Mild Cognitive Impairment

Back

Practice Essentials

In mild cognitive impairment (MCI), the changes in cognition exceeds the normal, expected changes related to age. In one classification of MCI, the amnestic form is distinguished from the nonamnestic form. The amnestic form often precedes Alzheimer disease.

Signs and symptoms

Symptoms of mild cognitive impairment (MCI) are often vague and include the following:

Ronald C. Petersen postulated that the defining element of MCI is a single sphere of slowly progressive cognitive impairment that is not attributable to motor or sensory deficits and to which other areas of involvement may eventually be added, before social or occupational impairment supervenes (because this occurrence marks the onset of dementia).[1]

Diagnosis

Although no single feature of the general physical examination characterizes MCI, the following should be included in the overall assessment of the patient:

No specific diagnostic studies exist for mild cognitive impairment. However, most clinicians perform a basic workup at minimum to exclude potential treatable causes (eg, thyroid disease, cobalamin deficiency). Research is ongoing in the search for biologic markers that may help differentiate between the large number of conditions that may progress from MCI to full dementia.

Brain imaging with magnetic resonance imaging (MRI) or computed tomography (CT) is often performed in patients with MCI. In general, MRI is preferred, as whole brain and hippocampal volume on MRI can predict progression from MCI to Alzheimer disease (AD).[2] However, there are no established parameters to integrate this finding into the routine diagnosis and management of MCI. In addition, there is also some preliminary evidence for the use of baseline FDG-PET of the brain in conjunction with episodic memory assessment to predict conversion to AD.[3]

There are no stipulated neuropsychological tests for patients with MCI, nor are there predetermined cutoff points (eg, 1.0, 1.5, or 2 standard deviations below the mean). However, clinicians use the results from standardized memory and cognitive tests to determine whether these data represent significant changes from a patient’s presumed baseline. In general, serial testing is required to establish whether the patient’s cognitive function is improving, staying stable, or progressing to full-blown clinical dementia.

Management

Although no established treatment exists for MCI, donepezil delays the progression to AD in MCI patients with depression without affecting their depressive symptoms, and some evidence suggests that cognitive interventions may have a positive effect.[4] Cholinesterase inhibitors have not been found to delay the onset of AD or dementia in MCI.[5]

Identify and monitor patients with MCI, because of their increased risk for AD (and, to a lesser extent, other dementing conditions). In addition, to the extent possible, correct any sensory and motor manifestations that compound their cognitive symptoms.

Diet and activity may have some positive effects in patients with MCI. The risk of developing MCI is lower in individuals who consume a Mediterranean diet,[6] and interactive, mentally challenging activities as well as moderate exercise have the potential to be helpful in MCI.[7]

Overview

Various terms have been employed to characterize the cognitive decline associated with aging, including benign senescent forgetfulness, age-associated memory impairment, and age-associated cognitive decline.[8, 9, 10] The term mild cognitive impairment (MCI) is intended to represent an intermediate stage between normal aging and the development of pathologic aging and dementia (eg, malignant senescent forgetfulness[9] ).

Other terms with connotations similar to those of MCI include isolated memory impairment, incipient dementia, and dementia prodrome. However, these terms are not nearly as widely accepted as MCI and should not be considered exact synonyms.

Of the normal memory functions, some decline substantially with increasing age, and some do not. Memory functions that remain relatively stable with increasing age include the following:

Memory functions that decrease with age include the following:

To demonstrate that a patient’s cognitive function is worse than would normally be expected for his or her age, neuropsychological testing is necessary so that the patient’s performance can be compared with that of an age-matched (and, ideally, education-matched) control group.

Mild degrees of cognitive impairment, particularly when self-reported by patients, pose a substantial challenge to the clinician. The physician may be dealing with a patient with a mild or transient condition, a drug-induced adverse effect, or a depressive disorder; the patient may be in the early stages of a condition that will eventually lead to a dementia; or the complaint may be due to a psychological condition rather than an organic brain disorder.

Because a variety of conditions may result in a complaint of cognitive impairment, an individualized workup for such conditions and a consensus on a therapeutic approach should be sought. To date, no medications have been approved by the US Food and Drug Administration (FDA) for the treatment of MCI.

For patient education resources, see the Dementia Center, as well as Possible Early Dementia.

Pathophysiology

In mild cognitive impairment (MCI), cognitive impairment exceeds the normal expected age-related changes, but functional activities are largely preserved; thus, MCI does not meet the criteria for dementia.[1] Different subtypes of MCI are recognized. One common classification distinguishes between amnestic and nonamnestic forms of MCI.

