Skip to main content
Erschienen in: BMC Nephrology 1/2022

Open Access 01.12.2022 | Research

Development of a risk prediction nomogram for sarcopenia in hemodialysis patients

verfasst von: Genlian Cai, Jinping Ying, Mengyan Pan, Xiabing lang, Weiping Yu, Qinqin Zhang

Erschienen in: BMC Nephrology | Ausgabe 1/2022

Abstract

Background

Sarcopenia is associated with various adverse outcomes in hemodialysis patients. However, current tools for assessing and diagnosing sarcopenia have limited applicability. In this study, we aimed to develop a simple and reliable nomogram to predict the risk of sarcopenia in hemodialysis patients that could assist physicians identify high-risk patients early.

Methods

A total of 615 patients undergoing hemodialysis at the First Affiliated Hospital College of Medicine Zhejiang University between March to June 2021 were included. They were randomly divided into either the development cohort (n = 369) or the validation cohort (n = 246). Multivariable logistic regression analysis was used to screen statistically significant variables for constructing the risk prediction nomogram for Sarcopenia. The line plots were drawn to evaluate the effectiveness of the nomogram in three aspects, namely differentiation, calibration, and clinical net benefit, and were further validated by the Bootstrap method.

Results

The study finally included five clinical factors to construct the nomogram, including age, C-reactive protein, serum phosphorus, body mass index, and mid-upper arm muscle circumference, and constructed a nomogram. The area under the ROC curve of the line chart model was 0.869, with a sensitivity and specificity of 77% sensitivity and 83%, the Youden index was 0.60, and the internal verification C-statistic was 0.783.

Conclusions

This study developed and validated a nomogram model to predict the risk of sarcopenia in hemodialysis patients, which can be used for early identification and timely intervention in high-risk groups.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
MHD
Maintenance hemodialysis
ESRD
End-stage renal disease
MRI
Magnetic resonance imaging
CT
Computed tomography
DXA
Dual-energy X-ray absorptiometry
BIA
Bioelectrical impedance analysis
SAS
Self-rating anxiety scale
SDS
Self-rating depression scale
BMI
Body mass index
ASM
Appendicular skeletal muscle mass
ROC
Receiver operating characteristic
AUC
The area under the receiver operating characteristic curve
DCA
Decision curve analysis
Kt/V
Urea clearance index
MAC
Mid-upper arm circumference
MAMC
Mid-upper arm muscle circumference
TSF
Triceps skin fold
PTH
Parathyroid hormone
HDL
High-density lipoprotein
LDL
Low-density lipoprotein
VLDL
Very low-density lipoprotein
SARC-F
Strength, assistance walking, rise from a chair, climb stairs and falls
SARC-CalF
Strength, assistance walking, rise from a chair, climb stairs, falls and calf

Introduction

Hemodialysis (HD) is one of the most common treatments for patients with end-stage renal disease (ESRD) [1]. Epidemiology studies [2] report that approximately 84% of all ESRD patients eventually receive hemodialysis treatment. Previous studies have indicated that patients with HD are predisposed to sarcopenia owing to the chronic inflammatory status, metabolic acidosis, malnutrition, and decreased physical activity [3]. Sarcopenia is characterized by a progressive and systemic loss of muscle mass and strength/function that is typically associated with numerous adverse outcomes [4, 5]. It has been estimated that 20–50% of all HD patients will develop sarcopenia, which is much higher than the general population [69]. Earlier studies have revealed that HD patients with comorbid sarcopenia had an increased risk of falls, fractures, and cardiovascular events as well as a higher likelihood re-hospitalization and death [1012]. Two recent systematic review and meta-analysis have also corroborated that sarcopenia is associated with a higher risk of death in hemodialysis patients [13, 14]. Other studies have demonstrated that that early identification and timely intervention can lower the occurrence and development of sarcopenia in hemodialysis patients [15, 16]. Therefore, the importance of early identification of high-risk groups of sarcopenia in hemodialysis patients cannot be overstated.
The diagnosis of sarcopenia is mainly based on low skeletal muscle mass, skeletal muscle strength, and physical performance [4]. Several technologies are currently being employed to estimate skeletal muscle mass, including magnetic resonance imaging (MRI), computed tomography (CT), dual-energy X-ray absorptiometry (DXA) and bioelectrical impedance analysis (BIA). Nevertheless, the use of DXA, MRI, and CT is limited in clinical practice by disadvantages such as high cost, complex procedures, and radiation exposure [17]. On the other hand, even though the BIA is convenient to perform, however, it was not applicable for the HD patients who experienced pacemaker implant and amputation [18]. The Asian Working Group for Sarcopenia (AWGS)2019 update proposes screening for sarcopenia with either SARC-F or SARC-CalF to facilitate earlier identification of high-risk individuals [15]. SARC-F is a self-reported questionnaire based on the patient’s perception of limitations in strength, walking ability, rising from a chair, stair climbing, and falls. The SARC-F score ≥ 4 indicates a risk of sarcopenia, however, numerous studies have demonstrated the low sensitivity and high specificity of SARC-F [19, 20]. SARC-CalF enhanced the sensitivity of SARC-F by the inclusion of calf circumference, with a score ≥ 11 indicates a risk of sarcopenia [21]. Despite the fact that SARC-F or SAC-CalF is simple to use and cost-free for identifying the risk of sarcopenia in hemodialysis patients, their results have been controversial [22, 23]. The relatively low sensitivity of these two scales renders a higher risk for miss diagnosis. As a result, the clinical medical staff cannot promptly identify the high-risk groups of sarcopenia in hemodialysis patients and thereby miss the window for early intervention.
To address the aforementioned challenges, we aimed to develop and validate a reliable nomogram model for predicting sarcopenia risk in hemodialysis patients. With this model, we hope to early identification of hemodialysis patients at high risk for sarcopenia and enable timely interventions to prevent or slow development and progression of sarcopenia. Thus improving patients adverse outcomes.

Methods

Study population

All patients undergoing hemodialysis at the First Affiliated Hospital College of Medicine Zhejiang University from March to June 2021 were enrolled in our study, and their medical records were retrospectively reviewed. Patients aged ≥ 18 years with a history of hemodialysis three times or more per week for at least 3 months were eligible to participate in this study. The exclusion criteria included implanted pacemaker or amputation surgeries; miss data of skeletal muscle mass index. Finally, 615 patients were enrolled in our study. They were randomly allocated to either the development cohort (n = 369) or the validation cohort (n = 246) in a 6:4 ratio. This study was performed in accordance with Declaration of Helsinki and was approved by the Clinical Research Ethics Committee of the First Affiliated Hospital of Zhejiang University School of Medicine (IIT20210808A), and all patients signed the informed consent form.

