Next Article in Journal
Association of Plasma Total Cysteine and Anthropometric Status in 6–30 Months Old Indian Children
Previous Article in Journal
Carbohydrate-Induced Insulin Signaling Activates Focal Adhesion Kinase: A Nutrient and Mechanotransduction Crossroads
Previous Article in Special Issue
HD-FFQ to Detect Nutrient Deficiencies and Toxicities for a Multiethnic Asian Dialysis Population
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Understanding Development of Malnutrition in Hemodialysis Patients: A Narrative Review

by
Sharmela Sahathevan
1,
Ban-Hock Khor
2,
Hi-Ming Ng
3,
Abdul Halim Abdul Gafor
2,
Zulfitri Azuan Mat Daud
4,
Denise Mafra
5 and
Tilakavati Karupaiah
6,*
1
Dietetics Program, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
2
Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaakob Latif, Bandar Tun Razak, Cheras, Kuala Lumpur 56000, Malaysia
3
School of Medicine, Faculty of Health & Medical Sciences, Taylor’s University Lakeside Campus, No 1, Jalan Taylors, Subang Jaya 47500, Malaysia
4
Department of Dietetics, Faculty of Medicine & Health Sciences, Universiti Putra Malaysia, UPM Serdang 43400, Malaysia
5
Post Graduation Program in Medical Sciences and Post-Graduation Program in Cardiovascular Sciences, (UFF), Federal Fluminense University Niterói-Rio de Janeiro (RJ), Niterói-RJ 24033-900, Brazil
6
School of BioSciences, Faculty of Health & Medical Sciences, Taylor’s University Lakeside Campus, No 1, Jalan Taylors, Subang Jaya 47500, Malaysia
*
Author to whom correspondence should be addressed.
Nutrients 2020, 12(10), 3147; https://doi.org/10.3390/nu12103147
Submission received: 23 September 2020 / Revised: 11 October 2020 / Accepted: 13 October 2020 / Published: 15 October 2020

Abstract

:
Hemodialysis (HD) majorly represents the global treatment option for patients with chronic kidney disease stage 5, and, despite advances in dialysis technology, these patients face a high risk of morbidity and mortality from malnutrition. We aimed to provide a novel view that malnutrition susceptibility in the global HD community is either or both of iatrogenic and of non-iatrogenic origins. This categorization of malnutrition origin clearly describes the role of each factor in contributing to malnutrition. Low dialysis adequacy resulting in uremia and metabolic acidosis and dialysis membranes and techniques, which incur greater amino-acid losses, are identified modifiable iatrogenic factors of malnutrition. Dietary inadequacy as per suboptimal energy and protein intakes due to poor appetite status, low diet quality, high diet monotony index, and/or psychosocial and financial barriers are modifiable non-iatrogenic factors implicated in malnutrition in these patients. These factors should be included in a comprehensive nutritional assessment for malnutrition risk. Leveraging the point of origin of malnutrition in dialysis patients is crucial for healthcare practitioners to enable personalized patient care, as well as determine country-specific malnutrition treatment strategies.

1. Introduction

The last three decades witnessed considerable growth in the global burden of chronic kidney disease (CKD), accounted for by 77.5% of end-stage kidney disease (ESKD) patients on kidney replacement therapy (KRT), with 43.1% alone provided by dialysis [1]. Hemodialysis (HD) forms 89% of the global treatment for ESKD patients [2]. The technological delivery of HD treatment to patients today is considered optimal as per medical guidelines for practice with regard to biocompatibility of dialyzer membranes, dialysis dose, frequency of dialyzer reuse, and duration of dialysis [3]. A significant problem faced by this patient group, however, is malnutrition with a global prevalence of 28–54% [4], facing a greater risk of mortality, varying from a likelihood of 1.61 to 4.08 [5,6].
Morbidity arising from malnutrition in these patients severely affects quality of life (QoL) [7], frailty, and increased risk of infections and mortality [8]. Malnutrition in patients on dialysis develops along different pathways from that observed in acute cases of hospitalization and critical illness. Its inception evolves from the early progressive nature of CKD itself [9], the implementation of a low protein diet to limit CKD progress [10], and the prolonged period of potentially lifesaving dialysis treatment for patients reaching ESKD [11]. In this context, the HD treatment itself in terms of dialysis-induced nutrient losses, multiple dialyzer reuse, dialysis-induced inflammation, efficacy of uremia and metabolic acidosis correction, and dialysis adequacy, frequency, and duration are inevitable iatrogenic factors contributing to malnutrition. Concurrently, prevailing non-iatrogenic factors such as suboptimal dietary intakes, taste alterations, poor appetite, insulin resistance, and psychosocial factors are also incriminated in the etiology of malnutrition.
Malnutrition occurrence in the dialysis population has generated much research. However, different definition terms exist for malnutrition such as protein-energy wasting, protein-energy malnutrition, malnutrition–inflammation complex syndrome, malnutrition–inflammation–atherosclerosis and uremic wasting syndrome depending on involvement of inflammation, hypercatabolism, and increased uremia [12]. Multiple factors are cited within the etiology of these descriptive malnutrition terms, and implication of some but not all these factors differentially indicates that there is no uniformity in the diagnosis of malnutrition [12]. Comorbidities such as heart failure (left-ventricular failure) and CKD-mineral bone disorder have a bidirectional association with nutritional status [13,14]. The related mechanisms include malabsorption due to gut edema, poor appetite due to cytokine production, and difficulty in oral intake and food preparation arising from fatigue and breathing difficulty [15]. However, malnutrition is acknowledged to be a kidney-specific risk factor for heart failure in CKD patients [14], and, with HD patients, there is lack of association between malnutrition assessment and echocardiographic findings [16] or cardiovascular disease (CVD) risk [17].
In essence, factors contributing to the development of malnutrition may be categorized as of iatrogenic and non-iatrogenic origin (Figure 1). Iatrogenic factors are an inadvertent consequence of dialysis for ESKD patients, whereas non-iatrogenic factors develop spontaneously from contributive factors accompanying the progression of CKD but not related to the primary treatment.
This review aims to provide a global view of the iatrogenic and non-iatrogenic causes of malnutrition, irrespective of its form described in current literature, as some aspects with regard to this topic have changed over recent years. Our approach discusses mechanisms elucidating the implication of each factor contributing to malnutrition. The origin of malnutrition contributed by both iatrogenic and non-iatrogenic factors is important to understand the implications for healthcare practitioners in performing the assessment and treatment for malnutrition. Before this categorization, malnutrition must also be delineated according to its development history. This review is, therefore, organized into (i) development of malnutrition in HD, (ii) iatrogenic factors of malnutrition, and (iii) non-iatrogenic factors of malnutrition.

2. Development of Malnutrition at the Time of HD Initiation and Indicators of Poor Nutritional Status

The decision to start dialysis for an ESKD patient varies across countries and is influenced by the local nephrology practice, healthcare policies, and cost for dialysis treatment [18]. The Dialysis Outcomes and Practice Patterns Study (DOPPS) Phase 2 with 12 participating countries indicated a greater mortality rate in patients new to dialysis compared to prevalent dialysis patients [19]. Early mortality at the time of dialysis initiation prevails with increased risk up to 80% within the first two months of HD initiation [20]. Apart from catheter vascular access [20] and pre-dialysis care [21], nutritional status is considered a potentially modifiable risk factor in early mortality [22]. Clearly, pre-existing malnutrition originates from progressive CKD stages 3 to 5 with vulnerability of the patient starting from the point of metabolic derangements associated with falling glomerular filtration rate, late nephrology access, and insufficient pre-dialysis dietetic care during this period [22,23].
Earlier opinion on dialysis initiation did consider poor nutritional status as a factor to initiate dialysis. However, this was not based on markers of malnutrition but rather signs and symptoms of malnutrition such as anorexia, nausea, and fatigue [24]. Van de Luijtgaarden et al. (2012) reported 53% of nephrologists agreeing to initiate dialysis in patients with poor nutritional status [25]. However, a review on dialysis initiation observed a lack of data on the benefits of early dialysis initiation in patients with low serum albumin level or in improving nutritional status [26]. The Canadian Society of Nephrology Guidelines (2014) [27] ceased to recommend dialysis initiation on the basis of a decline in nutritional status as indicated by serum albumin, lean body mass, or subjective global assessment (SGA), whereas the Caring for Australians with Renal Impairment Guidelines [28] recommend dialysis with glomerular filtration rate (GFR) < 10 mL/min per 1.73 m2 to reduce uremic symptoms or signs of malnutrition.
Dialysis treatment is expected to improve nutritional status for patients with a more liberal protein prescription compared to the pre-dialysis stage [29,30,31]. However, dialysis treatment is cited to contribute to malnutrition burden [8], and newly dialyzing patients are at risk of the early mortality attributed to malnutrition as evidenced by diagnostic assessment of nutrition risk screening using SGA [32], low body mass index (BMI), low mid-arm muscle circumference (MAMC) [22,33,34], low albumin [20], low cholesterol levels [32], and reduced food intake [22,33,34,35], as shown in Table 1.

3. Iatrogenic Factors of Malnutrition

ESKD patients with pre-existing malnutrition on maintenance dialysis become additionally vulnerable over time to the catabolic effects of the dialysis treatment, which predispose the patient to greater mortality and morbidity in long-term dialysis. The concern is that the presence of poor nutritional status in dialysis patients predicts increased mortality risk.
Kwon et al. (2016) prospectively used SGA to monitor patients, observing that those in SGA B and C categories at baseline almost tripled their risk of mortality by 12 months [32]. HD patients experiencing declines in BMI and serum albumin levels over a 6 month follow-up also had increased mortality risk [36]. Table 2 summarizes studies reporting various indicators of poor nutritional status as strong predictors of mortality in maintenance HD patients.
Iatrogenic malnutrition or “physician-induced malnutrition” is the development of malnutrition arising from medical procedures, pharmacological treatment, prolonged hospitalization, nosocomial infections, or delayed wound healing [40]. Similarly, aspects of the dialysis procedure contribute to malnutrition, which is unavoidable as it occurs as part of the treatment [8]. The iatrogenic aspects of dialysis procedure are detailed in the sections below.

3.1. Dialysis-Induced Nutrient Losses

The dialysis process is instrumental to chronic nutrient losses, particularly protein and amino acids. About 6–12 g of amino acids and 7–8 g of protein losses occurring during each dialysis session [41,42,43,44,45] may contribute to hypoalbuminemia, a strong predictor of malnutrition and mortality [11,33,36]. Optimal dietary protein intake (DPI) may replenish low plasma amino acids. However, DPI inadequacy is a common issue in HD patients, affecting 32–81% of HD populations globally [31]. Suboptimal DPI associated with dialysis-induced amino-acid losses [46] promotes protein catabolism through increased whole-body and muscle protein proteolysis [46,47].
Nutrient losses via dialysis depend on the mechanism of solute removal and the pore size of the dialyzer membrane, which determines solute removal [48]. However, increasing the pore size of dialyzer membranes to enable greater removal of middle molecules also increases involuntary albumin losses, estimated between 2 and 14 g depending on the degree of membrane permeability [49]. As such bioincompatible membranes [45], high flux membrane, hemofiltration (HF) and hemodiafiltration (HDF) techniques [46,50], or multiple dialyzer reuse practice [43] induce greater membrane permeability and facilitate greater losses of amino acids into the dialysate [50]. Table 3 shows the degree of protein and amino-acid losses associated with membranes characteristics.
Dialysis performed during the 1960s used low-flux membranes [55], which efficiently removed uremic solutes with low molecular weight (<0.5 kDa) but not the middle molecules of 0.5–60 kDa size [56,57]; with advanced technology, greater removal of larger uremic solutes using high-flux membranes and, now, membranes with medium (MCO) and high cutoffs (HCO) are available [53], combining larger pore sizes with improved HF and HDF techniques [41,58].
With highly permeable membranes or HF and HDF techniques, patients are reported to achieve better intradialytic and hemodynamic stability [59], alongside improvements in nutritional status as evidenced by gains in BMI, dry weight, and appetite [60]. Improvements are attributed to greater removal of middle molecules using HDF. However, these membranes and/or techniques, in addition to increasing the cost burden [57,59,60,61,62], induce greater albumin losses of 3.5 to 9.0 g per HD session [63,64], along with involuntary removal of vitamins, larger protein molecules, and lipids [62].
Contrarily, two studies reported a significant reduction in serum albumin levels in patients dialyzing with HCO membranes [63,65]. The risk–benefit balance by using dialyzer membranes with greater permeability for improved uremic solute removal versus greater albumin losses remains unknown [41]. Similarly, advanced techniques using either HF or HDF may also pose long-term risk of malnutrition development in HD patients. Given that the permissible threshold for tolerance of albumin losses with highly permeable membrane remains unclear, long-term use of MCO [66] or HCO membranes with HDF [63] techniques may pose malnutrition risk.

3.2. Multiple Dialyzer Reuse

In low-to-middle-income countries, the practice of dialyzer reuse is common [67,68]. However, multiple dialyzer reuse may contribute to negative outcomes [69] such as infection risks, biochemical and immunologic reactions, improper sterilization, increased membrane permeability [49], and loss of performance leading to inadequate dialysis adequacy. These issues are believed to arise from the reprocessing procedure involving sanitizing agents [67,70]. However, two studies have indicated that single, minimal (>6 times), or multiple dialyzer reuse carries no impact on dialysis adequacy, body weight, and serum albumin level [71,72].

3.3. Dialysis-Induced Inflammation

Many factors lead to inflammation in HD patients such as biocompatibility of the dialyzer membrane [73,74,75], infection related to dialysis access [73], and impure dialysate containing cytokine-inducing substances labeled as endotoxins [73,76].
Whether dialysis access directly contributes to malnutrition has not been shown. Arteriovenous fistula (AVF) failures were not influenced by markers of poor nutritional status except for high cholesterol (p = 0.034) and low normalized protein catabolic rate (nPCR) levels (p = 0.029) [77]. HD patients with catheter access compared to fistula and graft had significantly higher malnutrition–inflammation score (MIS) and lower serum albumin levels [78]. In fact, patients with AVF have 52% greater survival rate compared to those on central venous catheter (CVC) irrespective of nutritional status, although malnutrition was found to lower survival rate by 2% [79]. Instead, catheter rather than graft and fistula access appears to be a significant predictor of greater inflammatory response, and it is associated with the highest all-cause mortality rate [78] mediated by infection [20]. Therefore, the route of dialysis access is rather associated with inflammation and mortality risk [78], whereby presence of malnutrition may influence the survival rate [79].
Direct effects of the membrane, the extent of complement stimulation induced by the membrane, and the degree of eosinophilia associated with the clearance of cytokines determine the magnitude of the inflammatory response during dialysis [74]. Inflammatory marker levels may be modulated by different types of dialyzer membranes (Table 4). Generally, the high-flux dialyzer membrane and HDF technique are associated with lower inflammation grade in HD patients when compared to the low-flux dialyzer membrane. These differences are attributed to processing technology for structuring and composition of the membrane, conferring attributes to the dialyzer in terms of biocompatibility, water permeability, clearance, and appropriate sieving coefficients for myoglobin or albumin [59].
Inflammation also occurs with dialysate contamination by microorganisms, which produce endotoxins that pass through the dialyzer membrane and enter into blood circulation [83], amplifying the production of proinflammatory cytokines [84] such as interleukin (IL)-1, IL-6 [85], and tumor necrosis factor (TNF)-α [86]. Infected or old clotted grafts may also contribute to inflammation [87]. Of note, these middle molecules such as IL and TNF-α are not effectively removed by dialysis treatment with low-flux membrane [88] and are accumulated.
Overall, dialysis patients are vulnerable to oxidative stress with a marked increase in reactive oxygen species (ROS) production and antioxidant depletion. ROS induces activation of nuclear factor kappa B (NF-κB), which is translocated to the cell nucleus stimulating cytokine production, in turn causing inflammation [89]. Indeed, HD patients have pronounced NF-κB gene expression compared to a healthy population [90].
Another impact of the HD treatment is the activation of polymorphonuclear white blood cells, which trigger production of ROS and other pro-oxidants [91]. Indeed, increased indices of oxidative damage along with decreased indices of antioxidant defense have been observed in HD patients post-dialysis [92]. Low antioxidant levels in HD patients may also occur from limited vegetable and fruit intakes preventing hyperkalemia [93]. Resultant low intakes of vitamin A, C, and E and selenium would affect antioxidant defense mechanisms [93,94]. Additionally, involuntary removal of vitamins also occurs with every HD session as mentioned in Section 3.1.
However, Silva et al. (2019) found no difference in markers of oxidative stress and antioxidant defenses in malnourished HD patients identified using global objective assessment compared to mild or well-nourished patients [95]. In contrast, accumulation of advanced glycation end products, a biomarker of oxidative stress measured using skin auto fluorescence, was significantly associated with markers of malnutrition in HD patients such as lower serum albumin, lower handgrip strength, and lower protein intake [96]. HD patients with protein-energy wasting were 5.2 times more likely to experience oxidative stress as demonstrated by high protein carbonyl levels (95% confidence interval (CI): 24.0–1.1, p = 0.039) [97].
When malnutrition coexists with inflammation in dialysis patients, the combination of both conditions is known as malnutrition–inflammation complex syndrome [98]. The inflammation results in a reduction in albumin production in the liver [99] and fosters poor appetite, a non-iatrogenic factor implicated in malnutrition [100]. The strong relationship between malnutrition and inflammation in dialysis patients is evident, as indicated by significant association (r = 0.65, p = 0.040) of malnourished HD patients categorized by SGA B and C with high C-reactive protein (CRP) levels [101].