Amnestic MCI, in which memory impairment predominates, is often a precursor of clinical Alzheimer disease (AD). Nonamnestic forms of MCI are characterized by a variety of cognitive impairments, the most common of which is probably impaired executive function. A substantial number of patients with MCI may be judged to have normal cognition on follow-up visits.

The pathophysiology of MCI is multifactorial. Most cases of amnestic MCI result from pathologic changes of AD that have not yet become severe enough to cause clinical dementia.[16] At least in specialty research populations, autopsies done on amnestic MCI patients have found the neuropathology to be typical of AD.[17] Nonamnestic MCI may be associated with cerebrovascular disease, frontotemporal dementias (as a precursor), or no specific pathology.

Etiology

Mild cognitive impairment (MCI) is heterogeneous both in its clinical manifestations and in its etiology. Given that amnestic MCI often results from Alzheimer disease (AD) pathology, it is not surprising that most patients with amnestic MCI progress to clinical AD within 6 years. Nonamnestic forms of MCI may be due to cerebrovascular disease, Lewy body dementia, Parkinson disease, frontotemporal dementias, atypical Alzheimer disease, or no specific underlying pathology.

Mood disorders, medical illness, and medications may affect cognition in such a way that a patient will meet criteria for MCI (usually nonamnestic MCI). Many such patients have normal neuropsychological test results when reevaluated a year later.

According to a large population-based study conducted in 2016, there is no significant link between exposure to general anesthesia and the development of MCI in individuals aged 40 years and older. Anesthesia exposure, assessed as a dichotomous variable, was not associated with MCI nor was there was a link between the number of anesthesia exposures and MCI.[18] A study in 2013 also had similar findings; those results showed exposure to general anesthesia during medical procedures after age 45 years is not a risk factor for dementia.[19]  However, these data do not exclude the possibility that anesthetic exposures occurring later in life may be associated with an increase in the rate of incident MCI.

Epidemiology

Annual prevalence estimates for mild cognitive impairment (MCI) range from 12% to 18% in persons older than 60 years, a finding reflected in multiple international studies.[20, 21, 22, 23] Among community-dwelling African Americans, the estimated prevalence is 19.2% for those aged 65–74 years, 27.6% for those aged 75–84 years, and 38% for those aged 85 years and older.[24]

The prevalence of mild cognitive impairment increases with age. The prevalence is 10% in those aged 70–79 years and 25% in those aged 80–89 years.[25] Many studies indicate that the risk of Alzheimer disease (AD) is significantly higher in women than in men, and it is therefore presumed that the likelihood of developing MCI is greater in women than in men. Virtually nothing is known about cultural and racial factors influencing the clinical manifestations of MCI.

Clinical Presentation

Patient history

Patients with mild cognitive impairment (MCI) often present with vague and subjective symptoms of declining cognitive performance, which may be difficult to distinguish from the typical performance decline in healthy older individuals. The most common symptom is said to be memory loss, in keeping with the prevalent view that amnestic MCI is the most common type. However, some authorities affirm that the most common form of MCI affects multiple spheres of cognition.

Less common presentations of MCI include language disturbance (eg, difficulty in finding words), attention deficit (eg, difficulty in following or focusing on conversations), and deterioration in visuospatial skills (eg, disorientation in familiar surroundings in the absence of motor and sensory conditions that would account for the complaint).

Dissociating purely cognitive symptoms from those attributable to various degrees of sensory deprivation (eg, hearing loss or loss of visual acuity) that tend to coexist in the same patient population is often difficult and may be compounded by motor deficits that also beset the same individuals.

In any case, the defining element of MCI, as postulated by Petersen, is a single sphere of slowly progressive cognitive impairment that is not attributable to motor or sensory deficits and to which other areas of involvement may eventually be added, before social or occupational impairment supervenes (because this occurrence marks the onset of dementia).[1] Virtually nothing is known about the average duration of these manifestations before they are brought to medical attention (if they ever are).

Clinicians should rely on their own judgment in deciding when safety-related questions that are appropriate for patients with dementia—for example, about weapons, driving, and possible home fires involving cigarettes, stoves, or fireplaces—should also be asked of patients with MCI.

Physical examination

No feature of the general physical examination is characteristic of MCI. Nevertheless, a thorough physical examination should be performed as part of the general evaluation in an effort to determine whether any conditions capable of causing MCI (eg, thyroid disease, cobalamin deficiency, or venereal disease) are present and whether there are any sensory and motor deficits that could explain or compound the symptoms. Mental status examination is also important for documenting the degree of cognitive dysfunction.

