Statins decrease neuroinflammation and prevent cognitive impairment after cerebral malaria

Patricia A Reis, Vanessa Estato, Tathiany I da Silva, Joana C d'Avila, Luciana D Siqueira, Edson F Assis, Patricia T Bozza, Fernando A Bozza, Eduardo V Tibiriça, Guy A Zimmerman, Hugo C Castro-Faria-Neto, Patricia A Reis, Vanessa Estato, Tathiany I da Silva, Joana C d'Avila, Luciana D Siqueira, Edson F Assis, Patricia T Bozza, Fernando A Bozza, Eduardo V Tibiriça, Guy A Zimmerman, Hugo C Castro-Faria-Neto

Abstract

Cerebral malaria (CM) is the most severe manifestation of Plasmodium falciparum infection in children and non-immune adults. Previous work has documented a persistent cognitive impairment in children who survive an episode of CM that is mimicked in animal models of the disease. Potential therapeutic interventions for this complication have not been investigated, and are urgently needed. HMG-CoA reductase inhibitors (statins) are widely prescribed for cardiovascular diseases. In addition to their effects on the inhibition of cholesterol synthesis, statins have pleiotropic immunomodulatory activities. Here we tested if statins would prevent cognitive impairment in a murine model of cerebral malaria. Six days after infection with Plasmodium berghei ANKA (PbA) mice displayed clear signs of CM and were treated with chloroquine, or chloroquine and lovastatin. Intravital examination of pial vessels of infected animals demonstrated a decrease in functional capillary density and an increase in rolling and adhesion of leukocytes to inflamed endothelium that were reversed by treatment with lovastatin. In addition, oedema, ICAM-1, and CD11b mRNA levels were reduced in lovastatin-treated PbA-infected mice brains. Moreover, HMOX-1 mRNA levels are enhanced in lovastatin-treated healthy and infected brains. Oxidative stress and key inflammatory chemokines and cytokines were reduced to non-infected control levels in animals treated with lovastatin. Fifteen days post-infection cognitive dysfunction was detected by a battery of cognition tests in animals rescued from CM by chloroquine treatment. In contrast, it was absent in animals treated with lovastatin and chloroquine. The outcome was similar in experimental bacterial sepsis, suggesting that statins have neuroprotective effects in severe infectious syndromes in addition to CM. Statin treatment prevents neuroinflammation and blood brain barrier dysfunction in experimental CM and related conditions that are associated with cognitive sequelae, and may be a valuable adjuvant therapeutic agent for prevention of cognitive impairment in patients surviving an episode of CM.

Conflict of interest statement

The authors have declared that no competing interests exist.