3.4. Efficacy of Uremia Correction

A major aim of dialysis therapy is to remove uremic waste products [57,102]. However, dialysis only reduces uremic burden through partial removal of uremic solutes [103]. Incomplete clearance of uremic solutes and the generation of urea from both dialysis-induced tissue degradation and dietary proteins contribute to uremic solute accumulation [104]. Excessive uremic solute burden influences amino-acid and protein metabolism by inhibiting transamination activities of enzymes such as threonine dehydratase and alanine and aspartate transferases [104], impairing membrane transport [105], inhibiting protein binding [106], and promoting muscle wasting [104].
The removal of uremic solutes depends on dialyzer membrane permeability. As mentioned earlier (see Section 3.1), uremic solutes with low molecular weight such as urea and creatinine are efficiently dialyzed via a low-flux dialyzer membrane [88], whereas membranes with higher permeability allow for greater clearance of small and large middle molecules. However, the threshold for clearance depends on uremic gains on non-dialysis days and total clearance achieved from the previous HD session [104,107]. Both these factors would determine the severity of uremia in HD patients. Aside from the permeability of the membrane, total clearance of uremic solutes is also determined by dialysis adequacy, dialysis frequency, and duration of dialysis session [108].

3.5. Dialysis Adequacy

Uremic solute clearance depends on dialysis adequacy, which refers to the frequency and duration of dialysis [102]. Expert guidelines for optimal uremic solute removal favor a three- to five-hourly dialysis session provided three times weekly [3] in order to meet dialysis adequacy by achieving a Kt/Vurea of 1.2, which designates the dialyzer urea clearance (K), time on dialysis (t), and total body water (V) [57,102].
However, the hemodialysis (HEMO) study in a 3 year follow-up of 1846 dialyzing patients established that neither high (Kt/V = 1.65) nor low (Kt/V = 1.25) dialysis dose significantly affected markers of nutritional status such as serum albumin, post-dialysis weight, dietary energy and protein intakes, calf and upper arm circumference, and appetite status (all p > 0.05) [109]. The only effect was on normalized protein catabolic rate (nPCR), a surrogate marker for DPI with greater decline in the low compared to high dialysis dose group (p = 0.007).
Of note, the calculation of Kt/Vurea is based on urea, a surrogate marker for clearance of small solutes. It does not represent removal of the more detrimental larger uremic solutes [107,110]. Inefficient removal of uremic toxins via dialysis is suggested to induce taste alteration in HD patients, which contributes to malnutrition [111,112].

3.6. Dialysis Frequency

Increasing the thrice-weekly frequency of dialysis sessions to >4 times may support better management of fluid removal [113] and lower systolic blood pressure, as well as improve QoL [23]. Alternately, a shorter but increased frequency of dialysis provided by six HD sessions two-hourly per week may benefit toward greater removal of uremic solutes [56], as shown in patients with lower trends in pre-dialysis serum levels of creatinine, urea, uric acid, and protein-bound solutes such as indole-3-acetic acid and indoxyl sulfate. However, serum albumin levels and post-dialysis weight did not improve for these patients [56]. In contrast, Rashidi et al. (2011) observed improved weight, BMI, and serum albumin status along with decreased serum CRP in patients converting to four HD sessions four-hourly per week from the standard dialysis regime by six weeks [114]. Dietary intake also improved albeit non-significantly. This effect may perhaps be explained by improved appetite occurring with greater removal of uremic compounds through more frequent dialysis [115].
In some countries, weekly frequency of dialysis may depend on the patient’s access to financial support. For example, dialysis frequency in low-income countries such as India and Pakistan may be offered as two sessions four-hourly per week [116,117] compared to the standard dialysis of three sessions four-hourly per week in developing countries [116,118]. Treatment affordability, poor access to nephrology care and dialysis centers [116], and inadequately equipped dialysis facilities [117] are reasons for lower dialysis frequency. Interestingly, Chauhan and Mendonca (2015) showed that, for 50 Indian HD patients undergoing dialysis twice a week, those achieving dialysis adequacy of Kt/V ≥ 2.0 significantly improved their serum albumin and hemoglobin levels [118].

3.7. Dialysis Duration

Increasing the dialysis duration results in greater removal of small and large uremic solutes compared to the standard HD regime [102,119]. Patients dialyzing for 8 h using a high-flux dialyzer membrane have shown greater total solute removal, dialyzer extraction ratios, and total cleared volumes for urea, creatinine, phosphorus, and β2-microglobulin compared to patients on standard dialysis, and this occurs without affecting dialysis adequacy [102]. Lower post-dialysis levels of the uremic toxin indoxyl sulfate have been observed in patients dialyzing for 8 h compared to the standard 4 h regime (17.2 ± 3.6 vs. 27.5 ± 3.2 g/mL, p = 0.049), despite both patient groups having similar pre-dialysis levels [120].
Nutritional marker improvements through higher serum albumin and hemoglobin levels and lower white blood cell count appear to be associated with longer dialysis duration, as indicated from combined data of the three DOPPS [121]. These improvements may be explained by greater removal of both small and large solutes with longer hours of dialysis [102].

3.8. Efficacy of Metabolic Acidosis Correction

Metabolic acidosis develops in the early stages of CKD from the kidney’s inability to excrete nonvolatile acids and synthesize bicarbonate to maintain acid–base balance [122]. HD treatment aims to correct metabolic acidosis via bicarbonate concentration of the dialysate [123], ultrafiltration rate [124], dialyzer membrane surface area and permeability [125], blood and dialysis flow rate [124], transmembrane concentration gradient set by the patient’s serum bicarbonate level and bicarbonate availability from the dialysate [125], and dialysis adequacy [122,123] through maintaining the pre-dialysis serum bicarbonate levels between 24 and 26 mmol/L as recommended by current opinion [126]. However, metabolic acidosis correction depends on patient-related determinants such as interdialytic weight gain [123], acid generation from high protein intake [122,127], or gastrointestinal losses of bicarbonate [122,125]. Individual fluctuation in patients’ bicarbonate levels challenges optimum management [122].
Metabolic acidosis contributes to malnutrition by reducing protein synthesis and increasing muscle degradation [123]. The malnutrition pathway in HD patients involves protein catabolism, secondary insulin resistance, inflammation, and increased serum leptin levels [122]. Lines of evidence using animal and human studies explain that increased muscle breakdown occurs during metabolic acidosis via two mechanisms. These involve increased activation of branched-chain ketoacid dehydrogenase (BCKAD) and the ATP-dependent ubiquitin–proteasome system (UPS) pathway [128]. Importantly, acidosis stimulates increased gene transcription and activity of BCKAD enzyme to degrade the branched-chain amino acids (BCAA), namely, leucine, isoleucine, and valine. BCAAs are important precursors for protein synthesis and are mainly metabolized in the muscle [50]. Increased BCAA oxidation, therefore, is the basis for a higher protein requirement for HD patients [129]. However, metabolic acidosis concomitant with dietary insufficiency and uremia further exacerbates protein catabolism in dialysis patients [130]. Metabolic acidosis activates UPS by increasing gene transcription of the proteasome and ATP-dependent ubiquitin, components involved in the muscle protein degradation pathway [128]. This chain leads to increased caspase-3 activity which promotes cleaving of muscle fibers, resulting in poor muscle mass [128].
Additionally, the acidic environment affects insulin binding to receptors, thus reducing tissue sensitivity to insulin and affecting glucose uptake [131]. Separately, metabolic acidosis also inhibits the anabolic effect of insulin, causing muscle depletion in dialysis patients [122]. Moreover, cell culture studies have shown that TNF-α and interleukins are generated in an acidic environment, triggering an inflammatory response [132,133].
The impact of metabolic acidosis on nutritional status of HD patients by assessment of serum bicarbonate levels may present anomalies in interpretation. In one study, patients with serum bicarbonate levels ≤22 mmol/L rather than serum bicarbonate levels >22 mmol/L had lower serum albumin levels (p = 0.046) [134]. In these patients, high serum bicarbonate levels correlated negatively with nPCR (r = −0.492, p = 0.045) but positively with serum albumin (r = 0.432, p = 0.019). Acidosis-led catabolism triggers breakdown of the endogenous proteins, which influence higher nPCR levels. In different malnourished HD populations, serum bicarbonate levels of >23 [135,136] or >27 mmol/L [127,136] have been associated with greater mortality risk. As malnutrition is a confounding factor for serum bicarbonate level, there is no ideal serum bicarbonate level that fits all dialysis patients [137].

4. Non-Iatrogenic Causes of Malnutrition

Comorbid non-iatrogenic factors may also contribute to malnutrition development in dialysis patients. These non-iatrogenic factors are elaborated on in the sections below.

4.1. Suboptimal Dietary Intake

Suboptimal dietary intake is a primary contributing factor to malnutrition [12] and is associated with increased mortality in HD patients [31,138]. Adult recommendations for dietary energy intake (DEI) and DPI to achieve nutrient adequacy have been proposed for HD patients by several expert groups, and these generally fall within 25–35 kcal/kg ideal body weight (IBW)/day for DEI and 1.0–1.2 g protein/kg IBW/day for DPI [126,139,140,141]. Requirements factor in criteria to maintain physiological balance, prevent deficiencies from dialysis-induced nutrient losses, and reduce risk of malnutrition and mortality [126].
However, achieving DEI and DPI adequacies remains a challenge for HD patients with intakes falling below recommendations as indicated by many studies (Table 5). This is evidenced by 70–90% of global HD populations reported with DEI inadequacy, whereas DPI inadequacy ranges between 30% and 80%.
Suboptimal DEI is of greater concern than DPI inadequacy, as gluconeogenesis is implied. Three studies reported HD patients achieving DPI adequacy >1.2g/kg/BW but failing to meet DEI adequacy [146,151,155]. Insufficient DEI, despite DPI adequacy, predisposes patients to negative nitrogen balance, resulting in both dietary protein and muscle protein to be diverted to fuel body energy requirements [154]. Additionally, amino-acid losses occurring through the dialysis procedure (see Section 3.1) affect protein synthesis, triggering muscle proteolysis to generate amino acids if there is low DPI [158]. Of concern, suboptimal dietary intake bears a negative impact on the survival rate of HD patients as indicated by some studies reporting patients with poor DEI and DPI (Table 6). Kang et al. (2017) found that HD patients with DEI < 25 kcal/kg BW/day and DPI < 0.8g/kg BW/day had 86% and 35% increased risk of mortality, respectively [138]. Similarly, those with DPI < 1.2g/kg BW/day had a 4.98-fold greater risk of mortality [159]. A recent metabolomics study reported higher concentrations of 3-hydroxybutyrate and tartrate along with low creatinine appearing in patients with protein energy wasting [160]. These metabolites are linked to gluconeogenesis and may be conditional to suboptimal DEI and DPI intakes [161].
The background of dietary inadequacy observed in HD patients may be attributed to monotonous dietary patterns [142,154,162], poor diet quality [154], anorexia [154], and alterations in taste [163]. A monotonous diet defines a dietary pattern with minimal variety of food groups [142,154]. Zimmerer et al. (2003) showed that HD patients with the highest Diet Monotony Index (DMI) had the lowest DEI (21 kcal/kg/day) and DPI (0.83 g/kg/day) [162]. Furthermore, a 5 point increase in DMI was associated with decreases in both energy and protein intakes by 10 kcal/kg BW/day (p = 0.004) and 0.43 g/kg BW/day (p = 0.006), respectively. Importantly, Sualaheen et al. (2020) investigating habitual dietary patterns of Malaysian HD patients showed that the highest vs. lowest tertiles of a balanced and varied dietary pattern were associated with lower DMI (T3 = 29.0 ± 1.1 vs. T1 = 33.0 ± 1.0, p = 0.030) and lower malnutrition risk identified via malnutrition–inflammation score (MIS) assessment (T3 = 4.9 ± 0.36 vs. T1 = 6.4 ± 0.34, p = 0.010) [142].
Suboptimal DEI from reduced food intake also affects patient adequacy for other essential nutrients, as the overall diet quality falls. Kim et al. (2015) reported that an insufficient DEI of 21.90 ± 6.70 kcal/kg BW affected adequacy for micronutrients such as vitamin A and C, thiamin, riboflavin, niacin, folate, calcium, phosphorus, and zinc, as well as dietary fiber [154].

4.2. Taste Alterations

Low palatability of diets is underscored by taste alterations experienced by HD patients, and this factor reportedly affects 31–44% of HD populations [165]. Lynch et al. (2013) found 34.6% of 1745 HD patients in the HEMO study self-reporting taste alteration [165]. Such patients compared to those reporting “no taste alterations” clearly signified poor nutritional status as indicated by lower dry weight, serum albumin, serum creatinine, nPCR, and DPI. Reported DEI values for both groups were similarly low (22.8 ± 9.8 vs. 23.1 ± 9.4 kcal/kg/day, p = 0.260) highlighting that energy inadequacy evidently prevailed for all patients. This study also reported a 71% increased risk of mortality (odds ratio (OR) = 1.71, 95% CI: 1.01–1.37) in patients with altered taste perception.
Taste alterations experienced by HD patients may be explained by food aversion learning [166]. Aversions toward protein-rich foods such as meat have been significantly associated with enhanced metallic taste in patients also reporting poor appetite [167]. Clearly, taste alterations in HD patients develop food aversion learning, which impacts appetite and reduces overall diet quality, thus contributing to malnutrition. The reduction in taste perception may also be related to zinc deficiency [168].