Differential Diagnosis

Mild cognitive impairment (MCI) may result from virtually any disorder that causes brain dysfunction. Common causes include the following:

According to an analysis of 5150 patients aged 65 years or older from the Cardiovascular Health Study, patients with atrial fibrillation (AF) reach clinical thresholds for cognitive impairment and dementia at an earlier age than patients without AF, even in the absence of clinical stroke.[27, 28]

Depressive disorders are particularly prevalent in older adults (approximately 15%), who frequently exhibit vague somatic symptoms and anxiety and report inability to concentrate and poor memory. These patients typically deny a sad mood but often admit to sleep symptoms, lack of interest in things they used to enjoy, loss of appetite, and lack of motivation. Depression may certainly be accompanied by cognitive dysfunction that abates with successful treatment of the depression.

The association between depression and AD and other dementias is likely to be complex, and depression may be misdiagnosed in the realm of dementia. Framingham data have helped bolster the epidemiologic association, documenting a 50% increase in AD and dementia in those who were depressed at baseline.[29] During a 17-year follow-up period, a total of 21.6% of participants who were depressed at baseline developed dementia, compared with 16.6% of those who were not depressed.

In another related study, recurrent depression was noted to be particularly pernicious: having 1 depression episode conferred an 87–92% increase in dementia risk, whereas having 2 or more episodes nearly doubled the dementia risk (but did not increase the risk of incident MCI).[30]

Laboratory Studies

No specific laboratory studies are indicated for mild cognitive impairment (MCI). Most practitioners perform at least a basic workup to rule out treatable conditions that may cause dementia, such as thyroid disease and cobalamin deficiency. These assessments are not mandatory, however.

A search for biologic markers of MCI that may help distinguish among the many conditions that lead from MCI to full-blown dementia is ongoing. As yet, however, no unanimous agreement has been reached, and potentially useful markers, such as functional and structural abnormalities found on imaging studies (eg, hippocampal atrophy and cerebral hypoperfusion) and putative biochemical markers (eg, apolipoprotein E epsilon 4 allele), remain controversial.

Neuropsychological Testing

Neuropsychological testing is required in instances of mild cognitive impairment (MCI) to demonstrate that the patient is below some cutoff point on standardized memory tests (as well as other cognitive tests). However, the exact cutoff point (be it 1.0, 1.5, or 2 standard deviations below the mean) and the particular neuropsychological tests to be used are not stipulated.

Because few MCI patients have undergone baseline testing on these measures before the onset of the impairment, the clinician will have to determine whether a particular score represents a significant change from a patient’s presumed baseline. Such determinations are inexact, and serial testing eventually will be needed to establish whether the patient’s cognitive function is improving, staying stable, or progressing to full-blown clinical dementia. A useful aspect of this testing is the ability of the neuropsychologist to establish a profile for the patient based on their gender, age, and education, and to then evaluate if their level of function is adequate for that profile.

The Alzheimer's Association released guidelines, including an algorithm, to help clinicians in the primary care setting detect cognitive impairment and determine whether referral or further testing is needed.[3] The algorithm includes the following components[2, 3] :

The following 3 cognitive assessment tools are recommended for routine use by primary care physicians[9] :

Additionally, the Alzheimer’s Association recommends the following 3 cognitive assessment tools for use with the patient's spouse, family, or friends[8, 9] :

Treatment and Management

Medical care

At present, no established treatment exists for mild cognitive impairment (MCI). Cholinesterase inhibitors have not been found to delay the onset of Alzheimer disease (AD) or dementia in individuals with MCI; however, donepezil has been found to delay the progression to AD in MCI patients with depression without affecting their symptoms of depression.[5] There is some evidence to suggest that cognitive interventions may have a positive effect.[4]

A practice parameter recommendation by the American Academy of Neurology states that patients with MCI should be identified and monitored because of their increased risk for AD and, to a lesser extent, other dementing conditions. Obviously, correcting (to the extent possible) any sensory and motor manifestations compounding the cognitive symptoms is important for minimizing their impact on MCI.

Particular attention should be given to the need to make a legal statement about the competency of patients to handle their own affairs. Because patients with MCI are by definition not demented, they usually do not need to assign power of attorney to anyone else—unlike patients with AD, who eventually will need such help.

The results of a study that included 361 subjects with AD, vascular dementia, or mixed dementias suggested that centrally acting angiotensin-converting enzyme inhibitors (CACE-Is) may reduce the rate of cognitive decline in patients with dementia, regardless of blood pressure levels at the time of their hypertension diagnosis.[31, 32] The rate of cognitive change was slowed in the first 6 months after dementia patients started taking these drugs.