Figures

Figure 1. Lovastatin treatment prevents contextual and…
Figure 1. Lovastatin treatment prevents contextual and aversive memory impairment after CM.
C57BL/6 mice (n = 12–20/group) were infected with PbA (106 PRBC). As a control, one group was inoculated with the same number of uninfected RBC (n = 6–12/group). Starting on day 6-post infection, uninfected and PbA-infected mice were divided into 2 groups and treated orally with chloroquine (25 mg/kg b.w.), or with the combination of chloroquine/lovastatin for 7 days. On days 15 and 16 post-infection all the animals were submitted to open field training and test sessions (A–B), and to inhibitory avoidance sessions tasks (C–D). Data are expressed as mean ± SEM of crossings (A) and rearings (B) in training (gray bars) and test (black bars) sessions. *Significant difference between groups in training and test sessions (p<0.05, Student's t test). C–D: animals were subjected to a training session in which the latency time on the platform is recorded when an electrical shock is given immediately after the mice step down onto the bars. (C) 1.5 (Short-term memory) and (D) 24 h (long-term memory) aversive memory was tested by recording the latency time on the platform (with a cut-off of 180 sec). Data are expressed as individual values and horizontal lines represent the mean of latency, in seconds; *Significant difference compared with uninfected controls and between infected groups (comparisons among groups were performed by Mann-Whitney U test, p<0.05).
Figure 2. Effect of lovastatin on parasitemia…
Figure 2. Effect of lovastatin on parasitemia and survival rate after infection with PbA (n = 12–20/group).
Data on parasitemia (A) are shown as mean ± SEM. On day 6 post-infection mice were separated in two groups and one received lovastatin (10–40 mg/kg b.w.) daily for 7 days. Comparisons of C57BL6-PbA versus C57BL6-PbA treated animals (*) were significant by Mann-Withney Comparison test. There was no difference in parasitaemia when PbA and PbA-lovastatin treatment were compared (Mann-Withney Comparison test). (B) No differences in survival were observed among the groups (Log-rank (Mantel-Cox) and Gehan-Breslow-Wilcoxon tests).
Figure 3. Lovastatin treatment prevents impairment of…
Figure 3. Lovastatin treatment prevents impairment of short and long-term aversive memory after sepsis.
C57BL/6 mice (n = 5–10/group) were inoculated with fecal supernatant (2.5 mg/g b.w.). For control, one group was inoculated with saline (0.9%). Control and mice challenged with fecal supernatant were divided and one group was treated with lovastatin (20 mg/kg b.w.) 1 hour prior to feces or saline injection respectively, and every 24 h for 3 days. All the animals received imipenem (10 mg/kg b.w.) 6 hours after inoculation and every 24 h for 3 days. On day 15 all animals were subjected to a training session of inhibitory avoidance task, where the latency time on the platform is recorded and an electrical shock is given immediately after the mice step down onto the bars. (A) 1.5 (Short-term memory) and (B) 24 h (long-term memory) aversive memory was then tested by recording the latency time on the platform (with a cut-off of 180 sec). Data are expressed as individual values and horizontal lines represent the mean of latency, in seconds; significant difference compared between saline versus feces injected mice (comparisons among groups were performed by Mann-Whitney U test, *p

Figure 4. Lovastatin improves microvascular function and…

Figure 4. Lovastatin improves microvascular function and decreases leukocyte rolling and adhesion during PbA infection.

Figure 4. Lovastatin improves microvascular function and decreases leukocyte rolling and adhesion during PbA infection.
A–J: representative images of fluorescence intravital microscopy of pial vessels of uninfected mice (A and B; Magnification 100×) or infected mice (C and D) treated with vehicle or lovastatin, respectively (Magnification 40×). Venules with adherent and rolling rhodamine-labeled leukocytes in uninfected mice (E and F) or infected mice (G and H) treated with vehicle or lovastatin, respectively (Magnification 200×). Rhodamine-labeled leukocytes were associated with the venular endothelium in animals treated with vehicle (G) whereas they were largely free in the blood stream in lovastatin-treated mice (H). Fluorescent Plasmodium berghei (GFP)-infected RBC in venules of mice treated with vehicle (I) or lovastatin (J) (Magnification 200×). Mean ± SEM of functional capillary density (K), of rolling-rhodamine labeled leukocytes (L), and of number of adherent leukocytes (M) in pial venules (n = 6/group); *p<0.05 in relation to control (RBC) and vehicle-treated groups (Bonferroni's Multiple Comparison Test) and between PbA and PbA-lovastatin group (Bonferroni's Multiple Comparison Test; Scale bar, 100 µm).

Figure 5. Lovastatin treatment decreases ICAM-1 and…

Figure 5. Lovastatin treatment decreases ICAM-1 and CD11b expression and vascular permeability and induce HMOX-1…

Figure 5. Lovastatin treatment decreases ICAM-1 and CD11b expression and vascular permeability and induce HMOX-1 expression in the brains of mice with CM.
Panels illustrate histological examinations of cerebral cortex on day 6 post-infection, immunostained for ICAM-1 (brown and arrow) and counterstained with hematoxylin-eosin. Brain histology was examined using tissue from the following groups of animals: uninfected (A), uninfected treated with lovastatin (B), PbA-infected (C), and PbA-infected treated with lovastatin (D). Vascular congestion and edema (*) were observed in all PbA-infected mice, but were not seen in controls or treated animals. Scale bar: 50 µm. E: Dose-response relationships for effects of lovastatin on edema formation measured by Evans Blue Dye accumulation in the brain tissue on day 7 post-infection. F–H: ICAM-1, CD11b, and HMOX-1 expression in brain samples evaluated by semi-quantitative PCR on day 6 post-infection; *p

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine…

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine levels in the brains of animals with CM.