4.3. Poor Appetite

HD patients reporting poor appetite experience significantly higher frequency of hospital admissions, longer duration of hospitalization, poor QoL, and nutritional outcomes such as lower normalized protein nitrogen appearance levels and high inflammatory marker levels than those reporting good appetite [163,165]. The immediate impact of poor appetite is reduced dietary adequacy and increased risk of malnutrition. This was shown in Malaysian HD patients where poor appetite compared to good appetite was significantly linked to lower DEI (14.34 vs. 23.12 kcal/kg/IBW/day, p = 0.049), DPI (0.45 vs. 0.94 g/kg IBW/day, p = 0.010) but higher MIS scores (9.5 vs. 6.6, p = 0.039) indicative of malnutrition [148]. Of concern, patients with diminished appetite faced 4.74 times greater risk of mortality [163].
The mechanism for poor appetite may be explained by changes in appetite hormones in HD patients. Ghrelin, an orexigenic hormone mainly secreted by the stomach, regulates appetite by stimulating spontaneous food intake [29,169]. Ghrelin present in its active form as des-acyl ghrelin has an anorexigenic effect, whereas acyl ghrelin as ghrelin in its inactive form is the main orexigenic molecule [169]. Together, these studies suggest that des-acyl ghrelin may have a negative effect on appetite, whilst high acyl ghrelin levels associated with adiposity indicate better nutritional status [29,169]. Moreover, an association among ghrelin, inflammation, and nutritional status has been reported [170].
Leptin, an adipokine, has an inhibitory effect on appetite in normal metabolism [171]. However, leptin’s role in regulating appetite in CKD is controversial. Hypoleptinemia has been associated with malnutrition in HD populations although its mechanistic involvement in causing poor nutritional status is unknown [172,173]. Low serum leptin levels were independently associated with high MIS status observed in 100 Taiwanese HD patients [172] and 65 Turkish HD patients [174]. However, these studies could not show any association of low leptin levels with inflammatory markers. It may be implied that low leptin levels may serve as a marker of poor nutritional status, whereas higher levels may indicate leptin resistance [175], as there is an attenuation of appetite suppression [172]. Iikuni et al. (2008) proposed leptin as the link between energy homeostasis and inflammation in normal metabolism, where greater leptin secretion occurs with greater adiposity, which in turn promotes production of inflammatory cytokines [176]. Applying this hypothesis to the CKD population, it may be inferred that malnourished HD patients with low BMI will have less leptin secreted by adipocytes, whereas the reverse may occur with better nutritional status and higher BMI.
Studies reporting the impact of poor appetite on malnutrition as indicated by various nutritional outcomes in HD patients are summarized in Table 7.

4.4. Insulin Resistance

Insulin resistance is implicated in the etiology of malnutrition in HD patients. Insulin at physiological levels bears both catabolic and anabolic effects on skeletal muscle. Insulin’s anabolic role is to promote BCAA transport and regulate protein synthesis in the muscle [180]. Another anabolic role of insulin is facilitating glucose transport and uptake by muscle tissues [181]. Reduced insulin secretion by the pancreatic β-cells or impaired tissue sensitivity to insulin at receptor and post-receptor levels in the heart, liver, or muscle are two pathways of insulin insufficiency [182,183]. More commonly, “uremic insulin resistance” through inflammatory pathways may occur from insufficient removal of dialyzable uremic solutes [180]. Of relevance, insulin resistance at receptor levels are traced to defects in the insulin receptor signaling pathway arising from metabolic derangements accompanying kidney disease such as uremia, metabolic acidosis, anemia, and inflammation [8,184].
Insulin resistance is associated with peripheral resistance of glucose uptake at the skeletal muscle site and manifests as impaired insulin signaling through the phosphorylation of insulin receptor substrate-1, which inhibits tyrosine kinase activity at the insulin receptor [181,183]. Another pathway of insulin resistance and impaired glucose metabolism may be suggested by high circulating retinol binding protein 4 (RBP4) [185]. Animal studies have demonstrated that RBP4′s inverse relationship with glucose transporter type 4 (GLUT4), which is insulin-dependent, induces glucose uptake in the fat and muscle [186,187]. High RBP4 has a role in glucose metabolism by inducing gluconeogenesis and inhibiting glucose uptake in the muscle via feedback suppression of adipose tissue expression, followed by reduced GLUT4 expression, which affects glucose uptake [185].
Reduced insulin sensitivity affects BCAA transport, blunting the anabolic effect of insulin for decreasing skeletal muscle breakdown [180]. Depletion in BCAA due to amino-acid losses via dialysis, along with suboptimal dietary intake lead to increased proteolysis to supply amino acids needed for protein synthesis [188]. Therefore, insulin resistance promotes muscle proteolysis, and this association is evident in HD patients with studies reporting a positive correlation between insulin resistance and muscle loss [189,190].
It is, thus, clear that, in ESKD patients, apart from chronic suboptimal food intake, gluconeogenesis may also be driven by insulin resistance associated with inflammation, uremia, and metabolic acidosis. Gluconeogenesis is a normal adaptive catabolic process to produce energy. Protein sparing under conditions of energy sufficiency occurs as the primary protein function for tissue synthesis and repair. With dietary energy insufficiency, amino acids and proteins derived from dietary protein or breakdown of skeletal muscle during starvation become new substrates for energy [158]. As HD patients are known to have suboptimal food intake, increased gluconeogenesis in these patients stimulates muscle proteolysis, leading to greater risk of malnutrition.

4.5. Psychosocial Factors

Psychosocial factors may negatively impact physical and emotional status, QoL, and nutritional status in HD patients [149,191,192], as shown in Table 8.

4.5.1. Depression

Depression is reported to be prevalent in 6% to 84% of HD patients [204] and arises from loss of the provider role within a family, unemployment, lack of social support, reduced mobility, physical strength, cognitive ability, and sexual function [204]. Additional factors are anxiety and stress from the burden of kidney failure followed by fluid and dietary restrictions, which are significantly associated with poor QoL in these patients [204,205].
Several studies have reported associations between depression and markers of malnutrition such as poor anthropometry measures, as well as low serum albumin, creatinine, hemoglobin, and nPCR levels with increased inflammation [191,192,193]. Separately, malnourished HD patients as indicated by MIS ≥ 6 faced 52% increased mortality risk (hazard ratio (HR) = 1.52, 95% CI: 1.13–2.05), along with higher depression symptoms and poorer QoL [194]. These findings suggest that depression should be considered as an independent risk factor for malnutrition. Indeed, malnutrition reversal in HD patients by antidepressant treatment has been observed with significant improvement in nPCR, serum albumin, and pre-dialysis blood urea nitrogen levels, along with a significant decrease in depression score compared to healthy controls [206]. Thus, assessment and treatment of depression should be considered as part of overcoming malnutrition in HD patients.

4.5.2. Lack of Social Support

ESKD patients exist in a complex matrix of relationships with family, friends, healthcare professionals, and financial support [207]. The quality of emotional and financial support provided by their social network influences stress management, QoL, health-promoting behaviors, malnutrition, and mortality in HD patients [196,201,207].
HD patients lacking social support have higher prevalence of diminished appetite, reduced physical functioning, and poor adherence to HD treatment [197,200,201]. Greater non-adherence toward nutritional recommendations found in HD patients without family support was due to absence of personal engagement and encouragement from family members to tackle these issues [197,198]. DOPPS Phases 1–3 found that HD patients with poor social support were more likely to experience serum albumin <3.5 g/dL (OR = 1.18, 95% CI: 1.02–1.37) [201]. In contrast, HD patients with social support achieved better social interactions and coping mechanisms toward kidney disease [202], as well as fewer depression symptoms [208]. Ultimately, presence of social support enables patient self-efficacy to reach better health status.

4.5.3. Financial Constraints

Financial constraints commonly faced by HD patients may be attributed to physical limitations to perform work tasks imposed by treatment and time commitments to dialysis treatment [200]. With unemployment, HD patients are dependent on financial support from caregivers or welfare agencies. Incurring financial dependence triggers loss of self-esteem and depression, leading to poor self-efficacy toward health management [195]. The consequence of limited financial resources is suboptimal dietary intake [200].
Employment status in 231 Chinese working-age HD patients was 51% prior to HD initiation, which fell to 11% once they began treatment [209]. These patients reported that the dialysis schedule and post-dialysis fatigue were major reasons for unemployment. HD patients need to strictly adhere to their dialysis schedule of three sessions per week, which consumes up to 18 working h a week [200]. Additionally, fatigue arising post dialysis affects their ability to work [210,211]. Data from the Finnish Registry for Kidney Diseases (n = 2637) found that peritoneal dialysis patients compared to HD had a higher employment rate (19% vs. 31%, p < 0.001) from greater flexibility in treatment schedule and mobility [212].
Lack of income is commonly prevalent in approximately 50% of the HD population [202,203]. Financial constraints were blamed for poor adherence to dietary recommendations as patients limited their access to healthy food choices on the basis of cost or these patients faced a dilemma on having to choose between spending on medicine or food [202]. Contrarily, HD patients with employment achieved greater DEI (+281 kcal/day, p < 0.01) despite perceiving their income to be insufficient to meet their needs [213].
The influence of financial status on dietary intake is unclear but forms a factor contributive to malnutrition [142,213]. Freitas et al. (2014) indicated that low income was associated with a 13% increase in malnutrition prevalence in 344 Brazilian HD patients [203]. Higher SGA and MIS scores of patients indicative of malnutrition were associated with socioeconomic-related nutritional barriers such as difficulty in purchasing food (OR = 1.89, 95% CI: 1.27–2.88, p = 0.002) and requiring assistance in meal preparation (OR = 1.15, 95% CI: 1.06–2.06, p = 0.001) [166]. Both factors highlighted the impact of financial constraints on nutritional status of HD patients.

4.5.4. Decreased Physical Functioning

Comorbidities associated with CKD such as sarcopenia, vascular dysfunction, inflammation, and malnutrition [214] negatively impacts the three components of physical functioning [215] which are related to body functions and structure, ability to perform, and participation in physical activity.
Fatigue is central to the reduced physical capacity to perform activities of daily living by HD patients and contributes to malnutrition [207,216,217]. The dialysis process itself may cause fatigue, stiffening of joints, and muscle cramping, thus affecting work task performance [218]. Indeed, fatigue was reported to affect the ability to prepare meals as indicated by 59% of HD patients reporting “being too tired to prepare meal” as a barrier toward dietary adherence, and this barrier was associated with lower DEI (r = −0.125, p = 0.002) [213]. Malnourished HD patients identified using Mini Nutritional Assessment < 19 and SGA > 8 had poor activities of daily living score [219], suggesting that ability to perform simple daily tasks was affected in these patients.

5. Conclusions

We presented a global view that malnutrition susceptibility in the HD community is either or both of iatrogenic and of non-iatrogenic origins. Keeping in view disparities in dialysis provision between upper- and lower–middle-income countries, leveraging the point of origin of malnutrition in dialysis patients by healthcare practitioners would enable personalized patient care, as well as country-specific malnutrition treatment strategies. Nutrition assessment is a critical first step to identify the factors of iatrogenic and/or non-iatrogenic origin implicated in malnutrition etiology. This is important as the next step of prevention or treatment for malnutrition depends on the identified factors and aligning effective strategies for nutritional intervention.
The iatrogenic factors that may be implicated in malnutrition may be (i) low dialysis adequacy resulting in poor uremia and metabolic acidosis correction, and/or (ii) low serum albumin levels for patients if dialyzing on highly permeable membranes or dialysis techniques that incur greater amino-acid losses. The non-iatrogenic approach should identify implications of (i) dietary inadequacy as per suboptimal DEIs and DPIs, (ii) poor appetite status, inflammation markers, low diet quality, and high diet monotony index, which indicate barriers to achieving dietary adequacy, and/or (iii) identification of psychosocial and financial barriers to nutritional optimization. These factors are modifiable and should be incorporated as part of a comprehensive nutritional assessment. Identification of factors causing malnutrition that are patient-specific would be crucial for healthcare practitioners to provide a more personalized patient care to treat malnutrition.

Author Contributions

Conceptualization, S.S. and T.K.; literature search, S.S.; data curation, S.S.; visualization, S.S., B.-H.K., H.-M.N. and T.K.; writing—original draft preparation, S.S., B.-H.K. and T.K.; writing—review and editing, S.S., B.-H.K., A.H.A.G., Z.A.M.D., D.M., and T.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

S.S. was a recipient of the MyBrain15 scholarship from the Ministry of Higher Education (Malaysia). B.-H.K. is a postdoctoral researcher (MI-2020-004) from Universiti Kebangsaan Malaysia for postgraduate study.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. GBD Chronic Kidney Disease Collaboration. Global, regional, and national burden of chronic kidney disease, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet 2020, 395, 709–733. [Google Scholar] [CrossRef] [Green Version]
  2. Liyanage, T.; Ninomiya, T.; Jha, V.; Neal, B.; Patrice, H.M.; Okpechi, I.; Zhao, M.; Lv, J.; Garg, A.X.; Knight, J.; et al. Worldwide access to treatment for end-stage kidney disease: A systematic review. Lancet 2015, 385, 1975–1982. [Google Scholar] [CrossRef]
  3. National Kidney Foundation. KDOQI Clinical Practice Guidelines for HD Adequacy: 2015 Update. Am. J. Kidney Dis. 2015, 66, 884–930. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Carrero, J.J.; Thomas, F.; Nagy, K.; Arogundade, F.; Avesani, C.M.; Chan, M.; Chmielewski, M.; Cordeiro, A.C.; Espinosa-Cuevas, A.; Fiaccadori, E.; et al. Global Prevalence of Protein-Energy Wasting in Kidney Disease: A Meta-analysis of Contemporary Observational Studies from the International Society of Renal Nutrition and Metabolism. J. Ren. Nutr. 2018, 28, 380–392. [Google Scholar] [CrossRef]
  5. Takahashi, H.; Inoue, K.; Shimizu, K.; Hiraga, K.; Takahashi, E.; Otaki, K.; Yoshikawa, T.; Furuta, K.; Tokunaga, C.; Sakakibara, T.; et al. on behalf of the Tokai Renal Nutrition Study Group. Comparison of Nutritional Risk Scores for Predicting Mortality in Japanese Chronic Hemodialysis Patients. J. Ren. Nutr. 2017, 27, 201–206. [Google Scholar] [CrossRef]
  6. Toledo, F.; Antunes, A.; Vannini, F.C.D.; Silveira, L.V.A.; Martin, L.C.; Barretti, P.; Caramori, J.C.T. Validity of malnutrition scores for predicting mortality in chronic hemodialysis patients. Int. Urol. Nephrol. 2013, 45, 1747–1752. [Google Scholar] [CrossRef]
  7. Uy, M.C.; Lim-Alba, R.; Chua, E. Association of Dialysis Malnutrition Score with Hypoglycemia and Quality of Life among Patients with Diabetes on Maintenance Hemodialysis. J. ASEAN Fed. Endocr. Soc. 2018, 33, 137–145. [Google Scholar] [CrossRef]
  8. Carrero, J.J.; Stenvinkel, P.; Cuppari, L.; Ikizler, T.A.; Kalantar-Zadeh, K.; Kaysen, G.; Mitch, W.E.; Price, S.R.; Wanner, C.; Wang, A.Y.; et al. Etiology of the protein-energy wasting syndrome in chronic kidney disease: A consensus statement from the International Society of Renal Nutrition and Metabolism (ISRNM). J. Ren. Nutr. 2013, 23, 77–90. [Google Scholar] [CrossRef] [Green Version]
  9. Tan, R.; Long, J.; Fang, S.; Mai, H.; Lu, W.; Liu, Y.; Wei, J.; Yan, F. Nutritional Risk Screening in patients with chronic kidney disease. Asia Pac. J. Clin. Nutr. 2016, 25, 249–256. [Google Scholar]
  10. Vanholder, R.; Fouque, D.; Glorieux, G.; Heine, G.H.; Kanbay, M.; Mallamaci, F.; Massy, Z.A.; Ortiz, A.; Rossignol, P.; Wiecek, A.; et al. For the European Renal Association European Dialysis and Transplant Association (ERA-EDTA) European Renal and Cardiovascular Medicine (EURECA-m) working group. Clinical management of the uraemic syndrome in chronic kidney disease. Lancet Diabetes Endocrinol. 2016, 4, 360–373. [Google Scholar] [CrossRef]
  11. Caetano, C.; Valente, A.; Oliveira, T.; Garagarza, C. Body composition and mortality predictors in hemodialysis patients. J. Ren. Nutr. 2016, 26, 81–86. [Google Scholar] [CrossRef]
  12. Fouque, D.; Kalantar-Zadeh, K.; Kopple, J. A proposed nomenclature and diagnostic criteria for protein-energy wasting in acute and chronic kidney disease. Kidney Int. 2008, 73, 391–398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Bonilla-Palomas, J.L.; Gámez-López, A.L.; Anguita-Sánchez, M.P.; Castillo-Domínguez, J.C.; García-Fuertes, D.; Crespin-Crespin, M.; López-Granados, A.; Suárez de Lezo, J. Impact of malnutrition on long-term mortality in hospitalized patients with heart failure. Rev. Esp. Cardiol. 2011, 64, 752–758. [Google Scholar] [CrossRef]
  14. Tuegel, C.; Bansal, N. Heart failure in patients with kidney disease. Heart 2017, 103, 1848–1853. [Google Scholar] [CrossRef] [PubMed]
  15. Grossniklaus, D.A.; O’Brien, M.C.; Clark, P.C.; Dunbar, S.B. Nutrient intake in heart failure patients. J. Cardiovasc. Nurs. 2008, 23, 357–363. [Google Scholar] [CrossRef] [Green Version]
  16. Naini, A.E.; Karbalaie, A.; Abedini, M.; Askari, G.; Moeinzadeh, F. Comparison of malnutrition in hemodialysis and peritoneal dialysis patients and its relationship with echocardiographic findings. J. Res. Med. Sci. 2016, 21, 78. [Google Scholar] [CrossRef]
  17. Spatola, L.; Finazzi, S.; Calvetta, A.; Reggiani, F.; Morenghi, E.; Santostasi, S.; Angelini, C.; Badalamenti, S.; Mugnai, G. Subjective Global Assessment-Dialysis Malnutrition Score and cardiovascular risk in hemodialysis patients: An observational cohort study. J. Nephrol. 2018, 31, 757–765. [Google Scholar] [CrossRef] [PubMed]
  18. Van der Tol, A.; Lameire, N.; Morton, R.L.; Biesen, M.V.; Vanholder, R. An International Analysis of Dialysis Services Reimbursement. Clin. J. Am. Soc. Nephrol. 2019, 14, 83–94. [Google Scholar] [CrossRef] [PubMed]
  19. Robinson, B.M.; Zhang, J.; Morgenstern, H.; Bradbury, B.D.; Ng, L.J.; McCullough, K.P.; Gillespie, B.W.; Hakim, R.; Rayner, H.; Fort, J.; et al. Worldwide, mortality risk is high soon after initiation of hemodialysis. Kidney Int. 2013, 85, 158–165. [Google Scholar] [CrossRef] [Green Version]
  20. Lukowsky, L.R.; Kheifets, L.; Arah, O.A.; Nissenson, A.R.; Kalantar-Zadeh, K. Patterns and Predictors of Early Mortality in Incident Hemodialysis Patients: New Insights. Am. J. Nephrol. 2012, 35, 548–558. [Google Scholar] [CrossRef] [Green Version]
  21. Goldstein, M.; Yassa, T.; Dacouris, N.; McFarlane, P. Multidisciplinary Predialysis Care and Morbidity and Mortality of Patients on Dialysis. Am. J. Kidney Dis. 2004, 44, 706–714. [Google Scholar] [CrossRef]
  22. McQuillan, R.; Trpeski, L.; Fenton, S.; Lok, C.E. Modifiable Risk Factors for Early Mortality on Hemodialysis. Int. J. Nephrol. 2012, 2012, 435736. [Google Scholar] [CrossRef] [PubMed]
  23. Slinin, Y.; Guo, H.; Gilbertson, D.T.; Mau, L.W.; Ensrud, K.; Collins, A.J.; Ishani, A. Prehemodialysis Care by Dietitians and First-Year Mortality After Initiation of Hemodialysis. Am. J. Kidney Dis. 2011, 58, 583–590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ledebo, I.; Kessler, M.; van Biesen, W.; Wanner, C.; Wiecek, A.; Prichard, S.; Argilés, A.; Ritz, E. Initiation of dialysis: Opinions from an international survey: Report on the dialysis opinion symposium at the ERA-EDTA Congress, 18 September 2000, Nice. Nephrol. Dial. Transplant. 2001, 16, 1132–1138. [Google Scholar] [CrossRef] [Green Version]
  25. van de Luijtgaarden, M.W.; Noordzij, M.; Tomson, C.; Couchoud, C.; Cancarini, G.; Ansell, D.; Bos, W.J.; Dekker, F.W.; Gorriz, J.L.; Iatrou, C.; et al. Factors Influencing the Decision to Start Renal Replacement Therapy: Results of a Survey Among European Nephrologists. Am. J. Kidney Dis. 2012, 60, 940–948. [Google Scholar] [CrossRef] [Green Version]
  26. Rosansky, S.J.; Cancarini, G.; Clark, W.F.; Eggers, P.; Germaine, M.; Glassock, R.; Goldfarb, D.S.; Harris, D.; Hwang, S.J.; Imperial, E.B.; et al. Dialysis Initiation: What’s the Rush? Semin. Dial. 2013, 26, 650–657. [Google Scholar] [CrossRef] [Green Version]
  27. Nesrallah, G.E.; Mustafa, R.A.; Clark, W.F.; Bass, A.; Barnieh, L.; Hemmelgarn, B.R.; Klarenbach, S.; Quinn, R.R.; Hiremath, S.; Ravani, P.; et al. Canadian Society of Nephrology 2014 clinical practice guideline for timing the initiation of chronic dialysis. CMAJ 2014, 186, 112–117. [Google Scholar] [CrossRef] [Green Version]
  28. Kelly, J.; Stanley, M.; Harris, D. Caring for Australians with Renal Impairment (CARI). The CARI guidelines. Acceptance into dialysis guidelines. Nephrology 2005, 10, S46–S60. [Google Scholar] [CrossRef]
  29. Carrero, J.J.; Nakashima, A.; Qureshi, A.R.; Lindholm, B.; Heimburger, O.; Barany, P.; Stenvinkel, P. Protein-energy wasting modifies the association of ghrelin with inflammation, leptin, and mortality in hemodialysis patients. Kidney Int. 2011, 79, 749–756. [Google Scholar] [CrossRef] [Green Version]
  30. Gama-Axelsson, T.; Heimb€urger, O.; Stenvinkel, P.; Barany, P.; Lindholm, B.; Qureshi, A.R. Serum albumin as predictor of nutritional status in patients with ESRD. Clin. J. Am. Soc. Nephrol. 2012, 7, 1446–1453. [Google Scholar] [CrossRef]
  31. Therrien, M.; Byham-Gray, L.; Beto, J. A Review of Dietary Intake Studies in Maintenance Dialysis Patients. J. Ren. Nutr. 2015, 25, 329–338. [Google Scholar] [CrossRef] [PubMed]
  32. Kwon, Y.E.; Yoon, C.Y.; Han, I.M.; Han, S.G.; Park, K.S.; Lee, M.J.; Park, J.T.; Han, S.H.; Yoo, T.H.; Kim, Y.L.; et al. Change of Nutritional Status Assessed Using Subjective Global Assessment Is Associated with All-Cause Mortality in Incident Dialysis Patients. Medicine 2016, 95, e2714. [Google Scholar] [CrossRef] [PubMed]
  33. Araujo, L.C.; Kamimira, M.A.; Draibe, S.A.; Canziani, M.E.F.; Manfredi, S.R.; Avesani, C.M.; Sesso, R.; Cuppari, L. Nutritional Parameters and Mortality in Incident Hemodialysis Patients. J. Ren. Nutr. 2016, 16, 27–35. [Google Scholar] [CrossRef]
  34. Bradbury, B.D.; Fissell, R.B.; Albert, J.M.; Anthony, M.S.; Critchlow, C.W.; Pisoni, R.L.; Port, F.K.; Gillespie, B.W. Predictors of Early Mortality among Incident US Hemodialysis Patients in the Dialysis Outcomes and Practice Patterns Study (DOPPS). Clin. J. Am. Soc. Nephrol. 2007, 2, 89–99. [Google Scholar] [CrossRef]
  35. Murray, D.P.; Young, L.; Waller, J.; Wright, S.; Colombo, R.; Baer, S.; Spearman, V.; Garcia-Torres, R.; Williams, K.; Kheda, M.; et al. Is Dietary Protein Intake Predictive of One-Year Mortality in Dialysis Patients? Am. J. Med. Sci. 2018, 356, 234–243. [Google Scholar] [CrossRef] [PubMed]
  36. Pifer, T.B.; Mccullough, K.P.; Port, F.K.; Goodkin, D.A.; Maroni, B.J.; Held, P.J.; Young, E.W. Mortality risk in hemodialysis patients and changes in nutritional indicators: DOPPS. Kidney Int. 2002, 62, 2238–2245. [Google Scholar] [CrossRef] [Green Version]
  37. Dekker, M.J.E.; Marcelli, D.; Canaud, B.; Konings, C.J.A.M.; Leunissen, K.M.; Levin, N.W.; Carioni, P.; Maheshwari, V.; Raimann, J.G.; van der Sande, F.M.; et al. Unraveling the relationship between mortality, hyponatremia, inflammation and malnutrition in hemodialysis patients: Results from the international MONDO initiative. Eur. J. Clin. Nutr. 2016, 70, 779–784. [Google Scholar] [CrossRef]
  38. Rosenberger, J.; Kissova, V.; Majernikova, M.; Straussova, Z.; Boldizsar, J. Body composition monitor assessing malnutrition in the hemodialysis population independently predicts mortality. J. Ren. Nutr. 2014, 24, 172–176. [Google Scholar] [CrossRef]
  39. Chertow, G.M.; Goldstein-Fuchs, D.J.; Lazarus, J.M.; Kaysen, G.A. Prealbumin, mortality and cause-specific hospitalization in hemodialysis patients. Kidney Int. 2005, 68, 2794–2800. [Google Scholar] [CrossRef] [Green Version]
  40. Butterworth, C.E. The skeleton in the hospital closet. Nutr. Today 1974, 9, 4–8. [Google Scholar] [CrossRef]
  41. Van Gelder, M.; Abrahams, A.C.; Joles, J.A.; Kaysen, G.A.; Gerritsen, K. Albumin handling in different hemodialysis modalities. Nephrol. Dial. Transplant. 2018, 33, 906–913. [Google Scholar] [CrossRef] [PubMed]
  42. Wolfson, M.; Jones, M.R.; Kopple, J.D. Amino acid losses during hemodialysis with infusion of amino acids and glucose. Kidney Int. 1982, 21, 500–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Ikizler, T.A.; Flakoli, P.J.; Parker, R.A.; Hakim, R.M. Amino acid and albumin losses during hemodialysis. Kidney Int. 1994, 46, 830–837. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Salame, C.; Eaton, S.; Grimble, G.; Davenport, A. Protein Losses and Urea Nitrogen Underestimate Total Nitrogen Losses in Peritoneal Dialysis and Hemodialysis Patients. J. Ren. Nutr. 2018, 28, 317–323. [Google Scholar] [CrossRef]
  45. Ikizler, T.A.; Pupim, L.B.; Brouillette, J.R.; Levenhagen, D.K.; Farmer, K.; Hakim, R.M.; Flakoll, P.J. Hemodialysis stimulates muscle and whole body protein loss and alters substrate oxidation. Am. J. Physiol.-Endocrinol. Metab. 2002, 282, E107–E116. [Google Scholar] [CrossRef] [Green Version]
  46. Murtas, S.; Aquilani, R.; Deiana, M.L. Differences in amino acid loss between high-efficiency hemodialysis and post-dilution and pre-dilution hemodiafiltration using high convection volume exchange-A new metabolic scenario? A pilot study. J. Ren. Nutr. 2019, 29, 126–135. [Google Scholar] [CrossRef] [Green Version]
  47. Pupim, L.B.; Flakoll, P.J.; Brouillette, J.R.; Levenhagen, D.K.; Hakim, R.M.; Ikizler, T.A. Intradialytic parenteral nutrition improves protein and energy homeostasis in chronic hemodialysis patients. J. Clin. Investig. 2002, 110, 483–492. [Google Scholar] [CrossRef]
  48. Kohlová, M.; Amorim, C.G.; Araújo, A.; Santos-Silva, A.; Solich, P.; Montenegro, M.C.B.S.M. The biocompatibility and bioactivity of hemodialysis membranes: Their impact in end-stage renal disease. J. Artif. Organs. 2019, 22, 14–28. [Google Scholar] [CrossRef]
  49. Krieter, D.H.; Canaud, B. High permeability of dialysis membranes: What is the limit of albumin loss? Nephrol. Dial. Transplant. 2003, 18, 651–654. [Google Scholar] [CrossRef] [Green Version]
  50. Fouque, D.; Pelletier, S.; Mafra, D.; Chauveau, P. Nutrition and chronic kidney disease. Kidney Int. 2011, 80, 348–357. [Google Scholar] [CrossRef] [Green Version]
  51. Honeich, N.A.; Woffindin, C.; Matthews, J.N.S.; Goldfinch, M.E.; Turnbull, J. Clinical comparison of high-flux cellulose acetate and synthetic membranes. Nephrol. Dial. Transplant. 1994, 9, 60–66. [Google Scholar]
  52. Gil, H.W.; Yang, J.O.; Lee, E.Y.; Lee, E.M.; Choi, J.S.; Hong, S.Y. The Effect of Dialysis Membrane Flux on Amino Acid Loss in Hemodialysis Patients. J. Korean Med. Sci. 2007, 22, 598–603. [Google Scholar] [CrossRef] [PubMed]
  53. Kirsch, A.H.; Lyko, R.; Nilsson, L.G.; Beck, W.; Amdahl, M.; Lechner, P.; Schneider, A.; Wanner, C.; Rosenkranz, A.R.; Krieter, D.H. Performance of hemodialysis with novel medium cut-off dialyzers. Nephrol. Dial. Transplant. 2017, 32, 165–172. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Meert, N.; Eloot, S.; Schepers, E.; Lemke, H.D.; Dhondt, A.; Glorieux, G.; Landschoot, M.V.; Waterloos, M.A.; Vanholder, R. Comparison of removal capacity of two consecutive generations of high-flux dialysers during different treatment modalities. Nephrol. Dial. Transplant. 2011, 26, 2624–2630. [Google Scholar] [CrossRef] [Green Version]
  55. Scribner, B.H. A personalized history of chronic hemodialysis. Am. J. Kidney Dis. 1990, 16, 511–519. [Google Scholar] [CrossRef]
  56. Fagugli, R.M.; De Smet, R.; Buoncristiani, U.; Lameire, N.; Vanholder, R. Behavior of Non–Protein-Bound and Protein-Bound Uremic Solutes During Daily Hemodialysis. Am. J. Kidney Dis. 2002, 40, 339–347. [Google Scholar] [CrossRef]
  57. Florens, N.; Juillard, L. Large Middle Molecule and Albumin Removal: Why Should We Not Rest on Our Laurels? Contrib. Nephrol. 2017, 191, 178–187. [Google Scholar]
  58. Krieter, D.H.; Hackl, A.; Rodriguez, A.; Chenine, L.; Moragues, H.L.; Lemke, H.D.; Wanner, C.; Canaud, B. Protein-bound uraemic toxin removal in haemodialysis and post-dilution haemodiafiltration. Nephrol. Dial. Transplant. 2010, 25, 212–218. [Google Scholar] [CrossRef]
  59. Locatelli, F.; Martin-Malo, A.; Hannedouche, T. Effect of membrane permeability on survival of hemodialysis patients. J. Am. Soc. Nephrol. 2009, 20, 645–654. [Google Scholar] [CrossRef] [Green Version]
  60. Tiranathanagul, K.; Praditpornsilpa, K.; Katavetin, P.; Srisawat, N.; Townamchai, N.; Susantitaphong, P.; Tungsanga, K.; Eiam-Ong, S. On-line Hemodiafiltration in Southeast Asia: A Three-year Prospective Study of a Single Center. Ther. Apher. Dial. 2009, 13, 56–62. [Google Scholar] [CrossRef]
  61. Boschetti-de-Fierro, A.; Beck, W.; Hildwein, H.; Krause, B.; Storr, M.; Zweigart, C. Membrane Innovation in Dialysis. Contrib. Nephrol. 2017, 191, 100–114. [Google Scholar] [PubMed]
  62. Slinin, Y.; Greer, N.; Ishani, A.; MacDonald, R.; Olson, C.; Rutks, I.; Wilt, T.J. Timing of Dialysis Initiation, Duration and Frequency of Hemodialysis Sessions, and Membrane Flux: A Systematic Review for a KDOQI Clinical Practice Guideline. Am. J. Kidney Dis. 2015, 66, 823–836. [Google Scholar] [CrossRef] [PubMed]
  63. Kneis, C.; Beck, W.; Boenisch, O.; Klefisch, F.; Deppisch, R.; Zickler, D.; Schindler, R. Elimination of Middle-Sized Uremic Solutes with High-Flux and High-Cut-Off Membranes: A Randomized in vivo Study. Blood Purif. 2013, 36, 287–294. [Google Scholar] [CrossRef] [PubMed]
  64. Gondouin, B.; Hutchison, C.A. High Cut-off Dialysis Membranes: Current Uses and Future Potential. Adv. Chronic Kidney Dis. 2011, 18, 180–187. [Google Scholar] [CrossRef] [PubMed]
  65. Girndt, M.; Fiedler, R.; Martus, P.; Pawlak, M.; Storr, M.; Bohler, T.; Glomb, M.A.; Liehr, K.; Henning, C.; Templin, M.; et al. High cut-off dialysis in chronic haemodialysis patients. Eur. J. Clin. Investig. 2015, 45, 1333–1340. [Google Scholar] [CrossRef]
  66. Zickler, D.; Schindler, R.; Willy, K.; Martus, P.; Pawlak, M.; Storr, M.; Hulko, M.; Boehler, T.; Glomb, M.A.; Liehr, K.; et al. Medium Cut-Off (MCO) Membranes Reduce Inflammation in Chronic Dialysis Patients: A Randomized Controlled Clinical Trial. PLoS ONE 2017, 12, e0169024. [Google Scholar] [CrossRef]
  67. Upadhyay, A.; Jaber, B.L. Reuse and Biocompatibility of Hemodialysis Membranes: Clinically Relevant? Semin. Dial. 2017, 30, 121–124. [Google Scholar] [CrossRef]
  68. Upadhyay, A. Dialyzer reuse: Is it safe and worth it? Braz. J. Nephrol. 2019, 41, 312–314. [Google Scholar] [CrossRef] [Green Version]
  69. Lacson, E., Jr.; Lazarus, J.M. Dialyzer best practice: Single use or reuse? Semin. Dial. 2006, 19, 120–128. [Google Scholar] [CrossRef]
  70. Ribeiro, I.C.; Roza, N.A.V.; Duarte, D.A.; Guadagnini, D.; Elias, R.M.; de Oliveira, R.B. Clinical and microbiological effects of dialyzers reuse in hemodialysis patients. Braz. J. Nephrol. 2019, 41, 384–392. [Google Scholar] [CrossRef]
  71. Argyropoulos, C.; Roumelioti, M.E.; Sattar, A.; Kellum, J.A.; Weissfeld, L.; Unruh, M.L. Dialyzer Reuse and Outcomes of High Flux Dialysis. PLoS ONE 2015, 10, e0129575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Hamid, A.; Dhrolia, M.F.; Imtiaz, S.; Qureshi, R.; Ahmad, A. Comparison of Adequacy of Dialysis between Single-use and Reused Hemodialyzers in Patients on Maintenance Hemodialysis. J. Coll. Physicians Surg. Pak. 2019, 29, 720–723. [Google Scholar] [CrossRef] [PubMed]
  73. Jofre, R.; Rodriguez-Benitez, P.; Lopez-Gomez, J.M.; Perez-Garcia, R. Inflammatory Syndrome in Patients on Hemodialysis. J. Am. Soc. Nephrol. 2006, 17, S274–S280. [Google Scholar] [CrossRef]
  74. Poppelaars, F.; Faria, B.; da Costa, M.G.; Franssen, C.F.M.; van Son, W.J.; Berger, S.P.; Daha, M.R.; Seelen, M.A. The Complement System in Dialysis: A Forgotten Story? Front. Immunol. 2018, 9, 71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Abe, M.; Hamano, T.; Wada, A.; Nakai, S.; Masakane, I. Effect of dialyzer membrane materials on survival in chronic hemodialysis patients: Results from the annual survey of the Japanese Nationwide Dialysis Registry. PLoS ONE 2017, 12, e0184424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Wong, J.; Vilar, E.; Farrington, K. Endotoxemia in End-Stage Kidney Disease. Semin. Dial. 2014, 28, 59–67. [Google Scholar] [CrossRef]
  77. Gagliardi, G.M.; Rossi, S.; Condino, F.; Mancuso, D.; Greco, F.; Tenuta, R.; Savino, O.; Bonofiglio, R.; Domma, F.; Latorre, G. Malnutrition, infection and arteriovenous fistula failure: Is there a link? J. Vasc. Access 2011, 12, 57–62. [Google Scholar] [CrossRef]
  78. Dukkipati, R.; Molnar, M.Z.; Park, J.; Jing, J.; Kovesdy, C.P.; Kajani, R.; Kalantar-Zadeh, K. Association of Vascular Access Type with Inflammatory Marker Levels in Maintenance Hemodialysis Patients. Semin. Dial. 2014, 27, 415–423. [Google Scholar] [CrossRef] [Green Version]
  79. Zavacka, M.; Zelko, A.; Madarasova Geckova, A.; Majernikova, M.; Pobehova, J.; Zavacky, P. Vascular access as a survival factor for the haemodialysis population: A retrospective study. Int. Angiol. 2020. [Google Scholar] [CrossRef]
  80. Debska-Slizien, A.; Malgorzewicz, S.; Dudziak, M.; Ksiazek, A.; Sulowicz, W.; Grzeszczak, W. Cardiovascular risk in patients undergoing maintenance hemodialysis with Helixone(R) membrane: A multicenter randomized study. Pol. Arch. Med. Wewn. 2014, 124, 593–598. [Google Scholar]
  81. Movilli, E.; Camerini, C.; Gaggia, P. Total convection affects serum beta2 microglobulin and C-reactive protein but not erythropoietin requirement following post-dilutional hemodiafiltration. Am. J. Nephrol. 2015, 41, 494–501. [Google Scholar] [CrossRef] [PubMed]
  82. Galli, F.; Benedetti, S.; Floridi, A.; Canestrari, F.; Piroddi, M.; Buoncristiani, E.; Buoncristiani, U. Glycoxidation and inflammatory markers in patients on treatment with PMMA-based protein-leaking dialyzers. Kidney Int. 2005, 67, 750–759. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Glorieux, G.; Neirynk, N.; Veys, N.; Vanholder, R. Dialysis water and fluid purity: More than endotoxin. Nephrol. Dial. Transplant. 2012, 27, 4010–4021. [Google Scholar] [CrossRef] [Green Version]
  84. Heumann, D.; Roger, T. Initial responses to endotoxins and Gram-negative bacteria. Clin. Chim. Acta 2002, 323, 59–72. [Google Scholar] [CrossRef]
  85. Bossola, M.; Stasio, E.D.; Sanguinetti, M.; Posteraro, B.; Antocicco, M.; Pepe, G.; Mello, E.; Bugli, F.; Vulpio, C. Serum Endotoxin Activity Measured with Endotoxin Activity Assay Is Associated with Serum Interleukin-6 Levels in Patients on Chronic Hemodialysis. Blood Purif. 2016, 42, 294–300. [Google Scholar] [CrossRef]
  86. El-Koraie, A.F.; Naga, Y.S.; Farahat, N.G.; Hazzah, W.A. Endotoxins and inflammation in hemodialysis patients. Hemodial. Int. 2013, 17, 359–365. [Google Scholar] [CrossRef]
  87. Horl, W.H. Hemodialysis Membranes: Interleukins, biocompatibility, and middle molecules. J. Am. Soc. Nephrol. 2002, 13, 62–71. [Google Scholar]
  88. Vanholder, R.; Pletinck, A.; Schepers, E.; Glorieux, G. Biochemical and Clinical Impact of Organic Uremic Retention Solutes: A Comprehensive Update. Toxins 2018, 10, 33. [Google Scholar] [CrossRef] [Green Version]
  89. Biswas, S.K. Does the Interdependence between Oxidative Stress and Inflammation Explain the Antioxidant Paradox? Oxid. Med. Cell Longev. 2016, 2016, 5698931. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Sanz, A.B.; Sanchez-Nino, M.D.; Ramos, A.M.; Moreno, J.A.; Santamaria, B.; Ruiz-Ortega, M.; Egido, J.; Ortiz, A. NF-κB in Renal Inflammation. J. Am. Soc. Nephrol. 2010, 21, 1254–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  91. Liakopoulos, V.; Roumeliotis, S.; Zarogiannis, S.; Eleftheriadis, T.; Mertens, P.R. Oxidative stress in hemodialysis: Causative mechanisms, clinical implications, and possible therapeutic interventions. Semin. Dial. 2019, 32, 58–71. [Google Scholar] [CrossRef] [PubMed]
  92. Navarro-García, J.A.; Rodríguez-Sánchez, E.; Aceves-Ripoll, J.; Abarca-Zabalía, J.; Susmozas-Sánchez, A.; González Lafuente, L.; Bada-Bosch, T.; Hernández, E.; Mérida-Herrero, E.; Praga, M.; et al. Oxidative Status before and after Renal Replacement Therapy: Differences between Conventional High Flux Hemodialysis and on-Line Hemodiafiltration. Nutrients 2019, 11, 2809. [Google Scholar]
  93. Fatonah, S.; Sulchan, M.; Sofro, M.A.U. Macronutrients, micronutrients intake and inflammation in hemodialysis patients. Potravin. Slovak J. Food Sci. 2019, 13, 891–897. [Google Scholar] [CrossRef] [Green Version]
  94. Barroso, C.F.; Pires, L.V.; Santos, L.B.; Henriques, G.S.; Pessoa, P.P.; de Araújo, G.N.; de Araújo, C.O.D.; Oliveira, C.M.C.; Maia, C.S.C. Selenium Nutritional Status and Glutathione Peroxidase Activity and Its Relationship with Hemodialysis Time in Individuals Living in a Brazilian Region with Selenium-Rich Soil. Biol. Trace Elem. Res. 2020. [Google Scholar] [CrossRef] [PubMed]
  95. Silva, R.E.; Simões-E-Silva, A.C.; Miranda, A.S.; Justino, P.B.I.; Brigagão, M.R.P.L.; Moraes, G.O.I.; Gonçalves, R.V.; Novaes, R.D. Potential Role of Nutrient Intake and Malnutrition as Predictors of Uremic Oxidative Toxicity in Patients with End-Stage Renal Disease. Oxid. Med. Cell. Longev. 2019, 2019, 7463412. [Google Scholar] [CrossRef] [PubMed]
  96. Viramontes Hörner, D.; Selby, N.M.; Taal, M.W. The Association of Nutritional Factors and Skin Autofluorescence in Persons Receiving Hemodialysis. J. Ren. Nutr. 2019, 29, 149–155. [Google Scholar] [CrossRef] [PubMed]
  97. Almeida, S.G.; Veiga, J.P.; Arruda, S.F.; Neves, C.F.; Siqueira, E.M. The association of markers of oxidative-inflammatory status with malnutrition in hemodialysis patients with serum ferritin lower than 500 ng/mL. J. Bras. Nefrol. 2013, 35, 6–12. [Google Scholar] [CrossRef] [Green Version]
  98. Kalantar-Zadeh, K.; Ikizler, T.A.; Block, G.; Avram, M.M.; Kopple, J.D. Malnutrition-inflammation complex syndrome in dialysis patients: Causes and consequences. Am. J. Kidney Dis. 2003, 42, 864–881. [Google Scholar] [CrossRef] [Green Version]
  99. Rippe, B.; Öberg, C.M. Albumin Turnover in Peritoneal and Hemodialysis. Semin. Dial. 2006, 29, 458–462. [Google Scholar] [CrossRef]
  100. Oliveira, C.M.C.; Kubrusly, M.; Lima, A.T.; Torres, D.M.; Cavalcante, N.M.R.; Jeronimo, A.L.C.; Oliveira, T.C.B. Correlation Between Nutritional Markers and Appetite Self-Assessments in Hemodialysis Patients. J. Ren. Nutr. 2015, 25, 301–307. [Google Scholar] [CrossRef]
  101. Essadik, R.; Msaad, R.; Lebrazi, H.; Taki, H.; Tahri, E.H.; Kettani, A.; Madkouri, G.; Ramdani, B.; Saïle, R. Assessing the prevalence of protein-energy wasting in haemodialysis patients: A cross-sectional monocentric study. Nephrol. Ther. 2017, 13, 537–543. [Google Scholar] [CrossRef] [PubMed]
  102. Eloot, S.; Biesen, W.V.; Dhondt, A.; de Wynkele, H.V.; Glorieux, G.; Verdonck, P.; Vanholder, R. Impact of hemodialysis duration on the removal of uremic retention solutes. Kidney Int. 2008, 73, 765–770. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Stegmayr, B. Dialysis Procedures Alter Metabolic Conditions. Nutrients 2017, 9, 548. [Google Scholar] [CrossRef]
  104. Depner, T.A. Uremic toxicity: Urea and beyond. Semin. Dial. 2008, 14, 246–251. [Google Scholar] [CrossRef] [PubMed]
  105. Jansen, J.; Jankowski, J.; Gajjala, P.R.; Wetzels, J.F.M.; Masereeuw, R. Disposition and clinical implications of protein-bound uremic toxins. Clin. Sci. 2017, 131, 1631–1647. [Google Scholar] [CrossRef]
  106. Deltombe, O.; de Loor, H.; Glorieux, G.; Dhondt, A.; Biesen, W.V.; Meijers, B.; Eloot, S. Exploring binding characteristics and the related competition of different protein-bound uremic toxins. Biochimie 2017, 139, 20–26. [Google Scholar] [CrossRef]
  107. Kuhlmann, M.K.; Kotanko, P.; Levin, N.W. Hemodialysis: Outcome and Adequacy. In Comprehensive Clinical Nephrology; Johnson, R.J., Feehally, J., Floege, J., Eds.; Elsevier: Philadelphia, PA, USA, 2015; pp. 1075–1083. [Google Scholar]
  108. Yu, X. The Evolving Patterns of Uremia: Unmet Clinical Needs in Dialysis. Contrib. Nephrol. 2017, 191, 1–7. [Google Scholar]
  109. Rocco, M.V.; Dwyer, J.T.; Larive, B.; Greene, T.; Cockram, D.B.; Chumlea, W.C.; Kusek, J.W.; Leung, J.; Burrowes, J.D.; Mcleroy, S.L.; et al. The effect of dialysis dose and membrane flux on nutritional parameters in hemodialysis patients: Results of the HEMO Study. Kidney Int. 2004, 65, 2321–2334. [Google Scholar] [CrossRef] [Green Version]
  110. Jones, C.B.; Bargman, M. Should we look beyond Kt/V urea in assessing dialysis adequacy? Semin. Dial. 2018, 31, 420–429. [Google Scholar] [CrossRef]
  111. Bossola, M.; Muscaritoli, M.; Tazza, L.; Giungi, S.; Tortorelli, A.; Fanelli, F.R.; Luciani, G. Malnutrition in Hemodialysis Patients: What Therapy? Am. J. Kidney Dis. 2005, 46, 371–386. [Google Scholar] [CrossRef]
  112. Carrero, J.J.; Aguilera, A.; Stenvinkel, P.; Gil, F.; Selgas, R.; Lindhlm, B. Appetite Discorders in Uremia. J. Ren. Nutr. 2008, 18, 107–112. [Google Scholar] [CrossRef] [PubMed]
  113. Daugirdas, J.T.; Greene, T.; Chertow, G.M.; Depner, T.A. Can rescaling dose with 6 of dialysis to body surface area in the HEMO study explain the different responses to dose in women versus men? Clin. J. Am. Soc. Nephrol. 2010, 5, 1628–1636. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Rashidi, A.A.; Soleimani, A.R.; Nikoueinejad, H.; Sarbolouki, S. The Evaluation of Increase in Hemodialysis Frequency on C-Reactive Protein Levels and Nutritional Status. Acta Med. Iran. 2013, 51, 119–124. [Google Scholar] [PubMed]
  115. Chazot, C.; Jean, G. The advantages and challenges of increasing the duration and frequency of maintenance dialysis sessions. Nature Clin. Pract. Nephrol. 2009, 5, 34–44. [Google Scholar] [CrossRef] [PubMed]
  116. Jha, V. Current status of end-stage renal disease care in India and Pakistan. Kidney Inter Suppl. 2013, 3, 157–160. [Google Scholar] [CrossRef] [Green Version]
  117. Jha, V. End-stage renal care in developing countries: The India experience. Ren. Fail. 2004, 26, 201–208. [Google Scholar] [CrossRef]
  118. Chauhan, R.; Mendonca, S. Adequacy of twice weekly hemodialysis in end stage renal disease patients at a tertiary care dialysis centre. Indian J. Nephrol. 2015, 25, 329–333. [Google Scholar] [CrossRef]
  119. Daugirdas, J.T. Hemodialysis Treatment Time: As Important as it seems? Semin. Dial. 2017, 30, 93–98. [Google Scholar] [CrossRef]
  120. Torigoe, A.; Sato, E.; Mori, T.; Ieiri, N.; Takahashi, C.; Ishida, Y.; Hotta, O.; Ito, S. Comparisons of amino acids, body constituents and antioxidative response between long-time HD and normal HD. Hemodial. Int. 2016, 20, S17–S24. [Google Scholar] [CrossRef]
  121. Tentori, F.; Zhang, J.; Li, Y.; Karaboyas, A.; Kerr, P.; Saran, R. Longer dialysis session length is associated with better intermediate outcomes and survival among patients on in-center three times per week hemodialysis: Results from the Dialysis Outcomes and Practice Patterns Study (DOPPS). Nephrol. Dial. Transplant. 2012, 27, 4180–4188. [Google Scholar] [CrossRef]
  122. Rezende, L.R.; de Souza, P.B.; Pereira, G.R.M.; Lugon, J.R. Metabolic acidosis in hemodialysis patients: A review. Braz. J. Nephrol. 2017, 39, 305–311. [Google Scholar] [CrossRef]
  123. Soudan, K.; Ricanati, E.S.; Leon, J.B.; Sehgal, A.R. Determinants of metabolic acidosis among hemodialysis patients. Hemodial. Int. 2006, 10, 209–214. [Google Scholar] [CrossRef]
  124. Abramowitz, M.K. Bicarbonate Balance and Prescription in ESRD. J. Am. Soc. Nephrol. 2017, 28, 726–734. [Google Scholar] [CrossRef]
  125. Gennari, F.J. Acid-base considerations in end stage renal disease. In Principles and Practice of Dialysis; Henrich, W.L., Ed.; Lippincott Williams & Wilkins: Philadelphia, PA, USA, 2004; pp. 393–407. [Google Scholar]
  126. Ikizler, T.A.; Burrowes, J.D.; Byham-Gray, L.D.; Campbell, K.L.; Carrero, J.J.; Chan, W.; Fouque, D.; Friedman, A.N.; Ghaddar, S.; Goldstein-Fuchs, D.J.; et al. KDOQI Clinical Practice Guideline for Nutrition in CKD: 2020 Update. Am J Kidney Dis. 2020, 3, S1–S107. [Google Scholar] [CrossRef] [PubMed]
  127. Thornley-Brown, D.; Saha, M. Dialysate content and risk of sudden cardiac death. Curr. Opin. Nephrol. Hypertens. 2015, 24, 557–562. [Google Scholar] [CrossRef]
  128. Mehrotra, R.; Kopple, J.D.; Wolfsin, M. Metabolic acidosis in maintenance dialysis patients: Clinical considerations. Kidney Int. 2003, 64, S13–S25. [Google Scholar] [CrossRef] [Green Version]
  129. Ikizler, T.A.; Cano, N.J.; Franch, H.; Fouque, D.; Himmelfarb, J.; Kalantar-Zadeh, K.; Kuhlmann, M.K.; Stenvinkel, P.; TerWee, P.; Teta, D.; et al. Prevention and treatment of protein energy wasting in chronic kidney disease patients: A consensus statement by the International Society of Renal Nutrition and Metabolism. Kidney Int. 2013, 84, 1096–1107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Stenvinkel, P.; Carrero, J.J.; von Walden, F.; Ikizler, T.A.; Nader, G.A. Muscle wasting in end-stage renal disease promulgates premature death: Established, emerging and potential novel treatment strategies. Nephrol. Dial. Transplant. 2016, 31, 1070–1077. [Google Scholar] [CrossRef] [Green Version]
  131. DeFronzo, R.A.; Tobin, J.D.; Rowe, J.W.; Andres, R. Glucose intolerance in uremia. Quantification of pancreatic beta cell sensitivity to glucose and tissue sensitivity to insulin. J. Clin. Investig. 1978, 62, 425–435. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Raj, S.; Scott, D.R.; Nguyen, T.; Sachs, G.; Kraut, J.A. Acid stress increases gene expression of proinflammatory cytokines in Madin-Darby canine kidney cells. Am. J. Physiol. Renal. Physiol. 2013, 304, F41–F48. [Google Scholar] [CrossRef] [Green Version]
  133. Kellum, J.A.; Song, M.; Li, J. Lactic and hydrochloric acids induce different patterns of inflammatory response in LPS-stimulated RAW 264.7 cells. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2004, 286, R686–R692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Soleymanian, T.; Ghods, A. The Deleterious Effect of Metabolic Acidosis on Nutritional Status of Hemodialysis Patients. Saudi J. Kidney Dis. Transplant. 2011, 22, 1149–1154. [Google Scholar]
  135. Wu, D.Y.; Shinaberger, C.S.; Regidor, D.L.; McAllister, C.J.; Kopple, J.D.; Kalantar-Zadeh, K. Association between serum bicarbonate and death in hemodialysis patients: Is it better to be acidotic or alkalotic? Clin. J. Am. Soc. Nephrol. 2006, 1, 70–78. [Google Scholar] [CrossRef] [Green Version]
  136. Bommer, J.; Locatelli, F.; Satayathum, S.; Keen, M.L.; Goodkin, D.A.; Saito, A.; Akiba, T.; Port, F.K.; Young, E.W. Association of Predialysis Serum Bicarbonate Levels with Risk of Mortality and Hospitalization in the Dialysis Outcomes and Practice Patterns Study (DOPPS). Am. J. Kidney Dis. 2004, 44, 661–671. [Google Scholar] [CrossRef]
  137. Misra, M. Pro: Higher serum bicarbonate in dialysis patients is protective. Nephrol. Dial. Transplant. 2016, 31, 1220–1224. [Google Scholar] [CrossRef]
  138. Kang, S.S.; Chang, J.W.; Park, Y. Nutritional Status Predicts 10-Year Mortality in Patients with End-Stage Renal Disease on Hemodialysis. Nutrients 2017, 9, 399. [Google Scholar] [CrossRef]
  139. James, G.; Jackson, H. European Guidelines for the Nutritional Care of Adult Renal Patients. EDTNA-ERCA Journal. 2003, 29, 23–43. [Google Scholar] [CrossRef]
  140. Malaysian Dietetic Association (MDA). Malaysian Medical Nutrition Therapy (MNT) Guidelines for Chronic Kidney Disease; MDA: Kuala Lumpur, Malaysia, 2005. [Google Scholar]
  141. Cano, N.; Fiaccadori, E.; Tesinsky, P.; Toigo, G.; Druml, W.; Kuhlmann, M.; Mann, H.; Horl, W.H. ESPEN Guidelines on Enteral Nutrition: Adult Renal Failure. Clin. Nutr. 2006, 25, 295–310. [Google Scholar] [CrossRef] [PubMed]
  142. Sualaheen, A.; Khor, B.H.; Balasubramanian, G.; Sahathevan, S.; Ali, M.S.M.; Narayanan, S.S.; Chinna, K.; Daud, A.A.M.; Khosla, P.; Karupaiah, T. Habitual Dietary Patterns of Patients on Haemodialysis Indicate Nutritional Risk. J. Ren. Nutr. 2020, 30, 322–332. [Google Scholar] [CrossRef]
  143. Burrowes, J.D.; Larive, B.; Cockram, D.B.; Dwyer, J.; Kusek, J.W.; McLeroy, S.; Poole, D.; Rocco, M.V. Effects of Dietary Intake, Appetite, and Eating Habits on Dialysis and Non–Dialysis Treatment Days in Hemodialysis Patients: Cross-Sectional Results From the HEMO Study. J. Ren. Nutr. 2003, 13, 191–198. [Google Scholar] [CrossRef]
  144. Harvinder, G.H.; Chee, W.S.S.; Karupaiah, T.; Sahathevan, S.; Chinna, K.; Ghazali, A.; Bavanandan, S.; Goh, B.L. Comparison of malnutrition prevalence between haemodialysis and continuous ambulatory peritoneal dialysis patients: A cross-sectional study. Malays J. Nutr. 2013, 19, 271–283. [Google Scholar]
  145. Ichikawa, Y.; Hiramatsu, F.; Hamada, H.; Sakai, A.; Hara, K.; Kogirima, M.; Kawahara, K.; Minakuchi, J.; Kawashima, S.; Yamamoto, S. Effect of protein and energy intakes of body composition in non-diabetic maintenance hemodialysis patients. J. Nutr. Sci. Vitaminol. 2007, 53, 410–418. [Google Scholar] [CrossRef] [Green Version]
  146. Moreira, A.C.; Carolino, E.; Domingos, F.; Gaspar, A.; Ponce, P.; Camili, M.E. Nutritional status influences generic and disease-specific quality of life measures in haemodialysis patients. Nutr. Hosp. 2013, 28, 951–957. [Google Scholar] [PubMed]
  147. Rocco, M.V.; Paranandi, L.; Burrowes, J.D.; Cockram, D.B.; Dwyer, J.T.; Kusek, J.W.; Leung, J.; Makoff, R.; Maroni, B.; Poole, D. Nutritional Status in the HEMO Study Cohort at baseline. Am. J. Kidney Dis. 2002, 39, 245–256. [Google Scholar] [CrossRef] [PubMed]
  148. Sahathevan, S.; Se, C.H.; Ng, S.H.; Chinna, K.; Harvinder, G.S.; Chee, W.S.S.; Goh, B.L.; Halim, A.G.; Bavanandan, S.; Ghazali, A.; et al. Assessing protein energy wasting in a Malaysian haemodialysis population using self-reported appetite rating: A cross sectional study. BMC Nephrol. 2015, 16, 99. [Google Scholar] [CrossRef] [Green Version]
  149. Adanan, N.I.H.; Ali, M.S.M.; Lim, J.H.; Zakaria, N.F.; Lim, C.T.S.; Yahya, R.; Gafr, A.H.A.; Karupaiah, T.; Daud, Z.A.M. Investigating Physical and Nutritional Changes During Prolonged Intermittent Fasting in Hemodialysis Patients: A Prospective Cohort Study. J. Ren. Nutr. 2020, 30, e15–e26. [Google Scholar] [CrossRef] [Green Version]
  150. Arslan, Y.; Kiziltan, G. Nutrition-Related Cardiovascular Risk Factors in Hemodialysis Patients. J. Ren. Nutr. 2010, 20, 185–192. [Google Scholar] [CrossRef]
  151. Chauveau, P.; Grigaut, E.; Kolko, A.; Wolff, P.; Combe, C.; Aparicio, M. Evaluation of nutritional status in patients with kidney disease: Usefulness of dietary recall. J. Ren. Nutr. 2007, 17, 88–92. [Google Scholar] [CrossRef] [PubMed]
  152. Johansson, L.; Hickson, M.; Brown, E.A. Influence of Psychosocial Factors on the Energy and Protein Intake of Older People on Dialysis. J. Ren. Nutr. 2013, 23, 348–355. [Google Scholar] [CrossRef] [PubMed]
  153. Kalantar-Zadeh, K.; Kopple, J.D.; Deepak, S.; Block, D.; Block, G. Characteristics of Hemodialysis Patients as Obtained by Food Frequency Questionnaire. J. Food Intake Ren. Nutr. 2002, 12, 17–31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Kim, H.; Lim, H.; Choue, R. A Better Diet Quality is Attributable to Adequate Energy Intake in Hemodialysis Patients. Clin. Nutr. Res. 2015, 4, 46–55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Morais, A.A.C.; Silva, M.A.T.; Faintuch, J.; Vidigal, E.J.; Costa, R.A.; Lyrio, D.C.; Trindade, C.R.; Pitanga, K.K. Correlation of nutritional status and food intake in hemodialysis patients. Clinics 2005, 60, 185–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Shapiro, B.B.; Bross, R.; Morrison, G.; Kalantar-Zadeh, K.; Kopple, J.D. Self-Reported Interview-Assisted Diet Records Underreport Energy Intake in Maintenance Hemodialysis Patients. J. Ren. Nutr. 2015, 25, 357–363. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Vijayan, M.; Abraham, G.; Alex, M.E.; Vijayshree, N.; Reddy, Y.; Fernando, E.; Mather, M.; Nair, S.; Yuvaraj, A. Nutritional status in stage V dialyzed patient versus CKD patient on conservative therapy across different economic status. Ren. Fail. 2014, 36, 384–389. [Google Scholar] [CrossRef]
  158. Stumvoll, M.; Meyer, C.; Perriello, G.; Kreider, M.; Welle, S.; Gerich, J. Human kidney and liver gluconeogenesis: Evidence for organ substrate selectivity. Am. J. Physiol. 1998, 274, E817–E826. [Google Scholar] [CrossRef]
  159. Antunes, A.A.; Delatim Vannini, F.; de Arruda Silveira, L.V.; Martin, L.C.; Barretti, P.; Caramori, J.C. Influence of protein intake and muscle mass on survival in chronic dialysis patients. Ren. Fail. 2010, 32, 1055–1059. [Google Scholar] [CrossRef] [Green Version]
  160. Pauzi, F.A.; Sahathevan, S.; Khor, B.H.; Narayanan, S.S.; Zakaria, N.F.; Abas, F.; Karupaiah, T.; Daud, Z.A.M. Exploring Metabolic Signature of Protein Energy Wasting in Hemodialysis Patients. Metabolites 2020, 10, 291. [Google Scholar] [CrossRef]
  161. Gordon, A. (Ed.) Biochemistry of Hypoglycin and Toxic Hypoglycemic Syndrome. In Food Safety and Quality Systems in Developing Countries; Academic Press: San Diego, CA, USA, 2015; pp. 47–61. [Google Scholar]
  162. Zimmerer, J.L.; Leon, J.B.; Covinsky, K.E.; Desai, U.; Sehgal, A.R. Diet Monotony as a Correlate of Poor Nutritional Intake Among Hemodialysis Patients. J. Ren. Nutr. 2003, 13, 72–77. [Google Scholar] [CrossRef]
  163. Kalantar-Zadeh, K.; Block, G.; McAllister, C.J.; Humphreys, M.H.; Kopple, J. Appetite and inflammation, nutrition, anemia and clinical outcome in hemodialysis patients. Am. J. Clin. Nutr. 2004, 80, 299–307. [Google Scholar] [CrossRef]
  164. Beberashvili, I.; Sinuani, I.; Azar, A.; Yasur, H.; Shapiro, G.; Feldman, L.; Averbukh, Z.; Weissgarten, J. IL-6 Levels, Nutritional Status, and Mortality in Prevalent Hemodialysis Patients. Clin. J. Am. Soc. Nephrol. 2011, 6, 2253–2263. [Google Scholar] [CrossRef] [Green Version]
  165. Lynch, K.E.; Lynch, R.; Curhan, G.C.; Brunelli, S.M. Altered Taste Perception and Nutritional Status among Hemodialysis Patients. J. Ren. Nutr. 2013, 23, 288–295. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Ekramzadeh, M.; Mazloom, Z.; Jafari, P.; Ayatollahi, M.; Sagheb, M.M. Major Barriers Responsible for Malnutrition in Hemodialysis Patients: Challenges to Optimal Nutrition. Nephro-Urol. Mon. 2014, 6, e23158. [Google Scholar] [CrossRef] [Green Version]
  167. Bossola, M.; Luciani, G.; Rosa, F.; Tazza, L. Appetite and Gastrointestinal Symptoms in Chronic Hemodialysis Patients. J. Ren. Nutr. 2011, 21, 448–454. [Google Scholar] [CrossRef]
  168. Tavares, A.P.D.S.R.; Mafra, D.; Leal, V.O.; Gama, M.D.S.; Vieira, R.M.M.F.; Brum, I.S.D.C.; Borges, N.A.; Silva, A.A. Zinc Plasma Status and Sensory Perception in Nondialysis Chronic Kidney Disease Patients. J. Ren. Nutr. 2020. [Google Scholar] [CrossRef] [PubMed]
  169. Ronveaux, C.C.; Tomé, D.; Raybould, H.E. Glucagon-Like Peptide 1 Interacts with Ghrelin and Leptin to Regulate Glucose Metabolism and Food Intake through Vagal Afferent Neuron Signaling. J Nutr. 2015, 145, 672–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  170. Vanitha, R.N.; Kavimani, S.; Soundararajan, P.; Chamundeeswari, D.; Kannan, G.; Rengarajan, S. Ghrelin and its Association with Nutritional and Inflammatory Status of Patients on Maintenance Hemodialysis in a South Indian Tertiary Care Hospital. Ann. Med. Health Sci. Res. 2016, 6, 146–155. [Google Scholar] [CrossRef] [Green Version]
  171. Klok, M.D.; Jakobsdottir, S.; Drent, M.L. The role of leptin and ghrelin in the regulation of food intake and body weight in humans: A review. Obes. Rev. 2007, 8, 21–34. [Google Scholar] [CrossRef]
  172. Ko, Y.T.; Lin, Y.L.; Kuo, C.H.; Lai, Y.H.; Wang, C.H.; Hsu, B.G. Low serum leptin levels are associated with malnutrition status according to malnutrition-inflammation score in patients undergoing chronic hemodialysis. Hemodial. Int. 2020, 24, 221–227. [Google Scholar] [CrossRef]
  173. Montazerifar, F.; Karajibani, M.; Gorgij, F.; Akbari, O. Malnutrition Markers and Serum Ghrelin Levels in Hemodialysis Patients. Int. Sch. Res. Not. 2014, 2014, 765895. [Google Scholar] [CrossRef] [Green Version]
  174. Kara, E.; Ahbap, E.; Sahutoglu, T.; Sakaci, T.; Basturk, Y.; Koc, Y.; Sevinc, M.; Akgol, C.; Ucar, Z.A.; Kayalar, A.O.; et al. Elevated serum leptin levels are associated with good nutritional status in non-obese chronic hemodialysis patients. Clin. Nephrol. 2015, 83, 147–153. [Google Scholar] [CrossRef]
  175. Mafra, D.; Guebre-Egziabher, F.; Cleaud, C.; Arkouche, W.; Mialon, A.; Drai, J.; Fouque, D. Obestatin and ghrelin interplay in hemodialysis patients. Nutrition 2010, 26, 1100–1104. [Google Scholar] [CrossRef] [PubMed]
  176. Iikuni, N.; Lam, Q.L.K.; Lu, L.; Matarese, G.; Cava, A.L. Leptin and Inflammation. Curr. Immunol. Rev. 2008, 4, 70–79. [Google Scholar] [CrossRef] [PubMed]
  177. Molfino, A.; Kaysen, G.A.; Chertow, G.M.; Doyle, J.; Delgado, C.; Dwyer, T.; Laviano, A.; Fanelli, F.R.; Johansen, K.L. Validating Appetite Assessment Tools Among Patients Receiving Hemodialysis. J. Ren. Nutr. 2016, 26, 103–110. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Perez-Fontan, M.; Cordido, F.; Rodriguez-Carmona, A.; Peteiro, J.; Garcia-Naveiro, R.; Garcia-Buela, J. Plasma ghrelin levels in patients undergoing haemodialysis and peritoneal dialysis. Nephrol. Dial. Transplant. 2004, 19, 2095–2100. [Google Scholar] [CrossRef] [Green Version]
  179. Kursat, S.; Colak, H.B.; Toraman, A.; Tekçe, H.; Ulman, C.; Bayturan, O. Relationship of insulin resistance in chronic haemodialysis patients with inflammatory indicators, malnutrition, echocardiographic parameters and 24 hour ambulatory blood pressure monitoring. Scand. J. Urol. Nephrol. 2010, 44, 257–264. [Google Scholar] [CrossRef]
  180. Mak, R.H.K.; DeFronzo, R.A. Glucose and insulin metabolism in urea. Nephron 1992, 61, 377–382. [Google Scholar] [CrossRef]
  181. Siew, E.D.; Ikizler, T.A. Insulin Resistance and Protein Energy Metabolism in Patients with Advanced Chronic Kidney Disease. Semin. Dial. 2010, 23, 378–382. [Google Scholar] [CrossRef]
  182. O’Sullivan, A.J.; Kelly, J.J. Insulin resistance and protein catabolism in non-diabetic hemodialysis patients. Kidney Int. 2007, 71, 98–100. [Google Scholar] [CrossRef] [Green Version]
  183. Hung, A.M.; Ikizler, T.A. Factors Determining Insulin Resistance in Chronic Hemodialysis Patients. Lipid Disord. Metab. 2011, 171, 127–134. [Google Scholar]
  184. Liao, M.T.; Sung, C.C.; Hung, K.C.; Wu, C.C.; Lo, L.; Lu, K.C. Insulin Resistance in Patients with Chronic Kidney Disease. J. Biomed. Biotechnol. 2012, 2012, 691369. [Google Scholar] [CrossRef] [Green Version]
  185. Grosjean, F.; Esposito, P.; Maccarrone, R.; Libetta, C.; Canton, A.D.; Rampino, T. RBP4: A Culprit for Insulin Resistance in End Stage Renal Disease That Can Be Cleared by Hemodiafiltration. BioMed Res. Int. 2017, 2017, 7270595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  186. Yang, Q.; Graham, T.E.; Mody, N. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 2005, 436, 356–362. [Google Scholar] [CrossRef] [PubMed]
  187. Wolf, G. Serum retinol-binding protein: A link between obesity, insulin resistance, and type 2 diabetes. Nutr. Rev. 2007, 65, 251–256. [Google Scholar] [CrossRef] [PubMed]
  188. Lecker, S.H.; Goldberg, A.L.; Mitch, W.E. Protein Degradation by the Ubiquitin-Proteasome Pathway in Normal and Disease States. J. Am. Soc. Nephrol. 2006, 17, 1807–1819. [Google Scholar] [CrossRef]
  189. Rasic-Milutinovic, Z.; Perunicic-Pekovic, G.; Ristic-Medic, D.; Popovic, T.; Glibetic, M.; Djuric, D.M. Insulin resistance and chronic inflammation are associated with muscle wasting in end-stage renal disease patients on hemodialysis. Gen. Physiol. Biophys. 2009, 28, 184–189. [Google Scholar]
  190. Hui-Ling, W.; Ting-Ting, D.; Shi, L.; Ye, X.; Jun, T.; Wei-Feng, H.; Jin-Yuan, Z. Muscle mass loss and intermuscular lipid accumulation were associated with insulin resistance in patients receiving hemodialysis. Chin. Med. J. 2013, 126, 4612–4617. [Google Scholar]
  191. Koo, J.R.; Yoon, J.W.; Kim, S.G.; Lee, Y.K.; Oh, K.H.; Kim, G.H.; Kim, H.J.; Chae, D.W.; Noh, J.W.; Lee, S.K.; et al. Association of Depression with Malnutrition in Chronic Hemodialysis Patients. Am. J. Kidney Dis. 2003, 41, 1037–1042. [Google Scholar] [CrossRef]
  192. Choi, M.J.; Seo, J.W.; Yoon, J.W.; Lee, S.K.; Kim, S.J.; Lee, Y.K.; Noh, J.W.; Koo, J.R. The Malnutrition-Inflammation-Depression- Arteriosclerosis Complex Is Associated with an Increased Risk of Cardiovascular Disease and All-Cause Death in Chronic Hemodialysis Patients. Nephron Clin. Pract. 2012, 122, 44–52. [Google Scholar] [CrossRef]
  193. Ogrizovic, S.S.; Jovanovic, D.; Dopsaj, V.; Radovic, M.; Sumarac, Z.; Bogavac, S.N.; Stosovic, M.; Stanojevic, M.; Nesic, V. Could depression be a new branch of MIA syndrome? Clin. Nephrol. 2008, 71, 164–172. [Google Scholar] [CrossRef]
  194. Lopes, M.B.; Silva, L.F.; Lopes, G.B.; Penalva, M.A.; Matos, C.M.; Robinson, B.M.; Lopes, A.A. Additional Contribution of the Malnutrition–Inflammation Score to Predict Mortality and Patient-Reported Outcomes as Compared With Its Components in a Cohort of African Descent Hemodialysis Patients. J. Ren. Nutr. 2017, 27, 45–52. [Google Scholar] [CrossRef]
  195. Natashia, D.; Yen, M.; Chen, H.M.; Fetzer, S.J. Self-Management Behaviors in Relation to Psychological Factors and Interdialytic Weight Gain among Patients Undergoing Hemodialysis in Indonesia. J. Nurs. Scholarsh. 2019, 51, 417–426. [Google Scholar] [CrossRef] [PubMed]
  196. Kiajamali, M.; Hosseini, M.; Estebsari, F.; Nasiri, M.; Ashktorab, T.; Abdi, A.; Mahmoudi, A.; Salimi, A.; Abadi, A. Correlation between social support, self-efficacy and health-promoting behavior in hemodialysis patients hospitalized in Karaj in 2015. Electron. Physician 2017, 9, 4820–4827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Kara, B.; Caglar, K.; Kilic, S. Nonadherence with diet and fluid restrictions and perceived social support in patients receiving hemodialysis. J. Nurs. Scholarsh. 2007, 39, 243–248. [Google Scholar] [CrossRef]
  198. Dilek, E.; Kocaoz, S. Adherence to diet and fluid restriction of individuals on hemodialysis treatment and affecting factors in Turkey. Jpn. J. Nurs. Sci. 2015, 12, 113–123. [Google Scholar]
  199. Anees, M.; Batool, S.; Imtiaz, M.; Ibrahim, M. Socio-economic factors affecting quality of life of hemodialysis patients and its effects on mortality. Pak. J. Med Sci. 2018, 34, 811–816. [Google Scholar] [CrossRef] [PubMed]
  200. Lopes, A.A.; Elder, S.J.; Ginsberg, N.; Andreucci, V.E.; Cruz, J.M.; Fukuhara, S.; Mapes, D.L.; Saito, A.; Pisoni, R.L.; Saran, R.; et al. Lack of appetite in haemodialysis patients—Associations with patient characteristics, indicators of nutritional status and outcomes in the international DOPPS. Nephrol. Dial. Transplant. 2007, 22, 3538–3546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  201. Untas, A.; Thumma, J.; Rascle, N.; Rayner, H.; Mapes, D.; Lopes, A.A.; Fukuhara, S.; Akizawa, T.; Morgenstern, H.; Robinson, B.M.; et al. The Associations of Social Support and Other Psychosocial Factors with Mortality and Quality of Life in the Dialysis Outcomes and Practice Patterns Study. Clin. J. Am. Soc. Nephrol. 2011, 6, 142–152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  202. Clark-Cutaia, M.N.; Sevick, M.A.; Thurheimer-Cacciotti, J.; Hoffman, L.A.; Snetselaar, L.; Burke, L.E.; Zickmund, S.L. Perceived Barriers to Adherence to Hemodialysis Dietary Recommendations. Clin. Nurs. Res. 2018, 28, 1009–1029. [Google Scholar] [CrossRef]
  203. Freitas, A.T.V.; Vaz, I.M.F.; Ferraz, S.F.; Peixoto, M.R.G.; Campos, M.I.V.M. Prevalence of malnutrition and associated factors in hemodialysis patients. Rev. Nutr. 2014, 27, 357–366. [Google Scholar] [CrossRef] [Green Version]
  204. Gebrie, M.H.; Ford, J.A. Depressive symptoms and dietary nonadherence among end stage renal disease patients undergoing hemodialysis therapy: Systematic review. BMC Nephrol. 2019, 20, 429. [Google Scholar] [CrossRef] [Green Version]
  205. Bujang, M.A.; Musa, R.; Liu, W.J.; Chew, T.F.; Lim, C.T.S.; Morad, Z. Depression, anxiety and stress among patients with dialysis and the association with quality of life. Asian J. Psychiatr. 2015, 18, 49–52. [Google Scholar] [CrossRef]
  206. Koo, J.R.; Yoon, J.Y.; Joo, M.H.; Lee, H.S.; Oh, J.E.; Kim, S.G.; Seo, J.W.; Kim, H.J.; Noh, J.W.; Lee, S.K.; et al. Treatment of Depression and Effect of Antidepression Treatment on Nutritional Status in Chronic Hemodialysis Patients. Am. J. Med. Sci. 2005, 329, 1–5. [Google Scholar] [CrossRef] [Green Version]
  207. Cukor, D.; Cohen, S.D.; Peterson, R.A.; Kimmel, P.L. Psychosocial Aspects of Chronic Disease: ESRD as a Paradigmatic Illness. J. Am. Soc. Nephrol. 2007, 18, 3042–3055. [Google Scholar] [CrossRef] [PubMed]
  208. Liu, X.; Yang, X.; Yao, L.; Zhang, Q.; Sun, D.; Zhu, X.; Xu, T.; Liu, Q.; Wang, L. Prevalence and related factors of depressive symptoms in hemodialysis patients in northern China. BMC Psychiatr. 2017, 17, 128. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  209. Huang, B.; Lai, B.; Xu, L.; Wang, Y.; Cao, Y.; Yan, P.; Chen, J. Low employment and low willingness of being reemployed in Chinese working-age maintained hemodialysis patients. Ren. Fail. 2017, 39, 607–612. [Google Scholar] [CrossRef]
  210. Tangvoraphonkchai, K.; Davenport, A. Extracellular Water Excess and Increased Self-Reported Fatigue in Chronic Hemodialysis Patients. Ther. Apher. Dial. 2018, 22, 152–159. [Google Scholar] [CrossRef] [PubMed]
  211. Hatthakit, U. Lived experiences of patients on hemodialysis: A meta-synthesis. Nephrol. Nurs. J. 2012, 39, 295–304. [Google Scholar]
  212. Helanterä, I.; Haapio, M.; Koskinen, P.; Grönhagen-Riska, C.; Finne, P. Employment of Patients Receiving Maintenance Dialysis and After Kidney Transplant: A Cross-sectional Study from Finland. Am. J. Kidney Dis. 2012, 59, 700–706. [Google Scholar] [CrossRef] [PubMed]
  213. Jules, D.E.S.; Woolf, K.; Pompeii, M.L.; Sevick, M.A. Exploring Problems in Following the Hemodialysis Diet and Their Relation to Energy and Nutrient Intakes: The BalanceWise Study. J. Ren. Nutr. 2016, 26, 118–124. [Google Scholar] [CrossRef] [Green Version]
  214. Painter, P.; Roshanravan, B. The association of physical activity and physical function with clinical outcomes in adults with chronic kidney disease. Curr. Opin. Nephrol. Hypertens. 2013, 22, 615–623. [Google Scholar] [CrossRef]
  215. Painter, P. Physical functioning in end-stage renal disease patients: Update 2005. Hemodial. Int. 2005, 9, 218–235. [Google Scholar] [CrossRef] [PubMed]
  216. Bossola, M.; Marzetti, E.; Stasio, E.D.; Monteburini, T.; Cenerelli, S.; Mazzoli, K.; Parodi, E.; Sirolli, V.; Santarelli, S.; Ippoliti, F.; et al. Prevalence and associated variables of postdialysis fatigue: Results of a prospective multicenter study. Nephrology 2018, 23, 552–558. [Google Scholar] [CrossRef] [PubMed]
  217. Bonner, A.; Wellard, S.; Caltabiano, M. The impact of fatigue on daily activity in people with chronic kidney disease. J. Clin. Nurs. 2010, 19, 3006–3015. [Google Scholar] [CrossRef] [PubMed]
  218. Tchape, O.D.M.; Tchapoga, Y.B.; Atuhaire, C.; Priebe, G.; Cumber, S.N. Physiological and psychosocial stressors among hemodialysis patients in the Buea Regional Hospital, Cameroon. Pan Afr. Med. J. 2018, 30, 49. [Google Scholar] [PubMed]
  219. Abdulan, I.M.; Onofriescu, M.; Stefaniu, R.; Mastaleru, A.; Mocanu, V.; Alexa, I.D.; Covic, A. The predictive value of malnutrition for functional and cognitive status in elderly hemodialysis patients. Int. Urol. Nephrol. 2019, 51, 155–162. [Google Scholar] [CrossRef]
Figure 1. Etiology of malnutrition in dialysis patients.
Figure 1. Etiology of malnutrition in dialysis patients.
Nutrients 12 03147 g001
Table 1. Mortality risk within 12 months of HD initiation according to nutritional indicators of malnutrition.
Table 1. Mortality risk within 12 months of HD initiation according to nutritional indicators of malnutrition.
ReferenceSample Size (n)Predictors of Mortality
Bradbury et al., 2007 [34]4802MonthsBMI < 20 kg/m2Ser.Alb < 3.5 g/L
(AHR 95% CI)
<40.98 (0.67–1.44)1.57 (1.18–2.09)
4–121.38 (0.98–1.94)1.27 (1.00–1.63)
>121.19 (0.93–1.53)1.41 (1.17–1.70)
Lukowsky et al., 2012 [20]18,707MonthsBMI increase by 2
index points
Ser.Alb < 3.5 g/LnPCR > 1.0 g/kg/day
(AHR 95% CI)
<30.92 (0.90–0.94)2.56 (2.30–2.84)1.21 (1.06–1.38)
4–60.93 (0.91–0.95)2.04 (1.81–2.31)0.96 (0.80–1.14)
7–120.94 (0.92–0.96)1.89 (1.70–2.10)0.89 (0.74–1.07)
McQuillan et al., 2015 [22]4807MonthsBMI <18.5 kg/m2
<3AHR (95% CI) = 4.22 (3.12–5.17)
Murray et al., 2018 [35]227ParametersCPHp-value
BMI0.97 (0.85–1.11)0.625
Ser.Alb0.40 (0.12–1.39)0.149
Undefined malnutrition (using clinical judgment)4.70 (0.25–88.78)0.302
Abbreviations: AHR, adjusted hazard ratio; BMI, body mass index; CI, confidence interval; CPH, Cox proportional hazard; HD, hemodialysis; nPCR, normalized protein catabolic rate; Ser.Alb, serum albumin.
Table 2. Mortality risk in maintenance HD patients according to nutritional indicators of malnutrition.
Table 2. Mortality risk in maintenance HD patients according to nutritional indicators of malnutrition.
Parameters Associated to Mortality RiskReferences
Body mass indexCaetano et al., 2016 [11]; Pifer et al., 2002 [36]
Mid-arm muscle circumferenceAraujo et al., 2006 [33]
Fat tissue indexCaetano et al., 2016 [11]
Lean tissue indexDekker et al., 2016 [37]; Rosenberger et al., 2014 [38]
Serum albuminAraujo et al., 2006 [33]; Pifer et al., 2002 [36],
Caetano et al., 2016 [11]
Serum prealbuminChertow et al., 2005 [39]
Modified subjective global assessment (severe malnutrition)Pifer et al., 2002 [36]
Geriatric Nutritional Risk IndexTakahashi et al., 2014 [5]
Dietary energy intakeAraujo et al., 2006 [33]
Abbreviation: HD, hemodialysis. Notes: decreased trend; increased trend.
Table 3. Protein and amino-acid losses according to types of dialyzer membranes.
Table 3. Protein and amino-acid losses according to types of dialyzer membranes.
Types of MembraneNutrient LossesReferences
Cellulosic7–8 g of amino acidsWolfson et al., 1982 [42]; Ikizler et al., 1994 [43]
Cellulose acetate with HF3 g of proteinHoneich et al., 1994 [51]
Cellulose triacetate with HF4 g of proteinHoneich et al., 1994 [51]
Low flux5–6 g of amino acidsIkizler et al., 1994 [43]; Gil et al., 2007 [52]
High flux5–8 g of amino acidsIkizler et al., 1994 [43]; Gil et al., 2007 [52]
3–8 g of proteinHoneich et al., 1994 [51]; Salame et al., 2018 [44]; Ikizler et al., 1994 [43]
Medium cutoff3–7 g of albuminKirsch et al., 2017 [53]
Hemodiafiltration4–6 g of albuminMeert et al., 2011 [54]
9 g of proteinSalame et al., 2018 [44]
Abbreviation: HF, high flux.
Table 4. Effects of type of dialyzer membranes on inflammation status.
Table 4. Effects of type of dialyzer membranes on inflammation status.
ReferenceTreatment Duration (Months)Type of MembraneInflammatory Marker Outcomes
Dębska-Ślizień et al.,
2014 [80]
6Polysulfone (low flux)CRP: 9.3 ± 19.5 to 6.0 ± 6.9 mg/dL
Polysulfone (high flux)CRP: 12.2 ± 27.8 to 6.5 ± 9.2 mg/dL
Movili et al.,
2015 [81]
12Usual hemodialysisCRP: 5.1 ± 6.8 to 5.3 ± 5.0 mg/dL
HemodiafiltrationCRP: 6.8 ± 7.0 to 2.3 ± 2.4 mg/dL
Zickler et al., 2017 [66]1Polyarylethersulfone/polyvinylpyrrolidone
(medium cutoff)
TNF-α: 24.1 ± 8.1 to 20.6 ± 5.8 pg/mL
IL-6: 9.0 ± 13.2 to 6.0 ± 5.9 pg/mL
CRP: 15.3 ± 30.0 to 9.3 ± 14.5 mg/dL
Polyarylethersulfone/polyvinylpyrrolidone
(high flux)
TNF-α: 23.4 ± 7.3 to 22.0 ± 6.0 pg/mL
IL-6: 9.8 ± 20.5 to 5.5 ± 4.5 pg/mL
CRP: 13.4 ± 25.5 to 9.6 ± 15.7 mg/dL
Galli et al.,
2005 [82]
6Polymethylmethacrylate
(high flux)
TNF-α: 18.7 ± 4.3 to 15.1 ± 3.1 a pg/mL
IL-6: 5.0 ± 1.9 to 3.1 ± 0.6 a pg/mL
CRP: 22.7 ± 33.9 to 12.1 ± 9.1 mg/dL
Cellulose acetate/cuprammonium rayon
(low flux)
TNF-α: 19.0 ± 4.0 to 21.5 ± 5.5 pg/mL
IL-6: 5.3 ± 2.1 to 5.8 ± 2.3 pg/mL
CRP: 25.8 ± 28.6 to 27.4 ± 24.0 mg/dL
Abbreviations: CRP, C-reactive protein; IL-6, interleukin-6; TNF-α, tumor necrosis factor alpha; a significantly different (p < 0.05) compared to pre-treatment.
Table 5. Inadequate DEI and DPI in global HD populations.
Table 5. Inadequate DEI and DPI in global HD populations.
Author/YearCountrySample
Size, n
DEI (kcal/kg BW)/dayDPI
(g/kg BW/day)
Dietary Inadequacy a
Large Cross-Sectional/Cohort Studies (n > 100)
Suaheleen et al., 2020 [142]Malaysia38224.9 ± 5.20.90 ± 0.29DEI: 52%
DPI: 40%
Burrowes et al., 2003 [143]United States190122.70 ± 8.300.93 ± 0.35-
Harvinder et al., 2013 b [144]Malaysia15525.5 ± 8.51.07 ± 0.47DEI: 75%
DPI: 67%
Ichikawa et al., 2007 b [145]Japan20029.31.08 ± 0.17-
Kang et al., 2017 [138]Korea144 25.8 ± 5.40.88 ± 0.23-
Moreira et al., 2013 [146]Portugal13025.81.27 DEI: 74.6%
DPI: 32.3%
Rocco et al., 2002 [147]United States100022.90 ± 8.400.93 ± 0.36DEI: 92%
DPI: 81%
Sahathevan et al., 2015 [148]Malaysia20523.12 ± 6.940.94 ± 0.39DEI: 65%
DPI: 42%
Small-Scale Studies (n < 100)
Adanan et al., 2019 [149]Malaysia5421.8 ± 4.80.7 ± 0.2-
Arslan and Kiziltan, 2010 [150]Turkey9334.20 ± 8.890.94 ± 0.26-
Chauveau et al., 2007 [151]France9929.80 ± 7.501.18 ± 0.28-
Johansson et al., 2013 b [152]England5324.30 ± 6.700.97 ± 0.25-
Kalantar-Zadeh et al., 2002 [153]United States3026.40 ± 15.300.88 ± 0.57-
Kim et al., 2015 [154]Korea6321.90 ± 6.700.90 ± 0.30-
Morais et al., 2005 [155]Brazil4420.70 ± 6.701.20 ± 0.60-
Shapiro et al., 2015 [156]United States1325.4 ± 7.41.03 ± 0.32-
Vijayan et al., 2014 [157]India9831.3 0.98-
Abbreviations: BW, body weight; DEI, dietary energy intake; DPI, dietary protein intake; HD, hemodialysis. a Cutoff for dietary inadequacy: DEI < 35 kcal/kg BW/day; DPI < 0.8 g/kg BW/day. b Used ideal body weight for calculation of dietary adequacy.
Table 6. Suboptimal dietary intakes and mortality in HD patients.
Table 6. Suboptimal dietary intakes and mortality in HD patients.
Author/YearCountryPatient No.Follow-UpDEI (kcal/ kg BW)/dayHazard Ratio
(95% CI)
DPI (g/kg BW/day)Hazard Ratio
(95% CI)
SurvivorsNon-SurvivorsSurvivorsNon-Survivors
Antunes et al., 2010 a [159]Brazil7933 (17–38) months25.9
(22.0–29.8)
22.0
(18.0–26.0) b
-1.20
(0.86–1.47)
0.93
(0.90–1.1)
DPI < 1.2g/kg:
4.98 (1.47–16.86) b
Araujo et al., 2006 [33]Brazil34410 years27.4 ± 8.923.5 ± 7.4 b0.96
(0.92–0.99) b
1.01 ± 0.380.92 ± 0.34 c-
Beberashvili et al., 2011 [164]Israel852 years20.8 ± 5.419.1 ± 1.4-0.88 ± 0.240.81 ± 0.10-
Kang et al., 2017 [138]Korea14410 years26.7 ± 5.824.3 ± 4.2 bDEI <25 kcal/kg:
1.86 (1.02–3.40) b
0.91 ± 0.210.82 ± 0.24 bDPI < 0.8g/kg:
1.35 (0.77–2.35)
Abbreviations: BW, body weight; CI, confidence interval; DEI, dietary energy intake; DPI, dietary protein intake; HD, hemodialysis. a Inclusive of HD and peritoneal dialysis patients. b Significant at p < 0.05. c Significant at p < 0.01.
Table 7. Nutritional outcomes associated with anorexia and appetite hormones in HD patients.
Table 7. Nutritional outcomes associated with anorexia and appetite hormones in HD patients.
AssociationsReferences
Poor Appetite
BMI, MAC, MAMC, and MAMABossola et al., 2011 [167]; Sahathevan et al., 2015 [148]
Muscle mass as per BCM, LTM, and LBM index measuresEkramzadeh et al., 2014 [166]; Sahathevan et al., 2015 [148]; Oliveira et al., 2015 [100]
Serum albumin, serum prealbumin and
nPCR/nPNA
Molfino et al., 2015 [177]; Oliveira et al., 2015 [100]; Bossola et al., 2011 [167]; Kalantar-Zadeh et al., 2004 [163]
hsCRP Sahathevan et al., 2015 [148]; Kalantar-Zadeh et al., 2004 [163]
DMS, MIS and PG-SGASahathevan et al., 2015 [148]
Ekramzadeh et al., 2014 [66]; Kalantar-Zadeh et al., 2004 [163]
GNRIOliveira et al., 2015 [100]
Overall food intake of < 50%Molfino et al., 2015 [177]
DEI and DPISahathevan et al., 2015 [148]
Ghrelin
BMIMafra et al., 2010 [175]
Serum albumin and nPNAPerez-Fontan et al., 2004 [178]
MISVanita et al., 2016 [170]
SGAPerez-Fontan et al., 2004 [178]
Leptin
BMI, leptinMontazerifar et al., 2015 [173]
Serum albumin, leptinMontazerifar et al., 2015 [173]
DMS and MIS, leptinKursat et al., 2010 [179]; Ko et al., 2020 [172]
Abbreviations: BCM, body cell mass; BMI, body mass index; DEI, dietary energy intake; DMS, Dialysis Malnutrition Score; DPI, dietary protein intake; GNRI, Geriatric Nutritional Risk Index; HD, hemodialysis; hsCRP, high-sensitivity C-reactive protein; LBM, lean body mass; LTM, lean tissue mass; MAMA, mid-arm muscle area; MAC, mid-arm circumference; MAMC, mid-arm muscle circumference; MIS, malnutrition–inflammation score; nPCR, normalized protein catabolic rate; nPNA, normalized protein nitrogen appearance, PG-SGA, patient-generated subjective global assessment; SGA, subjective global assessment. Notes: decreased trend; increased trend.
Table 8. Impact of psychosocial factors on nutritional outcomes.
Table 8. Impact of psychosocial factors on nutritional outcomes.
AssociationsReferences
Depression
BMI, TSF, and MAMCKoo et al., 2003 [191]
Serum albumin, creatinine,
hemoglobin, nPCR
Koo et al., 2003 [191]; Choi et al., 2012 [192]; Ogrizovic et al., 2008 [193]
Inflammation markersChoi et al., 2012 [192]; Ogrizovic et al., 2008 [193]
SGA Koo et al., 2003 [191]
MISLopes et al., 2017 [194]
QoLNatashia et al., 2019 [195]
Lack of social support
Adherence to dietary restriction Kiajamali et al., 2017 [196]; Kara et al., 2007 [197]; Dilek and Kocaoz, 2015 [198]; Aness et al., 2018 [199]
AppetiteLopes et al., 2007 [200]
QoLUntas et al., 2011 [201]
Financial constraints
Access to purchase foodClark-Cutaia et al., 2018 [202]; Ekramzadeh et al., 2014 [166]
Adherence to dietary restrictionClark-Cutaia et al., 2018 [202]
SGA Freitas et al., 2014 [203]
MISFreitas et al., 2014 [203]
Abbreviations: BMI, body mass index; MAC, mid-arm circumference; MAMC, mid-arm muscle circumference; MIS, malnutrition–inflammation score; nPCR, normalized protein catabolic rate; QoL, qualify of life; SGA, subjective global assessment; TSF, triceps skinfold. Notes: decreased trend; increased trend.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Sahathevan, S.; Khor, B.-H.; Ng, H.-M.; Abdul Gafor, A.H.; Mat Daud, Z.A.; Mafra, D.; Karupaiah, T. Understanding Development of Malnutrition in Hemodialysis Patients: A Narrative Review. Nutrients 2020, 12, 3147. https://doi.org/10.3390/nu12103147

AMA Style

Sahathevan S, Khor B-H, Ng H-M, Abdul Gafor AH, Mat Daud ZA, Mafra D, Karupaiah T. Understanding Development of Malnutrition in Hemodialysis Patients: A Narrative Review. Nutrients. 2020; 12(10):3147. https://doi.org/10.3390/nu12103147

Chicago/Turabian Style

Sahathevan, Sharmela, Ban-Hock Khor, Hi-Ming Ng, Abdul Halim Abdul Gafor, Zulfitri Azuan Mat Daud, Denise Mafra, and Tilakavati Karupaiah. 2020. "Understanding Development of Malnutrition in Hemodialysis Patients: A Narrative Review" Nutrients 12, no. 10: 3147. https://doi.org/10.3390/nu12103147

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop