Back to Journals » Diabetes, Metabolic Syndrome and Obesity » Volume 17

The New Challenge of Obesity - Obesity-Associated Nephropathy

Authors Hao M, Lv Y , Liu S, Guo W

Received 12 October 2023

Accepted for publication 28 February 2024

Published 6 May 2024 Volume 2024:17 Pages 1957—1971

DOI https://doi.org/10.2147/DMSO.S433649

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Prof. Dr. Juei-Tang Cheng



Mengjin Hao,1,2 You Lv,1 Siyuan Liu,1 Weiying Guo1

1Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, 130021, People’s Republic of China; 2Department of Endocrinology, Jining No. 1 People’s Hospital, Jining, Shandong, 272000, People’s Republic of China

Correspondence: Weiying Guo, Department of Endocrinology and Metabolism, The First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China, Email [email protected]

Abstract: In recent years, obesity has become one of the major diseases that affect human health and consume human health resources, especially when it causes comorbidities such as hypertension, diabetes, cardiovascular disease and kidney disease. Many studies have demonstrated that obesity is associated with the development of chronic kidney disease and can exacerbate the progression of end-stage renal disease. This review described the mechanisms associated with the development of obesity-associated nephropathy and the current relevant therapeutic modalities, with the aim of finding new therapeutic targets for obesity-associated nephropathy. The mechanisms of obesity-induced renal injury include, in addition to the traditional alterations in renal hemodynamics, the involvement of various mechanisms such as macrophage infiltration in adipose tissue, alterations in adipokines (leptin and adiponectin), and ectopic deposition of lipids. At present, there is no “point-to-point” treatment for obesity-induced kidney injury. The renin-angiotensin-aldosterone system (RAAS) inhibitors, sodium-dependent glucose transporter 2 (SGLT-2) inhibitors and bariatric surgery described in this review can reduce urinary protein to varying degrees and delay the progression of kidney disease. In addition, recent studies on the therapeutic effects of intestinal flora on obesity may reduce the incidence of obesity-related kidney disease from the perspective of primary prevention. Both of these interventions have their own advantages and disadvantages, so the continuous search for the mechanism of obesity-induced related kidney disease will be extremely helpful for the future treatment of obesity-related kidney disease.

Keywords: obesity, kidney injury, inflammation, oxidative stress, adiponectin, leptin

Introduction

Obesity has become a world health problem that affects health. Especially in such a fast-paced and high-stress living environment, eating disorders or overeating have further contributed to the increase of people with obesity.1 The prevalence of obesity worldwide has shown a continuous increase over the past 50 years. According to a summary analysis of body mass indices of adolescents and children in various countries around the world from 1975 to 2016, the prevalence of obesity has increased in all countries, with some regional differences.2 The prevalence of obesity is higher in females than in males.3 Income level is positively correlated with the prevalence of obesity and overweight, and the prevalence of obesity in high-income countries increased significantly in the past, but the prevalence of obesity and overweight children in some high-income countries, such as the United States, Denmark, and Sweden, has gradually stabilized since 2000. Meanwhile, the prevalence of obesity in low- and middle-income countries has been on the rise. Even in India, where the prevalence of obesity among women of childbearing age is 5.1% and the prevalence of overweight is 15.5%, which is related to the level of urbanization and economic development.2,4 Current trends suggest that by 2025 the global prevalence of adult obesity will reach 18% among men and 21% among women.5 Obesity causes several complications such as hyperinsulinemia, disorders of lipid metabolism, nonalcoholic fatty liver, coronary artery disease, cardiovascular disease, various types of cancer and chronic kidney disease (CKD).6–9 In recent years, with the increase in the people with obesity, obesity has also become an important cause of CKD. An analysis of data from a UK biobank showed that for every 0.06 increase in waist-to-hip ratio estimated using genetic methods, the risk of CKD would increase by 30%; and for every 5 kg/m2 increase in body mass index (BMI), the risk of CKD would increase by 50%.10 In the UK, about one-third of CKD patients are associated with overweight or obesity.11 Another 14-year cohort study from Korean adults showed that high baseline levels of BMI and waist-to-hip ratio were independent risk factors for the development of CKD, and obesity was associated with the incidence of CKD.12 The fact that BMI is associated with obesity-related kidney injury is reflected in the assessment of the degree of obesity. However, BMI is indistinguishable when a person’s body composition changes, such as changes in the distribution of muscle and adipose tissue, visceral adipose tissue (VAT), and subcutaneous adipose tissue (SAT). Excessive VAT is associated with more insulin resistance and increased risk of kidney disease compared to SAT.13,14 Some studies have shown that abdominal or central obesity, reflected as waist circumference or waist-to-hip ratio, is a better indicator of cardiometabolic risk and mortality than BMI, and is more strongly associated with renal impairment.15,16 Obesity-induced renal injury (as shown in Figure 1) is mainly characterized by inflammation and oxidative stress, lipid accumulation in the glomerulus, and altered renal hemodynamics.6 In this paper, we describe the mechanisms associated with the development of obesity-associated nephropathy and the current relevant therapeutic modalities, with the aim of finding new therapeutic targets for obesity-associated nephropathy.

Figure 1 Obesity-induced kidney injury.

Histopathology of Obesity-Associated Nephropathy

The characteristic histopathologic manifestations of obesity-associated nephropathy are glomerulomegaly, focal segmental glomerulosclerosis (FSGS), and hypodensity of podocytes. In a large study of renal biopsies by Kambham et al17 it was found that, when matched for age and sex, the mean glomerular diameter was larger in biopsy samples from patients with obesity-associated nephropathy compared with normal controls (226 μm vs 169 μm). In an animal experimental model of obesity-associated nephropathy, glomerular cluster volume increases exponentially with body weight, which is associated with an increase in the number and surface area of glomerular capillaries induced by obese glomerular hyperfiltration.18 The increase in glomerular volume causes figures are alterations in the podocytes, which are reflected in increased width of the peduncle, mild and segmental loss of the peduncle, decreased density and number of podocytes, and even detachment of the podocytes from the basement mem- brane.19,20 These podocyte alterations are associated with adaptive hypertrophy brought about by glomerulomegaly. Podocytes are unable to proliferate, their hypertrophic capacity is limited, and as the glomerular volume expands, mechanical strain due to tensile tension and shear stress reaches a breaking point, allowing individual podocytes to fail and detach. Reduced podocyte density may reflect an adaptive hypertrophic response to glomerulomegaly and continued podocyte depletion that has not yet reached the threshold for the development of segmental sclerosis.21,22 This podocyte alteration ruptures glomerular filtration and plays an important role in the development of proteinuria and obesity-associated nephropathy.

Focal segmental glomerulosclerosis (FSGS) is another pathologic feature of obesity-associated nephropathy, but FSGS caused by obesity-associated nephropathy is milder and slower in progression than primary FSGS.23 Among the five histologic subtypes of FSGS (not otherwise specified, perihilar, cellular, tip variant, and collapsing variant), obesity-associated nephropathy exhibits a preponderance of the perihilar variant.24 In a Chinese cohort of patients with obesity-related nephropathy, the mean percentage of glomeruli with FSGS was only 6%.25 In a Spanish study, 95 extremely obese patients with normal renal function (mean BMI of 53.6 kg/m2), with a mean duration of obesity of 20 years, had pathologic findings that showed glomerulomegaly in 38% of cases and FSGS lesions in only 5%.26 This suggests that unlike primary FSGS, the renal changes caused by obesity-related nephropathy are adaptive FSGS.27 In adaptive FSGS, podocyte injury is more heterogeneous and less severe, and the pedicle is predominantly intact except for irreversible podocyte denudation. Intercellular proliferation of podocyte injury adjacent to initial foci of podocyte depletion may be mediated by interruption of pro-survival signaling by renin or by increased noxious stimuli (tensile tension and angiotensin). Proliferation of podocyte stress zones in obesity-associated nephropathy promotes the expansion of segmental lesions, ultimately leading to total glomerular fibrosis.28

