リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

リケラボ 全国の大学リポジトリにある学位論文・教授論文を一括検索するならリケラボ論文検索大学・研究所にある論文を検索できる

大学・研究所にある論文を検索できる 「Hemophagocytosis in visceral leishmaniasis」の論文概要。リケラボ論文検索は、全国の大学リポジトリにある学位論文・教授論文を一括検索できる論文検索サービスです。

コピーが完了しました

URLをコピーしました

論文の公開元へ論文の公開元へ
書き出し

Hemophagocytosis in visceral leishmaniasis

森本, 彩子 東京大学 DOI:10.15083/0002002309

2021.10.13

概要

Visceral leishmaniasis (VL), also known as kala-azar, is caused by parasitic protozoa of the genus Leishmania. Endemic countries of VL include India, Bangladesh, Nepal, Brazil, Ethiopia and Sudan. It is estimated that there are 300,000 new cases of VL and 20,000 deaths annually (WHO, 2012). In sand flies, the insect vector for Leishmania, the parasites develop as promastigotes in the midgut. Once transmitted to mammalian hosts through blood feeding by sand flies, these parasites proliferate as amastigotes within macrophages in the spleen, liver and bone marrow. VL is characterized by clinical manifestations such as fever, weight loss, hepatosplenomegaly and anemia. Anemia often occurs in infectious diseases, whereas the mechanisms of infection-associated anemia may be variable depending on the diseases. In VL, the mechanisms of anemia remained still elusive due to lack of appropriate animal model. I have established a mouse model of VL exhibiting anemia in my master thesis, and using this model it was demonstrated that hemophagocytosis in the spleen of infected mice is a possible cause of anemia during VL. The aim of Ph. D thesis is to examine the contribution of hemophagocytes to anemia during VL, molecular mechanisms of hemophagocytosis during VL and the role of hemophagocytosis in Leishmania survival using an experimental model. First, more detailed analyses on anemia and hemophagocytosis during VL were performed. At 24 weeks of L. donovani infection, BALB/cA mice exhibited splenomegaly, hepatomegaly and anemia. Bone marrow histology and iron status of both infected and uninfected mice were examined but there were no significant histopathological changes and serum iron level or MCV of infected mice were comparable to that of naïve mice. On the other hand, serum indirect bilirubin increased in infected mice. There were no apparent signs of defective erythropoiesis in the infected mice but rather indication of up-regulated hemolysis. Histological analyses on the spleen of infected mice demonstrated macrophages phagocytosing erythrocytes. Autoantibodies to erythrocytes or damages on erythrocyte membrane were not detected, implying those are not related the induced hemophagocytosis. The spleen was the major place for hemophagocytosis when compared with the liver and bone marrow, the other major tissues for parasitization; there, multinucleated giant cells heavily infected with amastigotes were markedly observed and were the major cell type phagocytosing erythrocytes. In addition, hemophagocytosis in the spleen and anemia of the infected mice were improved by treatment of the mice with an anti-leishmanial drug, suggesting that direct infection by parasites is one of the key factors causing hyper-activation of host macrophages to engulf blood cells. On the other hand, Ld-infected nu/nu mice didn’t show anemia and had quite low frequency of hemophagocytosis. From this result, it is suggested that secondary signals from T-cells are also required for the hyper-activation. Taken together, these results suggest that heavy infection of macrophages with Leishmania parasites in the spleen triggers phagocytosis of erythrocytes resulting in anemia during experimental VL. Up-regulated hemophagocytosis occurs in several infectious diseases or familial diseases. However, the detailed mechanisms of infection-associated hemophagocytosis are not fully understood. In order to identify essential factors for generation of hemophagocytosis directly, to reproduce hemophagocytosis in vitro by L. donovani only or in the presence of other factors. In my master thesis, in vitro L. donovani infection-induced hemophagocytosis cell culture model was reproduced. Next, the mechanism of hemophagocytosis during VL was analyzed using this model and mouse model. Contrary to the result from nude mice, without any extra signal, stimulation with L. donovani only caused hemophagocytosis in vitro. Phagocytosis assay for polystyrene beads was performed and phagocytic activity to beads was not up-regulated in Ld-infected macrophages. From this result, it is revealed