Diet

Roberts et al found that the risk of developing MCI is lower in individuals who consume a Mediterranean diet, which is high in vegetables and unsaturated fats.[6]

A randomized, double-blind, placebo-controlled trial involving 25 elderly subjects with MCI determined that dietary supplementation with an oily emulsion of docosahexaenoic acid (DHA)-phospholipids containing melatonin and tryptophan yielded significant improvements in several measures of cognitive function as compared with supplementation with the placebo.[33]

According to one study of 663 adults in Singapore 60 years of age and older, consuming more than two standard portions of mushrooms per week may reduce odds of developing MCI by as much as 50%.[34]

Activity

Because physical, social, and mental activity are often recommended for patients with AD and because MCI often heralds AD, many experts have suggested that mentally challenging activities (eg, crossword puzzles and brain teasers) may be helpful for patients with MCI. Although there is no definitive proof that these exercises are efficacious, recommending them to patients with MCI seems advisable.

Such exercises should be kept to a level of difficulty that is reasonable for the patient. Ideally, they should be interactive rather than passive, and they should be administered in a fashion that does not cause excessive frustration. If an activity is not enjoyable or stimulating for the patient, it is unlikely to offer much cognitive benefit. In such cases, searching for other similar cognitive activities may be beneficial.

Social isolation can be minimized through referral to senior community centers or a day treatment program. Cognitive retraining and rehabilitative strategies offer considerable promise in MCI[35] and are therefore being explored.

A growing body of evidence suggests that physical activity and exercise are beneficial for brain health. A prospective study suggested that engaging in moderate exercise of any frequency in midlife or late life was associated with reduced odds of having MCI.[7]  According to one study, aerobic exercise was associated with a slight improvement in cognition.[36]  The 2017 update of the American Academy of Neurology guideline on mild cognitive impairment (MCI), which is endorsed by the Alzheimer's Association, recommends that patients with MCI exercise regularly as part of an overall approach to managing their symptoms.[37]  However, a 2018 study published in BMJ suggests rigorous exercise is not effective against dementia. Researchers assigned 329 participants to an aerobic and strength exercise program and 165 to usual care (n=494). The primary outcome was score on the Alzheimer’s disease assessment scale-cognitive subscale (ADAS-cog) at 12 months. Secondary outcomes included activities of daily living, neuropsychiatric symptoms, health-related quality of life, and carer quality of life and burden. Results show mean ADAS-cog score had increased to 25.2 (SD 12.3) in the exercise arm and 23.8 (SD 10.4) in the usual care arm, which indicates greater cognitive impairment in the exercise group, but the clinical significance of this finding is uncertain. Overall, the study found that a moderate to high intensity aerobic and strength exercise training program does not slow cognitive impairment in people with mild to moderate dementia.[38]

Another study showed certain activities to lower risk of MCI in cognitively normal individuals older than 70 years. These included playing games, reading magazines, being engaged in crafts, computer use, and social activities. Among these, being social and using computers were shown to reduce risk of MCI in people who were APOE4 carriers as well.[39]

Prognosis

Many patients with mild cognitive impairment (MCI) eventually experience progressive deterioration in their abilities to perform activities of daily living, cognition, and behavior.

Subtypes of MCI progress to Alzheimer disease (AD) at different rates. A study by Rountree et al showed that the conversion rate to AD was 56% for amnestic MCI, 50% for amnestic-subthreshold MCI, and 52% for nonamnestic MCI.[40] For all MCI subtypes, the 4-year conversion rate to dementia was 56% (14% annually), and that to Alzheimer disease was 46% (11% annually).In comparison, healthy elderly individuals develop AD at a rate of 1-2% per year.

Boyle et al reported that patients with MCI are almost 7 times more likely to develop AD than are older individuals without cognitive impairment.[41] Of patients with MCI, 80% are said to progress to dementia after approximately 6 years. This is a significant finding, given that AD is often cited as the fourth leading cause of death in the United States.

At least one well-designed study has shown MCI, as identified by the Short Portable Mental Status Questionnaire, to be an independent predictor of mortality.[42] Wilson et al reported that in both African American and white patients, the risk of death was increased by about 50% among individuals with MCI and was nearly 3 times higher among those with AD.[43]

Ultimately, long-term follow-up and eventual autopsy are necessary to distinguish between patients experiencing MCI due to preclinical AD and patients experiencing MCI due to less frequently occurring conditions. However, there are some factors that can be helpful in predicting the likelihood of progression.

