• 검색 결과가 없습니다.

To observe the cytokine expression patterns in BV2 cells induced by N.

fowleri ESPs, BV2 cell were treated with N. fowleri ESPs (100 μg/ml) for 12 h and 24 h, and culture supernatant was analyzed by using mouse cytokine array kit. The BV2 cells secreted various cytokines and chemokines (Fig. 4.) The cytokines and chemokines expression were increased time-dependent manners (untreated, ESP treated for 12 and 24 h) (Fig. 4a and 4b). The chemotactic cytokine JE and MIP1- α were secreted from untreated BV2 cells. However, several cytokine such as IFN-γ, IL-1α, IL-1ra and TNF-α secretion were increased from BV2 cell after treated with N. fowleri ESPs for 12 h. In particularly, pro-inflammatory cytokine IL-1α and TNF-α were significantly increased in Nf-ESPs treated BV2 cells for 24 h

23

Fig. 4. Profiles of cytokines and chemokines induced by Nf-ESPs. BV2 cells were either untreated or treated with 100 μg/ml of N. fowleri ESP for 12 and 24 h, respectively. Mouse cytokine array (a) and the graph shows densitometric analysis of mean density (b).

24

D. mRNA expression of cytokine genes

Mouse cytokine array showed that various cytokine and chemokine expressed in BV2 cells after Nf-ESPs treatment. Especially, pro-inflammatory cytokine IL-lα and TNF-α important cytokines in initiation of inflammatory response were detected. To confirm the cytokine expression in the analysis by cytokine array, RT-PCR was performed. RT-PCR analysis suggested that Nf-ESPs strongly induced mRNA expression of IL-1α and TNF-α in BV2 microglial cells (Fig. 5).

The highest expression levels of IL-1α and TNF-α mRNA were identified at 6 h and 3 h after Nf-ESPs treatment, respectively.

25

Fig. 5. IL-1α and TNF-α mRNA expression in BV2 cells by RT-PCR. BV2 cells were treated with Nf-ESPs (100 μg/ml) for 0, 1, 3, 6, 9 (0-9 h). β-actin was used as a control for equal cDNA loading. PCR product were analyzed on 1.5% agarose gel and stained with EtBr (a). The graphs indicate densitometric analysis of IL-1α and TNF-α relative to β-actin, respectively (B) and (c).

26

E. Nf-ESPs induced MAPKs activation in BV2 cells

Mitogen activated protein kinase (MAPKs) are intracellular signal transduction factor, and they mediate pro-inflammatory process. Furthermore, MAPKs regulate cell function such as proliferation, gene expression, mitosis and apoptosis. To analyze the MAPKs signal transduction pathway induced by Nf-ESPs stimulation, phosphorylation of several mitogen-activated protein kinase P38, JNK and ERK were analyzed in BV2 cells treated with Nf-ESPs (100 μg/ml) in a time-dependent manner (Fig. 6a). P38 phosphorylation was increased by treatment of Nf-ESPs for 1 h and the highest level was identified at 3 h (Fig. 6b). The JNK phosphorylation was slightly increased when BV2 cells were treated with Nf-ESPs for 1 h and maximum level of phosphorylation was observed 6 h (Fig. 6c).

Meanwhile, ERK phosphorylation was not clearly identified (Fig. 6d).

27

Fig. 6. Activation of MAPKs (P38, JNK and ERK) in BV2 cells treated with Nf-ESPs. Mouse BV2 cells were treated with Nf-ESPs (100 μg/ml) for varying time periods (0-9 h). The phosphorylation levels of MAPKs were analyzed by western-blot (a). The densitometric analysis graph (b; P38, c; JNK, d; ERK). The graph values were shown phosphorylation /total rate.

28

IV. Discussion

Naegleria fowleri is the causal agent of PAM in experimental animals and humans. Naegleria spp. are widely found in soil and water (Schuster and Visvesvara, 2004). N. fowleri usually propagates in warm, stagnant bodies of freshwater (typically during the summer season), and opportunistically infects the central nervous system via insufflation of water conjugated with the amoeba (CDC, 2008). The amoeba attaches to the olfactory nerve, migrates through the cribriform plate and then reaches to the olfactory bulbs of the forebrain (Cervantes-Sandoval et al., 2008), where it actively multiplies by feeding the nerve tissue. During this stage, usually occurring approximately 3–7 days post-infection, the infected person showed typical symptoms such as parosmia, rapidly progressing anosmia (with resultant ageusia) as the nerve cells of the olfactory bulbs are disrupted and replaced with necrotic lesions.