that hemophagocytosis during VL is not up-regulation of universal phagocytosis. Alteration of gene expression levels in macrophages caused by L. donovani infection was analyzed by RNA-Seq to seek hemophagocytosis-related molecules. Expression levels of genes involved in familial hemophagocytin lymphohistiocytosis were not changed by the infection. Although the results from phagocytic activity assay implied that infected macrophages fail to recognize self RBC correctly, neither the mRNA levels of phagocytosis promoting nor inhibiting gene cluster was significantly changed after infection. Independent analysis of molecules involved in phagocytosis inhibiting was performed and Immunoblot of naïve/infected RAW264.7 demonstrated that SIRPα; one of the phagocytosis inhibiting receptors, was down-regulated in infected macrophages. Immunohistochemistry analysis revealed that in addition to in vitro Leishmania-infected RAW264.7, in MGCs in infected mice, SIRPα expression was also lower than other splenic monocytes/macrophages. Since contrary to down-regulation of SIRPα protein, mRNA level of sirpα is comparable between infected/uninfected macrophages, the mRNA levels of protease family which is responsible for SIRPα proteolysis were examined by RNA-Seq. although Adam10 which is known to proteolyse human SIRPα is up-regulated, Adam17 for mouse SIRPα is rather down-regulated. In order to examine the involvement of these protease to SIRPα proteolysis in infected macrophages, analysis at protein expression level or activity status is needed. From the results, It is suggested that Leishmania donovani infection down-regulate SIRPα in macrophages subsequently misrecognition of RBCs leading to hemophagocytosis by infected macrophages. However, whether if hemophagocytosis is harmful or beneficial to parasite remained elusive. Therefore, the survival of Leishmania in hemophagocytes was compared to those in non-hemophagocytes. By treat RBCs with glutaraldehyde, hemophagocytosis by RAW264.7 were artificially induced. These hemophagocytes and normal RAW264.7 were infected with L. donovani, and after 72h the number of amastigotes in both cells were compared. Both the ratio of infected macrophages with L. donovani in total macrophages and the number of amastigotes per infected macrophage were higher in the artificially induced hemophagocytes. In order to examine if RBCs in infected macrophages were digested and contributed to parasites survival, degradation status of RBCs was also tested on tissue section. Besides intact RBCs, there were the fragmented and colorless RBC or black depositions in MGCs, suggesting RBCs were considered to be degraded after engulfment by MGCs. In order to identify the structure of black depositions, histopathological analysis was performed. Prussian blue staining was performed for ferrous iron, and the depositions were not stained. They were also not stained with Hall staining for biliverdin whereas disappeared by treatment with hydrogen peroxode. As a result, they were suggested to be not ferrous iron, biliverdin or other endogenous pigment (e. g. melanin pigment). In addition, since they have polarization which is corresponded with hemozoin or formalin pigment, the black deposition seems to have similar composition with heme. Next, In order to explore the change in environment of hemophagocytes between naïve RAW264.7 cells affects the survivability of Leishmania, mRNA level of one of the antioxidant molecules, hmox1 was tested using artificially introduced hemophagocytosis assay. In RBC-supplemented and Ld-infected macrophages, the mRNA level of hmox1 was higher than Ld-only infected culture. These results implied that hemophagocytosis is beneficial for Leishmania survival by providing nutrients and low oxidative stress. Together, through the presented study, it was suggested that hemophagocytosis in VL has influence on symptoms of the disease through direct mechanism (i.e., anemia) or through indirect mechanisms by offering Leishmania parasites an incubator for growth. Further studies on this topic may contribute to not only understanding of symptoms other than anemia during VL but also elucidation of hemophagocytosis due to other causes. Moreover, in the era that antileishmanial chemotherapy is the main stream of treatment for VL, this study will offer the alternative choice for disease management by revealing molecular mechanisms of immunopathology for targets of symptom relief drugs.

この論文で使われている画像

参考文献

1. Goto Y, Cheng J, Omachi S, Morimoto A. Prevalence, severity, and pathogeneses of anemia in visceral leishmaniasis. Parasitol Res. 2017 Feb;116(2):457-64.

2. Ogawa S IY, Sofue H, Shiozawa M, Imawari M, Ota k, Ozawa T, Goto H, Chiba T, Hanabusa T, Ban S, Fujita T, Mimori H and Yamamoto K, editor. Internal Medicine. Seventh ed2009.

3. Al-Ghazaly J, Al-Selwi AH, Abdullah M, Al-Jahafi AK, Al-Dubai W, Al-Hashdi A. Pattern of haematological diseases diagnosed by bone marrow examination in Yemen: a developing country experience. Clin Lab Haematol. 2006 Dec;28(6):376-81.

4. Dhingra KK, Gupta P, Saroha V, Setia N, Khurana N, Singh T. Morphological findings in bone marrow biopsy and aspirate smears of visceral Kala Azar: a review. Indian J Pathol Microbiol. 2010 Jan-Mar;53(1):96-100.

5. Elnour IB, Akinbami FO, Shakeel A, Venugopalan P. Visceral leishmaniasis in Omani children: a review. Ann Trop Paediatr. 2001 Jun;21(2):159-63.

6. Cachia EA, Fenech FF. A Review of Kala-Azar in Malta from 1947 to 1962. Trans R Soc Trop Med Hyg. 1964 May;58:234-41.

7. Samanta S, Ghoshal A, Bhattacharya K, Saha B, Walden P, Mandal C. Sialoglycosylation of RBC in visceral leishmaniasis leads to enhanced oxidative stress, calpain-induced fragmentation of spectrin and hemolysis. PLoS One. 2012;7(7):e42361.

8. Pontes De Carvalho LC, Badaro R, Carvalho EM, Lannes-Vieira J, Vinhaes L, Orge G, et al. Nature and incidence of erythrocyte-bound IgG and some aspects of the physiopathogenesis of anaemia in American visceral leishmaniasis. Clin Exp Immunol. 1986 Jun;64(3):495-502.

9. Vilela RB, Bordin JO, Chiba AK, Castelo A, Barbosa MC. RBC-associated IgG in patients with visceral leishmaniasis (kala-azar): a prospective analysis. Transfusion. 2002 Nov;42(11):1442-7.

10. Djoko-Tamnou J, Leclerc C, Modabber F, Chedid L. Studies on visceral Leishmania tropica infection in BALB/c mice. I. Clinical features and cellular changes. Clin Exp Immunol. 1981 Dec;46(3):493-8.

11. Lafuse WP, Story R, Mahylis J, Gupta G, Varikuti S, Steinkamp H, et al. Leishmania donovani infection induces anemia in hamsters by differentially altering erythropoiesis in bone marrow and spleen. PLoS One. 2013;8(3):e59509.

12. Agu WE, Farrell JP, Soulsby EJ. Pathogenesis of anaemia in hamsters infected with Leishmania donovani. Z Parasitenkd. 1982;68(1):27-32.

13. Napier LE, Sharma LR. The Anaemia of the Leishmania-Infected Hamster. Ind Med Gaz. 1933 Dec;68(12):690.

14. Morimoto A, Omachi S, Osada Y, Chambers JK, Uchida K, Sanjoba C, et al. Hemophagocytosis in Experimental Visceral Leishmaniasis by Leishmania donovani. PLoS Negl Trop Dis. 2016 Mar;10(3):e0004505.

15. Szempruch AJ, Sykes SE, Kieft R, Dennison L, Becker AC, Gartrell A, et al. Extracellular Vesicles from Trypanosoma brucei Mediate Virulence Factor Transfer and Cause Host Anemia. Cell. 2016 Jan 14;164(1-2):246-57.

16. Pandey K, Yanagi T, Pandey BD, Mallik AK, Sherchand JB, Kanbara H. Characterization of Leishmania isolates from Nepalese patients with visceral leishmaniasis. Parasitol Res. 2007 May;100(6):1361-9.

17. Katakura K. An experimental challenge model of visceral leishmaniasis by Leishmania donovani promastigotes in mice. Parasitol Int. 2016 Oct;65(5 Pt B):603-6.

18. Zander R, Lang W, Wolf HU. Alkaline haematin D-575, a new tool for the determination of haemoglobin as an alternative to the cyanhaemiglobin method. I. Description of the method. Clin Chim Acta. 1984 Jan 16;136(1):83-93.

19. Alberts B JA, Lewis J, Raff M, Roberts K,, editor. Molecular Biology of THE CELL. fourth ed2004.

20. Huff CG, Nolf LO, Porter RJ, Read CP, Richards AG, Riker AJ, et al. An approach toward a course in the principles of parasitism. J Parasitol. 1958 Feb;44(1):28-45.

21. Stijlemans B, Cnops J, Naniima P, Vaast A, Bockstal V, De Baetselier P, et al. Development of a pHrodo-based assay for the assessment of in vitro and in vivo erythrophagocytosis during experimental trypanosomosis. PLoS Negl Trop Dis. 2015 Mar;9(3):e0003561.

22. Kumakura S, Ishikura H, Kondo M, Murakawa Y, Masuda J, Kobayashi S. Autoimmune-associated hemophagocytic syndrome. Mod Rheumatol. 2004;14(3):205-15.

