Skip to main content

Red blood cell distribution width to albumin ratio (RAR) is associated with low cognitive performance in American older adults: NHANES 2011–2014

Abstract

Background

The red blood cell distribution width to albumin ratio (RAR) is a novel comprehensive biomarker of inflammation and nutrition, which has emerged as a reliable prognostic indicator for adverse outcomes and mortality in patients with various diseases. However, the association between RAR and low cognitive performance in older adults remains unclear. This study aims to investigate the relationship between RAR and low cognitive performance among older adults in the United States.

Methods

This study, a retrospective analysis, included 2,765 participants aged 60 years and older from the National Health and Nutrition Examination Survey (NHANES) conducted between 2011 and 2014. Low cognitive performance was assessed using word learning subset from the Consortium to Establish a Registry for Alzheimer’s Disease (CERAD), the Digit Symbol Substitution Test (DSST), and the Animal Fluency Test (AFT). Low cognitive performance was defined as scores below the lowest quartile in each cognitive test. The association between RAR and low cognitive performance was evaluated using weighted multivariable logistic regression, restricted cubic splines (RCS), and subgroup analyses.

Results

After adjusting for all potential confounders, RAR was independently and linearly positively associated with both low DSST performance and low AFT performance. Specifically, compared to participants in the first quartile of RAR, those in the fourth quartile had adjusted ORs (95% CIs) of 1.81 (1.03, 3.20) for low DSST performance and 1.68 (1.05, 2.67) for low AFT performance. Subgroup analysis did not reveal significant interactions between stratification variables.

Conclusion

RAR is significantly linearly positively associated with low cognitive performance. Maintaining a lower RAR may be a crucial strategy for mitigating the risk of cognitive decline in the elderly population.

Peer Review reports

Introduction

Cognitive impairment (CI) is a common and concerning aspect of the aging process, characterized by declines in learning, memory, attention, and executive function, leading to impairment of function [1, 2]. Research indicates that declines in CI are closely associated with an increased risk of developing dementia [3], and over 50% of individuals with CI are expected to progress to dementia within 5 years [4]. In individuals with mild cognitive impairment (MCI), functional independence is generally preserved, whereas in dementia, patients experience progressive deterioration in memory and other cognitive functions, severely impairing quality of life and the ability to live independently [5]. With the increasing population aging, dementia has become a significant global public health challenge, posing serious difficulties for individuals, families, and society [6]. According to the 2022 World Alzheimer Report, it is estimated that there are currently approximately 50 million people with dementia worldwide, with this number projected to reach 152 million by 2050 [7]. In 2020, over 6 million individuals aged 65 and older in the United States were diagnosed with Alzheimer’s disease, and this number is estimated to rise to 13.85 million by 2060 [8].

The etiology of dementia remains unclear, and its pathogenesis is complex [8]. There are currently no effective treatments to completely reverse dementia, and up to 60% of dementia patients remain undiagnosed, so early detection of dementia symptoms and modifiable risk factors is essential [9, 10]. Notably, chronic inflammation is considered a significant component in the pathogenesis of dementia [11]. Research indicates that neuroinflammation caused by excessive activation of microglia in the central nervous system is a critical mechanism leading to neuronal structural and functional damage, which in turn results in CI [12,13,14,15]. In addition to central nervous system inflammation [16], a growing body of research has confirmed the association between peripheral inflammation and dementia [17,18,19,20]. A study from the UK has shown that peripheral inflammatory cytokines are significantly elevated in patients with early-stage dementia compared to healthy controls [20]. Several studies [21,22,23,24] have confirmed the association between various inflammatory markers and CI. Additionally, a large cohort study from the UK Biobank involving 84,342 older adults, utilized a population-based measure of the dietary inflammatory index (DII). This index assessed 31 inflammation-related dietary parameters, such as macronutrients, micronutrients, bioactive components, and specific foods. The study demonstrated that an anti-inflammatory diet can significantly suppress systemic inflammation, particularly in individuals with cardiometabolic diseases, thereby slowing the progression of dementia [25].

The red blood cell distribution width (RDW) to albumin ratio (RAR) is a novel comprehensive biomarker of inflammation and nutrition, used to evaluate immune status and immune response. Specifically, RDW serves as a biomarker that reflects the heterogeneity in red blood cell (RBC) volume [26]. Multiple large-scale cohort studies and Mendelian randomization analyses have demonstrated a causal relationship between RDW and CI, as well as Alzheimer’s disease [27, 28]. Serum albumin stands as a pivotal biomarker for assessing nutritional status and inflammation. Numerous studies have demonstrated a correlation between serum albumin levels and both neurodegenerative diseases and blood-brain barrier permeability [29,30,31]. Previous studies have identified that RDW and serum albumin levels serve as biomarkers for multidimensional dysfunctions related to inflammation, oxidative stress, and nutrition [32, 33], but they represent these pathological aspects from different perspectives. It is noteworthy that the RAR, which combines RDW and serum albumin, has demonstrated significant predictive value in the prognosis of stroke [34]and acute respiratory distress syndrome (ARDS) [35]. Recent studies have found that RAR can accurately predict the prognosis of patients with community-acquired bacteremia [36], acute pancreatitis [37], and rheumatic diseases [38]. However, the relationship between RAR and low cognitive performance in older adults remains unclear. Therefore, this study aims to investigate the association between RAR and low cognitive ability in the elderly, with the hope of contributing to the understanding of the pathogenesis of low cognitive ability and early identification.

Methods

Study population

The NHANES is a continuous, publicly accessible, nationwide research program conducted by the Centers for Disease Control and Prevention (CDC), from which researchers worldwide can freely access the data. It employs a complex, multistage probability sampling design to evaluate the nutritional status and overall health of both adults and children in the United States. The research protocol was approved by the National Center for Health Statistics (NCHS) Research Ethics Review Board (https://www.cdc.gov/nchs/nhanes/irba98.htm) and informed consent was obtained from all participants. We selected data from NHANES 2011–2014 (https://www.cdc.gov/nchs/nhanes/index.htm), ultimately including 2,765 participants (Fig. 1).

Fig. 1
figure 1

Flow chart of the study participants

Assessment of RAR

In the NHANES survey, highly trained medical personnel measured participants’ peripheral blood RDW (%) using a Coulter analyzer at the Mobile Examination Center (MEC). The concentration of albumin was measured using the DcX 800 method, a bichromatic digital endpoint method. In this process, albumin forms a complex with bromocresol purple (BCP) reagent, and the system monitors changes in absorbance at 600 nm. This absorbance is directly proportional to the concentration of albumin in the blood sample, reflecting serum albumin levels (g/dL). The RAR is calculated using the following formula: [RDW (%) / serum albumin (g/dL)]. Based on the quartile of RAR, participants were categorized into four groups (Q1, Q2, Q3, and Q4) as follows: Q1 (< 2.95), Q2 (2.95–3.16), Q3 (3.16–3.40), and Q4 (> 3.40), with Q1 serving as the reference group.

Assessment of low cognitive performance

The NHANES assesses cognitive function in participants aged 60 and older, utilizing three components: the word learning and recall modules from the Consortium to Establish a Registry for Alzheimer’s disease (CERAD), the Digit Symbol Substitution Test (DSST), and the Animal Fluency Test (AFT). Notably, the selection of cognitive assessment tools for the NHANES was based on the criteria that these tools should be concise, easily understood by diverse populations, and both simple to administer and to score [39]. Higher scores on these tests indicate better cognitive function. The CERAD Word Learning subtest (CERAD-WL) includes three consecutive immediate recall learning trials (CERAD-IR) and one delayed recall test (CERAD-DR), to assess an individual’s immediate and delayed learning abilities for new verbal information (memory sub-domain). The scores for the three CERAD-IR tests range from 0 to 10 points each, while the CERAD-DR test score ranges from 0 to 10 points [40]. The CERAD-WL score is the sum of the CERAD-IR and CERAD-DR scores [41]. The DSST is used to assess an individual’s processing speed, sustained attention, and working memory. The test involves a paper form with a keyboard containing nine digits and symbols. Participants are required to fill in 133 boxes with the corresponding symbols within a 2-minute time frame to match the digits. The total number of correct matches constitutes the DSST score [42]. The AFT primarily evaluates participants’ verbal fluency [43]. Participants are asked to name as many animals as possible within one minute. The AFT score is determined by the number of animal names provided. This study uses the scores from the three cognitive tests as outcome variables. Since NHANES does not specify a particular threshold for low cognitive performance in each cognitive test, we have defined low cognitive performance as the lowest quartile for each cognitive assessment within the full sample. The lowest quartile may be a preferable choice for selecting participants with low cognitive performance. This approach is consistent with multiple previously published studies and is widely recognized [39, 44].

Assessment of covariates

This study considered various covariates including demographic and health-related information, as follows: age; sex, categorized as male or female; race, classified as Mexican American, non-Hispanic White, non-Hispanic Black, or other; education, divided into less than high school, high school or equivalent, or college and above; marital status, categorized as married/ living with partner, widowed/divorced/separated, or never married. The poverty income ratio (PIR) was divided into low income (< 1.3), moderate income (1.3–3.5), and high income (> 3.5). Body mass index (BMI) was calculated as weight (kg)/height (m2), and categorized as normal/underweight (< 25), overweight (25–30), or obese (> 30). Smoking status was classified as never (lifetime consumption of fewer than 100 cigarettes), former (lifetime consumption of more than 100 cigarettes but currently not smoking), or current (lifetime consumption of more than 100 cigarettes and currently smoking daily). Drinking status was categorized as yes or no based on whether the individual drank more than 12 drinks per year. Physical activity (PA) was assessed using the Global Physical Activity Questionnaire (GPAQ) and classified according to the Metabolic Equivalent of Task (MET) adult guidelines (≥ 600 MET minutes/week, equivalent to 150 min/week of moderate-intensity or 75 min/week of high-intensity physical activity) [45, 46]. Hypertension was defined based on self-reported history of high blood pressure, use of antihypertensive medications, or systolic blood pressure (SBP) > 130 mmHg or diastolic blood pressure (DBP) > 80 mmHg [47]. Hyperlipidemia was defined as self-reported high cholesterol levels, use of cholesterol-lowering medications, or laboratory results showing triglycerides ≥ 150 mg/dl, low density lipoprotein cholesterol (LDL-C) ≥ 130 mg/dl, high density lipoprotein cholesterol (HDL-C) < 40 mg/dl (male) or < 50 mg/dl (female), and total cholesterol (TC) ≥ 200 mg/dl. Diabetes was defined based on self-reported history of diabetes, use of insulin or antidiabetic medications, or laboratory results indicating hemoglobin A1c (HbA1c) ≥ 6.5%, fasting blood glucose (FBG) ≥ 7.0 mmol/L, or 2-hour postprandial glucose (2hPG) ≥ 11.1 mmol/L [48].

Statistical analyses

To ensure that the research findings accurately represent the U.S. civilian non-institutionalized population, our study incorporated sample weights, stratification, and clustering in the analysis. Participants were divided into four groups (Q1-Q4) based on RAR quartiles. Continuous variables were reported as means and standard deviations (SDs), while categorical variables were presented as frequencies and percentages in the baseline characteristics. Differences of continuous variables were assessed using one-way ANOVA, and differences of categorical variables were evaluated using Pearson chi-square (χ2) tests. To mitigate the reduction in sample size due to missing covariate data, we employed multiple imputation by chained equations (MICE) to address the missing values. The proportion of missing covariates and the imputation methods used are detailed in Additional file: Table S1 and Fig. S1. We conducted tests for multicollinearity on all variables, and the variance inflation factors (VIFs) for each variable were below 2, indicating that no significant multicollinearity issues were detected (Additional file: Table S2). We employed weighted multivariable logistic regression to calculate odds ratios (ORs) and 95% confidence intervals (95%CIs) to estimate the independent associations between RAR quartiles and scores of three cognitive tests. Model 1 did not adjust for any covariates. Model 2 adjusted for age, sex, race, education, marital status, PIR, and BMI. Model 3 further adjusted for smoking status, drinking status, physical activity, hypertension, hyperlipidemia, and diabetes mellitus (DM). We employed restricted cubic spline (RCS) fitting logistic regression model to investigate the dose-response relationship between RAR and low cognitive performance. We further stratified the study population into two subgroups based on gender and conducted separate dose-response analyses to examine the association between RAR and low cognitive performance within each gender. Additionally, exploratory subgroup analyses were conducted stratified by age, sex, education, BMI, drinking status, smoking status, hypertension, and DM to evaluate the relationship between RAR and low cognitive performance across different population characteristics. Likelihood ratio tests were utilized to assess interactions between grouping variables and RAR. We also conducted a further analysis using the Receiver Operating Characteristic (ROC) curve to assess the diagnostic capability of RAR in detecting low cognitive performance. Finally, we conducted several sensitivity analyses to assess the stability of the association between RAR and low cognitive performance. First, we excluded missing values for any covariates to mitigate the potential impact of data missingness on the primary results. Second, we removed all extreme values of RAR (defined as mean ± 3SD) to minimize their influence on the significance and stability of the results. Third, to account for the potential impact of cancer on cognitive function, we excluded participants who self-reported a malignant tumor at baseline. Bonferroni correction was applied to adjust for multiple comparisons, setting the threshold for statistical significance for subgroup interaction tests at α = 0.05/8 = 0.006. All analyses were performed using R software (version 4.1.2), with P-values < 0.05 (two-sided) considered statistically significant.

Results

Characteristics of the study participants

This study included 2,765 participants with a mean age of 69.19 ± 6.64 years, and 46.2% of them were male. The mean RAR of the participants was 3.22 ± 0.40. Table 1 presents the baseline characteristics of the participants. They were divided into four groups based on RAR quartiles. Compared to participants in the first quartile, those in the fourth quartile of RAR were generally older, more likely to be female, non-Hispanic Black, and exhibited higher levels of waist circumference (WC), creatinine (Cr), and HbA1c, as well as lower levels of HDL-C and hemoglobin. Additionally, participants with higher RAR levels generally demonstrated lower cognitive performance, as evidenced by low scores on the AFT, DSST, and CERAD-WL. Moreover, a higher quartile of RAR was associated with increased numbers of participants who exhibited obesity, drinking, lack of physical activity, and diabetes. This suggests a potential association between unhealthy lifestyle habits and elevated RAR levels.

Table 1 Baseline characteristics according to RAR quartiles

Association between RAR and low cognitive performance

We employed weighted multivariable logistic regression analysis to estimate the independent association between RAR quartiles and low cognitive performance. The relationship between RAR and low cognitive performance is illustrated in Table 2. After adjusting for multiple variables (Model 3), no significant association was found between RAR quartiles and low CERAD-WL performance (P > 0.05). For low DSST performance, the ORs (95% CIs) were 1.00 (reference), 0.97 (0.56, 1.70), 1.03 (0.60, 1.78), and 1.81 (1.03, 3.20). For low AFT performance, the ORs (95% CIs) were 1.00 (reference), 1.38 (0.81, 2.34), 1.27 (0.76, 2.11), and 1.68 (1.05, 2.67). The results indicate that higher levels of RAR (Q4 vs. Q1) are associated with an 81% increased risk of low DSST performance and a 68% increased risk of low AFT performance.

Table 2 ORs (95% CIs) for low cognitive performance according to RAR quartiles

Dose-response analysis of RAR with low cognitive performance

We employed RCS fitted for logistic regression models to further investigate the relationship between RAR and low cognitive performance. The results revealed a significant linear relationship between RAR and low CERAD-WL performance. (Fig. 2A), low DSST performance (Fig. 2B), and low AFT performance (Fig. 2C) (P for nonlinear > 0.05). After adjusting for all confounding factors, we observed a generally linear positive association between RAR and both low DSST performance and low AFT performance, with an increased risk of low cognitive performance as RAR levels rise. Furthermore, we estimated the relationship between RAR and low cognitive performance across different genders. The results showed that RAR was linearly associated with low cognitive performance in both genders, with an increased risk of low DSST performance (Fig. 2E) and low AFT performance (Fig. 2F) as RAR levels increased across different gender groups.

Fig. 2
figure 2

Restricted cubic splines were used to test the associations between RAR and low cognitive performance among all participants and different genders. low CERAD-WL performance (A), low DSST performance (B), low AFT performance (C), low CERAD-WL performance between different genders (D), low DSST performance between different genders (E), low AFT performance between different genders (F). The analysis was adjusted for sex, age, race, education, marital status, PIR, BMI, smoking status, drinking status, physical activity, hypertension, hyperlipidemia, and DM. PIR poverty income ratio, BMI body mass index, DM diabetes mellitus, RAR red blood cell distribution width to albumin ratio, CERAD-WL consortium to establish a registry for Alzheimer’s disease word learning test, DSST digit symbol substitution test, AFT animal fluency test

Subgroup analyses

To further elucidate whether the association between RAR and low cognitive performance is consistent across different populations, we conducted a series of subgroup analyses. In these analyses (Fig. 3), we stratified by age, gender, education, BMI, drinking status, smoking status, hypertension, and DM. After applying the Bonferroni correction for statistical significance, the results demonstrated that the associations between RAR and low performance on the CERAD-WL, DSST, and AFT were consistent across all subgroups, with no significant interaction observed between RAR and the stratification variables (P for interaction > 0.006).

Fig. 3
figure 3

Subgroup analysis of the associations between RAR and low cognitive performance. The analysis was adjusted for age, gender, education, BMI, smoking status, drinking status, smoking status, hypertension, and DM. BMI body mass index, DM diabetes mellitus, RAR The red blood cell distribution width to albumin ratio, CERAD-WL consortium to establish a registry for alzheimer’s disease word learning test, DSST digit symbol substitution test, AFT animal fluency test

Receiver operating characteristic (ROC) curve analysis

The results demonstrated that RAR exhibited the optimal independent predictive ability for low DSST performance. The area under the ROC curve (AUC) and its 95% CI were 0.845 (95% CI: 0.829–0.860), indicating a strong predictive power. The AUC and 95% CI for the RAR in relation to low CERAD-WL performance and low AFT performance were 0.730 (95% CI: 0.710–0.750) and 0.750 (95% CI: 0.731–0.770), respectively. These values indicate that the model demonstrates a certain level of discriminative ability (Fig. 4).

Fig. 4
figure 4

ROC curve analysis for low cognitive performance. low CERAD-WL performance (A), low DSST performance (B), low AFT performance (C). The analysis was adjusted for sex, age, race, education, marital status, PIR, BMI, smoking status, drinking status, physical activity, hypertension, hyperlipidemia, and DM. PIR poverty income ratio, BMI body mass index, DM diabetes mellitus, RAR The red blood cell distribution width to albumin ratio, CERAD-WL consortium to establish a registry for Alzheimer’s disease word learning test, DSST digit symbol substitution test, AFT animal fluency test

Sensitivity analysis

To assess the stability of our results, we conducted several sensitivity analyses. After excluding participants with missing values for covariates, 2,265 participants were included in the study. Adjusting for a range of confounding factors, the association between RAR and low cognitive performance remained relatively stable (Additional file: Table S3). Following the removal of extreme RAR values, 2,707 participants were included, and after adjusting for various covariates, the results were consistent with the preliminary analysis (Additional file: Table S4). Finally, excluding participants who self-reported having cancers at baseline, 2,226 participants were included, and after adjusting for all confounding factors, the relationship between RAR and low cognitive performance remained robust (Additional file: Table S5).

Discussion

To the best of our knowledge, this study is the first cross-sectional research revealing the association between RAR and low cognitive performance. The results indicate that the RAR levels are independently and linearly positively associated with low DSST performance and low AFT performance among older adults in the United States. As RAR levels increase, the risk of low cognitive performance rises. Our findings underscore the importance of RAR as a novel comprehensive biomarker of inflammation and nutrition, providing some evidence of the role of inflammation and nutritional status in the development of low cognitive performance.

Recent studies have shown that RAR is closely associated with the prognosis of cardiovascular and cerebrovascular diseases, such as coronary artery disease, myocardial infarction, and stroke, and lower RAR level is linked to higher short-term and long-term survival rates in patients [34, 49, 50]. As a novel biomarker, RAR combines two classical clinical parameters, RDW and albumin, and demonstrates a superior predictive ability for adverse disease outcomes and all-cause mortality compared to either RDW or albumin alone [49]. Our study also suggests that RAR may serve as an important predictive factor for the risk of low cognitive performance.

The specific biological mechanisms about association between higher RAR and increased risk of low cognitive performance have not yet been fully elucidated, but may be related to chronic inflammation and poor nutritional status [51, 52]. RAR is a composite indicator integrating RDW and albumin, with increases in RDW and decreases in albumin both contributing to elevated RAR levels. Several studies have indicated that elevated RDW is associated with cognitive decline [53, 54]. A cross-sectional study [55]indicated a “J-shaped” relationship between elevated RDW levels and the likelihood of dementia in older adults. Additionally, a prospective cohort study highlighted a causal impact of RDW on the risk of Alzheimer’s disease [27]. Elevated RDW is commonly associated with systemic inflammatory states, which can affect erythropoiesis, red blood cell lifespan, and red blood cell deformability, leading to increased cell anisotropy and subsequently raising RDW levels [56]. Chronic inflammation is considered one of the key mechanisms underlying low cognitive performance. Abnormal deposition of amyloid proteins can lead to excessive activation of microglia and astrocytes in the central nervous system, triggering neuroinflammatory responses. Prolonged inflammatory conditions result in structural and functional damage to neurons, further contributing to cognitive decline [28]. Additionally, RDW is used in the differential diagnosis of anemia, which is considered a contributing factor to CI and Alzheimer’s disease [28]. In the condition of anemia, pathological changes in red blood cells, including alterations in quantity, volume, distribution, and overall mass, can reduce the blood’s oxygen-carrying capacity. Hypoxic changes in cerebral blood vessels may be a crucial mechanism in the pathogenesis of neurodegenerative diseases, leading to low cognitive performance and dementia [57]. Our study also found that participants with higher RAR often had lower hemoglobin levels and a higher risk of low cognitive performance.

Albumin is an essential protein in human plasma. Hypoalbuminemia, often caused by inflammatory conditions, reflects the presence of inflammation and is associated with increased mortality rates in various diseases and among healthy individuals [58]. Research indicates that albumin can predict cognitive decline, and increasing albumin levels may improve Alzheimer’s disease-related pathology and CI [59, 60]. Albumin plays a crucial role in regulating inflammatory responses, and a decrease in its levels may lead to uncontrolled inflammation, which can subsequently impact cognitive function [50]. Additionally, albumin has antioxidant properties and is capable of neutralizing free radicals and peroxides in the body. When albumin levels decrease, the body’s antioxidant capacity is reduced, leading to increased oxidative stress and contributing to cognitive decline [61]. Furthermore, low albumin levels reflect malnutrition or inadequate protein intake. Such nutritional deficiencies can lead to damage to brain structure and function, thereby affecting cognitive abilities [62]. In summary, previous studies have highlighted the significant independent associations of RDW and albumin with CI. Our research demonstrates that RAR can serve as a valuable biomarker for identifying the risk of low cognitive performance more comprehensively and intuitively than RDW and albumin alone. Elevated RAR may reflect underlying chronic inflammation, oxidative stress, and poor nutritional status, with these combined factors potentially leading to central nervous system damage and cognitive decline.

In addition to central nervous system inflammation, peripheral inflammation also plays an indirect role in the development of CI. Research indicates that increased peripheral inflammation is a driving factor for the onset of dementia and Alzheimer’s disease [63, 64], higher rates of CI have been observed in patients with rheumatoid arthritis and skin inflammation [24, 65]. Furthermore, systemic inflammatory response syndrome (SIRS) is associated with acute CI [66], and persistent systemic inflammation remains a key characteristic of long-term CI [67]. This may be related to increased peripheral inflammation, which elevates the circulating levels of pro-inflammatory cytokines, damages the blood-brain barrier, and subsequently induces central nervous system inflammation, thereby contributing to the development of CI [24]. It is noteworthy that research indicates a dynamic relationship between central and peripheral inflammation during the progression of dementia. Early in the disease, activation of microglia in the central nervous system leads to increased central inflammation. In later stages, as cognitive function declines, central inflammation may diminish while T-cell-related peripheral inflammation intensifies [68]. Thus, the type and level of inflammation can vary across different stages of CI, yet chronic inflammation remains a crucial factor in its onset and progression. Additionally, various inflammatory markers, such as the dietary inflammatory index [69], blood inflammatory index [70], systemic immune-inflammation index [71], and periodontal disease [72] are all considered to significantly impact cognitive function. This further supports the role of RAR as a novel and comprehensive inflammatory marker associated with low cognitive performance.

It is noteworthy that our study found RAR to be more strongly associated with low AFT performance and low DSST performance, while no significant correlation was observed with low CERAD-WL performance. The influence of RAR on cognitive function may be more closely related to language production assessed by AFT, as well as attention and working memory capacity evaluated by DSST, rather than immediate learning and delayed recall abilities assessed by CERAD-WL. Similar studies [73, 74] have indicated that the dietary inflammatory index is positively correlated with aspects of low cognitive performance reflected by AFT and DSST, while showing no correlation with low CERAD-WL performance. These findings are consistent with our study’s conclusions. It is important to emphasize that our study shows that, compared to the lowest RAR quartile, the highest RAR quartile is associated with an 81% increased risk of low DSST performance and a 68% increased risk of low AFT performance. This finding partially confirms that elevated RAR may be a significant risk factor for low cognitive performance, underscoring its importance.

Through gender stratification, our study also found that the risk of low AFT performance and low DSST performance increases with higher RAR levels in both male and female. This finding is consistent with conclusions from most studies on CI [43, 75, 76]. In the subgroup analyses, we did not observe significant interactions between RAR and the stratification variables. Sensitivity analysis results indicate that the relationship between RAR and low cognitive performance remains relatively stable.

Strengths and limitations

This study has several notable strengths. First, we evaluate the association between RAR and the risk of low cognitive performance. RAR, as a novel comprehensive indicator of inflammation and nutrition, is simple, reliable, practical, and easily obtainable, providing significant predictive value for low cognitive performance in the older adults. Second, the NHANES database employs a complex multistage probability sampling design, and weighting of the analyses allows for generalization of the findings to the U.S. non-institutionalized civilian population. To enhance the reliability of our analysis, we constructed and adjusted multiple confounding factor models and conducted stratified analyses to explore the relationship between RAR and different cognitive performance tests. However, this study has several limitations. First, as a cross-sectional study, it cannot establish a causal relationship between RAR and low cognitive performance, necessitating further large-scale cohort studies to assess the predictive capability of RAR for low cognitive performance. Second, despite adjusting for numerous confounding factors, we cannot completely rule out the influence of unknown or unmeasurable confounders on the results. Third, while we conducted exploratory subgroup analyses and adjusted p-values to control for the risk of false positives due to multiple comparisons, these results require validation in future clinical studies. Finally, we did not adjust the cognitive function assessment results based on age, gender, and education level, which may have a certain impact on the research results.

Conclusions

We conducted a cross-sectional study based on the NHANES database and found a significant linear positive association between RAR and low cognitive performance in older adults. As RAR levels increase, the risk of low DSST performance and low AFT performance also rises. Our study highlights that maintaining lower RAR could be an important strategy for reducing the risk of low cognitive performance. In the future, large-scale, multicenter prospective studies are warranted to assess the predictive value of RAR for cognitive function in the elderly and to further explore the specific mechanisms by which low RAR levels lead to low cognitive performance.

Data availability

The dataset used for this study analysis can be found on the official website of the National Health and Nutrition Examination Survey (https://www.cdc.gov/nchs/nhanes/index.htm).

Abbreviations

AFT:

Animal Fluency Test

ARDS:

Acute respiratory distress syndrome

BMI:

Body mass index

BCP:

Bromocresol purple

CI:

Cognitive impairment

CERAD:

Consortium to Establish a Registry for Alzheimer’s Disease

CERAD-WL:

Consortium to Establish a Registry for Alzheimer’s Disease-Word Learning subtest

CERAD-IR:

Consortium to Establish a Registry for Alzheimer’s Disease-Immediate Recall Test

CERAD-DR:

Consortium to Establish a Registry for Alzheimer’s Disease-Delayed Recall Test

CIs:

Confidence intervals

Cr:

Creatinine

DSST:

Digit Symbol Substitution Test

DBP:

Diastolic blood pressure

DM:

Diabetes mellitus

FBG:

Fasting blood glucose

GGT:

Gamma-glutamyl transferase

GPAQ:

Global physical activity questionnaire

HbA1c:

Hemoglobin A1c

HDL-C:

High density lipoprotein- cholesterol

LDL-C:

Low density lipoprotein- cholesterol

MCI:

Mild cognitive impairment

MEC:

Mobile Examination Center

MICE:

Multiple imputation by chained equations

MET:

Metabolic equivalent of task

NHANES:

National Health and Nutrition Examination Survey

ORs:

Odds ratios

PIR:

Poverty income ratio

PA:

Physical activity

RAR:

Red blood cell distribution width to albumin ratio

RCS:

Restricted cubic spline

RDW:

Red blood cell distribution width

RBC:

Red blood cell

SBP:

Systolic blood pressure

SD:

Standard deviations

SIRS:

Systemic inflammatory response syndrome

TC:

Total cholesterol

UA:

Uric acid

VIF:

Variance inflation factor

WC:

Waist circumference

References

  1. Langa KM, Levine DA. The diagnosis and management of mild cognitive impairment: a clinical review. JAMA. 2014;312:2551–61.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Mukadam N, Anderson R, Walsh S, Wittenberg R, Knapp M, Brayne C, et al. Benefits of population-level interventions for dementia risk factors: an economic modelling study for England. Lancet Healthy Longev. 2024;5:100611.

  3. Collyer TA, Murray AM, Woods RL, Storey E, Chong TT-J, Ryan J, et al. Association of dual decline in cognition and gait speed with risk of dementia in older adults. JAMA Netw Open. 2022;5:e2214647.

    Article  PubMed  PubMed Central  Google Scholar 

  4. Gauthier S, Reisberg B, Zaudig M, Petersen RC, Ritchie K, Broich K, et al. Mild cognitive impairment. Lancet. 2006;367:1262–70.

    Article  PubMed  Google Scholar 

  5. Petersen RC. Clinical practice. Mild cognitive impairment. N Engl J Med. 2011;364:2227–34.

    Article  CAS  PubMed  Google Scholar 

  6. GBD 2017 US Neurological Disorders Collaborators, Feigin VL, Vos T, Alahdab F, Amit AML, Bärnighausen TW, et al. Burden of neurological disorders across the US from 1990–2017: A global burden of disease study. JAMA Neurol. 2021;78:165–76.

    Article  Google Scholar 

  7. Kivipelto M, Mangialasche F, Snyder HM, Allegri R, Andrieu S, Arai H, et al. World-Wide FINGERS network: A global approach to risk reduction and prevention of dementia. Alzheimers Dement. 2020;16:1078–94.

    Article  PubMed  Google Scholar 

  8. Rajan KB, Weuve J, Barnes LL, McAninch EA, Wilson RS, Evans DA. Population estimate of people with clinical Alzheimer’s disease and mild cognitive impairment in the united States (2020–2060). Alzheimers Dement. 2021;17:1966–75.

    Article  PubMed  Google Scholar 

  9. Bynum JPW, Benloucif S, Martindale J, O’Malley AJ, Davis MA. Regional variation in diagnostic intensity of dementia among older U.S. Adults: an observational study. Alzheimers Dement. 2024;20:1–10.

    Google Scholar 

  10. Ye Y, Lei M, Chen L, Song R, Zhao F, Zhang L. Efficacy of technology-based cognitive and exercise interventions for mild cognitive impairment: A systematic review, network meta-analysis, and meta-regression of randomized controlled trials. Ageing Res Rev. 2024;100:102438.

    Article  PubMed  Google Scholar 

  11. Gulen MF, Samson N, Keller A, Schwabenland M, Liu C, Glück S, et al. cGAS-STING drives ageing-related inflammation and neurodegeneration. Nature. 2023;620:374–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. De Felice FG, Ferreira ST. Inflammation, defective insulin signaling, and mitochondrial dysfunction as common molecular denominators connecting type 2 diabetes to alzheimer disease. Diabetes. 2014;63:2262–72.

    Article  PubMed  Google Scholar 

  13. Mostafavi S, Gaiteri C, Sullivan SE, White CC, Tasaki S, Xu J, et al. A molecular network of the aging human brain provides insights into the pathology and cognitive decline of Alzheimer’s disease. Nat Neurosci. 2018;21:811–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tian Y, Jing G, Ma M, Yin R, Zhang M. Microglial activation and polarization in type 2 diabetes-related cognitive impairment: A focused review of pathogenesis. Neurosci Biobehav Rev. 2024;165:105848.

    Article  CAS  PubMed  Google Scholar 

  15. Zuliani G, Ranzini M, Guerra G, Rossi L, Munari MR, Zurlo A, et al. Plasma cytokines profile in older subjects with late onset Alzheimer’s disease or vascular dementia. J Psychiatr Res. 2007;41:686–93.

    Article  CAS  PubMed  Google Scholar 

  16. Appleton J, Finn Q, Zanotti-Fregonara P, Yu M, Faridar A, Nakawah MO, et al. Brain inflammation co-localizes highly with Tau in mild cognitive impairment due to early-onset Alzheimer’s disease. Brain. 2024;148:119–32.

  17. Besteher B, Rocktäschel T, Garza AP, Machnik M, Ballez J, Helbing D-L, et al. Cortical thickness alterations and systemic inflammation define long-COVID patients with cognitive impairment. Brain Behav Immun. 2024;116:175–84.

    Article  CAS  PubMed  Google Scholar 

  18. Chu M, Wen L, Jiang D, Liu L, Nan H, Yue A, et al. Peripheral inflammation in behavioural variant frontotemporal dementia: associations with central degeneration and clinical measures. J Neuroinflamm. 2023;20:65.

    Article  Google Scholar 

  19. Trares K, Bhardwaj M, Perna L, Stocker H, Petrera A, Hauck SM, et al. Association of the inflammation-related proteome with dementia development at older age: results from a large, prospective, population-based cohort study. Alzheimers Res Ther. 2022;14:128.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Wood H, Dementia. Peripheral inflammation could be a prodromal indicator of dementia. Nat Rev Neurol. 2018;14:127.

    PubMed  Google Scholar 

  21. Darweesh SKL, Wolters FJ, Ikram MA, de Wolf F, Bos D, Hofman A. Inflammatory markers and the risk of dementia and Alzheimer’s disease: A meta-analysis. Alzheimers Dement. 2018;14:1450–9.

    Article  PubMed  Google Scholar 

  22. Ozawa M, Shipley M, Kivimaki M, Singh-Manoux A, Brunner EJ. Dietary pattern, inflammation and cognitive decline: the Whitehall II prospective cohort study. Clin Nutr. 2017;36:506–12.

    Article  PubMed  PubMed Central  Google Scholar 

  23. Sun M, Wang L, Hu Y, Wang X, Yan S, Guo Y, et al. Cognitive impairment mediates the association between dietary inflammation and depressive symptoms in the elderly. Nutrients. 2022;14:5118.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Wen S, Elias PM, Wakefield JS, Mauro TM, Man M-Q. The link between cutaneous inflammation and cognitive impairment. J Eur Acad Dermatol Venereol. 2022;36:1705–12.

    Article  CAS  PubMed  Google Scholar 

  25. Dove A, Dunk MM, Wang J, Guo J, Whitmer RA, Xu W. Anti-Inflammatory diet and dementia in older adults with cardiometabolic diseases. JAMA Netw Open. 2024;7:e2427125.

    Article  PubMed  PubMed Central  Google Scholar 

  26. Vaya A, Hernández JL, Zorio E, Bautista D. Association between red blood cell distribution width and the risk of future cardiovascular events. Clin Hemorheol Microcirc. 2012;50:221–5.

    Article  PubMed  Google Scholar 

  27. Qiang YX, Deng YT, Zhang YR, Wang HF, Zhang W, Dong Q, et al. Associations of blood cell indices and anemia with risk of incident dementia: A prospective cohort study of 313,448 participants. Alzheimers Dement. 2023;19:3965–76.

    Article  PubMed  Google Scholar 

  28. Winchester LM, Powell J, Lovestone S, Nevado-Holgado AJ. Red blood cell indices and anaemia as causative factors for cognitive function deficits and for Alzheimer’s disease. Genome Med. 2018;10:51.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Yang H, Liao Z, Zhou Y, Gao Z, Mao Y. Non-linear relationship of serum albumin-to-globulin ratio and cognitive function in American older people: a cross-sectional National health and nutrition examination survey 2011–2014 (NHANES) study. Front Public Health. 2024;12:1375379.

    Article  PubMed  PubMed Central  Google Scholar 

  30. Brown RB, Tozer DJ, Loubière L, Harshfield EL, Hong YT, Fryer TD, et al. MINocyclinE to reduce inflammation and blood-brain barrier leakage in small vessel disease (MINERVA): A phase II, randomized, double-blind, placebo-controlled experimental medicine trial. Alzheimers Dement. 2024;20:3852–63.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tian X, Zhao Y, Zhu Y, Cui M. Association between elevated blood-brain barrier permeability and the risk of progressive cognitive decline: A longitudinal study. Arch Gerontol Geriatr. 2024;124:105441.

    Article  PubMed  Google Scholar 

  32. Fanali G, di Masi A, Trezza V, Marino M, Fasano M, Ascenzi P. Human serum albumin: from bench to bedside. Mol Aspects Med. 2012;33:209–90.

    Article  CAS  PubMed  Google Scholar 

  33. Salvagno GL, Sanchis-Gomar F, Picanza A, Lippi G. Red blood cell distribution width: A simple parameter with multiple clinical applications. Crit Rev Clin Lab Sci. 2015;52:86–105.

    Article  PubMed  Google Scholar 

  34. Zhao N, Hu W, Wu Z, Wu X, Li W, Wang Y, et al. The red blood cell distribution Width-Albumin ratio: A promising predictor of mortality in stroke patients. Int J Gen Med. 2021;14:3737–47.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Yoo J-W, Ju S, Lee SJ, Cho YJ, Lee JD, Kim HC. Red cell distribution width/albumin ratio is associated with 60-day mortality in patients with acute respiratory distress syndrome. Infect Dis. 2020;52:266–70.

    Article  Google Scholar 

  36. Shan X, Jiang J, Li W, Dong L. Red blood cell distribution width to albumin ratio as a predictor of mortality in ICU patients with community acquired bacteremia. Sci Rep. 2024;14:28596.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Ren D, Zhang Q, Zhang J, Meng Q. Value of red-blood-cell distribution width-to-albumin ratio combined with BISAP score in assessing the severity of acute pancreatitis. Asian J Surg. 2024;47:3923.

    Article  PubMed  Google Scholar 

  38. Yin L, Min J, Zhong L, Shen Q. The correlation between red cell distribution width to albumin ratio and all-cause mortality in critically ill patients with rheumatic diseases: a population-based retrospective study. Front Med (Lausanne). 2023;10:1199861.

    Article  PubMed  Google Scholar 

  39. Brody DJ, Kramarow EA, Taylor CA, McGuire LC. Cognitive performance in adults aged 60 and over: National health and nutrition examination survey, 2011–2014. Natl Health Stat Rep. 2019;Sep(126):1–23.

  40. Morris JC, Heyman A, Mohs RC, Hughes JP, van Belle G, Fillenbaum G, et al. The consortium to Establish a registry for Alzheimer’s disease (CERAD). Part I. Clinical and neuropsychological assessment of Alzheimer’s disease. Neurology. 1989;39:1159–65.

    Article  CAS  PubMed  Google Scholar 

  41. Zhang J, Yu C, Zhang X, Chen H, Dong J, Lu W, et al. Porphyromonas gingivalis lipopolysaccharide induces cognitive dysfunction, mediated by neuronal inflammation via activation of the TLR4 signaling pathway in C57BL/6 mice. J Neuroinflamm. 2018;15:37.

    Article  Google Scholar 

  42. Ryan JJ, Lopez SJ. Wechsler adult intelligence Scale-III. In: Dorfman WI, Hersen M, editors. Understanding psychological assessment. Boston, MA: Springer US; 2001. pp. 19–42.

    Chapter  Google Scholar 

  43. Dong X, Li S, Sun J, Li Y, Zhang D. Association of coffee, decaffeinated coffee and caffeine intake from coffee with cognitive performance in older adults: National health and nutrition examination survey (NHANES) 2011–2014. Nutrients. 2020;12:840.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Gong HJ, Tang X, Chai YH, Qiao YS, Xu H, Patel I, et al. Predicted lean body mass in relation to cognitive function in the older adults. Front Endocrinol. 2023;14:1172233.

    Article  Google Scholar 

  45. Liu C, Hua L, Xin Z. Synergistic impact of 25-hydroxyvitamin D concentrations and physical activity on delaying aging. Redox Biol. 2024;73:103188.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Wei X, Min Y, Xiang Z, Zeng Y, Wang J, Liu L. Joint association of physical activity and dietary quality with survival among US cancer survivors: a population-based cohort study. Int J Surg. 2024;110:5585–94.

    Article  PubMed  PubMed Central  Google Scholar 

  47. Whelton PK, Carey RM, Aronow WS, Casey DE, Collins KJ, Dennison Himmelfarb C, et al. 2017 ACC/AHA/AAPA/ABC/ACPM/AGS/APhA/ASH/ASPC/NMA/PCNA guideline for the prevention, detection, evaluation, and management of high blood pressure in adults: A report of the American college of cardiology/american heart association task force on clinical practice guidelines. J Am Coll Cardiol. 2018;71:e127–248.

  48. American Diabetes Association. 2. Classification and diagnosis of diabetes: standards of medical care in Diabetes-2021. Diabetes Care. 2021;44(Suppl 1):S15–33.

    Article  Google Scholar 

  49. Hong J, Hu X, Liu W, Qian X, Jiang F, Xu Z, et al. Impact of red cell distribution width and red cell distribution width/albumin ratio on all-cause mortality in patients with type 2 diabetes and foot ulcers: a retrospective cohort study. Cardiovasc Diabetol. 2022;21:91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Huang M, Liu F, Li Z, Liu Y, Su J, Ma M, et al. Relationship between red cell distribution width/albumin ratio and carotid plaque in different glucose metabolic States in patients with coronary heart disease: a RCSCD-TCM study in China. Cardiovasc Diabetol. 2023;22:39.

    Article  PubMed  PubMed Central  Google Scholar 

  51. Chen S, Guan S, Yan Z, Ouyang F, Li S, Liu L, et al. Prognostic value of red blood cell distribution width-to-albumin ratio in ICU patients with coronary heart disease and diabetes mellitus. Front Endocrinol (Lausanne). 2024;15:1359345.

    Article  PubMed  Google Scholar 

  52. Tan M, You R, Cai D, Wang J, Dai W, Yang R, et al. The red cell distribution width to albumin ratio: A novel prognostic Indicator in hepatitis B Virus-Related hepatocellular carcinoma. Int J Med Sci. 2025;22:441–50.

    Article  PubMed  PubMed Central  Google Scholar 

  53. Kim KM, Lui L-Y, Browner WS, Cauley JA, Ensrud KE, Kado DM, et al. Association between variation in red cell size and multiple Aging-Related outcomes. J Gerontol Biol Sci Med Sci. 2021;76:1288–94.

    Article  Google Scholar 

  54. Weuve J, Mendes de Leon CF, Bennett DA, Dong X, Evans DA. The red cell distribution width and anemia in association with prevalent dementia. Alzheimer Dis Assoc Disord. 2014;28:99–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Jiang Z, Han X, Wang Y, Hou T, Cong L, Tang S, et al. Red cell distribution width and dementia among Rural-Dwelling older adults: the MIND-China study. J Alzheimers Dis. 2021;83:1187–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Li D, Ruan Z, Wu B. Association of red blood cell distribution Width-Albumin ratio for acute myocardial infarction patients with mortality: A retrospective cohort study. Clin Appl Thromb Hemost. 2022;28:10760296221121286.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Taniguchi Y, Shinkai S, Nishi M, Murayama H, Nofuji Y, Yoshida H, et al. Nutritional biomarkers and subsequent cognitive decline among community-dwelling older Japanese: a prospective study. J Gerontol Biol Sci Med Sci. 2014;69:1276–83.

    Article  CAS  Google Scholar 

  58. Gatta A, Verardo A, Bolognesi M, Hypoalbuminemia. Intern Emerg Med. 2012;7(Suppl 3):S193–199.

    Article  PubMed  Google Scholar 

  59. Su Q, Li T, He PF, Lu XC, Yu Q, Gao QC, et al. Trichostatin A ameliorates Alzheimer’s disease-related pathology and cognitive deficits by increasing albumin expression and Aβ clearance in APP/PS1 mice. Alzheimers Res Ther. 2021;13:7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Shen J, Amari N, Zack R, Skrinak RT, Unger TL, Posavi M, et al. Plasma MIA, CRP, and albumin predict cognitive decline in Parkinson’s disease. Ann Neurol. 2022;92:255–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Boada M, López OL, Olazarán J, Núñez L, Pfeffer M, Paricio M, et al. A randomized, controlled clinical trial of plasma exchange with albumin replacement for Alzheimer’s disease: primary results of the AMBAR study. Alzheimers Dement. 2020;16:1412–25.

    Article  PubMed  Google Scholar 

  62. Murayama H, Shinkai S, Nishi M, Taniguchi Y, Amano H, Seino S, et al. Albumin, hemoglobin, and the trajectory of cognitive function in Community-Dwelling older Japanese: A 13-Year longitudinal study. J Prev Alzheimers Dis. 2017;4:93–9.

    CAS  PubMed  Google Scholar 

  63. Irwin MR, Vitiello MV. Implications of sleep disturbance and inflammation for Alzheimer’s disease dementia. Lancet Neurol. 2019;18:296–306.

    Article  CAS  PubMed  Google Scholar 

  64. Mason A, Holmes C, Edwards CJ. Inflammation and dementia: using rheumatoid arthritis as a model to develop treatments? Autoimmun Rev. 2018;17:919–25.

    Article  CAS  PubMed  Google Scholar 

  65. Won W, Choi H-J, Yoo J-Y, Kim D, Kim TY, Ju Y, et al. Inhibiting peripheral and central MAO-B ameliorates joint inflammation and cognitive impairment in rheumatoid arthritis. Exp Mol Med. 2022;54:1188–200.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Milosevich E, Demeyere N, Pendlebury ST. Infection, inflammation, and poststroke cognitive impairment. J Am Heart Assoc. 2024;13:e9130.

    Article  PubMed  PubMed Central  Google Scholar 

  67. Greene C, Connolly R, Brennan D, Laffan A, O’Keeffe E, Zaporojan L, et al. Blood-brain barrier disruption and sustained systemic inflammation in individuals with long COVID-associated cognitive impairment. Nat Neurosci. 2024;27:421–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Surendranathan A, Su L, Mak E, Passamonti L, Hong YT, Arnold R, et al. Early microglial activation and peripheral inflammation in dementia with lewy bodies. Brain. 2018;141:3415–27.

    Article  PubMed  PubMed Central  Google Scholar 

  69. Azarmanesh D, Bertone-Johnson ER, Pearlman J, Liu Z, Carbone ET. Association of the dietary inflammatory index with depressive symptoms among Pre- and Post-Menopausal women: findings from the National health and nutrition examination survey (NHANES) 2005–2010. Nutrients. 2022;14:1980.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Li W, Li S, Shang Y, Zhuang W, Yan G, Chen Z, et al. Associations between dietary and blood inflammatory indices and their effects on cognitive function in elderly Americans. Front Neurosci. 2023;17:1117056.

    Article  PubMed  PubMed Central  Google Scholar 

  71. Chen W, Sun X, Han J, Wu X, Wang Q, Li M, et al. Joint effect of abnormal systemic immune-inflammation index (SII) levels and diabetes on cognitive function and survival rate: A population-based study from the NHANES 2011–2014. PLoS ONE. 2024;19:e0301300.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Marruganti C, Baima G, Aimetti M, Grandini S, Sanz M, Romandini M. Periodontitis and low cognitive performance: A population-based study. J Clin Periodontol. 2023;50:418–29.

    Article  PubMed  Google Scholar 

  73. Zhang Y, Peng Y, Deng W, Xiang Q, Zhang W, Liu M. Association between dietary inflammatory index and cognitive impairment among American elderly: a cross-sectional study. Front Aging Neurosci. 2024;16:1371873.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Frith E, Shivappa N, Mann JR, Hébert JR, Wirth MD, Loprinzi PD. Dietary inflammatory index and memory function: population-based National sample of elderly Americans. Br J Nutr. 2018;119:552–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Chen SP, Bhattacharya J, Pershing S. Association of vision loss with cognition in older adults. JAMA Ophthalmol. 2017;135:963–70.

    Article  PubMed  PubMed Central  Google Scholar 

  76. Handing EP, Small BJ, Reynolds SL, Kumar NB. Impact of dietary factors and inflammation on cognition among older adults. J Prev Alzheimers Dis. 2015;2:220–6.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We sincerely express our gratitude to the participants and investigators of the NHANES study for their invaluable contributions, which have provided significant support and assistance to our research endeavors.

Funding

This research was funded by the Innovation Fund of the China Academy of Chinese Medical Sciences (CI2021A04701) and the National Key Research and Development Program of China (2021YFF 0901404).

Author information

Authors and Affiliations

Authors

Contributions

The study was conceived by BY, ML, and RG3, who were responsible for performing the data analysis and manuscript writing. ZY and HZ extracted the data from the official NHANES website. RG2 contributed to the revision and review of the manuscript. XF and AG conducted a repeat analysis of the data and verified the results. All authors have reviewed and approved the final version of the manuscript.

Corresponding authors

Correspondence to Rui Gao or Rui Gao.

Ethics declarations

Ethics approval and consent to participate

The NHANES protocol was approved by the National Center for Health Statistics and the Institutional Review Board. All participants provided written informed consent.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

12877_2025_5800_MOESM1_ESM.docx

Additional file: Table S1: The proportion of missing covariates and imputation methods; Table S2: Assessment of multicollinearity among independent variables; Table S3: ORs (95% CIs) for low cognitive performance according to RAR quartiles after excluding participants with any missing covariate values; Table S4: ORs (95% CIs) for low cognitive performance according to RAR quartiles after excluding extreme values (mean ± 3 standard deviations) of RAR; Table S5: ORs (95% CIs) for low cognitive performance according to RAR quartiles after excluding participants with self-reported cancer; Figure S1: Specific distribution of missing covariate values.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yu, B., Li, M., Yu, Z. et al. Red blood cell distribution width to albumin ratio (RAR) is associated with low cognitive performance in American older adults: NHANES 2011–2014. BMC Geriatr 25, 157 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12877-025-05800-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12877-025-05800-4

Keywords