Microglial cells are the resident macrophages of the brain and spinal cord, and play as first-like immune defense cells in the CNS. The primary role of microglia, involves the engulfing of various materials including cellular debris, lipid apoptotic cell death in the non-inflamed state, and invading virus, bacteria, or other foreign materials in the inflamed state. In addition, microglia also play various roles in the CNS including cytotoxicity for infectious organisms, antigen presenting, synaptic stripping, promotion of repair and extracellular signaling and homeostasis in

non-29

infected region and promoting inflammation infected or damaged tissue (Gehrmann et al., 1995). Extracellular signaling is complicated connected with other microglia, astrocytes, nerves, T-cells and progenitor cells. Microglia activation is induced by IFN-γ, and this activation increases IFN-γ release into the extracellular environment.

The activated microglia induces expressions of several cytokines, and they rapidly activate nearby microglia. Microglia-induced TNF-α causes neural tissue to undergo apoptosis and induces inflammation. Another cytokine IL-1 inhibits IL-10 and TGF-β, which down-regulate antigen presentation and pro-inflammatory signaling. The pro-inflammatory cytokines IL-1α, IL-1β and TNF-α induced by microglia stimulation of CNS, which play a potential role in neurodegeneration when microglia remain in a sustained activated state (Wood, 1998; Aloisi, 2001).

Elucidation of pathogenicity-related factors in PAM is important for understanding the mechanism of N. fowleri-host interactions. Amoebic pathogenicity may consist of complex processes that include both contact-dependent and contact-incontact-dependent mechanisms that lead to host cell death. In contact-dependent mechanism, Nfa1 protein, HSP 70s and Nf-actin, which are closely related with phagocytic food-cup formation, seem to play critical roles (Shin et al., 2001; Song et al., 2006; Sohn et al., 2010). Meanwhile, in a contact-independent mechanism, Nf-ESPs, which consisted of peroxiredoxins, proteases and thrombin receptor, may play a role in host cell invasion and lytic activity for host immunoglobulin and immune evasion (Kim et al., 2009).

30

IL1-β and IL-6 are produced in primary astrocytes stimulated with N. fowleri lysate (Kim et al., 2013). It is also reported that IL1-β, IL-6 and TNF-α were produced in primary microglia co-cultured with N. fowleri trophozoite. IL-18 and IFN-γ were also induced in microglial cell line by N. fowleri lysate (Oh et al., 2005). However, no studies have been on inflammatory responses against Nf-ESPs., In this study, the changes and immune responses in microglia cell line, BV2 cells, treated with Nf-ESPs were analyzed. BV2 cells treatment with Nf-ESPs markedly reduced cell viability in dose dependent manners, which supporting that Nf-ESPs is a significance effector factor for cell damages observed in PAM. The BV2 cells secreted various cytokines and chemokines in response to Nf-ESPs.

Especially, expressions of pro-inflammatory cytokines IL-1α and TNF-α were highly increased. To confirm mRNA expressions of IL-1α and TNF-α, RT-PCR was performed. Both IL-1α and TNF-α expressions was increased in BV2 cells in a time dependent manner by treatment of Nf-ESPs.

IL-1α is the prototypic pro-inflammatory cytokine and effective nearly every cell type, often in concert with another pro-inflammatory cytokine, TNF-α (Dinarello, 1997). Previous study of Leshmaniasis, IL-1 production was induced in monocytes (Crawford et al., 1985). Leshmania mexicana lipophosphoglican (LPG) induced the production of TNF-α, IL-1β, IL-12P70 and IL-10 when human macrophage was stimulated with L. mexicana LPG (Rojas-Bernabe et al., 2014).

Another study showed that IL-1α and TNF-α production increased in human oral

31

and vaginal epithelial cells during Candida albicans infection (Steele and Fidel, 2002). Pro-inflammatory cytokine such as IL-1α, IL-1β and TNF-α mRNA expression increased in primary rat-microglia co-cultured with Acanthamoeba castellanii (Marciano-carbral et al., 2000). In experiment of chlamydia trachomatis, IL-1α was increased in both apical and basolaterial of C. trachomatis infected polarized, immotalized, endocervical epithelial cell model (polA2EN) (Buckner et al., 2013). Some case report of carmeroonians infected by Onchocerca volvulus, observed that IL-1α, IL-6, IL-10 and IL-13 were detected from blood sample by Enzyme-linked immunoabsorbant assay (ELISA) (Nmorsi et al., 2012). In addition, some reports have described that proinflammatory cytokines IL-1, IL-6 and TNF-α level were increased mainly in the aged hippocampus but also in cortical regions (Murray et al., 1997; Katafuchi et al., 2003; Maher et al., 2004; Sierra et al., 2007).

