2007 Progress Report on Alzheimer's Disease: Discovery and Hope -- 3 Outlook for the Future, References
OUTLOOK FOR THE FUTURE

After 30 years of intensive study, momentum in AD research is palpable. Today, scientists are increasingly optimistic about the promise of AD research. Initially, they focused on defining the major characteristics of the disease, its course, and aspects of its etiology. Since then, they have built an enormous base of knowledge about AD and the many factors that contribute to the damage it causes to the brain and body. Researchers are now beginning to apply that knowledge to treatment and prevention strategies. To sustain this momentum, NIH is moving ahead on a number of fronts, described in the sections below.


Improving Basic Understanding

Research must continue to refine the mechanistic understanding of AD. Though we understand much about the disease, important gaps in knowledge remain. Basic research is central to the search for effective new therapies because it allows scientists to understand better the normal biology of the aging brain and what goes wrong in a disease like AD. One promising area of basic research involves studies to learn more about the molecular basis of cognition and how it changes with age. Another area of interest is how the brain adapts to injury. Researchers know that the brain is a remarkably adaptive organ, and they want to learn more about the components that are important to adaptability, the molecular basis of cognitive reserve, and whether strategies to build cognitive reserve can prevent or delay the negative consequences of AD pathology. The Cognitive Aging Summit sponsored by NIA and the McKnight Brain Research Foundation in Washington, D.C., in October 2007 helped identify topics in these areas of basic research that are most likely to bear fruit.


Identifying Genetic Causes and Risk Factors

With the establishment of the Alzheimer’s Disease Genetics Initiative and the Alzheimer’s Disease Genetics Consortium, scientists hope to learn considerably more about the major risk-factor genes for late-onset AD and for cognitive decline. Understanding more about the genetics of cognitive decline and AD will shed light on how much genetic risk they share. Knowing the risk-factor genes will help to pinpoint new pathways that contribute to the early development of AD and to identify people at the greatest genetic risk of cognitive decline or AD.


Understanding Disease Progression

Findings from the Alzheimer’s Disease Neuroimaging Initiative reported over the next few years will likely indicate which combinations of clinical, neuropsychological, imaging, and biomarker tests best predict who will progress from aMCI to AD and how this progression occurs. Improved knowledge in this area also will help accelerate and refine the conduct of clinical trials. A greater understanding of disease progression will eventually allow clinicians to start therapies much earlier in the disease process, when changes in the brain associated with AD are still minimal.


Making the Most of Translational Research and Clinical Trials

Translational initiatives and federally supported clinical trials should provide important new approaches for AD prevention and treatment because they will ensure that researchers with promising therapies have opportunities to develop them. Many compounds that test well in animal models and that have a sound theoretical basis fail in clinical trials because of safety or efficacy problems. It is important to understand why this happens. Many different therapeutic targets must be pursued besides the obvious ones of beta-amyloid and tau.

By funding clinical trials, NIA also hopes to provide the necessary foundation for private industry or NIH to support “follow-up” clinical studies of the most promising leads.


Cultivating Research Infrastructure and Resources

NIH will continue to provide the framework through which investigators can conduct interdisciplinary and collaborative AD research. For example, in 1999, NIA established the National Alzheimer’s Coordinating Center (NACC) (www.alz.washington.edu) so that data on healthy people and patients from the Alzheimer’s Disease Centers could be pooled and shared. By 2005, information on more than 75,000 Alzheimer’s Disease Center study participants and neuropathologic data on more than 9,000 brains from autopsied participants had been collected. Much of this material is available for research by qualified scientists.

In 2006, NACC launched the Alzheimer’s Disease Center Uniform Data Set (UDS), a system that improved upon a previous data collection system by standardizing data collection across research sites. Initially developed to gather information on healthy participants and those with aMCI and early AD, the UDS has since expanded to collect data on participants with frontotemporal dementia, Lewy body dementia, and vascular dementia. More than 15,000 individuals are now being evaluated using the UDS. Data are available to qualified researchers through NACC.

