Chronic kidney disease (CKD) is a risk factor for end-stage renal disease (ESRD) and cardiovascular disease (CVD). ESRD or CVD develop in a substantial proportion of patients with CKD receiving standard-of-care therapy, and mortality in CKD remains unchanged. These data suggest that key pathogenetic mechanisms underlying CKD progression go unaffected by current treatments. Growing evidence suggests that nonalcoholic fatty liver disease (NAFLD) and CKD share common pathogenetic mechanisms and potential therapeutic targets. Common nutritional conditions predisposing to both NAFLD and CKD include excessive fructose intake and vitamin D deficiency. Modulation of nuclear transcription factors regulating key pathways of lipid metabolism, inflammation, and fibrosis, including peroxisome proliferator–activated receptors and farnesoid X receptor, is advancing to stage III clinical development. The relevance of epigenetic regulation in the pathogenesis of NAFLD and CKD is also emerging, and modulation of microRNA21 is a promising therapeutic target. Although single antioxidant supplementation has yielded variable results, modulation of key effectors of redox regulation and molecular sensors of intracellular energy, nutrient, or oxygen status show promising preclinical results. Other emerging therapeutic approaches target key mediators of inflammation, such as chemokines; fibrogenesis, such as galectin-3; or gut dysfunction through gut microbiota manipulation and incretin-based therapies. Furthermore, NAFLD per se affects CKD through lipoprotein metabolism and hepatokine secretion, and conversely, targeting the renal tubule by sodium–glucose cotransporter 2 inhibitors can improve both CKD and NAFLD. Implications for the treatment of NAFLD and CKD are discussed in light of this new therapeutic armamentarium.

Chronic kidney disease (CKD) affects up to 8% of the world’s adult population, with its prevalence increasing in an aging population beset by lifestyle-associated diseases such as obesity, the metabolic syndrome, diabetes, hypertension, and smoking (1). CKD may progress to end-stage renal disease (ESRD) and is an important cardiovascular disease (CVD) risk factor. Importantly, most patients with CKD die as a result of CVD before renal replacement therapy is initiated (1).

There is potential for improving recognition and treatment of CKD. In the Third National Health and Nutrition Survey, awareness among patients with stage 3 CKD was <8% (1). Furthermore, ESRD or CVD develop in a substantial proportion of patients with CKD receiving standard-of-care therapy, and all-cause mortality remains unchanged in the CKD population (2). These data suggest that key pathogenetic mechanisms underlying renal disease progression are unaffected by current treatment and prompt the search for easily identifiable risk factors and novel pharmacological targets.

The presence and severity of nonalcoholic fatty liver disease (NAFLD) has been related to the incidence and stage of CKD (3) independently of traditional CKD risk factors; conversely, the presence of CKD increases overall mortality in patients with NAFLD compared with the general population (4). Further supporting a pathogenic link between NAFLD and CKD, nonalcoholic steatohepatitis (NASH)–related cirrhosis carries a higher risk of renal failure than other etiologies of cirrhosis, is an increasing indication for simultaneous liver-kidney transplantation, and is an independent risk factor for kidney graft loss and CVD (5,6). Collectively, these data suggest that common pathogenic mechanisms underlie both liver and kidney injury and could be targeted to retard the progression of both NAFLD and CKD.

Nutritional Factors in NAFLD and CKD: Fructose and Vitamin D Intake

Dietary intake of fructose, the main constituent of sugar sweeteners, increased twofold over the past decade (7). Fructose may contribute to liver and kidney injury through several mechanisms, including uric acid overproduction, and consistently, uric acid–lowering agents have improved fructose-induced experimental NAFLD and CKD (8,9) (Table 1). On this basis, the impact of xanthine oxidase inhibitors on CKD progression is being evaluated in the trials CKD-FIX (Controlled Trial of Kidney Disease Progression From the Inhibition of Xanthine Oxidase; clinical trial reg. no. ACTRN12611000791932, www.anzctr.org.au) and FEATHER (Febuxostat Versus Placebo Randomized Controlled Trial Regarding Reduced Renal Function in Patients With Hyperuricemia Complicated by Chronic Kidney Disease Stage 3; clinical trial reg. no. UMIN000008343, www.umin.ac.jp/ctr) (Table 2).

Table 1

Nutritional factors involved in liver and renal disease in NAFLD and CKD

Biological effect
Dietary fructose  
 Cellular effects  
  Hypothalamus Weight gain 
   ↑ Dopaminergic tone→↑ appetite and calorie intake  
  Hepatocyte, skeletal myocyte Ectopic fat deposition 
   ↓ FFA oxidation and REE  
  Adipocyte Insulin resistance 
   Adipose tissue dysfunction→↓ adiponectin secretion  
  Liver and kidney cells (uric acid mediated) Inflammation 
   NLRP3 inflammasome activation→↑ IL-1 secretion→↑ macrophage accumulation  
   ↓ AMPK activity→ ↓ FFA oxidation Fibrosis 
   SREBP-1c activation→↑ de novo lipogenesis  
   NOX activation→ ROS generation→ podocyte loss, tubule cell EMT, endothelial injury Albuminuria 
  Pancreatic β-cell  
   ↓ Postprandial insulin response Hyperglycemia 
Vitamin D deficiency  
 Cellular effects  
  Skeletal myocyte  
   ↓ IRS-1 activity→↓ insulin signaling Insulin resistance 
  Hepatocyte, Kupffer cell  
   ↑ TLR-2/4/9 expression→↑ sensitivity to LPS and FFA-induced inflammatory cytokines ↑ ROS production→↑ hepatocyte apoptosis, ↑ inflammatory cell recruitment Ectopic fat deposition 
  Hepatic stellate cell  
   VDR downregulation→ ↑ TGF-β secretion and SMAD2 activation→↑ fibrogenesis Inflammation 
  Glomerular podocyte  
   ↑ p38- and ERK-mediated apoptosis→↑ podocyte injury→ glomerular barrier disruption  
   ↑ RAS activation→ glomerulosclerosis Hepatic fibrosis 
  Renal tubule cell and interstitial macrophage  
   ↑ SREBP-1c/SREBP-2→↑ toxic cholesterol and FFA lipid accumulation  
   ↓ FXR/PPAR-α activation→↑ toxic cholesterol and FFA lipid accumulation Proteinuria 
   ↑ NF-κB pathway activation→↑ inflammation and macrophage accumulation  
Biological effect
Dietary fructose  
 Cellular effects  
  Hypothalamus Weight gain 
   ↑ Dopaminergic tone→↑ appetite and calorie intake  
  Hepatocyte, skeletal myocyte Ectopic fat deposition 
   ↓ FFA oxidation and REE  
  Adipocyte Insulin resistance 
   Adipose tissue dysfunction→↓ adiponectin secretion  
  Liver and kidney cells (uric acid mediated) Inflammation 
   NLRP3 inflammasome activation→↑ IL-1 secretion→↑ macrophage accumulation  
   ↓ AMPK activity→ ↓ FFA oxidation Fibrosis 
   SREBP-1c activation→↑ de novo lipogenesis  
   NOX activation→ ROS generation→ podocyte loss, tubule cell EMT, endothelial injury Albuminuria 
  Pancreatic β-cell  
   ↓ Postprandial insulin response Hyperglycemia 
Vitamin D deficiency  
 Cellular effects  
  Skeletal myocyte  
   ↓ IRS-1 activity→↓ insulin signaling Insulin resistance 
  Hepatocyte, Kupffer cell  
   ↑ TLR-2/4/9 expression→↑ sensitivity to LPS and FFA-induced inflammatory cytokines ↑ ROS production→↑ hepatocyte apoptosis, ↑ inflammatory cell recruitment Ectopic fat deposition 
  Hepatic stellate cell  
   VDR downregulation→ ↑ TGF-β secretion and SMAD2 activation→↑ fibrogenesis Inflammation 
  Glomerular podocyte  
   ↑ p38- and ERK-mediated apoptosis→↑ podocyte injury→ glomerular barrier disruption  
   ↑ RAS activation→ glomerulosclerosis Hepatic fibrosis 
  Renal tubule cell and interstitial macrophage  
   ↑ SREBP-1c/SREBP-2→↑ toxic cholesterol and FFA lipid accumulation  
   ↓ FXR/PPAR-α activation→↑ toxic cholesterol and FFA lipid accumulation Proteinuria 
   ↑ NF-κB pathway activation→↑ inflammation and macrophage accumulation  

ERK, extracellular signal–regulated kinase; IL, interleukin; IRS-1, insulin receptor substrate 1; NLRP3, NOD-like receptor family, pyrin domain containing 3; NOX, NADPH oxidase; RAS, renin-angiotensin system; REE, resting energy expenditure; TLR, Toll-like receptor; VDR, vitamin D receptor.

Table 2

Mechanisms of action and developmental stages of drugs targeting both NAFLD and CKD

Developmental stage
Mechanism of actionMoleculeNAFLDCKD
Xanthine oxidase inhibitors Allopurinol, febuxostat Preclinical IIa 
   CKD-FIX (clinical trial reg. no. ACTRN12611000791932) 
   FEATHER (clinical trial reg. no. UMIN000008343) 
Vitamin D supplementation Natural (ergocalciferol, cholecalciferol, calcitriol), IIa IIa 
 Synthetic VDR agonists (doxercalciferol, paricalcitol) Clinical trial reg. no. NCT02098317 Clinical trial reg. nos. NCT00893451 and NCT01623024 
PPAR-δ agonists GW0742 — Preclinical (50
 GW610742 — Preclinical (51
PPAR-α/δ agonists GFT505 IIb GOLDEN-505 (10— 
PPAR- α/γ agonists Saroglitazar IIa (clinical trial reg. no. CTRI/2010/091/000108) — 
 Aleglitazar — IIb (54
SREBP-2 antagonists Natural antioxidants (myricetin) — Preclinical (13
FXR agonists Obeticholic acid IIa FLINT (17Preclinical (56,57
miRNA21 antagonists Antagomir-21 Preclinical (59Preclinical (60
AMPK activators Natural: curcumin berberine, monascin ankaflavin Preclinical (19Preclinical (20
Synthetic: oltipraz IIa (clinical trial reg. no. NCT01373554) — 
HIF-1α inhibitors YC-1 Preclinical (65— 
Dual mTORC1/2 inhibitor Rapamycin Preclinical (24Preclinical (25
ASK1 inhibitor GS-4997 IIa (clinical trial reg. no. NCT02466516) IIa (clinical trial reg. no. NCT02177786) 
Nrf2 activators Oltipraz IIa  
 Bardoxolone methyl Clinical trial reg. no. NCT01373554 Clinical trial reg. no. NCT02316821 
CCR2 receptor antagonist CCX140-B — IIa (82
CCR2/5 receptor antagonist Cenicriviroc IIa: CENTAUR (clinical trial reg. no. NCT02217475)  
 BMS-813160  IIa 
 PF-04634817 — Clinical trial reg. nos. NCT01752985 and NCT01712061 
Galectin-3 inhibitors GM-CT-01 Preclinical (84— 
 GR-MD-02 Phase I (clinical trial reg. no. NCT01899859) — 
 GCS-100 — IIa (clinical trial reg. no. NCT01843790) 
Incretin-based therapies Incretin mimetics   
  Liraglutide, exenatide IIa LEAN (92IIa (93,94
 DPP-4 inhibitors   
  Saxagliptin, linagliptin Preclinical (90IIa (MARLINA-T2D, CARMELINA) 
Gut microbiota manipulation Prebiotics, probiotics, synbiotics IIa (106IIa (clinical trial reg. no. ACTRN12613000493741) 
CETP inhibitors Fc-CETP6 Preclinical (112— 
Inhibitors of syndecan-1 shedding Sphingosine-1-phosphate — Preclinical (119
FGF21 analogs PEG-FGF21, recombinant FGF21, anti-FGFR1 mAb IIa (123— 
SGLT2 inhibitors Remogliflozin etabonate, luseogliflozin, ipragliflozin IIa (131,132— 
 Dapagliflozin, canagliflozin, empagliflozin — IIa (127, CREDENCE trial) 
Developmental stage
Mechanism of actionMoleculeNAFLDCKD
Xanthine oxidase inhibitors Allopurinol, febuxostat Preclinical IIa 
   CKD-FIX (clinical trial reg. no. ACTRN12611000791932) 
   FEATHER (clinical trial reg. no. UMIN000008343) 
Vitamin D supplementation Natural (ergocalciferol, cholecalciferol, calcitriol), IIa IIa 
 Synthetic VDR agonists (doxercalciferol, paricalcitol) Clinical trial reg. no. NCT02098317 Clinical trial reg. nos. NCT00893451 and NCT01623024 
PPAR-δ agonists GW0742 — Preclinical (50
 GW610742 — Preclinical (51
PPAR-α/δ agonists GFT505 IIb GOLDEN-505 (10— 
PPAR- α/γ agonists Saroglitazar IIa (clinical trial reg. no. CTRI/2010/091/000108) — 
 Aleglitazar — IIb (54
SREBP-2 antagonists Natural antioxidants (myricetin) — Preclinical (13
FXR agonists Obeticholic acid IIa FLINT (17Preclinical (56,57
miRNA21 antagonists Antagomir-21 Preclinical (59Preclinical (60
AMPK activators Natural: curcumin berberine, monascin ankaflavin Preclinical (19Preclinical (20
Synthetic: oltipraz IIa (clinical trial reg. no. NCT01373554) — 
HIF-1α inhibitors YC-1 Preclinical (65— 
Dual mTORC1/2 inhibitor Rapamycin Preclinical (24Preclinical (25
ASK1 inhibitor GS-4997 IIa (clinical trial reg. no. NCT02466516) IIa (clinical trial reg. no. NCT02177786) 
Nrf2 activators Oltipraz IIa  
 Bardoxolone methyl Clinical trial reg. no. NCT01373554 Clinical trial reg. no. NCT02316821 
CCR2 receptor antagonist CCX140-B — IIa (82
CCR2/5 receptor antagonist Cenicriviroc IIa: CENTAUR (clinical trial reg. no. NCT02217475)  
 BMS-813160  IIa 
 PF-04634817 — Clinical trial reg. nos. NCT01752985 and NCT01712061 
Galectin-3 inhibitors GM-CT-01 Preclinical (84— 
 GR-MD-02 Phase I (clinical trial reg. no. NCT01899859) — 
 GCS-100 — IIa (clinical trial reg. no. NCT01843790) 
Incretin-based therapies Incretin mimetics   
  Liraglutide, exenatide IIa LEAN (92IIa (93,94
 DPP-4 inhibitors   
  Saxagliptin, linagliptin Preclinical (90IIa (MARLINA-T2D, CARMELINA) 
Gut microbiota manipulation Prebiotics, probiotics, synbiotics IIa (106IIa (clinical trial reg. no. ACTRN12613000493741) 
CETP inhibitors Fc-CETP6 Preclinical (112— 
Inhibitors of syndecan-1 shedding Sphingosine-1-phosphate — Preclinical (119
FGF21 analogs PEG-FGF21, recombinant FGF21, anti-FGFR1 mAb IIa (123— 
SGLT2 inhibitors Remogliflozin etabonate, luseogliflozin, ipragliflozin IIa (131,132— 
 Dapagliflozin, canagliflozin, empagliflozin — IIa (127, CREDENCE trial) 

