Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Sep 22.
Published in final edited form as: Drugs Future. 2009 Apr 1;34(4):333–340. doi: 10.1358/dof.2009.034.04.1358595

SOLUBLE CD40 LIGAND IN DEMENTIA

B Giunta 1,*, KP Figueroa 2, T Town 2,3,4, J Tan 5
PMCID: PMC2748392  NIHMSID: NIHMS137345  PMID: 19777117

Abstract

Some 15–20% of the population over the age of 65 years suffer from dementia, currently one of the leading causes of death behind cardiovascular diseases, cancer and cerebrovascular diseases. The major forms of dementia share in common overactivation of the CD40-CD40-L complex, leading to high levels of proinflammatory cytokine production by immune cells of the central nervous system (CNS), including microglia and astrocytes. Consequently, both neuronal survival and signaling are negatively affected, leading to the characteristic progressive loss of higher cortical functions. We have reviewed the literature concerning the involvement of this complex in the pathology of three major forms of dementia: Alzheimer's-type, HIV-associated and vascular dementia. This is followed by a discussion of current preclinical and clinical therapies that may influence this interaction, and thus point the way toward a future neuroimmunological approach to inhibiting the effects of CD40-CD40-L in neuropsychiatric disease.

INTRODUCTION

As the number of elderly individuals continues to rapidly increase, dementia has led to a massive socioeconomic burden which is projected to worsen. Specifically, some 15–20% of the population over the age of 65 years suffer from dementia (1, 2). Its presentation is heterogeneous, as it is caused by multiple disorders. Alzheimer's disease (AD) and vascular dementia (VAD) are the two main causes, affecting 25–45% and 15–35%, respectively, of all patients suffering from dementia (3). Among dementias where brain infectious viruses are etiological, HIV-1-associated dementia (HAD) is the most common cause of dementia (4).

All three of these disorders share a common neuropathological hallmark: microglial activation associated with increased CD40 pathway signaling. The interaction of membrane-bound CD40 (expressed by innate immune cells, including microglia) with its cognate CD40 ligand (CD40-L) plays a critical role in microglial phenotypic transformation from a ramified (resting) to an activated macrophage morphology (5), and enhances the surface expression of inflammatory markers (6, 7). These cell-surface molecules in turn enhance the inflammatory cycle. Conversely, low levels of molecules involved in antigen presentation, including CD40, are expressed by resting microglia (6, 8). CD40 is a 45- to 50-kDa type I integral membrane glycoprotein and a member of the tumor necrosis factor receptor (TNFR) superfamily. Landmark studies originally identified CD40 on B lymphocytes, where it was shown to mediate T-cell-dependent B-cell activation and differentiation. It is found on most immune and some nonimmune cells, including macrophages, dendritic cells, endothelial cells, smooth muscle cells and B cells, as well as astrocytes and microglia (9-15). CD40-L, also known as CD154, is a trimeric 33-kDa type II membrane glycoprotein that is predominantly expressed by activated T and B cells, astrocytes and platelets (13, 16, 17). Importantly, CD40-L also occurs in a soluble, secreted form (sCD40-L) that retains biological activity to bind and activate membrane-bound CD40 (18, 19).

It is now generally accepted that the CD40-CD40-L interaction is central to the pathophysiology of risk factor-related vascular damage (20, 21). Cerebral perfusion abnormalities are seen in the earliest phases of AD (22) and are a well-established hallmark of VAD. This is not surprising, as the majority (> 95%) of circulating CD40-L is bound to platelets. After activation, CD40-L is rapidly translocated to the platelet surface and is then cleaved, giving rise to sCD40-L over a period of minutes to hours (20). Additionally, circulating sCD40-L is an independent predictor of restenosis following percutaneous transluminal angioplasty (23). The clinical association between sCD40-L and VAD is also strengthened by evidence of sCD40-L engagement of CD40 on endothelial cells, resulting in various proinflammatory responses, including the expression of adhesion molecules and tissue factors, the release of cytokines/ chemokines and the expression of vessel-remodeling metalloproteinases (24, 25). The interaction between sCD40-L or CD40-L and CD40 activates various signaling cascades via the TNFR-associated factor (TRAF) family members (26). As sCD40-L also negatively affects vascular endothelial cell functions (24, 27), it has even further potential to interfere with microcirculation, increasing the risk of VAD development.

HIV-ASSOCIATED DEMENTIA

HIV-1 enters the central nervous system (CNS) early after infection, persists in the brain compartment for decades and generates clinical symptoms that include behavioral, motor and cognitive deficits in a significant proportion of infected patients (28). Together these clinical features are referred to as HIV-associated dementia, or HAD. The severity of HAD is strongly correlated with the number of activated macrophages and microglia within the basal ganglia and frontal lobes (29). Indeed, the hallmarks of HAD are HIV-1-infected macrophages and microglia in the brain, the formation of syncytial, multinucleated giant cells (30-32), perivascular infiltrates of inflammatory cells, astrogliosis and neuronal damage and/or loss - especially in the hippocampus, basal ganglia and cerebral cortex (4).

The introduction of highly active antiretroviral therapy (HAART) in the mid-90s dramatically reduced mortality associated with HIV-1 infection (32); however, it fails to eradicate the virus (33). Furthermore, many HAART drugs fail to adequately penetrate the CNS - likely allowing virus to replicate in the brain (4). The use of HAART has resulted in a significant decline in the incidence of HAD, but the prevalence has actually risen as HIV-infected individuals experience longer lifespans (34). Currently, more subtle forms of CNS disease, known as minor cognitive motor disorder (MCMD), have become more common in HIV-1 patients receiving HAART (35).

Productive CNS infection by HIV occurs primarily in perivascular macrophages and microglia, but not neurons. Accordingly, neuronal damage and death occur via indirect pathways relying on the intrinsic toxicity of secreted viral proteins and the excessive release of inflammatory mediators by infected/activated cells (36). Two such inflammatory mediators that work in synergy are sCD40-L and the HIV-1 transactivating protein Tat (37, 38). Both play important roles in HAD and HIV-related CNS disorders because of their ability to chemoattract specific subsets of leukocytes that may be infected, and because they activate resident CNS cells (37, 39-41), which in turn fuel excessive release of inflammatory mediators. Along these lines, Sui et al. demonstrated that CD40-L synergizes with Tat protein, causing elevated TNF-α production by human monocytes and microglia. This occurred via Tat upregulation of CD40 expression on these cells, sensitizing them to CD40-L. Studies by this same group on the neurotoxicity of Tat/CD40-L-exposed monocyte conditioned medium (CM) revealed a significantly higher level of neurotoxicity than CM from monocytes exposed to Tat or CD40-L alone. Furthermore, immunodepletion of TNF-α from CM nearly completely mitigated neurotoxicity (38).