Workshops and meetings, such as the AD Planning Meeting held in October 2006 and the Cognitive Aging Summit held in October 2007, provide an opportunity for experts to gather and assess basic, translational, and clinical research efforts and to assess future strategies for maximizing public investment.

NIA and other NIH Institutes and Centers that conduct research on AD collaborate with many others to push the boundaries of our knowledge about this disease. For example, the Institutes work with the Foundation for the National Institutes of Health to identify appropriate funding opportunities with private industry. NIA collaborates with private foundations, such as the Alzheimer’s Association and the Alzheimer’s Drug Discovery Foundation, on specific initiatives. NIA has worked with both organizations on the Alzheimer’s Disease Neuroimaging Initiative through the Foundation for the National Institutes of Health and with the AD Discovery Foundation on translational research projects. NIH scientists also work with colleagues at the Centers for Disease Control and Prevention, the FDA, and other Federal agencies to ensure expeditious efforts to find the best combinations of behavioral and drug interventions for AD and age-related cognitive decline.



CONCLUSION

The theme of the 2007 Alzheimer’s Disease Progress Report is “Discovery and Hope.” It’s a fitting description for, during 2007, AD researchers continued to push the boundaries of knowledge through discoveries on a number of fronts—basic and genetics research on the fundamental nature of the disease and its relationship to normal aging; epidemiologic studies on associations between AD risk, lifestyle factors, and chronic disease; studies on early physiologic changes that signal AD development; translational research studies; clinical trials; and studies to help caregivers.

These discoveries are fostering hope that our richer understanding of the disease (and our growing appreciation of its complexities) are leading to the development of improved techniques for diagnosing AD early and accurately and to the identification of a range of potential therapeutic targets. There is also new hope for the development of effective strategies for helping caregivers cope and even for the identification of possible factors that may reduce AD risk.

At the same time, we cannot forget that AD remains an urgent health problem for our Nation and that we still have much to learn about the disease. Our task is to build on these recent discoveries through continued support for multidisciplinary, collaborative AD research so that its potential will become a reality for millions of older adults.



REFERENCES

Bennett DA, Schneider JA, Arvanitakis Z, Kelly JF, Aggarwal NT, Shah RC, Wilson RS. Neuropathology of older persons without cognitive impairment from two community-based studies. Neurology 2006a;66(12):1837-1844.

Bennett DA, Schneider JA, Tang Y, Arnold SE, Wilson RS. The effect of social networks on the relation between Alzheimer’s disease pathology and level of cognitive function in old people: a longitudinal cohort study. Lancet Neurology 2006b;5(5):406-412.

Blacker D, Lee H, Muzikansky A, Martin EC, Tanzi R, McArdle JJ, Moss M, Albert M. Neuropsychological measures in normal individuals that predict subsequent cognitive decline. Archives of Neurology 2007;64(6):862-871.

Blackwell T, Yaffe K, Ancoli-Israel S, Schneider JL, Cauley JA, Hillier TA, Fink HA, Stone KL. Poor sleep is associated with impaired cognitive function in older women: the study of osteoporotic fractures. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 2006;61(4):405-410.

Buchman AS, Boyle PA, Wilson RS, Tang Y, Bennett DA. Frailty is associated with incident Alzheimer’s disease and cognitive decline in the elderly. Psychosomatic Medicine 2007;69(5):483-489.

Caselli RJ, Reiman EM, Locke DE, Hutton ML, Hentz JG, Hoffman-Snyder C, Woodruff BK, Alexander GE, Osborne D. Cognitive domain decline in healthy apolipoprotein E epsilon4 homozygotes before the diagnosis of mild cognitive impairment. Archives of Neurology 2007;64(9):1306-1311.

Cohen E, Bieschke J, Perciavalle RM, Kelly JW, Dillin A. Opposing activities protect against age-onset proteotoxicity. Science 2006;313(5793):1604-1610.