FGFR, FGF21 receptor; mAb, monoclonal antibodies; PEG, pegylated; VDR, vitamin D receptor; YC-1, 1-benzyl-3-(substituted aryl)-5-methylfuro[3,2-c]pyrazole.

Vitamin D attracted considerable interest because of its pleiotropic functions, with roles in regulation of cell proliferation, differentiation, immunity, inflammation, fibrogenesis, and metabolism (Table 1), concurrent with an unsuspected high prevalence of vitamin D deficiency, approaching 25% of the general adult population (37). Vitamin D deficiency has been linked to the pathogenesis and severity of NAFLD and CKD by observational and experimental data (3840) (Table 1). However, the few trials of vitamin D supplementation yielded mixed results, and the benefit of vitamin D supplementation remains uncertain (41). NAFLD and CKD are characterized by vitamin D resistance, which is partly determined by impaired hepatic 25 hydroxylation and increased renal tubular 25 hydroxyvitamin D loss, and may require higher-dose supplementation of vitamin D, calcitriol, or vitamin D receptor agonists (e.g., paricalcitol) to be overcome (42,43). The effects of vitamin D supplementation in CKD and NASH are being evaluated in several clinical trials (clinical trial reg. nos. NCT00893451, NCT01623024, and NCT02098317, www.clinicaltrials.gov).

Reversing Ectopic Fat Deposition by Targeting Nuclear Transcription Factors in NAFLD and CKD

NAFLD and CKD are characterized by ectopic toxic lipid accumulation, which is determined by an extensive derangement in hepatic and renal lipid metabolism and triggers lipoperoxidative stress, cell apoptosis, inflammation, and fibrosis (4446) (Fig. 1). Underlying these abnormalities is an extensive deregulation of nuclear transcription factors that regulate lipid metabolism, inflammation, and fibrogenesis, including peroxisome proliferator–activated receptor (PPAR)-α, PPAR-δ, and PPAR-γ; SREBP-2; and farnesoid X receptor (FXR), which represent an attractive target for the treatment of NAFLD and CKD (47,48) (Fig. 1).

Figure 1

The role of nuclear transcription factors in the pathogenesis of NAFLD and CKD. Each nuclear transcription factor is reported in the text box at the center of the scheme and is marked with a superscript number. The various molecular pathways affected by each nuclear transcription factor are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding nuclear transcription factor affecting the pathway. eNOS, endothelial nitric oxide synthase; LPL, lipoprotein lipase; NLRP3, NOD-like receptor family, pyrin domain containing 3; NOS, nitric oxide synthase; NOX, NADPH oxidase; PAI-1, plasminogen activator inhibitor 1.

Figure 1

The role of nuclear transcription factors in the pathogenesis of NAFLD and CKD. Each nuclear transcription factor is reported in the text box at the center of the scheme and is marked with a superscript number. The various molecular pathways affected by each nuclear transcription factor are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding nuclear transcription factor affecting the pathway. eNOS, endothelial nitric oxide synthase; LPL, lipoprotein lipase; NLRP3, NOD-like receptor family, pyrin domain containing 3; NOS, nitric oxide synthase; NOX, NADPH oxidase; PAI-1, plasminogen activator inhibitor 1.

Close modal

On the basis of the finding that PPAR-α and PPAR-δ are downregulated in NAFLD and CKD (47,49), potent, selective PPAR-α, PPAR-δ, and dual PPAR-α/δ agonists (K-877, GW501516, MBX-8025 and GFT505, respectively) were evaluated in these two conditions, with encouraging results in preclinical models (4951). Some of these compounds advanced to the clinical stage of development, and GFT505, a dual PPAR-α/δ agonist, improved steatohepatitis, fibrosis, and the glycolipid profile in the recently completed GOLDEN-505 trial (10) (Table 2).

The PPAR-γ agonists thiazolidinediones comprise another pharmacological class that has significantly improved NASH and albuminuria in clinical trials (11,12), but their clinical use was limited by their side effects. These drawbacks prompted the development of new compounds, including dual PPAR-α/γ agonists, which maintained the therapeutic effectiveness of PPAR-γ agonists but were devoid of their unwanted effects (Fig. 1). Saroglitazar, a potent PPAR-α/γ agonist, did not induce weight gain, peripheral edema, or other adverse events after 1 year (52) and improved markers of NAFLD in patients with diabetes (53), whereas aleglitazar slowed estimated glomerular filtration rate (eGFR) decline in diabetic nephropathy (54) (Table 2). A small, phase IIa trial of patients with biopsy-proven NASH has been completed, but the results are not yet available (clinical trial reg. no. CTRI/2010/091/000108, www.ctri.nic.in). Larger and longer randomized clinical trials (RCTs) are needed to evaluate long-term clinical safety and effectiveness of these compounds in patients with and without diabetes.

Among various lipotoxic species accumulating in NAFLD and CKD, free cholesterol is believed to play a key pathogenic role in liver and renal injury (13,55). Ectopic cholesterol accumulation is driven by an inappropriate upregulation of transcription factor SREBP-2, with consequently increased cholesterol synthesis, influx, and retention and reduced cholesterol excretion by liver and renal cells (13,55) (Fig. 1). Such pervasive deregulation in all steps of cholesterol metabolism may diminish the effectiveness of available cholesterol-lowering drugs that target single steps in cholesterol metabolism (14). Therefore, modulation of SREBP-2 activity represents an attractive therapeutic tool. Although selective SREBP-2 antagonists are under development, several natural antioxidants, such as myricetin, repress SREBP-2 expression and ameliorate cholesterol-induced inflammation and fibrosis in experimental models (13) (Table 2).

FXR is a nuclear transcription factor with prominent insulin sensitizing, antilipogenic, anti-inflammatory, and antifibrotic properties; furthermore, its activation improves endothelial function (16,56,57) (Fig. 1). FXR expression is downregulated in the liver and kidney of patients with NAFLD and CKD, respectively, and is inversely related to disease severity. On this basis, potent semisynthetic bile acid FXR agonists have been developed (Table 2). Obeticholic acid (or INT-747), a semisynthetic chenodeoxycholic acid derivative, improved liver histology in the phase IIa, multicenter, randomized FXR Ligand NASH Treatment (FLINT) trial and in ameliorated renal histology and proteinuria in nutritional models of CKD (16,57). Several issues remain, however, including the effectiveness of FXR agonists in subjects without diabetes and the impact of HDL cholesterol reduction on long-term CVD risk.

Epigenetic Regulation in NASH and CKD

microRNAs (miRNAs) are small (∼22 base pairs) endogenous noncoding RNAs that regulate gene expression of at least 60% of protein coding genes. miRNAs recognize mRNA targets through sequence complementarity between the miRNA and binding sites in the 3′ untranslated regions of the target mRNAs or through interaction with RNA-binding proteins.

miRNAs regulate gene expression in one of two ways, depending on the degree of complementarity between the miRNA and its target. miRNAs that bind to mRNA targets with imperfect complementarity block target gene expression through translational silencing while miRNAs binding their mRNA targets with perfect complementarity enhance target gene expression (15,58). To date, >2,500 miRNAs have been identified in the human genome, and each can regulate several hundred target genes involved in diverse developmental and cellular processes, including cellular metabolism proliferation, differentiation, and apoptosis. Several miRNAs have been found to be dysregulated in NAFLD and CKD (15,58). On this basis, approaches inhibiting overexpressed miRNAs by antisense oligonucleotides or restoring the expression of downregulated miRNAs by synthetic miRNA mimics have been attempted. miRNA21 in particular is an attractive target because it regulates key metabolic, proinflammatory, and profibrogenic pathways and its hepatic and renal overexpression in NASH and CKD leads to PPAR-α downregulation, SREBP-2 upregulation, mitochondrial dysfunction, and profibrogenic hepatic stellate cell activation and proximal tubular cell epithelial-to-mesenchymal transition (EMT) (59,60). Consistently, in experimental models of NASH and CKD, anti-miRNA21 antisense oligonucleotides induced weight loss, normalized metabolic dysregulation, and improved hepatic and renal inflammation and fibrosis, effects at least partly mediated by PPAR-α upregulation (59,60). Despite these premises, several issues remain, including the stability and selective delivery of the pharmacological modulators to the target organs and long-term safety of this approach because miRNAs also regulate cell proliferation and cell cycle progression in diverse tissues and long-term consequences of its modulation on tumor onset and progression are unclear (15,59).

In mammals, cellular metabolism is finely orchestrated by molecular sensors of energy, nutrient, and oxygen status to adapt to changing substrate availability. The dysregulation of some of these sensors, including AMPK, hypoxia-inducible factor (HIF)-1α, and mammalian target of rapamycin (mTOR), has been implicated in the pathogenesis of NAFLD and CKD and could be targeted for the treatment of NAFLD and CKD. AMPK is a ubiquitous kinase that preserves cell survival under calorie restriction or high energy demand (18). In response to cellular ATP depletion or to an increase in AMP/ATP ratio, AMPK activation enhances substrate oxidation and has antioxidant, anti-inflammatory, and antifibrotic effects (18,20,61) (Fig. 2A). On this basis, several natural AMPK activators were assessed in preclinical models of NASH and CKD, with encouraging results (18,20,61), and the synthetic AMPK activator oltipraz is being evaluated in patients with noncirrhotic NAFLD (clinical trials reg. no. NCT01373554, www.clinicaltrials.gov) (Table 2).

Figure 2

Mechanisms connecting cellular sensors AMPK (A) and HIF-1α and mTORC1 (B) in the pathogenesis of NAFLD and CKD. HIF-1α and mTORC1 are reported in the text box at the center of the scheme and are marked with a superscript number. The various molecular pathways affected by each sensor are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding cellular sensor affecting the pathway. eNOS, endothelial nitric oxide synthase; LOXL, lysyl oxidase-like; NOX, NADPH oxidase; PAI-1, plasminogen activator inhibitor 1; PGC-1α, peroxisome proliferator–activated receptor γ coactivator 1α.

Figure 2

Mechanisms connecting cellular sensors AMPK (A) and HIF-1α and mTORC1 (B) in the pathogenesis of NAFLD and CKD. HIF-1α and mTORC1 are reported in the text box at the center of the scheme and are marked with a superscript number. The various molecular pathways affected by each sensor are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding cellular sensor affecting the pathway. eNOS, endothelial nitric oxide synthase; LOXL, lysyl oxidase-like; NOX, NADPH oxidase; PAI-1, plasminogen activator inhibitor 1; PGC-1α, peroxisome proliferator–activated receptor γ coactivator 1α.