Data collection

Based on the relevant literature [3, 6, 8, 12, 24] and consulting experts, a total of 27 predictors for sarcopenia were identified. The following data were collected from each patient: ① baseline demographics: age, gender, primary disease, duration of dialysis, etc.; ② self-rating anxiety scale (SAS) and self-rating depression scale (SDS) psychological status assessment indicators; ③ anthropometric indicators: height, post-dialysis weight, skeletal muscle mass index (SMI), handgrip strength, gait speed, mid-upper arm circumference, triceps skinfold thickness, body mass index (BMI) and mid-upper arm muscle circumference (MAMC),which was calculated as mid-upper arm circumference − 3.14 × triceps skinfold thickness [18]. ④ laboratory examination parameters measured before a mid-week predialysis session following enrolment: serum creatinine, serum uric acid, serum urea nitrogen, hemoglobin, serum albumin, serum prealbumin, C-reactive protein, serum phosphorus, serum calcium, blood lipid, serum potassium, parathyroid hormone and urea clearance index.
Handgrip strength test was measured before hemodialysis with an electronic grip strength meter (EH101; CAMRY). The patient was required to stand the feet and arms naturally positioned. Three measurements with an interval of 5 s were taken, and the maximum value was recorded. The gait speed test was also evaluated before hemodialysis, without the assistance of any tools, the patient walked 6 m twice at a normal gait speed, each gait speed was observed, and the mean value of two consecutive measurements was documented. Triceps skinfold thickness and upper arm circumference were measured at the end of hemodialysis. In addition, BIA (InbodyS10, BiospaceCo,Korea) was performed 15–20 min after hemodialysis to measure appendicular skeletal muscle mass (ASM), which was later used to calculate SMI as follows: SMI = ASM/height2 [15].
The Zung self-rating depression scale (SDS) and Zung self-rating anxiety scale (SAS) were employed to assess patients' depression and anxiety levels; SAS and SDS have been found to have robust internal consistency with a Cronbach s alpha of 0.82 and 0.68, respectively. Both the SAS and SDS are measured on a scale containing 20 items, with each item scored on a four-point Likert scale, with “1” indicating no or little time, “2” representing a small amount of time, “3” referring to a lot of time, and “4” representing most or all of the time. The raw scores range from 20 to 80, which are subsequently converted into standard scores by dividing the sum of the raw scores by 80 and multiplying by 100 for further evaluation. the thresholds for identifying depression and anxiety were 50 points [25, 26].
Demographic characteristics and laboratory examination parameters were collected from medical records. The anthropometric indicators and SAS and SDS scales were determined by experienced nursing researchers; SAS and SDS were explained to patients by researchers using standardized guiding terms and were then filled in by the patients.

Diagnosis

In this study, the diagnosis of sarcopenia was based on the AWGS 2019 criteria [27]. ①low appendicular skeletal muscle mass (ASM): BIA < 7.0 kg/m2 in males and < 5.7 kg/m2 in females ②low muscle strength: handgrip strength < 28 kg in males and < 18 kg in females; ③low physical performance: gait speed (6-m walk): < 1.0 m/s. The diagnosis of sarcopenia was made in patients with low ASM + low muscle strength OR low physical performance.

Statistical analysis

SPSS 26 software (IBM Corporation, Armonk, NY, USA) and R software (version 4.2.1, R Foundation for Statistical Computing, Vienna, Austria) were used to analyze the collected data. Among the 27 predictor variables, prealbumin, urea clearance index, parathyroid hormone, upper arm circumference, and skinfold thickness were identified with less than 5% missing data, which were replaced by the series mean of the continuous variables before the model was developed. To ensure more convenient clinical use, continuous variables such as age, serum albumin, serum creatinine, C-reactive protein, serum phosphorus, serum calcium, parathyroid hormone, triglycerides, cholesterol, high-density lipoprotein, low-density lipoprotein, and very-low-density lipoprotein, BMI were converted into categorical variables according to clinical references [2730]. Other continuous variables were converted to dichotomous variables according to the receiver operating characteristic curve using the optimal cut-off points of the analyzed variables with the Youden index criterion. Frequency or constituent ratio was used for statistical description, and the chi-square test was used for comparison between groups. Through univariate logistic regression analysis, variables with univariate analysis results of p < 0.05 were included in the multivariate logistic regression analysis. The backward stepwise regression method was then applied to select statistically significant factors used to construct the clinical risk prediction nomogram. The predictive nomogram was assessed in three aspects: discrimination, calibration, and clinical net benefit. The area under the receiver operating characteristic curve (ROC) was utilized to assess discriminatory ability. The calibration curve and Hosmer–Lemeshow goodness of fit test were used to assess calibration ability. The decision curve analysis (DCA) was used to assess clinical effectiveness. The model was internally validated using the Bootstrap method(resampling = 1000). A p-value < 0.05 was considered statistically significant.

Results

Demographic parameters

A total of 615 patients undergoing hemodialysis were eventually included in this study and were randomly divided into the development cohort (n = 369) and the validation cohort (n = 246) in a 6:4 ratio, as illustrated in (Fig. 1). Among the enrolled patients, 381 (62%) were male and 234 (38%) were female. The mean patient age was 60.07 ± 14.34 years, the median duration of dialysis was 60 months (interquartile range 21–110). The most common reason for dialysis was chronic glomerulonephritis (430 patients), followed by diabetic nephropathy (101 patients), and other causes (hypertensive nephropathy, polycystic kidney disease, lupus nephritis, etc.). There were 102 patients (16.60%) diagnosed with sarcopenia and the demographic parameters of patients with or without sarcopenia are summarized in (Table 1).
Table 1
Baseline characteristics of the development and validation cohorts (n,%)
Characteristics
Development cohort(N = 369)
Validation cohort(N = 246)
P value
Sociodemographic
Gender
 Female
138(37.40)
96(39.00)
0.684
 Male
231(62.60)
150(61.00)
 
Age (≥ 60 years)
176(47.70)
126(51.20)
0.392
Sarcopenia
 No
310(84.00)
203(82.50)
0.626
 Yes
59(16.00)
43(17.50)
 
Dialysis and clinical
Primary disease
 Chronic glomerulonephrit
259(70.20)
171(69.50)
0.110
 Diabetic nephropathy
67(18.20)
34(13.80)
 
 Other
43(11.70)
41(16.70)
 
 Duration of dialysis (> 60 months)
179(48.50)
128(52.00)
0.392
Biochemical
 Hemoglobin (< 110 g/L)
149(40.40)
97(39.40)
0.814
 Creatinine (< 884 μmol/L)
175(47.40)
108(43.90)
0.390
 Uric acid (> 420 μmol/L)
201(54.50)
147(59.80)
0.195
 Urea nitrogen (> 20 mmol/L)
107(29.00)
69(28.00)
0.799
 Kt/V (> 1.4)
248(67.20)
168(68.30)
0.778
 Serum Albumin(< 38 g/L)
107(29.00)
85(34.60)
0.145
 CRP (≥ 3 mg/L)
140(37.90)
101(41.10)
0.438
 Alkaline phosphatase (> 70.5U/L)
194(52.60)
137(55.70)
0.448
 Prealbumin (≤ 30 mg/dL)
140(37.90)
85(34.60)
0.393
 Ferritin (< 200 ng/mL)
251(68.00)
171(69.50)
0.696
 Potassium (> 5.5 mmol/L)
104(28.20)
51(20.70)
0.037
Phosphorus
 1.13–1.78 mmol/L
148(40.10)
91(36.99)
0.707
  > 1.78 mmol/L
208(56.40)
147(59.76)
 
  < 1.13 mmol/L
13(3.50)
8(3.25)
 
Calcium
 2.1–2.5 mmol/L
243(65.86)
146(59.30)
0.149
  > 2.5 mmol/L
63(17.07)
43(17.50)
 
  < 2.1 mmol/L
63(17.07)
57(23.20)
 
PTH
 150-300 pg/L
138(37.40)
84(34.14)
0.180
  > 300 pg/L
74(20.10)
65(26.43)
 
  < 150 pg/L
157(42.50)
97(39.43)
 