Cohen-Mansfield J, Libin A, Marx M. Nonpharmacological treatment of agitation: a controlled trial of systematic individualized intervention. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 2007;62(8):908-916.

Colcombe SJ, Erickson KI, Scalf PE, Kim JS, Prakash R, McAuley E, Elavsky S, Marquez DX, Hu L, Kramer AF. Aerobic exercise training increases brain volume in aging humans. The Journals of Gerontology. Series A, Biological Sciences and Medical Sciences 2006;61(11):1166-1170.

Damjanovic AK, Yang Y, Glaser R, Kiecolt-Glaser JK, Nguyen H, Laskowski B, Zou Y, Beversdorf DQ, Weng N. Accelerated telomere erosion is associated with a declining immune function of caregivers of Alzheimer’s disease patients. Journal of Immunology 2007;179(6):4249-4254.

DeCarli C, Frisoni GB, Clark CM, Harvey D, Grundman M, Petersen RC, Thal LJ, Jin S, Jack CR, Scheltens P, Alzheimer’s Disease Cooperative Study Group. Qualitative estimates of medial temporal lobe atrophy as a predictor of progression from mild cognitive impairment to dementia. Archives of Neurology 2007;64(1):108-115.

Devanand DP, Pradhaban G, Liu X, Khandji A, De Santi S, Segal S, Rusinek H, Pelton GH, Honig LS, Mayeux R, Stern Y, Tabert MH, de Leon MJ. Hippocampal and entorhinal atrophy in mild cognitive impairment: prediction of Alzheimer disease. Neurology 2007;68(11):828-836.

Dowling GA, Graf CL, Hubbard EM, Luxenberg JS. Light treatment for neuropsychiatric behaviors in Alzheimer’s disease. Western Journal of Nursing Research 2007;29(8):961-975.

Erickson KI, Colcombe SJ, Wadhwa R, Bherer L, Peterson MS, Scalf PE, Kim JS, Alvarado M, Kramer AF. Training-induced functional activation changes in dual-task processing: an fMRI study. Cerebral Cortex 2007;17(1):192-204.

Evans DA, Scherr PA, Cook NR, Albert MS, Funkenstein HH, Smith LA, Hebert LE, Wetle TT, Branch LG, Chown M, Hennekens CH, Taylor JO. Estimated prevalence of Alzheimer’s disease in the United States. Milbank Quarterly 1990;68:267-289.

Fagan AM, Roe, CM, Xiong C, Mintun MA, Morris JC, Holtzman DM. Cerebrospinal fluid tau/β-Amyloid42 ratio as a prediction of cognitive decline in nondemented older adults. Archives of Neurology 2007;64(3):343-349.

Farran CJ, Gilley DW, McCann JJ, Bienias JL, Lindeman DA, Evans DA. Efficacy of behavioral interventions for dementia caregivers. Western Journal of Nursing Research 2007;29(8):944-960.

Femia EE, Zarit SH, Stephens MA, Greene R. Impact of adult day services on behavioral and psychological symptoms of dementia. Gerontologist 2007;47(6):775-788.

Fleisher AS, Sowell BB, Taylor C, Gamst AC, Petersen RC, Thal LJ, Alzheimer's Disease Cooperative Study. Clinical predictors of progression to Alzheimer disease in amnestic mild cognitive impairment. Neurology 2007;68(19):1588-1595.

Gallardo G, Schlüter OM, Südhof TC. A molecular pathway of neurodegeneration linking alpha-synuclein to ApoE and Abeta peptides. Nature Neuroscience 2008;11(3):301-308.

Galvin JE, Roe CM, Coats MA, Morris JC. Patient’s rating of cognitive ability: using the AD8, a brief informant interview, as a self-rating tool to detect dementia. Archives of Neurology 2007;64(5):725-730.

Galvin JE, Roe CM, Xiong C, Morris JC. Validity and reliability of the AD8 informant interview in dementia. Neurology 2006;67(11):1942-1948.