23. Pilonieta MC, Moreland SM, English CN, Detweiler CS. Salmonella enterica infection stimulates macrophages to hemophagocytose. MBio. 2014;5(6):e02211.

24. Brown DE, McCoy MW, Pilonieta MC, Nix RN, Detweiler CS. Chronic murine typhoid fever is a natural model of secondary hemophagocytic lymphohistiocytosis. PLoS One. 2010;5(2):e9441.

25. Cnops J, De Trez C, Stijlemans B, Keirsse J, Kauffmann F, Barkhuizen M, et al. NK-, NKT- and CD8-Derived IFNgamma Drives Myeloid Cell Activation and Erythrophagocytosis, Resulting in Trypanosomosis-Associated Acute Anemia. PLoS Pathog. 2015 Jun;11(6):e1004964.

26. Chang CS, Chang KP. Heme requirement and acquisition by extracellular and intracellular stages of Leishmania mexicana amazonensis. Mol Biochem Parasitol. 1985 Sep;16(3):267-76.

27. Most J, Neumayer HP, Dierich MP. Cytokine-induced generation of multinucleated giant cells in vitro requires interferon-gamma and expression of LFA-1. Eur J Immunol. 1990 Aug;20(8):1661-7.

28. McNally AK, Anderson JM. Interleukin-4 induces foreign body giant cells from human monocytes/macrophages. Differential lymphokine regulation of macrophage fusion leads to morphological variants of multinucleated giant cells. Am J Pathol. 1995 Nov;147(5):1487-99.

29. DeFife KM, Jenney CR, McNally AK, Colton E, Anderson JM. Interleukin-13 induces human monocyte/macrophage fusion and macrophage mannose receptor expression. J Immunol. 1997 Apr 1;158(7):3385-90.

30. Jordan MB, Hildeman D, Kappler J, Marrack P. An animal model of hemophagocytic lymphohistiocytosis (HLH): CD8+ T cells and interferon gamma are essential for the disorder. Blood. 2004 Aug 1;104(3):735-43.

31. Zoller EE, Lykens JE, Terrell CE, Aliberti J, Filipovich AH, Henson PM, et al. Hemophagocytosis causes a consumptive anemia of inflammation. J Exp Med. 2011 Jun 6;208(6):1203-14.

32. Milner JD, Orekov T, Ward JM, Cheng L, Torres-Velez F, Junttila I, et al. Sustained IL-4 exposure leads to a novel pathway for hemophagocytosis, inflammation, and tissue macrophage accumulation. Blood. 2010 Oct 7;116(14):2476-83.

33. Reed SG, Roters SB, Inverso JA, Jones TC, Goidl EA. Immune responses to T-dependent and T-independent antigens during visceral leishmaniasis in mice: evidence for altered T-cell regulation of immune responses to non-parasite antigens. Cell Immunol. 1985 Nov;96(1):12-25.

34. Stager S, Alexander J, Carter KC, Brombacher F, Kaye PM. Both interleukin-4 (IL-4) and IL-4 receptor alpha signaling contribute to the development of hepatic granulomas with optimal antileishmanial activity. Infect Immun. 2003 Aug;71(8):4804-7.

35. Engwerda CR, Ato M, Cotterell SE, Mynott TL, Tschannerl A, Gorak-Stolinska PM, et al. A role for tumor necrosis factor-alpha in remodeling the splenic marginal zone during Leishmania donovani infection. Am J Pathol. 2002 Aug;161(2):429-37.

36. Omachi S, Fujii W, Azuma N, Morimoto A, Sanjoba C, Matsumoto Y, et al. B-cell activating factor deficiency suppresses splenomegaly during Leishmania donovani infection. Biochem Biophys Res Commun. 2017 Aug 5;489(4):528-33.

37. Fathalla M, Hashim J, Alkindy H, Wali Y. Cerebrospinal fluid involvement in a case of visceral leishmaniasis associated with hemophagocytic lymphohistiocytosis. Sultan Qaboos Univ Med J. 2007 Dec;7(3):253-6.

38. Kilani B, Ammari L, Kanoun F, Ben Chaabane T, Abdellatif S, Chaker E. Hemophagocytic syndrome associated with visceral leishmaniasis. Int J Infect Dis. 2006 Jan;10(1):85-6.

39. Ozyurek E, Ozcay F, Yilmaz B, Ozbek N. Hemophagocytic lymphohistiocytosis associated with visceral leishmaniasis: a case report. Pediatr Hematol Oncol. 2005 Jul-Aug;22(5):409-14.