 Triglycerides (> 1.7 mmol/L)
175(47.40)
119(48.40)
0.818
 Cholesterol (< 2.59 mmol/L)
41(11.10)
36(14.60)
0.196
 HDL (< 1.03 mmol/L)
141(38.20)
93(37.80)
0.919
 LDL (> 3.37 mmol/L)
7(1.90)
7(2.80)
0.440
 VLDL (> 0.78 mmol/L)
171(46.30)
115(46.70)
0.921
Anthropometric indicators
 MAMC (< 22.64 cm)
151(40.90)
112(45.50)
0.258
Body mass index
  
0.978
 18.5–24.9 kg/m2
243(65.85)
164(66.67)
 
  > 25 kg/m2
55(14.91)
36(14.63)
 
  < 18.5 kg/m2
71(19.24)
46(18.70)
 
Psychological scores
 SAS (> 40)
23(6.20)
13(5.30)
0.624
 SDS (> 41)
41(11.10)
33(13.40)
0.390
Kt/V Urea clearance index, CRP C-reactive protein, MAMC Mid-upper arm muscle circumference, SAS Self-rating anxiety scale, SDS Self-rating depression scale, PTH Parathyroid hormone, HDL High-density lipoprotein, LDL Low-density lipoprotein, VLDL Very low-density lipoprotein

Construction of the predictive nomogram

Univariate logistic regression analysis was performed with the included 27 independent variables, and the results demonstrated that age, urea clearance index, serum creatinine, upper arm muscle circumference, as well as levels of blood uric acid, albumin, C-reactive protein, serum phosphorus, alkaline phosphatase, BMI, prealbumin, and triglycerides were significantly different between the two groups (P < 0.05), as presented in (Table 2). Next, significant independent variables obtained from the above univariate logistic regression analysis were included in multivariate logistic regression analysis following a backward stepwise regression method. The results exposed that age, C-reactive protein, and serum phosphorus levels, as well as BMI and MAMC, were independent risk factors for sarcopenia in HD patients, as outlined in Table 2.
Table 2
Univariate and multivariate analysis of Sarcopenia in the Development cohort (n = 369)
 
Univariate analysis
Multivariate analysis
Characteristics
β
OR
95%CI
P
β
OR
95%
P
Gender (Female)
0.006
1.01
0.57–1.79
0.985
    
Age(≥ 60 years)
0.901
2.46
1.37–4.42
0.002
0.651
1.92
0.94–3.93
0.075
Diabetic nephropathy
0.58
1.79
0.91–3.51
0.091
    
Other
0.348
1.42
0.61–3.3
0.42
    
Duration of dialysis (> 60 months)
0.273
1.31
0.75–2.3
0.338
    
Hemoglobin (< 110 g/L)
0.344
1.41
0.81–2.47
0.228
    
Creatinine (< 884 μmol/L)
1.388
4.01
2.14–7.5
 < 0.001
    
Uric acid (> 420 μmol/L)
-0.58
0.56
0.32–0.98
0.043
    
Urea nitrogen (> 20 mmol/L)
0.542
1.72
0.97–3.07
0.067
    
KTV (> 1.4)
1.297
3.66
1.68–7.98
0.001
    
Serum Albumin (< 38 g/L)
0.886
2.43
1.37–4.3
0.002
    
Alkaline phosphatase (> 70.5U/L)
0.58
1.79
1–3.18
0.049
    
CRP (≥ 3 mg/L)
0.962
2.62
1.49–4.61
0.001
1.168
3.22
1.58–6.54
0.001
Prealbumin (≤ 30 mg/dL)
0.714
2.04
1.16–3.58
0.013
    
Ferritin (< 200 ng/mL)
-0.369
0.69
0.39–1.23
0.21
    
Potassium (> 5.5 mmol/L)
-0.167
0.85
0.45–1.6
0.607
    
Phosphorus (> 1.78 mmol/L)
-0.871
0.42
0.23–0.77
0.005
-1.011
0.36
0.18–0.74
0.006
Phosphorus (< 1.13 mmol/L)
2.18
8.85
2.55–30.72
0.001
2.769
15.94
3.28–77.35
0.001
Calcium (> 2.5 mmol/L)
-0.304
0.74
0.33–1.67
0.466
    
Calcium (< 2.1 mmol/L)
0.071
1.07
0.52–2.23
0.85
    
PTH (> 300 pg/L)
-0.697
0.5
0.21–1.16
0.106
    
PTH (< 150 pg/L)
-0.299
0.74
0.4–1.36
0.333
    
Triglycerides (> 1.7 mmol/L)
-0.668
0.51
0.29–0.92
0.025
    
Cholesterol (< 2.59 mmol/L)
-0.35
0.7
0.26–1.88
0.484
    
HDL (< 1.03 mmol/L)
-0.536
0.59
0.33–1.03
0.061
    
LDL (> 3.37 mmol/L)
-0.135
0.87
0.1–7.38
0.901
    
VLDL (> 0.78 mmol/L)
-0.358
0.7
0.4–1.23
0.218
    
MAMC (< 22.64 cm)
1.933
6.91
3.58–13.35
 < 0.001
1.422
4.15
1.82–9.05
0.001
BMI (> 25 kg/m2)
-1.99
0.14
0.02–1.02
0.053
-2.122
0.12
0.01–1.14
0.065
BMI (< 18.5 kg/m2)
1.628
5.1
2.76–9.39
 < 0.001
1.428
4.15
1.91–6.21
 < 0.001
SAS (> 40)
0.668
1.95
0.74–5.18
0.179
    
SDS (> 41)
0.447
1.56
0.7–3.47
0.272
    
Kt/V Urea clearance index, CRP C-reactive protein, MAMC Mid-upper arm muscle circumference, SAS Self-rating anxiety scale, SDS Self-rating depression scale, PTH Parathyroid hormone, HDL High-density lipoprotein, LDL Low-density lipoprotein, VLDL Very low-density lipoprotein
The risk prediction model for sarcopenia in HD patients was then established based on the formula: P = 1/(1 + eY) where e stands for the base of natural logarithm, Y = -3.316 + 0.651 × age ≥ 60 years + 1.168 × C-reactive protein ≥ 3 mg/L + 2.769 × serum phosphorus < 1.13 mmol/L-1.011 × serum phosphorus 1.13 ~ 1.78 mmol/L + 1.428 × BMI < 18.5 kg/m2 -2.122 × BMI 18.5 ~ 24.9 kg/m2 + 1.422 × MAMC < 22.64 cm. A nomogram of the risk prediction model for sarcopenia in HD patients was also plotted, as depicted in (Fig. 2). The corresponding score for each variable was obtained by crossing to the nomogram, and the total score was used to predict the risk of developing sarcopenia in HD patients.

Validation of the predictive model

The validation of this prediction model was performed by area under the curve and the calibration curve. Our data indicated that the AUC for this model was 0.869 (95% CI (0.822 to 0.915) (Fig. 3), with a sensitivity, specificity, and Youden index of 77% and 83%, and 0.60, respectively, in the development cohort. Similarly, the AUC of the validation cohort was 0.832 (95% CI (0.765 to 0.900) (Fig. 4), with a sensitivity of 70%, a specificity of 88%, and a Youden index of 0.58. In addition, the Hosmer–Lemeshow goodness-of-fit test showed promising fit (development cohort \(\chi\) 2 = 4.001, P = 0.911; validation cohort \(\chi\) 2 = 13.941, P = 0.124). In the development cohort and validation cohort, the calibration curve showed agreement between the observation and prediction as displayed in (Figs. 5 and 6). The model was then internally validated using the Bootstrap method with an internal validation C-statistic of 0.783.