Moreover, the peripheral blood mononuclear cells (PBMC) from parturient and non-pregnant woman was exposed with live tarchyzoites of Toxoplasmma gondii, pro-inflammatory cytokines such as TNF-α and IL-2 was produced significantly higher level (Rezende-Oliveira et al., 2012). Moreover, release of IL-1α, IL-6 and GM-CSF activity were increased in the astroglial cells infected by T. gondii (Fischer et al., 1997). So I suggest that pro-inflammatory cytokine IL-1α and TNF-α may play as an inducer of inflammatory response during the N. fowleri infection.

MAPK are serine/threonine/tyrosine-specific protein kinase belonging to the CDK, MAPK, GSK and CLK kinase group (Manning et al., 2002). MAPKs

32

signaling pathway is essential in regulating diverse cellular processes including inflammatory response, cell differentiation, proliferation and death. MAPKs signaling modulated which mediate extracellular signals into the nucleus to turn on the responsive genes in mammalians cells, including P38, JNK and ERK kinase.

MAPKs phosphorylate a variety of intracellular targets including transcription factors, nuclear pore proteins, membrane transporters, cytoskeletal elements, and other protein kinases. Especially, MAPKs activation was related with pro-inflammatory response in astrocyte (Kim et al., 2013). Activation of MAPKs is also crucial for regulating cytokine expressions. Treatment of Nf-ESPs induced high level expressions of several cytokines, especially IL-1α and TNF-α.

IL-1 is responsible for inflammation as well as the promotion of fever and sepsis. Especially, IL-1α is mainly produce by activated macrophages, neutrophils, epithelial cells and endothelial cells. It plays one of the central roles in the regulation of immune response and possesses metabolic, physiological, haematopoietic activities (Bankers-Fulbright et al., 1996; Dinarello, 1997) and it is on the pathway that activates TNF-α. The role of TNF-α is regulation of immune cells, being an endogenous pyrogen, can induce fever, apoptotic cell death, inflammation and inhibit tumorgenesis. In addition, TNF-α activation closely relate with various human disease including Alzheimer’s disease (Locksley et al., 2001), cancer (Dowlati et al., 2010), major depression (Bryuskov et al., 2002) and inflammatory bowel disease (IBD) (Mikocka-walus et al., 2007). In conclusion,

33

Nf-ESPs induced inflammatory responses in BV2 microglial cells. Increased levels of several cytokines and chemokines were identified and IL-1α and TNF-α were the most predominant cytokines effected by Nf-ESPs. The expression of these cytokines is likely to be regulated by MAPKs signal pathway, but more detailed study on interaction of transcription factors such as AP-1 and NF-κB would be necessary to fully understand the precise mechanism for up-regulation of cytokine production. The result obtained, in this study suggest that Nf-ESPs may play an important role in contact-independent pathogenicity of N. fowleri in PAM.

34

V. Conclusion

To understand contact-independent pathogenesis of N. fowleri in PAM, the change and immune responses in BV2 cells induced by Nf-ESPs were analyzed.

The treatment of Nf-ESPs to BV2 cells markedly reduced cell viability of the cells, which suggesting possible roles of Nf-ESPs in cell damages observed in PAM.

Treatment of Nf-ESPs induced increased expressions of, especially IL-1α and TNF- α, various cytokine and chemokines in BV2 cells. To further understand the mechanism of expression of pro-inflammatory cytokines, MAPK such as P38, JNK and ERK activation were slightly increased by treatment of Nf-ESPs. These results suggested that Nf-ESPs induced productions of pro-inflammatory cytokines, which will further activate inflammation responses in the microglia during the PAM. Therefore, Nf-EPSs can be an important factor in contact-independent pathogenic mechanism of N. fowleri. Considering to Nf-ESPs is a mixture of various proteins secreted by the amoeba, further detailed studies to determine which protein is closely related to the pathogenic mechanism of Nf-ESPs should be needed.