Untreated HIV-1 infection is also associated with an elevated release of soluble markers for both endothelial and platelet activation, including sCD40-L (38, 40, 42-44). Others include the possible HIV-1 neurotoxin platelet-activating factor (PAF) (45), as well as soluble VCAM and von Willebrand factor (42, 43). HAART has been shown to reduce the latter two markers in HIV-1-infected patients (46, 47), but to have no effect on soluble platelet activation markers, including PAF acetylhydrolase (48) and sCD40-L (44).

Similar to HIV-1 Tat protein, sCD40-L also interacts with the chemokine IL-8 (also known as CXCL8). IL-8 is a chemotactic activating factor for primarily neutrophils (49, 50), and is also a chemoattractant and activating factor for T cells (51) and monocytes (52). IL-8 expression is increased in HAD brain tissue and plasma compared with uninfected tissue (53, 54), colocalizes with CD68/CD40-positive cells in HAD brain tissue, and stimulates HIV-1 replication in macrophages and T cells (55, 56). Soluble CD40-L is integral to the actions of IL-8, as microglia only secrete it after ligation of surface CD40 with CD40-L (55). Furthermore, sCD40-L is present at elevated levels in the plasma and cerebrospinal fluid (CSF) of HIV-1-infected individuals with cognitive impairment compared to HIV-1-positive subjects with normal cognitive function (38). Moreover, in at least one study, HAART administration has been positively associated with an increase in serum sCD40-L levels (40). Perhaps these phenomena may contribute to the increased prevalence of HIV-1-related cognitive impairment in the era of HAART.

ALZHEIMER'S DISEASE

Deposition of β-amyloid peptide (Aβ) into senile plaques and intracellular accumulation of neurofibrillary tangles (principally composed of phosphorylated tau protein) are the hallmark features of Alzheimer's disease (AD), the most common form of dementia (57, 58). Also, cerebrovascular dysregulation likely contributes to the development of the neurodegeneration that characterizes this dementing illness (59). As in HAD, a continuous inflammatory cycle exists in the AD brain, marked by chronic, low-level secretion of proinflammatory cytokines and acute-phase reactants around amyloid deposits (60). The inflammatory response is largely mounted by reactive glia, principally microglia, which are in an activated state surrounding “senile” plaques that are largely comprised of deposited 40- to 42-amino-acid Aβ peptides. We and others have demonstrated that microglia are activated at a low level by Aβ exposure in vitro, and that costimulation with interferon gamma (IFN-γ) or CD40-L synergistically affects microglial activation (61, 62). CD40 protein induction on microglia and vascular endothelial cells occurs in the presence of low levels of more soluble forms of Aβ, suggesting that this may occur prior to Aβ deposition (62, 63).

The pattern of expression of both CD40 and CD40-L is altered in the brains of AD patients, as well as in several animal models of AD (17, 64). It has recently been suggested that sCD40-L could be a potential AD biomarker, thereby allowing early identification of subjects requiring treatment. Specifically, it was found that patients with plasma sCD40-L concentrations of ≥ 6.0 ng/mL experience a 3-fold greater risk of suffering cognitive decline in the following 2 years (65). Importantly the subjects in this study were free of any drugs that could potentially affect sCD40-L, including statins, antioxidants and/or anti-inflammatory agents. Because of the close risk association between sCD40-L and both AD and vascular disease (66), the study could not completely exclude the possibility that silent brain infarcts could have occurred in some patients and contributed to a worse AD clinical presentation (67).

Aβ-induced CD40 is inserted into the cell membrane and is constitutively expressed at low levels on vascular smooth muscle and endothelial cells (63, 66, 68). Thus, not surprisingly, the risk relationship between sCD40-L and AD mirrors the positive correlation between sCD40-L levels and vascular disease, as well as sCD40-L levels and the risk of cognitive impairment in HIV infection. Furthermore, endothelial cells are a key component of the blood-brain barrier, suggesting that there could be alterations of this physiological unit under conditions of increased sCD40-L expression or function. Accordingly, it was previously demonstrated that Aβ increases both CD40 mRNA transcription and CD40 protein expression in cultured human vascular endothelial cells within 48 h of exposure (63, 68). Interestingly, these findings further suggest that factors which increase the risk for vascular disease also increase the risk for AD (69, 70). For example, brain ischemia induced by infarction is associated with worse cognitive decline in AD (71), and alterations in cerebral blood flow precede the onset of the disease (72). Such findings suggest that the role of platelet-derived sCD40-L should be considered in the context of AD neuropathogenesis. This concept is supported by findings from Sevush et al., which showed multiple abnormalities in platelet activation in AD patients (73). Taken together, the soluble or membrane-bound CD40-L and CD40 on smooth muscle and endothelial cells seem to work with Aβ peptides to promote a feed-forward, self-perpetuating inflammatory cascade in both the vascular wall and brain parenchyma, which in turn may initiate AD pathogenesis and/or propagate its progression (63, 68, 74, 75).

A second route by which sCD40-L may modulate AD pathogenesis is via T cells, which, like microglia, are increased in number and demonstrate altered activated profiles both in the circulation and brain parenchyma of AD patients (76). Activated T cells are one of the key sources of membrane-bound CD40-L and may also shed sCD40-L, and it has been shown that intercellular adhesion molecule 1 (ICAM-1) facilitates the migration of T lymphocytes across the brain endothelium (77). Ligation of endothelial cell CD40 by sCD40-L may upregulate ICAM-1 (78), pointing to a mechanism by which sCD40-L derived from brain-penetrating T lymphocytes might also promote neuroinflammation (76).

Finally, we and others have shown that the CD40-CD40-L complex also acts synergistically with Aβ peptide to promote proinflammatory activation of microglia, leading to brain inflammation and neuronal injury. Specifically, when cultured microglia are coincubated with Aβ peptide and CD40-L in the presence of primary neurons, the microglia synergistically secrete TNF-α, resulting in neuronal death (62). Furthermore, the expression of genes involved in amyloid precursor protein (APP) processing and tau phosphorylation is disturbed in cultured human microglia following challenge with CD40-L (62, 79-81). Laporte et al. demonstrated that mice deficient in CD40 and carrying the human “Swedish” mutant APP (APPsw; a mutant form of APP that increases the production of Aβ) show significantly less microgliosis, astrocytosis and Aβ load compared to CD40-sufficient littermates (82). This work validates earlier work in our laboratory that demonstrated that anti-CD40-L treatment causes a reduction in cerebral Aβ pathology and improved performance in several cognitive tasks in the double transgenic PSAPP mouse model expressing mutant human presenilin-1 and human APPsw (unpublished results). Further supporting this line of evidence, a CD40-L-neutralizing antibody reduced AD-like pathology and improved cognition in the double transgenic PSAPP AD mouse model (82, 83). Specifically, Todd Roach et al. showed that PSAPP mice given a regimen of anti-CD40-L antibody commencing at an age when initial Aβ deposition occurs had superior spatial and nonspatial memory compared to control PSAPP mice (83).