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine levels in the brains of animals with CM.
IL-1β (A), TNF-α (B), IL-12 (C) levels were determined by ELISA in brains of mice 6 days after infection with PbA (106 PRBC, n = 6–10/group). Lovastatin (10–40 mg/kg) was orally administered on day 6 post-infection together with chloroquine (25 mg/kg). *p<0.05 or less (Tukey's Multiple Comparison test) in relation to non-infected group and #p<0.05 or less in relation to PbA-infected group (Mann-Withney U Test).

Figure 7. Lovastatin treatment ameliorates oxidative stress…

Figure 7. Lovastatin treatment ameliorates oxidative stress in the brains of mice with CM.

Oxidative…

Figure 7. Lovastatin treatment ameliorates oxidative stress in the brains of mice with CM.
Oxidative stress was assayed by measuring MDA formation (A) and free thiols levels (B) in brains 6 days post-infection of mice with PbA (106 PRBC, n = 10–15/group). Control groups received the same number of uninfected RBC (106). Results are expressed as mean ± S.E.M. and * represents p<0.05 compared to RBC group and # in relation to PbA-infected group, according to Tukey's Multiple Comparison Test.
All figures (7)
Similar articles
References
    1. Kappe SH, Vaughan AM, Boddey JA, Cowman AF (2010) That was then but this is now: malaria research in the time of an eradication agenda. Science 328: 862–866. - PubMed
    1. Rosenthal PJ (2008) Artesunate for the treatment of severe falciparum malaria. N Engl J Med 358: 1829–1836. - PubMed
    1. de Souza JB, Hafalla JC, Riley EM, Couper KN (2010) Cerebral malaria: why experimental murine models are required to understand the pathogenesis of disease. Parasitology 137: 755–772. - PubMed
    1. Golenser J, McQuillan J, Hee L, Mitchell AJ, Hunt NH (2006) Conventional and experimental treatment of cerebral malaria. Int J Parasitol 36: 583–593. - PubMed
    1. Carter JA, Mung'ala-Odera V, Neville BG, Murira G, Mturi N, et al. (2005) Persistent neurocognitive impairments associated with severe falciparum malaria in Kenyan children. J Neurol Neurosurg Psychiatry 76: 476–481. - PMC - PubMed
Show all 78 references
Publication types
MeSH terms
Substances
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Figure 4. Lovastatin improves microvascular function and…
Figure 4. Lovastatin improves microvascular function and decreases leukocyte rolling and adhesion during PbA infection.
A–J: representative images of fluorescence intravital microscopy of pial vessels of uninfected mice (A and B; Magnification 100×) or infected mice (C and D) treated with vehicle or lovastatin, respectively (Magnification 40×). Venules with adherent and rolling rhodamine-labeled leukocytes in uninfected mice (E and F) or infected mice (G and H) treated with vehicle or lovastatin, respectively (Magnification 200×). Rhodamine-labeled leukocytes were associated with the venular endothelium in animals treated with vehicle (G) whereas they were largely free in the blood stream in lovastatin-treated mice (H). Fluorescent Plasmodium berghei (GFP)-infected RBC in venules of mice treated with vehicle (I) or lovastatin (J) (Magnification 200×). Mean ± SEM of functional capillary density (K), of rolling-rhodamine labeled leukocytes (L), and of number of adherent leukocytes (M) in pial venules (n = 6/group); *p<0.05 in relation to control (RBC) and vehicle-treated groups (Bonferroni's Multiple Comparison Test) and between PbA and PbA-lovastatin group (Bonferroni's Multiple Comparison Test; Scale bar, 100 µm).
Figure 5. Lovastatin treatment decreases ICAM-1 and…
Figure 5. Lovastatin treatment decreases ICAM-1 and CD11b expression and vascular permeability and induce HMOX-1 expression in the brains of mice with CM.
Panels illustrate histological examinations of cerebral cortex on day 6 post-infection, immunostained for ICAM-1 (brown and arrow) and counterstained with hematoxylin-eosin. Brain histology was examined using tissue from the following groups of animals: uninfected (A), uninfected treated with lovastatin (B), PbA-infected (C), and PbA-infected treated with lovastatin (D). Vascular congestion and edema (*) were observed in all PbA-infected mice, but were not seen in controls or treated animals. Scale bar: 50 µm. E: Dose-response relationships for effects of lovastatin on edema formation measured by Evans Blue Dye accumulation in the brain tissue on day 7 post-infection. F–H: ICAM-1, CD11b, and HMOX-1 expression in brain samples evaluated by semi-quantitative PCR on day 6 post-infection; *p

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine…

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine levels in the brains of animals with CM.

Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine levels in the brains of animals with CM.
IL-1β (A), TNF-α (B), IL-12 (C) levels were determined by ELISA in brains of mice 6 days after infection with PbA (106 PRBC, n = 6–10/group). Lovastatin (10–40 mg/kg) was orally administered on day 6 post-infection together with chloroquine (25 mg/kg). *p<0.05 or less (Tukey's Multiple Comparison test) in relation to non-infected group and #p<0.05 or less in relation to PbA-infected group (Mann-Withney U Test).

Figure 7. Lovastatin treatment ameliorates oxidative stress…

Figure 7. Lovastatin treatment ameliorates oxidative stress in the brains of mice with CM.

Oxidative…

Figure 7. Lovastatin treatment ameliorates oxidative stress in the brains of mice with CM.
Oxidative stress was assayed by measuring MDA formation (A) and free thiols levels (B) in brains 6 days post-infection of mice with PbA (106 PRBC, n = 10–15/group). Control groups received the same number of uninfected RBC (106). Results are expressed as mean ± S.E.M. and * represents p<0.05 compared to RBC group and # in relation to PbA-infected group, according to Tukey's Multiple Comparison Test.
All figures (7)
Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine…
Figure 6. Lovastatin treatment reduces pro-inflammatory cytokine levels in the brains of animals with CM.
IL-1β (A), TNF-α (B), IL-12 (C) levels were determined by ELISA in brains of mice 6 days after infection with PbA (106 PRBC, n = 6–10/group). Lovastatin (10–40 mg/kg) was orally administered on day 6 post-infection together with chloroquine (25 mg/kg). *p<0.05 or less (Tukey's Multiple Comparison test) in relation to non-infected group and #p<0.05 or less in relation to PbA-infected group (Mann-Withney U Test).
Figure 7. Lovastatin treatment ameliorates oxidative stress…
Figure 7. Lovastatin treatment ameliorates oxidative stress in the brains of mice with CM.
Oxidative stress was assayed by measuring MDA formation (A) and free thiols levels (B) in brains 6 days post-infection of mice with PbA (106 PRBC, n = 10–15/group). Control groups received the same number of uninfected RBC (106). Results are expressed as mean ± S.E.M. and * represents p<0.05 compared to RBC group and # in relation to PbA-infected group, according to Tukey's Multiple Comparison Test.

References

    1. Kappe SH, Vaughan AM, Boddey JA, Cowman AF (2010) That was then but this is now: malaria research in the time of an eradication agenda. Science 328: 862–866.
    1. Rosenthal PJ (2008) Artesunate for the treatment of severe falciparum malaria. N Engl J Med 358: 1829–1836.
    1. de Souza JB, Hafalla JC, Riley EM, Couper KN (2010) Cerebral malaria: why experimental murine models are required to understand the pathogenesis of disease. Parasitology 137: 755–772.
    1. Golenser J, McQuillan J, Hee L, Mitchell AJ, Hunt NH (2006) Conventional and experimental treatment of cerebral malaria. Int J Parasitol 36: 583–593.
    1. Carter JA, Mung'ala-Odera V, Neville BG, Murira G, Mturi N, et al. (2005) Persistent neurocognitive impairments associated with severe falciparum malaria in Kenyan children. J Neurol Neurosurg Psychiatry 76: 476–481.
    1. John CC, Bangirana P, Byarugaba J, Opoka RO, Idro R, et al. (2008) Cerebral malaria in children is associated with long-term cognitive impairment. Pediatrics 122: e92–99.
    1. Zimmerman GA, Castro-Faria-Neto H (2010) Persistent cognitive impairment after cerebral malaria: models, mechanisms and adjunctive therapies. Expert Rev Anti Infect Ther 8: 1209–1212.
    1. Boivin MJ, Bangirana P, Byarugaba J, Opoka RO, Idro R, et al. (2007) Cognitive impairment after cerebral malaria in children: a prospective study. Pediatrics 119: e360–366.
    1. Reis PA, Comim CM, Hermani F, Silva B, Barichello T, et al. (2010) Cognitive dysfunction is sustained after rescue therapy in experimental cerebral malaria, and is reduced by additive antioxidant therapy. PLoS Pathog 6: e1000963.
    1. Langhorne J, Buffet P, Galinski M, Good M, Harty J, et al. (2011) The relevance of non-human primate and rodent malaria models for humans. Malar J 10: 23.
    1. Craig AG, Grau GE, Janse C, Kazura JW, Milner D, et al. (2012) The role of animal models for research on severe malaria. PLoS Pathog 8: e1002401.
    1. Hunt NH, Grau GE, Engwerda C, Barnum SR, van der Heyde H, et al. (2010) Murine cerebral malaria: the whole story. Trends Parasitol 26: 272–274.
    1. Carvalho LJ (2010) Murine cerebral malaria: how far from human cerebral malaria? Trends Parasitol 26: 271–272.
    1. Nie CQ, Bernard NJ, Norman MU, Amante FH, Lundie RJ, et al. (2009) IP-10-mediated T cell homing promotes cerebral inflammation over splenic immunity to malaria infection. PLoS Pathog 5: e1000369.
    1. Ebersoldt M, Sharshar T, Annane D (2007) Sepsis-associated delirium. Intensive Care Med 33: 941–950.
    1. Semmler A, Hermann S, Mormann F, Weberpals M, Paxian SA, et al. (2008) Sepsis causes neuroinflammation and concomitant decrease of cerebral metabolism. J Neuroinflammation 5: 38.
    1. van Gool WA, van de Beek D, Eikelenboom P (2010) Systemic infection and delirium: when cytokines and acetylcholine collide. Lancet 375: 773–775.
    1. Terblanche M, Almog Y, Rosenson RS, Smith TS, Hackam DG (2007) Statins and sepsis: multiple modifications at multiple levels. Lancet Infect Dis 7: 358–368.
    1. Morandi A, Hughes CG, Girard TD, McAuley DF, Ely EW, et al. (2011) Statins and brain dysfunction: a hypothesis to reduce the burden of cognitive impairment in patients who are critically ill. Chest 140: 580–585.
    1. Barichello T, Machado RA, Constantino L, Valvassori SS, Reus GZ, et al. (2007) Antioxidant treatment prevented late memory impairment in an animal model of sepsis. Crit Care Med 35: 2186–2190.
    1. Pino P, Taoufiq Z, Nitcheu J, Vouldoukis I, Mazier D (2005) Blood-brain barrier breakdown during cerebral malaria: suicide or murder? Thromb Haemost 94: 336–340.