Ge S, Yang CH, Hsu KS, Ming GL, Song H. A critical period for enhanced synaptic plasticity in newly generated neurons of the adult brain. Neuron 2007;54(4):559-566.

Gidalevitz T, Ben-Zvi A, Ho KH, Brignull HR, Morimoto RI. Progressive disruption of cellular protein folding in models of polyglutamine diseases. Science 2006;311(5766):1471-1474.

Hebert LE, Scherr PA, Bienias JL, Bennett DA, Evans DA. Alzheimer disease in the US population: prevalence estimates using the 2000 census. Archives of Neurology 2003;60(8):1119-1122.

Hu W, Ranaivo HR, Roy SM, Behanna HA, Wing LK, Munoz L, Guo L, Van Eldik LJ, Watterson DM. Development of a novel therapeutic suppressor of brain proinflammatory cytokine up-regulation that attenuates synaptic dysfunction and behavioral deficits. Bioorganic & Medicinal Chemistry Letters 2007;17(2):414-418.

Jara JH, Singh BB, Floden AM, Combs CK. Tumor necrosis factor alpha stimulates NMDA receptor activity in mouse cortical neurons resulting in ERK-dependent death. Journal of Neurochemistry 2007;100(5):1407-1420.

Katzman R. The prevalence and malignancy of Alzheimer’s disease: a major killer. Archives of Neurology 1976;33(4):217-218.

Khan AA, Mao XO, Banwait S, Jin K, Greenberg DA. Neuroglobin attenuates beta-amyloid neurotoxicity in vitro and transgenic Alzheimer phenotype in vivo. Proceedings of the NationalAcademy of Sciences of the United States of America 2007;104(48):19114-19119.

Klunk WE, Engler H, Nordberg A, Wang Y, Blomqvist G, Holt DP, Bergström M, Savitcheva I, Huang GF, Estrada S, Ausén B, Debnath ML, Barletta J, Price JC, Sandell J, Lopresti BJ, Wall A, Koivisto P, Antoni G, Mathis CA, Långström B. Imaging brain amyloid in Alzheimer’s disease with Pittsburgh compound-B. Archives of Neurology 2004;55(3):306-319.

Lacor PN, Buniel MC, Furlow PW, Clemente AS, Velasco PT, Wood M, Viola KL, Klein WL. Abeta oligomer-induced aberrations in synapse composition, shape, and density provide a molecular basis for loss of connectivity in Alzheimer’s disease. Journal of Neuroscience 2007;27(4):796-807.

Larson EB, Wang L, Bowen JD, McCormick WC, Teri L, Crane P, Kulull W. Exercise is associated with reduced risk for incident dementia among persons 65 years of age and older. Annals of Internal Medicine 2006;144(2):73-81.

Lee JH, Cheng R, Honig LS, Vonsattel JP, Clark L, Mayeux R. Association between genetic variants in SORL1 and autopsy-confirmed Alzheimer disease. Neurology. 2008;70(11):887-889.

Lee JH, Cheng R, Schupf N, Manly J, Lantigua R, Stern Y, Rogaeva E, Wakutani Y, Farrer L, St George-Hyslop P, Mayeux R. The association between genetic variants in SORL1 and Alzheimer disease in an urban, multiethnic, community-based cohort. Archives of Neurology 2007;64(4):501-506.

Li T, Wen H, Brayton C, Laird FM, Ma G, Peng S, Placanica L, Wu TC, Crain BJ, Price DL, Eberhart CG, Wong PC. Moderate reduction of gamma-secretase attenuates amyloid burden and limits mechanism-based liabilities. Journal of Neuroscience 2007;27(40):10849-10859.

Luchsinger JA, Reitz C, Patel B, Tang MX, Manly JJ, Mayeux R. Relation of diabetes to mild cognitive impairment. Archives of Neurology 2007;64(4):570-575.

Lyketsos CG, Samus QM, Baker A, McNabney M, Onyike CU, Mayer LS, Brandt J, Rabins P, Rosenblatt A. Effect of dementia and treatment of dementia on time to discharge from assisted living facilities: the Maryland assisted living study. Journal of the American Geriatrics Society 2007;55(7):1031-1037.