FUTURE APPLICATIONS FOR SCD40-L AS A BIOMARKER AND TREATMENT TARGET FOR DEMENTIA

Biomarker and therapeutic target

Many groups, with the goal of identifying objective quantitative markers of neuropsychiatric illnesses, have measured CSF cytokines in various psychiatric conditions. These types of studies have been plagued by problems of reproducibility, as the literature reporting intrathecal or circulating cytokine levels in such diseases is often discordant. This could be due to the relatively small sample sizes used, the nature of cytokine activity (characterized by a short half-life) and signaling that occurs mainly in an autocrine or paracrine fashion. Additionally, cytokines can be buffered by soluble receptors in the periphery, which bind to the cytokine and can therefore obscure detection. Although the initial event(s) triggering the neurodegenerative processes in VAD, AD, HAD and other dementing illness are likely different, it is reasonable to hypothesize that such event(s) may converge on a common cascade of proinflammatory cytokine production in response to neurodegenerative changes, whether it be from a ruptured vascular plaque, HIV-1 proteins or neuritic Aβ plaques. The cytokines released in the CNS may, in turn, act to amplify certain neuropathological events. However, it is worth noting that a common cytokine release pattern “footprint” in the CSF has yet to be identified for different neurodegenerative diseases (84).

Another approach that we present here is to measure peripheral levels of proinflammatory molecules such as sCD40-L as an index of CNS changes associated with neurodegenerative diseases. A recent study by Ray et al. took an unbiased proteomics approach to screen 120 proteins and identified a panel of 18 markers (many of which mediate immune/inflammatory events) that correctly identify AD approximately 90% of the time (85). Along these lines, the finding of increased peripheral sCD40-L levels in HAD and AD could potentially be of clinical relevance as a biomarker, as the assessment of sCD40-L plasma levels might allow for early identification of those at risk for cognitive impairment, such that therapy could be initiated prior to the onset of full-blown disease.

Regarding the focus on the CD40 pathway as a therapeutic target for dementia, epidemiological studies suggest that statin administration might protect against the development of AD (86). Interestingly, we showed that the commonly used cholesterol-lowering agent lovastatin suppresses IFN-γ-induced CD40 expression. Additionally, it markedly inhibits IFN-γ-induced phosphorylation of JAK/STAT1 (Janus kinase/signal transducer and activator of transcription 1), a key intracellular proinflammatory signaling pathway. Furthermore, lovastatin is able to suppress microglial TNF-α, IL-1β and IL-6 production promoted either by IFN-γ or by Aβ peptide challenge in the presence of CD40 stimulation, and lovastatin markedly blunts CD40-mediated inhibition of microglial Aβ clearance from the brain (87).

Other statins have been shown to have somewhat similar effects. In a small pilot study, patients with mild to moderate AD who were administered 80 mg of atorvastatin daily over 1 year experienced significant clinical improvement (88). As sCD40-L is involved in both the activation of microglia and the induction of vascular inflammation, this clinical benefit may be secondary not only to the effect of lowering cholesterol on the reduction of vascular risk factors, but also to reduced Aβ deposition in brain parenchyma, as demonstrated in our experimental models (87). Individuals enrolled in a randomized, controlled trial testing two anti-inflammatory agents for the primary prevention of AD (Alzheimer's Disease Anti-inflammatory Prevention Trial; ADAPT) were allowed the elective use of statins to assess whether their use was associated with a reduced incidence of AD. It was found that elective statin use was associated with a significantly reduced risk of AD after adjustment for age, gender, education and apolipoprotein E (Apo-E) genotype. The findings were similar when comparing the use of lipid-lowering agents (statins and nonstatins) to the use of non-lipid-lowering agents (89). An international, multicenter, double-blind, randomized trial (LEADe, Lipitor's Effect in Alzheimer's Dementia) is under way in 641 subjects with mild to moderate AD treated with atorvastatin in addition to donepezil to definitively evaluate the potential of statins in AD (90).

Additionally, because statins have lipid-independent anti-inflammatory properties, including reduction of circulating sCD40-L (91, 92), these compounds may provide multiple mechanisms for therapeutic benefit in neurodegenerative diseases. Most recently, the Rotterdam study addressed the conflict between some cross-sectional reports suggesting that statin users are less likely to develop AD with prospective studies providing inconsistent evidence. In this prospective, population-based study (N = 6,992) patients were followed for 12–15 years for the incidence of AD. A distinction was made between statins, lipophilic and hydrophilic statins, and nonstatin cholesterol-lowering drugs. Compared with a total lack of use of cholesterol-lowering drugs, statin use was associated with a decreased risk of AD (hazard ratio [HR]: 0.57; 95% confidence interval [CI]: 0.37–0.90), but nonstatin cholesterol-lowering drug use was not (HR: 1.05; 95% CI: 0.45–2.44). The authors concluded that in the general population, statin use, but not the use of nonstatin cholesterol-lowering drugs, was associated with a lower risk of AD compared with no use of cholesterol-lowering drugs, independent of the lipophilicity of the statins (93). Taken together, these results suggest that statins, through both CD40-dependent and -independent pathways, may be a viable therapeutic avenue for neurodegenerative diseases, including AD.

Human umbilical cord blood cell transplantation

An additional therapeutic avenue, which relies in part on suppression of CD40 signaling, is human umbilical cord blood cell (HUCBC) transplantation. HUCBCs have unique immunomodulatory potential. Previous experiments in our laboratory have shown that these cells mitigate AD-like pathology after infusion into the PSAPP mouse model. Specifically, we observed a marked reduction in parenchymal plaques and associated astrocytosis following multiple low-dose infusions of HUCBC. HUCBC infusions also reduced cerebrovascular Aβ deposits in the Tg2576 AD mouse model. These effects were associated with suppression of the CD40-CD40-L interaction, as evidenced by decreased circulating and brain sCD40-L, elevated systemic immunoglobulin M (IgM) levels, attenuated CD40-L-induced inflammatory responses and reduced surface expression of CD40 on microglia. Interestingly, CD40 deficiency abolishes the effect of HUCBC on elevated plasma Aβ levels, demonstrating the requirement for intact CD40-CD40-L signaling for this beneficial effect. Moreover, microglia isolated from HUCBC-infused PSAPP mice demonstrated increased phagocytic clearance of Aβ, and sera from HUCBC-infused PSAPP significantly increased microglial phagocytosis of the Aβ peptide and inhibition of IFN-γ-induced microglial CD40 expression. Increased microglial phagocytic activity in this scenario was inhibited by the addition of recombinant CD40-L protein, again delineating the key role of the CD40 pathway in this effect (94).