40. Gagnaire MH, Galambrun C, Stephan JL. Hemophagocytic syndrome: A misleading complication of visceral leishmaniasis in children--a series of 12 cases. Pediatrics. 2000 Oct;106(4):E58.

41. Marom D, Offer I, Tamary H, Jaffe CL, Garty BZ. Hemophagocytic lymphohistiocytosis associated with visceral leishmaniasis. Pediatr Hematol Oncol. 2001 Jan-Feb;18(1):65-70.

42. Bode SF, Bogdan C, Beutel K, Behnisch W, Greiner J, Henning S, et al. Hemophagocytic lymphohistiocytosis in imported pediatric visceral leishmaniasis in a nonendemic area. J Pediatr. 2014 Jul;165(1):147-53 e1.

43. Henter JI, Horne A, Arico M, Egeler RM, Filipovich AH, Imashuku S, et al. HLH-2004: Diagnostic and therapeutic guidelines for hemophagocytic lymphohistiocytosis. Pediatr Blood Cancer. 2007 Feb;48(2):124-31.

44. Tabata R, Tabata C, Terada M, Nagai T. Hemophagocytic syndrome in elderly patients with underlying autoimmune diseases. Clin Rheumatol. 2009 Apr;28(4):461-4.

45. Unal S, Cetin M, Kutlay NY, Elmas SA, Gumruk F, Tukun A, et al. Hemophagocytosis associated with leukemia: a striking association with juvenile myelomonocytic leukemia. Ann Hematol. 2010 Apr;89(4):359-64.

46. Molad Y, Stark P, Prokocimer M, Joshua H, Pinkhas J, Sidi Y. Hemophagocytosis by small cell lung carcinoma. Am J Hematol. 1991 Feb;36(2):154-6.

47. Tsoi WC, Feng CS. Hemophagocytosis by rhabdomyosarcoma cells in bone marrow. Am J Hematol. 1997 Apr;54(4):340-2.

48. Vykuntaraju KN, Avinash T, Saini J, Appaji L. Secondary Hemophagocytic Lymphohistiocytosis Associated with Epstein-Barr Virus. Indian J Pediatr. 2015 Feb 25.

49. La Gruta NL, Kedzierska K, Stambas J, Doherty PC. A question of self-preservation: immunopathology in influenza virus infection. Immunol Cell Biol. 2007 Feb-Mar;85(2):85-92.

50. Mallory FB. A Histological Study of Typhoid Fever. J Exp Med. 1898 Nov 1;3(6):611-38.

51. Balasubramanian S, Kaarthigeyan K, Aparna V, Srinivas S. Tuberculosis associated hemophagocytic syndrome in infancy. Indian Pediatr. 2008 Jul;45(7):593-5.

52. Slovut DP, Benedetti E, Matas AJ. Babesiosis and hemophagocytic syndrome in an asplenic renal transplant recipient. Transplantation. 1996 Aug 27;62(4):537-9.

53. Granert C, Elinder G, Ost A, Henter JI. Kala-azar in a one-year-old Swedish child. Diagnostic difficulties because of active hemophagocytosis. Acta Paediatr. 1993 Sep;82(9):794-6.

54. Nix RN, Altschuler SE, Henson PM, Detweiler CS. Hemophagocytic macrophages harbor Salmonella enterica during persistent infection. PLoS Pathog. 2007 Dec;3(12):e193.

55. Hayashi K, Ohara N, Teramoto N, Onoda S, Chen HL, Oka T, et al. An animal model for human EBV-associated hemophagocytic syndrome: herpesvirus papio frequently induces fatal lymphoproliferative disorders with hemophagocytic syndrome in rabbits. Am J Pathol. 2001 Apr;158(4):1533-42.

56. Sato K, Misawa N, Nie C, Satou Y, Iwakiri D, Matsuoka M, et al. A novel animal model of Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis in humanized mice. Blood. 2011 May 26;117(21):5663-73.

57. Fujiwara F, Hibi S, Imashuku S. Hypercytokinemia in hemophagocytic syndrome. Am J Pediatr Hematol Oncol. 1993 Feb;15(1):92-8.

58. Silva-Herzog E, Detweiler CS. Intracellular microbes and haemophagocytosis. Cell Microbiol. 2008 Nov;10(11):2151-8.

59. Stepp SE, Dufourcq-Lagelouse R, Le Deist F, Bhawan S, Certain S, Mathew PA, et al. Perforin gene defects in familial hemophagocytic lymphohistiocytosis. Science. 1999 Dec 3;286(5446):1957-9.

60. Goransdotter Ericson K, Fadeel B, Nilsson-Ardnor S, Soderhall C, Samuelsson A, Janka G, et al. Spectrum of perforin gene mutations in familial hemophagocytic lymphohistiocytosis. Am J Hum Genet. 2001 Mar;68(3):590-7.

61. Molleran Lee S, Villanueva J, Sumegi J, Zhang K, Kogawa K, Davis J, et al. Characterisation of diverse PRF1 mutations leading to decreased natural killer cell activity in North American families with haemophagocytic lymphohistiocytosis. J Med Genet. 2004 Feb;41(2):137-44.

62. Feldmann J, Callebaut I, Raposo G, Certain S, Bacq D, Dumont C, et al. Munc13-4 is essential for cytolytic granules fusion and is mutated in a form of familial hemophagocytic lymphohistiocytosis (FHL3). Cell. 2003 Nov 14;115(4):461-73.

63. zur Stadt U, Schmidt S, Kasper B, Beutel K, Diler AS, Henter JI, et al. Linkage of familial hemophagocytic lymphohistiocytosis (FHL) type-4 to chromosome 6q24 and identification of mutations in syntaxin 11. Hum Mol Genet. 2005 Mar 15;14(6):827-34.

64. Okuno T, Goto Y, Matsumoto Y, Otsuka H. Applications of recombinant Leishmania amazonensis expressing egfp or the beta-galactosidase gene for drug screening and histopathological analysis. Exp Anim. 2003 Apr;52(2):109-18.

65. Ravetch JV, Lanier LL. Immune inhibitory receptors. Science. 2000 Oct 6;290(5489):84-9.

66. Adams S, van der Laan LJ, Vernon-Wilson E, Renardel de Lavalette C, Dopp EA, Dijkstra CD, et al. Signal-regulatory protein is selectively expressed by myeloid and neuronal cells. J Immunol. 1998 Aug 15;161(4):1853-9.

67. Ashwin H, Seifert K, Forrester S, Brown N, MacDonald S, James S, et al. Tissue and host species-specific transcriptional changes in models of experimental visceral leishmaniasis [version 1; referees: 3 approved, 1 approved with reservations]. Wellcome Open Research. 2018;3(135).

68. Elward K, Gasque P. "Eat me" and "don't eat me" signals govern the innate immune response and tissue repair in the CNS: emphasis on the critical role of the complement system. Mol Immunol. 2003 Sep;40(2-4):85-94.

69. Jaiswal S, Jamieson CH, Pang WW, Park CY, Chao MP, Majeti R, et al. CD47 is upregulated on circulating hematopoietic stem cells and leukemia cells to avoid phagocytosis. Cell. 2009 Jul 23;138(2):271-85.

70. Ishikawa-Sekigami T, Kaneko Y, Okazawa H, Tomizawa T, Okajo J, Saito Y, et al. SHPS-1 promotes the survival of circulating erythrocytes through inhibition of phagocytosis by splenic macrophages. Blood. 2006 Jan 1;107(1):341-8.

71. Okazawa H, Motegi S, Ohyama N, Ohnishi H, Tomizawa T, Kaneko Y, et al. Negative regulation of phagocytosis in macrophages by the CD47-SHPS-1 system. J Immunol. 2005 Feb 15;174(4):2004-11.

72. Inagaki K, Yamao T, Noguchi T, Matozaki T, Fukunaga K, Takada T, et al. SHPS-1 regulates integrin-mediated cytoskeletal reorganization and cell motility. EMBO J. 2000 Dec 15;19(24):6721-31.

73. Kong XN, Yan HX, Chen L, Dong LW, Yang W, Liu Q, et al. LPS-induced down-regulation of signal regulatory protein {alpha} contributes to innate immune activation in macrophages. J Exp Med. 2007 Oct 29;204(11):2719-31.

74. Londino JD, Gulick D, Isenberg JS, Mallampalli RK. Cleavage of Signal Regulatory Protein alpha (SIRPalpha) Enhances Inflammatory Signaling. J Biol Chem. 2015 Dec 25;290(52):31113-25.

75. Shirakabe K, Omura T, Shibagaki Y, Mihara E, Homma K, Kato Y, et al. Mechanistic insights into ectodomain shedding: susceptibility of CADM1 adhesion molecule is determined by alternative splicing and O-glycosylation. Sci Rep. 2017 Apr 10;7:46174.

