Skip to main content
AAN.com
Articles
March 26, 2008
Letter to the Editor

Central obesity and increased risk of dementia more than three decades later

September 30, 2008 issue
71 (14) 1057-1064

Abstract

Background: Numerous reports show that a centralized distribution of adiposity is a more dangerous risk factor for cardiovascular disease and diabetes than total body obesity. No studies have evaluated whether the same pattern exists with dementia. The objective was to evaluate the association between midlife central obesity and risk of dementia three decades later.
Methods: A longitudinal analysis was conducted of 6,583 members of Kaiser Permanente of Northern California who had their sagittal abdominal diameter (SAD) measured in 1964 to 1973. Diagnoses of dementia were from medical records an average of 36 years later, January 1, 1994, to June 16, 2006. Cox proportional hazard models adjusted for age, sex, race, education, marital status, diabetes, hypertension, hyperlipidemia, stroke, heart disease, and medical utilization were conducted.
Results: A total of 1,049 participants (15.9%) were diagnosed with dementia. Compared with those in the lowest quintile of SAD, those in the highest had nearly a threefold increased risk of dementia (hazard ratio, 2.72; 95% CI, 2.33–3.33), and this was only mildly attenuated after adding body mass index (BMI) to the model (hazard ratio, 1.92; 95% CI, 1.58–2.35). Those with high SAD (>25 cm) and normal BMI had an increased risk (hazard ratio, 1.89; 95% CI, 0.98–3.81) vs those with low SAD (<25 cm) and normal BMI (18.5–24.9 kg/m2), whereas those both obese (BMI >30 kg/m2) and with high SAD had the highest risk of dementia (HR, 3.60; 95% CI, 2.85–4.55).
Conclusions: Central obesity in midlife increases risk of dementia independent of diabetes and cardiovascular comorbidities. Fifty percent of adults have central obesity; therefore, mechanisms linking central obesity to dementia need to be unveiled.

Get full access to this article

View all available purchase options and get full access to this article.

REFERENCES

1.
Yarnell JW, Patterson CC, Thomas HF, Sweetnam PM. Central obesity: predictive value of skinfold measurements for subsequent ischaemic heart disease at 14 years follow-up in the Caerphilly Study. Int J Obes Relat Metab Disord 2001;25:1546–1549.
2.
Ramachandran A, Snehalatha C, Viswanathan V, Viswanathan M, Haffner SM. Risk of noninsulin dependent diabetes mellitus conferred by obesity and central adiposity in different ethnic groups: a comparative analysis between Asian Indians, Mexican Americans and Whites. Diabetes Res Clin Pract 1997;36:121–235.
3.
Onat A, Avci GS, Barlan MM, Uyarel H, Uzunlar B, Sansoy V. Measures of abdominal obesity assessed for visceral adiposity and relation to coronary risk. Int J Obes Relat Metab Disord 2004;28:1018–1025.
4.
Smith DA, Ness EM, Herbert R, et al. Abdominal diameter index: a more powerful anthropometric measure for prevalent coronary heart disease risk in adult males. Diabetes Obes Metab 2005;7:370–380.
5.
Rondinone CM. Adipocyte-derived hormones, cytokines, and mediators. Endocrine 2006;29:81–90.
6.
Whitmer RA. The epidemiology of adiposity and dementia. Curr Alzheimer Res 2007;4:117–122.
7.
Gustafson D. Adiposity indices and dementia. Lancet Neurol 2006;5:713–720.
8.
Whitmer RA, Gunderson EP, Barrett-Connor E, Quesenberry CP Jr, Yaffe K. Obesity in middle age and future risk of dementia: a 27 year longitudinal population based study. BMJ 2005;330:1360.
9.
Gustafson D, Rothenberg E, Blennow K, Steen B, Skoog I. An 18-year follow-up of overweight and risk of Alzheimer disease. Arch Intern Med 2003;163:1524–1528.
10.
Gustafson D, Lissner L, Bengtsson C, Bjorkelund C, Skoog I. A 24-year follow-up of body mass index and cerebral atrophy. Neurology 2004;63:1876–1881.
11.
Kivipelto M, Ngandu T, Fratiglioni L, et al. Obesity and vascular risk factors at midlife and the risk of dementia and Alzheimer disease. Arch Neurol 2005;62:1556–1560.
12.
Whitmer R, Gunderson EP, Quesenberry C, Zhou J, Yaffe K. Body mass index in midlife and risk of Alzheimer disease and vascular dementia. Curr Alzheimer Res 2007;4:103–109.
13.
Baumgartner RN, Heymsfield SB, Roche AF. Human body composition and the epidemiology of chronic disease. Obes Res 1995;3:73–95.
14.
Snijder MB, van Dam RM, Visser M, Seidell JC. What aspects of body fat are particularly hazardous and how do we measure them? Int J Epidemiol 2006;35:83–92.
15.
Krieger N. Overcoming the absence of socioeconomic data in medical records: validation and application of a census-based methodology. Am J Public Health 1992;82:703–710.
16.
Collen MFe. Multiphasic Health Testing Services. New York: John Wiley & Sons, 1978.
17.
Collen MF, Davis LF. The multitest laboratory in health care. J Occup Med 1969;11:355–360.
18.
Collen M, Rubin L, Neyman J, Dantzig G, Siegelaub AB. Automated multiphasic screening and diagnosis. Am J Public Health 1964;54:741–750.
19.
Collen M. Multiphasic Health Testing Services. Circulation New York: John Wiley & Sons, 1998:1837–1847.
20.
Iribarren C, Sidney S, Sternfeld B, Browner WS. Calcification of the aortic arch: risk factors and association with coronary heart disease, stroke, and peripheral vascular disease. JAMA 2000;283:2810–2815.
21.
Pouliot MC, Despres JP, Lemieux S, et al. Waist circumference and abdominal sagittal diameter: best simple anthropometric indexes of abdominal visceral adipose tissue accumulation and related cardiovascular risk in men and women. Am J Cardiol 1994;73:460–468.
22.
Arellano MG, Petersen GR, Petitti DB, Smith RE. The California Automated Mortality Linkage System (CAMLIS). Am J Public Health 1984;74:1324–1330.
23.
Riserus U, Arnlov J, Brismar K, Zethelius B, Berglund L, Vessby B. Sagittal abdominal diameter is a strong anthropometric marker of insulin resistance and hyperproinsulinemia in obese men. Diabetes Care 2004;27:2041–2046.
24.
Snijder MB, Visser M, Dekker JM, et al. Low subcutaneous thigh fat is a risk factor for unfavourable glucose and lipid levels, independently of high abdominal fat. The Health ABC Study. Diabetologia 2005;48:301–308.
25.
Snijder MB, Dekker JM, Visser M, et al. Associations of hip and thigh diameters independent of waist circumference with the incidence of type 2 diabetes: the Hoorn Study. Am J Clin Nutr 2003;77:1192–1197.
26.
Luchsinger JA, Tang MX, Stern Y, Shea S, Mayeux R. Diabetes mellitus and risk of Alzheimer’s disease and dementia with stroke in a multiethnic cohort. Am J Epidemiol 2001;154:635–641.
27.
Breteler MM. Vascular involvement in cognitive decline and dementia. Epidemiologic evidence from the Rotterdam Study and the Rotterdam Scan Study. Ann NY Acad Sci 2000;903:457–465.
28.
Yaffe K, Blackwell T, Kanaya AM, Davidowitz N, Barrett-Connor E, Krueger K. Diabetes, impaired fasting glucose, and development of cognitive impairment in older women. Neurology 2004;63:658–663.
29.
Whitmer RA, Sidney S, Selby J, Johnston SC, Yaffe K. Midlife cardiovascular risk factors and risk of dementia in late life. Neurology 2005;64:277–281.
30.
Chaldakov GN, Stankulov IS, Hristova M, Ghenev PI. Adipobiology of disease: adipokines and adipokine-targeted pharmacology. Curr Pharm Des 2003;9:1023–1031.
31.
Prins JB. Adipose tissue as an endocrine organ. Best Pract Res Clin Endocrinol Metab 2002;16:639–651.
32.
Landin K, Stigendal L, Eriksson E, et al. Abdominal obesity is associated with an impaired fibrinolytic activity and elevated plasminogen activator inhibitor-1. Metabolism 1990;39:1044–1048.
33.
Yaffe K, Lindquist K, Penninx BW, et al. Inflammatory markers and cognition in well-functioning African-American and white elders. Neurology 2003;61:76–80.
34.
Funahashi H, Yada T, Suzuki R, Shioda S. Distribution, function, and properties of leptin receptors in the brain. Int Rev Cytol 2003;224:1–27.
35.
Harvey J. Novel actions of leptin in the hippocampus. Ann Med 2003;35:197–206.
36.
Fewlass DC, Noboa K, Pi-Sunyer FX, Johnston JM, Yan SD, Tezapsidis N. Obesity-related leptin regulates Alzheimer’s Abeta. FASEB J 2004;18:1870–1878.
37.
Braak E, Griffing K, Arai K, Bohl J, Bratzke H, Braak H. Neuropathology of Alzheimer’s disease: what is new since A. Alzheimer? Eur Arch Psychiatry Clin Neurosci 1999;249 (suppl 3):14–22.
38.
Ward MA, Carlsson CM, Trivedi MA, Sager MA, Johnson SC. The effect of body mass index on global brain volume in middle-aged adults: a cross sectional study. BMC Neurol 2005;5:23.
39.
Jagust W, Harvey D, Mungas D, Haan M. Central obesity and the aging brain. Arch Neurol 2005;62:1545–1548.
40.
Wang PN, Yang CL, Lin KN, Chen WT, Chwang LC, Liu HC. Weight loss, nutritional status and physical activity in patients with Alzheimer’s disease. A controlled study. J Neurol 2004;251:314–320.
41.
Bunout D, Fjeld C. Nutritional reversion of cognitive impairment in the elderly. Nestle Nutr Workshop Ser Clin Perform Programme 2001;5:263–77; discussion 277-281.
42.
Gonzalez-Gross M, Marcos A, Pietrzik K. Nutrition and cognitive impairment in the elderly. Br J Nutr 2001;86:313–321.
43.
Winocur G, Greenwood CE. Studies of the effects of high fat diets on cognitive function in a rat model. Neurobiol Aging 2005;26(suppl 1):46–49.
44.
Sugerman H, Windsor A, Bessos M, Wolfe L. Intra-abdominal pressure, sagittal abdominal diameter and obesity comorbidity. J Intern Med 1997;241:71–79.
45.
Zamboni M, Turcato E, Armellini F, et al. Sagittal abdom-inal diameter as a practical predictor of visceral fat. Int J Obes Relat Metab Disord 1998;22:655–660.
46.
Empana JP, Ducimetiere P, Charles MA, Jouven X. Sagittal abdominal diameter and risk of sudden death in asymptomatic middle-aged men: the Paris Prospective Study I. Circulation 2004;110:2781–2785.
47.
Ohrvall M, Berglund L, Vessby B. Sagittal abdominal diameter compared with other anthropometric measurements in relation to cardiovascular risk. Int J Obes Relat Metab Disord 2000;24:497–501.
Letters to the Editor
26 June 2008
Central obesity and increased risk of dementia more than three decades later
Azeez Farooki, Memorial Sloan Kettering Cancer Center

I read with interest the longitudinal study [1] of US patients which adds dementia to the other well known negative outcomes associated with visceral obesity. [5] I would like to add a useful caveat: waist circumference is most accurately interpreted in the context of ethnicity.

Current ATP-III criteria for metabolic syndrome define abdominal obesity as a waist circumference in men >102 cm (40 in) and in women >88 cm (35 in). [6] The International Diabetes Foundation created definitions for four different ethnic groups and may include three more groups when data become available. [7] Using ATP-III criteria, a patient of Asian origin, for example, might be misclassified as having "normal" waist circumference.

For many patients with visceral obesity, losing weight through diet control and exercise is difficult but this study provides yet another motivation to pursue these therapeutic lifestyle changes.

References

5. Lapidus L, Bengtsson C, Larsson B, et al. Distribution of adipose tissue and risk of cardiovascular disease and death: a 12 year follow up of participants in the population study of women in Gothenburg, Sweden. Br Med J (Clin Res Ed) 1984;10:289:1257-1261.

6. Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults. Executive summary of the Third Report of the National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, and Treatment of High Blood Cholesterol in Adults (Adult Treatment Panel III). JAMA 2001;285:2486-2497.

7. George, K, Alberti, MM, Zimmet, P, et al. The metabolic syndrome - a new worldwide definition. Lancet 2005;336:1059.

Disclosure: The author reports no disclosures.

26 June 2008
Reply from the authors
Rachael A. Whitmer, Kaiser Permanente Division of Research
D. R. Gustafson, E. Barrett-Connor, M. N. Haan, E. P. Gunderson, and K. Yaffe

We thank Drs. Cereda, Farooki, and Tezapsidis for their comments on our article. [1]

We agree with Cereda et al. that obesity and central obesity confers with it a state of inflammation and this may also contribute to cognitive impairment and dementia risk. Unfortunately measures of physical activity were not available so it is unknown what effect this would have on the association between midlife central obesity status and dementia risk.

We also concur that visceral adipose tissue itself is the likely culprit in the association between central obesity and increased dementia risk. Hopefully, future studies will investigate possible actions of visceral adipocytes on the brain and effects of an altered adipokine milieu on cognitive status. [12]

Regarding comments from Farooki et al., ethnic specific anthropometric measurements are the most precise. Although there are standard ethnic specific guidelines for waist circumference in defining central obesity, there are not published ethnic specific guidelines for sagittal abdominal diameter, the anthropometric measurement used in our study. Future studies examining central obesity and disease outcomes should incorporate these guidelines.

With respect to comments from Tezapsidis concerning leptin as a possible therapeutic for patients with AD, leptin has been shown to be beneficial to neurons. However, obesity is associated with 'leptin resistance' so it is possible that obese individuals may not respond to possible neuroprotective effects of leptin. However, a recent study in non-demented elders demonstrated that higher serum leptin levels are associated with better cognitive performance independent of total body weight and total body fat. [13] More work is needed on how states of leptin resistance as induced by chronic obesity may contribute to brain aging.

References

12. Whitmer, RA. Epidemiology of Adiposity and Dementia, Current Alzheimer's Research, 2007;4:117-122

13. Holden KF, Lindquist K, Tylavsky FA, Rosano C, Harris TB, Yaffe K; for the Health ABC study. Serum leptin level and cognition in the elderly: Findings from the Health ABC Study.Neurobiol Aging. 2008 Mar 19. [Epub ahead of print].

Disclosure: The authors report no disclosures.

26 June 2008
Central obesity and increased risk of dementia more than three decades later
Emanuele Cereda, International Center for the Assessment of Nutritional Status (ICANS), University of Milan, Italy.
Manuela Chiara Sacchi (Division of Clinical Nutrition, Niguarda Ca' Granda Hospital, Milan, Italy) Alexis Elias Malavazos (Endocrinology Unit, Department of Medical and Surgical Sciences, Uni

We read with interest the recent article by Whitmer et al. who provide evidence of a longitudinal link between middle-life visceral obesity and dementia risk. [1] It is now apparent that intra-abdominal fat may play a role in other health complications in addition to cardiovascular disease (CVD). Unfortunately, the underlying mechanisms are still unclear.

To explain the observations that overall adiposity is linked both to cognitive decline and CVD and that concurrent CVD is often seen in older dementia patients, we recently reviewed current physiopathologic theories. [2,3] We concluded that visceral adipose tissue (VAT) is consistently implicated in cognitive decline rather than body mass index.

In addition, women who experience changes in body composition and fat distribution after menopause transition are at an increased risk of AD. This is due to a stronger link to vascular factors (pro-inflammatory molecules) and metabolic complications (insulin resistance, dyslipidemia, hypertension). [3]

Luchsinger et al. have hypothesized that in people over 65, a short-term (5-year follow-up) prospective association seems modified by age and is different depending on the anthropometric measure. [4] However, a life course contribution to chronic diseases is recognized. [2] The study by Whitmer et al. [1] supports the previously suggested theory [2,3] particularly when multiple adjustments for concurrent VAT-related complications (diabetes, hypertension, hyperlipidemia and CVDs) are considered. In this respect, the role of VAT as endocrine organ, which is able to release a number of mediators, appears fundamental.

The confounding effect of insulin resistance should be recognized but it should also be noted that a chronic low-grade inflammation is a precondition associated with obesity duration. Reduction in VAT, particularly through physical activity, is efficacious in improving metabolic profile and inflammation. However, long-term results are difficult to maintain and protective effects are still unclear. [3] It is possible that further adjustment for physical activity would have strengthened the authors' evidence. This warrants further investigation and anthropometric data is not readily available.

Despite this study's limitations, the suggested use of available surrogates of visceral adiposity other than sagittal abdominal diameter in a neurological setting is justified. [2]

References

1. Whitmer RA, Gustafson DR, Barrett-Connor E, et al. Central obesity and increased risk of dementia more than three decades later. Neurology. 2008; (in press)

2. Cereda E, Sansone V, Meola G, Malavazos AE. Increased visceral adipose tissue rather than BMI as a risk factor for dementia. Age Ageing 2007;36:488-491.

3. Cereda E, Battezzati A, Bertoli S, Malavazos AE, Testolin G. A life-course contribution of nutrition to future cognitive decline. In: Bernhardt NE and Kasko AM editors. Nutrition for the Middle-Aged and Elderly. Hauppauge NY: Nova Science Publishers; 2008 (in press).

4. Luchsinger JA, Patel B, Tang MX, Schupf N, Mayeux R. Measures of adiposity and dementia risk in elderly persons. Arch Neurol 2007;64:392-398.

Disclosure: The authors report no disclosures.

26 June 2008
Central obesity and increased risk of dementia more than three decades later
Nikolaos Tezapsidis, Neurotez, Inc.
Mark A. Smith (Case Western Reserve University), J. Wes Ashford (Stanford University)

Whitmer et al. demonstrated a significant correlation between mid-life obesity and dementia later in life and suggested that visceral adipocyte-derived factors may be neurotoxic, contributing to the slow deterioration of the CNS. [1]

However, it has been shown that some of these factors including leptin may be beneficial to neurons. Leptin is capable of reducing the brain Aß load [8], is neuroprotective and improves cognitive performance of aged rodents. [9]

Obesity is often characterized by some form of leptin resistance that may be attributable to CRP (C-reactive protein), which is elevated in obesity. [10] CRP can bind to leptin preventing its binding to the leptin receptor. Therefore, obese subjects may be deprived from leptin's beneficial action, propagating obesity and neurodegeneration.

It has also been documented that obese individuals who eventually developed AD may experience a loss in weight prior to the onset of dementia. [11] A gradual selective neuronal loss in the hippocampus, hypothalamus, or both due to the absence of centrally acting leptin may lead to disturbances in appetite.

In AD patients, leptin levels are reduced whereas blood CRP levels are normal so leptin therapy may benefit AD patients. Additional benefits may include ability to increase insulin sensitivity and alleviate insulin resistance which are common in AD.

References

8. Fewlass DC, Noboa K, Pi-Sunyer FX, Johnston JM, Yan SD, Tezapsidis N. Obesity-related leptin regulates Alzheimer's Abeta. Faseb J 2004;18:1870-1878.

9. Harvey J. Leptin: a diverse regulator of neuronal function. J Neurochem 2007;100:307-313.

10. Chen K, Li F, Li J, et al. Induction of leptin resistance through direct interaction of C-reactive protein with leptin. Nat Med 2006;12:425-432.

11. Gustafson S. Adiposity indices and dementia. Lancet Neurol 2006;5:713-720.

Disclosure: Dr. Tezapsidis is the founder of Neurotez, Inc., a private CNS biotechnology corporation (www.neurotez.com) that is pursuing leptin as a novel therapeutic for AD. Drs. Mark Smith and Wes Ashford are affiliated with Neurotez, Inc.

Information & Authors

Information

Published In

Neurology®
Volume 71Number 14September 30, 2008
Pages: 1057-1064
PubMed: 18367704

Publication History

Published online: March 26, 2008
Published in print: September 30, 2008

Permissions

Request permissions for this article.

Notes

Authors

Affiliations & Disclosures

R. A. Whitmer, PhD
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.
D. R. Gustafson, PhD
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.
E. Barrett-Connor, MD
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.
M. N. Haan, DrPH
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.
E. P. Gunderson, PhD
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.
K. Yaffe, MD
From Kaiser Permanente Division of Research (R.A.W., E.P.G.), Oakland, CA; Goteberg University (D.R.G.), Goteberg, Sweden; the Department of Family Medicine (E.B.-C.), University of California, La Jolla; the Department of Epidemiology (M.N.H.), University of Michigan, Ann Arbor; and the Departments of Psychiatry, Neurology and Epidemiology (K.Y.), University of California, San Francisco.

Notes

Address correspondence and reprint requests to Dr. Rachel A. Whitmer, Kaiser Permanente Division of Research, 2000 Broadway, Oakland, CA 94612 [email protected]

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

Cited By
  1. The Effect of a Combination of Eucommia ulmoides and Achyranthes japonica (KGC08EA) on Body Weight in High-Fat Diet-Induced Obese Mice , Journal of the Korean Society of Food Science and Nutrition, 52, 11, (1197-1205), (2023).https://doi.org/10.3746/jkfn.2023.52.11.1197
    Crossref
  2. Exploring the Effects of a Mediterranean Diet and Weight Loss on the Gut Microbiome and Cognitive Performance in Older, African American Obese Adults: A Post Hoc Analysis, Nutrients, 15, 15, (3332), (2023).https://doi.org/10.3390/nu15153332
    Crossref
  3. Adipocyte-Derived Small Extracellular Vesicles from Patients with Alzheimer Disease Carry miRNAs Predicted to Target the CREB Signaling Pathway in Neurons, International Journal of Molecular Sciences, 24, 18, (14024), (2023).https://doi.org/10.3390/ijms241814024
    Crossref
  4. Metabolic Syndrome as a Risk Factor for Alzheimer’s Disease: A Focus on Insulin Resistance, International Journal of Molecular Sciences, 24, 5, (4354), (2023).https://doi.org/10.3390/ijms24054354
    Crossref
  5. Inflammatory Pathways in Overweight and Obese Persons as a Potential Mechanism for Cognitive Impairment and Earlier Onset Alzeihmer’s Dementia in the General Population: A Narrative Review, Biomedicines, 11, 12, (3233), (2023).https://doi.org/10.3390/biomedicines11123233
    Crossref
  6. Poor Cognitive Agility Conservation in Obese Aging People, Biomedicines, 11, 1, (138), (2023).https://doi.org/10.3390/biomedicines11010138
    Crossref
  7. The Contribution of Hippocampal All-Trans Retinoic Acid (ATRA) Deficiency to Alzheimer’s Disease: A Narrative Overview of ATRA-Dependent Gene Expression in Post-Mortem Hippocampal Tissue, Antioxidants, 12, 11, (1921), (2023).https://doi.org/10.3390/antiox12111921
    Crossref
  8. The complex relationship between obesity and neurodegenerative diseases: an updated review, Frontiers in Cellular Neuroscience, 17, (2023).https://doi.org/10.3389/fncel.2023.1294420
    Crossref
  9. Cellular senescence in brain aging and cognitive decline, Frontiers in Aging Neuroscience, 15, (2023).https://doi.org/10.3389/fnagi.2023.1281581
    Crossref
  10. Obesity reduces hippocampal structure and function in older African Americans with the APOE-ε4 Alzheimer’s disease risk allele, Frontiers in Aging Neuroscience, 15, (2023).https://doi.org/10.3389/fnagi.2023.1239727
    Crossref
  11. See more
Loading...

View Options

Get Access

Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to get full access to it.

Purchase Access, $39 for 24hr of access

View options

Full Text

View Full Text

Full Text HTML

View Full Text HTML

Media

Figures

Other

Tables

Share

Share

Share article link

Share