Aβ vaccination

Aβ immunization efficiently reduces amyloid plaque load and memory impairment in transgenic mouse models of AD (95-97). Active Aβ immunization, accomplished by administration of Aβ peptide plus adjuvant, has also yielded favorable results in a subset of AD patients (98). However, a small percentage (~6%) of patients receiving the AN-1792 active Aβ vaccine developed severe aseptic meningoencephalitis associated with brain inflammation and infiltration of T cells. We recently demonstrated that genetic or pharmacological interruption of the CD40-CD40-L complex enhances the efficacy of Aβ1–42 immunization in reducing cerebral amyloidosis in the PSAPP and Tg2576 mouse models of AD. Potentially deleterious proinflammatory immune responses seen in the previous Elan/Wyeth AN-1792 trial (cerebral amyloid angiopathy and cerebral microhemorrhage) were reduced or absent in these combined approaches when tested in animals, raising the possibility that this combined approach may be both safe and effective in AD patients. In particular, pharmacological blockade of CD40-L decreased T-cell neurotoxicity to Aβ-producing neurons. Furthermore, reduction of cerebral amyloidosis in Aβ-immunized PSAPP mice homozygously deficient in CD40 was correlated with anti-inflammatory cytokine profiles and reduced plasma sCD40-L. These results suggest that CD40-CD40-L blockade promotes anti-inflammatory cellular immune responses, likely resulting in the promotion of microglial phagocytic activity and Aβ clearance, without the generation of neurotoxic Aβ-reactive T cells. Thus, combined approaches of Aβ immunotherapy and CD40 pathway blockade may provide for a safer and more effective Aβ vaccine in the future (99).

CONCLUSIONS

As the number of elderly individuals continues to increase, the prevention and treatment of dementia must be improved, as the illness is currently one of the leading causes of death after cardiovascular diseases, cancer and cerebrovascular diseases (2). Although the presentations of AD, VAD and HAD may be heterogeneous, we have attempted to highlight the importance of targeting inhibition of the CD40-CD40-L complex since all three of these disorders share a common neuropathological hallmark: microglial activation associated with increased CD40 pathway signaling.

Acknowledgments

DISCLOSURE B. Giunta is supported by an NIH/NIMH Clinical Scientist Award (1 K08 MH082642-01A1). T. Town is supported by an Alzheimer's Association Grant, an NIH/NIA “Pathway to Independence” award (4R00AG029726-02). J. Tan is supported by an NIH-NINDS Ro1 grant (5RO1NS048335-04).

REFERENCES

  • 1.Brookmeyer R, Gray S, Kawas C. Projections of Alzheimer's disease in the United States and the public health impact of delaying disease onset. Am J Public Health. 1998;88(9):1337–42. doi: 10.2105/ajph.88.9.1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Alzheimer's Association: 2009 Alzheimer's Disease Facts and Figures doi: 10.1016/j.jalz.2009.03.001. www.alz.org/alzheimers_disease_facts_figures.asp. [DOI] [PubMed]
  • 3.Burns A, Iliffe S. Dementia. BMJ. 2009;338:b75. doi: 10.1136/bmj.b75. [DOI] [PubMed] [Google Scholar]
  • 4.Ghafouri M, Amini S, Khalili K, Sawaya BE. HIV-1 associated dementia: Symptoms and causes. Retrovirology. 2006;3:28. doi: 10.1186/1742-4690-3-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Ponomarev ED, Shriver LP, Dittel BN. CD40 expression by microglial cells is required for their completion of a two-step activation process during central nervous system autoimmune inflammation. J Immunol. 2006;176(3):1402–10. doi: 10.4049/jimmunol.176.3.1402. [DOI] [PubMed] [Google Scholar]
  • 6.Aloisi F, De Simone R, Columba-Cabezas S, Penna G, Adorini L. Functional maturation of adult mouse resting microglia into an APC is promoted by granulocyte-macrophage colony-stimulating factor and interaction with Th1 cells. J Immunol. 2000;164(4):1705–12. doi: 10.4049/jimmunol.164.4.1705. [DOI] [PubMed] [Google Scholar]
  • 7.D'Aversa TG, Weidenheim KM, Berman JW. CD40-CD40L interactions induce chemokine expression by human microglia: Implications for human immunodeficiency virus encephalitis and multiple sclerosis. Am J Pathol. 2002;160(2):559–67. doi: 10.1016/S0002-9440(10)64875-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kawahara K, Yoshida A, Koga K, et al. Marked induction of inducible nitric oxide synthase and tumor necrosis factor-alpha in rat CD40+ microglia by comparison to CD40- microglia. J Neuroimmunol. 2009;208(1–2):70–9. doi: 10.1016/j.jneuroim.2009.01.007. [DOI] [PubMed] [Google Scholar]
  • 9.Townsend KP, Shytle DR, Bai Y, et al. Lovastatin modulation of microglial activation via suppression of functional CD40 expression. J Neurosci Res. 2004;78(2):167–76. doi: 10.1002/jnr.20234. [DOI] [PubMed] [Google Scholar]
  • 10.Chakrabarti S, Rizvi M, Pathak D, Kirber MT, Freedman JE. Hypoxia influences CD40-CD40L mediated inflammation in endothelial and monocytic cells. Immunol Lett. 2009;122(2):170–84. doi: 10.1016/j.imlet.2008.12.010. [DOI] [PubMed] [Google Scholar]
  • 11.Dubois B, Vanbervliet B, Fayette J, et al. Dendritic cells enhance growth and differentiation of CD40-activated B lymphocytes. J Exp Med. 1997;185(5):941–51. doi: 10.1084/jem.185.5.941. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Abdel-Haq N, Hao HN, Lyman WD. Cytokine regulation of CD40 expression in fetal human astrocyte cultures. J Neuroimmunol. 1999;101(1):7–14. doi: 10.1016/s0165-5728(99)00124-1. [DOI] [PubMed] [Google Scholar]
  • 13.Mach F, Schonbeck U, Sukhova GK, et al. Functional CD40 ligand is expressed on human vascular endothelial cells, smooth muscle cells, and macrophages: Implications for CD40-CD40 ligand signaling in atherosclerosis. Proc Natl Acad Sci USA. 1997;94(5):1931–6. doi: 10.1073/pnas.94.5.1931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Kim KM, Min HY, Jung SH, Lee TH, Kim JG, Kang CY. Characterization of an immunosuppressive anti-CD40 ligand monoclonal antibody. Hybridoma. 1998;17(5):463–70. doi: 10.1089/hyb.1998.17.463. [DOI] [PubMed] [Google Scholar]
  • 15.D'Aversa TG, Eugenin EA, Berman JW. NeuroAIDS: Contributions of the human immunodeficiency virus-1 proteins Tat and gp120 as well as CD40 to microglial activation. J Neurosci Res. 2005;81(3):436–46. doi: 10.1002/jnr.20486. [DOI] [PubMed] [Google Scholar]
  • 16.D'Aversa TG, Eugenin EA, Berman JW. CD40-CD40 ligand interactions in human microglia induce CXCL8 (interleukin-8) secretion by a mechanism dependent on activation of ERK1/2 and nuclear translocation of nuclear factor-kappaB (NFkappaB) and activator protein-1 (AP-1). J Neurosci Res. 2008;86(3):630–9. doi: 10.1002/jnr.21525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Calingasan NY, Erdely HA, Altar CA. Identification of CD40 ligand in Alzheimer's disease and in animal models of Alzheimer's disease and brain injury. Neurobiol Aging. 2002;23(1):31–9. doi: 10.1016/s0197-4580(01)00246-9. [DOI] [PubMed] [Google Scholar]
  • 18.Obradovic SD, Antovic JP, Antonijevic NM, et al. Elevations in soluble CD40 ligand in patients with high platelet aggregability undergoing percutaneous coronary intervention. Blood Coagul Fibrinolysis. 2009 doi: 10.1097/MBC.0b013e328329f28c. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 19.Lessiani G, Dragani A, Falco A, Fioritoni F, Santilli F, Davi G. Soluble CD40 ligand and endothelial dysfunction in aspirin-treated polycythaemia vera patients. Br J Haematol. 2009 doi: 10.1111/j.1365-2141.2009.07636.x. Epub ahead of print. [DOI] [PubMed] [Google Scholar]
  • 20.Andre P, Nannizzi-Alaimo L, Prasad SK, Phillips DR. Platelet-derived CD40L: The switch-hitting player of cardiovascular disease. Circulation. 2002;106(8):896–9. doi: 10.1161/01.cir.0000028962.04520.01. [DOI] [PubMed] [Google Scholar]
  • 21.Perry EK, Perry RH. Neurochemistry of consciousness: Cholinergic pathologies in the human brain. Prog Brain Res. 2004;145:287–99. doi: 10.1016/S0079-6123(03)45020-6. [DOI] [PubMed] [Google Scholar]
  • 22.Johnson KA, Albert MS. Perfusion abnormalities in prodromal AD. Neurobiol Aging. 2000;21(2):289–92. doi: 10.1016/s0197-4580(00)00137-8. [DOI] [PubMed] [Google Scholar]
  • 23.Türker S, Güneri S, Akdeniz B, et al. Usefulness of preprocedural soluble CD40 ligand for predicting restenosis after percutaneous coronary intervention in patients with stable coronary artery disease. Am J Cardiol. 2006;97(2):198–202. doi: 10.1016/j.amjcard.2005.08.024. [DOI] [PubMed] [Google Scholar]
  • 24.Henn V, Slupsky JR, Grafe M, et al. CD40 ligand on activated platelets triggers an inflammatory reaction of endothelial cells. Nature. 1998;391(6667):591–4. doi: 10.1038/35393. [DOI] [PubMed] [Google Scholar]