76. Malemud CJ. Matrix metalloproteinases: role in skeletal development and growth plate disorders. Front Biosci. 2006 May 1;11:1702-15.

77. Schif-Zuck S, Gross N, Assi S, Rostoker R, Serhan CN, Ariel A. Saturated-efferocytosis generates pro-resolving CD11b low macrophages: modulation by resolvins and glucocorticoids. Eur J Immunol. 2011 Feb;41(2):366-79.

78. Nilsson A, Vesterlund L, Oldenborg PA. Macrophage expression of LRP1, a receptor for apoptotic cells and unopsonized erythrocytes, can be regulated by glucocorticoids. Biochem Biophys Res Commun. 2012 Jan 27;417(4):1304-9.

79. Kraal G, Rep M, Janse M. Macrophages in T and B cell compartments and other tissue macrophages recognized by monoclonal antibody MOMA-2. An immunohistochemical study. Scand J Immunol. 1987 Dec;26(6):653-61.

80. Lang T, Ave P, Huerre M, Milon G, Antoine JC. Macrophage subsets harbouring Leishmania donovani in spleens of infected BALB/c mice: localization and characterization. Cell Microbiol. 2000 Oct;2(5):415-30.

81. Kropf P, Freudenberg MA, Modolell M, Price HP, Herath S, Antoniazi S, et al. Toll-like receptor 4 contributes to efficient control of infection with the protozoan parasite Leishmania major. Infect Immun. 2004 Apr;72(4):1920-8.

82. Alberts B JA, Lewis J, Raff M, Roberts K, Walter P. Molecular Biology of The Cell. 2004.

83. Hand WL, King-Thompson NL. Effect of erythrocyte ingestion on macrophage antibacterial function. Infect Immun. 1983 Jun;40(3):917-23.

84. Slater AF. Malaria pigment. Exp Parasitol. 1992 May;74(3):362-5.

85. Das NK, Biswas S, Solanki S, Mukhopadhyay CK. Leishmania donovani depletes labile iron pool to exploit iron uptake capacity of macrophage for its intracellular growth. Cell Microbiol. 2009 Jan;11(1):83-94.

86. Flannery AR, Huynh C, Mittra B, Mortara RA, Andrews NW. LFR1 ferric iron reductase of Leishmania amazonensis is essential for the generation of infective parasite forms. J Biol Chem. 2011 Jul 1;286(26):23266-79.

87. Huynh C, Sacks DL, Andrews NW. A Leishmania amazonensis ZIP family iron transporter is essential for parasite replication within macrophage phagolysosomes. J Exp Med. 2006 Oct 2;203(10):2363-75.

88. Huynh C, Yuan X, Miguel DC, Renberg RL, Protchenko O, Philpott CC, et al. Heme uptake by Leishmania amazonensis is mediated by the transmembrane protein LHR1. PLoS Pathog. 2012;8(7):e1002795.

89. Campos-Salinas J, Cabello-Donayre M, Garcia-Hernandez R, Perez-Victoria I, Castanys S, Gamarro F, et al. A new ATP-binding cassette protein is involved in intracellular haem trafficking in Leishmania. Mol Microbiol. 2011 Mar;79(6):1430-44.

90. Paramchuk WJ, Ismail SO, Bhatia A, Gedamu L. Cloning, characterization and overexpression of two iron superoxide dismutase cDNAs from Leishmania chagasi: role in pathogenesis. Mol Biochem Parasitol. 1997 Dec 1;90(1):203-21.

91. Plewes KA, Barr SD, Gedamu L. Iron superoxide dismutases targeted to the glycosomes of Leishmania chagasi are important for survival. Infect Immun. 2003 Oct;71(10):5910-20.

92. Ghosh S, Goswami S, Adhya S. Role of superoxide dismutase in survival of Leishmania within the macrophage. Biochem J. 2003 Feb 1;369(Pt 3):447-52.

93. Genetu A, Gadisa E, Aseffa A, Barr S, Lakew M, Jirata D, et al. Leishmania aethiopica: strain identification and characterization of superoxide dismutase-B genes. Exp Parasitol. 2006 Aug;113(4):221-6.

94. Ivens AC, Peacock CS, Worthey EA, Murphy L, Aggarwal G, Berriman M, et al. The genome of the kinetoplastid parasite, Leishmania major. Science. 2005 Jul 15;309(5733):436-42.

95. Gobert AP, Verriere T, Asim M, Barry DP, Piazuelo MB, de Sablet T, et al. Heme oxygenase-1 dysregulates macrophage polarization and the immune response to Helicobacter pylori. J Immunol. 2014 Sep 15;193(6):3013-22.