35

References

1. Aichelburg AC, Walochnik J, Assadian O, Prosch H, Steuer A, Perneczky G, Visvesvara GS, Aspock H, Vetter N: Successful treatment of disseminated Acanthamoeba sp. infection with miltefosine. Emerg Infect Dis 14(11): 1743-1746, 2008

2. Aloisi F: Immune function of microglia. Glia 36(2): 165-179, 2001.

3. Bankers-Fulbright JL, Kalli KR, McKean DJ: Interleukin-1 signal transduction.

Life Sci 59(2): 61-83, 1996

4. Bauman RW; "Microbial Diseases of the Nervous System and Eyes".

Microbiology, With Diseases by Body System (2nd ed.). San Francisco:

Pearson Education. p. 617, 2009

5. Brown T: Observations by immunofluorscence microscopy and electron microscopy on the cytopathogenicity of Naegleria fowleri in mouse embryo culutres. J Med Microbiol 12:355-362, 1979

6. Brynskov J, Foegh P, Pedersen G, Ellervik C. Kirkegaard T, Bingham A, Saemark T: Tumour necrosis factor alpha converting enzyme (TACE) activity in the colonic mucosa of patients with inflammatory bowel disease. Gut 51(1):

37-43, 2002

7. Buckner LR, Lewis ME, Greene SJ, Foster TP, Quayle AJ: Chlamydia trachomatis infection results in a modest pro-inflammatory cytokine response

36

and a decrease in T cell chemokine secretion in human polarized endocervical epithelial cells. Cytokine 63(2): 151-165, 2013

8. Carter RF: Description of a Naegleria species isolated from two cases of primary amoebic meningoencephalitis and of the experimental pathological changes induced by it. J Pathol 100: 217-244, 1970

9. Carter RF: Primary amoebic meningo-encephalitis. An appraisal of present knowledge. Trans R Soc Trop Med Hyg 66: 193-213, 1972

10. Carter RF: Primary amoebic meningoencephalitis: clinical, pathological and epidemiological features of six fatal cases. J Pathol Bacteriol 96: 1-25, 1968 11. Centers for Disease Control Prevention (CDC): Primary amebic

meningoencephalitis—Arizona, Florida, and Texas, 2008. MMWR Morb Mortal Wkly Rep 57: 573-577, 2007

12. Cervantes-Sandoval L, Serrano-Luna Jde J, Garcia-Latorre E, Tsutsumi V, Shibayama M: Mucins in the host defence against Naegleria fowleri and mucinolytic activity as a possible means of evasion. Microbiology 154(Pt 12):

3895-3904, 2008

13. Chao CC, Anderson WR, Hu S, Gekker G, Martella A, Peterson PK: Acvated microglia inhibit multiplication of Toxoplasma gondii via a nitric oxide mechanism. Clin Immunol Immunopathol 67: 178-183, 1993

14. Chao CC, Gerkker G, Hu S, Peterson PK: Human microglial cell defense against Toxoplasma gondii. The role of cytokines. J Immunol 152: 1246-1252,

37

1994

15. Chao CC, Hu S, Close K, Choi CS, Molter TW, Novick WJ, Peterson PK:

Cytokine release from microglia: differential inhibition by pentoxifylline and dexamethasone. J Infect Dis 166: 847-853, 1992

16. Chao CC, Hu S, Peterson PK: Glias: the not so innocent bystanders. J Neurovirol 2: 234-239, 1996

17. Crawford GD,Whler DJ, Dinarello CA: Parasite-monocyte interactions in human leishmaniasis: production of interleukin-1 in vitro. J Infect Dis 152(2):

315-322, 1985

18. Culberson GC: The pathogenicity of soil amoebas. Annu Rev Microbiol 25:

231-254, 1971

19. da Rocha-Azevedo B, Tanowitz HB, Marciano-Cabral F. Diagnosis of infections caused by pathogenic free-living amoebae. Interdiscip Perspect Infect Dis 251406, 2009

20. De Jonckheere JF: Characterization of Naegleria species by restriction endonuclease digestion fo whole-cell DNA. Mol Biochem Parasitol 24: 55-66, 1987

21. De Jonckheere JF: Isoenzyme patterns of pathogenic and non-pathogenic Naegleria spp. Using agarose isoelectirc focusing. Ann Microbiol 133(2):

3193-3142, 1982

22. Deetz TR, Sawyer MH, Billman G, Schuster FL, Visvesvara GS: Successful

38

treatment of Balamuthia amoebic encephalitis: presentation of 2 cases. Clinical Infect Dis 37 (10): 1304-1312, 2003

23. Derr-Harf and De Jonckheere JF: Isolation of pathogenic Naegleria australiensis (Amoebida, Vahlkampfidae) from the Rhine. Protistologica 302:

489-494, 1984

24. Dinarello CA: Interleukin-1 and its biologically related cytokines. Adv Immunol 44: 153-205, 1989

25. Dinarello CA: Interleukin-1 in the pathogenesis and treatment of inflammatory diseases. Blood 117(14): 3720-3732, 2011

26. Dinarello CA: Interneukin-1. Cytokine Growth Facor Rev 8(4): 253-265, 1997 27. Dinarello CA: Role of pro- and anti-inflammatory cytokines during

inflammation: experimental and clinical findings. J Biol Regul Homeost Agents 11(3): 91-103, 1997

28. Dingle AD and Fulton C: Development of the flagellar apparatus of Naegleria.

J Cell Biol 31: 43-54, 1966

29. Dowlati Y, Herrmann N, Swardfager W, Liu H, Sham L, Reim EK, Lanctot KL:

A meta-analysis of cytokines in major depression. Biol Psychiatry 67(5): 446-457, 2010

30. Ferrant A and Bates EJ: Elastase in the pathogenic free-living amebae Naegleria and Acanthamoeba spp. Infec Immun 56: 3320-3321, 1988

31. Fischer HG, Nitzgen B, Reichmann G, Hadding U: Cytokine responses

39

induced by Toxoplsma gondii in astrocytes and microglial cells. Eur J Immunol 27(6): 1539-1548, 1997

32. Fulton C: Cell differentiation in Naegleria gruberi. Annu Rev Microbiol 31:

597-629, 1977

33. Gehrmann J, Matsumoto Y, Kreutzberg GW: Microglia: intrinsic immuneffector cell of the brain. Brain Res Brain Res Rev 20(3): 269-287, 1995 34. Hadas E and Mazur T: Characterization of ameboid microglia isolated from

developing mammalian brain. Trop Med Parasitol 44: 197-200, 1993

35. Hu S, Sheng WS, Peterson PK, Chao CC: Cytokine modulation of rat microglial cell superoxide production. Glia 13: 45-50, 1995

36. John DT, Cole TB Jr, Bruner RA: Amebostomes of Naegleria fowleri. J protozool 32: 12-19, 1985

37. John DT, Cole TB Jr, Marciano-Cabral FM: Sucker-like structures on the pathogenic amoeba Naegleria fowleri. Appl Environ Microbiol 47: 12-14, 1984 38. John DT: Primary amebic meningoencephalitis and the biology of Naegleria

fowleri. Annu Rev Microbiol 36: 101-123, 1982

39. Jung SY, Kim JH, Lee YJ, Song KJ, Kim K, Park S, Im KI, Shin HJ: Naegleria fowleri: nfa1 gene knock-down by double-stranded RNAs. Exp Parasitol 118:

208-213, 2008

40. Kaminsky R: Miltefosine Zentaris. Curr Opin Investig Drug 3: 550-554, 2002 41. Katafuchi T, Takaki A, Take S, Kondo T, Yhshimura M: Endotoxin inhibitor

40

blocks heat exposure-induced expression of brain cytokine mRNA in aged rat.

Brain Res Mol Brain Res 118(1-2): 24-32, 2003

42. Khan NA, Jarrol EL, Panjwani N, Cao Z, Paget TA: Protease as markers for differentiation of pathogenic and non-pathogenic species of Acanthamoeba. J Clin Microbiol 43: 391-318, 2000

43. Kim JH, Jung SY, Lee YJ, Song KJ, Kwon D, Kim K, Park S, Im KI, Shin HJ:

Effect of therapeutic chemical agents in vitro and on experimental meningoencephalitis due to Naegleria fowleri. Antimicrob Agents Chemother 52: 4010-4016, 2008

44. Kim JH, Song AR, Sohn HJ, Lee J, Yoo JK, Kwon D, Shin HJ: IL-1beta and IL-6 activate inflammatory response of astrocytes against Naegleria fowleri infection via the modulation of MAPKs and AP-1. Parasite Immunol 35(3-4):

120-128, 2013

45. Kim JH, Yang AH, Sohn HJ, Kim D, Song KJ, Shin HJ: Immunodominant antigens in Naegleria fowleri excretory--secretory proteins were potential pathogenic factors. Parasitol Res 105(6):1675-1681, 2009

46. Lattyak M, Cabral G, Marciano-Cabral F: Scanning electron microscopy of trophozoites of Naegleria species. Proc Elect Microsc Soc Am 43: 642-643, 1985