Hemodynamic Changes in Obesity-Associated Nephropathy

Many studies have shown that obesity is one of the main causes of essential hypertension.29,30 Hypertension is one of the main risk factors for the development of cardiovascular disease and kidney diseases.31 Increased renal and perirenal adipose tissue in people with obesity causes compression of the kidneys, leading to increased sodium reabsorption by the kidneys.32 Early obesity is in a state of hypoxia and increased metabolic demand, manifested by increased renal plasma flow and increased renal glomerular filtration rate. The high filtration stimulates the glomerular-tubular balance, causing increased sodium-water reabsorption by the proximal tubules, which raises blood pressure and contributes to the progression of CKD.33,34 In addition, plasma levels of natriuretic peptide are associated with the development of hypertension in obese people. Natriuretic peptides are polypeptide hormones produced by the heart, including atrial natriuretic peptide and B- type natriuretic peptide. It binds to natriuretic peptide receptors to regulate water and salt metabolism balance in the body by relaxing vascular smooth muscle and promoting renal sodium excretion and drainage.35 In people with obesity, the concentration of natriuretic peptide receptor type C in adipose tissue increases and natriuretic peptide receptor C-mediated clearance increases, causing a decrease in plasma natriuretic peptide concentration. That weakens the inhibitory effect on the renin-angiotensin-aldosterone system (RAAS) and the diastolic effect on vascular smooth muscle and increasing sodium and water retention, causing the occurrence of hypertension.36,37 Activation of the RAAS in obesity is the main mechanism for the development of hypertension. It has been shown that activation of sympathetic nerve activity (SNA) plays a role in the development of obesity hypertension.38,39 Activation of the SNA in turn stimulates an increase in renin levels, further contributing to an increase in blood pressure.40 Activation of the SNA may be associated with increased leptin, insulin resistance, and activation of peripheral chemoreceptors due to hypoxia in people with obesity.41,42 It has been shown that when a high-salt diet was changed to a low-salt diet, blood pressure decreased significantly more in the group of people with obesity than in the non-obesity group, suggesting that obesity-related hypertension is associated with salt sensitivity.43 The development of salt-sensitive hypertension may be related to glomerular hyperfiltration, metabolic changes and neurohumoral mechanisms caused by long-term chronic obesity.32 In a cross-sectional study by verhave et al, the high sodium intake was found to be positively associated with urinary albumin excretion in people with obesity, suggesting that increased salt sensitivity due to obesity is associated with renal injury.44 Many studies have confirmed that plasma aldosterone levels are increased in people with obesity.45,46 Tuck et al found that reduced plasma renin and aldosterone levels were associated with weight loss through diet control for weight loss.47 Traditionally, aldosterone production is increased by activation of the RAAS system, which binds to the mineralocorticoid receptor (MR) to exert its sodium-saving and potassium-excreting effects. Later Goodfriend et al confirmed through a series of experimental studies that elevated levels of plasma free fatty acids (FFAs) stimulate an increase in aldosterone levels.48,49 Shibata et al found an increase in blood pressure and proteinuria in rats given a high-salt diet and aldosterone infusion, but no significant changes in blood pressure and proteinuria in rats given a low-salt diet and aldosterone infusion, suggesting that the aldosterone-induced increase in blood pressure and renal injury may be dependent on the increasing of salt sensitivity.50 Activation of the RAAS induces the production of fibrogenic factors (eg, TGF-b) that cause matrix in deposition in the kidney. It has been shown that blocking MR reduces tubulointerstitial fibrosis. Angiotensinogen II (Ang II) can cause functional and interstitial fibrosis in the proximal tubule through phenotypic transformation of renal epithelial-mesenchymal change (EMT) to renal injury.51,52 In addition, podocytes play an important role in glomerular exertion of filtration function. Obesity causes changes such as hyperfiltration in the kidney, increased glomerular capillary blood pressure and expansion of the basement membrane, causing deformation and detachment of the podocytes, further aggravating renal function injury.53 In addition to the alterations in podocytes caused by pathophysiological conditions, recent studies have shown that lipid accumulation and inflammatory vesicles in people with obesity are also important contributors to podocyte injury.54

Inflammation and Oxidative Stress of Obesity-Associated Nephropathy

The role of obesity in driving inflammation and oxidative stress in CKD was illustrated in a cross-sectional study conducted by Luis et al.55 Also, in a cross-sectional study conducted by Chen et al in 2324 subjects, it was confirmed that inflammation plays an important role in the increased risk of obesity-induced CKD.56 The inflammatory response people with obesity is low-grade, chronic and localized and is accompanied by macrophage infiltration, the degree of obesity is proportional to the degree of macrophage infiltration,57 and there is a greater increase in macrophages in visceral fat compared to subcutaneous adipose tissue.58,59 Macrophage infiltration plays an important role in inflammation caused by adipose tissue.60,61 Macrophage infiltration in the people with obesity is closely related to the injury and apoptosis of adipocytes caused by obesity-induced vascular thinning of adipose tissue and hypoxia.62,63 Macrophages have two phenotypes, M1 and M2, with M1 having pro-inflammatory and M2 anti-inflammatory effects.64,65 Obesity can contribute to the conversion of M2 to M1 phenotype,65,66 which causes an increase in pro-inflammatory factors (eg, TNF-α, IL-6, etc.) and leads to an increased production of intracellular reactive oxygen species (ROS). It in turn causes cellular injury and oxidative stress.63,67 Furthermore, the release of free fatty acids (FFAs) from adipose tissue in the people with obesity promotes the polarization of macrophages within adipose tissue toward the M1 phenotype.65 Recently, Chen et al showed through animal studies that Kdm6a (histone lysine demethylase 6a) regulates M1 and M2 imbalance within adipose tissue by regulating the expression of Ire1α(a transmembrane protein located in the endoplasmic reticulum that senses endoplasmic reticulum stress).68 The phenotypic conversion of macrophages is a complex process,69 but a better understanding of the process could be useful in the treatment of obesity-associated nephropathy in terms of inhibiting the conversion of macrophages to M1 type and achieving a reduction in inflammation. Wang et al70 by injecting an anti-inflammatory small molecule dye (IR-61) into mice by peritoneal administration, confirmed that IR-61 has the function of directly targeting the mitochondria of adipose tissue macrophages (ATMs) and inhibits macrophage activation by increasing mitochondrial oxidative phosphorylation, and also inhibits the expression of pro-inflammatory factors in visceral fat in mice on high-fat diet (HFD), which achieves inhibition of obesity-induced inflammation and obesity-associated diseases. In addition, miRNAs have been found to play an important role in the polarization of macrophages,71 and more studies are needed to confirm whether macrophage-induced related diseases are treated by targeting miRNAs.

In recent years, the pathogenic effects of nucleotide-binding oligomerized structural domain-like receptor protein 3 (NLRP3) inflammasome on the kidney have attracted extensive attention from researchers. The NLRP3 inflammasome is an intracellular multiprotein complex (NLRP3/ASC/caspase-1 complex) containing NLRP3, apoptosis-associated speckled protein (ASC), and caspase-1.72 NLRP3 inflammasome can be activated by many exogenous and endogenous substances, and the activation of NLRP3 inflammasome in obesity-associated nephropathy may be related to hyperfiltration caused by the RAAS system, mitochondrial dysfunction, and activation of endoplasmic reticulum.73 NLRP3 inflammasome acts on the progression of obesity-associated nephropathy through the caspase-1-IL-1β/IL-18 axis after being activated by signaling.74 Inflammatory cytokines such as IL-1β and IL-18 act in an autocrine or paracrine manner to induce podocyte damage and dysfunction. Additionally, it may be associated with non-inflammatory effects of NLRP3 inflammasome, such as pyroptosis cell death, cytoskeletal changes and altered cellular metabolism.75,76 A recent study showed that hyperfiltration remained unchanged in obese subjects whose IL-1β/Caspase-1 levels remained high after undergoing bariatric surgery, suggesting that inflammasome signaling plays a role in the development of obesity-associated nephropathy as early as the glomerular hyperfiltration stage.77 The exact pathogenic mechanisms of inflammasome at various stages of the development of obesity-associated nephropathy remain to be explored.