    1. Dorovini-Zis K, Schmidt K, Huynh H, Fu W, Whitten RO, et al. (2011) The neuropathology of fatal cerebral malaria in malawian children. Am J Pathol 178: 2146–2158.
    1. Faille D, El-Assaad F, Alessi MC, Fusai T, Combes V, et al. (2009) Platelet-endothelial cell interactions in cerebral malaria: the end of a cordial understanding. Thromb Haemost 102: 1093–1102.
    1. Ley K, Laudanna C, Cybulsky MI, Nourshargh S (2007) Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 7: 678–689.
    1. Hsu M, Muchova L, Morioka I, Wong RJ, Schroder H, et al. (2006) Tissue-specific effects of statins on the expression of heme oxygenase-1 in vivo. Biochem Biophys Res Commun 343: 738–744.
    1. Bu DX, Griffin G, Lichtman AH (2011) Mechanisms for the anti-inflammatory effects of statins. Curr Opin Lipidol 22: 165–170.
    1. Kouroumichakis I, Papanas N, Proikaki S, Zarogoulidis P, Maltezos E (2011) Statins in prevention and treatment of severe sepsis and septic shock. Eur J Intern Med 22: 125–133.
    1. Nath N, Giri S, Prasad R, Singh AK, Singh I (2004) Potential targets of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor for multiple sclerosis therapy. J Immunol 172: 1273–1286.
    1. Barichello T, Fortunato JJ, Vitali AM, Feier G, Reinke A, et al. (2006) Oxidative variables in the rat brain after sepsis induced by cecal ligation and perforation. Crit Care Med 34: 886–889.
    1. Carter JA, Lees JA, Gona JK, Murira G, Rimba K, et al. (2006) Severe falciparum malaria and acquired childhood language disorder. Dev Med Child Neurol 48: 51–57.
    1. Iwashyna TJ, Ely EW, Smith DM, Langa KM (2010) Long-term cognitive impairment and functional disability among survivors of severe sepsis. JAMA 304: 1787–1794.
    1. Sacanella E, Perez-Castejon JM, Nicolas JM, Masanes F, Navarro M, et al. (2011) Functional status and quality of life 12 months after discharge from a medical ICU in healthy elderly patients: a prospective observational study. Crit Care 15: R105.
    1. van der Heyde HC, Nolan J, Combes V, Gramaglia I, Grau GE (2006) A unified hypothesis for the genesis of cerebral malaria: sequestration, inflammation and hemostasis leading to microcirculatory dysfunction. Trends Parasitol 22: 503–508.
    1. Dobbie M, Crawley J, Waruiru C, Marsh K, Surtees R (2000) Cerebrospinal fluid studies in children with cerebral malaria: an excitotoxic mechanism? Am J Trop Med Hyg 62: 284–290.
    1. Souraud JB, Briolant S, Dormoi J, Mosnier J, Savini H, et al. (2012) Atorvastatin treatment is effective when used in combination with mefloquine in an experimental cerebral malaria murine model. Malar J 11: 13.
    1. Bienvenu AL, Picot S (2008) Statins alone are ineffective in cerebral malaria but potentiate artesunate. Antimicrob Agents Chemother 52: 4203–4204.
    1. Wurtz N, Briolant S, Gil M, Parquet V, Henry M, et al. (2010) Synergy of mefloquine activity with atorvastatin, but not chloroquine and monodesethylamodiaquine, and association with the pfmdr1 gene. J Antimicrob Chemother 65: 1387–1394.
    1. Taoufiq Z, Pino P, N'Dilimabaka N, Arrouss I, Assi S, et al. (2011) Atorvastatin prevents Plasmodium falciparum cytoadherence and endothelial damage. Malar J 10: 52.
    1. Greenwood J, Steinman L, Zamvil SS (2006) Statin therapy and autoimmune disease: from protein prenylation to immunomodulation. Nat Rev Immunol 6: 358–370.
    1. Niessner A, Steiner S, Speidl WS, Pleiner J, Seidinger D, et al. (2006) Simvastatin suppresses endotoxin-induced upregulation of toll-like receptors 4 and 2 in vivo. Atherosclerosis 189: 408–413.
    1. Hunt NH, Grau GE (2003) Cytokines: accelerators and brakes in the pathogenesis of cerebral malaria. Trends Immunol 24: 491–499.
    1. Yadava A, Kumar S, Dvorak JA, Milon G, Miller LH (1996) Trafficking of Plasmodium chabaudi adami-infected erythrocytes within the mouse spleen. Proc Natl Acad Sci U S A 93: 4595–4599.
    1. Yoshimoto T, Takahama Y, Wang CR, Yoneto T, Waki S, et al. (1998) A pathogenic role of IL-12 in blood-stage murine malaria lethal strain Plasmodium berghei NK65 infection. J Immunol 160: 5500–5505.
    1. Lou J, Lucas R, Grau GE (2001) Pathogenesis of cerebral malaria: recent experimental data and possible applications for humans. Clin Microbiol Rev 14: 810–820, table of contents.
    1. John CC, Panoskaltsis-Mortari A, Opoka RO, Park GS, Orchard PJ, et al. (2008) Cerebrospinal fluid cytokine levels and cognitive impairment in cerebral malaria. Am J Trop Med Hyg 78: 198–205.
    1. Armah H, Dodoo AK, Wiredu EK, Stiles JK, Adjei AA, et al. (2005) High-level cerebellar expression of cytokines and adhesion molecules in fatal, paediatric, cerebral malaria. Ann Trop Med Parasitol 99: 629–647.
    1. Willimann K, Matile H, Weiss NA, Imhof BA (1995) In vivo sequestration of Plasmodium falciparum-infected human erythrocytes: a severe combined immunodeficiency mouse model for cerebral malaria. J Exp Med 182: 643–653.
    1. McGown CC, Brown NJ, Hellewell PG, Reilly CS, Brookes ZL (2010) Beneficial microvascular and anti-inflammatory effects of pravastatin during sepsis involve nitric oxide synthase III. Br J Anaesth 104: 183–190.
    1. Brown H, Hien TT, Day N, Mai NT, Chuong LV, et al. (1999) Evidence of blood-brain barrier dysfunction in human cerebral malaria. Neuropathol Appl Neurobiol 25: 331–340.
    1. Brown H, Rogerson S, Taylor T, Tembo M, Mwenechanya J, et al. (2001) Blood-brain barrier function in cerebral malaria in Malawian children. Am J Trop Med Hyg 64: 207–213.
    1. Newton CR, Krishna S (1998) Severe falciparum malaria in children: current understanding of pathophysiology and supportive treatment. Pharmacol Ther 79: 1–53.
    1. Millan JM, San Millan JM, Munoz M, Navas E, Lopez-Velez R (1993) CNS complications in acute malaria: MR findings. AJNR Am J Neuroradiol 14: 493–494.
    1. Cordoliani YS, Sarrazin JL, Felten D, Caumes E, Leveque C, et al. (1998) MR of cerebral malaria. AJNR Am J Neuroradiol 19: 871–874.
    1. Looareesuwan S, Wilairatana P, Krishna S, Kendall B, Vannaphan S, et al. (1995) Magnetic resonance imaging of the brain in patients with cerebral malaria. Clin Infect Dis 21: 300–309.
    1. Patankar TF, Karnad DR, Shetty PG, Desai AP, Prasad SR (2002) Adult cerebral malaria: prognostic importance of imaging findings and correlation with postmortem findings. Radiology 224: 811–816.
    1. Penet MF, Viola A, Confort-Gouny S, Le Fur Y, Duhamel G, et al. (2005) Imaging experimental cerebral malaria in vivo: significant role of ischemic brain edema. J Neurosci 25: 7352–7358.
    1. Penet MF, Kober F, Confort-Gouny S, Le Fur Y, Dalmasso C, et al. (2007) Magnetic resonance spectroscopy reveals an impaired brain metabolic profile in mice resistant to cerebral malaria infected with Plasmodium berghei ANKA. J Biol Chem 282: 14505–14514.
    1. Paul R, Angele B, Popp B, Klein M, Riedel E, et al. (2007) Differential regulation of blood-brain barrier permeability in brain trauma and pneumococcal meningitis-role of Src kinases. Exp Neurol 203: 158–167.
    1. Papadopoulos G, Sgouropoulou S, Arnaoutoglou E, Petrou A (2002) Postoperative hypoxaemia in a patient with patent foramen ovale. Eur J Anaesthesiol 19: 152–154.
    1. Lu D, Goussev A, Chen J, Pannu P, Li Y, et al. (2004) Atorvastatin reduces neurological deficit and increases synaptogenesis, angiogenesis, and neuronal survival in rats subjected to traumatic brain injury. J Neurotrauma 21: 21–32.
    1. Lu D, Qu C, Goussev A, Jiang H, Lu C, et al. (2007) Statins increase neurogenesis in the dentate gyrus, reduce delayed neuronal death in the hippocampal CA3 region, and improve spatial learning in rat after traumatic brain injury. J Neurotrauma 24: 1132–1146.
    1. Lu D, Mahmood A, Goussev A, Schallert T, Qu C, et al. (2004) Atorvastatin reduction of intravascular thrombosis, increase in cerebral microvascular patency and integrity, and enhancement of spatial learning in rats subjected to traumatic brain injury. J Neurosurg 101: 813–821.
    1. Tapia-Perez JH, Sanchez-Aguilar M, Torres-Corzo JG, Gordillo-Moscoso A, Martinez-Perez P, et al. (2008) Effect of rosuvastatin on amnesia and disorientation after traumatic brain injury (NCT003229758). J Neurotrauma 25: 1011–1017.
    1. Hunt NH, Stocker R (2007) Heme moves to center stage in cerebral malaria. Nat Med 13: 667–669.
    1. Tenhunen R, Marver HS, Schmid R (1968) The enzymatic conversion of heme to bilirubin by microsomal heme oxygenase. Proc Natl Acad Sci U S A 61: 748–755.
    1. Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, et al. (2007) Heme oxygenase-1 and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat Med 13: 703–710.
    1. Stocker R, Perrella MA (2006) Heme oxygenase-1: a novel drug target for atherosclerotic diseases? Circulation 114: 2178–2189.
    1. Ali F, Zakkar M, Karu K, Lidington EA, Hamdulay SS, et al. (2009) Induction of the cytoprotective enzyme heme oxygenase-1 by statins is enhanced in vascular endothelium exposed to laminar shear stress and impaired by disturbed flow. J Biol Chem 284: 18882–18892.
    1. Irwin S (1968) Comprehensive observational assessment: Ia. A systematic, quantitative procedure for assessing the behavioral and physiologic state of the mouse. Psychopharmacologia 13: 222–257.
    1. Lackner P, Beer R, Heussler V, Goebel G, Rudzki D, et al. (2006) Behavioural and histopathological alterations in mice with cerebral malaria. Neuropathol Appl Neurobiol 32: 177–188.
    1. Greenwood J, Walters CE, Pryce G, Kanuga N, Beraud E, et al. (2003) Lovastatin inhibits brain endothelial cell Rho\ediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitis. FASEB J 17: 905–907.
    1. Levasseur JE, Wei EP, Raper AJ, Kontos AA, Patterson JL (1975) Detailed description of a cranial window technique for acute and chronic experiments. Stroke 6: 308–317.
    1. Araujo CV, Estato V, Tibirica E, Bozza PT, Castro-Faria-Neto HC, et al. (2012) PPAR gamma activation protects the brain against microvascular dysfunction in sepsis. Microvasc Res 84: 218–21.
    1. Carvalho-Tavares J, Hickey MJ, Hutchison J, Michaud J, Sutcliffe IT, et al. (2000) A role for platelets and endothelial selectins in tumor necrosis factor-alpha-induced leukocyte recruitment in the brain microvasculature. Circ Res 87: 1141–1148.
    1. Sabino B, Lessa MA, Nascimento AR, Rodrigues CA, Henriques MG, et al. (2008) Effects of antihypertensive drugs on capillary rarefaction in spontaneously hypertensive rats: intravital microscopy and histologic analysis. J Cardiovasc Pharmacol 51: 402–409.
    1. Rozen S, Skaletsky H (2000) Primer3 on the WWW for general users and for biologist programmers. Methods Mol Biol 132: 365–386.
    1. Draper HH, Hadley M (1990) Malondialdehyde determination as index of lipid peroxidation. Methods Enzymol 186: 421–431.
    1. Puhl H, Waeg G, Esterbauer H (1994) Methods to determine oxidation of low-density lipoproteins. Methods Enzymol 233: 425–441.

Source: PubMed

3
Abonnieren