Maier M, Seabrook TJ, Lazo ND, Jiang L, Das P, Janus C, Lemere CA. Short amyloid-β (Aβ) immunogens reduce cerebral Aβ load and learning deficits in an Alzheimer's disease mouse model in the absence of an Aβ-specific cellular immune response.Journal of Neuroscience 2006;26(18):4717-4728.

Mapstone M, Logan D, Duffy CJ. Cue integration for the perception and control of self-movement in ageing and Alzheimer’s disease. Brain 2006;129(Pt 11):2931-2944.

McKee AC, Au R, Cabral HJ, Kowall NW, Seshadri S, Kubilus CA, Drake J, Wolf PA. Visual association pathology in preclinical Alzheimer disease. Journal of Neuropathology and Experimental Neurolology 2006;65(6):621-630.

Meng Y, Lee JH, Cheng R, St. George-Hyslop P, Mayeux R, Farrer LA. Association between SORL1 and Alzheimer’s disease in a genome-wide study. Neuroreport 2007;18(17):1761-1764.

Mintun MA, Larossa GN, Sheline YI, Dence CS, Lee SY, Mach RH, Klunk WE, Mathis CA, DeKosky ST, Morris JC. PIB in a nondemented population: potential antecedent marker of Alzheimer disease. Neurology 2006;67(3):446-452.

Mittelman MS, Haley WE, Clay OJ, Roth DL. Improving caregiver well-being delays nursing home placement of patients with Alzheimer disease. Neurology 2006;67(9):1592-1599.

Mittelman MS, Roth DL, Clay OJ, Haley WE. Preserving health of Alzheimer caregivers: impact of a spouse caregiver intervention. American Journal of Geriatric Psychiatry 2007;15(9):780-789.

Neumann M, Sampathu DM, Kwong LK, Truax AC, Micsenyi MC, Chou TT, Bruce J, Schuck T, Grossman M, Clark CM, McCluskey LF, Miller BL, Masliah E, Mackenzie IR, Feldman H, Feiden W, Kretzschmar HA, Trojanowski JQ, Lee VM. Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 2006;314(5796):130-133.

Palop JJ, Chin J, Roberson ED, Wang J, Thwin MT, Bien-Ly N, Yoo J, Ho KO, Yu GQ, Kreitzer A, Finkbeiner S, Noebels JL, Mucke L. Aberrant excitatory neuronal activity and compensatory remodeling of inhibitory hippocampal circuits in mouse models of Alzheimer’s disease. Neuron 2007;55(5):697-711.

Palumbo C, Beiser A, Au R, DeCarli C, Seshadri S, Wolf PA. Apolipoprotein E modifies the relationship of brain MRI to cognition: the Framingham Heart Study. Neurology 2007;68(Suppl 1):A166-A167. Poster presented at 59th Annual American Academy of Neurology Meeting, May 2, 2007.

Peavy GM, Lange KL, Salmon DP, Patterson TL, Goldman S, Gamst AC, Mills PJ, Khandrika S, Galasko D. The effects of prolonged stress and APOE genotype on memory and cortisol in older adults. Biological Psychiatry 2007;62(5):472-478.

Pedersen WA, McMillan PJ, Kulstad JJ, Leverenz JB, Craft S, Haynatzki GR. Rosiglitazone attenuates learning and memory deficits in Tg2576 Alzheimer mice. Experimental Neurology 2006;199(2):265-273.

Plassman BL, Langa KM, Fischer GG, Heeringa SG, Weir DR, Ofstedal MB, Burke JR, Hurd MD, Potter GG, Rodgers WL, Steffens DC, Willis RJ, Wallace RB. Prevalence of dementia in the United States: the Aging, Demographics, and Memory Study. Neuroepidemiology 2007;29(1-2):125-132.