Clinical validity

The clinical validity of this predictive model was assessed using decision curve analysis (DCA). The DCA for the development and validation cohorts are displayed in (Figs. 7 and 8), which signaled that patients could benefit from this novel predictive model when the threshold was set to 10 ~ 80% and 10 ~ 70% for the development and validation cohorts, respectively. The positive predictive value, negative predictive value, positive likelihood ratio, negative likelihood ratio for this nomogram were 45%,93%,4.34,0.418, Accuracy was 82%.
To make this predictive model more convenient for physicians to use in clinical practice, we modified the nomogram into a scoring system with integer points: age ≥ 60 years(18points), C-reactive protein ≥ 3 mg/L (30points), serum phosphorus < 1.13 mmol/L (100points), serum phosphorus 1.13 ~ 1.78 mmol/L (27 points) and BMI < 18.5 kg/m2 (93 points), BMI 18.5 ~ 24.9 kg/m2 (56 points), MAMC < 22.64 cm(37 points). Then, the risk score model of sarcopenia in hemodialysis patients was established. The total score was 0–350 points of the nomogram, and the higher of the total score, the higher the risk of sarcopenia. The weights for each feature are list in Table 3 for calculation without a nomogram. the AUC of the scoring system was 0.867 (95% CI (0.830 to 0.903), with a sensitivity, specificity, and Youden index of 88% and 73%, and 0.611. The optimal cutoff value of 121 was taken as the risk threshold, if the total score < 121 points was classified as the low-risk group of sarcopenia, and the total score > 121 points was classified as the high-risk group.
Table 3
The points for predictors
Predictor
points
Age
  < 60 years
0
  ≥ 60 years
18
CRP
  < 3 mg/L
0
  ≥ 3 mg/L
30
Serum Phosphorus
 1.13–1.78 mmol/L
27
  > 1.78 mmol/L
0
  < 1.13 mmol/L
100
BMI
 18.5–24.9 kg/m2
56
  ≥ 25 kg/m2
0
  < 18.5 kg/m2
93
MAMC
  ≥ 22.64 cm
0
  < 22.64 cm
37
Total points
Risk of sarcopenia
112
0.1
133
0.2
147
0.3
159
0.4
170
0.5
181
0.6
192
0.7
206
0.8
228
0.9
CRP C-reactive protein, BMI Body mass index, MAMC Mid-upper arm muscle circumference

Discussion

Herein, we developed and validated a simple nomogram to predict the risk of developing sarcopenia in HD patients with a total of five clinically relevant variables, including age, C-reactive protein, serum phosphorus, BMI, and MAMC. The AUC, internal validation C-statistic, calibration curve, and DCA curve were constructed to validate the reliability as well as the accuracy of this model. The nomogram model can early identify hemodialysis patients at risk of sarcopenia, and enable timely interventions to prevent or slow development and progression. thus improving patients adverse outcomes.
Nomograms can predict the probability of disease by analyzing and integrating identified disease risk factors, thus providing valuable information for better clinical decisions. It has been extensively used in oncology and chronic diseases worldwide [24]. For instance, Cheng et al. [31] designed a nomogram to predict the risk of initiating renal replacement therapy within 3 years in diabetic nephropathy patients, while Jing et al. [32] developed a nomogram comprising multiple echocardiographic measures to assess 3-year all-cause mortality in hemodialysis patients, both of which showed favorable accuracy and reliability. Ouyang et al. [33]developed and validated an easy-to-use nomogram that can accurately predict 1-year, 5-year, and 10-year survival in hemodialysis patients. In addition, Mo, et al. [34] reported the development of nomograms to predict sarcopenia in community older adults. However, to the best of our knowledge, no study has been conducted to construct a nomogram that can predict the risk of developing sarcopenia in HD patients.
In this study, the multivariate logistic regression analysis indicated that age was a risk factor for sarcopenia in HD patients. Aging is a well-known independent risk factor for sarcopenia, With the increase of age, the protein breakdown and anabolic metabolism of patients are gradually unbalanced. In addition, mitochondrial dysfunction and hormonal changes caused by aging are also related to sarcopenia [5]. Our findings on the relationship between age and sarcopenia are consistent with previous studies [6, 8, 35]. However, there are some controversies, Wang et al. [14] systematic review have shown the age of HD patients was no significant influence on sarcopenia prevalence.
Hyperphosphatemia is a common complication in HD patients and is closely related to an increased risk of vascular calcification and cardiovascular mortality [36]. Interestingly, our study indicated that lower serum phosphorus level correlated with the development of sarcopenia in HD patients, which was in line with the findings of Ren et al. [8]. We hypothesized that a high-protein diet is the main source of phosphorus for uremic patients who often suffer from loss of appetite or anorexia. A decrease in food intake will inevitably lead to a decrease in serum phosphorus, malnutrition, and protein-energy expenditure in patients, ultimately resulting in sarcopenia [37]. Previous studies have shown that shown protein restriction to correlate with increased mortality in patients undergoing HD, restricting dietary protein to help control phosphorus levels in patients undergoing maintenance HD may be more harmful than beneficial [36]. Balancing Nutrition and Serum Phosphorus in HD patients requires an individualized approach, involving a combination of adequate dietary advice, phosphatebinder use, and adjustments to dialysis prescription.
BMI and MAMC are conventional nutritional assessments for HD patients, and previous studies [3840] indicated that both are independent predictors of survival. The study conducted by Su et al. [41] exposed that the decrease in MAC was associated with increased all-cause mortality and cardiac events in HD patients, especially in those with low BMI. Unsurprisingly, the data in our study showed that HD patients with decreased BMI and MAMC were more likely to develop sarcopenia, which is consistent with previous studies [42, 43].
Consistent with previous studies [12, 38], This study showed that the level of C-reactive protein was increased in HD patients who developed sarcopenia compared to non-sarcopenia patients. the role of inflammation as a risk factor for malnutrition has been more and more recognized, C-reactive protein is one of the most frequently utilized biochemical indicators to examine inflammation. It has been well established in the field that hemodialysis patients are often under micro inflammatory state for multiple reasons [44]. Indeed, the close association between inflammation and sarcopenia has been well documented. Inflammatory factors can activate numerous signaling pathways involved in the pathogenesis of sarcopenia, resulting in decreased anabolism and increased catabolism of proteins [45]. A systematic review shows that exercise training can be beneficial for both the body composition and nutritional status in hemodialysis patients, the MAMC, BMI, Serum albumin increase and C-reactive protein decrease after resistance exercise [46]. Therefore, attention should be paid to these high-risk patients and early interventions should be taken to improve their outcomes.
Hemodialysis patients often have report significant psychological distress, Depression is common in HD patients [47]. depression patients are significantly less involved in social, professional, and recreational activities resulting in less physical activity [48], Infection [47], these factors are related to the development of sarcopenia. Several previous studies have shown an association between depression and sarcopenia in hemodialysis patients [4951], there is a higher prevalence of depression in sarcopenia patients. however, we did not find any significant association between depression and sarcopenia. This may be due to differences in study populations and assessment tools.
Currently, commonly used scales for sarcopenia are the SARC-F score and the modified SARC-assisted Cal F score. A meta-analysis [23] revealed that the sensitivity of SARC-F was low to moderate (28.9% – 55.3%), and so was its specificity (68.9% – 88.9%). Although SARC-CalF is associated with higher specificity (87.7% – 91.3%), its sensitivity is not satisfactory (45.9% – 57.2%). The relatively low sensitivity of these two scales renders a higher risk for misdiagnosis. On the contrary, our novel nomogram provided an alternative method with increased clinical efficacy. The AUC of our constructed nomogram model was 0.869 in the development cohort and 0.832 in the validation cohort with an internal validation C-statistic of 0.783. The validity of this novel model was further verified by calibration and DCA curves. The sensitivity, specificity, positive predictive value, negative predictive value, positive likelihood ratio, negative likelihood ratio of our constructed nomogram model were 77%,83%,45%,93%,4.34,0.418, Accuracy was 82%. More importantly, all five variables included in this model are laboratory and anthropometric measurements that are routinely determined in clinical practice and do not require additional examinations or costs. To make this predictive model more convenient for physicians to use in clinical practice, we modified the nomogram into a scoring system with integer points, the scoring system has ease of visualization. If the total score > 121 points indicates that the patient has a high risk of sarcopenia, should be timely comprehensive interventions, such as exercise training, nutritional interventions to reduce the occurrence of sarcopenia.
However, this study has some limitations that need to be taken into account. First, even though the number of enrollments was relatively large, it was conducted at a single center that might not be representative of the HD patients in other areas. Second, our study was retrospectively constructed and hemodialysis patients level of physical activity, serum bicarbonate, vitamin D level and nutritional status were not included in the analysis, potentially reducing the performance of the model. Third, the constructed nomogram model was not validated using external data. Fourth, this study excluded cases with missing data, which may have led to selection bias. Therefore, this model should be validated through prospective, multicenter clinical studies in the future.