  • 25.Benveniste EN, Nguyen VT, Wesemann DR. Molecular regulation of CD40 gene expression in macrophages and microglia. Brain Behav Immun. 2004;18(1):7–12. doi: 10.1016/j.bbi.2003.09.001. [DOI] [PubMed] [Google Scholar]
  • 26.Mukundan L, Bishop GA, Head KZ, Zhang L, Wahl LM, Suttles J. TNF receptor-associated factor 6 is an essential mediator of CD40-activated proinflammatory pathways in monocytes and macrophages. J Immunol. 2005;174(2):1081–90. doi: 10.4049/jimmunol.174.2.1081. [DOI] [PubMed] [Google Scholar]
  • 27.Urbich C, Dernbach E, Aicher A, Zeiher AM, Dimmeler S. CD40 ligand inhibits endothelial cell migration by increasing production of endothelial reactive oxygen species. Circulation. 2002;106(8):981–6. doi: 10.1161/01.cir.0000027107.54614.1a. [DOI] [PubMed] [Google Scholar]
  • 28.Kaul M, Lipton SA. Mechanisms of neuronal injury and death in HIV-1 associated dementia. Curr HIV Res. 2006;4(3):307–18. doi: 10.2174/157016206777709384. [DOI] [PubMed] [Google Scholar]
  • 29.Avison MJ, Nath A, Greene-Avison R, et al. Inflammatory changes and breakdown of microvascular integrity in early human immunodeficiency virus dementia. J Neurovirol. 2004;10(4):223–32. doi: 10.1080/13550280490463532. [DOI] [PubMed] [Google Scholar]
  • 30.Bolokadze N, Gabunia P, Ezugbaia M, Gatserelia L, Khechiashvili G. Neurological complications in patients with HIV/AIDS. Georgian Med News. 2008;(165):34–8. [PubMed] [Google Scholar]
  • 31.Huang Y, Erdmann N, Peng H, Zhao Y, Zheng J. The role of TNF related apoptosis-inducing ligand in neurodegenerative diseases. Cell Mol Immunol. 2005;2(2):113–22. [PubMed] [Google Scholar]
  • 32.Gray F, Bazille C, Adle-Biassette H, Mikol J, Moulignier A, Scaravilli F. Central nervous system immune reconstitution disease in acquired immunodeficiency syndrome patients receiving highly active antiretroviral treatment. J Neurovirol. 2005;11(Suppl 3):16–22. doi: 10.1080/13550280500511741. [DOI] [PubMed] [Google Scholar]
  • 33.Lambotte O, Deiva K, Tardieu M. HIV-1 persistence, viral reservoir, and the central nervous system in the HAART era. Brain Pathol. 2003;13(1):95–103. doi: 10.1111/j.1750-3639.2003.tb00010.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.McArthur JC, McDermott MP, McClernon D, et al. Attenuated central nervous system infection in advanced HIV/AIDS with combination antiretroviral therapy. Arch Neurol. 2004;61(11):1687–96. doi: 10.1001/archneur.61.11.1687. [DOI] [PubMed] [Google Scholar]
  • 35.Fischer-Smith T, Rappaport J. Evolving paradigms in the pathogenesis of HIV-1-associated dementia. Expert Rev Mol Med. 2005;7(27):1–26. doi: 10.1017/S1462399405010239. [DOI] [PubMed] [Google Scholar]
  • 36.Rumbaugh JA, Nath A. Developments in HIV neuropathogenesis. Curr Pharm Des. 2006;12(9):1023–44. doi: 10.2174/138161206776055877. [DOI] [PubMed] [Google Scholar]
  • 37.El-Hage N, Bruce-Keller AJ, Knapp PE, Hauser KF. CCL5/RANTES gene deletion attenuates opioid-induced increases in glial CCL2/MCP-1 immunoreactivity and activation in HIV-1 Tat-exposed mice. J Neuroimmune Pharmacol. 2008;3(4):275–85. doi: 10.1007/s11481-008-9127-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Sui Z, Sniderhan LF, Schifitto G, Phipps RP, et al. Functional synergy between CD40 ligand and HIV-1 Tat contributes to inflammation: Implications in HIV type 1 dementia. J Immunol. 2007;178(5):3226–36. doi: 10.4049/jimmunol.178.5.3226. [DOI] [PubMed] [Google Scholar]
  • 39.Sawada M. Neuroprotective and toxic changes in microglia in neurodegenerative disease. Parkinsonism Relat Disord. 2009;15(Suppl 1):S39–41. doi: 10.1016/S1353-8020(09)70011-2. [DOI] [PubMed] [Google Scholar]
  • 40.Sipsas NV, Sfikakis PP, Kontos A, Kordossis T. Levels of soluble CD40 ligand (CD154) in serum are increased in human immunodeficiency virus type 1-infected patients and correlate with CD4(+) T-cell counts. Clin Diagn Lab Immunol. 2002;9(3):558–61. doi: 10.1128/CDLI.9.3.558-561.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Eugenin EA, Osiecki K, Lopez L, Goldstein H, Calderon TM, Berman JW. CCL2/monocyte chemoattractant protein-1 mediates enhanced transmigration of human immunodeficiency virus (HIV)-infected leukocytes across the blood-brain barrier: A potential mechanism of HIV-CNS invasion and NeuroAIDS. J Neurosci. 2006;26(4):1098–106. doi: 10.1523/JNEUROSCI.3863-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.de Gaetano Donati K, Rabagliati R, Iacoviello L, Cauda R. HIV infection, HAART, and endothelial adhesion molecules: Current perspectives. Lancet Infect Dis. 2004;4(4):213–22. doi: 10.1016/S1473-3099(04)00971-5. [DOI] [PubMed] [Google Scholar]
  • 43.van Vonderen MG, Hassink EA, van Agtmael MA, et al. Increase in carotid artery intima-media thickness and arterial stiffness but improvement in several markers of endothelial function after initiation of antiretroviral therapy. J Infect Dis. 2009;199(8):1186–94. doi: 10.1086/597475. [DOI] [PubMed] [Google Scholar]
  • 44.Wolf K, Tsakiris DA, Weber R, Erb P, Battegay M. Swiss HIV Cohort Study. Antiretroviral therapy reduces markers of endothelial and coagulation activation in patients infected with human immunodeficiency virus type 1. J Infect Dis. 2002;185(4):456–62. doi: 10.1086/338572. [DOI] [PubMed] [Google Scholar]
  • 45.Sallem W, Serradji N, Dereuddre-Bosquet N, Dive G, Clayette P, Heymans F. Structure-activity relationships in platelet-activating factor. Part 14: Synthesis and biological evaluation of piperazine derivatives with dual anti-PAF and anti-HIV-1 activity. Bioorg Med Chem. 2006;14(23):7999–8013. doi: 10.1016/j.bmc.2006.07.043. [DOI] [PubMed] [Google Scholar]
  • 46.Aukrust P, Bjornsen S, Lunden B, et al. Persistently elevated levels of von Willebrand factor antigen in HIV infection. Downregulation during highly active antiretroviral therapy. Thromb Haemost. 2000;84(2):183–7. [PubMed] [Google Scholar]
  • 47.de Larranaga GF, Bocassi AR, Puga LM, Alonso BS, Benetucci JA. Endothelial markers and HIV infection in the era of highly active antiretroviral treatment. Thromb Res. 2003;110(2–3):93–8. doi: 10.1016/s0049-3848(03)00291-3. [DOI] [PubMed] [Google Scholar]