The severity of memory impairment is predictive of progression to AD: patients with more severe memory impairment are more likely to progress. There are certain neuroradiologic features that predict progression of MCI. These include MRI findings of atrophy and volume loss in the medial temporal lobe as well as a hypometabolic pattern on FDG-PET scan.[44, 45, 3] In addition, APOE4 genotype carriers are at higher risk of progression, but APoE4 testing is not recommended for routine use.[46] A new modality that might prove useful in predicting and monitoring progression of MCI is a new PET tracer focusing on the role of tau. Early data suggests that spread of tau laterally, outside the medial temporal lobe, may predict a poor prognosis and a more rapid progression.[47]

Guidelines Summary

Alzheimer’s Association

The Alzheimer's Association guidelines, including an algorithm, help clinicians in the primary care setting detect cognitive impairment and determine whether referral or further testing is needed.[8, 9] The algorithm includes the following components[8, 9] :

The following 3 cognitive assessment tools are recommended for routine use by primary care physicians[48, 49] :

Additionally, the Alzheimer’s Association recommends the following 3 cognitive assessment tools for use with the patient's spouse, family, or friends[48, 49] :

American Academy of Neurology

The 2017 update of the American Academy of Neurology guideline on mild cognitive impairment (MCI), which is endorsed by the Alzheimer's Association, recommends that patients with MCI exercise regularly as part of an overall approach to managing their symptoms.[37] The guideline also advises clinicians to do the following:

What is mild cognitive impairment (MCI)?Which memory functions remain stable with increasing age?Which memory functions decrease with age?How is mild cognitive impairment assessed?What is the clinical presentation of mild cognitive impairment (MCI)?What are the signs and symptoms of mild cognitive impairment (MCI)?How is mild cognitive impairment (MCI) defined?What is included in the clinical assessment of mild cognitive impairment (MCI)?How is mild cognitive impairment (MCI) diagnosed?How is mild cognitive impairment (MCI) treated?What is the pathophysiology of mild cognitive impairment (MCI)?What causes mild cognitive impairment (MCI)?What is the prevalence of mild cognitive impairment (MCI)?Which clinical history findings are characteristic of mild cognitive impairment (MCI)?Which physical findings are characteristic of mild cognitive impairment (MCI)?Which disorders are associated with mild cognitive impairment (MCI)?What is the role of lab testing in the workup of mild cognitive impairment (MCI)?What are the Alzheimer's Association guidelines for detection of mild cognitive impairment (MCI)?What is the role of neuropsychological testing in the diagnosis of mild cognitive impairment (MCI)?Which cognitive assessment tools should be used by primary care physicians to detect mild cognitive impairment (MCI)?Which cognitive assessment tools can be used with family and friends to detect mild cognitive impairment (MCI)?What is the prognosis of mild cognitive impairment (MCI)?What are the Alzheimer’s Association algorithm for detecting mild cognitive impairment (MCI) in primary care?Which assessment tools for detecting mild cognitive impairment (MCI) are recommended by the Alzheimer’s Association?What are the AAN guidelines on mild cognitive impairment (MCI)?What is the approach to treatment of mild cognitive impairment (MCI)?Which dietary modifications are used in the treatment of mild cognitive impairment (MCI)?Which activity modifications are used in the treatment of mild cognitive impairment (MCI)?

Author

Sonal Mehta, MD, Clinical Assistant Professor, Department of Neurology, University of South Carolina School of Medicine

Disclosure: Nothing to disclose.

Coauthor(s)

Priyantha Herath, MD, PhD, Director of Movement Disorders Clinic, Attenting Neurologist, Department of Neurology, University of South Carolina School of Medicine at Columbia

Disclosure: Nothing to disclose.

Chief Editor

Jasvinder Chawla, MD, MBA, Chief of Neurology, Hines Veterans Affairs Hospital; Professor of Neurology, Loyola University Medical Center

Disclosure: Nothing to disclose.

Additional Contributors

Heather S Anderson, MD, Associate Professor, Staff Neurologist, Department of Neurology, University of Kansas Alzheimer's Disease Center

Disclosure: Nothing to disclose.

Acknowledgements

Richard J Caselli, MD Professor, Department of Neurology, Mayo Medical School, Rochester, MN; Chair, Department of Neurology, Mayo Clinic of Scottsdale

Richard J Caselli, MD is a member of the following medical societies: American Academy of Neurology, American Association of Neuromuscular and Electrodiagnostic Medicine, American Medical Association, American Neurological Association, and Sigma Xi

Disclosure: Nothing to disclose.