Close modal

HIF-1α is a ubiquitous oxygen-sensitive protein that regulates transcription of genes involved in metabolic adaptation, energy conservation, angiogenesis, and cell survival in response to cellular hypoxia and nonhypoxic stimuli like cholesterol overload (21). Inappropriate HIF-1α activation following such stimuli as chronic intermittent hypoxia and cholesterol overload has been found to be involved in NASH pathogenesis (22,62), whereas chronic hypoxia has been implicated in renal injury in early diabetic and obesity-related CKD (63,64) (Fig. 2B). On this basis, small-molecule synthetic HIF-1α inhibitors, including YC-1, have been developed and show potent antifibrotic properties in experimental models of NASH (65) (Table 2). Several issues remain, however. In some studies, HIF-1α activation protected against kidney injury (66), suggesting that HIF-1α may not be the sole, or the best, therapeutic target for reversing cellular effects of hypoxia in CKD.

mTOR is a serine/threonine kinase that in response to changes in cellular nutrient levels, growth factors like insulin and IGF and other stressors associates with companion proteins to form two distinct signaling molecular complexes, mTOR complex 1 (mTORC1) and mTORC2 (23). mTORC1 has been more extensively studied and found to promote cellular anabolism by stimulating synthesis of protein, lipid, and nucleotides and blocking catabolic processes. Inappropriate mTORC1 activation in NAFLD and CKD inhibits autophagy (23,67) and promotes insulin resistance, ectopic lipid accumulation, lipotoxicity, and proinflammatory monocyte recruitment in the liver and kidney (Fig. 2B) (24,25). Consistently, mTORC1 inhibition has reversed metabolic abnormalities and attenuated lipid accumulation, inflammation, and fibrosis in diverse models of NASH and CKD (68,69) and may represent a therapeutic option for NAFLD and CKD (70) (Table 2).

Although increased oxidative stress is believed to play a central role in NASH and CKD progression, single antioxidant supplementation strategies have yielded variable results (11), and other approaches targeting common effectors of redox regulation, like apoptosis signal-regulating kinase 1 (ASK1) and nuclear erythroid 2–related factor 2 (Nrf2), are being investigated. ASK1 is a serine/threonine kinase belonging to the mitogen-activated protein kinase (MAPK) kinase kinase family, which is activated in response to stresses like reactive oxygen species (ROS), tumor necrosis factor-α (TNF-α), lipopolysaccharide (LPS), and endoplasmic reticulum (ER) stress (71). ASK1 activates downstream terminal MAPK kinases p38 and c-Jun N-terminal kinase (JNK), which promote insulin resistance, cell death, proinflammatory cytokine/chemokine production, and fibrogenesis (71) (Fig. 3).

Figure 3

Role of effectors of redox regulation ASK1 and Nrf2 and role of galectin-3 in the pathogenesis of NAFLD and CKD. ASK1, Nrf2, and galectin-3 are reported in the text box at the center of the scheme and are marked with a superscript number. The various molecular pathways affected by these three molecules are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding factor affecting the pathway. COX, cyclooxygenase; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal–related kinase; G6PD, glucose-6-phosphate dehydrogenase; Glt-Px, glutathione peroxidase; Glt-R, glutathione reductase; GST, glutathione S-transferase; HPC, hepatic progenitor cells; iNOS, inducible nitric oxide synthase; IRS, insulin receptor substrate; NKT, natural killer T; NOS, nitric oxide synthase; PAI-1, plasminogen activator inhibitor 1; TXN-R, thioredoxin reductase.

Figure 3

Role of effectors of redox regulation ASK1 and Nrf2 and role of galectin-3 in the pathogenesis of NAFLD and CKD. ASK1, Nrf2, and galectin-3 are reported in the text box at the center of the scheme and are marked with a superscript number. The various molecular pathways affected by these three molecules are shown in the boxes surrounding the central box, with the superscript number referring to the corresponding factor affecting the pathway. COX, cyclooxygenase; eNOS, endothelial nitric oxide synthase; ERK, extracellular signal–related kinase; G6PD, glucose-6-phosphate dehydrogenase; Glt-Px, glutathione peroxidase; Glt-R, glutathione reductase; GST, glutathione S-transferase; HPC, hepatic progenitor cells; iNOS, inducible nitric oxide synthase; IRS, insulin receptor substrate; NKT, natural killer T; NOS, nitric oxide synthase; PAI-1, plasminogen activator inhibitor 1; TXN-R, thioredoxin reductase.

Close modal

Data have implicated ASK1 activation in oxidative stress–induced inflammation and fibrogenesis in NASH and CKD, and pharmacological ASK1 inhibition prevented diet-induced NASH and halted progression of diabetic and nondiabetic experimental CKD (72,73). On this basis, the highly selective oral ASK1 inhibitor GS-4997 is being evaluated in patients with NASH with moderate advanced fibrosis (clinical trial reg. no. NCT02466516, www.clinicaltrials.gov) and in patients with diabetes with stage 3/4 nephropathy (clinical trial reg. no. NCT02177786, www.clinicaltrials.gov) (Table 2). Several issues need to be clarified, however. ASK1 inhibition did not improve podocyte loss and albuminuria in experimental diabetic CKD, suggesting that this kinase is not central for glomerular injury (73). Furthermore, the impact of ASK1 inhibitors in nondiabetic CKD is unknown.

Nrf2 is a transcription factor expressed ubiquitously in human tissues and most abundantly in the liver (74), where it regulates the expression of several antioxidant and detoxifying enzymes and has direct metabolic, anti-inflammatory, and proautophagic actions (74) (Fig. 3). Under basal conditions, Nrf2 is kept transcriptionally inactive through binding to its inhibitor, Kelch-like ECH-associated protein 1 (KEAP1) (74). ROS and reactive nitrogen species interact with KEAP1 and cause the loosening of Nrf-2, which translocates to the nucleus and modulates transcription of its target genes (Fig. 3). The relevance of Nrf2 in the pathogenesis of liver and kidney injury has emerged in diverse diet-induced models of NASH and CKD, where disease progression was accelerated by Nrf2 deletion and prevented by Nrf2 activators (75,76). On this basis, several natural and synthetic small-molecule Nrf2 activators are currently being evaluated in patients with NASH and CKD (Table 2). Following early encouraging data, clinical development of bardoxolone methyl, a synthetic Nrf2 activator, was interrupted for safety concerns related to heart failure (77). A reanalysis of the potential risk/benefit ratio of the drug for a Japanese population and the observation that most of the severe adverse effects occurred in the first month of therapy prompted initiation of a dose-escalating RCT in Japan (clinical trial reg. no. NCT02316821, www.clinicaltrials.gov). Another potent synthetic Nrf2 activator, oltipraz, is being evaluated in patients with NAFLD (clinical trial reg. no. NCT01373554, www.clinicaltrials.gov).

Chemokines are small proteins that regulate leukocyte migration into tissues and consequent inflammation, tissue remodeling, and fibrosis (78). Among the >40 chemokine ligands and 20 chemokine receptors currently identified, chemokine (C-C motif) ligand 2 (CCL2 or MCP-1) and its receptor CCR2 have been implicated in the pathogenesis of NASH and CKD. In NAFLD, hepatic cells and adipocytes secrete CCL2, which attracts proinflammatory cells to the liver to promote NASH development (79,80), whereas genetic or pharmacological inhibition of the CCL2/CCR2 axis reverses steatohepatitis and advanced hepatic fibrosis (80). In the kidney, tubule cells and podocytes secrete chemokines CCL2 and CCL5 in response to diverse proinflammatory stimuli to promote tubulointerstitial inflammation and fibrosis, which are reversed by chemokine antagonists (81). On this basis, chemokine antagonists are advancing to the clinical stage of development: The small-molecule CCR2 antagonist CCX140-B has reduced albuminuria and slowed eGFR decline in diabetic nephropathy (82), whereas the dual chemokine receptor CCR2/CCR5 antagonists BMS-813160, PF-04634817, and cenicriviroc are being evaluated in diabetic nephropathy (clinical trial reg. nos. NCT01752985 and NCT01712061, www.clinicaltrials.gov) and in NASH (clinical trial reg. no. NCT02217475, www.clinicaltrials.gov) (Table 2).

Galectin-3 is a lectin broadly expressed by immune and epithelial cells where it localizes mainly in the cytoplasm as well as in the nucleus, cell surface, and extracellular space (83). Galectin-3 regulates cell proliferation, apoptosis, cell adhesion, and affinity for advanced glycation end products (AGEs), exerting multiple and sometimes contrasting effects, depending on its cellular location, cell type, and mechanisms of injury (83) (Fig. 3). Galectin-3 is upregulated in the liver and kidney of patients with NASH and CKD and correlates with the severity of liver and renal disease (26,84). Furthermore, circulating galectin-3 levels predict renal function decline and cardiovascular and all-cause mortality in patients with CKD (85). Consistent with epidemiological data, functional galectin-3 manipulation has shown important proinflammatory and profibrotic effects of this lectin (26,83,84). On this basis, pharmacological galectin-3 inhibition with small-molecule competitive inhibitors, including GR-MD-02 (galactoarabino-rhamnogalaturonan), GM-CT-01 (galactomannan), and N-acetyllactosamine, prevents hypertensive nephropathy (86) and reverses diet-induced NASH and cirrhosis (87). GR-MD-02 was well tolerated and improved markers of hepatic fibrosis in a phase I RCT enrolling patients with NASH and advanced fibrosis (clinical trial reg. no. NCT01899859, www.clinicaltrials.gov), whereas a phase IIa RCT is exploring the effect of the galectin-3 antagonist GCS-100 on eGFR in CKD (clinical trial reg. no. NCT01843790, www.clinicaltrials.gov) (Table 2).

Data on galectin-3 inhibition are not univocal, however, and galectin-3 deletion exacerbates systemic inflammation, hyperglycemia, and liver and kidney injury in diet-induced obese rodents (88). It has been suggested that inhibition of AGE uptake by the liver, which clears >90% of these end products from the circulation, promotes the systemic accumulation of AGEs and receptor for AGE (RAGE)–mediated uptake by other tissues, thereby aggravating extrahepatic toxicity of these molecules. Because both NASH and CKD are characterized by AGE accumulation, a better understanding of the impact of galectin-3 inhibitors on AGE-mediated tissue injury in vivo is warranted.

Incretin-based therapies, including GLP-1 mimetics and dipeptidyl peptidase 4 (DPP-4) inhibitors, increase insulin release from the pancreas, reduce glucagon production, and possess numerous extrapancreatic metabolic benefits, which prompted the evaluation for the treatment of NAFLD and CKD (89,90) (Table 3). Preliminary human data suggested that incretin mimetics improve NAFLD. A meta-analysis of six RCTs from the Liraglutide Effect and Action in Diabetes (LEAD) program found a significant improvement in biochemical and radiological features of steatosis (91). Furthermore, in the Liraglutide Efficacy and Action in NASH (LEAN) trial, liraglutide 1.8 mg/day for 48 weeks induced NASH resolution and improved markers of lipotoxicity, inflammation, and metabolic dysfunction compared with placebo (92) (Table 2).

Table 3

Role of incretin-based therapies and gut microbiota in the pathogenesis of NAFLD and CKD

Biological effect
Incretin analogs: GLP-1 receptor agonists  
 Cellular mechanism  
  Hypothalamus ↓ Calorie intake 
   ↓ Appetite  
  Skeletal myocyte ↑ Insulin sensitivity 
   ↑ Glucose uptake  
   ↑ Glycogen synthesis  
  Hepatocyte ↑ Insulin sensitivity 
   ↑ Glycogen synthesis and ↓ gluconeogenesis  
   ↑ Autophagy ↓ Hepatic steatosis 
   ↑ cAMP→↑AMPK and SIRT-1 activity  
   ↓ FGF21 secretion  
  Adipocyte ↑ Insulin sensitivity 
   ↑ Lipolysis  
   ↑ Glucose uptake ↓ Adipose tissue 
   ↑ Lipogenesis (↑ FFA synthesis, uptake, and reesterification)  
   ↑ LPL activity Dysfunction 
   ↑ Adiponectin secretion  
  Proximal tubule cell ↑ Na delivery to distal tubule→ 
   ↓ NHE3 activity→↑ Na uresis→↓ Na hyperreabsorption ↓ Tubuloglomerular feedback→ 
   ↑ PPAR-α→↑ FFA oxidation ↓ Glomerular hyperfiltration 
  Glomerular endothelial cell, mesangial cell, tubule cell ↓ Blood pressure 
   ↓ Apoptosis ↓ Fat infiltration 
   ↓ AGE/RAGE axis activation ↓ Glomerular sclerosis 
   ↓ IL-1/MCP-1/TGF-β secretion→↓ monocyte recruitment and fibrogenesis ↓ Inflammation and fibrosis 
   ↑ NOS activity ↓ Endothelial dysfunction 
   ↑ cAMP→ NOX downregulation→↓ ROS generation ↑ Insulin sensitivity 
   ↓ AngII activity→↓ IRS-1 phosphorylation→↑ IRS-1 signaling  
Incretin analogs: DPP-4 inhibitors  
Liver, kidney  
  ↑ GLP-1 activity  
Kidney Same effects of GLP-1R agonists 
  ↑ BNP→↑ natriuresis and vasodilation, ↓ RAAS and sympathetic activity  
  ↑ NPY and PYY→↑ AngII-mediated vasoconstriction  
  ↑ SDF-1a→↑ mesenchymal stem cells recruitment Ultimate effect depends on local tissue activity of various substrates inactivated by DPP-4 
Altered gut microbiota composition and intestinal barrier disruption  
 Cellular mechanism  
  Hepatocyte, macrophage, HSC, renal vascular endothelial cell, podocytes  
   ↑ LPS-TLR-4 axis activation→ ↑ proinflammatory cytokines/TGF-β Hepatic and renal inflammation, hepatic and renal fibrogenesis 
   ↑ LPS-TLR-4 axis activation→ ↑ NOX type II activity→ ROS production Endothelial dysfunction 
  Enterocyte Gut barrier disruption 
   ↓ SCFA production→↑ epithelial injury and ↓ GLP-1 secretion  
  Hepatocyte  
   ↑ Conversion of intestinal TMA to TMAO by flavin-containing monooxygenases Hepatic steatosis 
   Hepatic necroinflammation 
  Tubule cells Atherosclerosis 
   ↑ TMAO-induced TGF-β/SMAD3 axis activation Tubule-interstitial fibrosis 
  Skeletal myocyte Insulin resistance 
   ↑ URM-induced phosphorylation of IRS-1→↓ IRS-1 signaling  
  Hepatocyte Insulin resistance 
   ↑ URM-induced de novo lipogenesis Hepatic steatosis 
   ↓ URM-induced VLDL secretion  
  Adipocyte  
   ↑ URM-induced leptin secretion Insulin resistance 
   ↓ URM-induced de novo lipogenesis Adipose tissue dysfunction 
   ↑ URM-induced zinc-α2-glycoprotein and ↓ perilipin expression→↑ lipolysis  
  Macrophage Adipose tissue and systemic inflammation 
   ↑ LPS-induced NF-κB activation  
Biological effect
Incretin analogs: GLP-1 receptor agonists  
 Cellular mechanism  
  Hypothalamus ↓ Calorie intake 
   ↓ Appetite  
  Skeletal myocyte ↑ Insulin sensitivity 
   ↑ Glucose uptake  
   ↑ Glycogen synthesis  
  Hepatocyte ↑ Insulin sensitivity 
   ↑ Glycogen synthesis and ↓ gluconeogenesis  
   ↑ Autophagy ↓ Hepatic steatosis 
   ↑ cAMP→↑AMPK and SIRT-1 activity  
   ↓ FGF21 secretion  
  Adipocyte ↑ Insulin sensitivity 
   ↑ Lipolysis  
   ↑ Glucose uptake ↓ Adipose tissue 
   ↑ Lipogenesis (↑ FFA synthesis, uptake, and reesterification)  
   ↑ LPL activity Dysfunction 
   ↑ Adiponectin secretion  
  Proximal tubule cell ↑ Na delivery to distal tubule→ 
   ↓ NHE3 activity→↑ Na uresis→↓ Na hyperreabsorption ↓ Tubuloglomerular feedback→ 
   ↑ PPAR-α→↑ FFA oxidation ↓ Glomerular hyperfiltration 
  Glomerular endothelial cell, mesangial cell, tubule cell ↓ Blood pressure 
   ↓ Apoptosis ↓ Fat infiltration 
   ↓ AGE/RAGE axis activation ↓ Glomerular sclerosis 
   ↓ IL-1/MCP-1/TGF-β secretion→↓ monocyte recruitment and fibrogenesis ↓ Inflammation and fibrosis 
   ↑ NOS activity ↓ Endothelial dysfunction 
   ↑ cAMP→ NOX downregulation→↓ ROS generation ↑ Insulin sensitivity 
   ↓ AngII activity→↓ IRS-1 phosphorylation→↑ IRS-1 signaling  
Incretin analogs: DPP-4 inhibitors  
Liver, kidney  
  ↑ GLP-1 activity  
Kidney Same effects of GLP-1R agonists 
  ↑ BNP→↑ natriuresis and vasodilation, ↓ RAAS and sympathetic activity  
  ↑ NPY and PYY→↑ AngII-mediated vasoconstriction  
  ↑ SDF-1a→↑ mesenchymal stem cells recruitment Ultimate effect depends on local tissue activity of various substrates inactivated by DPP-4 
Altered gut microbiota composition and intestinal barrier disruption  
 Cellular mechanism  
  Hepatocyte, macrophage, HSC, renal vascular endothelial cell, podocytes  
   ↑ LPS-TLR-4 axis activation→ ↑ proinflammatory cytokines/TGF-β Hepatic and renal inflammation, hepatic and renal fibrogenesis 
   ↑ LPS-TLR-4 axis activation→ ↑ NOX type II activity→ ROS production Endothelial dysfunction 
  Enterocyte Gut barrier disruption 
   ↓ SCFA production→↑ epithelial injury and ↓ GLP-1 secretion  
  Hepatocyte  
   ↑ Conversion of intestinal TMA to TMAO by flavin-containing monooxygenases Hepatic steatosis 
   Hepatic necroinflammation 
  Tubule cells Atherosclerosis 
   ↑ TMAO-induced TGF-β/SMAD3 axis activation Tubule-interstitial fibrosis 
  Skeletal myocyte Insulin resistance 
   ↑ URM-induced phosphorylation of IRS-1→↓ IRS-1 signaling  
  Hepatocyte Insulin resistance 
   ↑ URM-induced de novo lipogenesis Hepatic steatosis 
   ↓ URM-induced VLDL secretion  
  Adipocyte  
   ↑ URM-induced leptin secretion Insulin resistance 
   ↓ URM-induced de novo lipogenesis Adipose tissue dysfunction 
   ↑ URM-induced zinc-α2-glycoprotein and ↓ perilipin expression→↑ lipolysis  
  Macrophage Adipose tissue and systemic inflammation 
   ↑ LPS-induced NF-κB activation  

BNP, brain-derived natriuretic peptide; HSC, hepatic stellate cell; IL, interleukin; IRS-1, insulin receptor substrate 1; LPL, lipoprotein lipase; NOS, nitric oxide synthase; NOX, NADPH oxidase; NPY, neuropeptide Y; PYY, peptide YY; SCFA, short-chain fatty acid; SDF-1a, stromal-derived factor 1a; SIRT, sirtuin; TLR, Toll-like receptor; TMA, trimethylamine.

Incretin-based therapies also have the potential for nephroprotection independent of improved glycemic control through several mechanisms (Table 3). GLP-1 administration induced natriuresis through inhibition of proximal tubular Na-H exchanger 3 (NHE3) and reduced activation of the angiotensin II (AngII) axis (93). These actions counteract the increase in proximal tubular sodium reabsorption, which has been hypothesized to trigger glomerular hyperfiltration, the functional defect believed to be central to obesity-associated and diabetic CKD (93). Furthermore, GLP-1 mimetics have demonstrated direct renal anti-inflammatory, antifibrotic, and antioxidative actions (27,94). Collectively, these properties may explain the attenuated progression of overt diabetic nephropathy in two small preliminary RCTs of treatment with liraglutide (27,94). In addition to inactivating GLP-1, DPP-4 cleaves multiple other peptides, including brain-derived natriuretic peptide, neuropeptide Y, peptide YY, and stromal-derived factor 1a (Table 3). Thus, the effect of DPP-4 inhibition depends on the actions of the various substrates inactivated in each tissue and organ. This may theoretically explain the slightly lower antihypertensive effect observed with DPP-4 inhibitors compared with GLP-1 mimetics (28). Although the impact of DPP-4 inhibitors on NAFLD must be assessed, results from the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus-Thrombolysis in Myocardial Infarction (SAVOR-TIMI) 53 trial (29) and from four other RCTs (95) have suggested that saxagliptin and linagliptin reduce the development and progression of albuminuria. Renal effects of linagliptin are currently being investigated in the MARLINA-T2D (Efficacy, Safety, & Modification of Albuminuria in Type 2 Diabetes Subjects With Renal Disease With Linagliptin; clinical trial reg. no. NCT01792518, www.clinicaltrials.gov) and CARMELINA (Cardiovascular and Renal Microvascular Outcome Study With Linagliptin in Patients With Type 2 Diabetes Mellitus; clinical trial reg. no. NCT01897532, www.clinicaltrials.gov) trials. In conclusion, incretin-based therapies address several pathophysiological mechanisms common to experimental NAFLD and CKD, but their impact on renal disease in nondiabetic CKD is unknown. Furthermore, incretin-based therapies did not affect the risk of CVD, a major cause of death in NAFLD and CKD (96).

The capacity for gut microbiota to interact with host metabolic and immune responses and to contribute to the development of obesity-associated disorders has been increasingly recognized (97). Patients with NAFLD and CKD exhibit an altered gut microbiota composition, with a relative decrease in healthy Bacteroidetes, Lactobacillaceae, and Prevotellaceae families and disruption of the normal gastrointestinal barrier (98,99). The resulting accumulation of gut-derived toxins induces inflammation (100), insulin resistance, and ectopic fat deposition in liver and muscle through several mechanisms (Table 3). Some of these molecules, including endotoxin, indoxylsulfate, p-cresyl sulfate, and trimethylamine-N-oxide (TMAO), have documented clinical relevance for the development and progression of CKD (101104).

CKD may also aggravate gut dysbiosis and systemic inflammation through accumulation of uremic toxic metabolites (URMs) normally eliminated by the kidneys, including urea and p-cresyl sulfate. The accumulation of urea may lead to influx into the gastrointestinal lumen where it is hydrolyzed to ammonia by microbial urease and then converted to ammonium hydroxide. Ammonia and ammonium hydroxide promote the growth of urea-metabolizing bacteria at the expense of carbohydrate-fermenting strains and disrupt intestinal epithelial tight junctions, enhancing passage of LPS and other toxic luminal compounds into the circulation (105). Further highlighting the relevance of this mechanism to systemic inflammation, administration of oral activated charcoal absorbent AST-120 improves intestinal barrier function and reduces systemic oxidative stress, inflammation, and endotoxemia in rodent models of CKD (105). Gut microbiota manipulation with probiotics or prebiotics improved surrogate markers of NAFLD in small RCTs of short duration (106) and reduced URM levels in patients with CKD (107). The impact of synbiotic administration on renal function in CKD is being investigated in the Synbiotics Easing Renal Failure by Improving Gut Microbiology (SYNERGY) trial (clinical trial reg. no. ACTRN12613000493741, www.anzctr.org.au).

In NAFLD, liver disease per se contributes to kidney injury through several mechanisms. The liver contains up to 80% of all macrophages of the body, and the steatotic liver may represent a more relevant source of proinflammatory cytokines than adipose tissue (30). Furthermore, the liver is a central regulator of lipoprotein metabolism and secretes hepatokines like fibroblast growth factor 21 (FGF21), which can modulate whole-body metabolism and inflammation.

Hepatic Secretion of VLDL, Cholesteryl Ester Transfer Protein, and Syndecan-1 in the Pathogenesis of Atherogenic Dyslipidemia and Kidney Injury

Atherogenic dyslipidemia is the most common lipid abnormality in CKD and an independent predictor of the incidence and progression of CKD and of CVD in patients with CKD (108,109). Atherogenic dyslipidemia promotes CKD through receptor-mediated uptake of qualitatively abnormal lipoproteins by glomerular and tubulointerstitial cells (110). NAFLD may promote atherogenic dyslipidemia through several mechanisms, which represent potential therapeutic targets. Liver fat accumulation per se proportionally increases the hepatic secretion rate of large VLDL1, which exchanges triglycerides with cholesterol contained in circulating LDL and HDL particles, resulting in small LDL and HDL3 formation (55). Furthermore, recent studies demonstrated that circulating cholesteryl ester transfer protein (CETP) derives largely from hepatic Kupffer cells, and its levels parallel the severity of histological necroinflammations in NASH (111). Of note, CETP inhibitors alleviate high-fat diet–induced steatohepatitis and fibrosis, possibly by reducing oxidized LDL uptake by hepatic Kupffer and stellate cells (112), and represent a potential therapeutic target for the treatment of both NAFLD and CKD (Table 2).

Syndecan-1 is another key mediator of hepatic clearance of triglyceride-rich lipoproteins (TRLPs) (113). It is a transmembrane heparan sulfate proteoglycan constitutively bound to hepatocyte membrane, where it binds lipoprotein lipase and apolipoprotein (apo) E through its heparan sulfate chains and internalizes apoE-containing lipoproteins (31). Sulfation by hepatic sulfotransferases and binding to hepatocyte membrane are required for syndecan-1 biological activity. NAFLD is characterized by an increased shedding of syndecan-1 (114), as a result of increased hepatic metalloproteinase activity, and by a defective syndecan-1 sulfation, as a result of defective hepatic sulfotransferase activity (115,116). These changes in hepatic syndecan-1 metabolism impair TRLP clearance and, accordingly, predicted atherogenic dyslipidemia in CKD (117). Beside mediating hepatic TRLP clearance, syndecan-1 is a key constituent of the endothelial glycocalyx layer, and its shedding has been associated with loss of endothelial barrier integrity and endothelial dysfunction across progressive CKD stages (118). Inhibitors of syndecan-1 shedding, including the phospholipid sphingosine-1-phosphate, restored endothelial integrity experimentally (119) and may represent a potential therapeutic tool for the treatment of CKD.

FGF21

FGFs are signaling proteins that regulate embryonic development, tissue regeneration, and diverse metabolic functions by binding extracellularly to four cell surface tyrosine kinase FGF receptors (FGFRs 1–4) (120). FGF21 is mainly secreted by the liver and exerts its multiple beneficial metabolic effects by binding to FGFRs in the presence of coreceptor β-Klotho (120). FGF21 administration ameliorates adipose and hepatic insulin sensitivity, suppresses hepatic gluconeogenesis and lipogenesis, and enhances free fatty acid (FFA) oxidation and mitochondrial function, at least in part by activating the AMPK-SIRT1-PGC-1α pathway (32,120). Furthermore, FGF21 has recently been demonstrated to direct anti-inflammatory and antifibrogenic activity by inhibiting the key nuclear factor κB (NF-κB) and transforming growth factor-β (TGF-β)/mothers against decapentaplegic homolog (SMAD) 2/3 signaling pathways (121). By virtue of these properties, FGF21 administration has improved experimental NASH and CKD (32,120,121). The clinical development of FGF21, however, is hampered by its short half-life (0.5–5 h) and by tissue FGF21 resistance, which is subtended by FGFR1 and β-Klotho downregulation (122) and is overcome by pharmacological doses of FGF21. To this aim, engineering of the native molecule yielded FGF21 analogs with improved biophysical properties, and one of these FGF21 analogs, LY2405319, ameliorated atherogenic dyslipidemia, insulin resistance, and adiponectin in obese patients with diabetes (123).

Sodium-glucose cotransporter-2 (SGLT2) inhibitors block the activity of the SGLT2 protein, which is expressed in the S1 segment of the renal proximal tubule, leading to substantial glucosuria and a reduction in plasma glucose levels. Experimental evidence has demonstrated that SGLT2 inhibitors confer nephroprotection independently of their glucose-lowering or blood pressure–lowering properties. SGLT2 inhibitors attenuate glomerular hyperfiltration, which is believed to be the initial pathogenic alteration in diabetic and obesity-related CKD. In diabetes, hyperglycemia induces an increase in SGLT2-mediated proximal tubule NaCl reabsorption. The consequent reduction in NaCl distal delivery to the macula densa decreases tubuloglomerular feedback–mediated afferent arteriolar vasoconstriction, thereby increasing glomerular afferent-to-efferent arteriolar tone, intraglomerular ultrafiltration pressure, and glomerular filtration rate (124). Proximal tubule SGLT2 upregulation and increased NaCl reabsorption also have been documented in obesity-related CKD as a result of enhanced sympathetic activity (33,125) and TGF-β1/SMAD3 axis activation (126). By virtue of these actions, SGLT2 inhibitors synergize with renin-angiotensin-aldosterone system (RAAS) inhibitors, and their combination may confer incremental renal benefits (127). Simultaneous SGLT2-RAAS blockade induces afferent arteriole constriction (SGLT2 inhibition) and efferent arteriole vasodilatation (RAAS blockade), thereby more thoroughly counteracting early intrarenal hemodynamic abnormalities underlying glomerular hyperfiltration in CKD. Additionally, SGLT2 inhibitors have decreased inflammatory and fibrogenic responses, oxidative stress, and cell apoptosis in diverse experimental models of CKD (128).

Available data on the impact of SGLT2 inhibitors on CKD have derived from an analysis of RCTs conducted with efficacy and safety end points in a diabetic population where SGLT2 inhibitors slowed renal function decline and reduced albuminuria independently of glycemic control (129), and dedicated nephroprotection trials are under way (Evaluation of the Effects of Canagliflozinon Renal and Cardiovascular Outcomes in Participants With Diabetic Nephropathy [CREDENCE]; clinical trial reg. no. NCT02065791, www.clinicaltrials.gov). Besides nephroprotection, SGLT2 inhibitors have the potential for cardiovascular protection. In the EMPA-REG OUTCOME [BI 10773 (Empagliflozin) Cardiovascular Outcome Event Trial in Type 2 Diabetes Mellitus Patients] trial, empagliflozin added to standard care was associated with a lower rate of the primary composite outcome of cardiovascular and all-cause mortality (130).

SGLT2 inhibitors may also ameliorate NAFLD. In a post hoc analysis of an RCT and a small RCT, remogliflozin etabonate and luseogliflozin improved liver fat accumulation and liver enzymes in patients with diabetes (131,132). The conclusions of these RCTs are supported by preclinical data, whereby SGLT2 inhibitors prevented diet-induced hepatic steatosis, inflammation, and fibrosis independently of antihyperglycemic action (34,35). Potential mechanisms underlying liver-related benefits of these drugs include insulin sensitizing (131) and body fat loss–inducing properties mediated by enhanced lipolysis and fatty acid oxidation (34,35), attenuation of adipose tissue dysfunction and inflammation (133), increased ACE2 activation (134), and intrinsic drug-specific antioxidant and anti-RAGE axis activation properties (36). Furthermore, SGLT2 mRNA expression has been documented in the liver, where its biological and clinical significance remain unknown (135). Several issues with SGLT2 inhibitors warrant assessment, including the renoprotective effects in patients with CKD but without diabetes, the impact on liver histology, long-term safety in patients with various degrees of renal and hepatic impairment, and risk of ketoacidosis, which led the U.S. Food and Drug Administration to issue a safety warning in 2015 (136).

Despite progress made in the past decade, CKD still remains a major health problem for two reasons. CKD often goes unrecognized, and the current therapeutic armamentarium has limited effectiveness in retarding disease progression. NAFLD is the most common chronic liver disease and, given the lack of an effective treatment, is becoming the leading indication for liver transplantation in the Western world (5). The shared unmet needs of NAFLD and CKD, therefore, are boosting research on novel therapeutic targets in these two conditions.

Epidemiological data suggest a tight relationship between the presence and severity of NAFLD and the presence and stage of CKD and place NAFLD as an important contributor to the development and progression of CKD independently of traditional risk factors. When analyzing the pathophysiological basis for this association, striking analogies can be found between fatty liver and CKD. Like NAFLD, CKD is characterized by a deranged cellular substrate metabolism; ectopic fat deposition, which triggers oxidative stress; and inflammatory and profibrotic responses to drive the progression of both disease processes. This review shows a wealth of cellular pathways and mechanisms that represent key contributors to liver and kidney injury and potential therapeutic targets (Fig. 4). Most of these targets currently are being evaluated in phase II RCTs, and some of them, such as PPAR-α/δ agonists, FXR agonists, and incretin analogs, have promising results (137), although few of them have advanced to the same developmental stage in both NAFLD and CKD, reflecting a continued low awareness of the similarities in the pathogenic mechanisms underlying these two conditions. Besides shared pathogenic mechanisms that promote both liver and kidney injury, the fatty liver per se may promote kidney injury and vice versa, with potentially relevant therapeutic implications. For example, SGLT2 inhibitors target the renal tubule but may improve both CKD and NAFLD. Whether the relative merits of the various therapeutic approaches will translate into a clinical benefit needs assessment in adequately powered, larger RCTs of longer duration with clinical end points. A key challenge to therapeutic success of these various approaches will be the selection of the optimal therapeutic strategy for each patient. NAFLD and CKD progression is likely a multifactorial process, involving varied molecular pathways that may operate in different patient subsets and at different stages of disease. Within this context, recent developments in metabolic phenotyping with metabolomics and systems biology technologies will hopefully enable individualized treatment tailored to individual profiles. Given the increasing prevalence of CKD and NAFLD and their direct effects on and acceleration of CVD, strategies to reduce the incidence, progression, and complications of these twin conditions are an important priority in health care.

Figure 4

Molecular pathways mediating the interplay among the liver, kidney, gut, and adipose tissue in the pathogenesis of NAFLD and CKD and the effect of their modulation. A: In NAFLD-associated CKD, the gut promotes liver and kidney injury by reduced incretin GLP-1 and by enhanced microbial production of LPS and URMs, the excretion of which is reduced in CKD, further increasing their systemic levels. Further injurious mechanisms contributing to NASH and CKD at a systemic level include inappropriate activation of SREBP-2, which promotes intracellular cholesterol accumulation; cellular sensors HIF-1 and mTORC1; and oxidative stress–activated ASK1. Collectively, these factors induce insulin resistance, ectopic lipid accumulation that triggers lipoperoxidative stress and enhances secretion of proinflammatory cytokines, chemokines CCL2 and CCL5, and profibrogenic factors such as miRNA21 and galectin-3. The liver promotes CKD through enhanced secretion of uric acid and proatherogenic factors, including VLDL1, CETP, and syndecan-1, which promote atherogenic dyslipidemia, endothelial dysfunction, and renal vascular disease. Additionally, in NAFLD, there is an impaired action of hepatokine FGF21, the systemic levels of which are elevated because of tissue FGF21 resistance and impaired renal FGF21 excretion in CKD. B: Besides directly antagonizing the action of noxious factors described in A, NAFLD and CKD can be ameliorated at intestinal levels by incretin mimetics and modulation of gut microbiota composition with prebiotics, probiotics, or synbiotics and at systemic levels by activating cellular energy sensor AMPK and several nuclear transcription factors, including PPAR-α, PPAR-δ, PPAR-γ, FXR, and Nrf2. Tables 13 and the text provide a detailed description of molecular mechanisms underlying the gut-liver-kidney connection. Chol, cholesterol; IR, insulin resistance.

Figure 4

Molecular pathways mediating the interplay among the liver, kidney, gut, and adipose tissue in the pathogenesis of NAFLD and CKD and the effect of their modulation. A: In NAFLD-associated CKD, the gut promotes liver and kidney injury by reduced incretin GLP-1 and by enhanced microbial production of LPS and URMs, the excretion of which is reduced in CKD, further increasing their systemic levels. Further injurious mechanisms contributing to NASH and CKD at a systemic level include inappropriate activation of SREBP-2, which promotes intracellular cholesterol accumulation; cellular sensors HIF-1 and mTORC1; and oxidative stress–activated ASK1. Collectively, these factors induce insulin resistance, ectopic lipid accumulation that triggers lipoperoxidative stress and enhances secretion of proinflammatory cytokines, chemokines CCL2 and CCL5, and profibrogenic factors such as miRNA21 and galectin-3. The liver promotes CKD through enhanced secretion of uric acid and proatherogenic factors, including VLDL1, CETP, and syndecan-1, which promote atherogenic dyslipidemia, endothelial dysfunction, and renal vascular disease. Additionally, in NAFLD, there is an impaired action of hepatokine FGF21, the systemic levels of which are elevated because of tissue FGF21 resistance and impaired renal FGF21 excretion in CKD. B: Besides directly antagonizing the action of noxious factors described in A, NAFLD and CKD can be ameliorated at intestinal levels by incretin mimetics and modulation of gut microbiota composition with prebiotics, probiotics, or synbiotics and at systemic levels by activating cellular energy sensor AMPK and several nuclear transcription factors, including PPAR-α, PPAR-δ, PPAR-γ, FXR, and Nrf2. Tables 13 and the text provide a detailed description of molecular mechanisms underlying the gut-liver-kidney connection. Chol, cholesterol; IR, insulin resistance.

Close modal

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. G.M. contributed to the study conception and design, literature search, data acquisition, critical analysis of the results, and drafting and final approval of the manuscript. M.C., S.C., F.D.M., S.P., F.S., and R.G. contributed to the literature search, data acquisition, critical analysis of the results, and drafting and final approval of the manuscript. G.M. is the guarantor of this work and, as such, had full access to all the data in the study and takes responsibility for the integrity of the data and the accuracy of the data analysis.

1.
McCullough
K
,
Sharma
P
,
Ali
T
, et al
.
Measuring the population burden of chronic kidney disease: a systematic literature review of the estimated prevalence of impaired kidney function
.
Nephrol Dial Transplant
2012
;
27
:
1812
1821
[PubMed]
2.
Palmer
SC
,
Mavridis
D
,
Navarese
E
, et al
.
Comparative efficacy and safety of blood pressure-lowering agents in adults with diabetes and kidney disease: a network meta-analysis
.
Lancet
2015
;
385
:
2047
2056
[PubMed]
3.
Musso
G
,
Gambino
R
,
Tabibian
JH
, et al
.
Association of non-alcoholic fatty liver disease with chronic kidney disease: a systematic review and meta-analysis
.
PLoS Med
2014
;
11
:
e1001680
[PubMed]
4.
Stepanova M, de Avila L, Birerdinc A, et al. In female patients with non-alcoholic fatty liver disease (NAFLD) presence, of type 2 diabetes (DM) and chronic kidney disease (CKD) are independently associated with the risk of mortality. Hepatology 2015;62(Suppl.):2205
5.
Singal
AK
,
Salameh
H
,
Kuo
YF
,
Wiesner
RH
.
Evolving frequency and outcomes of simultaneous liver kidney transplants based on liver disease etiology
.
Transplantation
2014
;
98
:
216
221
[PubMed]
6.
Mikolasevic
I
,
Racki
S
,
Zaputovic
L
,
Lukenda
V
,
Sladoje-Martinovic
B
,
Orlic
L
.
Nonalcoholic fatty liver disease (NAFLD) and cardiovascular risk in renal transplant recipients
.
Kidney Blood Press Res
2014
;
39
:
308
314
[PubMed]
7.
Sugar and Sweeteners Team, Market and Trade Economics, Economic Research Service, US Department of Agriculture. US per capita caloric sweeteners estimated deliveries for domestic food and beverage use, by calendar year. Available at: http://www.ers.usda.gov/data-products/sugar-and-sweeteners-yearbook-tables.aspx
8.
Johnson
RJ
,
Nakagawa
T
,
Sanchez-Lozada
LG
, et al
.
Sugar, uric acid, and the etiology of diabetes and obesity
.
Diabetes
2013
;
62
:
3307
3315
[PubMed]
9.
Fan
CY
,
Wang
MX
,
Ge
CX
,
Wang
X
,
Li
JM
,
Kong
LD
.
Betaine supplementation protects against high-fructose-induced renal injury in rats
.
J Nutr Biochem
2014
;
25
:
353
362
[PubMed]
10.
Sanyal
AJ
,
Harrison
SA
,
Francque
SM
, et al
.
The hepatic and extrahepatic profile of resolution of steatohepatitis induced by GFT-505
.
Hepatology
2015
;
62
:
1252A
11.
Musso
G
,
Cassader
M
,
Rosina
F
,
Gambino
R
.
Impact of current treatments on liver disease, glucose metabolism and cardiovascular risk in non-alcoholic fatty liver disease (NAFLD): a systematic review and meta-analysis of randomised trials
.
Diabetologia
2012
;
55
:
885
904
[PubMed]
12.
Sarafidis
PA
,
Stafylas
PC
,
Georgianos
PI
,
Saratzis
AN
,
Lasaridis
AN
.
Effect of thiazolidinediones on albuminuria and proteinuria in diabetes: a meta-analysis
.
Am J Kidney Dis
2010
;
55
:
835
847
[PubMed]
13.
Kandasamy
N
,
Ashokkumar
N
.
Renoprotective effect of myricetin restrains dyslipidemia and renal mesangial cell proliferation by the suppression of sterol regulatory element binding proteins in an experimental model of diabetic nephropathy
.
Eur J Pharmacol
2014
;
743
:
53
62
[PubMed]
14.
Chen
Y
,
Zhao
L
,
Li
Q
, et al
.
Inflammatory stress reduces the effectiveness of statins in the kidney by disrupting HMGCoA reductase feedback regulation
.
Nephrol Dial Transplant
2014
;
29
:
1864
1878
[PubMed]
15.
Trionfini
P
,
Benigni
A
,
Remuzzi
G
.
MicroRNAs in kidney physiology and disease
.
Nat Rev Nephrol
2015
;
11
:
23
33
[PubMed]
16.
Li
J
,
Wilson
A
,
Gao
X
, et al
.
Coordinated regulation of dimethylarginine dimethylaminohydrolase-1 and cationic amino acid transporter-1 by farnesoid X receptor in mouse liver and kidney and its implication in the control of blood levels of asymmetric dimethylarginine
.
J Pharmacol Exp Ther
2009
;
331
:
234
243
[PubMed]
17.
Neuschwander-Tetri
BA
,
Loomba
R
,
Sanyal
AJ
, et al.;
NASH Clinical Research Network
.
Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial
.
Lancet
2015
;
385
:
956
965
[PubMed]
18.
Musso
G
,
Gambino
R
,
Cassader
M
.
Emerging molecular targets for the treatment of nonalcoholic fatty liver disease
.
Annu Rev Med
2010
;
61
:
375
392
[PubMed]
19.
Li
J
,
Pan
Y
,
Kan
M
, et al
.
Hepatoprotective effects of berberine on liver fibrosis via activation of AMP-activated protein kinase
.
Life Sci
2014
;
98
:
24
30
[PubMed]
20.
Soetikno
V
,
Sari
FR
,
Sukumaran
V
, et al
.
Curcumin decreases renal triglyceride accumulation through AMPK-SREBP signaling pathway in streptozotocin-induced type 1 diabetic rats
.
J Nutr Biochem
2013
;
24
:
796
802
[PubMed]
21.
Anavi
S
,
Hahn-Obercyger
M
,
Madar
Z
,
Tirosh
O
.
Mechanism for HIF-1 activation by cholesterol under normoxia: a redox signaling pathway for liver damage
.
Free Radic Biol Med
2014
;
71
:
61
69
[PubMed]
22.
Musso
G
,
Cassader
M
,
Olivetti
C
,
Rosina
F
,
Carbone
G
,
Gambino
R
.
Association of obstructive sleep apnoea with the presence and severity of non-alcoholic fatty liver disease. A systematic review and meta-analysis
.
Obes Rev
2013
;
14
:
417
431
[PubMed]
23.
Kim
YC
,
Guan
KL
.
mTOR: a pharmacologic target for autophagy regulation
.
J Clin Invest
2015
;
125
:
25
32
[PubMed]
24.
Sapp
V
,
Gaffney
L
,
EauClaire
SF
,
Matthews
RP
.
Fructose leads to hepatic steatosis in zebrafish that is reversed by mechanistic target of rapamycin (mTOR) inhibition
.
Hepatology
2014
;
60
:
1581
1592
[PubMed]
25.
Chen
H
,
Zhu
J
,
Liu
Y
, et al
.
Lipopolysaccharide induces chronic kidney injury and fibrosis through activation of mTOR signaling in macrophages
.
Am J Nephrol
2015
;
42
:
305
317
[PubMed]
26.
Calvier
L
,
Martinez-Martinez
E
,
Miana
M
, et al
.
The impact of galectin-3 inhibition on aldosterone-induced cardiac and renal injuries
.
JACC Heart Fail
2015
;
3
:
59
67
[PubMed]
27.
Imamura
S
,
Hirai
K
,
Hirai
A
.
The glucagon-like peptide-1 receptor agonist, liraglutide, attenuates the progression of overt diabetic nephropathy in type 2 diabetic patients
.
Tohoku J Exp Med
2013
;
231
:
57
61
[PubMed]
28.
Bergenstal
RM
,
Wysham
C
,
Macconell
L
, et al.;
DURATION-2 Study Group
.
Efficacy and safety of exenatide once weekly versus sitagliptin or pioglitazone as an adjunct to metformin for treatment of type 2 diabetes (DURATION-2): a randomised trial
.
Lancet
2010
;
376
:
431
439
[PubMed]
29.
Scirica
BM
,
Bhatt
DL
,
Braunwald
E
, et al.;
SAVOR-TIMI 53 Steering Committee and Investigators
.
Saxagliptin and cardiovascular outcomes in patients with type 2 diabetes mellitus
.
N Engl J Med
2013
;
369
:
1317
1326
[PubMed]
30.
Musso
G
,
Cassader
M
,
Cohney
S
,
Pinach
S
,
Saba
F
,
Gambino
R
.
Emerging liver-kidney interactions in nonalcoholic fatty liver disease
.
Trends Mol Med
2015
;
21
:
645
662
[PubMed]
31.
Stanford
KI
,
Wang
L
,
Castagnola
J
, et al
.
Heparan sulfate 2-O-sulfotransferase is required for triglyceride-rich lipoprotein clearance
.
J Biol Chem
2010
;
285
:
286
294
[PubMed]
32.
Xu
J
,
Lloyd
DJ
,
Hale
C
, et al
.
Fibroblast growth factor 21 reverses hepatic steatosis, increases energy expenditure, and improves insulin sensitivity in diet-induced obese mice
.
Diabetes
2009
;
58
:
250
259
[PubMed]
33.
Zingerman
B
,
Herman-Edelstein
M
,
Erman
A
, et al
.
Effect of acetazolamide on obesity-induced glomerular hyperfiltration: a randomized controlled trial
.
PLoS One
2015
;
10
:
e0137163
[PubMed]
34.
Hayashizaki-Someya
Y
,
Kurosaki
E
,
Takasu
T
, et al
.
Ipragliflozin, an SGLT2 inhibitor, exhibits a prophylactic effect on hepatic steatosis and fibrosis induced by choline-deficient l-amino acid-defined diet in rats
.
Eur J Pharmacol
2015
;
754
:
19
24
[PubMed]
35.
Yokono
M
,
Takasu
T
,
Hayashizaki
Y
, et al
.
SGLT2 selective inhibitor ipragliflozin reduces body fat mass by increasing fatty acid oxidation in high-fat diet-induced obese rats
.
Eur J Pharmacol
2014
;
727
:
66
74
[PubMed]
36.
Ojima
A
,
Matsui
T
,
Nishino
Y
,
Nakamura
N
,
Yamagishi
S
.
Empagliflozin, an inhibitor of sodium-glucose cotransporter 2 exerts anti-inflammatory and antifibrotic effects on experimental diabetic nephropathy partly by suppressing AGEs-receptor axis
.
Horm Metab Res
2015
;
47
:
686
692
[PubMed]
37.
Yetley
EA
.
Assessing the vitamin D status of the US population
.
Am J Clin Nutr
2008
;
88
:
558S
564S
[PubMed]
38.
Barchetta
I
,
Carotti
S
,
Labbadia
G
, et al
.
Liver vitamin D receptor, CYP2R1, and CYP27A1 expression: relationship with liver histology and vitamin D3 levels in patients with nonalcoholic steatohepatitis or hepatitis C virus
.
Hepatology
2012
;
56
:
2180
2187
[PubMed]
39.
Beilfuss A, Sowa JP, Sydor S. Vitamin D counteracts fibrogenic TGF-β signalling in human hepatic stellate cells both receptor-dependently and independently. Gut 2015;64:791–799
40.
Wang
XX
,
Jiang
T
,
Shen
Y
, et al
.
Vitamin D receptor agonist doxercalciferol modulates dietary fat-induced renal disease and renal lipid metabolism
.
Am J Physiol Renal Physiol
2011
;
300
:
F801
F810
[PubMed]
41.
Xu
L
,
Wan
X
,
Huang
Z
, et al
.
Impact of vitamin D on chronic kidney diseases in non-dialysis patients: a meta-analysis of randomized controlled trials
.
PLoS One
2013
;
8
:
e61387
[PubMed]
42.
Dasarathy
J
,
Varghese
R
,
Kalinina
I
,
Lopez
R
,
McCullough
AJ
.
Hypovitaminosis D in NAFLD requires high dose supplementation to reverse deficiency
.
Hepatology
2015
;
62
:
1273A
43.
Parikh A, Chase HS, Vernocchi L, Stern L. Vitamin D resistance in chronic kidney disease (CKD). BMC Nephrol 2014 19;15:47
44.
Xu
Y
,
Huang
J
,
Xin
W
, et al
.
Lipid accumulation is ahead of epithelial-to-mesenchymal transition and therapeutic intervention by acetyl-CoA carboxylase 2 silence in diabetic nephropathy
.
Metabolism
2014
;
63
:
716
726
[PubMed]
45.
Xin
W
,
Zhao
X
,
Liu
L
, et al
.
Acetyl-CoA carboxylase 2 suppression rescues human proximal tubular cells from palmitic acid induced lipotoxicity via autophagy
.
Biochem Biophys Res Commun
2015
;
463
:
364
369
[PubMed]
46.
Herman-Edelstein
M
,
Scherzer
P
,
Tobar
A
,
Levi
M
,
Gafter
U
.
Altered renal lipid metabolism and renal lipid accumulation in human diabetic nephropathy
.
J Lipid Res
2014
;
55
:
561
572
[PubMed]
47.
Souza-Mello
V
.
Peroxisome proliferator-activated receptors as targets to treat non-alcoholic fatty liver disease
.
World J Hepatol
2015
;
7
:
1012
1019
[PubMed]
48.
Pawlak
M
,
Baugé
E
,
Bourguet
W
, et al
.
The transrepressive activity of peroxisome proliferator-activated receptor alpha is necessary and sufficient to prevent liver fibrosis in mice
.
Hepatology
2014
;
60
:
1593
1606
[PubMed]
49.
Zhou Y, Lin S, Zhang L, Li Y. Resveratrol prevents renal lipotoxicity in high-fat diet-treated mouse model through regulating PPAR-α pathway. Mol Cell Biochem 2016;411:143–150
50.
Collino
M
,
Benetti
E
,
Rogazzo
M
, et al
.
Reversal of the deleterious effects of chronic dietary HFCS-55 intake by PPAR-δ agonism correlates with impaired NLRP3 inflammasome activation
.
Biochem Pharmacol
2013
;
85
:
257
264
[PubMed]
51.
Lee
EY
,
Kim
GT
,
Hyun
M
, et al
.
Peroxisome proliferator-activated receptor-δ activation ameliorates albuminuria by preventing nephrin loss and restoring podocyte integrity in type 2 diabetes
.
Nephrol Dial Transplant
2012
;
27
:
4069
4079
[PubMed]
52.
Shashank R. Saroglitazar in diabetic dyslipidemia. Abstract 126-LB presented at the 75th Scientific Sessions of the American Diabetes Association, 5–9 June 2015, Boston, Massachusetts
53.
Banshi D. To assess the effect of 4mg saroglitazar on patients of diabetes dyslipidemia with nonalcoholic fatty liver disease for 24 weeks. Abstract 712-P presented at the 75th Scientific Sessions of the American Diabetes Association, 5–9 June 2015, Boston, Massachusetts
54.
Ruilope
L
,
Hanefeld
M
,
Lincoff
AM
, et al
.
Effects of the dual peroxisome proliferator-activated receptor-α/γ agonist aleglitazar on renal function in patients with stage 3 chronic kidney disease and type 2 diabetes: a Phase IIb, randomized study
.
BMC Nephrol
2014
;
15
:
180
[PubMed]
55.
Musso
G
,
Gambino
R
,
Cassader
M
.
Cholesterol metabolism and the pathogenesis of non-alcoholic steatohepatitis
.
Prog Lipid Res
2013
;
52
:
175
191
[PubMed]
56.
Hu
Z
,
Ren
L
,
Wang
C
,
Liu
B
,
Song
G
.
Effect of chenodeoxycholic acid on fibrosis, inflammation and oxidative stress in kidney in high-fructose-fed Wistar rats
.
Kidney Blood Press Res
2012
;
36
:
85
97
[PubMed]
57.
Wang
XX
,
Jiang
T
,
Shen
Y
, et al
.
The farnesoid X receptor modulates renal lipid metabolism and diet-induced renal inflammation, fibrosis, and proteinuria
.
Am J Physiol Renal Physiol
2009
;
297
:
F1587
F1596
[PubMed]
58.
Gerhard
GS
,
DiStefano
JK
.
Micro RNAs in the development of non-alcoholic fatty liver disease
.
World J Hepatol
2015
;
7
:
226
234
[PubMed]
59.
Dattaroy
D
,
Pourhoseini
S
,
Das
S
, et al
.
Micro-RNA 21 inhibition of SMAD7 enhances fibrogenesis via leptin-mediated NADPH oxidase in experimental and human nonalcoholic steatohepatitis
.
Am J Physiol Gastrointest Liver Physiol
2015
;
308
:
G298
G312
[PubMed]
60.
Gomez
IG
,
MacKenna
DA
,
Johnson
BG
, et al
.
Anti-microRNA-21 oligonucleotides prevent Alport nephropathy progression by stimulating metabolic pathways
.
J Clin Invest
2015
;
125
:
141
156
[PubMed]
61.
Hsu
WH
,
Chen
TH
,
Lee
BH
,
Hsu
YW
,
Pan
TM
.
Monascin and ankaflavin act as natural AMPK activators with PPARα agonist activity to down-regulate nonalcoholic steatohepatitis in high-fat diet-fed C57BL/6 mice
.
Food Chem Toxicol
2014
;
64
:
94
103
[PubMed]
62.
Musso
G
,
Olivetti
C
,
Cassader
M
,
Gambino
R
.
Obstructive sleep apnea-hypopnea syndrome and nonalcoholic fatty liver disease: emerging evidence and mechanisms
.
Semin Liver Dis
2012
;
32
:
49
64
[PubMed]
63.
Shoji
K
,
Tanaka
T
,
Nangaku
M
.
Role of hypoxia in progressive chronic kidney disease and implications for therapy
.
Curr Opin Nephrol Hypertens
2014
;
23
:
161
168
[PubMed]
64.
Luo
R
,
Zhang
W
,
Zhao
C
, et al
.
Elevated endothelial hypoxia-inducible factor-1α contributes to glomerular injury and promotes hypertensive chronic kidney disease
.
Hypertension
2015
;
66
:
75
84
[PubMed]
65.
Xiao
J
,
Jin
C
,
Liu
Z
, et al
.
The design, synthesis, and biological evaluation of novel YC-1 derivatives as potent anti-hepatic fibrosis agents
.
Org Biomol Chem
2015
;
13
:
7257
7264
[PubMed]
66.
Nordquist
L
,
Friederich-Persson
M
,
Fasching
A
, et al
.
Activation of hypoxia-inducible factors prevents diabetic nephropathy
.
J Am Soc Nephrol
2015
;
26
:
328
338
[PubMed]
67.
Xu Y, Liu L, Xin W, et al. The renoprotective role of autophagy activation in proximal tubular epithelial cells in diabetic nephropathy. J Diabetes Complications 2015;29:976–983
68.
Jiang
H
,
Westerterp
M
,
Wang
C
,
Zhu
Y
,
Ai
D
.
Macrophage mTORC1 disruption reduces inflammation and insulin resistance in obese mice
.
Diabetologia
2014
;
57
:
2393
2404
[PubMed]
69.
Wang
L
,
Liu
X
,
Nie
J
, et al
.
ALCAT1 controls mitochondrial etiology of fatty liver diseases, linking defective mitophagy to steatosis
.
Hepatology
2015
;
61
:
486
496
[PubMed]
70.
Torricelli C, Daveri E, Salvadori S, et al. Phosphorylation-independent mTORC1 inhibition by the autophagy inducer Rottlerin. Cancer Lett 2015;360:17–27
71.
Kawarazaki
Y
,
Ichijo
H
,
Naguro
I
.
Apoptosis signal-regulating kinase 1 as a therapeutic target
.
Expert Opin Ther Targets
2014
;
18
:
651
664
[PubMed]
72.
Yamamoto
E
,
Dong
YF
,
Kataoka
K
, et al
.
Olmesartan prevents cardiovascular injury and hepatic steatosis in obesity and diabetes, accompanied by apoptosis signal regulating kinase-1 inhibition
.
Hypertension
2008
;
52
:
573
580
[PubMed]
73.
Tesch
GH
,
Ma
FY
,
Han
Y
,
Liles
JT
,
Breckenridge
DG
,
Nikolic-Paterson
DJ
.
ASK1 inhibitor halts progression of diabetic nephropathy in Nos3-deficient mice
.
Diabetes
2015
;
64
:
3903
3913
[PubMed]
74.
Tebay
LE
,
Robertson
H
,
Durant
ST
, et al
.
Mechanisms of activation of the transcription factor Nrf2 by redox stressors, nutrient cues, and energy status and the pathways through which it attenuates degenerative disease
.
Free Radic Biol Med
2015
;
88
:
108
146
[PubMed]
75.
Shimozono
R
,
Asaoka
Y
,
Yoshizawa
Y
, et al
.
Nrf2 activators attenuate the progression of nonalcoholic steatohepatitis-related fibrosis in a dietary rat model
.
Mol Pharmacol
2013
;
84
:
62
70
[PubMed]
76.
Choi
BH
,
Kang
KS
,
Kwak
MK
.
Effect of redox modulating NRF2 activators on chronic kidney disease
.
Molecules
2014
;
19
:
12727
12759
[PubMed]
77.
de Zeeuw
D
,
Akizawa
T
,
Agarwal
R
, et al
.
Rationale and trial design of bardoxolone methyl evaluation in patients with chronic kidney disease and type 2 diabetes: the occurrence of renal events (BEACON)
.
Am J Nephrol
2013
;
37
:
212
222
[PubMed]
78.
Kufareva
I
,
Salanga
CL
,
Handel
TM
.
Chemokine and chemokine receptor structure and interactions: implications for therapeutic strategies
.
Immunol Cell Biol
2015
;
93
:
372
383
[PubMed]
79.
Oh
DY
,
Morinaga
H
,
Talukdar
S
,
Bae
EJ
,
Olefsky
JM
.
Increased macrophage migration into adipose tissue in obese mice
.
Diabetes
2012
;
61
:
346
354
[PubMed]
80.
Baeck
C
,
Wei
X
,
Bartneck
M
, et al
.
Pharmacological inhibition of the chemokine C-C motif chemokine ligand 2 (monocyte chemoattractant protein 1) accelerates liver fibrosis regression by suppressing Ly-6C(+) macrophage infiltration in mice
.
Hepatology
2014
;
59
:
1060
1072
[PubMed]
81.
Moreno
JA
,
Moreno
S
,
Rubio-Navarro
A
, et al
.
Targeting chemokines in proteinuria-induced renal disease
.
Expert Opin Ther Targets
2012
;
16
:
833
845
[PubMed]
82.
de Zeeuw D, Bekker P, Henkel E, et al. The effect of CCR2 inhibitor CCX140-B on residual albuminuria in patients with type 2 diabetes and nephropathy: a randomised trial. Lancet Diabetes Endocrinol 2015;3:687–696
83.
Pugliese
G
,
Iacobini
C
,
Pesce
CM
,
Menini
S
.
Galectin-3: an emerging all-out player in metabolic disorders and their complications
.
Glycobiology
2015
;
25
:
136
150
[PubMed]
84.
Iacobini
C
,
Menini
S
,
Ricci
C
, et al
.
Galectin-3 ablation protects mice from diet-induced NASH: a major scavenging role for galectin-3 in liver
.
J Hepatol
2011
;
54
:
975
983
[PubMed]
85.
Drechsler
C
,
Delgado
G
,
Wanner
C
, et al
.
Galectin-3, renal function, and clinical outcomes: results from the LURIC and 4D studies
.
J Am Soc Nephrol
2015
;
26
:
2213
2221
[PubMed]
86.
Frenay
AR
,
Yu
L
,
van der Velde
AR
, et al
.
Pharmacological inhibition of galectin-3 protects against hypertensive nephropathy
.
Am J Physiol Renal Physiol
2015
;
308
:
F500
F509
[PubMed]
87.
Traber
PG
,
Chou
H
,
Zomer
E
, et al
.
Regression of fibrosis and reversal of cirrhosis in rats by galectin inhibitors in thioacetamide-induced liver disease
.
PLoS One
2013
;
8
:
e75361
[PubMed]
88.
Pang
J
,
Rhodes
DH
,
Pini
M
, et al
.
Increased adiposity, dysregulated glucose metabolism and systemic inflammation in Galectin-3 KO mice
.
PLoS One
2013
;
8
:
e57915
[PubMed]
89.
Zhou
SW
,
Zhang
M
,
Zhu
M
.
Liraglutide reduces lipid accumulation in steatotic L‑02 cells by enhancing autophagy
.
Mol Med Rep
2014
;
10
:
2351
2357
[PubMed]
90.
Hwang
HJ
,
Jung
TW
,
Kim
BH
, et al
.
A dipeptidyl peptidase-IV inhibitor improves hepatic steatosis and insulin resistance by AMPK-dependent and JNK-dependent inhibition of LECT2 expression
.
Biochem Pharmacol
2015
;
98
:
157
166
[PubMed]
91.
Armstrong
MJ
,
Houlihan
DD
,
Rowe
IA
, et al
.
Safety and efficacy of liraglutide in patients with type 2 diabetes and elevated liver enzymes: individual patient data meta-analysis of the LEAD program
.
Aliment Pharmacol Ther
2013
;
37
:
234
242
[PubMed]
92.
Armstrong MJ, Hull D, Guo K, et al. Glucagon-like peptide 1 decreases lipotoxicity in non-alcoholic steatohepatitis. J Hepatol 2016;64:399–408
93.
Skov
J
,
Dejgaard
A
,
Frøkiær
J
, et al
.
Glucagon-like peptide-1 (GLP-1): effect on kidney hemodynamics and renin-angiotensin-aldosterone system in healthy men
.
J Clin Endocrinol Metab
2013
;
98
:
E664
E671
[PubMed]
94.
Zhang
H
,
Zhang
X
,
Hu
C
,
Lu
W
.
Exenatide reduces urinary transforming growth factor-β1 and type IV collagen excretion in patients with type 2 diabetes and microalbuminuria
.
Kidney Blood Press Res
2012
;
35
:
483
488
[PubMed]
95.
Groop
PH
,
Cooper
ME
,
Perkovic
V
,
Emser
A
,
Woerle
HJ
,
von Eynatten
M
.
Linagliptin lowers albuminuria on top of recommended standard treatment in patients with type 2 diabetes and renal dysfunction
.
Diabetes Care
2013
;
36
:
3460
3468
[PubMed]
96.
Sanofi. Sanofi announces top-line results for cardiovascular outcomes study of Lyxumia® (lixisenatide), 2015. Available from http://www.news.sanofi.us/2015-03-19-Sanofi-Announces-Top-Line-Results-for-Cardiovascular-Outcomes-Study-of-Lyxumia-lixisenatide. Accessed 14 September 2015
97.
Musso
G
,
Gambino
R
,
Cassader
M
.
Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes
.
Annu Rev Med
2011
;
62
:
361
380
[PubMed]
98.
Wong
J
,
Piceno
YM
,
Desantis
TZ
,
Pahl
M
,
Andersen
GL
,
Vaziri
ND
.
Expansion of urease- and uricase-containing, indole- and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD
.
Am J Nephrol
2014
;
39
:
230
237
[PubMed]
99.
Vaziri
ND
,
Yuan
J
,
Nazertehrani
S
,
Ni
Z
,
Liu
S
.
Chronic kidney disease causes disruption of gastric and small intestinal epithelial tight junction
.
Am J Nephrol
2013
;
38
:
99
103
[PubMed]
100.
Adesso
S
,
Popolo
A
,
Bianco
G
, et al
.
The uremic toxin indoxyl sulphate enhances macrophage response to LPS
.
PLoS One
2013
;
8
:
e76778
[PubMed]
101.
Koppe
L
,
Pillon
NJ
,
Vella
RE
, et al
.
p-Cresyl sulfate promotes insulin resistance associated with CKD
.
J Am Soc Nephrol
2013
;
24
:
88
99
[PubMed]
102.
Wu
IW
,
Hsu
KH
,
Lee
CC
, et al
.
p-Cresyl sulphate and indoxyl sulphate predict progression of chronic kidney disease
.
Nephrol Dial Transplant
2011
;
26
:
938
947
[PubMed]
103.
Nymark
M
,
Pussinen
PJ
,
Tuomainen
AM
,
Forsblom
C
,
Groop
PH
,
Lehto
M
;
FinnDiane Study Group
.
Serum lipopolysaccharide activity is associated with the progression of kidney disease in Finnish patients with type 1 diabetes
.
Diabetes Care
2009
;
32
:
1689
1693
[PubMed]
104.
Tang
WH
,
Wang
Z
,
Kennedy
DJ
, et al.;
Gut Microbiota-Dependent Trimethylamine
.
Gut microbiota-dependent trimethylamine N-oxide (TMAO) pathway contributes to both development of renal insufficiency and mortality risk in chronic kidney disease
.
Circ Res
2015
;
116
:
448
455
[PubMed]
105.
Vaziri
ND
,
Yuan
J
,
Khazaeli
M
,
Masuda
Y
,
Ichii
H
,
Liu
S
.
Oral activated charcoal adsorbent (AST-120) ameliorates chronic kidney disease-induced intestinal epithelial barrier disruption
.
Am J Nephrol
2013
;
37
:
518
525
[PubMed]
106.
Eslamparast
T
,
Poustchi
H
,
Zamani
F
,
Sharafkhah
M
,
Malekzadeh
R
,
Hekmatdoost
A
.
Synbiotic supplementation in nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled pilot study
.
Am J Clin Nutr
2014
;
99
:
535
542
[PubMed]
107.
Rossi M, Klein K, Johnson DW, Campbell KL. Pre-, pro-, and synbiotics: do they have a role in reducing uremic toxins? A systematic review and meta-analysis. Int J Nephrol 2012;2012:673631
108.
Penno
G
,
Solini
A
,
Zoppini
G
, et al.;
Renal Insufficiency And Cardiovascular Events (RIACE) Study Group
.
Hypertriglyceridemia is independently associated with renal, but not retinal complications in subjects with type 2 diabetes: a cross-sectional analysis of the Renal Insufficiency And Cardiovascular Events (RIACE) Italian Multicenter Study
.
PLoS One
2015
;
10
:
e0125512
[PubMed]
109.
Sonmez
A
,
Yilmaz
MI
,
Saglam
M
, et al
.
The role of plasma triglyceride/high-density lipoprotein cholesterol ratio to predict cardiovascular outcomes in chronic kidney disease
.
Lipids Health Dis
2015
;
14
:
29
[PubMed]
110.
Gyebi
L
,
Soltani
Z
,
Reisin
E
.
Lipid nephrotoxicity: new concept for an old disease
.
Curr Hypertens Rep
2012
;
14
:
177
181
[PubMed]
111.
Wang
Y
,
van der Tuin
S
,
Tjeerdema
N
, et al
.
Plasma cholesteryl ester transfer protein is predominantly derived from Kupffer cells
.
Hepatology
2015
;
62
:
1710
1722
[PubMed]
112.
Liaw
YW
,
Lin
CY
,
Lai
YS
, et al
.
A vaccine targeted at CETP alleviates high fat and high cholesterol diet-induced atherosclerosis and non-alcoholic steatohepatitis in rabbit
.
PLoS One
2014
;
9
:
e111529
[PubMed]
113.
Deng
Y
,
Foley
EM
,
Gonzales
JC
,
Gordts
PL
,
Li
Y
,
Esko
JD
.
Shedding of syndecan-1 from human hepatocytes alters very low density lipoprotein clearance
.
Hepatology
2012
;
55
:
277
286
[PubMed]
114.
Yilmaz
Y
,
Eren
F
,
Colak
Y
,
Senates
E
,
Celikel
CA
,
Imeryuz
N
.
Hepatic expression and serum levels of syndecan 1 (CD138) in patients with nonalcoholic fatty liver disease
.
Scand J Gastroenterol
2012
;
47
:
1488
1493
[PubMed]
115.
Okazaki
I
,
Noro
T
,
Tsutsui
N
, et al
.
Fibrogenesis and carcinogenesis in nonalcoholic steatohepatitis (NASH): involvement of matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinase (TIMPs)
.
Cancers (Basel)
2014
;
6
:
1220
1255
[PubMed]
116.
Hardwick
RN
,
Ferreira
DW
,
More
VR
, et al
.
Altered UDP-glucuronosyltransferase and sulfotransferase expression and function during progressive stages of human nonalcoholic fatty liver disease
.
Drug Metab Dispos
2013
;
41
:
554
561
[PubMed]
117.
Adepu
S
,
Katta
K
,
Tietge
UJ
, et al
.
Hepatic syndecan-1 changes associate with dyslipidemia after renal transplantation
.
Am J Transplant
2014
;
14
:
2328
2338
[PubMed]
118.
Padberg
JS
,
Wiesinger
A
,
di Marco
GS
, et al
.
Damage of the endothelial glycocalyx in chronic kidney disease
.
Atherosclerosis
2014
;
234
:
335
343
[PubMed]
119.
Zeng
Y
,
Adamson
RH
,
Curry
FR
,
Tarbell
JM
.
Sphingosine-1-phosphate protects endothelial glycocalyx by inhibiting syndecan-1 shedding
.
Am J Physiol Heart Circ Physiol
2014
;
306
:
H363
H372
[PubMed]
120.
Zhang
J
,
Li
Y
.
Fibroblast growth factor 21 analogs for treating metabolic disorders
.
Front Endocrinol (Lausanne)
2015
;
6
:
168
[PubMed]
121.
Xu P, Zhang Y, Liu Y, et al. Fibroblast growth factor 21 attenuates hepatic fibrogenesis through TGF-β/smad2/3 and NF-κB signaling pathways. Toxicol Appl Pharmacol 2016;290:43–53
122.
Fisher
FM
,
Chui
PC
,
Antonellis
PJ
, et al
.
Obesity is a fibroblast growth factor 21 (FGF21)-resistant state
.
Diabetes
2010
;
59
:
2781
2789
[PubMed]
123.
Gaich
G
,
Chien
JY
,
Fu
H
, et al
.
The effects of LY2405319, an FGF21 analog, in obese human subjects with type 2 diabetes
.
Cell Metab
2013
;
18
:
333
340
[PubMed]
124.
Skrtić
M
,
Yang
GK
,
Perkins
BA
, et al
.
Characterisation of glomerular haemodynamic responses to SGLT2 inhibition in patients with type 1 diabetes and renal hyperfiltration
.
Diabetologia
2014
;
57
:
2599
2602
[PubMed]
125.
Elliott
RH
,
Matthews
VB
,
Rudnicka
C
,
Schlaich
MP
.
Is it time to think about the sodium glucose co-transporter 2 sympathetically
?
Nephrology (Carlton)
2016
;21:286–294
[PubMed]
126.
Panchapakesan
U
,
Pegg
K
,
Gross
S
, et al
.
Effects of SGLT2 inhibition in human kidney proximal tubular cells—renoprotection in diabetic nephropathy
?
PLoS One
2013
;
8
:
e54442
[PubMed]
127.
Lambers Heerspink
HJ
,
Johnsson
E
,
Gause-Nilsson
I
,
Johannson
K
,
Sjostrom
CD
.
Dapagliflozin reduces albuminuria on top of renin-angiotensin system blockade in hypertensive diabetic patients
.
Diabetes
2015
;
64
:
A303
128.
Vallon
V
,
Gerasimova
M
,
Rose
MA
, et al
.
SGLT2 inhibitor empagliflozin reduces renal growth and albuminuria in proportion to hyperglycemia and prevents glomerular hyperfiltration in diabetic Akita mice
.
Am J Physiol Renal Physiol
2014
;
306
:
F194
F204
[PubMed]
129.
Yale
JF
,
Bakris
G
,
Cariou
B
, et al.;
DIA3004 Study Group
.
Efficacy and safety of canagliflozin over 52 weeks in patients with type 2 diabetes mellitus and chronic kidney disease
.
Diabetes Obes Metab
2014
;
16
:
1016
1027
[PubMed]
130.
Zinman
B
,
Wanner
C
,
Lachin
JM
, et al
.
Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes
.
N Engl J Med
2015
;373:2117–2128
131.
Wilkison W, Cheatham B, Walker S. Remogliflozin etabonate reduces insulin resistance and liver function enzymes: Role for treatment of NASH. J Hepatology 2015;62(Suppl. 2): S211–S212
132.
Fushimi
N
,
Shibuya
T
,
Takeishi
S
,
Itou
S
,
Kawai
H
,
Mori
A
.
Hepatic fat deposition is improved more with SGLT2 inhibitor luseogliflozin compared with sitagliptin: a randomised, crossover, controlled study using computed tomography
.
Diabetologia
2015
;
58
(
Suppl. 1
):
S392
133.
Qiang
S
,
Nakatsu
Y
,
Seno
Y
, et al
.
Treatment with the SGLT2 inhibitor luseogliflozin improves nonalcoholic steatohepatitis in a rodent model with diabetes mellitus
.
Diabetol Metab Syndr
2015
;
7
:
104
[PubMed]
134.
Cherney
DZI
,
Perkins
BA
,
Soleymanlou
N
, et al
.
Sodium glucose cotransport-2 inhibition and intrarenal RAS activity in people with type 1 diabetes
.
Kidney Int
2014
;
86
:
1057
1058
[PubMed]
135.
Dransfeld
O
,
Gehrmann
T
,
Köhrer
K
, et al
.
Oligonucleotide microarray analysis of differential transporter regulation in the regenerating rat liver
.
Liver Int
2005
;
25
:
1243
1258
[PubMed]
136.
U.S. Food and Drug Administration. FDA drug safety communication: FDA warns that SGLT2 inhibitors for diabetes may result in a serious condition of too much acid in blood [Internet], 2015. Silver Spring, MD, U.S. Food and Drug Administration. Available from http://www.fda.gov/Drugs/DrugSafety/ucm446845.htm. Accessed 30 January 2016
137.
Musso
G
,
Cassader
M
,
Gambino
R
.
Nonalcoholic steatohepatitis: emerging molecular targets and therapeutic strategies
.
Nat Rev Drug Discov
2016
;
15
:
249
274
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at http://www.diabetesjournals.org/content/license.