  • 48.Khovidhunkit W, Memon RA, Shigenaga JK, et al. Plasma platelet-activating factor acetylhydrolase activity in human immunodeficiency virus infection and the acquired immunodeficiency syndrome. Metabolism. 1999;48(12):1524–31. doi: 10.1016/s0026-0495(99)90240-8. [DOI] [PubMed] [Google Scholar]
  • 49.Yoshimura T, Matsushima K, Oppenheim JJ, Leonard EJ. Neutrophil chemotactic factor produced by lipopolysaccharide (LPS)-stimulated human blood mononuclear leukocytes: Partial characterization and separation from interleukin 1 (IL 1) J Immunol. 2005;175(9):5569–74. [PubMed] [Google Scholar]
  • 50.Baggiolini M, Walz A, Kunkel SL. Neutrophil-activating peptide-1/interleukin 8, a novel cytokine that activates neutrophils. J Clin Invest. 1989;84(4):1045–9. doi: 10.1172/JCI114265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Leonard EJ, Skeel A, Yoshimura T, Noer K, Kutvirt S, Van Epps D. Leukocyte specificity and binding of human neutrophil attractant/activation protein-1. J Immunol. 1990;144(4):1323–30. [PubMed] [Google Scholar]
  • 52.Gerszten RE, Garcia-Zepeda EA, Lim YC, et al. MCP-1 and IL-8 trigger firm adhesion of monocytes to vascular endothelium under flow conditions. Nature. 1999;398(6729):718–23. doi: 10.1038/19546. [DOI] [PubMed] [Google Scholar]
  • 53.Sanders VJ, Pittman CA, White MG, Wang G, Wiley CA, Achim CL. Chemokines and receptors in HIV encephalitis. AIDS. 1998;12(9):1021–6. [PubMed] [Google Scholar]
  • 54.Xiong H, Boyle J, Winkelbauer M, et al. Inhibition of long-term potentiation by interleukin-8: Implications for human immunodeficiency virus-1-associated dementia. J Neurosci Res. 2003;71(4):600–7. doi: 10.1002/jnr.10503. [DOI] [PubMed] [Google Scholar]
  • 55.Zheng JC, Huang Y, Tang K, et al. HIV-1-infected and/or immune-activated macrophages regulate astrocyte CXCL8 production through IL-1β and TNF-α: Involvement of mitogen-activated protein kinases and protein kinase R. J Neuroimmunol. 2008;200(1–2):100–10. doi: 10.1016/j.jneuroim.2008.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Lane BR, Lore K, Bock PJ, et al. Interleukin-8 stimulates human immunodeficiency virus type 1 replication and is a potential new target for antiretroviral therapy. J Virol. 2001;75(17):8195–202. doi: 10.1128/JVI.75.17.8195-8202.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Selkoe DJ. Developing preventive therapies for chronic diseases: Lessons learned from Alzheimer's disease. Nutr Rev. 2007;65(12 Pt 2):S239–43. doi: 10.1111/j.1753-4887.2007.tb00370.x. [DOI] [PubMed] [Google Scholar]
  • 58.Selkoe DJ. Alzheimer's disease: Genes, proteins, and therapy. Physiol Rev. 2001;81(2):741–66. doi: 10.1152/physrev.2001.81.2.741. [DOI] [PubMed] [Google Scholar]
  • 59.Kalaria RN. Cerebral vessels in ageing and Alzheimer's disease. Pharmacol Ther. 1996;72(3):193–214. doi: 10.1016/s0163-7258(96)00116-7. [DOI] [PubMed] [Google Scholar]
  • 60.Akiyama H, Barger S, Barnum S, et al. Inflammation and Alzheimer's disease. Neurobiol Aging. 2000;21(3):383–421. doi: 10.1016/s0197-4580(00)00124-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Meda L, Cassatella MA, Szendrei GI, et al. Activation of microglial cells by beta-amyloid protein and interferon-gamma. Nature. 1995;374(6523):647–50. doi: 10.1038/374647a0. [DOI] [PubMed] [Google Scholar]
  • 62.Tan J, Town T, Paris D, et al. Microglial activation resulting from CD40-CD40L interaction after beta-amyloid stimulation. Science. 1999;286(5448):2352–5. doi: 10.1126/science.286.5448.2352. [DOI] [PubMed] [Google Scholar]
  • 63.Tan J, Town T, Suo Z, et al. Induction of CD40 on human endothelial cells by Alzheimer's beta-amyloid peptides. Brain Res Bull. 1999;50(2):143–8. doi: 10.1016/s0361-9230(99)00122-7. [DOI] [PubMed] [Google Scholar]
  • 64.Togo T, Akiyama H, Kondo H, et al. Expression of CD40 in the brain of Alzheimer's disease and other neurological diseases. Brain Res. 2000;885(1):117–21. doi: 10.1016/s0006-8993(00)02984-x. [DOI] [PubMed] [Google Scholar]
  • 65.Desideri G, Cipollone F, Necozione S, et al. Enhanced soluble CD40 ligand and Alzheimer's disease: Evidence of a possible pathogenetic role. Neurobiol Aging. 2008;29(3):348–56. doi: 10.1016/j.neurobiolaging.2006.10.019. [DOI] [PubMed] [Google Scholar]
  • 66.Tousoulis D, Antoniades C, Nikolopoulou A, et al. Interaction between cytokines and sCD40L in patients with stable and unstable coronary syndromes. Eur J Clin Invest. 2007;37(8):623–8. doi: 10.1111/j.1365-2362.2007.01834.x. [DOI] [PubMed] [Google Scholar]
  • 67.Vermeer SE, Hollander M, van Dijk EJ, et al. Silent brain infarcts and white matter lesions increase stroke risk in the general population: The Rotterdam Scan Study. Stroke. 2003;34(5):1126–9. doi: 10.1161/01.STR.0000068408.82115.D2. [DOI] [PubMed] [Google Scholar]
  • 68.Suo Z, Tan J, Placzek A, Crawford F, Fang C, Mullan M. Alzheimer's beta-amyloid peptides induce inflammatory cascade in human vascular cells: The roles of cytokines and CD40. Brain Res. 1998;807(1–2):110–7. doi: 10.1016/s0006-8993(98)00780-x. [DOI] [PubMed] [Google Scholar]
  • 69.Breteler MM. Vascular risk factors for Alzheimer's disease: An epidemiologic perspective. Neurobiol Aging. 2000;21(2):153–60. doi: 10.1016/s0197-4580(99)00110-4. [DOI] [PubMed] [Google Scholar]
  • 70.de la Torre JC. Critically attained threshold of cerebral hypoperfusion: The CATCH hypothesis of Alzheimer's pathogenesis. Neurobiol Aging. 2000;21(2):331–42. doi: 10.1016/s0197-4580(00)00111-1. [DOI] [PubMed] [Google Scholar]
  • 71.Snowdon DA, Greiner LH, Mortimer JA, Riley KP, Greiner PA, Markesbery WR. Brain infarction and the clinical expression of Alzheimer disease. The Nun Study. JAMA. 1997;277(10):813–7. [PubMed] [Google Scholar]
  • 72.Kalaria RN. The role of cerebral ischemia in Alzheimer's disease. Neurobiol Aging. 2000;21(2):321–30. doi: 10.1016/s0197-4580(00)00125-1. [DOI] [PubMed] [Google Scholar]
  • 73.Sevush S, Jy W, Horstman LL, Mao WW, Kolodny L, Ahn YS. Platelet activation in Alzheimer disease. Arch Neurol. 1998;55(4):530–6. doi: 10.1001/archneur.55.4.530. [DOI] [PubMed] [Google Scholar]
  • 74.Town T, Tan J, Mullan M. CD40 signaling and Alzheimer's disease pathogenesis. Neurochem Int. 2001;39(5–6):371–80. doi: 10.1016/s0197-0186(01)00044-4. [DOI] [PubMed] [Google Scholar]
  • 75.Tan J, Town T, Mullan M. CD40-CD40L interaction in Alzheimer's disease. Curr Opin Pharmacol. 2002;2(4):445–51. doi: 10.1016/s1471-4892(02)00180-7. [DOI] [PubMed] [Google Scholar]
  • 76.Town T, Tan J, Flavell RA, Mullan M. T-cells in Alzheimer's disease. Neuromol Med. 2005;7(3):255–64. doi: 10.1385/NMM:7:3:255. [DOI] [PubMed] [Google Scholar]
  • 77.Greenwood J, Wang Y, Calder VL. Lymphocyte adhesion and transendothelial migration in the central nervous system: The role of LFA-1, ICAM-1, VLA-4 and VCAM-1. Immunology. 1995;86(3):408–15. [PMC free article] [PubMed] [Google Scholar]
  • 78.Saito M, Ota Y, Ohashi H, et al. CD40-CD40 ligand signal induces the intercellular adhesion molecule-1 expression through nuclear factor-kappa B p50 in cultured salivary gland epithelial cells from patients with Sjögren's syndrome. Mod Rheumatol. 2007;17(1):45–53. doi: 10.1007/s10165-006-0538-7. [DOI] [PubMed] [Google Scholar]
  • 79.Mandrekar S, Jiang Q, Lee CY, Koenigsknecht-Talboo J, Holtzman DM, Landreth GE. Microglia mediate the clearance of soluble Abeta through fluid phase macropinocytosis. J Neurosci. 2009;29(13):4252–62. doi: 10.1523/JNEUROSCI.5572-08.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Tan J, Town T, Paris D, et al. Activation of microglial cells by the CD40 pathway: Relevance to multiple sclerosis. J Neuroimmunol. 1999;97(1–2):77–85. doi: 10.1016/s0165-5728(99)00053-3. [DOI] [PubMed] [Google Scholar]
  • 81.Ait-Ghezala G, Mathura VS, Laporte V, et al. Genomic regulation after CD40 stimulation in microglia: Relevance to Alzheimer's disease. Mol Brain Res. 2005;140(1–2):73–85. doi: 10.1016/j.molbrainres.2005.07.014. [DOI] [PubMed] [Google Scholar]
  • 82.Laporte V, Ait-Ghezala G, Volmar CH, Mullan M. CD40 deficiency mitigates Alzheimer's disease pathology in transgenic mouse models. J Neuroinflammation. 2006;3:3. doi: 10.1186/1742-2094-3-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Todd Roach J, Volmar CH, Dwivedi S, et al. Behavioral effects of CD40-CD40L pathway disruption in aged PSAPP mice. Brain Res. 2004;1015(1–2):161–8. doi: 10.1016/j.brainres.2004.05.004. [DOI] [PubMed] [Google Scholar]
  • 84.Tarkowski E, Liljeroth AM, Minthon L, Tarkowski A, Wallin A, Blennow K. Cerebral pattern of pro- and anti-inflammatory cytokines in dementias. Brain Res Bull. 2003;61(3):255–60. doi: 10.1016/s0361-9230(03)00088-1. [DOI] [PubMed] [Google Scholar]
  • 85.Ray S, Britschgi M, Herbert C, et al. Classification and prediction of clinical Alzheimer's diagnosis based on plasma signaling proteins. Nat Med. 2007;13(11):1359–62. doi: 10.1038/nm1653. [DOI] [PubMed] [Google Scholar]
  • 86.Cramer C, Haan MN, Galea S, Langa KM, Kalbfleisch JD. Use of statins and incidence of dementia and cognitive impairment without dementia in a cohort study. Neurology. 2008;71(5):344–50. doi: 10.1212/01.wnl.0000319647.15752.7b. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Townsend KP, Shytle DR, Bai Y, et al. Lovastatin modulation of microglial activation via suppression of functional CD40 expression. J Neurosci Res. 2004;78(2):167–76. doi: 10.1002/jnr.20234. [DOI] [PubMed] [Google Scholar]
  • 88.Sparks DL, Sabbagh MN, Connor DJ, et al. Atorvastatin for the treatment of mild to moderate Alzheimer disease: Preliminary results. Arch Neurol. 2005;62(5):753–7. doi: 10.1001/archneur.62.5.753. [DOI] [PubMed] [Google Scholar]
  • 89.Sparks DL, Kryscio RJ, Sabbagh MN, Connor DJ, Sparks LM, Liebsack C. Reduced risk of incident AD with elective statin use in a clinical trial cohort. Curr Alzheimer Res. 2008;5(4):416–21. doi: 10.2174/156720508785132316. [DOI] [PubMed] [Google Scholar]
  • 90.Jones RW, Kivipelto M, Feldman H, et al. The Atorvastatin/Donepezil in Alzheimer's Disease Study (LEADe): Design and baseline characteristics. Alzheimers Dement. 2008;4(2):145–53. doi: 10.1016/j.jalz.2008.02.001. [DOI] [PubMed] [Google Scholar]
  • 91.Santini E, Madec S, Corretti V, Ferrannini E, Solini A. Effect of statins on soluble CD40 ligand in hypercholesterolemic type 2 diabetic patients. J Endocrinol Invest. 2008;31(7):660–5. doi: 10.1007/BF03345621. [DOI] [PubMed] [Google Scholar]
  • 92.Blanco-Colio LM, Martin-Ventura JL, de Teresa E, et al. Atorvastatin decreases elevated soluble CD40L in subjects at high cardiovascular risk. Atorvastatin on inflammatory markers study: A substudy of ACTFAST. Kidney Int Suppl. 2008;(111):S60–3. doi: 10.1038/ki.2008.514. [DOI] [PubMed] [Google Scholar]
  • 93.Haag MD, Hofman A, Koudstaal PJ, Stricker BH, Breteler MM. Statins are associated with a reduced risk of Alzheimer disease regardless of lipophilicity. The Rotterdam Study. J Neurol Neurosurg Psychiatry. 2009;80(1):13–7. doi: 10.1136/jnnp.2008.150433. [DOI] [PubMed] [Google Scholar]
  • 94.Nikolic WV, Hou H, Town T, et al. Peripherally administered human umbilical cord blood cells reduce parenchymal and vascular beta-amyloid deposits in Alzheimer mice. Stem Cells Dev. 2008;17(3):423–39. doi: 10.1089/scd.2008.0018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schenk D, Barbour R, Dunn W, et al. Immunization with amyloid-beta attenuates Alzheimer-disease-like pathology in the PDAPP mouse. Nature. 1999;400(6740):173–7. doi: 10.1038/22124. [DOI] [PubMed] [Google Scholar]
  • 96.Morgan D, Diamond DM, Gottschall PE, et al. A beta peptide vaccination prevents memory loss in an animal model of Alzheimer's disease. Nature. 2000;408(6815):982–5. doi: 10.1038/35050116. [DOI] [PubMed] [Google Scholar]
  • 97.Janus C, Pearson J, McLaurin J, et al. A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer's disease. Nature. 2000;408(6815):979–82. doi: 10.1038/35050110. [DOI] [PubMed] [Google Scholar]
  • 98.Hock C, Konietzko U, Streffer JR, et al. Antibodies against beta-amyloid slow cognitive decline in Alzheimer's disease. Neuron. 2003;38(4):547–54. doi: 10.1016/s0896-6273(03)00294-0. [DOI] [PubMed] [Google Scholar]
  • 99.Obregon D, Hou H, Bai Y, et al. CD40L disruption enhances Abeta vaccine-mediated reduction of cerebral amyloidosis while minimizing cerebral amyloid angiopathy and inflammation. Neurobiol Dis. 2008;29(2):336–53. doi: 10.1016/j.nbd.2007.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES