• 검색 결과가 없습니다.

Age-related Aβ deposition and Aβ-associated gliosis in the cerebral cortex of

In transgenic mice carrying AD-linked APP and PS1 mutations, we observed a rapid accumulation of fibrillar Aβ from 6 months of age up to the oldest age studied, 17 months (Fig. 1 A). By immunohistochemical study using Iba1 antibody, we investigated microglial activation in the cerebral cortex of APP/PS1 transgenic mouse at the different time points (3, 6, 10, 14 and 17 months). An aggregate of activated microglia were increased synchronously with Aβ deposition. GFAP-immunopositive reactive astrocytes were also observed in parallel with Aβ deposition. At 14-17 months, microglia and astrocytes were morphologically damaged (Fig. 1 B). Iba1-positive microglia and GFAP-positive astrocyte were abundant and closely associated with fibrillar (thioflavin S-positive) Aβ deposits throughout the cortex (Fig. 1 C).

10

11

Fig. 1. Age-related Aβ deposition and Aβ-associated gliosis in the cerebral cortex of APP/PS1 mice. Tissue sections were obtained from cerebral cortex of APP/PS1 transgenic mouse at the different time points (3, 6, 10, 14 and 17 months). (A) Representative images of thioflavine S staining in the cerebral cortex. Scale bar: 200 μm.

(B) Immunohistochemistry staining with antibodies to Iba1 for microglia and GFAP for astrocyte at the indicated time point. Scale bar: 20 μm. (C) Iba1-positive microglia and GFAP-positive astrocyte immunoreactivity (red) showed in the area surrounding the plaque (thioflavine S, green) at 10 months in the APP/PS1 mice. Scale bar: 20 μm.

12

B. Expression of pro-inflammatory mediators and IL-10 in the cerebral cortex of APP/PS1 mice

To elucidate the mechanisms involved in Aβ-mediated inflammation, we investigated the expression of IL-1β, TNF-α, iNOS and IL-10 in the APP/PS1 transgenic mouse. The level of mRNA and protein expression was detected in brain sections from cortex regions at various postnatal ages ranging from 3 to 17 months. The mRNA levels of IL-1β, TNF-α and iNOS were detected from 3 months, and there was increased with the increasing age.

The mRNA of IL-10 was detectable in 6 month-old transgenic mouse, which is 3 months later than the expression of pro-inflammatory mediators (Fig. 2 A, C). The protein levels of IL-1β, TNF-α, iNOS and IL-10 were also similarly reflected at the mRNA expression. The protein levels of IL-1β, TNF-α and iNOS were detected from 3 months, and there was increased with age. The protein levels of IL-10 were detectable in 6 month-old transgenic mouse, which is 3 months later than the expression of pro-inflammatory mediators (Fig. 2 B, D).

13

Fig. 2. Expression of pro-inflammatory mediators and IL-10 in the cerebral cortex of APP/PS1 mice. Tissue lysates were obtained from cerebral cortex of APP/PS1 transgenic mouse at the different time points (3, 6, 8, 10, 12, 14 and 17 months). (A) mRNA expression of IL-1β, TNF-α, iNOS and IL-10 in the cerebral cortex. (B) Western blot analysis of IL-1β, TNF-α, iNOS and IL-10 expression. (C) Intensity of PCR bands was quantified using ImageQuant software (*p<0.01). Differences among means ± standard error of means (S.E.M.) were analyzed using ANOVA followed by Tukey-Kramer multiple comparisons test (n=3-5). (D) Intensity of western bands was quantified using ImageQuant software (*p<0.01). Differences among means ± standard error of means (S.E.M.) were analyzed using ANOVA followed by Tukey-Kramer multiple comparisons test (n=3-5).