Conclusion

In this study, a predictive nomogram model was constructed based on conventional serology and noninvasive anthropometric measurements that could accurately predict the risk of developing sarcopenia in HD patients. Our model maybe assist physicians in identifying the high risk of sarcopenia in hemodialysis patients earlier and adopt effective intervention strategies to improving patients adverse outcomes. In the future, it is necessary to further optimize the model through prospective, multi-center clinical research.

Acknowledgements

We would like to acknowledge all the participants in this study.

Declarations

The research protocol was approved by the Institutional Review Board of the First Affiliated Hospital, Zhejiang University School of Medicine, and conducted according to the principles of the Declaration of Helsinki. Due to the retrospective nature of the study, written informed consent for participation was waived by the Institutional Review Board of the First Affiliated Hospital, Zhejiang University School of Medicine.
Not applicable.

Competing interests

The authors declare that they have no conflict of interest.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Liyanage T, Ninomiya T, Jha V, Neal B, Patrice HM, Okpechi I, Zhao MH, Lv J, Garg AX, Knight J, et al. Worldwide access to treatment for end-stage kidney disease: a systematic review. The Lancet. 2015;385(9981):1975–82.CrossRef Liyanage T, Ninomiya T, Jha V, Neal B, Patrice HM, Okpechi I, Zhao MH, Lv J, Garg AX, Knight J, et al. Worldwide access to treatment for end-stage kidney disease: a systematic review. The Lancet. 2015;385(9981):1975–82.CrossRef
2.
Zurück zum Zitat Kramer A, Pippias M, Noordzij M, Stel VS, Andrusev AM, Aparicio-Madre MI, Arribas Monzon FE, Asberg A, Barbullushi M, Beltran P, et al. The European Renal Association - European Dialysis and Transplant Association (ERA-EDTA) registry annual report 2016: a summary. Clin Kidney J. 2019;12(5):702–20.PubMedPubMedCentralCrossRef Kramer A, Pippias M, Noordzij M, Stel VS, Andrusev AM, Aparicio-Madre MI, Arribas Monzon FE, Asberg A, Barbullushi M, Beltran P, et al. The European Renal Association - European Dialysis and Transplant Association (ERA-EDTA) registry annual report 2016: a summary. Clin Kidney J. 2019;12(5):702–20.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Fahal IH. Uraemic sarcopenia: aetiology and implications. Nephrol Dial Transplant. 2014;29(9):1655–65.PubMedCrossRef Fahal IH. Uraemic sarcopenia: aetiology and implications. Nephrol Dial Transplant. 2014;29(9):1655–65.PubMedCrossRef
4.
Zurück zum Zitat Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412–23.PubMedPubMedCentralCrossRef Cruz-Jentoft AJ, Baeyens JP, Bauer JM, Boirie Y, Cederholm T, Landi F, Martin FC, Michel JP, Rolland Y, Schneider SM, et al. Sarcopenia: European consensus on definition and diagnosis: Report of the European Working Group on Sarcopenia in Older People. Age Ageing. 2010;39(4):412–23.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Cruz-Jentoft AJ, Sayer AA. Sarcopenia. The Lancet. 2019;393(10191):2636–46.CrossRef Cruz-Jentoft AJ, Sayer AA. Sarcopenia. The Lancet. 2019;393(10191):2636–46.CrossRef
6.
Zurück zum Zitat Kim JK, Choi SR, Choi MJ, Kim SG, Lee YK, Noh JW, Kim HJ, Song YR. Prevalence of and factors associated with sarcopenia in elderly patients with end-stage renal disease. Clin Nutr. 2014;33(1):64–8.PubMedCrossRef Kim JK, Choi SR, Choi MJ, Kim SG, Lee YK, Noh JW, Kim HJ, Song YR. Prevalence of and factors associated with sarcopenia in elderly patients with end-stage renal disease. Clin Nutr. 2014;33(1):64–8.PubMedCrossRef
7.
Zurück zum Zitat Mori K, Nishide K, Okuno S, Shoji T, Emoto M, Tsuda A, Nakatani S, Imanishi Y, Ishimura E, Yamakawa T, et al. Impact of diabetes on sarcopenia and mortality in patients undergoing hemodialysis. BMC Nephrol. 2019;20(1):105.PubMedPubMedCentralCrossRef Mori K, Nishide K, Okuno S, Shoji T, Emoto M, Tsuda A, Nakatani S, Imanishi Y, Ishimura E, Yamakawa T, et al. Impact of diabetes on sarcopenia and mortality in patients undergoing hemodialysis. BMC Nephrol. 2019;20(1):105.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Ren H, Gong D, Jia F, Xu B, Liu Z. Sarcopenia in patients undergoing maintenance hemodialysis: incidence rate, risk factors and its effect on survival risk. Ren Fail. 2016;38(3):364–71.PubMedCrossRef Ren H, Gong D, Jia F, Xu B, Liu Z. Sarcopenia in patients undergoing maintenance hemodialysis: incidence rate, risk factors and its effect on survival risk. Ren Fail. 2016;38(3):364–71.PubMedCrossRef
9.
Zurück zum Zitat Shafiee G, Keshtkar A, Soltani A, Ahadi Z, Larijani B, Heshmat R. Prevalence of sarcopenia in the world: a systematic review and meta- analysis of general population studies. J Diabetes Metab Disord. 2017;16:21.PubMedPubMedCentralCrossRef Shafiee G, Keshtkar A, Soltani A, Ahadi Z, Larijani B, Heshmat R. Prevalence of sarcopenia in the world: a systematic review and meta- analysis of general population studies. J Diabetes Metab Disord. 2017;16:21.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Kim JK, Kim SG, Oh JE, Lee YK, Noh JW, Kim HJ, Song YR. Impact of sarcopenia on long-term mortality and cardiovascular events in patients undergoing hemodialysis. Korean J Intern Med. 2019;34(3):599–607.PubMedCrossRef Kim JK, Kim SG, Oh JE, Lee YK, Noh JW, Kim HJ, Song YR. Impact of sarcopenia on long-term mortality and cardiovascular events in patients undergoing hemodialysis. Korean J Intern Med. 2019;34(3):599–607.PubMedCrossRef
11.
Zurück zum Zitat Lai S, Muscaritoli M, Andreozzi P, Sgreccia A, De Leo S, Mazzaferro S, Mitterhofer AP, Pasquali M, Protopapa P, Spagnoli A, et al. Sarcopenia and cardiovascular risk indices in patients with chronic kidney disease on conservative and replacement therapy. Nutrition. 2019;62:108–14.PubMedCrossRef Lai S, Muscaritoli M, Andreozzi P, Sgreccia A, De Leo S, Mazzaferro S, Mitterhofer AP, Pasquali M, Protopapa P, Spagnoli A, et al. Sarcopenia and cardiovascular risk indices in patients with chronic kidney disease on conservative and replacement therapy. Nutrition. 2019;62:108–14.PubMedCrossRef