96. Liu G, Friggeri A, Yang Y, Park YJ, Tsuruta Y, Abraham E. miR-147, a microRNA that is induced upon Toll-like receptor stimulation, regulates murine macrophage inflammatory responses. Proc Natl Acad Sci U S A. 2009 Sep 15;106(37):15819-24.

97. Department of Pathology KUSoMDoDP, Keio University Hospital, editor. How to make pathological specimens1968.

98. Mulaw T, Tariku A, Tsegaye AT, Abebe Z. Effect of iron-folic acid supplementation on change of hemoglobin among visceral Leishmaniasis patients in northwest Ethiopia: a retrospective follow up study. BMC Hematol. 2018;18:29.

99. Pham NK, Mouriz J, Kima PE. Leishmania pifanoi amastigotes avoid macrophage production of superoxide by inducing heme degradation. Infect Immun. 2005 Dec;73(12):8322-33.

100. Luz NF, Andrade BB, Feijo DF, Araujo-Santos T, Carvalho GQ, Andrade D, et al. Heme oxygenase-1 promotes the persistence of Leishmania chagasi infection. J Immunol. 2012 May 1;188(9):4460-7.

101. Almeida BFM, Silva KLO, Venturin GL, Chiku VM, Leal AAC, Bosco AM, et al. Induction of haem oxygenase-1 increases infection of dog macrophages by L. infantum. Parasite Immunol. 2017 Dec;39(12).

102. Quintela-Carvalho G, Luz NF, Celes FS, Zanette DL, Andrade D, Menezes D, et al. Heme Drives Oxidative Stress-Associated Cell Death in Human Neutrophils Infected with Leishmania infantum. Front Immunol. 2017;8:1620.

103. Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, Epiphanio S, et al. Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat Med. 2007 Jun;13(6):703-10.

104. Vogl T, Tenbrock K, Ludwig S, Leukert N, Ehrhardt C, van Zoelen MA, et al. Mrp8 and Mrp14 are endogenous activators of Toll-like receptor 4, promoting lethal, endotoxin-induced shock. Nat Med. 2007 Sep;13(9):1042-9.

105. Tsai SY, Segovia JA, Chang TH, Morris IR, Berton MT, Tessier PA, et al. DAMP molecule S100A9 acts as a molecular pattern to enhance inflammation during influenza A virus infection: role of DDX21-TRIF-TLR4-MyD88 pathway. PLoS Pathog. 2014 Jan;10(1):e1003848.

106. Mizobuchi H, Fujii W, Isokawa S, Ishizuka K, Wang Y, Watanabe S, et al. Exacerbation of hepatic injury during rodent malaria by myeloid-related protein 14. PLoS One. 2018;13(6):e0199111.

107. White NJ. Anaemia and malaria. Malar J. 2018 Oct 19;17(1):371.

108. Coombs RR. Cytomegalic inclusion-body disease associated with acquired autoimmune haemolytic anaemia. Br Med J. 1968 Jun 22;2(5607):743-4.

109. Michels K, Nemeth E, Ganz T, Mehrad B. Hepcidin and Host Defense against Infectious Diseases. PLoS Pathog. 2015 Aug;11(8):e1004998.

110. Oldenborg PA, Zheleznyak A, Fang YF, Lagenaur CF, Gresham HD, Lindberg FP. Role of CD47 as a marker of self on red blood cells. Science. 2000 Jun 16;288(5473):2051-4.

111. Sabbatini A, Bombardieri S, Migliorini P. Autoantibodies from patients with systemic lupus erythematosus bind a shared sequence of SmD and Epstein-Barr virus-encoded nuclear antigen EBNA I. Eur J Immunol. 1993 May;23(5):1146-52.

112. Fenderson PG, Fischetti VA, Cunningham MW. Tropomyosin shares immunologic epitopes with group A streptococcal M proteins. J Immunol. 1989 Apr 1;142(7):2475-81.

113. Karlsen AE, Dyrberg T. Molecular mimicry between non-self, modified self and self in autoimmunity. Semin Immunol. 1998 Feb;10(1):25-34.

114. Bull PC, Abdi AI. The role of PfEMP1 as targets of naturally acquired immunity to childhood malaria: prospects for a vaccine. Parasitology. 2016 Feb;143(2):171-86.

参考文献をもっと見る

全国の大学の
卒論・修論・学位論文

一発検索!

この論文の関連論文を見る