ABSTRACT
Autosomal dominant polycystic kidney disease (ADPKD) is characterized by the progressive growth of renal cysts, leading to the loss of functional nephrons. Recommendations for individuals with ADPKD to maintain a healthy diet and lifestyle are largely similar to those for the general population. However, recent evidence from preclinical models suggests that more tightly specified dietary regimens, including caloric restriction, intermittent fasting, and ketogenic diets, hold promise to slow disease progression, and the results of ongoing human clinical trials are eagerly awaited. These dietary interventions directly influence nutrient signaling and substrate availability in the cystic kidney, while also conferring systemic metabolic benefits. The present review focuses on the importance of local and systemic metabolism in ADPKD and summarizes current evidence for dietary interventions to slow disease progression and improve quality of life.
Keywords: diet, polycystic kidney, ADPKD, hereditary kidney disease, fasting, time-restricted feeding, time-restricted eating, ketosis, beta-hydroxybutyrate, slow progression
Statement of Significance: We offer the first comprehensive review of the current evidence for dietary interventions to slow ADPKD progression from preclinical animal models and early-stage clinical trials, and elucidate potential common mechanisms underlying their effect.
Introduction
Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic kidney disease. It is characterized by growth of cysts originating in the epithelia of a small proportion of nephrons (1). Over time, cyst growth damages healthy tissue and results in the progressive loss of kidney function, often culminating in end-stage renal disease (2). Disease presentation and progression are highly variable among individuals with ADPKD. Although associations between genotype and disease outcomes are clear, there is still variability in disease severity among individuals sharing the same mutation, and environmental factors amenable to lifestyle modification can meaningfully impact disease progression (1).
The worldwide prevalence of obesity has risen rapidly in recent decades, as have rates of associated metabolic diseases (3). Recently, obesity was found to be associated with a greater rate of kidney enlargement and functional decline in early-stage ADPKD (4). Components of the metabolic syndrome, including hypertension and impaired glucose tolerance, are also associated with more severe ADPKD (5). In addition to accelerating disease progression, obesity may independently increase the kidney pain experienced by those affected by ADPKD, potentially through increased mechanical stress and chronic inflammation. Patients with ADPKD most frequently report pain in the low back (6), and post hoc analysis of the Halt Progression of Polycystic Kidney Disease (HALT-PKD) trials revealed higher BMI is associated with more frequent back pain, independently of height-adjusted kidney and liver volumes (7). As the prevalence of obesity and metabolic disease continues to rise, interventions to slow disease progression and improve quality of life in patients with ADPKD and comorbid obesity are crucially needed.
General guidelines for lifestyle modifications in ADPKD do not currently include recommendations for a particular diet (8). At the same time, a growing body of evidence highlights the role of metabolic alterations in fueling cyst growth. The changes in nutrient signaling pathways and basic metabolism that occur in ADPKD are responsive to the systemic metabolic state, which is determined over time in large part by diet. Importantly, studies in preclinical animal models have shown highly promising effects of slowing disease progression through dietary interventions, including caloric restriction (CR), time-restricted feeding (TRF), and a ketogenic diet (KD). Human data support the existence of metabolic reprogramming in the cystic kidney and overall metabolic health as a modifier of the rate of disease progression, and a number of early-stage clinical trials of dietary interventions in ADPKD are in progress. If proven effective, specific diets may soon become key tools in ADPKD management. Here, we describe possible mechanisms of action and review the current evidence supporting their use.
Metabolic Reprogramming in ADPKD
The kidney has exceptionally high metabolic demands, particularly in the proximal tubule. It is second only to the heart in mitochondrial number (9), and relies heavily on fatty acid oxidation (FAO) for efficient ATP production (10). Alteration of cellular metabolism in cyst-lining epithelial cells is becoming recognized as an important pathophysiologic feature of ADPKD (11–14). Several metabolic abnormalities have been described, notably dependence on aerobic glycolysis and defective FAO. Together, these promote cyst growth at the detriment of healthy tissue function. Understanding these metabolic abnormalities is key to discerning and targeting the intersection of energy metabolism and ADPKD.
Aerobic glycolysis and glucose dependence
The reliance on glycolysis rather than oxidative phosphorylation (OXPHOS) in aerobic conditions, or the Warburg effect, was first described in cancer (15). This change in fuel source preference occurs in highly proliferative cells to support biosynthesis from glycolytic intermediates (16). In the context of ADPKD, aerobic glycolysis was first demonstrated by Rowe et al. (17) who showed increased glucose uptake and conversion to lactate in polycystin-1 (Pkd1) knockout mouse embryonic fibroblasts (MEFs) and in the cystic kidneys of Pkd1 knockout mice using 13C isotope tracing. Upregulation of genes encoding glycolytic enzymes was also observed in the renal cysts of patients with PKD1 mutations (Figure 1, arrow 1). Other studies have corroborated these results. Lactate concentrations were elevated in the cyst fluid of ADPKD patients at the time of transplant (18) (Figure 1, arrow 2). In an orthologous (Pkd1+/-) mini-pig model, expression of glycolytic enzymes [hexokinase (HK) 1, phosphofructokinase 1 (PFK1), pyruvate kinase isoform M2 (PKM2)] was increased, and the activity of electron transport complexes was decreased (19). Moreover, truncation ofPkd2in HEK-293 cells altered mitochondrial morphology and resulted in more rapid acidification of the extracellular medium, suggesting that this mutation resulted in aerobic glycolysis (20). However, 2 independent reports found no increase in glycolysis in Pkd1-deficient kidney tissues or cells (21, 22). Even so, they reported reduced OXPHOS and FAO (21) or upregulation of HK2 (22), results that are consistent with a metabolic shift towards greater reliance on glycolysis (12).
FIGURE 1.
Altered cellular metabolism in ADPKD. Cyst-lining cells exhibit the Warburg effect; aerobic glycolysis is increased (red arrow 1), resulting in the extracellular accumulation of lactate (red arrow 2). In addition to glucose, highly proliferative cells have increased demands for glutamine, reflected by upregulation of GLS (red arrow 3). At the same time, fatty acid oxidation (red arrow 4) and oxidative phosphorylation (red arrow 5) are impaired. ADPKD, autosomal dominant polycystic kidney disease; GLS, glutaminase; GLUD, glutamate dehydrogenase; TCA cycle, tricarboxylic acid cycle; αKG, alpha-ketoglutarate.
The strongest evidence for glucose reliance in Pkd-deficient renal cells comes from experiments using 2-deoxyglucose (2DG). This molecule competitively inhibits the phosphorylation of glucose to glucose-6-phosphate (G6P), thereby inhibiting glycolysis (23). Two days of postnatal treatment with 2DG reduced the weight of kidneys, but not other organs, and lowered the cystic index in 2 orthologous models, one of which was slowly progressive (17). Increased uptake of 13C-glucose and production of 13C-lactate were confirmed in an orthologous and slowly progressive inducible Pkd1 knockout; 2DG treatment for 2 mo reduced kidney volume, leukocyte infiltration, and cystic index (24). Similar improvements in kidney volume and cystic index were reported with 2DG treatment in non-orthologous Han:SPRD-Cy rats (25) and mice with conditional loss of Pkd1 or Pkd2 (26). In a mini-pig model with inducible Pkd1 knockout, 2DG effectively slowed disease, and synergistic effects were observed when 2DG was combined with metformin (27). Co-treatment with low-dose 2DG with metformin in vitro also synergistically inhibited proliferation in cyst-lining epithelia from ADPKD patients (28). An ongoing phase I clinical trial aims to assess the safety and tolerability of 3-mo 2DG treatment in 18 patients with ADPKD (29).
Glutamine dependence
In addition to a greater reliance on glycolysis, proliferating cells have higher glutamine demands to support the increased biosynthesis of amino acids, nucleic acids, and glutathione (16, 30). Glutamine dependence (Figure 1, arrow 3) has been demonstrated in Pkd1-deficient kidneys. Glutamine restriction slowed cystogenesis in explants, and the glutaminase (GLS) inhibitor slowed cyst growth in Pkhd1-Cre;Pkd1fl/fl mice (31, 32), but not in the more aggressive Aqp2-Cre;Pkd1fl/fl model (32). Although glutamine needs were found not to differ between wild-type (WT) and Pkd1del2/del2 MEFs (22), the GLS1-GAC isoform of GLS is upregulated in cyst-lining epithelia of human ADPKD (32). Thus, glutamine dependence is observed in some but not all animal models of ADPKD, and there is some evidence that GLS is upregulated in human ADPKD.
Certain cancers, including glioblastoma and breast cancer, rely on glutamine as a fermentable fuel. Pharmacologic strategies to deplete glutamine have been used in conjunction with limiting dietary glucose intake (KD) in this setting (33, 34). This treatment reduces glycolytic and pentose phosphate pathway flux, both of which are also aberrantly upregulated in ADPKD (13), while at the same time providing ketones as a nonfermentable fuel source for cells with intact mitochondrial OXPHOS (33). Similar to cancer cells, changes in substrate dependence in cyst-lining epithelia may render them susceptible to reduced glutamine and glucose availability (discussed in Ketosis—potential mechanisms).
Defective FAO
A second hallmark of metabolic dysregulation in ADPKD is defective FAO. A lost capacity for mitochondrial B-oxidation, the preferred energy source of the kidney, is a critical step in the pathogenesis of multiple kidney diseases (35). Impaired FAO was inferred from high concentrations of urinary acetylcarnitine in early-onset Pkd1 deletion mice, and OXPHOS was significantly downregulated (36) (Figure 1, arrows 4 and 5). This same study identified transcription factor hepatocyte nuclear factor 4ɑ (HNF-4α), an important regulator of FAO, as a transcriptional network node in ADPKD, and double knockout of Pkd1 and Hnf4a worsened cystic phenotype. Defective FAO in the renal epithelia of Pkd1 knockout mice was later confirmed, a metabolic shift which may preserve lipids for growing cysts (21).
Renal cysts occur in inherited disorders of FAO, further implicating defective FAO in the pathogenesis of ADPKD. These disorders include carnitine palmitoyl transferase (CPT) 2 deficiency (37) and multiple acyl-CoA dehydrogenase deficiency (MADD) (38–40). The prenatal manifestations of these congenital FAO enzyme deficiencies are very similar to polycystic kidney disease (41). Moreover, impaired FAO and peroxisome proliferator-activated receptor γ coactivator 1ɑ (Pgc1α) downregulation have been identified as signatures of renal tubule fibrosis (10, 42), while increased FAO in the tubular epithelia (through overexpression of CPT1A) was protective in 3 fibrosis models (43). Thus, the defective FAO observed in ADPKD may contribute to cystogenesis and fibrosis.
The downregulation of transcription factor peroxisome proliferator activated receptor ɑ (PPARα) is likely to be involved in ADPKD-associated FAO defects [reviewed in (44)]. PPARα downregulation is observed in human cysts (45) and rodent models (46, 47), potentially due to post-transcriptional inhibition by the microRNAs miR-17∼92 and miR-21 (46, 48). Treatment with PPARα agonist fenofibrate improved disease phenotype in both aggressive (46) and slowly progressing ADPKD mice (47), and ameliorated tubulointerstitial fibrosis associated with reduced FAO (10). Importantly, age-associated fibrosis in rats was slowed by CR (49), as circulating nonesterified fatty acids are endogenous PPARα ligands (50). Taken together, the above studies suggest that defects in FAO may contribute to progressive cytogenesis and kidney fibrosis in ADPKD.