14

C. IL-1β, TNF-α and IL-10 expression in microglia

To identify the specific cell types expressing TNF-α, IL-1β and IL-10 in the cerebral cortex, double immunofluorescence staining with Iba1 for microglia and TNF-α, IL-1β, IL-10 (Fig. 3 A) or GFAP for astrocyte and IL-1β, TNF-α, IL-10 (Fig. 3 B) was performed at 3, 6, 10, 14 and 17 months. Fluorescence images from each channel of the double-labeled sections were merged. The results showed that IL-1β, TNF-α and IL-10 expression was localized mainly in Iba1-immunopositive microglia.

15

16

17

Fig. 3. IL-1β, TNF-α and IL-10 expression in microglia. Double-immunofluorescence staining with antibodies to (A) Iba1 for microglia and IL-1β, TNF-α and IL-10, (B) GFAP for astrocyte and IL-1β, TNF-α and IL-10 at the different time points (3, 6, 10, 14 and 17 months). Images from the double-labeled tissue were merged. Scale bar: 10 μm.

18

D. ROS production and neuronal damage in the cerebral cortex of APP/PS1 mice 1-42 was found to generate ROS through NADPH oxidase, leading to neurodegeneration in rat brain in vivo (Rosales-Corral et al., 2004) and in vitro (Abramov et al., 2007). Thus, we investigated whether fibrillar Aβ mediates the production of ROS in the cerebral cortex of transgenic mouse model of AD (APP/PS1). To examine this possibility, we performed in situ analysis of ROS production by hydroethidine histochemistry (Wu et al., 2003). The fluorescent products of oxidized hydroethidine were detected from 6 months, and there were significantly increased between the ages of 14 months and 17 months (Fig. 4 A). Recent studies have demonstrated that NADPH oxidase is a significant source of ROS in Alzhimer’s disease (Wilkinson et al., 2006).

Double-immunofluorescence staining was performed with a combination of antibodies against Iba1 and gp91phox or GFAP and gp91phox. gp91phox protein was localized to Iba1-immunopositive microglia in the cerebral cortex of 14 month-old APP/PS1 mice (Fig. 4 B). Next, we have investigated the neuronal damage in the APP/PS1 transgenic mouse model of AD. Cerebral cortex of 14, 17-month-old APP/PS1 transgenic mice showed significant decreases in NeuN-positive neurons and MAP2-immunoreactive dendrites (Fig. 4 C).

19

20

Fig. 4. ROS production and neuronal damage in the cerebral cortex of APP/PS1 mice. (A) In situ visualization of O2- and O2--derived oxidant production. Animals were injected with hydroethidine at 3, 6, 8, 10, 12, 14 and 17 months. Confocal micrographs show ethidium fluorescence (red). Nuclei were counterstained with Hocheset33258 (blue).

Scale bar: 100 μm. (B) Double-immunofluorescence staining with antibodies to Iba1 for microglia and gp91phox at 14 months. Images from the double-labled tissue were merged.

Scale bar: 10 μm. (C) Immunohistochemistry staining with antibodies to NeuN (neuronal nuclei) and MAP2 (microtubule associated protein 2) for neuron at the different time points (3, 6, 8, 10, 12, 14 and 17 months). Scale bar: 50 μm., 100 μm.

21

. D

ISCUSSION

Microglia and astrocyte are a key component of the inflammatory response inthe brain and are associated with Aβ plaques in Alzheimer'sdisease (AD). Although there is evidence that glial activationis important for the pathogenesis of AD, the role of gliain cerebral amyloidosis remains obscure. The present study wasundertaken to investigate the temporal relationship between Aβ deposition and glial activation in the cerebral cortex of APP/PS1 transgenic mice. In transgenic mice, we showed accumulation of fibrillar Aβ from 6 months of age up to the oldest age studied, 17 months. Activated microglia and astrocytes increased synchronously with Aβ deposits and were abundant and closely associated with senile plaques. At 14-17 months, microglia and astrocytes were morphologically damaged and excessively activated (Fig. 1). Recent studies showed that despite the capability of microglia to degrade amyloid deposits, microglial cells are unable to effectively reduce the Aβ deposition in the brains of APP23 transgenic mice and human AD patients (Bornemann et al., 2001; Fiala et al., 2005).

