Literature
首页医源资料库在线期刊美国临床营养学杂志2000年71卷第2期

Weight loss in Alzheimer disease

来源:《美国临床营养学杂志》
摘要:ABSTRACTBackground:EpidemiologicstudieshaveshownthatweightlossiscommonlyassociatedwithAlzheimerdisease(AD)andisamanifestationofthediseaseitself。TheetiologyofweightlossinADappearsmultifactorial。Hypothesestoexplaintheweightlosshavebeensuggested(eg,atrophy......

点击显示 收起

Sophie Gillette-Guyonnet, Fatemeh Nourhashémi, Sandrine Andrieu, Isabelle de Glisezinski, Pierre Jean Ousset, Daniel Rivière, Jean-Louis Albarède and Bruno Vellas

1 From the Departments of Gerontology and Internal Medicine and Exploration of Respiratory Function and Sports Medicine, Purpan University Hospital, Toulouse, France, and INSERM U 518, Toulouse, France.

2 Reprints not available. Address correspondence to B Vellas, University of Toulouse, Toulouse, France. E-mail: 101333.1462{at}CompuServe.com.


ABSTRACT  
Background: Epidemiologic studies have shown that weight loss is commonly associated with Alzheimer disease (AD) and is a manifestation of the disease itself. The etiology of weight loss in AD appears multifactorial. Hypotheses to explain the weight loss have been suggested (eg, atrophy of the mesial temporal cortex, biological disturbances, and higher energy expenditure); however, none have been proven.

Objective: In the first part of this article, we describe weight loss in AD (epidemiologic data and hypotheses to explain weight loss and anorexia in AD). In the second part we report the results of a longitudinal study of the changes in nutritional variables in a cohort of patients with a probable diagnosis of AD.

Design: We followed subjects with AD (based on criteria of the National Institute of Neurological and Communicative Disorders and Stroke/Alzheimer's Disease and Related Disorders Association) who were recruited from the Alzheimer's Disease Center in Toulouse. All subject underwent a nutritional, neuropsychologic, and functional evaluation. The Zarit scales were used to assess caregiver burden and caregiver reactions to the patients' behavioral and autonomic disorders.

Results: We showed that only results of the Burden Interview and the Memory and Behavior Problems Checklist, which explored caregiver burden, predicted weight loss in AD. It is possible that caregivers who consider themselves overburdened by the disease process are not willing to invest adequate resources to allow AD patients to properly nourish themselves.

Conclusion: Nutritional education programs for the caregivers of AD patients seem to be the best way to prevent weight loss and improve the nutritional status of these patients.

Key Words: Alzheimer disease • weight loss • caregiver burden • elderly • nutrition education • aging


INTRODUCTION  
Alzheimer disease (AD) is a growing health issue and is one of the leading causes of death among elderly people. Three million people are currently affected in Europe. This disease is associated with cognitive and behavioral disorders and, frequently, with nutritional disorders such as weight loss and eating disorders. It seems important to have a better understanding of the pathogenesis of this AD-associated weight loss because it leads to reduced muscle mass, a loss of autonomy, and an increased risk of falls, decubitus ulcers, and systemic infection, which increase the burden of the disease and worsen the patient's and caregiver's quality of life.


WEIGHT LOSS IN ALZHEIMER DISEASE  
Epidemiology
Since the beginning of the 1980s, many studies have been carried out to compare the annual weight variation in AD patients and healthy control subjects (1–12). These studies reported that weight loss frequently occurs in the first stages of the disease (3–12), although patients usually have adequate energy intakes (5). Weight loss is actually listed as a symptom consistent with the diagnosis of AD in the most commonly used criteria for diagnosis, those of the National Institute of Neurological and Communicative Disorders and Stroke/Alzheimer's Disease and Related Disorders Association Work Group (13). Using data collected by the Consortium to Establish a Registry for Alzheimer's Disease, White et al (14, 15) examined the natural history of weight change and the frequency and severity of weight loss. They recruited 362 AD patients and 317 control subjects and determined the percentage of patients who lost or gained 5% of their body weight during participation in the study. The entry criteria excluded persons with advanced dementia. White et al found that almost twice as many AD patients as control subjects lost 5% of their initial body weight. The same finding was observed for a loss of 10% of initial body weight. Surprisingly, AD patients were also more likely to gain 5% of their initial body weight than were control subjects. In a recent study, White et al (16) determined the association of weight change in AD with the severity of the disease and mortality. They showed that the risk of weight loss tends to increase with the severity and progression of AD. According to these authors, weight loss is a predictor of mortality in subjects with AD, whereas weight gain appears to have a protective effect.

Individuals with dementia frequently develop serious feeding difficulties as their dementia progresses; accordingly, their weight probably decreases more over time than does that of nondemented individuals, and their survival might be compromised. To test this hypothesis, Wang et al (17) compared the weight variations over 48 mo of 2 institutionalized cohorts of subjects with dementia (n = 31 subjects with severe dementia and 48 with less-severe dementia) with those of a third cohort of institutionalized nondemented subjects (n = 26). Most of the AD patients had been diagnosed several years before the study began ( ± SD: 9.2 ± 5.4 y). The authors showed that men had a significantly higher mean body weight than did women. The longitudinal weight trends of the men and women were significantly different by two-factor repeated measures analysis of variance. The nondemented subjects had a significantly higher mean weight than did the demented patients at admission and throughout the longitudinal study period. All 3 cohorts maintained stable body weights throughout their institutional stay. The mean weights of the patients with severe dementia were similar to those with less-severe dementia. The authors concluded that dementia was not necessarily associated with continuous weight loss during institutionalization, despite the frequent occurrence of eating disorders and episodes of weight loss.

Wang et al (17) reported that of the 105 subjects they studied, 3 women developed dementia after they were institutionalized. The mean (±SD) weight of these 3 women at the time of entry to the study was 58.5 ± 18 kg compared with 64 ± 21 kg for the 18 women who did not develop dementia during the 4 y of the study. These results suggest, as described previously (18), that weight loss may precede dementia. Indeed, Barrett-Connor et al (18) studied weight changes in 299 free-living elderly subjects who were followed for 20 y before they were determined to be cognitively intact or to have mild-to-moderate dementia. For all of the men and women who received a diagnosis of AD (n = 60), there was a significant decrease in weight (5 kg) since the first evaluation.

Some hypotheses to explain weight loss
Weight loss in AD occurs frequently in the first stages of the disease, although patients usually have adequate energy intakes. Many hypotheses have been proposed to explain the weight loss.

Atrophy of the mesial temporal cortex
The mesial temporal cortex (MTC), which is involved in feeding behavior and memory, is affected in the primary stages of AD and continues to be a site of major AD pathology as the disease worsens. Grundman et al (19) showed in AD that a low body mass index correlates best and specifically with atrophy of the MTC. They suggested that there is a connection between limbic system damage and low body weight in AD. Atrophy of the MTC might contribute to weight loss through additional mechanisms other than cognitive impairments (eg, lack of food preparation, diminished skills, reduced appetite, and increased physical activity). Moreover, some disturbances associated with weight loss, such as increased cortisol (20) and tumor necrosis factor (21) concentrations or decreased estrogen concentrations (22), worsen atrophy of the MTC and, consequently, dementia itself.

Higher energy requirements
It has been suggested that AD patients have higher energy requirements than do healthy individuals, which may contribute to their unexplained weight loss (23–25). Donaldson et al (26) examined whether the resting metabolic rate, the largest component of daily energy expenditure, was higher in 25 AD patients [Mini-Mental State Examination (MMSE) score (27): 17 ± 8 (possible score of 0–30, with 30 being the best score.] than in 73 healthy, older, control subjects. Donaldson et al showed that the resting metabolic rate was not significantly different between AD patients and control subjects when measured on an absolute basis or when normalized for body composition and age. In AD patients, resting energy expenditure was adapted to body composition.

Poehlman et al (28) tested the hypothesis that daily energy expenditure would be higher in 30 AD patients with mild-to-moderate AD (MMSE score: 16 ± 8) than in 103 healthy subjects. Fat-free mass tended to be lower in AD patients than in control subjects, whereas no significant differences were noted in fat mass. Daily energy expenditure was 14% lower in AD patients than in healthy elderly subjects. This difference was explained by a lower resting metabolic rate and a lower physical activity expenditure in the AD patients. No significant difference in energy expenditure was found between the AD patients and the control subjects after adjustment for body composition. Thus, there was no evidence of an increase in daily energy expenditure linked to AD to explain the weight loss. In AD patients, energy expenditure is appropriate for their body size.

Biological disturbances
Hyperinsulinemia and insulin resistance have been reported in subjects with AD (29). However, these metabolic disturbances may be due to weight gain or physical inactivity rather than to AD per se. Furthermore, neuropeptide Y (NPY) may directly affect energy balance because of its potent effects on food intake, energy expenditure, and body weight (30). A decline in orexigenic factor concentrations, such as NPY and norepinephrine, has been reported in AD patients and may be involved in anorexia, which frequently occurs at the end of the disease (31).

Anorexia in Alzheimer disease
AD is generally associated with a progressive change in nutritional behavior. Anorexia may occur in the late stage of the disease and contribute to the associated weight loss. It may be due to physical changes [eg, decreased taste and smell functions (32), decreased appetite related to a decline in endogenous opioids, and increased satiety related to an increase in sensitivity to cholecystokinin], neuropsychiatric disorders associated with the disease (eg, memory loss, disorientation, mood disorders, indifference, and impaired judgment), a change in autonomy and dietary habits, or changes in neurotransmitter concentrations (eg, NPY and norepinephrine).

Dronabinol, one of the main components of marijuana, is associated with increased food intake. It also improves appetite and the sense of well-being and causes weight gain. Volicer et al (33) studied the effect of dronabinol in 12 hospitalized AD patients (MMSE score: 4.0 ± 7.4) who refused to eat. The study used a double-bind, placebo-controlled, crossover design with each study period lasting 6 wk. During the study period, nutritional status was monitored by weekly measurements of body weight and triceps skinfold thickness and by biological markers (eg, albumin and lymphocytes). Energy intake was also calculated. Disturbed behavior was measured by the Cohen-Mansfield Agitation Inventory (34; possible score: 0–91, with 0 being the best score). Body weight of study subjects increased more during dronabinol treatment than during the placebo periods. Energy intake did not change significantly during the study period and was similar in the placebo and dronabinol periods. Dronabinol treatment decreased the severity of disturbed behavior and this effect persisted during the placebo period in patients who received dronabinol first. It is possible, according to the authors, that the weight gain was partly caused by decreased agitation in the patients, which could have decreased their energy requirements. These results indicate that dronabinol is a promising novel therapeutic agent to treat anorexia but also to improve disturbed behavior observed in AD patients.

In summary, several epidemiologic studies have indicated an association between AD and weight loss. This weight loss could occur throughout the disease process, but it could be more common as the illness becomes more severe and anorexia develops. According to some authors (18), weight loss could be a manifestation of the disease itself. The etiology of weight loss in AD appears multifactorial and some hypotheses have been proposed to explain it (eg, atrophy of the MTC, biological disturbances, and high energy expenditure); however, none of these hypotheses has been proven.


A LONGITUDINAL STUDY OF NUTRITIONAL STATUS IN ALZHEIMER DISEASE  
No work to date has prospectively studied the evolution of nutritional variables (weight, anthropometric and biological markers, and energy intakes) in AD patients; therefore, this longitudinal study was conducted, which followed the nutritional status of a cohort of patients diagnosed with probable AD.

Subjects and methods
Subjects
Subjects were recruited from the Alzheimer's Disease Center in Toulouse. All patients met the National Institute of Neurological and Communicative Disorders/Alzheimer's Disease and Related Disorders Association criteria for probable AD (13). Assessment batteries included a detailed case history and physical examination; cerebral computed tomography to exclude strokes, tumors, etc; blood chemistry screening to rule out thyroid dysfunction, vitamin B-12 deficiency, and other metabolic and hematologic abnormalities; as well as neuropsychologic tests to establish the above cited criteria. Patients were all community dwelling and had a responsible caregiver who lived with them and who was available to inform us about the activities of the AD patients.

Methods
All subjects had a nutritional assessment, which included measurements of anthropometric (weight, height, body mass index, skinfold thickness, and circumferences) and biological (albumin, prealbumin, C-reactive protein, and orosomucoid) markers. A Mini Nutritional Assessment (MNA; 35), used to evaluate the nutritional state of the elderly, was also conducted on every patient. Dietary assessment was accomplished as follows: for a period of 3 d, caregivers kept a food diary of everything the patients drank or ate. Portion size was expressed in household units. The ingredients of mixed dishes were specified. A dietitian collected and inspected these nutritional surveys and the results were analyzed by using the Dupin food-composition table (36) augmented with certain regional foods (37). The data for the dietary investigations are only available for all the subjects for the first year of follow-up.

A neuropsychologic and functional evaluation was performed on each patient by using many standardized and validated tools, such as the MMSE. The neuropsychologic evaluation included the following: the Cornell scale for depression (38; possible score: 0–38, with 0 being the best score), an adaptation of the Cohen-Mansfield Agitation Inventory for behavioral problems, the Reisberg scale (39; possible score: 0–7, with 0 being the best score), and the Hierarchic Dementia Scale (HDS; possible score: 0–200, with 0 being the best score) for dementia severity (40). Patient autonomy was screened by using the activities of daily living (ADL; possible score: 0–6, with 6 being the best score) and instrumental ADL (IADL; possible score: 0–8, with 8 being the best score) scales (41, 42). The Zarit scales were used to assess caregiver burden (Burden Interview; possible score: 0–88, with 0 being the best) and caregiver reactions to the AD patients' behavioral and autonomic disorders (Memory and Behavior Problems Checklist; possible score: 0–240, with 0 being the best) (43).

To study weight change, we considered a weight loss of 4% of body weight in 1 y to be significant (44). We researched significant weight loss during the first year of follow-up to identify subjects with weight variations and those whose weight remained stable throughout the study. We used the following equation:

REFERENCES  

  1. Morgan DB, Hullin RP. The body composition of the chronically mentally ill. Hum Nutr Clin Nutr 1982;36:439–48.
  2. Seth RV. Review: weight loss in Alzheimer's disease. Int J Geriatr Psychiatry 1994;9:605–10.
  3. Sandman PO, Adolfsson R, Nygren C, et al. Nutritional status and dietary intake in institutionalized patients with Alzheimer's disease and multiinfarct dementia. J Am Geriatr Soc 1987;35:31–8.
  4. Tavares AR, Rabins PV. Weight loss in Alzheimer's disease: a longitudinal study. Zeitschr Alternsforsch 1987;42:165–7.
  5. Singh S, Mulley GP, Losowsky MS. Why are Alzheimer patients thin? Age Ageing 1988;17:21–8.
  6. Burns A, Marsh GP, Bender D. Dietary intake and clinical anthropometric and biochemical indices of malnutrition in elderly demented patients and non-demented subjects. Psychol Med 1989;19:383–91.
  7. O'Neill D, McKiernan M, Gibney M, et al. Dietary and anthropometric measures in mild to moderate senile dementia of the Alzheimer type (SDAT). J Hum Nutr Dietetics 1990;3:177–81.
  8. Morgan DB, Nexton HMV, Schorah CJ, et al. Abnormal indices of nutrition in the elderly: a study of different clinical groups. Age Ageing 1986;15:65–7.
  9. Renvall MJ, Spindler AA, Ramsdell JW, Paskvan M. Nutritional status of free-living Alzheimer's patients. Am J Med Sci 1989;298:20–7.
  10. Berlinger WG, Potter JF. Low body mass index in demented outpatients. J Am Geriatr Soc 1991;39:973–8.
  11. Wolf-Klein GP, Silverstone FA, Levy AP. Nutritional patterns and weight change in Alzheimer patients. Int Psychogeriatr 1992;4:103–11.
  12. Du W, DiLuca C, Growdon JH. Weight loss in Alzheimer's disease. J Geriatr Psychiatr Neurol 1993;6:34–7.
  13. McKhann G, Drachman D, Folstein M, et al. Clinical diagnosis of Alzheimer's disease: report of the NINCDS-ADRDA Work Group under the auspices of Department of Health and Human Services Task Force on Alzheimer's disease. Neurology 1984;34:939–44.
  14. White H, Pieper C, Schmader K, Fillenbaum G. A longitudinal analysis of weight change in Alzheimer's disease. J Am Geriatr Soc 1995;43:SA5 (abstr A17).
  15. White H, Pieper C, Schmader K, Fillenbaum G. Weight change in Alzheimer's disease. J Am Geriatr Soc 1996;44:1232–4.
  16. White H, Pieper C, Schmader K. The association of weight change in Alzheimer's disease with severity of disease and mortality: a longitudinal analysis. J Am Geriatr Soc 1998;46:1223–7.
  17. Wang SY, Fukagawa N, Hossain M, Ooi WL. Longitudinal weight changes, length of survival, and energy requirements of long term care residents with dementia. J Am Geriatr Soc 1997;45:1189–95.
  18. Barrett-Connor E, Edelstein SL, Corey-Bloom J, Wiederholt WC. Weight loss precedes dementia in community-dwelling older adults. J Am Geriatr Soc 1996;44:1147–52.
  19. Grundman M, Corey-Bloom J, Jernigan T, Archibald S, Thal LJ. Low body weight in Alzheimer's disease is associated with mesial temporal cortex atrophy. Neurology 1996;46:1585–91.
  20. Weiner MF, Vobach S, Svetlick D, Risser RC. Cortisol secretion and Alzheimer's disease progression: a preliminary report. Biol Psychol 1993;34:158–61.
  21. Fillit H, Wanhong D, Buee L, et al. Elevated circulating tumor necrosis factor levels in Alzheimer's disease. Neurosci Lett 1991; 129:318–20.
  22. Honjo H, Ogino Y, Naitoh K, et al. In vivo effects by estrone sulfate on central nervous system-senile dementia (Alzheimer's type). J Steroid Biochem 1989;34:521–5.
  23. Prentice AM, Leavesley K, Murgatroyd PR, et al. Is severe wasting in elderly mental patients caused by excessive energy requirements? Age Ageing 1989;18:158–67.
  24. Niskanen L, Piirainen M, Koljonen M, Usitupa M. Resting energy expenditure in relation to energy intake in patients with Alzheimer's disease, multi-infarct dementia and in control women. Age Ageing 1993;22:132–7.
  25. Poehlman ET. Regulation of energy expenditure in aging humans. J Am Geriatr Soc 1993;41:552–9.
  26. Donaldson KE, Carpenter WH, Toth MJ, et al. No evidence for a higher resting metabolic rate in noninstitutionalized Alzheimer's disease patients. J Am Geriatr Soc 1996;44:1232–4.
  27. Folstein FM, Folstein SE, McHugh PR. Mini-Mental state. A practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res 1975;12:189–98.
  28. Poehlman ET, Toth MJ, Goran MI, et al. Daily energy expenditure in free-living non-institutionalized Alzheimer's patients: a doubly labeled water study. Neurology 1997;48:997–1002.
  29. Razay G, Gordon KW. Hyperinsulinaemia and Alzheimer's disease. Age Ageing 1994;23:396–9.
  30. Billington CJ, Briggs JE, Harker S, et al. Neuropeptide Y in hypothalamic paraventricular nucleus: a center coordinating energy metabolism. Am J Physiol 1994;266:R1765–70.
  31. Valenti G. Neuropetide changes in dementia: pathogenetic implications and diagnostic value. Gerontology 1996;42:241–56.
  32. Knupfer L, Spiegel R. Differences in olfactory test performance between normal aged, Alzheimer and vascular type dementia individuals. Int Geriatr Psychiatry 1994;1:3–14.
  33. Volicer L, Stelly M, Morris J, et al. Effects of dronabinol on anorexia and disturbed behavior in patients with Alzheimer's disease. Int J Geriatr Psychiatry 1997;12:913–9.
  34. Cohen D, Eisdorfer C, Gorelick P, et al. Psychopathology associated with Alzheimer's disease and related disorders. J Gerontol 1993; 48:255–60.
  35. Guigoz Y, Vellas B, Garry PJ. Mini Nutritional Assessment: a practical assessment tool for grading the nutritional state of elderly patients. Facts Res Gerontol 1994;2:15–59.
  36. Dupin H, Abraham J, Giachetti I. Apports nutritionnels conseillés pour la population Francaise. (Recommended dietary allowances for the French population.) Paris: CNRS-CNERNA, 1992 (in French).
  37. Decarli B. Les enquêtes de consommation alimentaire. (Dietary intakes evaluated by 3 day diet records.) L'Année Gérontol 1994; 179–189 (in French).
  38. Alexoupoulos GS, Abrams RC, Young CA. Cornell scale for depression in dementia. Biol Psychiatr 1988;23:271–81.
  39. Reisberg B, Ferris SH, Mony D, De Leon J, Crook T. The global deterioration scale for assessment of primary degenerative dementia. Am J Psychiatry 1982;139:1136–9.
  40. Cole MG, Dastoor D. The hierarchic dementia scale. J Clin Exp Gerontol 1983;5:219–34.
  41. Katz S, Ford A, Moskowitz R. The index of ADL: a standardized measure of biological and psychosocial function. JAMA 1963; 185:914–9.
  42. Lawton MP, Moss M, Fulcomer M, Kleban MH. A research and service oriented multiple assessment instrument. J Gerontol 1982; 37:91–9.
  43. Zarit S, Zarit J. The memory and behavior problems checklist and the burden interview. University Park, PA: The Pennsylvania State University, 1983.
  44. Wallace JI, Schwartz RS, LaCroix AZ, et al. Involuntary weight loss in older outpatients: incidence and clinical significance. J Am Geriatr Soc 1995;43:329–37.
  45. Guyonnet S, Nourhashemi F, Andrieu S, et al. A prospective study of changes in nutritional status in Alzheimer's patients. Arch Gerontol Geriatr 1998;(suppl):255–62.
  46. Schulz R, O'Brien AT, Bookwala J, Fleissner K. Psychiatric and physical morbidity effects of dementia caregiving: prevalence, correlates, and causes. Gerontologist 1995;35:771–91.
  47. Vitaliano PP, Young HM, Russo J, et al. Does expressed emotion in spouses predict subsequent problems among care recipients with Alzheimer's disease? J Geriatr 1993;48:202–9.
  48. Dunkin JJ, Anderson-Hanley C. Dementia caregiver burden. A review of the literature and guidelines for assessment and intervention. Neurology 1998;51(suppl):S53–60.
  49. Fredman L, Daly MP. Weight change: a measure of caregiver stress. J Aging Health 1997;9:43–9.
  50. Rivière S, Lauque S, Vellas B. Health promotion programme: nutrition and Alzheimer's disease. In: Vellas B, Rivière S, Fitten J, eds. Research and practice in Alzheimer's disease focused on weight loss and eating behaviour in Alzheimer's patients. Paris: Serdi Publisher, 1998:93–103.

作者: Sophie Gillette-Guyonnet
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具