Pollock BG, Mulsant BH, Rosen J, Mazumdar S, Blakesley RE, Houck PR, Huber KA. A double-blind comparison of citalopram and risperidone for the treatment of behavioral and psychotic symptoms associated with dementia. American Journal of Geriatric Psychiatry 2007;15(11):942-952.

Prada CM, Garcia-Alloza M, Betensky RA, Zhang-Nunes SX, Greenberg SM, Bacskai BJ, Frosch MP. Antibody-mediated clearance of amyloid-beta peptide from cerebral amyloid angiopathy revealed by quantitative in vivo imaging. Journal of Neuroscience 2007;27(8):1973-1980.

Ramirez-Amaya V, Marrone DF, Gage FH, Worley PF, Barnes CA. Integration of new neurons into functional neural networks. Journal of Neuroscience 2006;26(47):12237-12241.

Roberson ED, Scearce-Levie K, Palop JJ, Yan F, Cheng IH, Wu T, Gerstein H, Yu GQ, Mucke L. Reducing endogenous tau ameliorates amyloid beta-induced deficits in an Alzheimer’s disease mouse model. Science 2007;316(5825):750-754.

Rocca WA, Bower JH, Maraganore DM, Ahlskog JE, Grossardt BR, de Andrade M, Melton LJ 3rd. Increased risk of cognitive impairment or dementia in women who underwent oophorectomy before menopause. Neurology 2007;69(11):1074-1083.

Rogaeva E, Meng Y, Lee JH, Gu Y, Kawarai T, Zou F, Katayama T, Baldwin CT, Cheng R, Hasegawa H, Chen F, Shibata N, Lunetta KL, Pardossi-Piquard R, Bohm C, Wakutani Y, Cupples LA, Cuenco KT, Green RC, Pinessi L, Rainero I, Sorbi S, Bruni A, Duara R, Friedland RP, Inzelberg R, Hampe W, Bujo H, Song YQ, Andersen OM, Willnow TE, Graff-Radford N, Petersen RC, Dickson D, Der SD, Fraser PE, Schmitt-Ulms G, Younkin S, Mayeux R, Farrer LA, St George-Hyslop P. The neuronal sortilin-related receptor SORL1 is genetically associated with Alzheimer disease. Nature Genetics 2007;39(2):168-177.

Rosenheck RA, Leslie DL, Sindelar JL, Miller EA, Tariot PN, Dagerman KS, Davis SM, Lebowitz BD, Rabins P, Hsiao JK, Lieberman JA, Schneider LS. Clinical antipsychotic trial of intervention effectiveness-Alzheimer's disease (CATIE-AD) investigators: cost-benefit analysis of second-generation antipsychotics and placebo in a randomized trial of the treatment of psychosis and aggression in Alzheimer disease. Archives of General Psychiatry 2007;64(11):1259-1268.

Rowe CC, Ng S, Ackermann U, Gong SJ, Pike K, Savage G, Savage G, Cowie TF, Dickinson KL, Maruff P, Darby D, Smith C, Woodward M, Merory J, Tochon-Danguy H, O’Keefe G, Klunk WE, Mathis CA, Price JC, Masters CL, Villemagne VL. Imaging beta-amyloid burden in aging and dementia. Neurology 2007;68(20):1718-1725.

Rowe WB, Blalock EM, Chen KC, Kadish I, Wang D, Barrett JE, Thibault O, Porter NM, Rose GM, Landfield PW. Hippocampal expression analyses reveal selective association of immediate-early, neuroenergetic, and myelinogenic pathways with cognitive impairment in aged rats. Journal of Neuroscience 2007;27(12):3098-3110.

Saganich MJ, Schroeder BE, Galvan V, Bredesen DE, Koo EH, Heinemann SF. Deficits in synaptic transmission and learning in amyloid precursor protein (APP) transgenic mice require C-terminal cleavage of APP. Journal of Neuroscience 2006;26(52):13428-13436.

Schneider JA, Boyle PA, Arvanitakis Z, Bienias JL, Bennett DA. Subcortical infarcts, Alzheimer’s disease pathology, and memory function in older persons. Archives of Neurology 2007;62(1):59-66.