Previous studies indicate the potentially toxic effects of activated microglia, due to their production of neurotoxic substances, such as glutamate (Piani et al., 1991; Piani et al., 1992), nitric oxide (Chao et al., 1992; Dawson et al., 1994), superoxide anion (Thery et al., 1991), and cytokines with neurotoxic actions (Banati et al., 1993). Pro-inflammatory cytokines such as IL-1β and TNF-α are over-expressed in activated microglia surrounding plaques in AD (McGeer et al., 2001). They can then also stimulate the production of Aβ peptides (Blasko et al., 1999). The balance between pro- and

anti-22

inflammatory cytokines determines the magnitude of the inflammatory response. IL-10 is known as an inhibitor of the synthesis of pro-inflammatory cytokine, including TNF-α and IL-1β (Bogdan et al., 1992; Wang et al., 1994; Bethea et al., 1999; Sawada et al., 1999). There is evidence that IL-10 has an anti-inflammatory role in the brain (Strle et al., 2001; Lee et al., 2007) and therefore might be important in down-regulating inflammatory processes associated whit AD pathology (Remarque et al., 2001). Of relevance to AD, IL-10 has been shown in vitro to inhibit Aβ-induced cytokine production in differentiated THP-1 monocytes and primary murine microglia (Szczepanik et al., 2001). We examined the endogenous expression of IL-10 in the cerebral cortex of APP/PS1 mice, but did not find of an association between IL-10 expression and down-regulation of brain inflammation. A notable feature is that the expression of IL-10 was detectable in 6 month-old transgenic mouse, which is 3 months later than expression of pro-inflammatory mediators (Fig. 2). Double immunoflorescence staining showed that IL-1β, TNF-α and IL-10 expression was localized mainly in Iba1-immunopositive microglia (Fig. 3). These results suggest that IL-10 production act as a negative feedback regulation in pro-inflammatory mediators expression.

In the present study, reactive oxygen species (ROS) production was detected from 6 months, and there significantly increased between the ages of 14 months and 17 months.

A major subunit of NADPH oxidase, gp91phox protein was localized to Iba1-immunopositive microglia in the APP/PS1 transgenic mice. Additionally, cerebral cortex of 14, 17 months showed significant decrease in NeuN-positive neurons and MAP2-immunoreactice dendrites (Fig 4). These results suggest that activated microglia

23

surrounding plaques induce excessive expression of pro-inflammatory mediators and ROS production, leading to neuronal damage.

24

. CONCLUSION

This study showed that the activated microglia surrounding plaques induced excessive expression of pro-inflammatory mediators such as IL-1β, TNF-α, iNOS and ROS, leading to neuronal damage in the cerebral cortex of APP/PS1 mice. Our previous study demonstrated that LPS-induced endogenous expression of IL-10 in microglia down-regulated brain inflammation and neuronal damage in the rat cerebral cortex. However, in the present study, we did not find of an association between IL-10 expression and down-regulation of brain inflammation. Therefore, further understanding the mechanisms of IL-10 expression in chronic brain inflammation may be an important point to comprehend the pathogenesis of Alzheimer’s disease.

25

REFERENCES

1. Abramov AY, Scorziello A, Duchen MR: Three distinct mechanisms generate oxygen free radicals in neurons and contribute to cell death during anoxia and reoxygenation.

J Neurosci 27: 1129-1138, 2007

2. Akiyama H, Barger S, Barnum S, Bradt B, Bauer J, Cole GM, Cooper NR, Eikelenboom P, Emmerling M, Fiebich BL, Finch CE, Frautschy S, Griffin WS, Hampel H, Hull M, Landreth G, Lue L, Mrak R, Mackenzie IR, McGeer PL, O'Banion MK, Pachter J, Pasinetti G, Plata-Salaman C, Rogers J, Rydel R, Shen Y, Streit W, Strohmeyer R, Tooyoma I, Van Muiswinkel FL, Veerhuis R, Walker D, Webster S, Wegrzyniak B, Wenk G, Wyss-Coray T: Inflammation and Alzheimer's disease. Neurobiol Aging 21: 383-421, 2000

3. Alpar A, Ueberham U, Bruckner MK, Seeger G, Arendt T, Gartner U: Different dendrite and dendritic spine alterations in basal and apical arbors in mutant human amyloid precursor protein transgenic mice. Brain Res 1099: 189-198, 2006

4. Bachis A, Colangelo AM, Vicini S, Doe PP, De Bernardi MA, Brooker G, Mocchetti I: Interleukin-10 prevents glutamate-mediated cerebellar granule cell death by blocking caspase-3-like activity. J Neurosci 21: 3104-3112, 2001

26

5. Banati RB, Gehrmann J, Schubert P, Kreutzgerg GW: Cytotoxicity of microglia. Glia 7: 111-118, 1993

6. Bethea JR, Nagashima H, Acosta MC, Briceno C, Gomez F, Marcillo AE, Loor K, Green J, Dietrich WD: Systemically administered interleukin-10 reduces tumor necrosis factor-alpha production and significantly improves functional recovery following traumatic spinal cord injury in rats. J Neurotrauma 16: 851-863, 1999

7. Blasko I, Marx F, Steiner E, Hartmann T, Grubeck-Loebenstein B: TNFalpha plus IFNgamma induce the production of Alzheimer beta-amyloid peptides and decrease the secretion of APPs. Faseb J 13: 63-68, 1999

8. Bogdan C, Paik J, Vodovotz Y, Nathan C: Contrasting mechanisms for suppression of macrophage cytokine release by transforming growth factor-beta and interleukin-10. J Biol Chem 267: 23301-23308, 1992

9. Boje KM, Arora PK: Microglial-produced nitric oxide and reactive nitrogen oxides mediate neuronal cell death. Brain Res 587: 250-256, 1992

10. Bornemann KD, Wiederhold KH, Pauli C, Ermini F, Stalder M, Schnell L, Sommer B, Jucker M, Staufenbiel M: Aβ-Induced Inflammatory Processes in Microglia Cells of APP23 Transgenic Mice. Am J Pathol 158: 63-73, 2001

27

11. Chao CC, Hu S, Molitor TW, Shaskan EG, Peterson PK: Activated microglia mediate neuronal cell injury via a nitric oxide mechanism. J Immunol 149: 2736-2741, 1992

12. Chao CC, Hu S, Peterson PK: Glia, cytokines, and neurotoxicity. Crit Rev Neurobiol 9: 189-205, 1995

13. Dawson VL, Brahmbhatt HP, Mong JA, Dawson TM: Expression of inducible nitric oxide synthase causes delayed neurotoxicity in primary mixed neuronal-glial cortical cultures. Neuropharmacology 33: 1425-1430, 1994

14. Dickson DW, Lee SC, Mattiace LA, Yen SH, Brosnan C: Microglia and cytokines in neurological disease, with special reference to AIDS and Alzheimer's disease. Glia 7:

75-83, 1993

15. Dietrich WD, Busto R, Bethea JR: Postischemic hypothermia and IL-10 treatment provide long-lasting neuroprotection of CA1 hippocampus following transient global ischemia in rats. Exp Neurol 158: 444-450, 1999

16. Fiala M, Liu QN, Sayre J, Pop V, Brahmandam V, Graves MC, Vinters HV:

Cyclooxygenase-2-positive macrophages infiltrate the Alzheimer's disease brain and damage the blood-brain barrier. Eur J Clin Invest 32: 360-371, 2002

28

17. Fiala M, Lin J, Ringman J, Kermani-Arab V, Tsao G, Patel A, Lossinsky AS, Graves MC, Gustavson A, Sayre J, Sofroni E, Suarez T, Chiappelli F, Bernard G: Ineffective phagocytosis of amyloid-β by macrophages of Alzheimer's disease patients. J Alzheimers Dis 7(3): 221-232; discussion 255-262, 2005

18. Fiorentino DF, Zlontnik A, Mossman TR, Howard M O’Garra A: IL-10 inhibits cytokine production by activated macrophages. J Immunol 147: 3815–3822, 1991

19. Gebicke-Haerter PJ, Spleiss O, Ren LQ, Li H, Dichmann S, Norgauer J, Boddeke HW: Microglial chemokines and chemokine receptors. Prog Brain Res 132: 525-532, 2001

20. Glezer I, Simard AR, Rivest S: Neuroprotective role of the innate immune system by microglia. Neuroscience 147: 867-883, 2007

21. Gonzalez-Scarano F, Baltuch G: Microglia as mediators of inflammatory and degenerative diseases. Annu Rev Neurosci 22: 219-240, 1999

22. Goodwin JL, Uemura E, Cunnick JE: Microglial release of nitric oxide by the synergistic action of beta-amyloid and IFN-gamma. Brain Res 692: 207-214, 1995

23. Grilli M, Barbieri I, Basudev H, Brusa R, Casati C, Lozza G, Ongini E:

Interleukin-29

10modulates neuronal threshold of vulnerability to ischaemic damage. Eur J Neurosci 12: 2265-2272, 2000

24. Haas J, Storch-Hagenlocher B, Biessmann A, Wildemann B: Inducible nitric oxide synthase and argininosuccinate synthetase: co-induction in brain tissue of patients with Alzheimer's dementia and following stimulation with beta-amyloid 1-42 in vitro.

Neurosci Lett 322: 121-5, 2002

25. Ii M, Sunamoto M, Ohnishi K, Ichimori Y: beta-Amyloid protein-dependent nitric oxide production from microglial cells and neurotoxicity. Brain Res 720: 93-100, 1996

26. Jankowsky JL, Slun HH, Gonzales V, Jenkins NA, Copeland NG, Borchelt DR: APP processing and amyloid deposition in mice haplo-insufficient for presenilin 1.

Neurobiol Aging 25: 885-892, 2004

27. Klegeris A, Walker DG, McGeer PL: Activation of macrophages by Alzheimer beta amyloid peptide. Biochem Biophys Res Commun 199: 984-991, 1994

28. Klegeris A, McGeer PL: beta-amyloid protein enhances macrophage production of oxygen free radicals and glutamate. J Neurosci Res 49: 229-235, 1997

30

29. Koshinaga M, Katayama Y, Fukushima M, Oshima H, Suma T, Takahata T: Rapid and widespread microglial activation induced by traumatic brain injury in rat brain slices.

J Neurotrauma 17: 185-192, 2000

30. Kremlev SG, Palmer C: Interleukin-10 inhibits endotoxin-induced proinflammatory cytokines in microglial cell cultures. J Neuroimmunol 162: 71-80, 2005

31. Lee HG, Park KW, Jin BK, Lee YB: Interleukin-10 endogenously expressed in microglia prevents lipopolysaccharide-induced neurodegeneration in the rat cerebral cortex in vivo. Exp Mol Med 39: 812-819, 2007

32. Liao YF, Wang BJ, Cheng HT, Kuo LH, Wolfe MS: Tumor necrosis factor-alpha, interleukin-1beta, and interferon-gamma stimulate gamma-secretase-mediated cleavage of amyloid precursor protein through a JNK-dependent MAPK pathway. J Biol Chem 279: 49523-49532, 2004

33. Mallat M, Chamak B: Brain macrophages: neurotoxic or neurotrophic effector cells?

J Leukoc Biol 56: 416-422, 1994

34. Martino G: How the brain repairs itself: new therapeutic strategies in inflammatory and degenerative CNS disorders. Lancet Neurol 3: 372-378, 2004

31

35. McGeer PL, McGeer EG: Inflammation, autotoxicity and Alzheimer disease.

Neurobiol Aging 22: 799-809, 2001

36. Meda L, Cassatella MA, Szendrei GI, Otvos L, Jr, Baron P, Villalba M, Ferrari D, Rossi F: Activation of microglial cells by beta-amyloid protein and interferon-gamma.

Nature 374: 647-650, 1995

37. Mizuno T, Sawada M, Marunouchi T, Suzumura A: Production of IL-10 by mouse glial cells in culture. Biochem Biophys Res Comm 205: 1907-1915, 1994

38. Molina-Holgado E, Vela JM, Arevalo-Martin A, Guaza C: LPS/IFN-gamma cytotoxicity in oligodendroglial cells: role of nitric oxide and protection by the anti-inflammatory cytokine IL-10. Eur J Neurosci 13: 493-502, 2001

39. Morgan JH, Gamblin TC, Adkins JR, Groves JR, Dalton ML, Ashley DW:

Norepinephrine is a more potent inhibitor of tumor necrosis factor over a range of doses than dopamine. Am Surg 70: 526-528, 2004

40. Mrak RE, Griffin WS: Interleukin-1 and the immunogenetics of Alzheimer disease. J Neuropathol Exp Neurol 59: 471-476, 2000

41. Nguyen MD, Julien JP, Rivest S: Innate immunity: the missing link in neuroprotection

32

and neurodegeneration? Nat Rev Neurosci 3: 216-227, 2002

42. Perry VH, Bolton SJ, Anthony DC, Betmouni S: The contribution of inflammation to acute and chronic neurodegeneration. Res Immunol 149: 721-725, 1998

43. Piani D, Frei K, Do KQ, Cuenod M, Fontana A: Murine brain macrophages induced NMDA receptor mediated neurotoxicity in vitro by secreting glutamate. Neurosci Lett 133: 159-162, 1991

44. Piani D, Spranger M, Frei K, Schaffner A, Fontana A: Macrophage-induced cytotoxicity of N-methyl-D-aspartate receptor positive neurons involves excitatory amino acids rather than reactive oxygen intermediates and cytokines. Eur J Immunol 22: 2429-2436, 1992

45. Polazzi E, Gianni T, Contestabile A: Microglial cells protect cerebellar granule neurons from apoptosis: evidence for reciprocal signaling. Glia 36: 271-280, 2001

46. Remarque EJ, Bollen EL, Weverling-Rijnsburger AW, Laterveer JC, Blauw GJ, Westendorp RG: atients with Alzheimer's disease display a pro-inflammatory phenotype. Exp Gerontol 36: 171-176, 2001

47. Rogers J, Cooper NR, Webster S, Schultz J, McGeer PL, Styren SD, Civin WH,

33

Brachova L, Bradt B, Ward P: Complement activation by beta-amyloid in Alzheimer disease. Proc Natl Acad Sci U S A 89: 10016-10020, 1992

48. Rosales-Corral S, Tan DX, Reiter RJ, Valdivia-Velazquez M, Acosta-Martinez JP, Ortiz GG: Kinetics of the neuroinflammation-oxidative stress correlation in rat brain following the injection of fibrillar amyloid-beta onto the hippocampus in vivo. J Neuroimmunol 150: 20-28, 2004

49. Ryu JK, Kim J, Choi SH, Oh YJ, Lee YB, Kim SU, Jin BK: ATP-induced in vivo neurotoxicity in the rat striatum via P2 receptors. Neuroreport 13: 1611-1615, 2002

50. Sawada M, Suzumura A, Hosoya H, Marunouchi T, Nagatsu T: Interleukin-10 inhibits both production of cytokines and expression of cytokine receptors in microglia. J Neurochem 72: 1466-1471, 1999

51. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, Hu K, Huang J, Johnson-Wood K, Khan K, Kholodenko D, Lee M, Liao Z, Lieberburg I, Motter R, Mutter L, Soriano F, Shopp G, Vasquez N, Vandevert C, Walker S, Wogulis M, Yednock T, Games D, Seubert P: Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400: 173-177, 1999

52. Selkoe DJ, Schenk D: Alzheimer's disease: molecular understanding predicts

34

amyloid-based therapeutics. Annu Rev Pharmacol Toxicol 43: 545-584, 2003

53. Strauss S, Bauer J, Ganter U, Jonas U, Berger M, Volk B: Detection of interleukin-6 and alpha 2-macroglobulin immunoreactivity in cortex and hippocampus of Alzheimer's disease patients. Lab Invest 66: 223-230, 1992

54. Strle K, Zhou JH, Shen WH, Broussard SR, Johnson RW, Freund GG, Dantzer R, Kelley KW: Interleukin-10 in the brain. Crit Rev Immunol 21: 427-449, 2001

55. Szczepanik AM, Funes S, Petko W, Ringheim GE: IL-4, IL-10 and IL-13 modulate A beta (1-42)-induced cytokine and chemokine production in primary murine microglia and a human monocyte cell line. J Neuroimmunol 113: 49-62, 2001

56. Thery C, Chamak B, Mallat M: Cytotoxic Effect of Brain Macrophages on Developing. Eur J Neurosci 3: 1155-1164, 1991

57. Tsunawaki S, Sporn M, Ding A, Nathan CF: Deactivation of macrophages by transforming growth factor β. Nature 334: 260–262, 1988

58. Van der Wal EA, Gomez-Pinilla F, Cotman CW: Transforming growth factor-beta 1 is in plaques in Alzheimer and down pathologies. Neuroreport 4: 69-72, 1993

35

59. Van Muiswinkel FL, Veerhuis R, Eikelenboom P: Amyloid beta protein primes cultured rat microglial cells for an enhanced phorbol 12-myristate 13-acetate-induced respiratory burst activity. J Neurochem 66: 2468-2476, 1996

60. Vila M, Jackson-Lewis V, Guegan C, Wu DC, Teismann P, Choi DK, Tieu K, Przedborski S: The role of glial cells in Parkinson's disease. Curr Opin Neurol 14:

483-489, 2001

61. Wang L, Goillot E, Tepper RI: IL-10 inhibits alloreactive cytotoxic T lymphocyte generation in vivo. Cell Immunol 159: 152-169, 1994

62. Wilkinson BL, Landreth GE: The microglial NADPH oxidase complex as a source of oxidative stress in Alzheimer's disease. J Neuroinflammation 3: 30, 2006

63. Wu DC, Teismann P, Tieu K, Vila M, Jackson-Lewis V, Ischiropoulos H, Przedborski S: NADPH oxidase mediates oxidative stress in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. Proc Natl Acad Sci U S A 100: 6145-6150, 2003

64. Xia MQ, Hyman BT: Chemokines/chemokine receptors in the central nervous system and Alzheimer's disease. J Neurovirol 5: 32-41, 1999

36

- 국문요약 -

현이 Iba1-면역양성을 보이는 마이크로글리아에서 대부분 배치됨을 관찰하였 다. 활성산소종 (ROS) 생성은 8개월부터 나타나며 14개월과 17개월 사이에서 두드러지게 증가되었다. NADPH oxidase의 주요 구성물질인 gp91phox 단백질 은 Iba1-면역양성을 보이는 마이크로글리아에서 대부분 배치됨을 관찰하였다.

또한, 14개월과 17개월의 대뇌피질에서 NeuN-양성적 뉴론과 MAP2-면역반 응적인 수지상 돌기가 상당히 감소되어 있음을 관찰하였다. 이 결과들은 알츠 하이머병 뇌조직에서 섬유성 아밀로이드 베타 (fibrillar β-amyloid)의 축적에

또한, 14개월과 17개월의 대뇌피질에서 NeuN-양성적 뉴론과 MAP2-면역반 응적인 수지상 돌기가 상당히 감소되어 있음을 관찰하였다. 이 결과들은 알츠 하이머병 뇌조직에서 섬유성 아밀로이드 베타 (fibrillar β-amyloid)의 축적에

관련 문서