Leptin in Obesity-Associated Nephropathy

Leptin is an adipose tissue-derived hormone and the amount of leptin in the blood is proportional to the fat content.78 Leptin is an important hormone for maintaining homeostasis by acting mainly on the central nervous system to suppress food intake and promote energy expenditure leading to weight loss.79 It has been shown that leptin gene expression is increased in adipose tissue in people with obesity.80 In people with obesity, there is “leptin resistance”, where weight and intake are not reduced even though leptin levels are increased in people with obesity.81 The central nervous system is the main part of leptin metabolism and exerts its effects by acting on the leptin receptor (LepR) (Figure 2). The arcuate nucleus of the hypothalamus (ARC) is the main site of leptin regulation.82 Leptin interacts with the receptor to activate Janus kinase 2 (JAK2), which downstream can cause transcriptional activator 3 (STAT3), insulin receptor substrate (IRS)-phosphatidylinositol 3 kinase (PI3K), sh2-containing protein tyrosine phosphatase 2 (SHP2)-mitogen-activated protein kinase (MAPK), and 5 ‘adenosine monophosphate-activated protein kinase. Activation of the JAK2 signaling pathway causes increased transcription of suppressor of cytokine signaling 3 (SOCS3), which in turn inhibits the JAK2 signaling pathway and reduces leptin action.78,79,83 In addition, protein tyrosine phosphatase 1B (PTP1B) is also involved in the inhibition of leptin signaling.79 In addition to the regulation of diet and body weight, leptin can also increase sympathetic nerve activity (SNA) in certain tissues, such as cardiovascular sympathetic nerve activity and renal sympathetic nerve activity (RSNA), through receptor action in the central nervous system.84,85 Harlan et al82 found in diet-induced mice that deletion of the highest concentration of leptin receptor in the ARC abolished the effect of leptin on increased renal sympathetic excitability. The increase in plasma leptin concentration can cause activation of the RAAS by increasing RSNA and affect renal hemodynamic changes,32,78,82 which may be related to the involvement of the IRS2-PI3K pathway.78,86 In addition, it has been shown that leptin is associated with glomerular fibrosis,87 which is related to the fact that leptin increases TGF-β expression in glomerular cells and activates the JAK/STAT signaling pathway.88 Also, leptin can affect renal endothelial cells and cause renal fibrosis by increasing reactive oxygen species (ROS).89 Recently, Liu et al significantly reduced RSNA and elevated blood pressure in HFD mice by siRNA knockdown of Sirt1(an energy-sensing enzyme located downstream of the leptin signaling pathway), and also reduced leptin-induced levels of tumor necrosis factor α (TNF-α), cytokine interleukin 6 (IL-6), and interleukin 1β (IL-1β), providing a treatment of leptin-induced RSNA and hypertension in the people with obesity.90,91 In addition, Heiss et al found that the gut microflora-GLP-1 axis could modulate diet-induced hypothalamic inflammation and improve leptin sensitivity.92

Figure 2 The basic pathway of leptin. Leptin binds to LepR to activate associated JAK2 phosphorylation, causing activation of the SHP2/MAPK pathway and the transcriptional activator STAT3. The SHP2/MAPK signaling pathway has an important role in the cardiometabolic effects of leptin. Activation of STAT3 regulates leptin-induced reduction in food intake and elevated blood pressure, and also induces transcription of SOCS3, which has an attenuating effect on LepR-mediated signaling. Activation of the IRS2-PI3K pathway contributes to the effects of leptin on SNA and blood pressure. PTP1B attenuates leptin signaling by dephosphorylating JAK2.

Abbreviations: LepR, leptin receptor; JAK2, Janus kinase 2; STAT3, transcriptional activator 3; IRS, insulin receptor substrate; PI3K, phosphatidylinositol 3 kinase; SHP2, sh2-containing protein tyrosine phosphatase 2; MAPK, mitog en-activated protein kinase; SOCS3, suppressor of cytokine signaling 3; mTOR, mammalian target of rapamycin; Akt, v-Akt murine thymoma viral oncogene homolog; PTP1B, protein tyrosine phosphatase 1B.

Characteristics of Adiponectin in the People with Obesity

Adiponectin is an amino acid protein secreted by adipose tissue. It has three structural forms: low molecular weight (LMW), which is trimeric, medium molecular weight (MMW), which is hexamers, and high molecular weight (HMW), which is multimeric. The globular adiponectin differs from the three oligomers in the above in that it is a C1q-like globular structural domain at the COOH terminus of adiponectin, present in the circulation, biologically active, and produced by hydrolysis of full-length adiponectin.93,94 Adiponectin has both anti-inflammatory and pro-inflammatory effects, which is one of the mechanisms that cause the development of various diseases.64,95 The reduced secretion of adiponectin in people with obesity may be related to the suppression of adiponectin transcription due to the persistent chronic inflammatory state of the body caused by obesity.64,96 Adiponectin exerts its function by interacting with adiponectin receptors (Figure 3).Currently known are adiponectin receptor 1 (AdipoR1), adiponectin receptor 2 (AdipoR2) and T-cadherin (T-cadherin), AdipoR1 and AdipoR2 both have seven transmembrane structural domains distinct from G protein-coupled receptors, which are widely expressed in skeletal muscle and liver and have ceramidase activity, playing an important role in the regulation of metabolic processes.64,97 The binding of adiponectin to AdipoR1 activates the APPL1 (a protein that interacts with the adiponectin receptor)-AMPK-acetyl coenzyme a carboxylase (ACC)/nitric oxide synthase (eNOS) pathway, enhancing cellular β-oxidation, reducing lipid accumulation, and improving endothelial cell function. Adiponectin binding to AdipoR2 activates APPL1-peroxisome proliferator-activated receptor alpha (PPAR-α), increasing fatty acid oxidation and glucose uptake.95,98 In addition, adiponectin can act on macrophages to promote conversion to the M2 phenotype and exert the anti-inflammatory effects.94,99 Adiponectin and its receptors (mostly AdipoR1) are mainly present in glomerular and proximal tubular cells, exerting their protective function on the kidney.98,100 Studies have shown that obesity is an independent risk factor for the development of CKD.101,102 Many studies have shown that adiponectin levels are elevated in patients with CKD, especially when the glomerular filtration rate (eGFR) is less than15 ml/min/1.73 m2, which are 2–3 times higher than normal in patients with end-stage renal disease (ESRD).64,103 Interestingly, Menon et al concluded through an MDRD study, based on a 10-year follow-up of CKD patients, that stage 3 and 4 mortality in CKD patients is strongly associated with increased adiponectin, which is very different from the anti-inflammatory protective function of adiponectin.104 Rhee et al through a prospective study of 500 patients with chronic kidney disease on regular hemodialysis found that a 10 μg/ml increase in adiponectin concentration was associated with an approximately 25% increase in the risk of death.105 Recently, Chen et al demonstrated through animal studies that adiponectin can upregulate AdipoR1 in renal tissues and further activate peroxisome proliferator-activated receptor γ coactivator α (PGC-1α) to improve mitochondrial function within renal tubular epithelial cells, which provides an idea for the treatment of diabetic nephropathy.106 Francisqueti et al showed that the γ Oz-treated group had lower proteinuria levels and increased glomerular filtration rate compared to the control group by using γ-glutamatergic (γ Oz) in obese mice with nephropathy for 10 weeks, it provides a new target for obesity-induced kidney injury.102

Figure 3 The mechanism of action of adiponectin: Adiponectin and insulin interact with their respective receptors to cause cascade signaling. Adiponectin binds to APL1 and APL2 to activate fatty acid oxidation and inhibit energy expenditure, exerting anti-inflammatory properties. Insulin regulates metabolism by causing glucose uptake and utilization, glycogen and protein synthesis through the PI3K/AKT pathway.

Abbreviations: AdipoR1, adiponectin receptor 1; AdipoR2, adiponectin receptor 2; APPL-1, a protein that interacts with the adiponectin receptor; ACC, acetyl coenzyme a carboxylase; eNOS, nitric oxide synthase; PPAR-α, peroxisome proliferator-activated receptor alpha.

The Impact of Gut Microbes on People with Obesity

There are many normal flora in the human intestine that have physiological roles in regulating host energy metabolism and immune function.107 Among them, four bacteria phyla play a major role in the adult gastrointestinal tract, including Firmicutes, Bacteroidetes, Actinobacteria, and Proteobacteria.108 Studies in animal models have shown that obesity and high-fat diets can cause changes in intestinal flora, including a decrease in Bacteroides and an increase in Firmicutes.109 In human studies, RNA sequencing of fecal samples from obese individuals has also shown a reduced proportion of Bacteroidetes compared to lean individuals.110 Altered intestinal microbes in the obese population disrupt the integrity of the intestinal barrier, causing pathological bacterial transfer into the blood and the initiation of inflammatory responses.111 Previously, animal studies have shown that a high-fat diet can cause an increase in plasma lipopolysaccharide (LPS) concentrations, which promotes the secretion of inflammatory cytokines, and induce inflammation and oxidative stress in the body.112 In addition, changes in intestinal flora cause an increase in the storage of triglycerides and the proportion of short-chain fatty acids in the host, regulating lipid metabolism.111 The altered intestinal flora in the obese population described above causes systemic inflammation and oxidative stress that can have an impact on the kidneys. The development of obesity-associated nephropathy can also cause dysbiosis of the intestinal flora. Therefore, by “reversing” the alteration of intestinal flora (including the intake of probiotics and prebiotics), organ damage and dysfunction caused by intestinal dysbiosis can be reduced.

The Role of Other Obesity-Induced Effects on the Development of Chronic Kidney Disease

In addition to the basic pathophysiological factors mentioned above, in people with obesity, excessive fat content can cause metabolic disorders and the phenomenon of “ectopic deposition”. In the kidney, the accumulation of free fatty acids (FFAs) and triglycerides can lead to inflammation, activation of oxidative stress and alteration of signaling pathways, which can further lead to apoptosis of kidney cells and fibrosis of the kidney.113 In addition to increasing macrophage infiltration through the IKKβ and JNK pathways, FFAs can act directly on TLR4 to exert an inflammatory cascade response. Other immune cells such as neutrophils, lymphocytes, and T cells also play a role in the production of inflammation.114,115 Recently, Watanabe et al showed that adipose tissue neutrophils promote macrophage infiltration and inflammation by activating NF-kB to enable IL-1β expression, revealing a role for neutrophils in adipose tissue inflammation.116 In addition, disturbed cholesterol metabolism in people with obesity results in cholesterol deposition and impaired renal function.117 In patients with advanced CKD, HDL levels are reduced and are accompanied by a significant decrease in antioxidant enzyme activity, resulting in impaired anti-inflammatory and antioxidant properties. Moreover, the anti-inflammatory effect of HDL was changed to pro-inflammatory by the modification of oxidative enzymes and apolipoproteins under the effect of systemic inflammation and oxidative stress.118,119

Advances in the Treatment of Obesity-Associated Nephropathy

Weight Loss

For kidney injury caused by obesity, weight loss is the most basic treatment.120 Common methods about weight loss include diet and exercise management, weight loss drugs, weight loss surgery. Many studies have shown that weight loss with diet management resulted in a significant reduction in proteinuria.121,122 In addition, weight loss can lower blood pressure, reduce renal hyperfiltration, and have a protective function for the kidneys.123 Studies have shown that a low glycemic index diet and a low-calorie diet not only reduce proteinuria but also have a significant effect on hypertension, insulin resistance, and hyperlipidemia in people with obesity (Table 1).124,125 In addition, behavioral interventions including goal setting, self-monitoring and stimulus control are also beneficial for weight loss.125 Bariatric surgery has been shown to have a positive effect on obesity-induced kidney disease.119,126,127 It has been shown that bariatric surgery is accompanied not only by a significant reduction in proteinuria, but also by significant changes in the reduction of hypertension and inflammatory factors, especially 1–5 years after surgery.128 Recently, endoscopic bariatric therapies have been rapidly developed to reduce gastric volume and delay gastric emptying, mainly through intragastric balloon placement,129 sleeve gastroplasty, and aspiration therapy.130

Table 1 Studies on the Effects of Different Treatments on Obesity-Induced Kidney Injury

Renin-Angiotensin-Aldosterone System (RAAS) Inhibitors

In the treatment of obesity-induced kidney injury, the control of proteinuria is an important way to slow the progression of kidney disease.120 RAAS inhibitors are the basic drugs for early protection of kidney function, and typical drugs are angiotensin-converting enzyme inhibitor (ACEI) and angiotensin-receptor blockers (ARB), which can reduce proteinuria, improve renal blood flow and reduce blood pressure to protect kidney function. In a meta-analysis of RAAS inhibitors on chronic kidney disease outcomes, RAAS inhibitors did not appear to have a more beneficial effect on renal prognosis with ACEI or ARB compared with other blood pressure-lowering drugs. However, compared with placebo, RAAS inhibitors were able to reduce blood pressure and urinary protein excretion with significant renoprotective effects, demonstrating the important role of blood pressure control in improving end-stage renal disease and urinary protein.140 In a clinical trial study by Morales et al showed that RAAS system inhibitors combined with spironolactone (mineralocorticoid receptor antagonist, MRA) consistently reduced proteinuria, lowered blood pressure, and delayed the progression of nephropathy.131 Finerenone (selective MRA) has been shown to reduce proteinuria in patients with CKD in the short term.132 Bakris et al showed a significantly lower incidence of the primary composite outcome (including renal failure, at least 40% reduction in eGFR at baseline, and death from nephropathy causes) with finerenone compared to placebo in a randomized double-blind trial than in the placebo group.133

Sodium-Dependent Glucose Transporter 2 (SGLT-2) Inhibitor

SGLT2 inhibitors act on proximal renal tubular SGLT2 to promote urinary glucose excretion by inhibiting glucose and sodium reabsorption, while also lowering blood pressure, improving glomerular hyperfiltration, and slowing the progression of nephropathy. As a novel hypoglycemic drug, SGLT2 inhibitors not only lower blood glucose, but also reduce the risk of proteinuria and death in obesity and diabetes and slow the progression of nephropathy.23,141 In a recent clinical trial of DAPA-CKD, results suggested that dapagliflozin significantly reduced the risk of cardiovascular hospitalization and the risk of adverse renal outcomes, regardless of concomitant cardiovascular disease.135 Interestingly, another 6-week randomized double-blind clinical trial of dapagliflozin in non-diabetic patients with combined CKD showed no significant difference in the short-term effect of dapagliflozin compared with placebo on proteinuria, but there was a significant reduction in eGFR and body weight, and the impact about renal outcomes needs to be explored in longer-term trials.134 Recently, in another DAPA-CKD trial with a median follow-up of 2.4 years, results showed that dapagliflozin reduced the incidence of adverse events and all-cause mortality in chronic kidney disease and cardiovascular disease in diabetic and non-diabetic patients.142 In addition, many studies have demonstrated that SGLT2 inhibitors improve renal function by reducing pro-inflammatory factors and oxidative stress and regulating lipid metabolism.143

Other Treatment Modalities

In addition to the above common drugs, there are some other drugs or modalities that have been proved to have impact on renal function. Early studies have confirmed that gut microbes play a role in the development of obesity,144,145 so regulating the gut microflora has also become one of the potential methods for treating obesity. Probiotics have been considered to have a positive effect on the gastrointestinal tract, and many studies have demonstrated that probiotic treatment reduces obesity-related inflammation and fat storage.136 Recently, it has been found that food can be used as a probiotic carrier for the treatment of obesity, with common foods such as fermented dairy products and fruit juices.146 Melatonin has been shown to have anti-inflammatory, antioxidant and endothelial protective functions in addition to regulating biological rhythms.113,147 Ishigaki et al improved renal RAAS activation and renal injury by 5/6 nephrectomized (Nx) rats as a chronic progressive CKD model, and after 4 weeks of melatonin treatment, the melatonin-treated group reversed the decrease in intrarenal antioxidant activity, the increase in oxidative stress and the increase in markers of renal interstitial fibrosis compared to the control group.137 In obesity-induced renal injury, melatonin exerts renoprotective effects by reducing pro-inflammatory factors and adipokines.148 In addition, some studies have confirmed that melatonin can also reduce obesity-associated nephropathy by inhibiting inflammatory vesicles.138 Meanwhile, the novel hypoglycemic drug GLP-1 receptor agonist may protect renal function by reducing RAAS system activation, acting as an anti-inflammatory and anti-fibrotic agent, and having a sustained weight loss effect, but the long-term prognosis for the kidney still lacks relevant clinical trials and needs to be explored.139

Conclusion

With the increasing prevalence of obesity and other health problems associated with obesity, more and more people are expressing their concern about obesity and its related diseases. The pathophysiological mechanisms of obesity-induced kidney injury are mainly inflammation and oxidative stress, alteration of adipokines, lipid deposition. Those mechanisms cause the alteration of signaling pathways, the glomerular and tubular vascular endothelial and cellular injury and the activation of RAAS system, which further cause kidney injury and fibrosis. Among them, macrophage infiltration and phenotypic transformation play an important role in the inflammatory response caused by obesity, leptin mainly acts on the activation of renal RAAS system through the increase of sympathetic nerve activity (SNA) caused by acting on hypothalamic leptin receptors. Adiponectin, as an anti-inflammatory factor, plays an anti-inflammatory, antioxidant and endothelial function-protecting role, in people with obesity. The decrease of adiponectin is more likely to cause kidney injury. In addition, lipid deposition and disorders of cholesterol metabolism can directly or indirectly cause injury to the kidney and cause the progression of kidney disease.

Practical Application

Clinically, the risk of kidney-related diseases is reduced by means of weight loss, and on the other hand, drugs such as RAAS system inhibitors and SGLT-2 inhibitors are used to lower blood pressure and reduce proteinuria to improve renal function. Both of these interventions have their own advantages and disadvantages, so the continuous search for the mechanism of obesity-induced related kidney disease will be extremely helpful for the future treatment of obesity-related kidney disease.

Disclosure

The authors report no conflicts of interest in this work.