Seshadri S, DeStefano AL, Au R, Massaro JM, Beiser AS, Kelly-Hayes M, Kase CS, D’Agostino RB, DeCarli C, Atwood LD, Wolf PA. Genetic correlates of brain aging on MRI and cognitive test measures: a genome-wide association and linkage analysis in the Framingham Study. BMC Medical Genetics 2007;8(Suppl 1):S15.

Siegel SJ, Bieschke J, Powers ET, Kelly JW. The oxidative stress metabolite 4-hydroxynonenal promotes Alzheimer protofibril formation. Biochemistry 2007;46(6):1503-1510.

Tan ZS, Beiser AS, Vasan RS, Roubenoff R, Dinarello CA, Harris TB, Benjamin EJ, Au R, Kiel DP, Wolf PA, Seshadri S. Inflammatory markers and the risk of Alzheimer disease: the Framingham Study. Neurology. 2007;68(22):1902-1908.

Thomas RM, Hotsenpiller G, Peterson DA. Acute psychosocial stress reduces cell survival in adult hippocampal neurogenesis without altering proliferation. Journal of Neuroscience 2007;27(11):2734-2743.

Unverzagt FW, Kasten L, Johnson KE, Rebok GW, Marsiske M, Koepke KM, Elias JW, Morris JN, Willis SL, Ball K, Rexroth DF, Smith DM, Wolinsky FD, Tennstedt SL. Effect of memory impairment on training outcomes in ACTIVE. Journal of the International Neuropsychological Society 2007;13(6):953-960.

Wang M, Ramos BP, Paspalas CD, Shu Y, Simen A, Duque A, Vijayraghavan S, Brennan A, Dudley A, Nou E, Mazer JA, McCormick DA, Arnsten AFT. Alpha2A-adrenoceptors strengthen working memory networks by inhibiting cAMP-HCN channel signaling in prefrontal cortex. Cell 2007;129(2):397-410.

Weeraratna AT, Kalehua A, Deleon I, Bertak D, Maher G, Wade MS, Lustig A, Becker KG, Wood W 3rd, Walker DG, Beach TG, Taub DD. Alterations in immunological and neurological gene expression patterns in Alzheimer's disease tissues. Experimental Cell Research 2007;313(3):450-461.

Whitmer RA, Gunderson EP, Quesenberry CP Jr, Zhou J, Yaffe K. Body mass index in midlife and risk of Alzheimer disease and vascular dementia. Current Alzheimer Research 2007;4(2):103-109.

Williamson JD, Miller ME, Bryan RN, Lazar RM, Coker LH, Johnson J, Cukierman T, Horowitz KR, Murray A, Launer LJ, ACCORD Study Group. The Action to Control Cardiovascular Risk in Diabetes—Memory in Diabetes Study (ACCORD-MIND): rationale, design, and methods. American Journal of Cardiology 2007;99(12A):112i-122i.

Wilson RS, Arnold SE, Schneider JA, Kelly JF, Tang Y, Bennett DA. Chronic psychological distress and risk of Alzheimer’s disease in old age. Neuroepidemiology 2006;27(3):143-153.

Wilson RS, Schneider JA, Arnold SE, Tang Y, Boyle PA, Bennett DA. Olfactory identification and incidence of mild cognitive impairment in older age. Archives of General Psychiatry 2007;64(7):802-808.

Wolf PA, Beiser A, Elias MF, Au R, Vasan RS, Seshadri S. Relation of obesity to cognitive function: importance of central obesity and synergistic influence of concomitant hypertension: the Framingham Heart Study. Current Alzheimer Research 2007;4(2):111-116.

Yaffe K, Blackwell T, Barnes DE, Ancoli-Israel S, Stone KL, Study of Osteoporotic Fractures Group. Preclinical cognitive decline and subsequent sleep disturbance in older women. Neurology 2007;69(3):237-242.
Comments: 0
Votes:11