Nutrient and inflammatory signaling
Changes in the metabolism of cyst-lining cells are intertwined with aberrant nutrient sensing pathway activity, notably decreases in AMP-activated protein kinase (AMPK) and increases in mammalian target of rapamycin complex 1 (mTORC1) (13). The role of altered nutrient signaling in potentiating cyst growth has made these key targets for therapeutic innovation in ADPKD (12). At a systemic level, reduced AMPK activity is a key feature of metabolic diseases and is associated with chronic inflammation (51). Metabolic reprogramming is also connected to proinflammatory signaling in the cystic kidney. For example, the proinflammatory cytokine macrophage migration inhibitory factor (MIF) activates aerobic glycolysis (52, 53). Deletion of the gene for MIF or pharmacological inhibition of MIF lowers glucose uptake, glycolysis, mammalian target of rapamycin (mTOR) activation, and AMPK inhibition (54) and slows kidney disease progression in multiple Pkd1-deficient models (55). Persistent injury of the renal tissue by cyst growth leads to continued macrophage accumulation and proinflammatory signaling in a maladaptive cycle (54), and rodent studies have established a causal role for macrophage accumulation in ADPKD progression (56–59). Lifestyle modifications to improve systemic metabolic health and reduce chronic inflammation are therefore pertinent in ADPKD.
Dietary Interventions for ADPKD
The majority of the metabolic and signaling pathways that are perturbed in ADPKD are not specific to renal cysts. This can be problematic for drug development due to increased potential for extrarenal side effects (60). However, in cases where obesity and metabolic disease co-occur with ADPKD, these common pathways become potentially important treatment targets, many of which are amenable to dietary interventions. Dietary interventions are relatively low cost and can be combined with drug therapy, permitting a lower dose of drug and thereby reducing side effects (14). This section highlights dietary interventions that aim to challenge the aberrant metabolism contributing to cyst growth while concurrently improving systemic metabolic health.
Caloric restriction
In general, CR has wide-ranging benefits for metabolic health and longevity (61, 62). Two independent groups recently showed impressive attenuation of disease in orthologous mouse models of ADPKD. Kipp et al. (63) found that 23% CR slowed cyst growth and prevented end-stage renal disease in 2 models through inhibition of mTORC1 signaling. Warner et al. (22) further showed dose-dependent reductions in cyst growth in mice with CR of 10–40%, although 2 mo of 20% CR approached 6 mo of 40% CR in efficacy. Furthermore, CR reduced proinflammatory cytokine expression [monocyte chemoattractant protein 1 (MCP-1), IL-6, TNF-ɑ] and restored expression of the glycolytic enzyme HK2 to WT levels. When applied at a later stage, 40% CR reduced the existing cystic burden. The mTORC1-S6K pathway was inhibited and liver kinase B1 (LKB1)–AMPK activated before kidney size changed, suggesting a potential causal role for the key nutrient signaling pathways.
A recently completed clinical trial (NCT03342742; Figure 2) investigated the feasibility and tolerability of a caloric deficit of ∼34% in 30 overweight/obese adults with ADPKD. This is based on novel preliminary data that overweight/obesity are independently associated with more rapid kidney growth, consistent with prior reports (4). Constant CR will be compared to restriction by 80% of caloric needs on 3 d per week, a form of intermittent fasting (IF).
FIGURE 2.
Dietary interventions for ADPKD: preclinical studies and clinical trials to date. NCT03342742 is completed, results not yet published. Note that none of the current clinical trials are sufficiently powered to test therapeutic efficacy. Numbers in parentheses are reference citations. ADPKD, autosomal dominant polycystic kidney disease; CR, caloric restriction; IF, intermittent fasting; KD, ketogenic diet; TRE, time-restricted eating.
Intermittent fasting and time-restricted feeding
Although CR shows promise, its implementation may be challenging due to side effects (64). Adherence to a special diet may be a particular concern, and loss of bone density and lean body mass in the elderly may contribute to increased risks of frailty (65). IF and TRF are alternative dietary interventions that produce many of the same effects through the periodic activation of fasting-responsive systems, such as the AMPK pathway, without requiring constant restriction. The term IF encompasses repeated fasting periods of a day or more [e.g., alternate-day fasting (ADF)], while TRF refers to a restricted daily eating window. For example, 16:8 TRF consists of 16 h of fasting per day and an 8-h ad libitum eating window (64).
An advantage of IF or TRF as compared with traditional CR is that rigorous tracking is not required to achieve reductions in food intake. Caloric intake was spontaneously reduced in trials of TRF [by 20% (66), 300 kcal/d (67), and 550 kcal/d (68)]. Compliance may also be better with IF/TRF due to appetite-reducing effects (69–71). Following an 8-wk trial of ADF or CR in obese adults, participants in the ADF group continued to lose fat and gain lean mass during a 24-wk unsupervised follow-up period, with many indicating they had continued to fast intermittently; the same was not observed in the CR group (72). Continued adherence without monitoring is noteworthy, given that weight-loss maintenance frequently requires extensive support (73). Thus, IF or TRF may be more feasible than CR for chronic conditions, including ADPKD and metabolic disease, where long-term adherence is necessary.
Like CR, IF has an array of demonstrated benefits for metabolic health and longevity. In mice, TRF prevents obesity and metabolic disease caused by a high-fat diet (74) or a high-fat, high-sucrose diet (75). Of note, it is essential that the eating window occurs within the active phase (i.e., during the night in mice, during the day in humans) in order to observe these benefits [reviewed in (76)]. A number of clinical trials of IF showed improvements in metabolic parameters, including blood pressure (77), insulin sensitivity/glucose homeostasis (72, 78, 79), and lipids (72, 77, 80, 81), even on the background of a high-fat diet (82). Clinical trials of TRF showed similar improvements; decreased blood pressure (67) and insulin resistance and oxidative stress (68) have been observed in healthy individuals and in prediabetic men (70), and lipids and glycated hemoglobin (HbA1c) were additionally improved in metabolic syndrome (83). Notably, a daily fast of ∼17 h during Ramadan had no negative effects on kidney function in patients with ADPKD, while proteinuria decreased (84).
There is reason to predict that TRF or IF would slow ADPKD in a similar fashion to CR. Fasting was reported to inhibit aerobic glycolysis and proliferation in colorectal cancer (85). Furthermore, most rodent studies of CR are also, in practice, a form of TRF because the daily ration of food is consumed within a few hours (86). This was noted in a study showing CR efficacy in orthologous ADPKD mice (63), leading the authors to conduct the only preclinical study to date on TRF in ADPKD (87). They showed promising results with 16:8 TRF in the non-orthologous Han:SPRD-cy rat (discussed below in “Preclinical evidence in ADPKD”). Future studies are required to replicate these results in orthologous models and test other IF regimens. A clinical trial of 8-h TRF in overweight or obese adults with ADPKD is currently underway (NCT04534985; Figure 2).
Ketogenic diet
Part of the adaptive response to fasting involves the metabolic switch to production and utilization of ketone bodies like β-hydroxybutyrate (BHB) and acetoacetate. In humans, fasting concentrations of BHB were elevated by 18:6 TRF (88), while BHB concentrations not only surge on fasting days but remain elevated throughout an ADF regimen (81). Ketogenesis can also be induced without fasting or CR through adherence to a high-fat, very-low-carbohydrate KD. Mounting evidence suggests that the KD also increases longevity (89, 90) and may benefit cases of obesity and metabolic disease [reviewed in (91, 92)].
The KD has been used to treat childhood epilepsy since the 1920s (93). From research in epilepsy, the safety and efficacy of the KD have been demonstrated in children (94, 95) and adults (96, 97). Furthermore, a hypocaloric KD has been demonstrated to be safe and effective for weight loss in obese individuals with mild kidney failure (stage 2 chronic kidney disease), nearly a third of whom had improved renal function after the 3-mo intervention (98). These results support the potential safety of the KD as an early intervention in ADPKD or in patients with comorbid obesity and metabolic disease.
The KD may have particular advantages for weight loss compared with conventional CR. In the context of overweight/obesity and type 2 diabetes, a hypocaloric KD was superior to standard CR for weight loss and glycemic control (99, 100). For weight loss in general, a low-fat diet or KD are equally effective given equal caloric intake; however, long-term compliance may be better with a KD due to its appetite-reducing effects (101–104), which counteract the increase in appetite that accompanies weight loss (105, 106). Thus, akin to IF/TRF, the KD presents advantages over conventional CR for weight loss and holds promise for addressing the intersection of ADPKD and obesity.
Preclinical evidence in ADPKD
Recently, Torres and colleagues (87) published evidence that inducing ketosis—whether by KD, TRF, acute fasting, or exogenous ketones—greatly improved the phenotype of 3 animal models of ADPKD. The most extensive evidence was in the non-orthologous Han:SPRD rat model. After 5 wk of isocaloric 16:8 TRF or ad libitum KD, relative kidney weight, cystic index, and epithelial proliferation were reduced; KD additionally reduced fibrosis and mTOR signaling, and increased creatinine clearance. Exogenous BHB (in the drinking water at 157.5 mM concentration) with normal chow produced similar effects. The KD also decreased kidney and cyst size, although not cyst number, in established disease, an effect that has previously only been seen with CR (22) or mTORC1 inhibition using high-dose rapamycin (60, 107).
Remarkably, a single acute 48-h feed deprivation reduced cystic kidney weight proportionately more than the weight changes in other organs or in WT kidneys, and cell death was increased selectively in cyst-lining cells of mutant kidneys (87). Lipid droplets accumulated within the cyst-lining cells, consistent with previous reports of defective FAO (21, 47, 108). Lipid-induced toxicity (lipotoxicity) in cyst-lining cells may be responsible for the increase in cell death. This weakens the epithelial barrier and allows cyst drainage, perhaps explaining the reductions in cyst size but not number. This was supported by increased cyst-lining cell death after 24-h feed deprivation in orthologous (Pkd1) mice, and a decrease in total kidney volume (TKV) and apparent cyst drainage after 72-h feed deprivation in naturally occurring orthologous PKD1 mutant Persian cats. These results are remarkable and need to be replicated independently.
Torres et al. (87) reported a minor increase in BHB concentrations on TRF, while others have reported that CR, but not TRF (1 meal/d), induced ketosis in mice (109). It will be important to distinguish whether the benefits of CR (22, 63) or TRF (87) in ADPKD are indeed primarily mediated by ketosis. Larger studies in orthologous animals are needed. Nevertheless, the results of these preclinical studies are promising and have led to the initiation of clinical trials.
Clinical trials
A phase II trial of the modified Atkins diet (MAD) in patients with rapidly progressive ADPKD is currently underway (110) (Figure 2). The MAD, which, unlike the classical KD, permits unrestricted intake of protein, fluids, and calories, was first introduced to induce ketosis in the outpatient setting (111) and has better compliance compared with the classical KD (112). This follows a small pilot study in which 3 mo of MAD (prescribed 20 g carbohydrate/d) was well tolerated, glycemia decreased over the study period, and BHB concentrations were consistently elevated (113). Transient side effects were minor and predictable (114). Total cholesterol increased, as has been previously reported with the KD (115, 116). However, KD-induced dyslipidemia may resolve in the longer term (117–119), and after 10 y of follow-up, the KD did not alter cardiovascular risk in other clinical settings (120). An independent group is currently recruiting for a trial of ketogenic dietary interventions in 63 patients with rapidly progressive ADPKD (NCT04680780). Patients will be randomly assigned to 3 mo of the classical KD, 3 mo with a 3-d water fast within the first 2 wk of each month, or a control arm. Adherence (ketone concentrations) and subjective feasibility will be measured, along with secondary outcomes including TKV and BMI.
Ketosis—potential mechanisms
Evidence that cystic epithelia are glucose-dependent and success in preclinical models with 2DG administration (reviewed above) suggest that the KD acts, in part, by limiting glucose availability. However, ketosis-inducing interventions (e.g., TRF, oral BHB) did not consistently decrease blood glucose concentrations, even though they improved the phenotype of Han:SPRD rats (87). In CR, serum glucose only trended downwards, and kidney glucose transport protein 4 (GLUT4) expression was upregulated, potentially compensating the reduced serum glucose (22). Nevertheless, in nondiabetic patients with ADPKD, higher serum glucose concentrations predicted a greater yearly increase in height-adjusted TKV (87, 121), suggesting that reducing glucose is advantageous. This has been achieved with the KD or IF/TRF in humans [e.g. (83, 91, 92)], and although BHB supplementation does not alter fasting blood glucose (122), it does lower blood glucose acutely (123) and reduces postprandial glucose excursions in healthy (124) and obese (125) adults.
The KD redirects full-body metabolism away from glucose oxidation, inducing a rapid and lasting switch to fat oxidation (126). As part of this metabolic switch, the KD also activates AMPK (87, 127, 128), induces PPAR⍺ and several of its target genes (129–131), and affects mTORC1 signaling in a tissue-dependent fashion (89, 132). Importantly, ketone bodies provided to kidney cortex ex vivo were used preferentially, providing up to 80% of the respiratory fuel (133). A pilot study with 11C-acetoacetate positron emission tomography confirmed the kidney actively consumes exogenous ketones, with uptake greater than that of the brain (134). Furthermore, BHB may inhibit cyst cell glycolysis, as has been shown in tumor cells (135), neurons (136), and heart and muscle (137). Thus, ketone bodies may sustain healthy tubular epithelia, while inhibition of glycolysis and a build-up of nonfermentable fuels reduce cyst cell growth and survival.
The common feature between CR, fasting, and the KD is elevation of BHB. In addition to providing a nonfermentable fuel to the kidney, BHB has signaling functions. It is one of few ligands for G-protein–coupled receptor GPR109A (138), the activation of which in kidney podocytes reduced inflammation and proteinuria (139); therefore, BHB may attenuate podocyte injury observed in early-stage ADPKD (140). GPR109A also has tumor-suppressor function in multiple tissues (141). Moreover, BHB is an inhibitor of class I histone deacetylases (HDACs 1–3 and 8) (142, 143) and therefore participates in a network of epigenetic regulation in ADPKD [reviewed in (144)]. Treatment with valproic acid, a class I HDAC inhibitor, inhibited cystogenesis in Pkd1 knockout mice (145). Additionally, HDACs deacetylate a number of non-histone proteins including c-MYC (146), the upregulation of which is a hallmark of ADPKD (147), and the tumor suppressors retinoblastoma protein (Rb) and p53 (148).
The regulation of p53 acetylation by BHB may be particularly important. Hypoacetylation of Rb and p53 increased proliferation and decreased cell death of the cystic epithelia, respectively (149). Mice with mutated p53 develop a renal cystic phenotype and metabolic abnormalities reminiscent of PKD (150), and it was recently shown that mTOR signaling contributes to uncontrolled growth in ADPKD cells by causing p53 degradation (151). However, in a small (n = 36) pilot trial of nicotinamide, an inhibitor of the sirtuin HDACs, temporary increases in p53 acetylation reverted within 6–12 mo (152). It is possible that BHB reverses the hypoacetylation of p53 to increase apoptosis of cystic cells. Such a mechanism could contribute to cystic cell death by fasting (87). Indeed, 1 mo of a KD increased acetyl-p53 by 10-fold in the mouse liver (89). On the other hand, BHB also causes β-hydroxybutyrylation (153), a post-translational modification shown in vitro to interfere with p53 acetylation (154). Potential p53-mediated effects of BHB in the polycystic kidney are a topic for future investigation.
BHB may also attenuate oxidative damage, which is increased in PKD and induces cyst growth (155, 156). Several antioxidant systems rely on NAD(P)H as an electron donor (glutathione, thioredoxin), and BHB reduces free cytoplasmic NADP+ to NAD(P)H (157). Additionally, BHB may reduce production of and directly scavenge mitochondrial reactive oxygen species [ROS; reviewed in (158)]. Elevation of BHB (through fasting, CR, or supplementation) protected kidneys against chemically induced oxidative damage (142, 159), in part through activation of antioxidant response regulator nuclear factor-erythroid factor 2–related factor 2 (Nrf2) (159). Nrf2 is also known to positively regulate mitochondrial biogenesis (158, 160), which is disrupted in ADPKD (12, 13). ROS production correlated with disease severity and inversely correlated with NRF2 abundance in human ADPKD; and in an orthologous mouse model, Nrf2 deletion increased, whereas Nrf2 induction decreased ROS production and cyst growth (161).
Other dietary considerations
Dietary fat
In contrast to the apparent benefits of the KD, increased fat intake appears to have detrimental effects in ADPKD when carbohydrate intake is not limited. In Pkd1-/- mice, FAO is dysfunctional, and a minor increase in the fat content of the diet (5.62–7.47%) fed to nursing mothers and young pups resulted in minor but significant increases in relative kidney weight (21). Feeding non-orthologous Han:SPRD-cy rats or pcy mice a diet higher in fat (20 g vs. 5 g/100 g) worsened the renal phenotype (162–164), although the fat intake achieved was much greater than that of a typical human diet (165).
Several studies have also examined the effects of fat source and generally reported improvements with omega-3-rich oils (Table 1). However, these studies were in non-orthologous rodent models, and benefits were not substantiated in orthologous mice (166). Certain fats were thought to confer benefits by altering the profile of oxylipins (bioactive lipid metabolites); however, in orthologous models, dietary effects on oxylipins were found to be independent of disease severity (167). This may be due to their complex role; signaling by the oxylipin prostaglandin E2 (PGE2) stimulated cystogenesis and proliferation in vitro, but its antagonism increased inflammation and worsened disease in Pkd1-deficient mice (168). Although omega-3 PUFAs support overall metabolic health (169, 170), recommendations to consume specific fatty acids for ADPKD are not well supported.
TABLE 1.
The effects of dietary fat composition and level on ADPKD1
| Dietary fat | Duration | Outcomes | References | |
|---|---|---|---|---|
| Non-orthologous | ||||
| Han:SPRD-cy rat | Flax vs. soybean oil | 8 wk | ↓ serum creatinine↓ cyst growth↓ fibrosis↓ macrophage infiltration↓ epithelial proliferation (F only) | Ogborn et al., 1999; 2002; 2006 (171–173) |
| Fish vs. soybean or cottonseed oil | 6 wk | ↓ kidney size↓ fluid content↓cyst volumesvs. cottonseed only:↓ fibrosis↓ inflammation | Lu et al., 2003 (164) | |
| Flax vs. corn oil | 7 wk + perinatal period (fed to mother 2 wk prior to conception until weaning) | ↓ cyst growth↓ epithelial proliferation↓ oxidant injury↓ fibrosis↓ glomerular hypertrophy↓ proteinuria↑ serum urea↑ serum creatinine | Sankaran et al., 2006 (174) | |
| Fish vs. soybean oil | 8 wk | No effect Altered eicosanoids | Ibrahim et al., 2014 (175) | |
| Han:SPRD-cy rat (162, 164),pcy mouse (163) | Fat level:Low (4–5 g/100 g) vs. high (20 g/100 g diet)Soybean oil (162–164)Fish oil (164)Cottonseed oil (164) | 6 wk (162, 164)130 d (163) | ↓ kidney weight (162–164)↓ fluid content (162)↓ cyst score (162)↓ serum urea (162, 163)↓ fibrosis (163, 164)↑ creatinine clearance (M only) (162, 164) | Jayapalan et al., 2000 (162)Lu et al.,2003 (164)Sankaran et al., 2004 (163) |
| pcy Mouse | EPA-enriched vs. sunflower seed vs. standard | 60 d | ↓ tubular dilation↓ cyst area (M only) | Yamaguchi et al., 1990 (176) |
| Fish oil vs. sunflower seed oil | Ad mortem | No effect (on proteinuria or survival) Altered renal fatty acid composition | Aukema et al., 1992 (177) | |
| Flaxseed vs. algal vs. corn oil | 8 wk | Flaxseed oil mitigated increased fibrosis with higher fat level Algal vs. corn oil↑ kidney weight↑ cyst volume | Sankaran et al., 2004 (163) | |
| Orthologous | ||||
| PCK rats (ARPKD) | Soybean/salmon oil blend vs. soybean vs. corn oil | 12 wk | No effect (on kidney weight, cyst size, inflammation, or fibrosis) | Maditz et al., 2013 (165) |
| Pkd2WS25/- mice (ADPKD),PCK rats (ARPKD) | Flax vs. fish vs. soybean oil | 12–13 wk | No benefit Pkd2WS25/-: fish oil↑ kidney size↑ fluid content PCK: fish and flax oil↑ kidney size↑ fluid content↑ cyst area | Yamaguchi et al., 2016 (166) |
| Clinical trial | ||||
| n = 41 patients with ADPKD,age 47 ± 12 y, stage 2–3 CKD | EPA capsules 2.4 g/d vs. no supplement No placebo control | 2 y | No effect (on kidney volume or creatinine clearance) | Higashihara et al., 2008 (178) |
1Benefits of lower fat intake and of replacement with omega-3 fatty acid-rich sources were observed in non-orthologous rodent models. However, these results were not replicated in orthologous models, and a clinical trial of supplemental EPA showed no benefit. ADPKD, autosomal dominant polycystic kidney disease; ARPKD, autosomal recessive polycystic kidney disease; CKD, chronic kidney disease; Pkd2, polycystin 2.
Protein restriction
Studies have also investigated the effect of protein source on ADPKD progression. Most report better outcomes with soy (plant) compared with casein (animal) protein (Table 2). However, this evidence again comes from non-orthologous models and was not replicated in orthologous rodents (166). Furthermore, in a large study of 200 patients with ADPKD, a low-protein diet [0.58 g/(kg/d)] showed no protective effect for a glomerular filtration rate (GFR) range of 25–55 mL/(min/1.73 m²) (179). A study following 589 patients with ADPKD for 4 y found no association between protein intake and annual change in estimated GFR (180).
TABLE 2.
The effects of dietary protein source and level on ADPKD1
| Dietary protein | Duration | Outcomes | References | |
|---|---|---|---|---|
| Non-orthologous | ||||
| Han:SPRD-cy rat | Soy vs. casein | 6–8 wk | ↓ serum creatinine↓ cyst size↓ fibrosis↓ macrophage infiltration↓ epithelial proliferation | Ogborn et al., 1998 & 2000 (181, 182) |
| 6 wk | ↓ kidney weight↓ fluid content↓ cyst size↓ serum urea and creatinine | Aukema & Housini, 2001 (183) | ||
| 1–3 wk | 1 wk:↓ fibrosis↑PGE23 wk:↓ cyst growth | Fair et al., 2004 (184) | ||
| 7 wk + perinatal period (2 wk preconception to weaning) | ↓ macrophage infiltration↓ epithelial proliferation↓ proteinuria | Cahill et al., 2007 (185) | ||
| 4 mo (began at 2 mo of age) | ↓ epithelial proliferation↓ oxidative damage ↓ inflammation | Sankaran et al., 2007 (186) | ||
| 8 wk | ↓ cyst growth↓ fibrosis↓ COX products↑ LOX products | Ibrahim et al., 2014 (175) | ||
| Protein level: high (50%) or low (4%)vs. control (23%) casein | 6 wk | High protein increased, low protein decreased relative kidney weight | Cowley et al., 1996 (187) | |
| pcy Mouse | Protein level: low (6%) vs. control (25%)casein | 105 d | ↓ relative kidney weight↓ fluid content↓ cyst area | Tomobe et al., 1994 (188) |
| Soy vs. casein | 90 d | ↓ relative kidney weight↓ fluid content↓ cyst volume | Tomobe et al., 1998 (189) | |
| 13 wk | Only if low protein (6% vs. 17.4%):↓ relative kidney weight (F only)↓ fluid content↓ cyst score | Aukema et al., 1999 (190) | ||
| Orthologous | ||||
| PCK rat (ARPKD) | Soy vs. casein | 12 wk | No effect (on relative kidney weight, cyst size, inflammation, or fibrosis) | Maditz et al., 2013 (165) |
| Pkd2WS25/- mice (ADPKD),PCK rats (ARPKD) | 12–13 wk | No effect | Yamaguchi et al., 2016 (166) | |
| Clinical trial | ||||
| n = 141 patients withADPKD, GFR13-24 mL/(min/1.73 m²) | Protein level: low-protein diet0.58 g/(kg/d) vs. usual-protein diet 1.3 g/(kg/d) | Mean 2.2 y follow-up | No effect (rate of GFR decline) | Klahr et al., 1995 (179) |
1Benefits of plant vs. animal protein and lower protein intake were observed in non-orthologous rodent models. However, plant protein conferred no benefit in orthologous rodent models, and a low-protein diet did not reduce rate of GFR decline in a clinical trial. ADPKD, autosomal dominant polycystic kidney disease; ARPKD, autosomal recessive polycystic kidney disease; COX, cyclooxygenase; GFR, glomerular filtration rate; LOX, lipoxygenase; PGE2, prostaglandin E2; Pkd2, polycystin 2.
While evidence supporting severe protein restriction is lacking, excessive protein intake may be detrimental. In an orthologous mouse model, supplementation with branched-chain amino acids upregulated mTORC1 and mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) signaling, increasing proliferation of cyst-lining cells (191). Moreover, excessive protein increases systemic acidity, osmolarity, and the nitrogenous waste burden on the kidney. A low osmole diet (protein 0.8–0.9 g/kg and sodium 1500 mg/d) suppressed ambulatory venous presure (AVP) (192, 193), although perhaps not enough to be clinically relevant (194). Considering the available evidence, moderate intake sufficient to meet nutritional needs should be emphasized, and avoidance of excess protein is advisable, as has also been concluded by others (195). This recommendation should be considered when implementing a low-carbohydrate, unrestricted protein diet such as the MAD.
Conclusions
Obesity and metabolic disease are increasingly common conditions with important implications for the progression of ADPKD. Abnormalities in metabolic regulation observed in the cystic kidney share common features with systemic changes observed in individuals with metabolic disease and associated with obesity, including alterations in nutrient-signaling pathways and the activation of chronic inflammation. The intersection of local and systemic metabolic abnormalities is a compelling treatment target in ADPKD with comorbidities such as obesity and/or metabolic disease. The diet is a pivotal point of intervention to this end, with the potential to improve systemic metabolic health while also preferentially targeting cyst-lining cells, which are vulnerable to changes in substrate availability. While investigations of the role of metabolism in ADPKD have focused on abnormalities in basic metabolic processes, including aerobic glycolysis, glutamine dependence, and impaired FAO, evidence from dietary intervention studies underlines the importance of further investigation into the role of ketosis in cyst metabolism and growth. Dietary regimens including CR, IF, and KDs have shown promise in slowing ADPKD progression in animal models. While caution is necessary when interpreting the results of preclinical studies, especially in non-orthologous animal models, clinical trials currently underway promise to improve our understanding of the impact of diet on ADPKD.
Patient adherence may limit the feasibility of a dietary approach to treating ADPKD. However, the majority of patients with ADPKD are highly motivated to implement lifestyle-based interventions, particularly when recommendations are clear and well-supported (196). Questions remain regarding the timing of interventions, although preclinical data suggest benefits even at later stages of disease. Monitoring to ensure adequate nutrition is important, especially in cases of existing renal functional impairment. This further emphasizes the need for dietary interventions to be discussed within the clinical setting to ensure proper formulation.
The implementation of dietary interventions and their mechanisms of action are of particular interest in light of the increasingly common presentation of ADPKD with comorbid obesity or metabolic disease. Indeed, many features of metabolic disease intersect with local metabolic abnormalities in the cystic kidney, pointing to critical pathways for therapeutic intervention.
Although dietary interventions seem especially suited to individuals with ADPKD who would also benefit from weight loss, the main mechanisms of benefit—altered nutrient availability and signaling in the cystic kidney, and systemic improvements in metabolic health—would also be expected to occur in normal-weight individuals. The results of adequately powered clinical trials in normal-weight and obese individuals with ADPKD are eagerly awaited.
Acknowledgments
The authors’ responsibilities were as follows—LP and H-KS: conceived and designed the research; LP: conducted the literature search, drafted the manuscript, and drew the figures; H-KS: reviewed and critically revised the manuscript; LP, I-AI, JS, YP, and H-KS: revised and edited the manuscript; and all authors: read and approved the final manuscript.
Notes
Supported, in part, by the Canadian Institutes of Health Research (CIHR; PJT-162083) of Canada and Sun Life Financial New Investigator Award of Banting & Best Diabetic Centre (BBDC), University of Toronto (to H-KS). Also partly supported by the CIHR Strategy for Patient Oriented Research (SPOR) program grant in Chronic Kidney Disease (CAN-Solve-CKD SCA-145103; to YP).
Author disclosures: YP received compensation for participation in advisory boards for Otsuka, Sanofi-Genzyme, and Reata Pharmaceuticals. The other authors report no conflicts of interest.
Abbreviations used: ADF, alternate-day fasting; ADPKD, autosomal dominant polycystic kidney disease; AMPK, AMP-activated protein kinase; BHB, β-hydroxybutyrate; CPT, carnitine palmitoyltransferase; CR, caloric restriction; FAO, fatty acid oxidation; GFR, glomerular filtration rate; GLS, glutaminase; HDAC, histone deacetylase; HK, hexokinase; Hnf4a/HNF-4ɑ, hepatocyte nuclear factor 4ɑ; IF, intermittent fasting; KD, ketogenic diet; MAD, modified Atkins diet; MEF, mouse embryonic fibroblast; MIF, macrophage migration inhibitory factor; mTOR, mammalian target of rapamycin; mTORC1, mammalian target of rapamycin complex 1; Nrf2, nuclear factor-erythroid factor 2-related factor 2; OXPHOS, oxidative phosphorylation; Pkd, polycystin; PPARa, peroxisome proliferator activated receptor ɑ; Rb, retinoblastoma protein; ROS, reactive oxygen species; TKV, total kidney volume; TRF, time-restricted feeding; WT, wild-type; 2DG, 2-deoxyglucose.
Contributor Information
Lauren Pickel, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada.
Ioan-Andrei Iliuta, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada.
James Scholey, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada; Department of Medicine, University of Toronto, Toronto, Ontario, Canada.
York Pei, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada.
Hoon-Ki Sung, Translational Medicine Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
References
- 1. Cornec-Le Gall E, Alam A, Perrone RD. Autosomal dominant polycystic kidney disease. Lancet North Am Ed. 2019;393(10174):919–35. [DOI] [PubMed] [Google Scholar]
- 2. Spithoven EM, Kramer A, Meijer E, Orskov B, Wanner C, Abad JM, Aresté N, Alonso de la Torre R, Caskey F, Couchoud Cet al. Renal replacement therapy for autosomal dominant polycystic kidney disease (ADPKD) in Europe: prevalence and survival—an analysis of data from the ERA-EDTA Registry. Nephrol Dial Transplant. 2014;29(Suppl 4):iv15–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Blüher M. Obesity: global epidemiology and pathogenesis. Nat Rev Endocrinol. 2019;15(5):288–98. [DOI] [PubMed] [Google Scholar]
- 4. Nowak KL, You Z, Gitomer B, Brosnahan G, Torres VE, Chapman AB, Perrone RD, Steinman TI, Abebe KZ, Rahbari-Oskoui FFet al. Overweight and obesity are predictors of progression in early autosomal dominant polycystic kidney disease. J Am Soc Nephrol. 2018;29(2):571–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Pietrzak-Nowacka M, Safranow K, Palacz J, Gołembiewska E, Marchelek-Myśliwiec M, Ciechanowski K. Association of kidney and cysts dimensions with anthropometric and biochemical parameters in patients with ADPKD. Ren Fail. 2015;37(5):798–803. [DOI] [PubMed] [Google Scholar]
- 6. Hogan MC, Norby SM. Evaluation and management of pain in autosomal dominant polycystic kidney disease. Adv Chronic Kidney Dis. 2010;17(3):e1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7. Nowak KL, Murray K, You Z, Gitomer B, Brosnahan G, Abebe KZ, Braun W, Chapman A, Harris PC, Miskulin Det al. Pain and obesity in autosomal dominant polycystic kidney disease: a post hoc analysis of the Halt Progression of Polycystic Kidney Disease (HALT-PKD) studies. Kidney Med. 2021;3(4):P536–45.E1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Chapman AB, Devuyst O, Eckardt KU, Gansevoort RT, Harris T, Horie S, Kasiske BL, Odland D, Pei Y, Perrone RDet al. Autosomal dominant polycystic kidney disease (ADPKD): executive summary from a Kidney Disease: Improving Global Outcomes (KDIGO) Controversies Conference. Kidney Int. 2015;88(1):17–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Forbes JM. Mitochondria—power players in kidney function?. Trends Endocrinol Metab. 2016;27(7):441–2. [DOI] [PubMed] [Google Scholar]
- 10. Kang HM, Ahn SH, Choi P, Ko YA, Han SH, Chinga F, Park AS, Tao J, Sharma K, Pullman Jet al. Defective fatty acid oxidation in renal tubular epithelial cells plays a key role in kidney fibrosis development. Nat Med. 2015;21(1):37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Menezes LF, Germino GG. The pathobiology of polycystic kidney disease from a metabolic viewpoint. Nat Rev Nephrol. 2019;15(12):735–49. [DOI] [PubMed] [Google Scholar]
- 12. Padovano V, Podrini C, Boletta A, Caplan MJ. Metabolism and mitochondria in polycystic kidney disease research and therapy. Nat Rev Nephrol. 2018;14(11):678–87. [DOI] [PubMed] [Google Scholar]
- 13. Podrini C, Cassina L, Boletta A. Metabolic reprogramming and the role of mitochondria in polycystic kidney disease. Cell Signal. 2020;67:109495. [DOI] [PubMed] [Google Scholar]
- 14. Nowak KL, Hopp K. Metabolic reprogramming in autosomal dominant polycystic kidney disease. Clin J Am Soc Nephrol. 2020;15(4):577–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Warburg O. On the origin of cancer cells. Science. 1956;123(3191):309–14. [DOI] [PubMed] [Google Scholar]
- 16. Lunt SY, Vander Heiden MG. Aerobic glycolysis: meeting the metabolic requirements of cell proliferation. Annu Rev Cell Dev Biol. 2011;27:1, 441–64. [DOI] [PubMed] [Google Scholar]
- 17. Rowe I, Chiaravalli M, Mannella V, Ulisse V, Quilici G, Pema M, Song XW, Xu H, Mari S, Qian Fet al. Defective glucose metabolism in polycystic kidney disease identifies a new therapeutic strategy. Nat Med. 2013;19(4):488–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Foxall PJD, Price RG, Jones JK, Nield GH, Thompson FD, Nicholson JK. High resolution proton magnetic resonance spectroscopy of cyst fluids from patients with polycystic kidney disease. Biochim Biophys Acta BBA Mol Basis Dis. 1992;1138(4):305–14. [DOI] [PubMed] [Google Scholar]
- 19. Lian X, Zhao J, Wu X, Zhang Y, Li Q, Lin S, Bai XY, Chen X. The changes in glucose metabolism and cell proliferation in the kidneys of polycystic kidney disease mini-pig models. Biochem Biophys Res Commun. 2017;488(2):374–81. [DOI] [PubMed] [Google Scholar]
- 20. Chumley P, Zhou J, Mrug S, Chacko B, Parant JM, Challa AK, Wilson LS, Berryhill TF, Barnes S, Kesterson RAet al. Truncating PKHD1 and PKD2 mutations alter energy metabolism. Am J Physiol Ren Physiol. 2019;316(3):F414–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Menezes LF, Lin C-C, Zhou F, Germino GG. Fatty acid oxidation is impaired in an orthologous mouse model of autosomal dominant polycystic kidney disease. EBioMedicine. 2016;5:183–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Warner G, Hein KZ, Nin V, Edwards M, Chini CC, Hopp K, Harris PC, Torres VE, Chini EN. Food restriction ameliorates the development of polycystic kidney disease. J Am Soc Nephrol. 2016;27(5):1437–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Wick AN, Drury DR, Nakada HI, Wolfe JB. Localization of the primary metabolic block produced by 2-deoxyglucose. J Biol Chem. 1957;224(2):963–69. [PubMed] [Google Scholar]
- 24. Chiaravalli M, Rowe I, Mannella V, Quilici G, Canu T, Bianchi V, Gurgone A, Antunes S, D'Adamo P, Esposito Aet al. 2-Deoxy-d-glucose ameliorates PKD progression. J Am Soc Nephrol. 2016;27(7):1958–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Riwanto M, Kapoor S, Rodriguez D, Edenhofer I, Segerer S, Wüthrich RP. Inhibition of aerobic glycolysis attenuates disease progression in polycystic kidney disease. PLoS One. 2016;11(1):e0146654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Nikonova AS, Deneka AY, Kiseleva AA, Korobeynikov V, Gaponova A, Serebriiskii IG, Kopp MC, Hensley HH, Seeger-Nukpezah TN, Somlo Set al. Ganetespib limits ciliation and cystogenesis in autosomal-dominant polycystic kidney disease (ADPKD). FASEB J. 2018;32(5):2735–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Lian X, Wu X, Li Z, Zhang Y, Song K, Cai G, Li Q, Lin S, Chen X, Bai XY. The combination of metformin and 2-deoxyglucose significantly inhibits cyst formation in miniature pigs with polycystic kidney disease. Br J Pharmacol. 2019;176(5):711–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28. Zhao J, Ma Y, Zhang Y, Fu B, Wu X, Li Q, Cai G, Chen X, Bai XY. Low-dose 2-deoxyglucose and metformin synergically inhibit proliferation of human polycystic kidney cells by modulating glucose metabolism. Cell Death Discov. 2019;5(1):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Testa F, Magistroni R. ADPKD current management and ongoing trials. J Nephrol. 2020;33(2):223–37. [DOI] [PubMed] [Google Scholar]
- 30. Choi Y-K, Park K-G. Targeting glutamine metabolism for cancer treatment. Biomol Ther. 2018;26(1):19–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Flowers EM, Sudderth J, Zacharias L, Mernaugh G, Zent R, DeBerardinis RJ, Carroll TJ. Lkb1 deficiency confers glutamine dependency in polycystic kidney disease. Nat Commun. 2018;9(1):814. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Soomro I, Sun Y, Li Z, Diggs L, Hatzivassiliou G, Thomas AG, Rais R, Slusher BS, Somlo S, Skolnik EY. Glutamine metabolism via glutaminase 1 in autosomal-dominant polycystic kidney disease. Nephrol Dial Transplant. 2018;33(8):1343–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Mukherjee P, Augur ZM, Li M, Hill C, Greenwood B, Domin MA, Kondakci G, Narain NR, Kiebish MA, Bronson RTet al. Therapeutic benefit of combining calorie-restricted ketogenic diet and glutamine targeting in late-stage experimental glioblastoma. Commun Biol. 2019;2(1):1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Seyfried TN, Mukherjee P, Iyikesici MS, Slocum A, Kalamian M, Spinosa JP, Chinopoulos C. Consideration of ketogenic metabolic therapy as a complementary or alternative approach for managing breast cancer. Front Nutr. 2020;7:21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Jang H-S, Noh MR, Kim J, Padanilam BJ. Defective mitochondrial fatty acid oxidation and lipotoxicity in kidney diseases. Front Med. 2020;7:65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Menezes LF, Zhou F, Patterson AD, Piontek KB, Krausz KW, Gonzalez FJ, Germino GG. Network Analysis of a Pkd1-mouse model of autosomal dominant polycystic kidney disease identifies HNF4α as a disease modifier. PLos Genet. 2012; 8(11):e1003053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Knottnerus SJ, Bleeker JC, Wüst RC, Ferdinandusse S, IJlst L, Wijburg FA, Wanders RJ, Visser G, Houtkooper RH. Disorders of mitochondrial long-chain fatty acid oxidation and the carnitine shuttle. Rev Endocr Metab Disord. 2018;19(1):93–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Whitfield J, Hurst D, Bennett MJ, Sherwood WG, Hogg R, Gonsoulin W. Fetal polycystic kidney disease associated with glutaric aciduria type II: an inborn error of energy metabolism. Am J Perinatol. 1996;13(03):131–4. [DOI] [PubMed] [Google Scholar]
- 39. Kjaergaard S, Graem N, Larsen T, Skovby F. Recurrent fetal polycystic kidneys associated with glutaric aciduria type II. APMIS. 1998;106(7-12):1188–93. [DOI] [PubMed] [Google Scholar]
- 40. Saudubray JM, Martin D, De Lonlay P, Touati G, Poggi-Travert F, Bonnet D, Jouvert P, Boutron M, Slama A, Vianey-Saban Cet al. Recognition and management of fatty acid oxidation defects: a series of 107 patients. J Inherit Metab Dis. 1999; 22(4):487–502. [DOI] [PubMed] [Google Scholar]
- 41. Hackl A, Mehler K, Gottschalk I, Vierzig A, Eydam M, Hauke J, Beck BB, Liebau MC, Ensenauer R, Weber LTet al. Disorders of fatty acid oxidation and autosomal recessive polycystic kidney disease—different clinical entities and comparable perinatal renal abnormalities. Pediatr Nephrol. 2017;32(5):791–800. [DOI] [PubMed] [Google Scholar]
- 42. Zhang J, Muise ES, Han S, Kutchukian PS, Costet P, Zhu Y, Kan Y, Zhou H, Shah V, Huang Yet al. Molecular profiling reveals a common metabolic signature of tissue fibrosis. Cell Rep Med. 2020;1(4):100056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Miguel V, Tituaña J, Herrero JI, Herrero L, Serra D, Cuevas P, Barbas C, Puyol DR, Márquez-Exposito L, Ruiz-Ortega Met al. Renal tubule Cpt1a overexpression protects from kidney fibrosis by restoring mitochondrial homeostasis. J Clin Invest. 2021;131(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Lakhia R. The role of PPARα in autosomal dominant polycystic kidney disease. Curr Opin Nephrol Hypertens. 2020;29(4):432–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Song X, Di Giovanni V, He N, Wang K, Ingram A, Rosenblum ND, Pei Y. Systems biology of autosomal dominant polycystic kidney disease (ADPKD): computational identification of gene expression pathways and integrated regulatory networks. Hum Mol Genet. 2009;18(13):2328–43. [DOI] [PubMed] [Google Scholar]
- 46. Hajarnis S, Lakhia R, Yheskel M, Williams D, Sorourian M, Liu X, Aboudehen K, Zhang S, Kersjes K, Galasso Ret al. microRNA-17 family promotes polycystic kidney disease progression through modulation of mitochondrial metabolism. Nat Commun. 2017;8(1):14395. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Lakhia R, Yheskel M, Flaten A, Quittner-Strom EB, Holland WL, Patel V. PPARα agonist fenofibrate enhances fatty acid β-oxidation and attenuates polycystic kidney and liver disease in mice. Am J Physiol Renal Physiol. 2018;314(1):F122–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Lakhia R, Hajarnis S, Williams D, Aboudehen K, Yheskel M, Xing C, Hatley ME, Torres VE, Wallace DP, Patel V. MicroRNA-21 aggravates cyst growth in a model of polycystic kidney disease. J Am Soc Nephrol. 2016;27(8):2319–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Chung KW, Lee EK, Lee MK, Oh GT, Yu BP, Chung HY. Impairment of PPARα and the fatty acid oxidation pathway aggravates renal fibrosis during aging. J Am Soc Nephrol. 2018;29(4):1223–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Busato S, Bionaz M. The interplay between non-esterified fatty acids and bovine peroxisome proliferator-activated receptors: results of an in vivo-in vitro hybrid approach. Curr Dev Nutr. 2020;4(Suppl 2):1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Day EA, Ford RJ, Steinberg GR. AMPK as a therapeutic target for treating metabolic diseases. Trends Endocrinol Metab. 2017;28(8):545–60. [DOI] [PubMed] [Google Scholar]
- 52. Toso C, Emamaullee JA, Merani S, Shapiro AMJ. The role of macrophage migration inhibitory factor on glucose metabolism and diabetes. Diabetologia. 2008;51(11):1937–46. [DOI] [PubMed] [Google Scholar]
- 53. Lan H, Luo L, Chen Y, Wang M, Yu Z, Gong Y. MIF signaling blocking alleviates airway inflammation and airway epithelial barrier disruption in a HDM-induced asthma model. Cell Immunol. 2020;347:103965. [DOI] [PubMed] [Google Scholar]
- 54. Weimbs T. Are cyst-associated macrophages in polycystic kidney disease the equivalent to TAMs in cancer?. J Am Soc Nephrol. 2018;29(10):2447–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Chen L, Zhou X, Fan LX, Yao Y, Swenson-Fields KI, Gadjeva M, Wallace DP, Peters DJ, Yu A, Grantham JJet al. Macrophage migration inhibitory factor promotes cyst growth in polycystic kidney disease. J Clin Invest. 2015;125(6):2399–412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Karihaloo A, Koraishy F, Huen SC, Lee Y, Merrick D, Caplan MJ, Somlo S, Cantley LG. Macrophages promote cyst growth in polycystic kidney disease. J Am Soc Nephrol. 2011;22(10):1809–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Swenson-Fields KI, Vivian CJ, Salah SM, Peda JD, Davis BM, Van Rooijen N, Wallace DP, Fields TA. Macrophages promote polycystic kidney disease progression. Kidney Int. 2013;83(5):855–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58. Cassini MF, Kakade VR, Kurtz E, Sulkowski P, Glazer P, Torres R, Somlo S, Cantley LG. Mcp1 promotes macrophage-dependent cyst expansion in autosomal dominant polycystic kidney disease. J Am Soc Nephrol. 2018;29(10):2471–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Viau A, Bienaimé F, Lukas K, Todkar AP, Knoll M, Yakulov TA, Hofherr A, Kretz O, Helmstädter M, Reichardt Wet al. Cilia-localized LKB1 regulates chemokine signaling, macrophage recruitment, and tissue homeostasis in the kidney. EMBO J. 2018;37(15):e98615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Weimbs T, Shillingford JM, Torres J, Kruger SL, Bourgeois BC. Emerging targeted strategies for the treatment of autosomal dominant polycystic kidney disease. Clin Kidney J. 2018;11(Suppl 1):i27–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Fontana L, Partridge L, Longo VD. Dietary restriction, growth factors and aging: from yeast to humans. Science. 2010;328(5976):321–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Picca A, Pesce V, Lezza AMS. Does eating less make you live longer and better? An update on calorie restriction. Clin Interv Aging. 2017;12:1887–902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Kipp KR, Rezaei M, Lin L, Dewey EC, Weimbs T. A mild reduction of food intake slows disease progression in an orthologous mouse model of polycystic kidney disease. Am J Physiol Ren Physiol. 2016;310(8):F726–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Lee JH, Verma N, Thakkar N, Yeung C, Sung H-K. Intermittent fasting: physiological implications on outcomes in mice and men. Physiology. 2020;35(3):185–95. [DOI] [PubMed] [Google Scholar]
- 65. Francesco AD, Germanio CD, Bernier M, Cabo R. A time to fast. Science. 2018;362(6416):770–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Gill S, Panda S. A smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Cell Metab. 2015;22(5):789–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Gabel K, Hoddy KK, Haggerty N, Song J, Kroeger CM, Trepanowski JF, Panda S, Varady KA. Effects of 8-hour time restricted feeding on body weight and metabolic disease risk factors in obese adults: a pilot study. Nutr Healthy Aging. 2018;4(4):345–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. Cienfuegos S, Gabel K, Kalam F, Ezpeleta M, Wiseman E, Pavlou V, Lin S, Oliveira ML, Varady KA. Effects of 4- and 6-h time-restricted feeding on weight and cardiometabolic health: a randomized controlled trial in adults with obesity. Cell Metab. 2020; 32(3):366–78.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Klempel MC, Bhutani S, Fitzgibbon M, Freels S, Varady KA. Dietary and physical activity adaptations to alternate day modified fasting: implications for optimal weight loss. Nutr J. 2010;9(35):1–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Sutton EF, Beyl R, Early KS, Cefalu WT, Ravussin E, Peterson CM. Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 2018;27(6):1212–21, e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Ravussin E, Beyl RA, Poggiogalle E, Hsia DS, Peterson CM.. Early time-restricted feeding reduces appetite and increases fat oxidation but does not affect energy expenditure in humans. Obesity. 2019;27(8):1244–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Catenacci VA, Pan Z, Ostendorf D, Brannon S, Gozansky WS, Mattson MP, Martin B, MacLean PS, Melanson EL, Troy Donahoo W. A randomized pilot study comparing zero-calorie alternate-day fasting to daily caloric restriction in adults with obesity. Obesity. 2016;24(9):1874–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Hall KD, Kahan S. Maintenance of lost weight and long-term management of obesity. Med Clin North Am. 2018;102(1):183–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Hatori M, Vollmers C, Zarrinpar A, DiTacchio L, Bushong EA, Gill S, Leblanc M, Chaix A, Joens M, Fitzpatrick JAet al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 2012;15(6):848–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Chaix A, Zarrinpar A, Miu P, Panda S. Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges. Cell Metab. 2014;20(6):991–1005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Pickel L, Sung H-K. Feeding rhythms and the circadian regulation of metabolism. Front Nutr. 2020;7:39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Varady KA, Bhutani S, Church EC, Klempel MC. Short-term modified alternate-day fasting: a novel dietary strategy for weight loss and cardioprotection in obese adults. Am J Clin Nutr. 2009;90(5):1138–43. [DOI] [PubMed] [Google Scholar]
- 78. Heilbronn LK, Smith SR, Martin CK, Anton SD, Ravussin E. Alternate-day fasting in nonobese subjects: effects on body weight, body composition, and energy metabolism. Am J Clin Nutr. 2005;81(1):69–73. [DOI] [PubMed] [Google Scholar]
- 79. Harvie M, Wright C, Pegington M, McMullan D, Mitchell E, Martin B, Cutler RG, Evans G, Whiteside S, Maudsley Set al. The effect of intermittent energy and carbohydrate restriction v. daily energy restriction on weight loss and metabolic disease risk markers in overweight women. Br J Nutr. 2013;110(8):1534–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Harvie MN, Pegington M, Mattson MP, Frystyk J, Dillon B, Evans G, Cuzick J, Jebb SA, Martin B, Cutler RGet al. The effects of intermittent or continuous energy restriction on weight loss and metabolic disease risk markers: a randomized trial in young overweight women. Int J Obes. 2011;35(5):714–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Stekovic S, Hofer SJ, Tripolt N, Aon MA, Royer P, Pein L, Stadler JT, Pendl T, Prietl B, Url Jet al. Alternate day fasting improves physiological and molecular markers of aging in healthy, non-obese humans. Cell Metab. 2019;30(3):462–76., e6. [DOI] [PubMed] [Google Scholar]
- 82. Klempel MC, Kroeger CM, Varady KA. Alternate day fasting (ADF) with a high-fat diet produces similar weight loss and cardio-protection as ADF with a low-fat diet. Metabolism. 2013;62(1):137–43. [DOI] [PubMed] [Google Scholar]
- 83. Wilkinson MJ, Manoogian EN, Zadourian A, Lo H, Fakhouri S, Shoghi A, Wang X, Fleischer JG, Navlakha S, Panda Set al. Ten-hour time-restricted eating reduces weight, blood pressure, and atherogenic lipids in patients with metabolic syndrome. Cell Metab. 2020;31(1):92–104., e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84. Ekinci I, Erkoc R, Gursu M, Dogan E, Kilic E, Cebeci E, Ozturk S, Kazancioglu R. Effects of fasting during the month of Ramadan on renal function in patients with autosomal dominant polycystic kidney disease. Clin Nephrol. 2018;89(02):103–12. [DOI] [PubMed] [Google Scholar]
- 85. Weng ML, Chen WK, Chen XY, Lu H, Sun ZR, Yu Q, Sun PF, Xu YJ, Zhu MM, Jiang Net al. Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1α pathway suppression. Nat Commun. 2020;11(1):1869. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 86. de Cabo R, Mattson MP. Effects of intermittent fasting on health, aging, and disease. N Engl J Med. 2019;381(26):2541–51. [DOI] [PubMed] [Google Scholar]
- 87. Torres JA, Kruger SL, Broderick C, Amarlkhagva T, Agrawal S, Dodam JR, Mrug M, Lyons LA, Weimbs T. Ketosis ameliorates renal cyst growth in polycystic kidney disease. Cell Metab. 2019;30(6):1007–23., e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Jamshed H, Beyl RA, Della Manna DL, Yang ES, Ravussin E, Peterson CM. Early time-restricted feeding improves 24-hour glucose levels and affects markers of the circadian clock, aging, and autophagy in humans. Nutrients. 2019;11(6):1234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Roberts MN, Wallace MA, Tomilov AA, Zhou Z, Marcotte GR, Tran D, Perez G, Gutierrez-Casado E, Koike S, Knotts TAet al. A ketogenic diet extends longevity and healthspan in adult mice. Cell Metab. 2017;26(3):539–46., e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Newman JC, Covarrubias AJ, Zhao M, Yu X, Gut P, Ng CP, Huang Y, Haldar S, Verdin E. Ketogenic diet reduces mid-life mortality and improves memory in aging mice. Cell Metab. 2017;26(3):547–57, e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Ludwig DS, Willett WC, Volek JS, Neuhouser ML. Dietary fat: from foe to friend?. Science. 2018;362(6416):764–70. [DOI] [PubMed] [Google Scholar]
- 92. Gershuni VM, Yan SL, Medici V. Nutritional ketosis for weight management and reversal of metabolic syndrome. Curr Nutr Rep. 2018;7(3):97–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Wheless JW. History of the ketogenic diet. Epilepsia. 2008;49(Suppl 8):3–5. [DOI] [PubMed] [Google Scholar]
- 94. Rezaei S, Abdurahman AA, Saghazadeh A, Badv RS, Mahmoudi M. Short-term and long-term efficacy of classical ketogenic diet and modified Atkins diet in children and adolescents with epilepsy: a systematic review and meta-analysis. Nutr Neurosci. 2019;22(5):317–34. [DOI] [PubMed] [Google Scholar]
- 95. Arya R, Peariso K, Gaínza-Lein M, Harvey J, Bergin A, Brenton JN, Burrows BT, Glauser T, Goodkin HP, Lai YCet al. Efficacy and safety of ketogenic diet for treatment of pediatric convulsive refractory status epilepticus. Epilepsy Res. 2018;144:1–6. [DOI] [PubMed] [Google Scholar]
- 96. Liu H, Yang YI, Wang Y, Tang H, Zhang F, Zhang Y, Zhao Y. Ketogenic diet for treatment of intractable epilepsy in adults: a meta-analysis of observational studies. Epilepsia Open. 2018;3(1):9–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Green SF, Nguyen P, Kaalund-Hansen K, Rajakulendran S, Murphy E. Effectiveness, retention, and safety of modified ketogenic diet in adults with epilepsy at a tertiary-care centre in the UK. J Neurol. 2020;267(4):1171–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Bruci A, Tuccinardi D, Tozzi R, Balena A, Santucci S, Frontani R, Mariani S, Basciani S, Spera G, Gnessi Let al. Very low-calorie ketogenic diet: a safe and effective tool for weight loss in patients with obesity and mild kidney failure. Nutrients. 2020;12(2):333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99. Hussain TA, Mathew TC, Dashti AA, Asfar S, Al-Zaid N, Dashti HM. Effect of low-calorie versus low-carbohydrate ketogenic diet in type 2 diabetes. Nutrition. 2012;28(10):1016–21. [DOI] [PubMed] [Google Scholar]
- 100. Goday A, Bellido D, Sajoux I, Crujeiras AB, Burguera B, García-Luna PP, Oleaga A, Moreno B, Casanueva FF. Short-term safety, tolerability and efficacy of a very low-calorie-ketogenic diet interventional weight loss program versus hypocaloric diet in patients with type 2 diabetes mellitus. Nutr Diabetes. 2016;6(9):e230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Foster GD, Wyatt HR, Hill JO, McGuckin BG, Brill C, Mohammed BS, Szapary PO, Rader DJ, Edman JS, Klein S. A randomized trial of a low-carbohydrate diet for obesity. N Engl J Med. 2003;348(21):2082–90. [DOI] [PubMed] [Google Scholar]
- 102. Mohorko N, Černelič-Bizjak M, Poklar-Vatovec T, Grom G, Kenig S, Petelin A, Jenko-Pražnikar Z. Weight loss, improved physical performance, cognitive function, eating behavior, and metabolic profile in a 12-week ketogenic diet in obese adults. Nutr Res. 2019;62:64–77. [DOI] [PubMed] [Google Scholar]
- 103. Deemer SE, Plaisance EP, Martins C. Impact of ketosis on appetite regulation—a review. Nutr Res. 2020;77:1–11. [DOI] [PubMed] [Google Scholar]
- 104. Kirkpatrick CF, Bolick JP, Kris-Etherton PM, Sikand G, Aspry KE, Soffer D E, Willard KE, Maki KC. Review of current evidence and clinical recommendations on the effects of low-carbohydrate and very-low-carbohydrate (including ketogenic) diets for the management of body weight and other cardiometabolic risk factors: a scientific statement from the National Lipid Association Nutrition and Lifestyle Task Force. J Clin Lipidol. 2019;13(5):689–711., e1. [DOI] [PubMed] [Google Scholar]
- 105. Gibson AA, Seimon RV, Lee CM, Ayre J, Franklin J, Markovic TP, Caterson ID, Sainsbury A. Do ketogenic diets really suppress appetite? A systematic review and meta-analysis. Obes Rev. 2015;16(1):64–76. [DOI] [PubMed] [Google Scholar]
- 106. Nymo S, Coutinho SR, Jørgensen J, Rehfeld JF, Truby H, Kulseng B, Martins C. Timeline of changes in appetite during weight loss with a ketogenic diet. Int J Obes. 2017;41(8):1224–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Shillingford JM, Piontek KB, Germino GG, Weimbs T. Rapamycin ameliorates PKD resulting from conditional inactivation of Pkd1. J Am Soc Nephrol. 2010;21(3):489–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Lin CC, Kurashige M, Liu Y, Terabayashi T, Ishimoto Y, Wang T, Choudhary V, Hobbs R, Liu LK, Lee PHet al. A cleavage product of polycystin-1 is a mitochondrial matrix protein that affects mitochondria morphology and function when heterologously expressed. Sci Rep. 2018;8(1):2743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Mitchell SJ, Bernier M, Mattison JA, Aon MA, Kaiser TA, Anson RM, Ikeno Y, Anderson RM, Ingram DK, de Cabo R. Daily fasting improves health and survival in male mice independent of diet composition and calories. Cell Metab. 2019;29(1):221–8., e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Testa F, Marchiò M, D'Amico R, Giovanella S, Ligabue G, Fontana F, Alfano G, Cappelli G, Biagini G, Magistroni R. GREASE II. A phase II randomized, 12-month, parallel-group, superiority study to evaluate the efficacy of a modified Atkins diet in autosomal dominant polycystic kidney disease patients. PharmaNutrition. 2020;13:100206. [Google Scholar]
- 111. Kossoff EH, McGrogan JR, Bluml RM, Pillas DJ, Rubenstein JE, Vining EP. A modified Atkins diet is effective for the treatment of intractable pediatric epilepsy. Epilepsia. 2006;47(2):421–4. [DOI] [PubMed] [Google Scholar]
- 112. Ye F, Li X-J, Jiang W-L, Sun H-B, Liu J. Efficacy of and patient compliance with a ketogenic diet in adults with intractable epilepsy: a meta-analysis. J Clin Neurol. 2015;11(1):26–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Testa F, Marchiò M, Belli M, Giovanella S, Ligabue G, Cappelli G, Biagini G, Magistroni R. A pilot study to evaluate tolerability and safety of a modified Atkins diet in ADPKD patients. PharmaNutrition. 2019;9:100154. [Google Scholar]
- 114. Bostock ECS, Kirkby KC, Taylor BV, Hawrelak JA. Consumer reports of “keto flu” associated with the ketogenic diet. Front Nutr. 2020;7:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Özdemir R, Güzel O, Küçük M, Karadeniz C, Katipoglu N, Yılmaz Ü, Yılmazer MM, Meşe T. The effect of the ketogenic diet on the vascular structure and functions in children with intractable epilepsy. Pediatr Neurol. 2016;56:30–4. [DOI] [PubMed] [Google Scholar]
- 116. Azevedo de Lima P, Baldini Prudêncio M, Murakami DK, Pereira de Brito Sampaio L, Figueiredo Neto AM, Teixeira Damasceno NR. Effect of classic ketogenic diet treatment on lipoprotein subfractions in children and adolescents with refractory epilepsy. Nutrition. 2017;33:271–7. [DOI] [PubMed] [Google Scholar]
- 117. Dashti HM, Mathew TC, Hussein T, Asfar SK, Behbahani A, Khoursheed MA, Al-Sayer HM, Bo-Abbas YY, Al-Zaid NS. Long-term effects of a ketogenic diet in obese patients. Exp Clin Cardiol. 2004;9(3):200–5. [PMC free article] [PubMed] [Google Scholar]
- 118. Peter O, Kwiterovich J, Vining EPG, Pyzik P, Richard Skolasky J, Freeman JM. Effect of a high-fat ketogenic diet on plasma levels of lipids, lipoproteins, and apolipoproteins in children. JAMA. 2003;290(7):912–20. [DOI] [PubMed] [Google Scholar]
- 119. Kapetanakis M, Liuba P, Odermarsky M, Lundgren J, Hallböök T. Effects of ketogenic diet on vascular function. Eur J Paediatr Neurol. 2014;18(4):489–494. [DOI] [PubMed] [Google Scholar]
- 120. Heussinger N, Della Marina A, Beyerlein A, Leiendecker B, Hermann-Alves S, Dalla Pozza R, Klepper J. 10 patients, 10 years—long term follow-up of cardiovascular risk factors in Glut1 deficiency treated with ketogenic diet therapies: a prospective, multicenter case series. Clin Nutr. 2018;37(6):2246–51. [DOI] [PubMed] [Google Scholar]
- 121. Alan SL, Shen C, Landsittel DP, Grantham JJ, Cook LT, Torres VE, Chapman AB, Bae KT, Mrug M, Harris PCet al. Long-term trajectory of kidney function in autosomal-dominant polycystic kidney disease. Kidney Int. 2019;95(5):1253–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Soto-Mota A, Vansant H, Evans RD, Clarke K. Safety and tolerability of sustained exogenous ketosis using ketone monoester drinks for 28 days in healthy adults. Regul Toxicol Pharmacol. 2019;109:104506. [DOI] [PubMed] [Google Scholar]
- 123. Stubbs BJ, Cox PJ, Evans RD, Santer P, Miller JJ, Faull OK, Magor-Elliott S, Hiyama S, Stirling M, Clarke K. On the metabolism of exogenous ketones in humans. Front Physiol. 2017;8:848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Myette-Côté É, Neudorf H, Rafiei H, Clarke K, Little JP. Prior ingestion of exogenous ketone monoester attenuates the glycaemic response to an oral glucose tolerance test in healthy young individuals. J Physiol. 2018;596(8):1385–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Myette-Côté É, Caldwell HG, Ainslie PN, Clarke K, Little JP.. A ketone monoester drink reduces the glycemic response to an oral glucose challenge in individuals with obesity: a randomized trial. Am J Clin Nutr. 2019;110(6):1491–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Hall KD, Chen KY, Guo J, Lam YY, Leibel RL, Mayer LE, Reitman ML, Rosenbaum M, Smith SR, Walsh BTet al. Energy expenditure and body composition changes after an isocaloric ketogenic diet in overweight and obese men. Am J Clin Nutr. 2016;104(2):324–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127. Kennedy AR, Pissios P, Otu H, Xue B, Asakura K, Furukawa N, Marino FE, Liu FF, Kahn BB, Libermann TAet al. A high-fat, ketogenic diet induces a unique metabolic state in mice. Am J Physiol Endocrinol Metab. 2007;292(6):E1724–39. [DOI] [PubMed] [Google Scholar]
- 128. Bae HR, Kim DH, Park MH, Lee B, Kim MJ, Lee EK, Chung KW, Kim SM, Im DS, Chung HY. β-Hydroxybutyrate suppresses inflammasome formation by ameliorating endoplasmic reticulum stress via AMPK activation. Oncotarget. 2016;7(41):66444–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Badman MK, Pissios P, Kennedy AR, Koukos G, Flier JS, Maratos-Flier E. Hepatic fibroblast growth factor 21 is regulated by PPARα and is a key mediator of hepatic lipid metabolism in ketotic states. Cell Metab. 2007;5(6):426–37. [DOI] [PubMed] [Google Scholar]
- 130. Grabacka M, Pierzchalska M, Dean M, Reiss K. Regulation of ketone body metabolism and the role of PPARα. Int J Mol Sci. 2016;17(12):2093. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Longo R, Peri C, Cricrì D, Coppi L, Caruso D, Mitro N, De Fabiani E, Crestani M. Ketogenic diet: a new light shining on old but gold biochemistry. Nutrients. 2019;11(10):2497. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. McDaniel SS, Rensing NR, Thio LL, Yamada KA, Wong M. The ketogenic diet inhibits the mammalian target of rapamycin (mTOR) pathway. Epilepsia. 2011;52(3):e7–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Weidemann MJ, Krebs HA. The fuel of respiration of rat kidney cortex. Biochem J. 1969;112(2):149–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. Cuenoud B, Hartweg M, Godin JP, Croteau E, Maltais M, Castellano CA, Carpentier AC, Cunnane SC. Metabolism of exogenous D-beta-hydroxybutyrate, an energy substrate avidly consumed by the heart and kidney. Front Nutr. 2020;7:13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Shukla SK, Gebregiworgis T, Purohit V, Chaika NV, Gunda V, Radhakrishnan P, Mehla K, Pipinos II, Powers R, Yu Fet al. Metabolic reprogramming induced by ketone bodies diminishes pancreatic cancer cachexia. Cancer Metab. 2014;2:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136. Lund TM, Ploug KB, Iversen A, Jensen AA, Jansen-Olesen I. The metabolic impact of β-hydroxybutyrate on neurotransmission: reduced glycolysis mediates changes in calcium responses and KATP channel receptor sensitivity. J Neurochem. 2015;132(5):520–31. [DOI] [PubMed] [Google Scholar]
- 137. Cox PJ, Kirk T, Ashmore T, Willerton K, Evans R, Smith A, Murray AJ, Stubbs B, West J, McLure SW, King MT. Nutritional ketosis alters fuel preference and thereby endurance performance in athletes. Cell Metab. 2016;24(2):256–68. [DOI] [PubMed] [Google Scholar]
- 138. Graff EC, Fang H, Wanders D, Judd RL. Anti-inflammatory effects of the hydroxycarboxylic acid receptor 2. Metabolism. 2016;65(2):102–13. [DOI] [PubMed] [Google Scholar]
- 139. Felizardo RJ, de Almeida DC, Pereira RL, Watanabe IK, Doimo NT, Ribeiro WR, Cenedeze MA, Hiyane MI, Amano MT, Braga TTet al. Gut microbial metabolite butyrate protects against proteinuric kidney disease through epigenetic- and GPR109a-mediated mechanisms. FASEB J. 2019;33(11):11894–908. [DOI] [PubMed] [Google Scholar]
- 140. Cebeci E, Ekinci I, Gursu M, Coskun C, Karadag S, Uzun S, Behlul A, Senel TE, Kazancioglu R, Ozturk S. Podocyte injury in autosomal dominant polycystic kidney disease. Nephron. 2019;142(4):311–9. [DOI] [PubMed] [Google Scholar]
- 141. Ristic B, Bhutia YD, Ganapathy V. Cell-surface G-protein-coupled receptors for tumor-associated metabolites: a direct link to mitochondrial dysfunction in cancer. Biochim Biophys Acta. 2017;1868(1):246–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142. Shimazu T, Hirschey MD, Newman J, He W, Shirakawa K, Le Moan N, Grueter CA, Lim H, Saunders LR, Stevens RDet al. Suppression of oxidative stress by β-hydroxybutyrate, an endogenous histone deacetylase inhibitor. Science. 2013;339(6116):211–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143. Newman JC, Verdin E. β-Hydroxybutyrate: a signaling metabolite. Annu Rev Nutr. 2017;37(1):51–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144. Li X. Epigenetics and cell cycle regulation in cystogenesis. Cell Signal. 2020;68:109509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145. Cao Y, Semanchik N, Lee SH, Somlo S, Barbano PE, Coifman R, Sun Z. Chemical modifier screen identifies HDAC inhibitors as suppressors of PKD models. Proc Natl Acad Sci. 2009;106(51):21819–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146. Glozak MA, Sengupta N, Zhang X, Seto E. Acetylation and deacetylation of non-histone proteins. Gene. 2005;363:15–23. [DOI] [PubMed] [Google Scholar]
- 147. Kurbegovic A, Trudel M. The master regulators Myc and p53 cellular signaling and functions in polycystic kidney disease. Cell Signal. 2020;71:109594. [DOI] [PubMed] [Google Scholar]
- 148. Mrakovcic M, Kleinheinz J, Fröhlich LF. p53 at the crossroads between different types of HDAC inhibitor-mediated cancer cell death. Int J Mol Sci. 2019;20(10):2415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149. Zhou X, Fan LX, Sweeney WE, Denu JM, Avner ED, Li X. Sirtuin 1 inhibition delays cyst formation in autosomal-dominant polycystic kidney disease. J Clin Invest. 2013;123(7):3084–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Li H, Zhang Y, Dan J, Zhou R, Li C, Li R, Wu X, Singh SK, Chang JT, Yang Jet al. p53 mutation regulates PKD genes and results in co-occurrence of PKD and tumorigenesis. Cancer Biol Med. 2019;16(1):79–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151. De Stephanis L, Mangolini A, Servello M, Harris PC, Dell'Atti L, Pinton P, Aguiari G. MicroRNA501-5p induces p53 proteasome degradation through the activation of the mTOR/MDM2 pathway in ADPKD cells. J Cell Physiol. 2018;233(9):6911–24. [DOI] [PubMed] [Google Scholar]
- 152. El Ters M, Zhou X, Lepping RJ, Lu P, Karcher RT, Mahnken JD, Brooks WM, Winklhofer FT, Li Xet al. Biological efficacy and safety of niacinamide in patients with ADPKD. Kidney Int Rep. 2020;5(8):1271–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Xie Z, Zhang D, Chung D, Tang Z, Huang H, Dai L, Qi S, Li J, Colak G, Chen Yet al. Metabolic regulation of gene expression by histone lysine β-hydroxybutyrylation. Mol Cell. 2016;62(2):194–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154. Liu K, Li F, Sun Q, Lin N, Han H, You K, Tian F, Mao Z, Li T, Tong Tet al. p53 β-hydroxybutyrylation attenuates p53 activity. Cell Death Dis. 2019;10(3):1–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. Maser RL, Vassmer D, Magenheimer BS, Calvet JP. Oxidant stress and reduced antioxidant enzyme protection in polycystic kidney disease. J Am Soc Nephrol. 2002;13(4):991–9. [DOI] [PubMed] [Google Scholar]
- 156. Schreiber R, Buchholz B, Kraus A, Schley G, Scholz J, Ousingsawat J, Kunzelmann K. Lipid peroxidation drives renal cyst growth in vitro through activation of TMEM16A. J Am Soc Nephrol. 2019;30(2):228–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157. Veech RL, King MT, Pawlosky R, Kashiwaya Y, Bradshaw PC, Curtis W. The “great” controlling nucleotide coenzymes. IUBMB Life. 2019;71(5):565–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Miller VJ, Villamena FA, Volek JS. Nutritional ketosis and mitohormesis: potential implications for mitochondrial function and human health. J Nutr Metab. 2018;2018:5157645. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Wei T, Tian W, Liu F, Xie G. Protective effects of exogenous β-hydroxybutyrate on paraquat toxicity in rat kidney. Biochem Biophys Res Commun. 2014;447(4):666–71. [DOI] [PubMed] [Google Scholar]
- 160. Edwards C, Canfield J, Copes N, Rehan M, Lipps D, Bradshaw PC. D-beta-hydroxybutyrate extends lifespan in C. elegans. Aging. 2014;6(8):621–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Lu Y, Sun Y, Liu Z, Lu Y, Zhu X, Lan B, Mi Z, Dang L, Li N, Zhan Wet al. Activation of NRF2 ameliorates oxidative stress and cystogenesis in autosomal dominant polycystic kidney disease. Sci Transl Med. 2020;12(554):eaba3613. [DOI] [PubMed] [Google Scholar]
- 162. Jayapalan S, Saboorian MH, Edmunds JW, Aukema HM. High dietary fat intake increases renal cyst disease progression in Han:SPRD-cy rats. J Nutr. 2000;130(9):2356–60. [DOI] [PubMed] [Google Scholar]
- 163. Sankaran D, Lu J, Bankovic-Calic N, Ogborn MR, Aukema HM. Modulation of renal injury in pcy mice by dietary fat containing n−3 fatty acids depends on the level and type of fat. Lipids. 2004;39(3):207–14. [DOI] [PubMed] [Google Scholar]
- 164. Lu J, Bankovic-Calic N, Ogborn M, Saboorian MH, Aukema HM. Detrimental effects of a high fat diet in early renal injury are ameliorated by fish oil in Han:SPRD-cy rats. J Nutr. 2003;133(1):180–6. [DOI] [PubMed] [Google Scholar]
- 165. Maditz KH, Gigliotti JC, Tou JC. Evidence for a role of proteins, lipids, and phytochemicals in the prevention of polycystic kidney disease progression and severity. Nutr Rev. 2013;71(12):802–14. [DOI] [PubMed] [Google Scholar]
- 166. Yamaguchi T, Devassy JG, Monirujjaman M, Gabbs M, Aukema HM. Lack of benefit of early intervention with dietary flax and fish oil and soy protein in orthologous rodent models of human hereditary polycystic kidney disease. PLoS One. 2016;11(5):e0155790. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Devassy JG, Yamaguchi T, Monirujjaman M, Gabbs M, Ravandi A, Zhou J, Aukema HM. Distinct effects of dietary flax compared to fish oil, soy protein compared to casein, and sex on the renal oxylipin profile in models of polycystic kidney disease. Prostaglandins Leukotrienes Essent Fatty Acids. 2017;123:1–13. [DOI] [PubMed] [Google Scholar]
- 168. Lannoy M, Valluru MK, Chang L, Abdela-Ali F, Peters DJ, Streets AJ, Ong AC. The positive effect of selective prostaglandin E2 receptor EP2 and EP4 blockade on cystogenesis in vitro is counteracted by increased kidney inflammation in vivo. Kidney Int. 2020;98(2):404–19. [DOI] [PubMed] [Google Scholar]
- 169. Albracht-Schulte K, Kalupahana NS, Ramalingam L, Wang S, Rahman SM, Robert-McComb J, Moustaid-Moussa N. Omega-3 fatty acids in obesity and metabolic syndrome: a mechanistic update. J Nutr Biochem. 2018;58:1–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170. Jang H, Park K. Omega-3 and omega-6 polyunsaturated fatty acids and metabolic syndrome: A systematic review and meta-analysis. Clin Nutr. 2020;39(3):765–773. [DOI] [PubMed] [Google Scholar]
- 171. Ogborn MR, Nitschmann E, Weiler H, Leswick D, Bankovic-Calic N. Flaxseed ameliorates interstitial nephritis in rat polycystic kidney disease. Kidney Int. 1999;55(2):417–23. [DOI] [PubMed] [Google Scholar]
- 172. Ogborn MR, Nitschmann E, Bankovic-Calic N, Weiler HA, Aukema H. Dietary flax oil reduces renal injury, oxidized LDL content, and tissue n-6/n-3 FA ratio in experimental polycystic kidney disease. Lipids. 2002;37(11):1059–65. [DOI] [PubMed] [Google Scholar]
- 173. Ogborn MR, Nitschmann E, Bankovic-Calic N, Weiler HA, Aukema HM. Effects of flaxseed derivatives in experimental polycystic kidney disease vary with animal gender. Lipids. 2006;41(12):1141–9. [DOI] [PubMed] [Google Scholar]
- 174. Sankaran D, Bankovic-Calic N, Peng CY-C, Ogborn MR, Aukema HM. Dietary flax oil during pregnancy and lactation retards disease progression in rat offspring with inherited kidney disease. Pediatr Res. 2006;60(6):729–33. [DOI] [PubMed] [Google Scholar]
- 175. Ibrahim NH, Jia Y, Devassy JG, Yamaguchi T, Aukema HM. Renal cyclooxygenase and lipoxygenase products are altered in polycystic kidneys and by dietary soy protein and fish oil treatment in the Han:SPRD-Cy rat. Mol Nutr Food Res. 2014;58(4):768–81. [DOI] [PubMed] [Google Scholar]
- 176. Yamaguchi T, Valli VE, Philbrick D, Holub B, Yoshida K, Takahashi H. Effects of dietary supplementation with n-3 fatty acids on kidney morphology and the fatty acid composition of phospholipids and triglycerides from mice with polycystic kidney disease. Res Commun Chem Pathol Pharmacol. 1990;69(3):335–51. [PubMed] [Google Scholar]
- 177. Aukema HM, Yamaguchi T, Takahashi H, Philbrick DJ, Holub BJ. Effects of dietary fish oil on survival and renal fatty acid composition in murine polycystic kidney disease. Nutr Res. 1992;12(11):1383–92. [Google Scholar]
- 178. Higashihara E, Nutahara K, Horie S, Muto S, Hosoya T, Hanaoka K, Tuchiya K, Kamura K, Takaichi K, Ubara Yet al. The effect of eicosapentaenoic acid on renal function and volume in patients with ADPKD. Nephrol Dial Transplant. 2008;23(9):2847–52. [DOI] [PubMed] [Google Scholar]
- 179. Klahr S, Breyer JA, Beck GJ, Dennis VW, Hartman JA, Roth D, Steinman TI, Wang SR, Yamamoto ME. Dietary protein restriction, blood pressure control, and the progression of polycystic kidney disease. Modification of Diet in Renal Disease Study Group. J Am Soc Nephrol. 1995;5(12):2037–47. [DOI] [PubMed] [Google Scholar]
- 180. Kramers BJ, Koorevaar IW, Drenth JP, de Fijter JW, Neto AG, Peters DJ, Vart P, Wetzels JF, Zietse R, Gansevoort RTet al. Salt, but not protein intake, is associated with accelerated disease progression in autosomal dominant polycystic kidney disease. Kidney Int. 2020;98(4):989–98. [DOI] [PubMed] [Google Scholar]
- 181. Ogborn MR, Bankovic-Calic N, Shoesmith C, Buist R, Peeling J. Soy protein modification of rat polycystic kidney disease. Am J Physiol. 1998;274(3):F541–9. [DOI] [PubMed] [Google Scholar]
- 182. Ogborn MR, Nitschmann E, Weiler HA, Bankovic-Calic N. Modification of polycystic kidney disease and fatty acid status by soy protein diet. Kidney Int. 2000;57(1):159–66. [DOI] [PubMed] [Google Scholar]
- 183. Aukema HM, Housini I. Dietary soy protein effects on disease and IGF-I in male and female Han:SPRD-cy rats. Kidney Int. 2001;59(1):52–61. [DOI] [PubMed] [Google Scholar]
- 184. Fair D E, Ogborn MR, Weiler HA, Bankovic-Calic N, Nitschmann EP, Fitzpatrick-Wong SC, Aukema HM. Dietary soy protein attenuates renal disease progression after 1 and 3 weeks in Han:SPRD-cy weanling rats. J Nutr. 2004;134(6):1504–7. [DOI] [PubMed] [Google Scholar]
- 185. Cahill LE, Peng CY-C, Bankovic-Calic N, Sankaran D, Ogborn MR, Aukema HM. Dietary soya protein during pregnancy and lactation in rats with hereditary kidney disease attenuates disease progression in offspring. Br J Nutr. 2007;97(1):77–84. [DOI] [PubMed] [Google Scholar]
- 186. Sankaran D, Bankovic-Calic N, Cahill L, Yu-Chen Peng C, Ogborn MR, Aukema HM. Late dietary intervention limits benefits of soy protein or flax oil in experimental polycystic kidney disease. Nephron Exp Nephrol. 2007;106(4):e122–8. [DOI] [PubMed] [Google Scholar]
- 187. Cowley BD, Grantham JJ, Muessel MJ, Kraybill AL, Gattone VH. Modification of disease progression in rats with inherited polycystic kidney disease. Am J Kidney Dis. 1996;27(6):865–79. [DOI] [PubMed] [Google Scholar]
- 188. Tomobe K, Philbrick D, Aukema HM, Clark WF, Ogborn MR, Parbtani A, Takahashi H, Holub BJ. Early dietary protein restriction slows disease progression and lengthens survival in mice with polycystic kidney disease. J Am Soc Nephrol. 1994;5(6):1355–60. [DOI] [PubMed] [Google Scholar]
- 189. Tomobe K, Philbrick DJ, Ogborn MR, Takahashi H, Holub BJ. Effect of dietary soy protein and genistein on disease progression in mice with polycystic kidney disease. Am J Kidney Dis. 1998;31(1):55–61. [DOI] [PubMed] [Google Scholar]
- 190. Aukema HM, Housini I, Rawling JM. Dietary soy protein effects on inherited polycystic kidney disease are influenced by gender and protein level. J Am Soc Nephrol. 1999;10(2):300–8. [DOI] [PubMed] [Google Scholar]
- 191. Yamamoto J, Nishio S, Hattanda F, Nakazawa D, Kimura T, Sata M, Makita M, Ishikawa Y, Atsumi T. Branched-chain amino acids enhance cyst development in autosomal dominant polycystic kidney disease. Kidney Int. 2017;92(2):377–87. [DOI] [PubMed] [Google Scholar]
- 192. Taylor JM, Hamilton-Reeves JM, Sullivan DK, Gibson CA, Creed C, Carlson SE, Wesson D E, Grantham JJ. Diet and polycystic kidney disease: a pilot intervention study. Clin Nutr. 2017;36(2):458–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193. Amro OW, Paulus JK, Noubary F, Perrone RD. Low osmolar diet and adjusted water intake for vasopressin reduction in autosomal dominant polycystic kidney disease: a pilot randomized controlled trial. Am J Kidney Dis. 2016;68(6):882–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Carriazo S, Perez-Gomez MV, Cordido A, García-González MA, Sanz AB, Ortiz A, Sanchez-Niño MD. Dietary care for ADPKD patients: current status and future directions. Nutrients. 2019;11(7):1576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Di Iorio BR, Cupisti A, D'Alessandro C, Bellasi A, Barbera V, Di Lullo L. Nutritional therapy in autosomal dominant polycystic kidney disease. J Nephrol. 2018;31(5):635–43. [DOI] [PubMed] [Google Scholar]
- 196. Tran W-C, Huynh D, Chan T, Chesla CA, Park M. Understanding barriers to medication, dietary, and lifestyle treatments prescribed in polycystic kidney disease. BMC Nephrology. 2017;18(1):214. [DOI] [PMC free article] [PubMed] [Google Scholar]


