, O. BrunaRomero, T. Nakayama, M. Taniguchi, A. Bendelac, L. Van Kaer, Y. Koezuka, M. Tsuji, a-Galactosylcer-amide-activated Va14 natural killer T cells mediate protection against murine malaria, Proc. Natl. Acad. Sci. USA 97 (2000) 8461– 8466. [20] Y. Kanda, H. Kawamura, H. Matsumoto, T. Kobayashi, T. Kawamura, T. Abo, Identification and characterization of autoantibody-producing B220low B (B-1) cells appearing in malarial infection, Cell. Immunol. 263 (2010) 49–54. [21] M.K. Mannoor, R.C. Halder, S.R.M. Morshed, A. Ariyasinghe, H.Y. Bakir, H. Kawamura, H. Watanabe, H. Sekikawa, T. Abo, Essential role of extrathymic T cells in protection against malaria, J. Immunol. 169 (2002) 301–306. [22] K. Mannoor, C. Li, M. Inafuku, T. Taniguchi, T. Abo, Y. Sato, H. Watanabe, Induction of ssDNA-binding autoantibody secreting B cell immunity during murine malaria infection is a critical part of the protective immune responses, Immunobiology 218 (2013) 10–20. [23] B.J. Yoder, K.J. Goodrum, Plasmodium chabaudi chabaudi: B-1 cell expansion correlates with semiresistance in BALB/cJ mice, Exp. Parasitol. 98 (2001) 71– 82. [24] K.N. Jhaveri, K. Ghosh, D. Mohanty, B.D. Parmar, R.R. Surati, H.M. Camoens, S.H. Joshi, Y.S. Lyer, A. Desai, S.S. Badakere, Autoantibodies, immunoglobulins, complement and circulating immune complexes in acute malaria, Natl. Med. J. India 10 (1997) 5–7. [25] A.O. Adebajo, P. Charles, R.N. Maini, B.L. Hazleman, Autoantibodies in malaria, tuberculosis and hepatitis B in a west African population, Clin. Exp. Immunol. 92 (1993) 73–76. [26] C. Daniel-Ribeiro, L. Ben Slama, M. Gentilini, Anti-nuclear and anti-smooth muscle antibodies in Caucasians, Africans and Asians with acute malaria, J. Clin. Lab. Immunol. 35 (1991) 109–112. [27] M. Zouali, P. Druilhe, A. Eyquem, IgG-subclass expression of anti-DNA and anti-ribonucleoprotein autoantibodies in human malaria, Clin. Exp. Immunol. 66 (1986) 273–278. [28] S. Boonpucknavig, G. Ekapanyakul, Autoantibodies in sera of Thai patients with Plasmodium falciparum infection, Clin. Exp. Immunol. 58 (1984) 77–82. [29] D. Adu, D.G. Williams, I.A. Quakyi, A. Voller, Y. Anim-Addo, A.A. Bruce-Tagoe, G.B. Johnson, E.J. Holborow, Anti-ssDNA and antinuclear antibodies in human malaria, Clin. Exp. Immunol. 49 (1982) 310–316. [30] H. Watanabe, A. Weerasinghe, C. Miyaji, H. Sekikawa, S. Toyabe, M.K. Mannor, S.R.M. Morshed, R.C. Halder, J. Kobayashi, H. Toma, Y. Sato, K. Iwai, H. Matsuoka, T. Abo, Expansion of unconventional T cells with natural killer markers in malaria patients, Parasitol. Int. 52 (2003) 61–70. [31] R.C. Halder, T. Abe, M.K. Mannoor, S.R.M. Morshed, A. Ariyasinghe, H. Watanabe, H. Kawamura, H. Sekikawa, H. Hamada, Y. Nishiyama, H.

[32] [33]

[34]

[35]

[36]

[37]

[38]

[39] [40] [41]

[42]

[43]

[44]

[45]

[46] [47] [48]

[49]

[50]

[51]

[52]

[53]

[54]

[55]

[56]

Ishikawa, K. Toba, T. Abo, Onset of hepatic erythropoiesis after malarial infection in mice, Parasitol. Int. 52 (2003) 259–268. T. Abo, T. Kawamura, H. Watanabe, Immunologic states of autoimmune diseases, Immunol. Res. 33 (2005) 23–34. A. Ariyasinghe, S.R.M. Morshed, M.K. Mannoor, H.Y. Bakir, H. Kawamura, C. Miyaji, T. Nagura, T. Kawamura, H. Watanabe, H. Sekikawa, T. Abo, Protection against malaria due to innate immunity enhanced by low-protein diet, J. Parasitol. 92 (2006) 531–538. Y. Fujii, H. Kawamura, T. Kawamura, Y. Kanda, H. Matsumoto, T. Kobayashi, T. Yamamoto, T. Aoyama, T. Abo, Co-appearance of autoantibody-producing B220low B cells with NKT cells in the course of hepatic injury, Cell. Immunol. 260 (2010) 105–112. M.F. Good, The implications for malaria vaccine programs if memory T cells from non-exposed humans can respond to malaria antigens, Curr. Opin. Immunol. 3 (1991) 496–502. L.A. Winger, R.E. Sinden, Immunoprotection in mice susceptible to waning memory against the pre-erythrocytic stages of malaria after validated immunization with irradiated sporozoites of Plasmodium berghei, Parasitol. Res. 78 (1992) 427–432. H. Mori, K. Natarajan, B. Betschart, N. Weiss, R.N. Franklin, Polyclonal B-cell activation and autoantibody formation during the course of mosquitotransmitted Plasmodium berghei infection in mice, Trop. Med. Parasitol. 38 (1987) 157–162. O. Simone, M.T. Bejarano, S.K. Pierce, S. Antonaci, M. Wahlgren, M. TroyeBlomberg, D. DonatI, TLRs innate immunereceptors and Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) CIDR1a-driven human polyclonal B-cell activation, Acta Trop. 119 (2011) 144–150. C.T. Daniel-Ribeiro, G. Zanini, Autoimmunity and malaria: what are they dong together?, Acta Trop 76 (2000) 205–221. C.T. Daniel-Ribeiro, Is there a role for autoimmunity in immune protection against malaria?, Mem Inst. Oswaldo Cruz 95 (2000) 199–207. E. Winograd, I.W. Sherman, Naturally occurring anti-band 3 autoantibodies recognize a high molecular weight protein on the surface of Plasmodium falciparum infected erythrocytes, Biochem. Biophys. Res. Commun. 160 (1989) 1357–1363. E. Winograd, J.R. Greenan, I.W. Sherman, Expression of senescent antigen on erythrocytes infected with a knobby variant of the human malaria parasite Plasmodium falciparum, Proc. Natl. Acad. Sci. USA 84 (1987) 1931–1935. H.J. Lustig, V. Nussenzweig, R.S. Nussenzweig, Erythrocyte membraneassociated immunoglobulins during malaria infection of mice, J. Immunol. 119 (1977) 210–216. V. Pradhan, S.S. Badakere, U. Shankarkumar, Y.S. Iyer, K. Ghosh, D. Karnad, Anti-neutrophil cytoplasmic antibodies (ANCA) in malaria, Indian J. Malariol. 39 (2002) 51–59. T.M. Yahya, S. Benedict, A. Shalabi, R. Bayoumi, Anti-neutrophil cytoplasmic antibody (ANCA) in malaria is directed against cathepsin G, Clin. Exp. Immunol. 110 (1997) 41–44. R. Conte, C. Tassi, D. Belletti, F. Ricci, P.L. Tazzari, Autoimmune thrombocytopenia in malaria, Vox Sang. 85 (2003) 221.  rensen, H. Mickley, K.G. Schmidt, Malaria-induced immune P.G. Sø thrombocytopenia, Vox Sang. 47 (1984) 68–72. T. Abo, H. Watanabe, K. Sato, T. Iiai, T. Moroda, K. Takeda, S. Seki, Extrathymic T cells stand at an intermediate phylogenetic position between natural killer cells and thymus-derived T cells, Nat. Immun. 14 (1995) 173–187. A. Tsukahara, S. Seki, T. Iiai, T. Moroda, H. Watanabe, S. Suzuki, T. Tada, H. Hiraide, K. Hatakeyama, T. Abo, Mouse liver T cells: their change with aging and in comparison with peripheral T cells, Hepatology 26 (1997) 301–309. C. Miyaji, H. Watanabe, H. Toma, M. Akisaka, K. Tomiyama, Y. Sato, T. Abo, Functional alteration of granulocytes, NK cells, and natural killer T cells in centenarians, Hum. Immunol. 61 (2000) 908–916. S.R.M. Morshed, K. Mannoor, R.C. Halder, H. Kawamura, M. Bannai, H. Sekikawa, H. Watanabe, T. Abo, Tissue-specific expansion of NKT and CD5+B cells at the onset of autoimmune disease in (NZBNZW)F1 mice, Eur. J. Immunol. 32 (2002) 2551–2561. K. Arai, S. Yamamura, S. Seki, T. Hanyu, H.E. Takahashi, T. Abo, Increase of CD57+ T cells in knee joints and adjacent bone marrow of rheumatoid arthritis (RA) patients: implication for an anti-inflammatory role, Clin. Exp. Immunol. 111 (1998) 345–352. R.C. Halder, T. Kawamura, M. Bannai, H. Watanabe, H. Kawamura, M.K. Mannoor, S.R.M. Morshed, T. Abo, Intensive generation of NK1.1 extrathymic T cells in the liver by injection of bone marrow cells isolated from mice with a mutation of polymorphic major histocompatibility complex antigens, Immunology 102 (2001) 450–459. R. Miyakawa, C. Miyaji, H. Watanabe, H. Yokoyama, C. Tsukada, H. Asakura., T. Abo, Unconventional NK1.1 intermediate TCR cells as major T lymphocytes expanding in chronic graft-versus-host disease, Eur. J. Immunol. 32 (2002) 2521–2531. M. Watanabe, C. Tomiyama-Miyaji, E. Kainuma, M. Inoue, Y. Kuwano, H.W. Ren, J.W. Shen, T. Abo, Role of a-adrenergic stimulus in stress-induced modulation of body temperature, blood glucose and innate immunity, Immunol. Lett. 115 (2008) 43–49. E. Kainuma, M. Watanabe, C. Tomiyama-Miyaji, M. Inoue, Y. Kuwano, H.W. Ren, T. Abo, Association of glucocorticoid with stress-induced modulation of body temperature, blood glucose and innate immunity, Psychoneuroendocrinology 34 (2009) 1459–1468.

Reactivity of autoantibodies against not only erythrocytes but also hepatocytes in sera of mice with malaria.

In order to further examine the reactivity of autoantibodies, mice were infected with a non-lethal strain of Plasmodium yoelii. Parasitemia appeared b...
2MB Sizes 0 Downloads 3 Views