References

1. Razzoli M, Pearson C, Crow S, Bartolomucci A. Stress, overeating, and obesity: insights from human studies and preclinical models. Neurosci Biobehav Rev. 2017;76(Pt A):154–162. doi:10.1016/j.neubiorev.2017.01.026

2. NCD. Risk Factor Collaboration (NCD-RisC).Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet. 2017;390(10113):2627–2642. doi:10.1016/S0140-6736(17)32129-3

3. Ataey A, Jafarvand E, Adham D, Moradi-Asl E. The relationship between obesity, overweight, and the human development index in world health organization eastern Mediterranean region countries. J Prev Med Public Health. 2020;53(2):98–105. doi:10.3961/jpmph.19.100

4. Al Kibria GM, Swasey K, Hasan MZ, Sharmeen A, Day B. Prevalence and factors associated with underweight, overweight and obesity among women of reproductive age in India. Glob Health Res Policy. 2019;4:24. doi:10.1186/s41256-019-0117-z

5. NCD Risk Factor Collaboration (NCD-RisC). Trends in adult body-mass index in 200 countries from 1975 to 2014: a pooled analysis of 1698 population-based measurement studies with 19·2 million participants. Lancet. 2016;387(10026):1377–1396. doi:10.1016/S0140-6736(16)30054-X

6. Chang Y, Ryu S, Choi Y, et al. Metabolically healthy obesity and development of chronic kidney disease: a cohort study. Ann Intern Med. 2016;164(5):305–312. doi:10.7326/M15-1323

7. Yazıcı D, Sezer H. Insulin resistance, obesity and lipotoxicity. Adv Exp Med Biol. 2017;960:277–304.

8. Alpert MA, Omran J, Bostick BP. Effects of obesity on cardiovascular hemodynamics, cardiac morphology, and ventricular function. Curr Obes Rep. 2016;5(4):424–434. doi:10.1007/s13679-016-0235-6

9. Kolb R, Sutterwala FS, Zhang W. Obesity and cancer: inflammation bridges the two. Curr Opin Pharmacol. 2016;29:77–89. doi:10.1016/j.coph.2016.07.005

10. Zhu P, Herrington WG, Haynes R, et al. Conventional and genetic evidence on the association between adiposity and CKD. J Am Soc Nephrol. 2021;32(1):127–137. doi:10.1681/ASN.2020050679

11. Herrington WG, Smith M, Bankhead C, et al. Body-mass index and risk of advanced chronic kidney disease: prospective analyses from a primary care cohort of 1.4 million adults in England. PLoS One. 2017;12(3):e0173515. doi:10.1371/journal.pone.0173515

12. Zhang HS, An S, Ahn C, Park SK, Park B. Obesity measures at baseline, their trajectories over time, and the incidence of chronic kidney disease: a 14 year cohort study among Korean adults. Nutr, Metab Cardiovasc Dis. 2021;31(3):782–792. doi:10.1016/j.numecd.2020.10.021

13. Bray GA, Heisel WE, Afshin A, et al. The science of obesity management: an endocrine society scientific statement. Endocr Rev. 2018;39(2):79–132. doi:10.1210/er.2017-00253

14. Piché ME, Tchernof A, Després JP. Obesity phenotypes, diabetes, and cardiovascular diseases. Circ Res. 2020;126(11):1477–1500. doi:10.1161/CIRCRESAHA.120.316101

15. Elsayed EF, Sarnak MJ, Tighiouart H, et al. Waist-to-Hip ratio, body mass index, and subsequent kidney disease and death. Am J Kidney Dis. 2008;52(1):29–38. doi:10.1053/j.ajkd.2008.02.363

16. Shen FC, Chiu YW, Kuo MC, et al. U-shaped association between waist-to-hip ratio and All-cause mortality in stage 3-5 chronic kidney disease patients with body mass index paradox. J Pers Med. 2021;11(12):1355. doi:10.3390/jpm11121355

17. Kambham N, Markowitz GS, Valeri AM, Lin J, D’Agati VD. Obesity-related glomerulopathy: an emerging epidemic. Kidney Int. 2001;59(4):1498–1509. doi:10.1046/j.1523-1755.2001.0590041498.x

18. Fukuda A, Chowdhury MA, Venkatareddy MP, et al. Growth-dependent podocyte failure causes glomerulosclerosis. J Am Soc Nephrol. 2012;23(8):1351–1363. doi:10.1681/ASN.2012030271

19. Chen HM, Liu ZH, Zeng CH, Li SJ, Wang QW, Li LS. Podocyte lesions in patients with obesity-related glomerulopathy. Am J Kidney Dis. 2006;48(5):772–779. doi:10.1053/j.ajkd.2006.07.025

20. Barutta F, Bellini S, Gruden G. Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci. 2022;136(7):493–520.

21. Tsuboi N, Okabayashi Y, Shimizu A, Yokoo T. The renal pathology of obesity. Kidney Int Rep. 2017;2(2):251–260. doi:10.1016/j.ekir.2017.01.007

22. Tsuboi N, Okabayashi Y. The renal pathology of obesity: structure-function correlations. Semin Nephrol. 2021;41(4):296–306. doi:10.1016/j.semnephrol.2021.06.002

23. Martínez-Montoro JI, Morales E, Cornejo-Pareja I, Tinahones FJ, Fernández-García JC. Obesity-related glomerulopathy: current approaches and future perspectives. Obes Rev. 2022;23(7):e13450. doi:10.1111/obr.13450

24. D’Agati VD, Fogo AB, Bruijn JA, Jennette JC. Pathologic classification of focal segmental glomerulosclerosis: a working proposal. Am J Kidney Dis. 2004;43(2):368–382. doi:10.1053/j.ajkd.2003.10.024

25. Chen HM, Li SJ, Chen HP, Wang QW, Li LS, Liu ZH. Obesity-related glomerulopathy in China: a case series of 90 patients. Am J Kidney Dis. 2008;52(1):58–65. doi:10.1053/j.ajkd.2008.02.303

26. Serra A, Romero R, Lopez D, et al. Renal injury in the extremely obese patients with normal renal function. Kidney Int. 2008;73(8):947–955. doi:10.1038/sj.ki.5002796

27. Rosenberg AZ, Kopp JB. Focal Segmental Glomerulosclerosis. Clin J Am Soc Nephrol. 2017;12(3):502–517. doi:10.2215/CJN.05960616

28. De Vriese AS, Wetzels JF, Glassock RJ, Sethi S, Fervenza FC. Therapeutic trials in adult FSGS: lessons learned and the road forward. Nat Rev Nephrol. 2021;17(9):619–630. doi:10.1038/s41581-021-00427-1

29. Ferrannini E, Buzzigoli G, Bonadonna R, et al. Insulin resistance in essential hypertension. N Engl J Med. 1987;317(6):350–357. doi:10.1056/NEJM198708063170605

30. Seravalle G, Grassi G. Obesity and hypertension. Pharmacol Res. 2017;122:1–7. doi:10.1016/j.phrs.2017.05.013

31. Landsberg L, Aronne LJ, Beilin LJ, et al. Obesity-related hypertension: pathogenesis, cardiovascular risk, and treatment--a position paper of the the obesity society and the American society of hypertension. Obesity. 2013;21(1):8–24. doi:10.1002/oby.20181

32. Hall JE, Do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity, kidney dysfunction and hypertension: mechanistic links. Nat Rev Nephrol. 2019;15(6):367–385. doi:10.1038/s41581-019-0145-4

33. Chagnac A, Zingerman B, Rozen-Zvi B, Herman-Edelstein M. Consequences of glomerular hyperfiltration: the role of physical forces in the pathogenesis of chronic kidney disease in diabetes and obesity. Nephron. 2019;143(1):38–42. doi:10.1159/000499486

34. Giannini G, Kopp JB, Rosenberg AZ. Podocytopathy in Obesity: challenges of Living Large. Semin Nephrol. 2021;41(4):307–317. doi:10.1016/j.semnephrol.2021.06.003

35. Goetze JP, Bruneau BG, Ramos HR, Ogawa T, de Bold MK, de Bold AJ. Cardiac natriuretic peptides. Nat Rev Cardiol. 2020;17(11):698–717. doi:10.1038/s41569-020-0381-0

36. Kovacova Z, Tharp WG, Liu D, et al. Adipose tissue natriuretic peptide receptor expression is related to insulin sensitivity in obesity and diabetes. Obesity. 2016;24(4):820–828. doi:10.1002/oby.21418

37. Januzzi JL, Mohebi R. Obesity-mediated disruption of natriuretic peptide-blood pressure rhythms. J Am Coll Cardiol. 2021;77(18):2304–2306. doi:10.1016/j.jacc.2021.03.317

38. Hall JE, Mouton AJ, da Silva AA, et al. Obesity, kidney dysfunction, and inflammation: interactions in hypertension. Cardiovasc Res. 2021;117(8):1859–1876. doi:10.1093/cvr/cvaa336