12.
Zurück zum Zitat Wilkinson TJ, Miksza J, Yates T, Lightfoot CJ, Baker LA, Watson EL, Zaccardi F, Smith AC. Association of sarcopenia with mortality and end-stage renal disease in those with chronic kidney disease: a UK Biobank study. J Cachexia Sarcopenia Muscle. 2021;12(3):586–98.PubMedPubMedCentralCrossRef Wilkinson TJ, Miksza J, Yates T, Lightfoot CJ, Baker LA, Watson EL, Zaccardi F, Smith AC. Association of sarcopenia with mortality and end-stage renal disease in those with chronic kidney disease: a UK Biobank study. J Cachexia Sarcopenia Muscle. 2021;12(3):586–98.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Ribeiro HS, Neri SGR, Oliveira JS, Bennett PN, Viana JL, Lima RM. Association between sarcopenia and clinical outcomes in chronic kidney disease patients: a systematic review and meta-analysis. Clin Nutr. 2022;41(5):1131–40.PubMedCrossRef Ribeiro HS, Neri SGR, Oliveira JS, Bennett PN, Viana JL, Lima RM. Association between sarcopenia and clinical outcomes in chronic kidney disease patients: a systematic review and meta-analysis. Clin Nutr. 2022;41(5):1131–40.PubMedCrossRef
14.
Zurück zum Zitat Shu X, Lin T, Wang H, Zhao Y, Jiang T, Peng X, Yue J. Diagnosis, prevalence, and mortality of sarcopenia in dialysis patients: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(1):145–58.PubMedPubMedCentralCrossRef Shu X, Lin T, Wang H, Zhao Y, Jiang T, Peng X, Yue J. Diagnosis, prevalence, and mortality of sarcopenia in dialysis patients: a systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(1):145–58.PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyere O, Cederholm T, Cooper C, Landi F, Rolland Y, Sayer AA, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(1):16–31.PubMedCrossRef Cruz-Jentoft AJ, Bahat G, Bauer J, Boirie Y, Bruyere O, Cederholm T, Cooper C, Landi F, Rolland Y, Sayer AA, et al. Sarcopenia: revised European consensus on definition and diagnosis. Age Ageing. 2019;48(1):16–31.PubMedCrossRef
16.
Zurück zum Zitat Yoshimura Y, Wakabayashi H, Yamada M, Kim H, Harada A, Arai H. Interventions for Treating Sarcopenia: A Systematic Review and Meta-Analysis of Randomized Controlled Studies. J Am Med Dir Assoc. 2017;18(6):553 e551-553 e516.CrossRef Yoshimura Y, Wakabayashi H, Yamada M, Kim H, Harada A, Arai H. Interventions for Treating Sarcopenia: A Systematic Review and Meta-Analysis of Randomized Controlled Studies. J Am Med Dir Assoc. 2017;18(6):553 e551-553 e516.CrossRef
17.
Zurück zum Zitat Buckinx F, Landi F, Cesari M, Fielding RA, Visser M, Engelke K, Maggi S, Dennison E, Al-Daghri NM, Allepaerts S, et al. Pitfalls in the measurement of muscle mass: a need for a reference standard. J Cachexia Sarcopenia Muscle. 2018;9(2):269–78.PubMedPubMedCentralCrossRef Buckinx F, Landi F, Cesari M, Fielding RA, Visser M, Engelke K, Maggi S, Dennison E, Al-Daghri NM, Allepaerts S, et al. Pitfalls in the measurement of muscle mass: a need for a reference standard. J Cachexia Sarcopenia Muscle. 2018;9(2):269–78.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Slee A, McKeaveney C, Adamson G, Davenport A, Farrington K, Fouque D, Kalantar-Zadeh K, Mallett J, Maxwell AP, Mullan R, et al. Estimating the prevalence of muscle wasting, weakness, and sarcopenia in hemodialysis patients. J Ren Nutr. 2020;30(4):313–21.PubMedCrossRef Slee A, McKeaveney C, Adamson G, Davenport A, Farrington K, Fouque D, Kalantar-Zadeh K, Mallett J, Maxwell AP, Mullan R, et al. Estimating the prevalence of muscle wasting, weakness, and sarcopenia in hemodialysis patients. J Ren Nutr. 2020;30(4):313–21.PubMedCrossRef
19.
Zurück zum Zitat Malmstrom TK, Morley JE. SARC-F: a simple questionnaire to rapidly diagnose sarcopenia. J Am Med Dir Assoc. 2013;14(8):531–2.PubMedCrossRef Malmstrom TK, Morley JE. SARC-F: a simple questionnaire to rapidly diagnose sarcopenia. J Am Med Dir Assoc. 2013;14(8):531–2.PubMedCrossRef
20.
Zurück zum Zitat Woo J, Leung J, Morley JE. Validating the SARC-F: a suitable community screening tool for sarcopenia? J Am Med Dir Assoc. 2014;15(9):630–4.PubMedCrossRef Woo J, Leung J, Morley JE. Validating the SARC-F: a suitable community screening tool for sarcopenia? J Am Med Dir Assoc. 2014;15(9):630–4.PubMedCrossRef
21.
Zurück zum Zitat Lim WS, Chew J, Lim JP, Tay L, Hafizah N, Ding YY. Letter to the editor: case for validated instead of standard cut-offs for SARC-CalF. J Nutr Health Aging. 2019;23(4):393–5.PubMedCrossRef Lim WS, Chew J, Lim JP, Tay L, Hafizah N, Ding YY. Letter to the editor: case for validated instead of standard cut-offs for SARC-CalF. J Nutr Health Aging. 2019;23(4):393–5.PubMedCrossRef
22.
Zurück zum Zitat Duarte PM, Ribeiro HS, Almeida LS, Baiao VM, Inda-Filho A, Avesani CM, Ferreira AP, Lima RM. SARC-F and SARC-CalF are associated with sarcopenia traits in hemodialysis patients. Nutr Clin Pract 2022. Duarte PM, Ribeiro HS, Almeida LS, Baiao VM, Inda-Filho A, Avesani CM, Ferreira AP, Lima RM. SARC-F and SARC-CalF are associated with sarcopenia traits in hemodialysis patients. Nutr Clin Pract 2022.
23.
Zurück zum Zitat Voelker SN, Michalopoulos N, Maier AB, Reijnierse EM. Reliability and concurrent validity of the SARC-F and its modified versions: a systematic review and meta-analysis. J Am Med Dir Assoc. 2021;22(9):1864-1876 e1816.PubMedCrossRef Voelker SN, Michalopoulos N, Maier AB, Reijnierse EM. Reliability and concurrent validity of the SARC-F and its modified versions: a systematic review and meta-analysis. J Am Med Dir Assoc. 2021;22(9):1864-1876 e1816.PubMedCrossRef
27.
Zurück zum Zitat Chen LK, Woo J, Assantachai P, Auyeung TW, Chou MY, Iijima K, Jang HC, Kang L, Kim M, Kim S, et al. Asian working group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J Am Med Dir Assoc. 2020;21(3):300-307 e302.PubMedCrossRef Chen LK, Woo J, Assantachai P, Auyeung TW, Chou MY, Iijima K, Jang HC, Kang L, Kim M, Kim S, et al. Asian working group for sarcopenia: 2019 consensus update on sarcopenia diagnosis and treatment. J Am Med Dir Assoc. 2020;21(3):300-307 e302.PubMedCrossRef