47. Lether H, Silvany R, Alizadeh H, Huang J, Niederkorn JY: Mannose induce the release of cytophathic factors from Acanthamoeba Keratitis. Infect Immun 66:

41

5-10, 1998

48. Locksley RM, Killeen N, Leonardo MJ: The TNF and TNF receptor superfamilies: integrating mammalian biology. Cell 104(4): 487-501, 2001 49. Ma P, Visvesvara GS, Martinez AJ, Theodore FH, Daggett PM, Sawyer TK:

Naegleria and Acanthamoeba infections: review. Reviews infect dis 12(3): 490-513, 1990

50. Maher FO, Martin DS, Lynch MA: Increased IL-1beta in cortex of aged rats is accompanied by downregulation of ERK and PI-3 kinase. Neurobiol Aging 25(6): 795-806, 2004

51. Manning G, Whyte DB, Martinez R, Hunter T, Sudarsanam S: The portein kinase complement of the human genome. Science 298(5600): 1912-1934, 2002

52. Marciano-Cabral F and Fulford DE: Cytopathology of pathogenic and nonpathogenic Naegleria species for cultured rat neuroblastoma cells. Appl Eviron Microbiol 51: 1133-1137, 1986

53. Marciano-Cabral F and John DT: Cytophathologenicity of Naegleria fowleri for rat neuroblastoma cell cultures: scanning electron microscopy study. Infect Immun 40: 1214-1217, 1983

54. Marciano-Cabral F, MacLean R, Mensah A, LaPat-Polasko L: Identification of Naegleria fowleri in domestic water sources by nested PCR. Appl Envion Microbiol 69: 5864-5869, 2003

42

55. Marciano-Cabral F, Patterson M, John DT, Bradley SG: Cytopathogenicity of Naegleria fowleri and Naegleria gruberi for established mammalian cell cultures. J Parasitol 68: 1110-1116, 1982

56. Marciano-Cabral F, Puffenbarger R, Cabral GA: The increasing importance of Acanthamoeba infections. J Eukaryot Microbiol 47(1): 29-36, 2000

57. Marciano-Cabral F: Biology of Naegleria spp. Microbiol Rev 52: 114-133, 1988

58. Martinez AJ: Free living amebas: natural history, prevention, diagnosis, pathology and treatment of the disease. Boca Raton: CRC Press Inc 156, 1985 59. Martinez DY, Seas C, Bravo F, Legua P, Ramos C. Cabello AM, Gotuzzo E:

Successful treatment of Balamuthia mandrillaria amoebic infection with extensive neurological and cutaneous involvement. Clin Infect Dis 51(2): e7-e11, 2010

60. Mikocka-Walus A, Turnbull DA, Moulding NT, Wilson IG, Andrews JM, Holtmann GJ: Controversies surrounding the comorbidity of depression and anxiety in inflammatory bowel disease patients: a literature review. Inflamm Bowel Dis 13(2): 225-234, 2007

61. Mitra MM, Alzadeh H, Gerard HR, Niederkorn JY: Characterization of plasminogen activator produced by Acanthamoeba castellanii. Mol Biochem Parasitol 73:157-164, 1995

62. Mitro K, Bhagavathiammai A, Zou OM, Bobbett G, McKERROW JH,

43

Chokshi R, Chokshi B, James ER: Partial characterization of the proteolytic secretions of Acanthamoeba polyphaga. Exp Parasitol 78: 377-385

63. Murray-Calderon P and Connolly MA: Interleukin-1 beta inhibits glutamate release in hippocamous of young, but not aged, rats. Neurobiol Aging 18(3):

343-348, 1997

64. Nakajuma K and Kohsaka S: Function roles of microglia in the brain: (Review).

Neurosci Res 17: 187, 1993

65. Nmorsi OP, Nkot BP, Che J: Relationship between pro-and anti-inflammatory cytokines profiles and some haematological parameters in some Cameroonians infected with Onchocerca volvulus. Asian Pac J Trop Med 5(9): 713-717, 2012 66. Oh YH, Jeong SR, Kim JH, Song KJ, Kim K, Park S, Sohn S, Shin HJ:

Cytopathic changes and pro-inflammatory cytokines induced by Naegleria fowleri trophozoites in rat microglial cells and protective effects of an anti-Nfa1 antibody. Parasite Immunol 27(12): 453-459, 2005

Cytopathic changes and pro-inflammatory cytokines induced by Naegleria fowleri trophozoites in rat microglial cells and protective effects of an anti-Nfa1 antibody. Parasite Immunol 27(12): 453-459, 2005

관련 문서