39. Grassi G, Biffi A, Seravalle G, et al. Sympathetic Neural Overdrive in the Obese and Overweight State. Hypertension. 2019;74(2):349–358. doi:10.1161/HYPERTENSIONAHA.119.12885

40. Susic D, Varagic J. Obesity: a perspective from hypertension. Med Clin North Am. 2017;101(1):139–157. doi:10.1016/j.mcna.2016.08.008

41. Armitage JA, Burke SL, Prior LJ, et al. Rapid onset of renal sympathetic nerve activation in rabbits fed a high-fat diet. Hypertension. 2012;60(1):163–171. doi:10.1161/HYPERTENSIONAHA.111.190413

42. Dewan NA, Nieto FJ, Somers VK. Intermittent hypoxemia and OSA: implications for comorbidities. Chest. 2015;147(1):266–274. doi:10.1378/chest.14-0500

43. Rocchini AP, Key J, Bondie D, et al. The effect of weight loss on the sensitivity of blood pressure to sodium in obese adolescents. N Engl J Med. 1989;321(9):580–585. doi:10.1056/NEJM198908313210905

44. Verhave JC, Hillege HL, Burgerhof JG, et al. Sodium intake affects urinary albumin excretion especially in overweight subjects. J Intern Med. 2004;256(4):324–330. doi:10.1111/j.1365-2796.2004.01390.x

45. Vogt B, Bochud M, Burnier M. The association of aldosterone with obesity-related hypertension and the metabolic syndrome. Semin Nephrol. 2007;27(5):529–537. doi:10.1016/j.semnephrol.2007.07.009

46. Flynn C. Increased aldosterone: mechanism of hypertension in obesity. Semin Nephrol. 2014;34(3):340–348. doi:10.1016/j.semnephrol.2014.04.009

47. Tuck ML, Sowers J, Dornfeld L, Kledzik G, Maxwell M. The effect of weight reduction on blood pressure, plasma renin activity, and plasma aldosterone levels in obese patients. N Engl J Med. 1981;304(16):930–933. doi:10.1056/NEJM198104163041602

48. Goodfriend TL, Egan BM, Kelley DE. Aldosterone in obesity. Endocr Res. 1998;24(3–4):789–796. doi:10.3109/07435809809032689

49. Goodfriend TL, Ball DL, Gardner HW. An oxidized derivative of linoleic acid affects aldosterone secretion by adrenal cells in vitro. Prostaglandins Leukot Essent Fatty Acids. 2002;67(2–3):163–167. doi:10.1054/plef.2002.0414

50. Shibata S, Mu S, Kawarazaki H, et al. Rac1 GTPase in rodent kidneys is essential for salt-sensitive hypertension via a mineralocorticoid receptor-dependent pathway. J Clin Invest. 2011;121(8):3233–3243. doi:10.1172/JCI43124

51. Brown NJ. Contribution of aldosterone to cardiovascular and renal inflammation and fibrosis. Nat Rev Nephrol. 2013;9(8):459–469. doi:10.1038/nrneph.2013.110

52. Whaley-Connell A, Sowers JR. Obesity and kidney disease: from population to basic science and the search for new therapeutic targets. Kidney Int. 2017;92(2):313–323. doi:10.1016/j.kint.2016.12.034

53. Kriz W, Lemley KV. A potential role for mechanical forces in the detachment of podocytes and the progression of CKD. J Am Soc Nephrol. 2015;26(2):258–269. doi:10.1681/ASN.2014030278

54. Zhao J, Rui HL, Yang M, Sun LJ, Dong HR, Cheng H. CD36-mediated lipid accumulation and activation of NLRP3 inflammasome lead to podocyte injury in obesity-related glomerulopathy. Mediators Inflamm. 2019;2019:3172647. doi:10.1155/2019/3172647

55. Ramos LF, Shintani A, Ikizler TA, Himmelfarb J. Oxidative stress and inflammation are associated with adiposity in moderate to severe CKD. J Am Soc Nephrol. 2008;19(3):593–599. doi:10.1681/ASN.2007030355

56. Chen S, Zhou S, Wu B, et al. Association between metabolically unhealthy overweight/obesity and chronic kidney disease: the role of inflammation. Diabetes Metab. 2014;40(6):423–430. doi:10.1016/j.diabet.2014.08.005

57. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest. 2003;112(12):1796–1808. doi:10.1172/JCI200319246

58. Harman-Boehm I, Blüher M, Redel H, et al. Macrophage infiltration into omental versus subcutaneous fat across different populations: effect of regional adiposity and the comorbidities of obesity. J Clin Endocrinol Metab. 2007;92(6):2240–2247. doi:10.1210/jc.2006-1811

59. Hardy OT, Perugini RA, Nicoloro SM, et al. Body mass index-independent inflammation in omental adipose tissue associated with insulin resistance in morbid obesity. Surg Obes Relat Dis. 2011;7(1):60–67. doi:10.1016/j.soard.2010.05.013

60. Schipper HS, Prakken B, Kalkhoven E, Boes M. Adipose tissue-resident immune cells: key players in immunometabolism. Trends Endocrinol Metab. 2012;23(8):407–415. doi:10.1016/j.tem.2012.05.011

61. González-Périz A, Clària J. Resolution of adipose tissue inflammation. ScientificWorldJournal. 2010;10:832–856. doi:10.1100/tsw.2010.77

62. Pasarica M, Sereda OR, Redman LM, et al. Reduced adipose tissue oxygenation in human obesity: evidence for rarefaction, macrophage chemotaxis, and inflammation without an angiogenic response. Diabetes. 2009;58(3):718–725. doi:10.2337/db08-1098

63. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nat Rev Immunol. 2011;11(2):85–97. doi:10.1038/nri2921

64. Choi HM, Doss HM, Kim KS. Multifaceted Physiological Roles of Adiponectin in Inflammation and Diseases. Int J Mol Sci. 2020;21(4):1219. doi:10.3390/ijms21041219

65. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007;117(1):175–184. doi:10.1172/JCI29881

66. Prieur X, Mok CY, Velagapudi VR, et al. Differential lipid partitioning between adipocytes and tissue macrophages modulates macrophage lipotoxicity and M2/M1 polarization in obese mice. Diabetes. 2011;60(3):797–809. doi:10.2337/db10-0705

67. Gordon S, Martinez FO. Alternative activation of macrophages: mechanism and functions. Immunity. 2010;32(5):593–604. doi:10.1016/j.immuni.2010.05.007

68. Chen J, Xu X, Li Y, et al. Kdm6a suppresses the alternative activation of macrophages and impairs energy expenditure in obesity. Cell Death Differ. 2021;28(5):1688–1704. doi:10.1038/s41418-020-00694-8

69. Orecchioni M, Ghosheh Y, Pramod AB, Ley K. Macrophage Polarization: different Gene Signatures in M1(LPS+) vs. Classically and M2(LPS-) vs. Alternatively Activated Macrophages. Front Immunol. 2019;10:1084. doi:10.3389/fimmu.2019.01084

70. Wang Y, Tang B, Long L, et al. Improvement of obesity-associated disorders by a small-molecule drug targeting mitochondria of adipose tissue macrophages. Nat Commun. 2021;12(1):102. doi:10.1038/s41467-020-20315-9

71. Shapouri-Moghaddam A, Mohammadian S, Vazini H, et al. Macrophage plasticity, polarization, and function in health and disease. J Cell Physiol. 2018;233(9):6425–6440. doi:10.1002/jcp.26429

72. Ma Q, Schuetz J. Pharmacological Inhibition of the NLRP3 Inflammasome: structure, Molecular Activation, and Inhibitor-NLRP3 Interaction. Pharmacol Rev. 2023;75(3):487–520. doi:10.1124/pharmrev.122.000629

73. Ke B, Shen W, Fang X, Wu Q. The NLPR3 inflammasome and obesity-related kidney disease. J Cell Mol Med. 2018;22(1):16–24. doi:10.1111/jcmm.13333

74. Li LC, Yang JL, Lee WC, et al. Palmitate aggravates proteinuria-induced cell death and inflammation via CD36-inflammasome axis in the proximal tubular cells of obese mice. Am J Physiol Renal Physiol. 2018;315(6):F1720–F1731. doi:10.1152/ajprenal.00536.2017

75. Lamkanfi M. Emerging inflammasome effector mechanisms. Nat Rev Immunol. 2011;11(3):213–220. doi:10.1038/nri2936

76. Takano Y, Yamauchi K, Hayakawa K, et al. Transcriptional suppression of nephrin in podocytes by macrophages: roles of inflammatory cytokines and involvement of the PI3K/Akt pathway. FEBS Lett. 2007;581(3):421–426. doi:10.1016/j.febslet.2006.12.051

77. Moriconi D, Antonioli L, Masi S, et al. Glomerular hyperfiltration in morbid obesity: role of the inflammasome signalling. Nephrology (Carlton). 2022;27(8):673–680. doi:10.1111/nep.14077

78. Bell BB, Rahmouni K. Leptin as a mediator of obesity-induced hypertension. Curr Obes Rep. 2016;5(4):397–404. doi:10.1007/s13679-016-0231-x

79. Friedman JM. Leptin and the endocrine control of energy balance. Nat Metab. 2019;1(8):754–764. doi:10.1038/s42255-019-0095-y

80. Lönnqvist F, Arner P, Nordfors L, Schalling M. Overexpression of the obese (ob) gene in adipose tissue of human obese subjects. Nat Med. 1995;1(9):950–953. doi:10.1038/nm0995-950

81. Myers MG, Leibel RL, Seeley RJ, Schwartz MW. Obesity and leptin resistance: distinguishing cause from effect. Trends Endocrinol Metab. 2010;21(11):643–651. doi:10.1016/j.tem.2010.08.002

82. Harlan SM, Morgan DA, Agassandian K, et al. Ablation of the leptin receptor in the hypothalamic arcuate nucleus abrogates leptin-induced sympathetic activation. Circ Res. 2011;108(7):808–812. doi:10.1161/CIRCRESAHA.111.240226

83. Park HK, Ahima RS. Physiology of leptin: energy homeostasis, neuroendocrine function and metabolism. Metabolism. 2015;64(1):24–34. doi:10.1016/j.metabol.2014.08.004

84. Henegar JR, Zhang Y, De Rama R, Hata C, Hall ME, Hall JE. Catheter-based radiorefrequency renal denervation lowers blood pressure in obese hypertensive dogs. Am J Hypertens. 2014;27(10):1285–1292. doi:10.1093/ajh/hpu048

85. Rahmouni K, Morgan DA. Hypothalamic arcuate nucleus mediates the sympathetic and arterial pressure responses to leptin. Hypertension. 2007;49(3):647–652. doi:10.1161/01.HYP.0000254827.59792.b2

86. Rahmouni K, Haynes WG, Morgan DA, Mark AL. Intracellular mechanisms involved in leptin regulation of sympathetic outflow. Hypertension. 2003;41(3):763–767. doi:10.1161/01.HYP.0000048342.54392.40

87. Wolf G, Hamann A, Han DC, et al. Leptin stimulates proliferation and TGF-beta expression in renal glomerular endothelial cells: potential role in glomerulosclerosis [seecomments]. Kidney Int. 1999;56(3):860–872. doi:10.1046/j.1523-1755.1999.00626.x

88. Han DC, Isono M, Chen S, et al. Leptin stimulates type I collagen production in db/db mesangial cells: glucose uptake and TGF-beta type II receptor expression. Kidney Int. 2001;59(4):1315–1323. doi:10.1046/j.1523-1755.2001.0590041315.x

89. Mao S, Fang L, Liu F, Jiang S, Wu L, Zhang J. Leptin and chronic kidney diseases. J Recept Signal Transduction Res. 2018;38(2):89–94. doi:10.1080/10799893.2018.1431278

90. Nillni EA. The metabolic sensor Sirt1 and the hypothalamus: interplay between peptide hormones and pro-hormone convertases. Mol Cell Endocrinol. 2016;438:77–88. doi:10.1016/j.mce.2016.09.002

91. Liu X, Zheng H. Modulation of sirt1 and FoxO1 on hypothalamic leptin-mediated sympathetic activation and inflammation in diet-induced obese rats. J Am Heart Assoc. 2021;10(14). doi:10.1161/JAHA.120.020667

92. Heiss CN, Mannerås-Holm L, Lee YS, et al. The gut microbiota regulates hypothalamic inflammation and leptin sensitivity in Western diet-fed mice via a GLP-1R-dependent mechanism. Cell Rep. 2021;35(8):109163. doi:10.1016/j.celrep.2021.109163

93. Achari AE, Jain SK. Adiponectin, a Therapeutic Target for Obesity, Diabetes, and Endothelial Dysfunction. Int J Mol Sci. 2017;18(6):1321. doi:10.3390/ijms18061321

94. Przybyciński J, Dziedziejko V, Puchałowicz K, Domański L, Pawlik A. Adiponectin in chronic kidney disease. Int J Mol Sci. 2020;21(24):9375. doi:10.3390/ijms21249375

95. Fang H, Judd RL. Adiponectin regulation and function. Compr Physiol. 2018;8(3):1031–1063.

96. Parida S, Siddharth S, Adiponectin SD. Obesity, and cancer: clash of the bigwigs in health and disease. Int J Mol Sci. 2019;20(10):2519. doi:10.3390/ijms20102519

97. Holland WL, Xia JY, Johnson JA, et al. Inducible overexpression of adiponectin receptors highlight the roles of adiponectin-induced ceramidase signaling in lipid and glucose homeostasis. Mol Metab. 2017;6(3):267–275. doi:10.1016/j.molmet.2017.01.002

98. Yang Q, Fu C, Zhang X, et al. Adiponectin protects against uric acid‑induced renal tubular epithelial inflammatory responses via the AdipoR1/AMPK signaling pathway. Int J Mol Med. 2019;43(3):1542–1552. doi:10.3892/ijmm.2019.4072

99. Xuan D, Han Q, Tu Q, et al. Epigenetic modulation in periodontitis: interaction of adiponectin and JMJD3-IRF4 axis in macrophages. J Cell Physiol. 2016;231(5):1090–1096. doi:10.1002/jcp.25201

100. Christou GA, Kiortsis DN. The role of adiponectin in renal physiology and development of albuminuria. J Endocrinol. 2014;221(2). doi:10.1530/JOE-13-0578

101. Carbone A, Al Salhi Y, Tasca A, et al. Obesity and kidney stone disease: a systematic review. Minerva Urol Nefrol. 2018;70(4):393–400. doi:10.23736/S0393-2249.18.03113-2

102. Francisqueti FV, Ferron AJT, Hasimoto FK, et al. Gamma oryzanol treats obesity-induced kidney injuries by modulating the adiponectin receptor 2/PPAR-α axis. Oxid Med Cell Longev. 2018;2018:1278392. doi:10.1155/2018/1278392

103. Guebre-Egziabher F, Bernhard J, Funahashi T, Hadj-Aissa A, Fouque D. Adiponectin in chronic kidney disease is related more to metabolic disturbances than to decline in renal function. Nephrol Dial Transplant. 2005;20(1):129–134. doi:10.1093/ndt/gfh568

104. Menon V, Li L, Wang X, et al. Adiponectin and mortality in patients with chronic kidney disease. J Am Soc Nephrol. 2006;17(9):2599–2606. doi:10.1681/ASN.2006040331

105. Rhee CM, Nguyen DV, Moradi H, et al. Association of adiponectin with body composition and mortality in hemodialysis patients. Am J Kidney Dis. 2015;66(2):313–321. doi:10.1053/j.ajkd.2015.02.325

106. Chen Y, Yang Y, Liu Z, He L. Adiponectin promotes repair of renal tubular epithelial cells by regulating mitochondrial biogenesis and function. Metabolism. 2022;128:154959. doi:10.1016/j.metabol.2021.154959

107. de Clercq NC, Frissen MN, Groen AK, Nieuwdorp M. Gut microbiota and the gut-brain axis: new insights in the pathophysiology of metabolic syndrome. Psychosom Med. 2017;79(8):874–879. doi:10.1097/PSY.0000000000000495

108. Qin J, Li R, Raes J, et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464(7285):59–65. doi:10.1038/nature08821

109. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A. 2005;102(31):11070–11075. doi:10.1073/pnas.0504978102

110. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: human gut microbes associated with obesity. Nature. 2006;444(7122):1022–1023. doi:10.1038/4441022a

111. Gomes AC, Hoffmann C, Mota JF. The human gut microbiota: metabolism and perspective in obesity. Gut Microbes. 2018;9(4):308–325. doi:10.1080/19490976.2018.1465157

112. Cani PD, Bibiloni R, Knauf C, et al. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes. 2008;57(6):1470–1481. doi:10.2337/db07-1403

113. Wang M, Wang Z, Chen Y, Dong Y. Kidney Damage Caused by Obesity and Its Feasible Treatment Drugs. Int J Mol Sci. 2022;23(2):747. doi:10.3390/ijms23020747

114. Lancaster GI, Langley KG, Berglund NA, et al. Evidence that TLR4 is not a receptor for saturated fatty acids but mediates lipid-induced inflammation by reprogramming macrophage metabolism. Cell Metab. 2018;27(5):1096–110.e5. doi:10.1016/j.cmet.2018.03.014

115. Kawai T, Autieri MV, Scalia R. Adipose tissue inflammation and metabolic dysfunction in obesity. Am J Physiol Cell Physiol. 2021;320(3):C375–c91. doi:10.1152/ajpcell.00379.2020

116. Watanabe Y, Nagai Y, Honda H, et al. Bidirectional crosstalk between neutrophils and adipocytes promotes adipose tissue inflammation. FASEB j. 2019;33(11):11821–11835. doi:10.1096/fj.201900477RR

117. Zheng Y, Tang L, Huang W, et al. Anti-inflammatory effects of Ang-(1-7) in Ameliorating HFD-induced renal injury through LDLr-SREBP2-SCAP pathway. PLoS One. 2015;10(8):e0136187. doi:10.1371/journal.pone.0136187

118. Brites F, Martin M, Guillas I, Kontush A. Antioxidative activity of high-density lipoprotein (HDL): mechanistic insights into potential clinical benefit. BBA Clin. 2017;8:66–77. doi:10.1016/j.bbacli.2017.07.002

119. Sharma I, Liao Y, Zheng X, Kanwar YS. New pandemic: obesity and associated nephropathy. Front Med Lausanne. 2021;8:673556. doi:10.3389/fmed.2021.673556

120. Wei L, Li Y, Yu Y, et al. Obesity-related glomerulopathy: from mechanism to therapeutic target. Diabetes Metab Syndr Obes. 2021;14:4371–4380. doi:10.2147/DMSO.S334199

121. Morales E, Valero MA, León M, Hernández E, Praga M. Beneficial effects of weight loss in overweight patients with chronic proteinuric nephropathies. Am J Kidney Dis. 2003;41(2):319–327. doi:10.1053/ajkd.2003.50039

122. Navaneethan SD, Yehnert H, Moustarah F, Schreiber MJ, Schauer PR, Beddhu S. Weight loss interventions in chronic kidney disease: a systematic review and meta-analysis. Clin J Am Soc Nephrol. 2009;4(10):1565–1574. doi:10.2215/CJN.02250409

123. Bolignano D, Zoccali C. Effects of weight loss on renal function in obese CKD patients: a systematic review. Nephrol Dial Transplant. 2013;28(4):iv82–98. doi:10.1093/ndt/gft302

124. Shen WW, Chen HM, Chen H, Xu F, Li LS, Liu ZH. Obesity-related glomerulopathy: body mass index and proteinuria. Clin J Am Soc Nephrol. 2010;5(8):1401–1409. doi:10.2215/CJN.01370210

125. Tchang BG, Saunders KH, Igel LI. Best practices in the management of overweight and obesity. Med Clin North Am. 2021;105(1):149–174. doi:10.1016/j.mcna.2020.08.018

126. Ritz E. Bariatric surgery and the kidney-much benefit, but also potential harm. Clin Kidney J. 2013;6(4):368–372. doi:10.1093/ckj/sfs161

127. Friedman AN, Wahed AS, Wang J, et al. Effect of Bariatric Surgery on CKD Risk. J Am Soc Nephrol. 2018;29(4):1289–1300. doi:10.1681/ASN.2017060707

128. Friedman AN. The case for a bariatric-centered approach to CKD care. Clin J Am Soc Nephrol. 2019;14(2):291–293. doi:10.2215/CJN.12061018

129. Abu Dayyeh BK, Maselli DB, Rapaka B, et al. Adjustable intragastric balloon for treatment of obesity: a multicentre, open-label, randomised clinical trial. Lancet. 2021;398(10315):1965–1973. doi:10.1016/S0140-6736(21)02394-1

130. Král J, Machytka E, Horká V, et al. Endoscopic treatment of obesity and nutritional aspects of bariatric endoscopy. Nutrients. 2021;13(12):4268. doi:10.3390/nu13124268

131. Morales E, Gutiérrez E, Caro J, Sevillano A, Rojas-Rivera J, Praga M. Beneficial long-term effect of aldosterone antagonist added to a traditional blockade of the renin-angiotensin-aldosterone system among patients with obesity and proteinuria. Nefrologia. 2015;35(6):554–561. doi:10.1016/j.nefro.2015.09.008

132. Bakris GL, Agarwal R, Chan JC, et al. Effect of finerenone on albuminuria in patients with diabetic nephropathy: a randomized clinical trial. JAMA. 2015;314(9):884–894. doi:10.1001/jama.2015.10081

133. Bakris GL, Agarwal R, Anker SD, et al. Effect of finerenone on chronic kidney disease outcomes in type 2 diabetes. N Engl J Med. 2020;383(23):2219–2229. doi:10.1056/NEJMoa2025845

134. Cherney DZI, Dekkers CCJ, Barbour SJ, et al. Effects of the SGLT2 inhibitor dapagliflozin on proteinuria in non-diabetic patients with chronic kidney disease (DIAMOND): a randomised, double-blind, crossover trial. Lancet Diabetes Endocrinol. 2020;8(7):582–593. doi:10.1016/S2213-8587(20)30162-5

135. McMurray JJV, Wheeler DC, Stefánsson BV, et al. Effect of dapagliflozin on clinical outcomes in patients with chronic kidney disease, with and without cardiovascular disease. Circulation. 2021;143(5):438–448. doi:10.1161/CIRCULATIONAHA.120.051675

136. Nihei N, Okamoto H, Furune T, et al. Dietary α-cyclodextrin modifies gut microbiota and reduces fat accumulation in high-fat-diet-fed obese mice. Biofactors. 2018;44:336–347. doi:10.1002/biof.1429

137. Ishigaki S, Ohashi N, Matsuyama T, et al. Melatonin ameliorates intrarenal renin-angiotensin system in a 5/6 nephrectomy rat model. Clin Exp Nephrol. 2018;22(3):539–549. doi:10.1007/s10157-017-1505-7

138. Liu Z, Gan L, Xu Y, et al. Melatonin alleviates inflammasome-induced pyroptosis through inhibiting NF-κB/GSDMD signal in mice adipose tissue. J Pineal Res. 2017;63(1). doi:10.1111/jpi.12414

139. Wilding JPH, Batterham RL, Calanna S, et al. Once-weekly semaglutide in adults with overweight or obesity. N Engl J Med. 2021;384(11):989. doi:10.1056/NEJMoa2032183

140. Casas JP, Chua W, Loukogeorgakis S, et al. Effect of inhibitors of the renin-angiotensin system and other antihypertensive drugs on renal outcomes: systematic review and meta-analysis. Lancet. 2005;366(9502):2026–2033. doi:10.1016/S0140-6736(05)67814-2

141. Novikov A, Vallon V. Sodium glucose cotransporter 2 inhibition in the diabetic kidney: an update. Curr Opin Nephrol Hypertens. 2016;25(1):50–58. doi:10.1097/MNH.0000000000000187

142. Wheeler DC, Stefánsson BV, Jongs N, et al. Effects of dapagliflozin on major adverse kidney and cardiovascular events in patients with diabetic and non-diabetic chronic kidney disease: a prespecified analysis from the DAPA-CKD trial. Lancet Diabetes Endocrinol. 2021;9(1):22–31. doi:10.1016/S2213-8587(20)30369-7

143. Wang XX, Levi J, Luo Y, et al. SGLT2 protein expression is increased in human diabetic nephropathy: SGLT2 protein inhibition decreases renal lipid accumulation, inflammation, and the development of nephropathy in diabetic mice. J Biol Chem. 2017;292(13):5335–5348. doi:10.1074/jbc.M117.779520

144. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444(7122):1027–1031. doi:10.1038/nature05414

145. Dinan TG, Cryan JF. Gut-brain axis in 2016: brain-gut-microbiota axis - mood, metabolism and behaviour. Nat Rev Gastroenterol Hepatol. 2017;14(2):69–70. doi:10.1038/nrgastro.2016.200

146. Green M, Arora K, Prakash S. Microbial medicine: prebiotic and probiotic functional foods to target obesity and metabolic syndrome. Int J Mol Sci. 2020;21(8):2890. doi:10.3390/ijms21082890

147. Prado NJ, Ferder L, Manucha W, Diez ER. Anti-inflammatory effects of melatonin in obesity and hypertension. Curr Hypertens Rep. 2018;20(5):45. doi:10.1007/s11906-018-0842-6

148. Yea JH, Yoon YM, Lee JH, Yun CW, Lee SH. Exosomes isolated from melatonin-stimulated mesenchymal stem cells improve kidney function by regulating inflammation and fibrosis in a chronic kidney disease mouse model. J Tissue Eng. 2021;12:20417314211059624. doi:10.1177/20417314211059624

Creative Commons License © 2024 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.