28.
Zurück zum Zitat Fouque D, Kalantar-Zadeh K, Kopple J, Cano N, Chauveau P, Cuppari L, Franch H, Guarnieri G, Ikizler TA, Kaysen G, et al. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int. 2008;73(4):391–8.PubMedCrossRef Fouque D, Kalantar-Zadeh K, Kopple J, Cano N, Chauveau P, Cuppari L, Franch H, Guarnieri G, Ikizler TA, Kaysen G, et al. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int. 2008;73(4):391–8.PubMedCrossRef
29.
Zurück zum Zitat Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Update Work Group. KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney Int Suppl. 2017;7:1–59. Kidney Disease: Improving Global Outcomes (KDIGO) CKD-MBD Update Work Group. KDIGO 2017 Clinical Practice Guideline Update for the Diagnosis, Evaluation, Prevention, and Treatment of Chronic Kidney Disease-Mineral and Bone Disorder (CKD-MBD). Kidney Int Suppl. 2017;7:1–59.
30.
Zurück zum Zitat Yang S, Su T, Huang L, Feng L-H, Liao T. A novel risk-predicted nomogram for sepsis associated-acute kidney injury among critically ill patients. BMC Nephrology. 2021;22(1):173.PubMedPubMedCentralCrossRef Yang S, Su T, Huang L, Feng L-H, Liao T. A novel risk-predicted nomogram for sepsis associated-acute kidney injury among critically ill patients. BMC Nephrology. 2021;22(1):173.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Cheng Y, Shang J, Liu D, Xiao J, Zhao Z. Development and validation of a predictive model for the progression of diabetic kidney disease to kidney failure. Ren Fail. 2020;42(1):550–9.PubMedPubMedCentralCrossRef Cheng Y, Shang J, Liu D, Xiao J, Zhao Z. Development and validation of a predictive model for the progression of diabetic kidney disease to kidney failure. Ren Fail. 2020;42(1):550–9.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Zhu J, Tang C, Ouyang H, Shen H, You T, Hu J. Prediction of all-cause mortality using an echocardiography-based risk score in hemodialysis patients. Cardiorenal Med. 2021;11(1):33–43.PubMedCrossRef Zhu J, Tang C, Ouyang H, Shen H, You T, Hu J. Prediction of all-cause mortality using an echocardiography-based risk score in hemodialysis patients. Cardiorenal Med. 2021;11(1):33–43.PubMedCrossRef
33.
Zurück zum Zitat Ouyang H, Shi Q, Zhu J, Shen H, Jiang S, Song K. Nomogram for predicting 1-, 5-, and 10-year survival in hemodialysis (HD) patients: a single center retrospective study. Ren Fail. 2021;43(1):1508–19.PubMedPubMedCentralCrossRef Ouyang H, Shi Q, Zhu J, Shen H, Jiang S, Song K. Nomogram for predicting 1-, 5-, and 10-year survival in hemodialysis (HD) patients: a single center retrospective study. Ren Fail. 2021;43(1):1508–19.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Mo YH, Su YD, Dong X, Zhong J, Yang C, Deng WY, Yao XM, Liu BB, Wang XH. Development and validation of a nomogram for predicting sarcopenia in community-dwelling older adults. J Am Med Dir Assoc. 2022;23(5):715-721 e715.PubMedCrossRef Mo YH, Su YD, Dong X, Zhong J, Yang C, Deng WY, Yao XM, Liu BB, Wang XH. Development and validation of a nomogram for predicting sarcopenia in community-dwelling older adults. J Am Med Dir Assoc. 2022;23(5):715-721 e715.PubMedCrossRef
35.
Zurück zum Zitat Sanchez-Tocino ML, Miranda-Serrano B, Lopez-Gonzalez A, Villoria-Gonzalez S, Pereira-Garcia M, Gracia-Iguacel C, Gonzalez-Ibarguren I, Ortiz-Arduan A, Mas-Fontao S, Gonzalez-Parra E. Sarcopenia and mortality in older hemodialysis patients. Nutrients. 2022;14(11):2354.PubMedPubMedCentralCrossRef Sanchez-Tocino ML, Miranda-Serrano B, Lopez-Gonzalez A, Villoria-Gonzalez S, Pereira-Garcia M, Gracia-Iguacel C, Gonzalez-Ibarguren I, Ortiz-Arduan A, Mas-Fontao S, Gonzalez-Parra E. Sarcopenia and mortality in older hemodialysis patients. Nutrients. 2022;14(11):2354.PubMedPubMedCentralCrossRef
36.
Zurück zum Zitat Palmer SC, Hayen A, Macaskill P, Pellegrini F, Craig JC, Elder GJ, Strippoli GF. Serum levels of phosphorus, parathyroid hormone, and calcium and risks of death and cardiovascular disease in individuals with chronic kidney disease: a systematic review and meta-analysis. JAMA. 2011;305(11):1119–27.PubMedCrossRef Palmer SC, Hayen A, Macaskill P, Pellegrini F, Craig JC, Elder GJ, Strippoli GF. Serum levels of phosphorus, parathyroid hormone, and calcium and risks of death and cardiovascular disease in individuals with chronic kidney disease: a systematic review and meta-analysis. JAMA. 2011;305(11):1119–27.PubMedCrossRef
37.
Zurück zum Zitat Fouque D, Horne R, Cozzolino M, Kalantar-Zadeh K. Balancing nutrition and serum phosphorus in maintenance dialysis. Am J Kidney Dis. 2014;64(1):143–50.PubMedCrossRef Fouque D, Horne R, Cozzolino M, Kalantar-Zadeh K. Balancing nutrition and serum phosphorus in maintenance dialysis. Am J Kidney Dis. 2014;64(1):143–50.PubMedCrossRef
38.
Zurück zum Zitat Lin TY, Lim PS, Hung SC. Impact of misclassification of obesity by body mass index on mortality in patients with CKD. Kidney Int Rep. 2018;3(2):447–55.PubMedCrossRef Lin TY, Lim PS, Hung SC. Impact of misclassification of obesity by body mass index on mortality in patients with CKD. Kidney Int Rep. 2018;3(2):447–55.PubMedCrossRef
39.
Zurück zum Zitat Noori N, Kopple JD, Kovesdy CP, Feroze U, Sim JJ, Murali SB, Luna A, Gomez M, Luna C, Bross R, et al. Mid-arm muscle circumference and quality of life and survival in maintenance hemodialysis patients. Clin J Am Soc Nephrol. 2010;5(12):2258–68.PubMedPubMedCentralCrossRef Noori N, Kopple JD, Kovesdy CP, Feroze U, Sim JJ, Murali SB, Luna A, Gomez M, Luna C, Bross R, et al. Mid-arm muscle circumference and quality of life and survival in maintenance hemodialysis patients. Clin J Am Soc Nephrol. 2010;5(12):2258–68.PubMedPubMedCentralCrossRef
40.
Zurück zum Zitat Stosovic M, Stanojevic M, Simic-Ogrizovic S, Jovanovic D, Djukanovic L. The predictive value of anthropometric parameters on mortality in haemodialysis patients. Nephrol Dial Transplant. 2011;26(4):1367–74.PubMedCrossRef Stosovic M, Stanojevic M, Simic-Ogrizovic S, Jovanovic D, Djukanovic L. The predictive value of anthropometric parameters on mortality in haemodialysis patients. Nephrol Dial Transplant. 2011;26(4):1367–74.PubMedCrossRef
41.
Zurück zum Zitat Su CT, Yabes J, Pike F, Weiner DE, Beddhu S, Burrowes JD, Rocco MV, Unruh ML. Changes in anthropometry and mortality in maintenance hemodialysis patients in the HEMO Study. Am J Kidney Dis. 2013;62(6):1141–50.PubMedCrossRef Su CT, Yabes J, Pike F, Weiner DE, Beddhu S, Burrowes JD, Rocco MV, Unruh ML. Changes in anthropometry and mortality in maintenance hemodialysis patients in the HEMO Study. Am J Kidney Dis. 2013;62(6):1141–50.PubMedCrossRef
42.
Zurück zum Zitat Hortegal EVF, Alves J, Santos EJF, Nunes LCR, Galvao JC, Nunes RF, Lula DA, Carvalho SCR, Franca A, Santos EMD, et al. Sarcopenia and inflammation in patients undergoing hemodialysis. Nutr Hosp. 2020;37(4):855–62.PubMed Hortegal EVF, Alves J, Santos EJF, Nunes LCR, Galvao JC, Nunes RF, Lula DA, Carvalho SCR, Franca A, Santos EMD, et al. Sarcopenia and inflammation in patients undergoing hemodialysis. Nutr Hosp. 2020;37(4):855–62.PubMed
43.
Zurück zum Zitat Lin YL, Liou HH, Wang CH, Lai YH, Kuo CH, Chen SY, Hsu BG. Impact of sarcopenia and its diagnostic criteria on hospitalization and mortality in chronic hemodialysis patients: a 3-year longitudinal study. J Formos Med Assoc. 2020;119(7):1219–29.PubMedCrossRef Lin YL, Liou HH, Wang CH, Lai YH, Kuo CH, Chen SY, Hsu BG. Impact of sarcopenia and its diagnostic criteria on hospitalization and mortality in chronic hemodialysis patients: a 3-year longitudinal study. J Formos Med Assoc. 2020;119(7):1219–29.PubMedCrossRef
44.
Zurück zum Zitat Carrero JJ, Johansen KL, Lindholm B, Stenvinkel P, Cuppari L, Avesani CM. Screening for muscle wasting and dysfunction in patients with chronic kidney disease. Kidney Int. 2016;90(1):53–66.PubMedCrossRef Carrero JJ, Johansen KL, Lindholm B, Stenvinkel P, Cuppari L, Avesani CM. Screening for muscle wasting and dysfunction in patients with chronic kidney disease. Kidney Int. 2016;90(1):53–66.PubMedCrossRef
45.
Zurück zum Zitat Crossland H, Skirrow S, Puthucheary ZA, Constantin-Teodosiu D, Greenhaff PL. The impact of immobilisation and inflammation on the regulation of muscle mass and insulin resistance: different routes to similar end-points. J Physiol. 2019;597(5):1259–70.PubMedCrossRef Crossland H, Skirrow S, Puthucheary ZA, Constantin-Teodosiu D, Greenhaff PL. The impact of immobilisation and inflammation on the regulation of muscle mass and insulin resistance: different routes to similar end-points. J Physiol. 2019;597(5):1259–70.PubMedCrossRef
46.
Zurück zum Zitat Bakaloudi DR, Siargkas A, Poulia KA, Dounousi E, Chourdakis M. the effect of exercise on nutritional status and body composition in hemodialysis: a systematic review. Nutrients. 2020;12(10):3071.PubMedCentralCrossRef Bakaloudi DR, Siargkas A, Poulia KA, Dounousi E, Chourdakis M. the effect of exercise on nutritional status and body composition in hemodialysis: a systematic review. Nutrients. 2020;12(10):3071.PubMedCentralCrossRef
47.
Zurück zum Zitat Tian N, Chen N, Li PK. Depression in dialysis. Curr Opin Nephrol Hypertens. 2021;30(6):600–12.PubMedCrossRef Tian N, Chen N, Li PK. Depression in dialysis. Curr Opin Nephrol Hypertens. 2021;30(6):600–12.PubMedCrossRef
48.
Zurück zum Zitat Purnell TS, Auguste P, Crews DC, Lamprea-Montealegre J, Olufade T, Greer R, Ephraim P, Sheu J, Kostecki D, Powe NR, et al. Comparison of life participation activities among adults treated by hemodialysis, peritoneal dialysis, and kidney transplantation: a systematic review. Am J Kidney Dis. 2013;62(5):953–73.PubMedCrossRef Purnell TS, Auguste P, Crews DC, Lamprea-Montealegre J, Olufade T, Greer R, Ephraim P, Sheu J, Kostecki D, Powe NR, et al. Comparison of life participation activities among adults treated by hemodialysis, peritoneal dialysis, and kidney transplantation: a systematic review. Am J Kidney Dis. 2013;62(5):953–73.PubMedCrossRef
49.
Zurück zum Zitat Li Z, Tong X, Ma Y, Bao T, Yue J. Prevalence of depression in patients with sarcopenia and correlation between the two diseases: systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(1):128–44.PubMedPubMedCentralCrossRef Li Z, Tong X, Ma Y, Bao T, Yue J. Prevalence of depression in patients with sarcopenia and correlation between the two diseases: systematic review and meta-analysis. J Cachexia Sarcopenia Muscle. 2022;13(1):128–44.PubMedPubMedCentralCrossRef
50.
Zurück zum Zitat Vettoretti S, Caldiroli L, Armelloni S, Ferrari C, Cesari M, Messa P. Sarcopenia is associated with malnutrition but not with systemic inflammation in older persons with advanced CKD. Nutrients. 2019;11(6):1378.PubMedCentralCrossRef Vettoretti S, Caldiroli L, Armelloni S, Ferrari C, Cesari M, Messa P. Sarcopenia is associated with malnutrition but not with systemic inflammation in older persons with advanced CKD. Nutrients. 2019;11(6):1378.PubMedCentralCrossRef
51.
Zurück zum Zitat Yuenyongchaiwat K, Jongritthiporn S, Somsamarn K, Sukkho O, Pairojkittrakul S, Traitanon O. Depression and low physical activity are related to sarcopenia in hemodialysis: a single-center study. PeerJ. 2021;9:e11695.PubMedPubMedCentralCrossRef Yuenyongchaiwat K, Jongritthiporn S, Somsamarn K, Sukkho O, Pairojkittrakul S, Traitanon O. Depression and low physical activity are related to sarcopenia in hemodialysis: a single-center study. PeerJ. 2021;9:e11695.PubMedPubMedCentralCrossRef
Metadaten
Titel
Development of a risk prediction nomogram for sarcopenia in hemodialysis patients
verfasst von
Genlian Cai
Jinping Ying
Mengyan Pan
Xiabing lang
Weiping Yu
Qinqin Zhang
Publikationsdatum
01.12.2022
Verlag
BioMed Central
Erschienen in
BMC Nephrology / Ausgabe 1/2022
Elektronische ISSN: 1471-2369
DOI
https://doi.org/10.1186/s12882-022-02942-0

Weitere Artikel der Ausgabe 1/2022

BMC Nephrology 1/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.