Joseph Quinn, MD Assistant Professor, Department of Neurology, Portland VA Medical Center, Oregon Health Sciences University

Joseph Quinn, MD is a member of the following medical societies: American Academy of Neurology, Society for Neuroscience, and Society for Pediatric Radiology

Disclosure: Nothing to disclose.

Francisco Talavera, PharmD, PhD Adjunct Assistant Professor, University of Nebraska Medical Center College of Pharmacy; Editor-in-Chief, Medscape Drug Reference

Disclosure: Medscape Salary Employment

References

  1. Petersen RC. Conceptual overview. Petersen RC. Mild Cognitive Impairment: Aging to Alzheimer's Disease. 1-14. New York, NY: Oxford University Press, Inc; 2003.
  2. Risacher SL, Saykin AJ, West JD, Shen L, Firpi HA, McDonald BC; Alzheimer's Disease Neuroimaging Initiative (ADNI). Baseline MRI predictors of conversion from MCI to probable AD in the ADNI cohort. Current Alzheimer Research. 2009 Aug. 6(4):347-61. [View Abstract]
  3. Landau SM, Harvey D, Madison CM, Reiman EM, Foster NL, Aisen PS. Comparing predictors of conversion and decline in mild cognitive impairment. Neurology. 2010 Jul 20. 75(3):230-8. [View Abstract]
  4. Simon SS, Yokomizo JE, Bottino CM. Cognitive intervention in amnestic Mild Cognitive Impairment: A systematic review. Neurosci Biobehav Rev. 2012 Feb 1. 36(4):1163-1178. [View Abstract]
  5. Panza F, Frisardi V, Capurso C, D'Introno A, Colacicco AM, Chiloiro R, et al. Effect of donepezil on the continuum of depressive symptoms, mild cognitive impairment, and progression to dementia. Journal of the American Geriatrics Society. 2010 Feb. 58(2):389-90. [View Abstract]
  6. Roberts RO, Geda YE, Cerhan JR, Knopman DS, Cha RH, Christianson TJ, et al. Vegetables, Unsaturated Fats, Moderate Alcohol Intake, and Mild Cognitive Impairment. Dementia and Geriatric Cognitive Disorders. 2010 May 22. 29(5):413-423. [View Abstract]
  7. Geda YE, Roberts RO, Knopman DS, Christianson TJ, Pankratz VS, Ivnik RJ, et al. Physical exercise, aging, and mild cognitive impairment: a population-based study. Archives of Neurology. 2010 Jan. 67(1):80-6. [View Abstract]
  8. Crook T, Bartus RT, Ferris SH. Age-associated memory impairment: proposed diagnostic criteria and measures of clinical change. Dev Neuropsychol. 1986. 2:261-276.
  9. KRAL VA. Senescent forgetfulness: benign and malignant. Can Med Assoc J. 1962 Feb 10. 86:257-60. [View Abstract]
  10. Levy R. Aging-associated cognitive decline. Working Party of the International Psychogeriatric Association in collaboration with the World Health Organization. Int Psychogeriatr. 1994 Spring. 6(1):63-8. [View Abstract]
  11. Luo L, Craik FI. Aging and memory: a cognitive approach. Can J Psychiatry. 2008 Jun. 53(6):346-53. [View Abstract]
  12. Jenkins L, Myerson J, Joerding JA, et al. Converging evidence that visuospatial cognition is more age-sensitive than verbal cognition. Psychol Aging. 2000 Mar. 15(1):157-75. [View Abstract]
  13. Head D, Rodrigue KM, Kennedy KM, et al. Neuroanatomical and cognitive mediators of age-related differences in episodic memory. Neuropsychology. 2008 Jul. 22(4):491-507. [View Abstract]
  14. Hannon B, Daneman M. Age-related changes in reading comprehension: an individual-differences perspective. Experimental Aging Research. 2009. 35(4):432-56. [View Abstract]
  15. Parks CM, Decarli C, Jacoby LL, Yonelinas AP. Aging effects on recollection and familiarity: the role of white matter hyperintensities. Neuropsychology, development, and cognition. Section B, Aging, neuropsychology and cognition. Feb 2010. 19:1-17. [View Abstract]
  16. Morris JC, Storandt M, Miller JP, et al. Mild cognitive impairment represents early-stage Alzheimer disease. Arch Neurol. 2001 Mar. 58(3):397-405. [View Abstract]
  17. Markesbery WR, Schmitt FA, Kryscio RJ, et al. Neuropathologic substrate of mild cognitive impairment. Arch Neurol. 2006 Jan. 63(1):38-46. [View Abstract]
  18. Sprung J, Roberts RO, Knopman DS, Olive DM, Gappa JL, Sifuentes VL, et al. Association of Mild Cognitive Impairment With Exposure to General Anesthesia for Surgical and Nonsurgical Procedures: A Population-Based Study. Mayo Clin Proc. 2016 Jan 2. [View Abstract]
  19. Sprung J, Jankowski CJ, Roberts RO, Weingarten TN, Aguilar AL, Runkle KJ, et al. Anesthesia and incident dementia: a population-based, nested, case-control study. Mayo Clin Proc. 2013 Jun. 88 (6):552-61. [View Abstract]
  20. Busse A, Hensel A, Gühne U, Angermeyer MC, Riedel-Heller SG. Mild cognitive impairment: long-term course of four clinical subtypes. Neurology. 2006 Dec 26. 67 (12):2176-85. [View Abstract]
  21. Di Carlo A, Lamassa M, Baldereschi M, Inzitari M, Scafato E, Farchi G, et al. CIND and MCI in the Italian elderly: frequency, vascular risk factors, progression to dementia. Neurology. 2007 May 29. 68 (22):1909-16. [View Abstract]
  22. Ganguli M, Chang CC, Snitz BE, Saxton JA, Vanderbilt J, Lee CW. Prevalence of mild cognitive impairment by multiple classifications: The Monongahela-Youghiogheny Healthy Aging Team (MYHAT) project. Am J Geriatr Psychiatry. 2010 Aug. 18 (8):674-83. [View Abstract]
  23. Larrieu S, Letenneur L, Orgogozo JM, Fabrigoule C, Amieva H, Le Carret N, et al. Incidence and outcome of mild cognitive impairment in a population-based prospective cohort. Neurology. 2002 Nov 26. 59 (10):1594-9. [View Abstract]
  24. Unverzagt FW, Gao S, Baiyewu O, et al. Prevalence of cognitive impairment: data from the Indianapolis Study of Health and Aging. Neurology. 2001 Nov 13. 57(9):1655-62. [View Abstract]
  25. Roberts RO, Geda YE, Knopman DS, et al. The Mayo Clinic Study of Aging: design and sampling, participation, baseline measures and sample characteristics. Neuroepidemiology. 2008. 30(1):58-69. [View Abstract]
  26. Cheng G, Huang C, Deng H, Wang H. Diabetes as a risk factor for dementia and mild cognitive impairment: a meta-analysis of longitudinal studies. Intern Med J. 2012 Feb 28. [View Abstract]
  27. Jeffrey S. AF linked to earlier, more rapid cognitive decline. Medscape Medical News. June 7, 2013.
  28. Thacker EL, McKnight B, Psaty BM, Longstreth WT Jr, Sitlani CM, Dublin S, et al. Atrial fibrillation and cognitive decline: A longitudinal cohort study. Neurology. 2013 Jun 5. [View Abstract]
  29. Saczynski JS, Beiser A, Seshadri S, Auerbach S, Wolf PA, Au R. Depressive symptoms and risk of dementia: the Framingham Heart Study. Neurology. 2010 Jul 6. 75(1):35-41. [View Abstract]
  30. Dotson VM, Beydoun MA, Zonderman AB. Recurrent depressive symptoms and the incidence of dementia and mild cognitive impairment. Neurology. 2010 Jul 6. 75(1):27-34. [View Abstract]
  31. Anderson P. ACE Inhibitors May Slow Cognitive Decline. Medscape Medical News. Available at http://www.medscape.com/viewarticle/808589. Accessed: August 7, 2013.
  32. Gao Y, O'Caoimh R, Healy L, Kerins DM, Eustace J, Guyatt G, et al. Effects of centrally acting ACE inhibitors on the rate of cognitive decline in dementia. BMJ Open. 2013. 3(7):[View Abstract]
  33. Mariangela R, Annalisa O, Milena F, Marco M, Neldo A, Roberta C, et al. Effects of a diet integration with an oily emulsion of DHA-phospholipids containing melatonin and tryptophan in elderly patients suffering from mild cognitive impairment. Nutr Neurosci. 2011 Dec 20. [View Abstract]
  34. Feng L, Cheah IK, Ng MM, Li J, Chan SM, Lim SL, et al. The Association between Mushroom Consumption and Mild Cognitive Impairment: A Community-Based Cross-Sectional Study in Singapore. J Alzheimers Dis. 2019. 68 (1):197-203. [View Abstract]
  35. Galante E, Venturini G, Fiaccadori C. Computer-based cognitive intervention for dementia: preliminary results of a randomized clinical trial. G Ital Med Lav Ergon. 2007 Jul-Sep. 29(3 Suppl B):B26-32. [View Abstract]
  36. Lautenschlager NT, Cox KL, Flicker L, Foster JK, van Bockxmeer FM, Xiao J, et al. Effect of physical activity on cognitive function in older adults at risk for Alzheimer disease: a randomized trial. JAMA. 2008 Sep 3. 300 (9):1027-37. [View Abstract]
  37. Petersen RC, Lopez O, Armstrong MJ, Getchius TSD, Ganguli M, Gloss D, et al. Practice guideline update summary: Mild cognitive impairment: Report of the Guideline Development, Dissemination, and Implementation Subcommittee of the American Academy of Neurology. Neurology. 2018 Jan 16. 90 (3):126-135. [View Abstract]
  38. Lamb SE, Sheehan B, Atherton N, et al. Dementia And Physical Activity (DAPA) trial of moderate to high intensity exercise training for people with dementia: randomised controlled trial. BMJ. May 16, 2018. 361:
  39. Krell-Roesch, J., Roberts, R., Pink, A., et al. Mentally Stimulating Activities in Late-Life and the Risk of Incident Mild Cognitive Impairment: A Prospective Cohort Study. Neurology. 2016. 86:S35-007.
  40. Rountree SD, Waring SC, Chan WC, et al. Importance of subtle amnestic and nonamnestic deficits in mild cognitive impairment: prognosis and conversion to dementia. Dement Geriatr Cogn Disord. 2007. 24(6):476-82. [View Abstract]
  41. Boyle PA, Wilson RS, Aggarwal NT, et al. Mild cognitive impairment: risk of Alzheimer disease and rate of cognitive decline. Neurology. 2006 Aug 8. 67(3):441-5. [View Abstract]
  42. Sachs GA, Carter R, Holtz LR, Smith F, Stump TE, Tu W, et al. Cognitive impairment: an independent predictor of excess mortality: a cohort study. Ann Intern Med. 2011 Sep 6. 155(5):300-8. [View Abstract]
  43. Wilson RS, Aggarwal NT, Barnes LL, Bienias JL, Mendes de Leon CF, Evans DA. Biracial population study of mortality in mild cognitive impairment and Alzheimer disease. Archives of Neurology. 2009 Jun. 66(6):767-72. [View Abstract]
  44. Weiner MW, Veitch DP, Aisen PS, Beckett LA, Cairns NJ, Cedarbaum J, et al. Impact of the Alzheimer's Disease Neuroimaging Initiative, 2004 to 2014. Alzheimers Dement. 2015 Jul. 11 (7):865-84. [View Abstract]
  45. Dickerson BC, Salat DH, Greve DN, Chua EF, Rand-Giovannetti E, Rentz DM, et al. Increased hippocampal activation in mild cognitive impairment compared to normal aging and AD. Neurology. 2005 Aug 9. 65 (3):404-11. [View Abstract]
  46. Boyle PA, Buchman AS, Wilson RS, Kelly JF, Bennett DA. The APOE epsilon4 allele is associated with incident mild cognitive impairment among community-dwelling older persons. Neuroepidemiology. 2010. 34(1):43-9. [View Abstract]
  47. Johnson KA, Schultz A, Betensky RA, Becker JA, Sepulcre J, et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease. Ann Neurol. 2016 Jan. 79 (1):110-9. [View Abstract]
  48. Jeffrey S. New guidelines on screening for cognitive impairment. Medscape Medical News. December 21, 2012. Available at http://www.medscape.com/viewarticle/776548?src=wnl_edit_medn_fmed&spon=34. Accessed: January 8, 2013.
  49. Cordell CB, Borson S, Boustani M, Chodosh J, Reuben D, Verghese J, et al. Alzheimer's Association recommendations for operationalizing the detection of cognitive impairment during the Medicare Annual Wellness Visit in a primary care setting. Alzheimers Dement. 2012 Dec 19. [View Abstract]
  50. Ganguli M, Dodge HH, Shen C, et al. Mild cognitive impairment, amnestic type: an epidemiologic study. Neurology. 2004 Jul 13. 63(1):115-21. [View Abstract]
  51. Anderson P. Fornix changes may be first clue to cognitive decline. Medscape Medical News. September 11, 2013.
  52. Fletcher E, Raman M, Huebner P, Liu A, Mungas D, Carmichael O, et al. Loss of Fornix White Matter Volume as a Predictor of Cognitive Impairment in Cognitively Normal Elderly Individuals. JAMA Neurol. 2013 Sep 9. [View Abstract]