Your new experience awaits. Try the new design now and help us make it even better

REVIEW article

Front. Endocrinol., 30 July 2025

Sec. Renal Endocrinology

Volume 16 - 2025 | https://doi.org/10.3389/fendo.2025.1631312

Research progress in the treatment of lipid metabolism disorder in patients with diabetic kidney disease by the integrated traditional Chinese and Western medicine

Lingli Sheng,&#x;Lingli Sheng1,2†Ziyi Cao&#x;Ziyi Cao3†Lin WangLin Wang2Youhua Xu*Youhua Xu1*Dingkun Gui,*Dingkun Gui1,4*
  • 1Faculty of Chinese Medicine, Macau University of Science and Technology, Macao, Macao SAR, China
  • 2Nephropathy Department II, Longhua Hospital Shanghai University of Traditional Chinese Medicine, Shanghai, China
  • 3College of Integrated Chinese and Western Medicine, Shaanxi University of Chinese Medicine, Xianyang, China
  • 4Department of Nephrology, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China

Diabetic kidney disease (DKD) is one of the most common and severe chronic microvascular complications of diabetes mellitus (DM). The pathogenesis of DKD is complex, and lipid metabolism disorders play an important role in the pathogenesis of DKD. DKD belongs to the category of “kidney deficiency”, “edema”, “guan ge” and other pathological factors secondary to “thirst quenching disease” in traditional Chinese medicine. The pathological factors mainly focus on blood stasis and toxicity, which is consistent with modern medical theory. At present, the efficacy and safety of integrated traditional Chinese and Western medicine in treating lipid metabolism disorders in patients with DKD have been extensively studied and confirmed. In this review, the application and possible mechanism of traditional Chinese patent medicines (Bailing Capsule, Shenyan Kangfu Tablets, Jinshuibao Capsule, Huangkui Capsule, Yi-Shen-Hua-Shi granule, Shenmai injection), Chinese medicine compound (Tangshen Formula, Danggui Buxue Decoction, Tangshenkang), single Chinese medicine (Astragalus membranaceus, Panax notoginseng, Salvia miltiorrhiza) combined with Western medicine in the treatment of DKD with lipid metabolism disorder were discussed, in order to provide ideas for clinical diagnosis and treatment of patients with DKD with lipid metabolism disorder.

1 Introduction

Diabetic kidney disease (DKD) is a serious microvascular complication of diabetes mellitus (DM) (1). With the increase of global prevalence of diabetes, about 30%~40% of DM patients will progress to DKD, which is the most common cause of end-stage renal disease (ESRD) (2). Therefore, early screening, control, and treatment of DKD is extremely important. The treatment strategy for early DKD is not just about controlling blood glucose, other related pathogenic factors cannot be ignored. The Chinese Guideline for the Prevention and Treatment of Type 2 diabetes (2020) proposed that lipid metabolism disorder is one of the risk factors of DKD, which should be paid more attention to and controlled.

Lipids are the significant component of the cell membrane, playing a crucial part in energy generation, cellular homeostasis, cell signal transduction and survival (3). Lipids have duality. Moderate increase has a positive effect, while overdose can arise oxidative damage, resulting in tissue lipid peroxidation and ultimately lipid toxicity. Lipid homeostasis is prominent in preventing lipid toxicity (4). Modern pathology believes that lipid deposition in the kidneys has become a momentous factor leading to renal damage in DKD. High levels of lipids in the human body can provoke renal tissue fat deposition, glomerulosclerosis, and mesangial dilation, exacerbating proteinuria and progression of tubulointerstitial fibrosis, and directly damaging podocytes, leading to irreversible damage to renal function (5, 6). Thus, regulation of lipid metabolism disorder is vital significance for reducing the incidence rate of lipid metabolism disorder in DKD and slowing its progress.

In recent years, molecular biology has been developing rapidly, With the widespread application of ultra-high performance liquid chromatography and mass spectrometry, the active ingredients, indication and mechanism of traditional Chinese medicine (TCM) have been further studied in depth. As primary or alternative therapy for DKD, TCM has shown good clinical efficacy. More and more studies emphasize the identification of bioactive compounds in TCM and the molecular mechanisms of renal protection (1).

This review aims to discuss the integrated treatment of traditional Chinese and Western medicine for lipid metabolism disorders in patients with DKD, emphasize the significance of prevention and treatment of DKD and the importance of simultaneous regulation of lipid metabolism disorder in the treatment process; TCM will play a leading part in the treatment of such diseases with its advantages of multi-target, multi-pathway, and compound efficacy. Then, we hope this review could provide reference for clinical diagnosis and scientific research.

2 The pathogenesis of DKD combined with lipid metabolism disorders

DKD patients typically suffer from lipid metabolism disorders, and dyslipidemia occurs throughout the entire process of illness (7). In the early stage of DKD, hypertriglyceridemia develops. Then overt DKD patients with macroalbuminuria have elevated levels of total cholesterol (TC) and significantly increased low-density lipoprotein (LDL) cholesterol. And renal failure increases lipoproteins rich in triglyceride (TG) (such as chylomicrons and very low density lipoprotein) and decreases high-density lipoprotein (HDL) cholesterol (8, 9). Ectopic lipid deposition refers to the excessive accumulation of lipids, notably TGs, in lean tissues like the kidneys, causing lipotoxic damage and then gives rise to a series of pathological and physiological changes (10).

The kidney is rich in mitochondria so that it is a highly active metabolic organ with high energy demands (11). The demands for ATP vary in different regions of the kidney, with glucose being the preferred energy matrix for renal substrates. The glomerulus tends to use glucose, while the tubules prefer fatty acids (FAs) (12). Podocytes, also known as glomerular epithelial cells, form a silt diaphragm (SD) with their foot processes, which are a portion of the glomerular filtration barrier (GFB) and restrict the filtration of albumin into urinary space (13). SD is assembled in lipid rafts, which are special membrane domains rich in cholesterol and sphingolipids (14, 15). However, lipid accumulation can induce podocyte hypertrophy, the disappearance of podocyte processes and apoptosis, causing irreversible damage to podocytes, resulting in a large amount of protein entering the urine and intensifying the progression of DKD (16). Renal tubular cells and podocytes are prone to lipid accumulation, inducing mitochondrial stress, inflammation, insulin resistance (IR), endoplasmic reticulum stress (ERS), and ultimately causing cell death (17). Under physiological conditions, TGs from the diet are decomposed into free fatty acids (FFAs) and glycerol in the cytoplasm. Fatty acid transporters are mainly responsible for FFA transport in renal tubular cells, for instance, fatty acid transport protein-2(FATP2) (18). And B-class scavenger receptors, such as CD36, mainly focus on FFA transport in podocytes (19). After being stimulated by palmitate (a saturated FFA), the expression of CD36 is upregulated in podocytes, speeding up the transport of CD36 from the cytoplasm to the cell membrane, then enhancing the lipid uptake of podocytes, leading to oxidative stress (20). Fatty acids (FAs) undergo beta oxidation in cells, also called FA oxidation (FAO), then form acetyl-CoA or are stored in the form of lipid droplets (LD), while glycerol directly enters the glycolytic pathway (21). Both incomplete (FAO and lipid peroxidation can bring about oxidative stress, endoplasmic reticulum stress, and activation of pro-inflammatory processes (22). If too much acetyl-CoA is produced, the tricarboxylic acid cycle (TCA cycle) will overload, provoking the conversion of acetyl-CoA to ketone bodies, which can be taken for a fuel origin at low glucose levels. Nevertheless, if glucose levels are high, excess acetyl-CoA can be transformed into FAs, TG, cholesterol, steroids, and bile salts, a process known as adipogenesis (21). That is, long-term hyperglycemia in diabetes patients will aggravate fatty acid synthesis and TG accumulation.

What’s more, sterol regulatory element binding proteins (SREBPs) are transcription factors participating in regulating lipid biosynthesis, among which SREBP-1a and SREBP-1c preferentially activate genes involved in fat production, while SREBP-2 is mainly responsible for transcriptional regulation of cholesterol homeostasis related genes (23). In the kidney, the increase of TG level is related to the increase of SREBP-1c expression level (24). Peroxisome proliferator activated receptor (PPAR)α, PPARβ/δ, and PPARγ are members of the ligand activated transcription factor nuclear receptor family (25). PPARα, a ligand dependent nuclear receptor, takes part in the process of lipid metabolism, which is sensitive to exogenous compounds such as FAs, and regulates FAO like PPARβ/δ (26, 27). PPARγ has two contrary effects on lipid metabolism. On the one hand, the elevation of PPARγ can bring about the expression of lipoprotein lipase (LPL), causing fat breakdown, reducing TG and LDL-C levels, then improving lipid imbalance (28). On the other hand, PPARγ activation can promote CD36 expression so that lipid deposition occurs in renal tubular cells (29). But the specific mechanism needs additional experimental verification. Furthermore, peroxisome proliferator activated receptor gamma coactivator-1α (PGC-1α) is related to mitochondrial biogenesis and fatty acid beta oxidation directly. Inhibiting PGC-1α can significantly accelerate the reduction of FAO and increase lipid accumulation (Figure 1) (30, 31). Meanwhile, PGC-1α can strengthen the expression of farnesoid x receptor (FXR) to regulate triglyceride metabolism (32). Accordingly, overexpression of PGC-1α can protect renal cells from lipotoxicity to a certain extent and play an important role in the pathogenesis of DKD. Moreover, stearoyl-CoA desaturase (SCD) is a rate limiting enzyme that converts saturated FAs into monounsaturated FAs, resulting in the formation of neutral LD (33). Overexpression of SCD1 can reduce apoptosis of proximal renal tubular epithelial cells induced by saturated FA (34).

Figure 1
Flowchart illustrating lipid metabolism pathways leading to lipid accumulation. Triglycerides break down into glycerol and free fatty acids (FFA). FFA promotes CD36 and lipid uptake, forming lipid droplets (LD), leading to accumulation. Glycerol enters the glycolytic pathway, and fatty acids (FA) undergo beta oxidation to acetyl-CoA, which also forms from high glucose. Acetyl-CoA contributes to TCA cycle overload, ketone bodies, and synthesis of fatty acids (FAs), triglycerides (TG), cholesterol, steroids, and bile salts, enhancing lipid accumulation.

Figure 1. The mechiasm of lipid accumulation. TGs from the diet are decomposed into free fatty acids (FFAs) and glycerol in the cytoplasm. After being stimulated by FFA, the expression of CD36 is upregulated in podocytes, speeding up the transport of CD36 from the cytoplasm to the cell membrane. Fatty acids (FAs) undergo beta oxidation in cells, also called FA oxidation (FAO), then form acetyl-CoA or are stored in the form of lipid droplets (LD), while glycerol directly enters the glycolytic pathway. If too much acetyl-CoA is produced, the tricarboxylic acid cycle (TCA cycle) will overload, provoking the conversion of acetyl-CoA to ketone bodies. if glucose levels are high, excess acetyl-CoA can be transformed into FAs, TG, cholesterol, steroids, and bile salts, a process known as adipogenesis.

HDL is a cholesterol reverse transporter protein which can unrestricted enter and exit the arterial wall, uptake harmful substances such as LDL, cholesterol, TGs, etc. deposited in the inner layer of the vascular wall, and transport them to the liver for decomposition and excretion (35). Apolipoprotein (APO) A-I is the main component of HDL. Studies have found that it can increase glucose tolerance and insulin sensitivity, inhibit liver gluconeogenesis, reduce liver glycogen output, and have an important protective effect on diabetes and its complications (36). At the same time, the decrease in HDL levels downregulated the levels of circulating APOM, APOE and APOL1 (21). ATP-binding cassette transporter A1 (ABCA1) controls cholesterol efflux by transporting free cholesterol and phospholipids from the cell to high-density lipoprotein particles containing APOA-I (37). The expression of ABCA1, participated in impaired cholesterol efflux, declines in DKD patients’ podocytes (38). Studies have shown that the deletion of Subtilisin/kexin type 9 serine protease (PCSK9) can improve dyslipidemia (39). Especially in the high-fat diet animal model of kidney injury, the reduction of PCSK9 level can promote renal lipid accumulation caused by CD36 (40). PCSK9 can reduce the level of LDL receptor on plasma membrane, thereby promoting cholesterol influx of circulating LDL (21). Elevated plasma LDL levels and the formation of foam cells (macrophages that ingest LDL) stimulate the release of pro-inflammatory cytokines and accelerate the inflammatory response, thereby causing kidney injury by affecting lipid metabolism and causing oxidative stress (41). LDL size is negatively regulated by serum TG levels, and LDL size is significantly reduced in hypertriglyceridemic subjects (42). Ox-LDL is easier to deposit on the inner wall of blood vessels than normal LDL, leading to atherosclerosis and other cardiovascular complications. Klotho is an important renoprotective protein, which can effectively eliminate renal ox-LDL deposition through IGF-1R/RAC1/OLR1 signaling axis, thereby improving podocyte injury (43). Liver x receptors (LXRs) are ligand activated nuclear receptor transcription factors that play a vital part in regulating cholesterol and triglyceride metabolism by regulating the expression of fatty acid synthesis related genes such as SREBP1-c (24). In addition, the LXR pathway may regulate the expression of ABCA1 gene through PPAR (28). FXR is a multifunctional transcription factor that regulates bile acid homeostasis and glucose and lipid metabolism in various tissues (44). It can also alleviate renal fibrosis (45), regulate mitochondrial biogenesis pathways (46), and reduce TGFβ-SMAD signal transduction and inflammatory response in renal mesangial cells (47), deferring the progression of DKD (48).

3 Integrated traditional Chinese and Western medicine treatment for DKD combined with lipid metabolism disorder

As mentioned earlier, DKD combined with lipid metabolism disorders involves multiple mechanisms such as lipid deposition, hyperglycemia, oxidative stress, inflammation, and fibrosis. At present, Western medicine clinical practice mainly reduces kidney damage and delays the progression of kidney disease by controlling blood lipids, blood sugar, and blood pressure levels.

At present, the clinical therapeutic drugs in Western medicine mainly include the following categories: fibrates, HMG-CoA reductase inhibitors (statins), proprotein convertase subtilisin-kexin type 9 (PCSK9) inhibitors, sodium-glucose transporter-2 inhibitors (SGLT2i), glucagon-like peptide-1 (GLP-1) receptor agonists, renin angiotensin aldosterone system (RAAS) blockers etc (49).

Firstly, as an activator of PPARα, fibrates can improve fatty acid β oxidation and lipid accumulation in DKD mice through AMPK/FOXA2/MACD pathway, and alleviate apoptosis of renal tubular cells (50). However, clinical trials have shown that fibrates have an improving effect on proteinuria, but further research is needed to investigate the impact of fibrates on eGFR (49). Secondly, it has been confirmed that statins reduce the risk of cardiovascular disease in patients with DM and hypertension (51). Studies have shown that statins can improve proteinuria and reduce estimated glomerular filtration rate (eGFR) decline rate in DKD patients (52, 53). However, the specific impact on DKD is still unclear. Then, PCSK9 inhibitors are considered as alternative or adjuvant drugs to statins, but there is insufficient data on the impact of PCSK9 inhibitors on the progression of DKD (49). In addition, the strong protective effect of anti-diabetic drug, SGLT2i, on the kidney has been proved, and its mechanism may include anti lipid toxicity (54). GLP-1 receptor agonists can reduce blood lipids and alleviate renal ectopic lipid deposition in DKD rats (55). At the same time, it can also improve the decrease in urinary albumin and significantly slow down the decrease in eGFR in type 2 DM patients (56). RAAS blockers have been used as therapeutic drugs for DKD for decades (57). Commonly used drugs include angiotensin converting enzyme inhibitors (ACEI), angiotensin receptor blockers (ARB), and mineralocorticoid receptor antagonists (MRA), all of which can improve proteinuria and kidney injury. However, it should be noted that spironolactone and eperenone have a risk of hyperkalemia (58).

According to the theory of TCM, lipid metabolism disorder belongs to the categories of “phlegm turbidity”, “qi stagnation” and “blood stasis”. These pathological factors can lead to impaired blood flow and poor circulation of qi and blood. Over time, there is turbidity and blood stasis in blood vessels, obstruction of the venation, which can then lead to various diseases. The pathogenesis of DKD is consistent with it, that is, phlegm turbidity affects the normal consolidation and drainage of the kidney, causes proteinuria, hinders the blood flow of the kidney for a long time, forms blood stasis, aggravates the burden of the kidney, and accelerates the disease process (59).

3.1 Traditional Chinese patent medicines combined with Western medicine in the treatment of DKD with lipid metabolism disorder

Traditional Chinese patent medicines refer to a kind of medicine with certain specifications which can be directly taken under the guidance of the theory of TCM, with Chinese herbal medicine as the raw material, according to the prescribed prescription compatibility and preparation standards.

3.1.1 Bailing capsule

Bailing Capsule is a capsule made from fermented Cordyceps sinensis powder (C. sinensis), mainly composed of mannitol, adenosine, and total amino acids. Its main functions are to nourish the lungs and kidneys, benefit essence and qi, and be commonly used as an adjuvant therapy for chronic renal insufficiency in clinical practice (60). It has anti-inflammatory, antioxidant, and proteinuria reducing functions (61). In addition, Bailing capsules are also used to treat chronic bronchitis, improve symptoms such as cough, asthma, and lower back pain in patients (60, 62). Multiple clinical studies have demonstrated that BC can deter the inflammatory process by reducing the expression of inflammatory factors such as hypersensitive C-reactive protein (hs-CRP), tumor necrosis factor-α (TNF-α) and interleukin(IL)-18, improve oxidative stress function, like a significant increase in superoxide dismutase (SOD) levels and a significant decrease in malondialdehyde (MDA) levels, reduce urinary protein, improve lipid metabolism and protect renal function (63, 64). Wang Wenru et al. (65) found that BC is superior in reducing TG through meta-analysis. A molecular biology study has shown that BC inhibits adipogenesis through triggering SCD and suppressing fatty acid synthase (FASN) expression, activates PPARα pathway and downstream acyl-coenzyme A oxidase 1 (ACOX1), a lipolytic enzyme regulated by PPARα in lipid oxidation, to enhance lipolysis, thereby delaying on the progression of DKD in rats and protecting renal function (66).

ARB are used to treat different types of hypertension, such as irbesartan, losartan, valsartan, etc. This class of drugs inhibits the production of angiotensin II, dilates the renal arteries, improves the state of hyperfiltration, hyperperfusion, and high pressure in the glomeruli, and significantly reduces the excretion of urinary protein (67). When BC is used in combination with ARB drugs, it can effectively improve the renal function of patients with DKD, reduce urinary protein excretion, and improve lipid metabolism disorders (68, 69). Dapagliflozin is a sodium-glucose cotransporter 2 inhibitor (SGLT2i), and the incidence of adverse reactions in the treatment of chronic kidney disease is relatively low. The combination of BC and dapagliflozin in the treatment of DKD can enhance the therapeutic effect, reduce renal injury, and delay the progression of the disease (70). Pancreatic kininogenase can prevent platelet formation and thrombosis, balance the hemodynamic disorders caused by the activation of the RAAS, reduce the leakage of urinary protein, and delay the progression of DKD. The combination of BC and pancreatic kininogenase can effectively improve hypertriglyceridemia in patients with early-onset DKD, reduce the level of proteinuria in stage III of DKD, and there are no adverse reactions during the medication period (71). Simvastatin and atorvastatin are commonly used statin drugs in clinical practice. When each of them is used in combination with BC, the improvement of blood lipids in DKD patients is better than when used alone, and it can significantly improve renal function and reduce the inflammatory response (72, 73). ,

3.1.2 Shenyan Kangfu tablets

SKT is composed of thirteen traditional Chinese medicines, including Salvia miltiorrhiza Bunge, Panax ginseng, Eucommia ulmoides, Scleromitrion diffusum, Radix Rehmanniae, Semen Sojae Nigrum, Rhizoma Dioscoreae, American Ginseng, Alisma plantago-aquatica, Rhizoma Smilacis Glabrae, Leonurus japonicus, and Radix Platycodonis. Its main ingredients are Tanshinone IIA, Ginsenoside Rb1, Ginsenoside Re, etc. It has effects on nourishing qi and yin, strengthening spleen and kidney, clearing residual toxins, and is commonly used in patients with chronic nephritis, proteinuria, and hematuria to improve symptoms such as edema, fatigue, dizziness, and tinnitus in clinical practice (60). Specifically, Wang X. et al. (74) used network pharmacology and metabolomics techniques to jointly elucidate the mechanism of SKT in improving DKD. The results showed that SKT can inhibit IR and correct abnormal metabolism in DKD mice through the starch sucrose metabolism pathway and the biosynthesis of unsaturated fatty acids. It mainly inhibits renal inflammation by regulating the TNF signaling pathway and Toll-like receptor signaling pathway, with IL-6, Mitogen-activated protein kinase-3 (MAPK3), vascular endothelial growth factor (VEGF), TNF, etc. being its main targets of action. In addition, Chen Q. et al. (75) found that SKT inhibits the expression of inflammatory mediators nuclear factor-κB (NF-κB), TNF-α, and IL-1β, and its anti-inflammatory activity goes hand in hand with the abundance of gut microbiota.

Glutathione is an effective antioxidant substance within cells, which can improve the functions of the glomeruli and renal tubules. When used in combination with SKT, it can significantly reduce the TC level in patients with DKD and remarkably improve urinary protein (76). Alprostadil is commonly used clinically to improve microcirculation. When combined with SKT, it has a better effect on improving the renal function and blood lipids of patients with early-stage DKD compared with using it alone, and it has good safety (77).

3.1.3 Jinshuibao capsule

According to the pharmacopoeia (60), JSB is made by packaging fermented cordyceps powder (CS-4) into capsules. Its main ingredients are uridine, guanosine, adenosine, ergosterol, etc. With the functions of nourishing the lungs and kidneys, and replenishing essence and qi, JSB is commonly used in clinical practice for diseases such as chronic renal insufficiency hyperlipidemia, chronic bronchitis, cirrhosis and other diseases. JSB have therapeutic effects on stable chronic obstructive pulmonary disease (COPD) by increasing FEV1% pred, FEV1/FVC ratio, FEV1, FVC, and PaO2 levels, while reducing PaCO2 levels (78). At the same time, JSB increases the levels of serum SOD and calcitonin gene-related peptide (CGRP) and reduces C-reactive protein (CRP) and endothelin (ET) -1 to enhance the body’s antioxidant capacity and reduce the severity of micro inflammatory reactions in the body and thus has a positive regulatory effect on renal hemodynamics with significant therapeutic effects (79, 80). What’s more, an in vitro experiment (81) has shown that fermented cordyceps has significant renal protective effects, which are achieved by facilitating proliferation and hindering apoptosis of proximal renal tubular cells, possibly through targeting Caspase-3, Bcl-2-associated X protein (Bax), VEGFA, and phosphatase and tensin homolog deleted on chromosome ten (PTEN). The protein kinase B (AKT) and extracellular regulated protein kinases (ERK) signaling pathways might be the key mechanisms for the therapeutic efficacy of fermented cordyceps in treating DKD.

Ramipril belongs to ACEI, which can effectively reduce the intraglomerular pressure and decrease the excretion of urinary protein (82). The combination of JSB and ramipril in the treatment of patients with DKD can significantly reduce urinary protein, regulate the disordered lipid metabolism, and have a positive regulatory effect on renal hemodynamics, with obvious therapeutic effects (80). Moreover, The combination of JSB with telmisartan and candesartan cilexetil can protect renal function, improve blood lipid levels, effectively slow down the progression of diabetic kidney disease (DKD), and its therapeutic effect is significantly better than that of using western medicine alone (83, 84). Sulodexide can achieve the goal of treating early-stage DKD through multiple pathways, such as improving the hypercoagulable state of the body’s blood, renal blood perfusion, reducing proteinuria, and antagonizing the apoptosis of glomerular cells (85). When combined with JSB, it can significantly reduce the urinary albumin excretion in patients with early-stage DKD, protect renal function, correct lipid metabolism disorders, improve endothelial function, alleviate the micro-inflammatory state, enhance the body’s antioxidant capacity, and regulate renal hemodynamics (79).

3.1.4 Huangkui capsule

HKC, extracted from Abelmoschus manihot, can promote diuresis, remove blood stasis, and clear heat. Its main component is flavonoids, which effectively treat damp heat syndrome and DKD (damp heat combined with blood stasis) (8688). Clinical studies demonstrated that HKC can improve renal function and lipid metabolism disorders in DKD patients significantly. The mechanism is probably reducing serum MDA, increasing serum GSH-Px and SOD, inhibiting oxidative stress, and thus exerting a renal protective effect (89, 90). Moreover, HKC treatment for DKD has a significant anti-inflammatory effect, manifested in the reduction of TNF-α, hs-CRP, IL-2, IL-6, IL-8, IL-1β, γ-glutamyl transferase (GGT), and advanced oxidation protein product (AOPP) levels, thereby increasing capillary permeability, reducing edema, improving inflammatory response and oxidative stress status in the body, ultimately alleviating kidney damage (9196). Animal experiments have shown that HKC treatment in DKD rats can improve kidney morphology and weight, as well as renal hypertrophy index, and alleviate glomerular hypertrophy, GBM pachynsis, and mild renal interstitial dilation. At the same time, podocyte foot process fusion is reduced, the proliferation of capillary endothelial cells is mild (97, 98). HKC increased the expression of PPARα and its target genes (such as LPL and aP2), PPARγ and its target genes (such as LPL, CPT-1, ACO, and CYP4A) in the liver and kidneys of DKD rats. These genes participate in entry, oxidation and hydroxylation of fatty acids, which may lead to the fall-off in fatty acid exposure in the kidneys of DKD rats (99). Therefore, these influences may trigger a decrease in the synthesis and secretion of triglycerides. In addition, the secreted protein acidic and rich in cysteine (SPARC) is secreted by adipocytes and its expression and secretion are impacted by fat mass, leptin, insulin, and glucose, and it is involved in the pathogenesis of tumorigenesis and renal and liver fibrosis (100, 101). HKC can also inhibit SPARC levels, inhibit Klotho, downregulate phosphorylated p38MAPK and phosphorylated Akt (p-Akt) in DKD rats to alleviate renal fibrosis (102104). Furthermore, HKC can alleviate epithelial-mesenchymal transition (EMT) in renal tubules of DKD rats by suppressing the activation of NLRP3 inflammasome and TLR4/NF-κB signaling transduction in the kidneys (105). W. Wu et al. (98) found that HKC can restrain the phosphorylation of Akt, mTOR and p70S6K in the kidneys, as well as the overexpression of TGF-β1 protein. Its active ingredient, hyperoside, can inhibit the phosphorylation of PI3K, Akt, mTOR and p70S6K in murine mesenchymal cells induced by high glucose, further demonstrating that HKC can safely and effectively relieve early DKD glomerular pathological changes. Additionally, HKC can readjust the activity of solute carriers (SLC) in both renal proximal and distal tubules, thereby inhibiting the development of DKD (106).

A Meta-analysis (107) has shown that the combination of HKC and ARB/ACEI drugs has obvious advantages in the treatment of early-stage DKD. It significantly improves the clinical treatment efficiency, and is more effective in reducing the levels of urinary albumin excretion rate (UAER), serum creatinine (Scr), blood urea nitrogen (BUN), TG and TC. Moreover, it does not increase the probability of occurrence of adverse reactions. Another systematic review also elaborated the efficacy of HKC combined with Western medicine in the treatment of DKD (108),

3.1.5 Yi-Shen-Hua-Shi granule

YSHS is composed of Panax ginseng, Astragalus membranaceus, Atractylodes macrocephala, Poria cocos, Panax quinquefolius, Pinellia ternata, Hansenia weberbaueriana, Angelica pubescens, Saposhnikovia divaricata, Bupleurum chinense, Coptis chinensis, Paeonia lactiflora, Citri reticulatae pericarpium, Radix glycyrrhizae preparata, Zingiberis rhizoma recens and Jujubae fructus, which is good at promoting yang and tonifying the spleen, nourishing kidneys and removing dampness, promoting diuresis and reducing swelling (109). It is commonly used in clinical practice to treat patients with chronic glomerulonephritis (Scr<2mg/dl) and improve their symptoms such as proteinuria, edema, and cold sensitivity. Its main ingredients are Ginsenoside Rg1, Ginsenoside Re, Ginsenoside Rb1 and Astragaloside (60). There are studies indicating that YSHS have the potential to treat IgA nephropathy and sepsis induced acute kidney injury (110, 111). After treatment, SOD levels increased; MDA, TNF-αand IL-6 levels decreased, and insulin-producing beta cell function index increased, suggesting that its mechanism perhaps has a bearing on improving oxidative stress levels and increasing insulin beta cell function index (112). Shen Zhen et al. (109) found that six pathways were simultaneously enriched through a combined analysis of metabolomics and transcriptomics, consisting of glycerophospholipid metabolism, arachidonic acid metabolism, purine metabolism, primary bile acid biosynthesis, ascorbic acid and aldehyde metabolism, and galactose metabolism. What’s more, studies have indicated that YSHS can improve gut microbiota translocation, regulate gut microbiota structure, increase microbiota diversity and increase the abundance of probiotics such as lactobacilli (which is instrumental in reducing fasting blood glucose, ameliorating glycolipid metabolism, alleviating insulin resistance and reducing tissue damage) (113) through the “gut kidney axis” in DKD rats, thereby improving glucose and lipid metabolism, lowering inflammation levels, lowering urinary toxins in the body, and then protecting kidney function (114, 115). YSHS can regulate the blood sugar and lipid levels of DKD rats, alleviate the symptoms of IR and podocyte reduction, and is conducive to the repair of GFB function, possibly correlated with the increase of nephrin and podocin protein levels and the decrease of platelet-derived growth factor receptorsβ (PDGFRβ) protein levels (116).

Clinical studies have suggested that YSHS can safely and effectively improve clinical symptoms, renal function, and blood lipid metabolism levels in DKD patients. It is more effective than using Western medicine alone, such as calcitriol, ligustrazine hydrochloride, and sulodexide. When combined with Tripterygium wilfordii Hook. f. polysaccharide tablets, YSHS has a synergistic and detoxifying effect (112, 117123).

3.1.6 Shenmai injection

SMI is made by mixing extracts of talinum paniculatum, radix ophiopogonis, etc. Among them, talinum paniculatum greatly replenishes qi, restores the pulse condition and solidifies the body, and enriches qi and blood; radix ophiopogonis nourishes yin and moistens the lungs, benefits the stomach and generates fluids, clears the heart and eliminates annoyance (124). Compatibility of the two has the functions of nourishing qi, strengthening the body constitution, nourishing yin and generating fluids, and promoting blood circulation. Modern pharmacological studies have demonstrated that the major active ingredients of SMI are Ginsenoside Rb1, Ginsenoside Rg1, Ginsenoside Re, and Saponin D, which interact directly with IL-6/STAT3 directly (125). In clinical practice, SMI is widely used for various diseases, such as ischemic stroke, viral myocarditis, and so on (126, 127).

A randomized controlled trial (128) involving 68 patients with DKD showed that the combination of SMI and alprostadil treatment can significantly improve the relevant indicators of qi deficiency and blood stasis type DKD patients, namely Scr, blood urea nitrogen (BUN), UAER, TC, TG, and effectively improve blood lipids and renal function in DKD patients.

Additionally, Qiyao Xiaoke Capsule (129), Soft Capsules of Compound Oil of Jujube, Arboruitae and Gardenia (130), Liuwei Dihuang Pills (131), Keluoxin Capsules (132), Compound Danshen Dropping Pills (133), Shenqi Jiangtang Granules (134), Tianqi Jiangtang Capsules (135, 136), these traditional Chinese patent medicines have been proved to improve the lipid metabolism disorder in DKD patients or animal models, improve renal function, and delay disease progression.

3.2 Traditional Chinese medicine compound combined with Western medicine in the treatment of DKD with lipid metabolism disorder

The traditional Chinese medicine compound is a clinically effective formula composed of two or more single herbs used according to the principles of TCM syndrome differentiation and treatment and the principles of composition and prescription. Specifically, it is formulated according to the “Monarch, Minister, Assistant and Guide” compatibility principles.

3.2.1 Tangshen formula

TSF is composed of Astragalus membranaceus, Ghost arrow feather, Rehmannia glutinosa, Fructus aurantii, Cornus officinalis, Rhubarb, and Panax notoginseng (137). Qin et al. (138) identified representative components of TSF, including loganin, calycosin-7-O-b-D-glucoside, naringenin-7-rhamnosidoglucoside, neohesperidin, naringenin and aloeemodin, by high-performance liquid chromatography (HPLC), which improved the blood lipid levels and reduced liver steatosis in db/db mice. Molecular biology studies (139, 140) have shown that TSF can significantly inhibit urinary albumin excretion and relieve kidney damage in DKD rats. It downregulates the expression of pro-inflammatory factors: GSDMD, NLRP3, IL-1β, IL-18 and Caspase-1 in vivo, and blocks renal inflammation driven by NF-κB and renal fibrosis mediated by TGF-β/Smad3 through blocking the Smad7 degradation pathway mediated by Smurf2, preventing apoptosis of renal tubular epithelial cells through regulating the TXNIP-NLRP3-GDMD axis in vitro. Liu Ping et al. (141) found that TSF treatment reduced serum LDL-C, TC and TG levels in db/db mice, while HDL-C levels had no significant effect. Upregulation of ABCA1 reduced cholesterol accumulation in the kidneys of db/db mice, indicating that the promotion of cholesterol efflux mediated by ABCA1 may contribute to the therapeutic effect of DKD.

A multi-center, randomized, double-blind, placebo-controlled clinical study has shown that the combination of TSF and irbesartan has demonstrated good effects in reducing proteinuria and increasing the estimated glomerular filtration rate in patients with DKD. Among them, the blood lipid levels of patients with massive proteinuria have been improved, while in patients with microalbuminuria, only the LDL has shown a significant change, and there are no significant differences in other blood lipid levels (142).

3.2.2 Danggui Buxue decoction

DBD is a classic formula for nourishing qi and blood, composed of Astragalus membranaceus and Angelica sinensis Radix in a ratio of 5:1. In this formula, Astragalus membranaceus is used to nourish the lungs and spleen, nourishing the source of biochemistry. Angelicae sinensis Radix is used to supplement blood and nourish the body. Thus, tangible blood is born from intangible qi, and replenishing qi generates blood (143). Moreover, a study on the application of DBD in combination with Jingui Shenqi Pill in 80 DKD patients indicated that the combination of the two could decrease the Scr, urea nitrogen and 24-hour urine protein content more than using Jingui Shenqi Pill alone. At the same time, TC, TG, LDL-C diminished, and HDL-C dramatically increased (144). A molecular study (145) has shown that DBD can alleviate endoplasmic reticulum stress in DKD rat models by inhibiting PERK pathway related proteins, protecting kidney tissue and function, improving TC and TG, and even improving MAU and HDL at high doses compared to gliclazide (146). A network pharmacology study (147) based on lipidomics and transcriptomics has shown that the improvement effect of DBD on DKD is cardinally bound up with adjusting glycerophospholipid and sphingolipid metabolism, attenuating inflammation and IR. However, the active ingredients of DBD may act on VEGFA, ACE, NOS2, NOS3 and MAP3K5 to downregulate the expression of Degs2 and Cers genes, reduce the amount of pc, PEs, Cers, and SMs, thereby alleviating the symptoms of DKD, some of which involve AGE-RAGE, sphingolipids and many kinds of inflammation related signaling pathways.

A study with DBD as the treatment group and irbesartan as the control group shows that DBD can improve the renal function and blood lipid levels of DKD patients with qi deficiency and blood stasis (148). Studies have shown that DBD combined with conventional Western medicine therapy has significant therapeutic effects on patients with DKD. It can effectively reduce blood lipids and proteinuria levels, inhibit systemic inflammatory reactions, improve renal blood circulation, and defend renal function (143).

3.2.3 Tangshenkang

TSK is made of Astragalus membranaceus, Cornus officinalis, Rehmanniae Radix, Common yam rhizome, Poria cocos, Polygonatum sibiricum, Atractylodes lancea, Ligusticum sinense, Prunus mume, Hirudo nipponica whitman and other medicines. It functions in supplementing qi, nourishing yin, invigorating blood circulation and removing blood stasis. As early as 1999, Song Haixiang and others (149, 150) discovered that TSK has an improving effect on renal function indicators and control of blood lipids in DKD patients, and directly inhibits the function and secretion of IL-6 and TGF- β 1 to protect the kidneys. Animal experiments suggested that TSK can significantly improve urinary microalbumin, urinary creatinine, 24-hour urinary protein, and renal function in DKD rats and mice, also improve blood glucose and lipids levels to varying degrees (151153). Moreover, TSK can significantly alleviate the pathological changes compared to Benazepril in kidneys of DKD rats, inhibit the expansion of mesangial area and glomerular area (154), significantly downregulate TNF-α values, increase SOD levels and reduce MDA levels, which is related to its anti-inflammatory effect and the reduction of oxidative stress levels (151). Shi Xiaowei et al. (152) found that TSK reduced the expression of α-SMA, TGF-β, and CD8+ in the kidneys of DKD mice, decreased ECM deposition in kidney tissue, and alleviated the degree of renal interstitial fibrosis and glomerulosclerosis.

Subsequently, multiple clinical studies have shown that taking TSK in addition to conventional Western medicine treatment can significantly improve renal function indicators, urinary microalbumin excretion, blood lipid levels, reduce blood viscosity, and effectively postpone disease progression of DKD patients (155, 156).

Besides, Huangqi Tang has shown potential in improving lipid metabolism disorders related to DKD by adjusting the expression of lipid metabolism interrelated genes and improving lipid mass spectrometry (157, 158). Xiaoke Shen`an Decoction may affect the activation of subsequent biological pathways by acting on RAF1 and BCL-2 targets, and improve lipid metabolism and renal function in DKD patients, thereby inhibiting the progression of DKD (159, 160). However, further research is needed on the efficacy and safety of the combination with Western Medicine.

3.3 Single Chinese medicine

TCM often exerts its efficacy through compatibility, so single TCM preparations are rarely used in clinic. However, owing to the complexity of pharmacology, the exact latent mechanisms of traditional Chinese medicine preparations are difficult to distinguish. In order to avoid adverse reactions, a single Chinese medicine is more suitable for elucidating the exact mechanism of action of DKD.

3.3.1 Astragalus membranaceus

As is the dried root of the leguminous plants Astragalus Membranaceus (Fisch.) Bge. var.mongholicus (Bge.) Hsiao or Astragalus Membranaceus (Fisch.) Bge. The main active ingredient of AS is Astragaloside IV, which has a mild temperature and a sweet taste. It has the effects of tonifying qi and promoting yang, strengthening exterior and reducing sweat, inducing diuresis for removing edema, generating fluids and nourishing blood, breaking stagnation and unblocking rheumatism, supporting toxins and eliminating pus, and promoting healing and muscle growth (60). Among them, AS IV improves DKD by counteracting oxidative stress, restoring mitochondrial homeostasis (inhibiting Drp-1 and PINK1/Parkin signaling pathways), weakening ERS, adjusting calcium homeostasis, relieving inflammation, and improving EMT and endothelial function (161). Proteomic studies have shown that AS IV treatment possibly reduces lipid deposition, lipid related protein and metabolite levels in the kidneys affected by DKD, which perchance defers the occurrence and progression of DKD through adjusting HMOX1 mediated lipid metabolism disorders (162). Part AS II enhances the ability of resistance to oxidative stress through the co - regulation of Nrf2 and PINK1, has beneficial effects on autophagy, and improves podocyte injury and mitochondrial dysfunction in diabetes rats induced by STZ (163).

3.3.2 Panax Notoginseng

According to the pharmacopoeia (60), Panax Notoginseng (Burk.) F.H. Chen, a plant in the Araliaceae family, is derived from the dry root and rhizome with a warm nature, sweet taste, and slight bitterness. It is used for dispersing blood stasis, stopping bleeding, reducing swelling and relieving pain. Its main ingredients are ginsenosides Rg1, Rb1, and R1. Xue Rui et al. (164) found that total saponins of Panax notoginseng can significantly improve proteinuria, mesangial dilation, podocyte apoptosis and morphological changes in DKD rats, and some of them exert antioxidant and anti-apoptotic effects by regulating the PTEN-PDK1-Akt mTOR signaling pathway and reducing the expression of Nox4, while reducing ROS markers such as 8-OHdG and 4-hydroxy-2-nonenal. Furthermore, Shenyilan et al. (165) demonstrated that Panax notoginseng saponins Fc, a novel saponin isolated from PNG, partially improved mitochondrial dysfunction and pyroptosis in GECs by regulating the HMGCS2 pathway. Moreover, researches have shown that Panax notoginseng polysaccharides (PNPs), the main byproduct of Panax notoginseng saponin extraction, have effects such as regulating lipid metabolism disorders, reducing inflammation, enhancing the body’s immunity, anti-tumor, anti-aging, etc (166). Li Yi et al. (167) found that PNPs can significantly improve IR levels and renal function in DKD rats, diminish the expression of inflammatory factors such as IL-1β, IL-6 and TNF-α, and also regulate lipid metabolism disorders through downregulating the levels of SREBP-1c and ACCα, reducing blood fat content, ultimately achieving the effect of treating kidney damage in T2DM rats.

3.3.3 Salvia miltiorrhiza

SM is the dried root and rhizome of Salvia miltiorrhiza Bge, a plant in the family Lamiaceae. Its main components are tanshinones, which are bitter in taste and slightly cold in nature (60). It has the functions of invigorating blood circulation, removing blood stasis, relieving pain, clearing the heart and eliminating annoyance, cooling the blood and eliminating carbuncles (168). Pharmacological studies have indicated that multiple active ingredients in SM can improve lipid metabolism disorders, such as Salvianolic acid A (SalA), which can improve lipid levels in STZ induced SD rats (169). Sal B can reduce total cholesterol and triglyceride levels and regulate dyslipidemia by regulating downstream effectors of AMPK, such as PPARα and acetyl CoA carboxylase (170). Clinical studies have shown that intravenous infusion of salvianolic acid salt can lower TC, TG, LDL-C, and increase HDL-C levels in DKD patients (171). At the same time, it can effectively reduce inflammatory response and alleviate oxidative stress through reducing CRP, TNF-α, IL-6, ROS, MDA levels and increasing T-AOC, SOD, GSH Px levels (172, 173). Tanshinone IIA can inhibit pyroptosis by adjusting the thioredoxin interacting protein Txnip/NLRP3 inflammasome, thereby retarding the progression of DKD. It can also affect the distribution of high-density lipoprotein subfractions in rats, thereby regulating cholesterol metabolism and reducing lipid deposition (174). Z. Xu et al. (175) discovered that the combination of salvianolic acid B and tanshinone IIA has a synergistic anti-inflammatory function, reforming glucose and lipid metabolism abnormalities and liver and kidney damage in early DKD rats. It improves urinary and serum metabolism disorders by unsaturated fatty acid biosynthesis, glycerophospholipid metabolism, steroid biosynthesis, alanine and arachidonic acid biosynthesis, and its mechanism is possibly bound up with the PI3K/Akt/NF-κB signaling pathway. What’s more, sodium tanshinone IIA sulfonate can be obtained by sulfonation of tanshinone IIA, which is currently the only monomeric chemical drug in China prepared from the lipophilic active ingredient of SM. It can significantly improve the blood lipid levels of DKD hemodialysis patients by inhibiting the reaction of NLRP3 to suppress the splicing of caspase-1, thereby reducing the maturation and release of IL-1β, achieving anti-inflammatory, anti-oxidant, and anti-pyroptosis effects, and delaying the progression of DKD (176178).

Besides, ligustri lucidi fructus (LLF) may affect the progression of DKD through inflammatory response (179). Moreover, the steamed LLF extract exhibits a stronger protective effect on the kidneys. This is manifested in the improvement of indicators such as Scr, BUN, and 24-hour urinary protein. It also shows better control of hyperlipidemia and a reduction in the release of pro-inflammatory mediators like TNF-α, IL-6, and IL-1β in DKD rats (180). Hirudin, the main active ingredient of Hirudo nipponica Whitman (HNW), is a natural thrombin inhibitor that can improve kidney injury and inhibit inflammation through the p38/NF-κB pathway in the DKD rat model (181). HNW freeze-dried powder has a dose-dependent effect on improving renal function in DKD rats, while TC, TG, and LDL are all reduced, with no statistically significant difference in HDL (182).

4 Discussion

DKD, as a common microvascular complication of diabetes, has become the main cause of ESRD. Its pathogenesis is complex, with multiple etiologies interacting with each other. Among them, lipid metabolism disorder is an important risk factor for the occurrence and development of DKD. Therefore, it is recommended to intervene in lifestyle and adjust medication while receiving routine medication treatment, and control blood lipid levels, thus effectively retarding the progression of renal disease. Along with the modernization of TCM, many kinds of traditional Chinese patent medicines, compound Chinese medicines and single Chinese medicine preparations are constantly gaining increasing attention, and their therapeutic effects and safety have also been largely verified (Table 1). Currently, there is no specific medicine for the clinical therapy of DKD, and Western medicine has a single mode of action and many adverse reactions. Many drugs cannot be used for a long time. Numerous researches have suggested that the combined use of TCM and Western medicine often yields better results than using TCM or Western medicine alone (Table 2). In general, it is of great significance to further pinpoint the pathogenesis of DKD combined with lipid metabolism disorders and better apply integrated Traditional Chinese and Western medicine to treat DKD.

Table 1
www.frontiersin.org

Table 1. The summary of TCM.

Table 2
www.frontiersin.org

Table 2. Clinical research.

Author contributions

LS: Writing – original draft, Writing – review & editing, Visualization. ZC: Writing – original draft, Writing – review & editing, Visualization. LW: Methodology, Writing – review & editing. YX: Conceptualization, Writing – original draft, Writing – review & editing. DG: Conceptualization, Visualization, Writing – original draft, Writing – review & editing.

Funding

The author(s) declare that financial support was received for the research and/or publication of this article. This study was supported by Science and Technology Development Fund of Macau (FDCT: 0022/2024/RIB1, 0104/2024/AGJ), Science and Technology Commission of Shanghai Municipality, Shanghai Science and Technology Innovation Action Plan (23430761300) and the Special Project for the Health Sector, Pudong New Area Health Commission (PW2023E-03).

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Tang G, Li S, Zhang C, Chen H, Wang N, and Feng Y. Clinical efficacies, underlying mechanisms and molecular targets of Chinese medicines for diabetic nephropathy treatment and management. Acta Pharm Sin B. (2021) 11:2749–67. doi: 10.1016/j.apsb.2020.12.020

PubMed Abstract | Crossref Full Text | Google Scholar

2. Guo M, He F, and Zhang C. Molecular therapeutics for diabetic kidney disease: an update. Int J Mol Sci. (2024) 25(18):10051. doi: 10.3390/ijms251810051

PubMed Abstract | Crossref Full Text | Google Scholar

3. Olzmann JA and Carvalho P. Dynamics and functions of lipid droplets. Nat Rev Mol Cell Biol. (2019) 20:137–55. doi: 10.1038/s41580-018-0085-z

PubMed Abstract | Crossref Full Text | Google Scholar

4. Li MT, Liu LL, Zhou Q, Huang LX, Shi YX, Hou JB, et al. Phyllanthus niruri L. Exerts protective effects against the calcium oxalate-induced renal injury via ellgic acid. Front Pharmacol. (2022) 13:891788. doi: 10.3389/fphar.2022.891788

PubMed Abstract | Crossref Full Text | Google Scholar

5. Gai Z, Wang T, Visentin M, Kullak-Ublick GA, Fu X, and Wang Z. Lipid accumulation and chronic kidney disease. Nutrients. (2019) 11(4):722. doi: 10.3390/nu11040722

PubMed Abstract | Crossref Full Text | Google Scholar

6. Thongnak L, Pongchaidecha A, and Lungkaphin A. Renal lipid metabolism and lipotoxicity in diabetes. Am J Med Sci. (2020) 359:84–99. doi: 10.1016/j.amjms.2019.11.004

PubMed Abstract | Crossref Full Text | Google Scholar

7. Zhou Y, Liu L, Jin B, Wu Y, Xu L, Chang X, et al. Metrnl alleviates lipid accumulation by modulating mitochondrial homeostasis in diabetic nephropathy. Diabetes. (2023) 72:611–26. doi: 10.2337/db22-0680

PubMed Abstract | Crossref Full Text | Google Scholar

8. Hayashi T, Hirano T, Taira T, Tokuno A, Mori Y, Koba S, et al. Remarkable increase of apolipoprotein B48 level in diabetic patients with end-stage renal disease. Atherosclerosis. (2008) 197:154–8. doi: 10.1016/j.atherosclerosis.2007.03.015

PubMed Abstract | Crossref Full Text | Google Scholar

9. Hirano T. Abnormal lipoprotein metabolism in diabetic nephropathy. Clin Exp Nephrol. (2014) 18:206–9. doi: 10.1007/s10157-013-0880-y

PubMed Abstract | Crossref Full Text | Google Scholar

10. Mutlu AS, Duffy J, and Wang MC. Lipid metabolism and lipid signals in aging and longevity. Dev Cell. (2021) 56:1394–407. doi: 10.1016/j.devcel.2021.03.034

PubMed Abstract | Crossref Full Text | Google Scholar

11. Su J, Ye D, Gao C, Huang Q, and Gui D. Mechanism of progression of diabetic kidney disease mediated by podocyte mitochondrial injury. Mol Biol Rep. (2020) 47:8023–35. doi: 10.1007/s11033-020-05749-0

PubMed Abstract | PubMed Abstract | Crossref Full Text | Google Scholar

12. Liu X, Du H, Sun Y, and Shao L. Role of abnormal energy metabolism in the progression of chronic kidney disease and drug intervention. Ren Fail. (2022) 44:790–805. doi: 10.1080/0886022X.2022.2072743

PubMed Abstract | Crossref Full Text | Google Scholar

13. Barutta F, Bellini S, and Gruden G. Mechanisms of podocyte injury and implications for diabetic nephropathy. Clin Sci (Lond). (2022) 136:493–520. doi: 10.1042/CS20210625

PubMed Abstract | Crossref Full Text | Google Scholar

14. Zhuang L, Kim J, Adam RM, Solomon KR, and Freeman MR. Cholesterol targeting alters lipid raft composition and cell survival in prostate cancer cells and xenografts. J Clin Invest. (2005) 115:959–68. doi: 10.1172/JCI200519935

PubMed Abstract | Crossref Full Text | Google Scholar

15. Castillo-Mancho V, Atienza-Manuel A, Sarmiento-Jiménez J, Ruiz-Gómez M, and Culi J. Phospholipid scramblase 1: an essential component of the nephrocyte slit diaphragm. Cell Mol Life Sci. (2024) 81:261. doi: 10.1007/s00018-024-05287-z

PubMed Abstract | Crossref Full Text | Google Scholar

16. Mundel P and Kriz W. Structure and function of podocytes: an update. Anat Embryol (Berl). (1995) 192:385–97. doi: 10.1007/BF00240371

PubMed Abstract | Crossref Full Text | Google Scholar

17. de Vries AP, Ruggenenti P, Ruan XZ, Praga M, Cruzado JM, Bajema IM, et al. Fatty kidney: emerging role of ectopic lipid in obesity-related renal disease. Lancet Diabetes Endocrinol. (2014) 2:417–26. doi: 10.1016/S2213-8587(14)70065-8

PubMed Abstract | Crossref Full Text | Google Scholar

18. Khan S, Gaivin R, Abramovich C, Boylan M, Calles J, and Schelling JR. Fatty acid transport protein-2 regulates glycemic control and diabetic kidney disease progression. JCI Insight. (2020) 5(15):e136845. doi: 10.1172/jci.insight.136845

PubMed Abstract | Crossref Full Text | Google Scholar

19. Yokoi H and Yanagita M. Targeting the fatty acid transport protein CD36, a class B scavenger receptor, in the treatment of renal disease. Kidney Int. (2016) 89:740–2. doi: 10.1016/j.kint.2016.01.009

PubMed Abstract | Crossref Full Text | Google Scholar

20. Hua W, Huang HZ, Tan LT, Wan JM, Gui HB, Zhao L, et al. CD36 mediated fatty acid-induced podocyte apoptosis via oxidative stress. PloS One. (2015) 10:e0127507. doi: 10.1371/journal.pone.0127507

PubMed Abstract | Crossref Full Text | Google Scholar

21. Mitrofanova A, Merscher S, and Fornoni A. Kidney lipid dysmetabolism and lipid droplet accumulation in chronic kidney disease. Nat Rev Nephrol. (2023) 19:629–45. doi: 10.1038/s41581-023-00741-w

PubMed Abstract | Crossref Full Text | Google Scholar

22. Ducasa GM, Mitrofanova A, and Fornoni A. Crosstalk between lipids and mitochondria in diabetic kidney disease. Curr Diabetes Rep. (2019) 19:144. doi: 10.1007/s11892-019-1263-x

PubMed Abstract | Crossref Full Text | Google Scholar

23. Nogalska A, Sucajtys-Szulc E, and Swierczynski J. Leptin decreases lipogenic enzyme gene expression through modification of SREBP-1c gene expression in white adipose tissue of aging rats. Metabolism. (2005) 54:1041–7. doi: 10.1016/j.metabol.2005.03.007

PubMed Abstract | Crossref Full Text | Google Scholar

24. Proctor G, Jiang T, Iwahashi M, Wang Z, Li J, and Levi M. Regulation of renal fatty acid and cholesterol metabolism, inflammation, and fibrosis in Akita and OVE26 mice with type 1 diabetes. Diabetes. (2006) 55:2502–9. doi: 10.2337/db05-0603

PubMed Abstract | Crossref Full Text | Google Scholar

25. Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schütz G, Umesono K, et al. The nuclear receptor superfamily: the second decade. Cell. (1995) 83:835–9. doi: 10.1016/0092-8674(95)90199-X

PubMed Abstract | Crossref Full Text | Google Scholar

26. Desvergne B and Wahli W. Peroxisome proliferator-activated receptors: nuclear control of metabolism. Endocr Rev. (1999) 20:649–88. doi: 10.1210/edrv.20.5.0380

PubMed Abstract | Crossref Full Text | Google Scholar

27. Wang Y, Liu T, Wu Y, Wang L, Ding S, Hou B, et al. Lipid homeostasis in diabetic kidney disease. Int J Biol Sci. (2024) 20:3710–24. doi: 10.7150/ijbs.95216

PubMed Abstract | Crossref Full Text | Google Scholar

28. Qiu YY, Zhang J, Zeng FY, and Zhu YZ. Roles of the peroxisome proliferator-activated receptors (PPARs) in the pathogenesis of nonalcoholic fatty liver disease (NAFLD). Pharmacol Res. (2023) 192:106786. doi: 10.1016/j.phrs.2023.106786

PubMed Abstract | Crossref Full Text | Google Scholar

29. Huang CC, Chou CA, Chen WY, Yang JL, Lee WC, Chen JB, et al. Empagliflozin ameliorates free fatty acid induced-lipotoxicity in renal proximal tubular cells via the PPARγ/CD36 pathway in obese mice. Int J Mol Sci. (2021) 22(22):12408. doi: 10.3390/ijms222212408

PubMed Abstract | Crossref Full Text | Google Scholar

30. Bhargava P and Schnellmann RG. Mitochondrial energetics in the kidney. Nat Rev Nephrol. (2017) 13:629–46. doi: 10.1038/nrneph.2017.107

PubMed Abstract | Crossref Full Text | Google Scholar

31. Qin X, Jiang M, Zhao Y, Gong J, Su H, Yuan F, et al. Berberine protects against diabetic kidney disease via promoting PGC-1α-regulated mitochondrial energy homeostasis. Br J Pharmacol. (2020) 177:3646–61. doi: 10.1111/bph.14935

PubMed Abstract | Crossref Full Text | Google Scholar

32. Zhang Y, Castellani LW, Sinal CJ, Gonzalez FJ, and Edwards PA. Peroxisome proliferator-activated receptor-gamma coactivator 1alpha (PGC-1alpha) regulates triglyceride metabolism by activation of the nuclear receptor FXR. Genes Dev. (2004) 18:157–69. doi: 10.1101/gad.1138104

PubMed Abstract | Crossref Full Text | Google Scholar

33. Dobrzyn A and Ntambi JM. The role of stearoyl-CoA desaturase in body weight regulation. Trends Cardiovasc Med. (2004) 14:77–81. doi: 10.1016/j.tcm.2003.12.005

PubMed Abstract | Crossref Full Text | Google Scholar

34. Iwai T, Kume S, Chin-Kanasaki M, Kuwagata S, Araki H, Takeda N, et al. Stearoyl-coA desaturase-1 protects cells against lipotoxicity-mediated apoptosis in proximal tubular cells. Int J Mol Sci. (2016) 17(11):1868. doi: 10.3390/ijms17111868

PubMed Abstract | Crossref Full Text | Google Scholar

35. von Eckardstein A, Nordestgaard BG, Remaley AT, and Catapano AL. High-density lipoprotein revisited: biological functions and clinical relevance. Eur Heart J. (2023) 44:1394–407. doi: 10.1093/eurheartj/ehac605

PubMed Abstract | Crossref Full Text | Google Scholar

36. Wong NKP, Nicholls SJ, Tan JTM, and Bursill CA. The role of high-density lipoproteins in diabetes and its vascular complications. Int J Mol Sci. (2018) 19(6):1680. doi: 10.3390/ijms19061680

PubMed Abstract | Crossref Full Text | Google Scholar

37. Chen Y, Chen M, Zhu W, Zhang Y, Liu P, and Li P. Morroniside attenuates podocytes lipid deposition in diabetic nephropathy: A network pharmacology, molecular docking and experimental validation study. Int Immunopharmacol. (2024) 138:112560. doi: 10.1016/j.intimp.2024.112560

PubMed Abstract | Crossref Full Text | Google Scholar

38. Pedigo CE, Ducasa GM, Leclercq F, Sloan A, Mitrofanova A, Hashmi T, et al. Local TNF causes NFATc1-dependent cholesterol-mediated podocyte injury. J Clin Invest. (2016) 126:3336–50. doi: 10.1172/JCI85939

PubMed Abstract | Crossref Full Text | Google Scholar

39. Haas ME, Levenson AE, Sun X, Liao WH, Rutkowski JM, de Ferranti SD, et al. The role of proprotein convertase subtilisin/kexin type 9 in nephrotic syndrome-associated hypercholesterolemia. Circulation. (2016) 134:61–72. doi: 10.1161/CIRCULATIONAHA.115.020912

PubMed Abstract | Crossref Full Text | Google Scholar

40. Byun JH, Lebeau PF, Platko K, Carlisle RE, Faiyaz M, Chen J, et al. Inhibitory antibodies against PCSK9 reduce surface CD36 and mitigate diet-induced renal lipotoxicity. Kidney360. (2022) 3:1394–410. doi: 10.34067/KID.0007022021

PubMed Abstract | Crossref Full Text | Google Scholar

41. Kaseda R, Tsuchida Y, Yang HC, Yancey PG, Zhong J, Tao H, et al. Chronic kidney disease alters lipid trafficking and inflammatory responses in macrophages: effects of liver X receptor agonism. BMC Nephrol. (2018) 19:17. doi: 10.1186/s12882-018-0814-8

PubMed Abstract | Crossref Full Text | Google Scholar

42. Hirano T. Pathophysiology of diabetic dyslipidemia. J Atheroscler Thromb. (2018) 25:771–82. doi: 10.5551/jat.RV17023

PubMed Abstract | Crossref Full Text | Google Scholar

43. Jiang W, Gan C, Zhou X, Yang Q, Chen D, Xiao H, et al. Klotho inhibits renal ox-LDL deposition via IGF-1R/RAC1/OLR1 signaling to ameliorate podocyte injury in diabetic kidney disease. Cardiovasc Diabetol. (2023) 22:293. doi: 10.1186/s12933-023-02025-w

PubMed Abstract | Crossref Full Text | Google Scholar

44. Zhou W and Anakk S. Enterohepatic and non-canonical roles of farnesoid X receptor in controlling lipid and glucose metabolism. Mol Cell Endocrinol. (2022) 549:111616. doi: 10.1016/j.mce.2022.111616

PubMed Abstract | Crossref Full Text | Google Scholar

45. Kim DH, Choi HI, Park JS, Kim CS, Bae EH, Ma SK, et al. Src-mediated crosstalk between FXR and YAP protects against renal fibrosis. FASEB J. (2019) 33:11109–22. doi: 10.1096/fj.201900325R

PubMed Abstract | Crossref Full Text | Google Scholar

46. Kim DH, Park JS, Choi HI, Kim CS, Bae EH, Ma SK, et al. The role of the farnesoid X receptor in kidney health and disease: a potential therapeutic target in kidney diseases. Exp Mol Med. (2023) 55:304–12. doi: 10.1038/s12276-023-00932-2

PubMed Abstract | Crossref Full Text | Google Scholar

47. Zhao K, He J, Zhang Y, Xu Z, Xiong H, Gong R, et al. Activation of FXR protects against renal fibrosis via suppressing Smad3 expression. Sci Rep. (2016) 6:37234. doi: 10.1038/srep37234

PubMed Abstract | Crossref Full Text | Google Scholar

48. Zhou B, Feng B, Qin Z, Zhao Y, Chen Y, Shi Z, et al. Activation of farnesoid X receptor downregulates visfatin and attenuates diabetic nephropathy. Mol Cell Endocrinol. (2016) 419:72–82. doi: 10.1016/j.mce.2015.10.001

PubMed Abstract | Crossref Full Text | Google Scholar

49. Schelling JR. The contribution of lipotoxicity to diabetic kidney disease. Cells. (2022) 11(20):3236. doi: 10.3390/cells11203236

PubMed Abstract | Crossref Full Text | Google Scholar

50. Tang C, Deng X, Qu J, Miao Y, Tian L, Zhang M, et al. Fenofibrate attenuates renal tubular cell apoptosis by up-regulating MCAD in diabetic kidney disease. Drug Des Devel Ther. (2023) 17:1503–14. doi: 10.2147/DDDT.S405266

PubMed Abstract | Crossref Full Text | Google Scholar

51. Adhyaru BB and Jacobson TA. Safety and efficacy of statin therapy. Nat Rev Cardiol. (2018) 15:757–69. doi: 10.1038/s41569-018-0098-5

PubMed Abstract | Crossref Full Text | Google Scholar

52. Fried LF, Orchard TJ, and Kasiske BL. Effect of lipid reduction on the progression of renal disease: a meta-analysis. Kidney Int. (2001) 59:260–9. doi: 10.1046/j.1523-1755.2001.00487.x

PubMed Abstract | Crossref Full Text | Google Scholar

53. Qin X, Dong H, Fang K, and Lu F. The effect of statins on renal outcomes in patients with diabetic kidney disease: A systematic review and meta-analysis. Diabetes Metab Res Rev. (2017) 33(6). doi: 10.1002/dmrr.2901

PubMed Abstract | Crossref Full Text | Google Scholar

54. Takata T and Isomoto H. Pleiotropic effects of sodium-glucose cotransporter-2 inhibitors: renoprotective mechanisms beyond glycemic control. Int J Mol Sci. (2021) 22(9):4374. doi: 10.3390/ijms22094374

PubMed Abstract | Crossref Full Text | Google Scholar

55. Su K, Yi B, Yao BQ, Xia T, Yang YF, Zhang ZH, et al. Liraglutide attenuates renal tubular ectopic lipid deposition in rats with diabetic nephropathy by inhibiting lipid synthesis and promoting lipolysis. Pharmacol Res. (2020) 156:104778. doi: 10.1016/j.phrs.2020.104778

PubMed Abstract | Crossref Full Text | Google Scholar

56. Naaman SC and Bakris GL. Diabetic nephropathy: update on pillars of therapy slowing progression. Diabetes Care. (2023) 46:1574–86. doi: 10.2337/dci23-0030

PubMed Abstract | Crossref Full Text | Google Scholar

57. Mazzieri A, Porcellati F, Timio F, and Reboldi G. Molecular targets of novel therapeutics for diabetic kidney disease: A new era of nephroprotection. Int J Mol Sci. (2024) 25(7):3969. doi: 10.3390/ijms25073969

PubMed Abstract | Crossref Full Text | Google Scholar

58. Malek V, Suryavanshi SV, Sharma N, Kulkarni YA, Mulay SR, and Gaikwad AB. Potential of renin-angiotensin-aldosterone system modulations in diabetic kidney disease: old players to new hope! Rev Physiol Biochem Pharmacol. (2021) 179:31–71. doi: 10.1007/112_2020_50

PubMed Abstract | Crossref Full Text | Google Scholar

59. Yangyang Z and Tongning Y. Research progress of traditional chinese medicine prevention and treatment of diabetic nephropathy based on lipid metabolism. Med Innovation China. (2024) 21:154–9. doi: 10.3969/j.issn.1674-4985.2024.35.035

Crossref Full Text | Google Scholar

60. Commission CP. Pharmacopoeia of the people’s republic of China 2020. Beijing: China Medical Science Press (2020).

Google Scholar

61. Sheng X, Dong Y, Cheng D, Wang N, and Guo Y. Efficacy and safety of Bailing capsules in the treatment of type 2 diabetic nephropathy: a meta-analysis. Ann Palliat Med. (2020) 9:3885–98. doi: 10.21037/apm-20-1799

PubMed Abstract | Crossref Full Text | Google Scholar

62. Shu X, Xu D, Qu Y, Shang X, Qiao K, Feng C, et al. Efficacy and safety of Cordyceps sinensis (Hirsutella sinensis, Cs-C-Q80) in chronic bronchitis. Front Pharmacol. (2024) 15:1428216. doi: 10.3389/fphar.2024.1428216

PubMed Abstract | Crossref Full Text | Google Scholar

63. Yuli T and Masichen. Effect of bailing capsule on inflammatory factors in patients with early diabetes nephropathy. Guangming J Chin Med. (2021) 36:1446–9. doi: 10.3969/j.issn.1003-8914.2021.09.029

Crossref Full Text | Google Scholar

64. Xiaoting G and Yanning W. Effects of Bailing capsule combined with sulodexide on vascular endothelial function and oxidative stress function in elderly patients with diabetes nephropathy. Drug Eval Res. (2020) 43:1864–7. doi: 10.7501/j.issn.1674-6376.2020.09.036

Crossref Full Text | Google Scholar

65. Wenru W, Xuming Z, Jixin L, Jiayi Y, Renhuan Y, and Yanming X. Network Meta-analysis of Qi-supplementing and Yin-nourishing Chinese patent medicines in treatment of early diabetic nephropathy. China J Chin Materia Medica. (2023) 48:3949–64. doi: 10.19540/j.cnki.cjcmm.20230314.501

PubMed Abstract | Crossref Full Text | Google Scholar

66. Zhang Q, Xiao X, Li M, Yu M, and Ping F. Bailing capsule (Cordyceps sinensis) ameliorates renal triglyceride accumulation through the PPARα pathway in diabetic rats. Front Pharmacol. (2022) 13:915592. doi: 10.3389/fphar.2022.915592

PubMed Abstract | Crossref Full Text | Google Scholar

67. Xubo J. Clinical analysis of Bailing capsule combined with valsartan capsule in the treatment of 50 cases of early diabetes nephropathy. J New Chin Med. (2016) 48:99–101. doi: 10.13457/j.cnki.jncm.2016.08.042

Crossref Full Text | Google Scholar

68. Jiayuan H and Canxin Z. Effect of bailing capsule and losartan on UMA and lipid metabolism in patients with early diabetes nephropathy. Chin J Biochem Pharmaceutics. (2016) 36:144–6. doi: 10.3969/j.issn.1005-1678.2016.12.041

Crossref Full Text | Google Scholar

69. Xiaojun H, Ping G, and Xiaofei X. Clinical efficacy of Bailing capsule combined with irbesartan in the treatment of elderly patients with diabetes nephropathy. Chin J Clin Rational Drug Use. (2023) 16:91–4. doi: 10.15887/j.cnki.13-1389/r.2023.21.026

Crossref Full Text | Google Scholar

70. Chaohui F and Huiping H. The therapeutic effect of dagelin combined with bailing capsule on diabetes nephropathy and its influence on renal function. Diabetes New World. (2022) 25:17–20. doi: 10.16658/j.cnki.1672-4062.2022.16.017

Crossref Full Text | Google Scholar

71. Haifang N. Clinical Observation of Bailing Capsule Combined with Pancreatic Kallidinogenase in the Treatment of Early onset Type 2 diabetes Nephropathy Phase III. J Med Forum. (2020) 41:94–6 + 100.

Google Scholar

72. Ruichun H, Guoqing X, Yanfang R, Ruiying Z, Jingxin, Haibo T, et al. Bailing capsule combined with simvastatin in the treatment of diabetes nephropathy. Chin J Clin Res. (2018) 31:1109–13. doi: 10.13429/j.cnki.cjcr.2018.08.026

Crossref Full Text | Google Scholar

73. Juan Z, Qiaoling O, and Qing L. Clinical observation on 57 cases of diabetes nephropathy with carotid atherosclerosis treated by integrated traditional chinese and western medicine. Chin J Ethnomedicine Ethnopharmacy. (2020) 29:84–7.

Google Scholar

74. Wang X, He Q, Chen Q, Xue B, Wang J, Wang T, et al. Network pharmacology combined with metabolomics to study the mechanism of Shenyan Kangfu Tablets in the treatment of diabetic nephropathy. J Ethnopharmacol. (2021) 270:113817. doi: 10.1016/j.jep.2021.113817

PubMed Abstract | Crossref Full Text | Google Scholar

75. Chen Q, Ren D, Wu J, Yu H, Chen X, Wang J, et al. Shenyan Kangfu tablet alleviates diabetic kidney disease through attenuating inflammation and modulating the gut microbiota. J Nat Med. (2021) 75:84–98. doi: 10.1007/s11418-020-01452-3

PubMed Abstract | Crossref Full Text | Google Scholar

76. Wenyu Z, Wenxiu C, and Wins HY. Clinical observation of Shenyan Kangfu Table combined with reduced glutathione in the treatment of diabetes nephropathy. Drugs Clinic. (2016) 31:683–6. doi: 10.7501/j.issn.1674-5515.2016.05.027

Crossref Full Text | Google Scholar

77. Yanna C and Xiaoli H. Clinical observation of Shenyan Kangfu Table combined with alprostadil in the treatment of early diabetes nephropathy. Modern J Integrated Traditional Chin Western Med. (2018) 27:1763–5. doi: 10.3969/j.issn.1008-8849.2018.16

Crossref Full Text | Google Scholar

78. Yin Y, Wang Y, Liu Y, Wang F, and Wang Z. Evidence construction of Jinshuibao capsules against sTabble chronic obstructive pulmonary disease: A systematic review and network pharmacology. Heliyon. (2024) 10:e34572. doi: 10.1016/j.heliyon.2024.e34572

PubMed Abstract | Crossref Full Text | Google Scholar

79. Jing L, Weixuan D, Yan G, Kenn, Zhe L, and Jiao Y. Clinical study of Jinshuibao capsule combined with sulodide in the treatment of early diabetes nephropathy. Drugs Clinic. (2019) 34:1483–7. doi: 10.7501/j.issn.1674-5515.2019.05.047

Crossref Full Text | Google Scholar

80. Tao J. Clinical observation of Jinshuibao capsule combined with ramipril in the treatment of early diabetes nephropathy. J Pract Traditional Chin Internal Med. (2021) 35:39–42. doi: 10.13729/j.issn.1671-7813.Z20201413

Crossref Full Text | Google Scholar

81. Zhang Y, Xu L, Lu Y, Zhang J, Yang M, Tian Y, et al. Protective effect of Cordyceps sinensis against diabetic kidney disease through promoting proliferation and inhibiting apoptosis of renal proximal tubular cells. BMC Complement Med Ther. (2023) 23:109. doi: 10.1186/s12906-023-03901-4

PubMed Abstract | Crossref Full Text | Google Scholar

82. Zhenhua W, Lixia Y, and Xiangyun M. Study on the mechanism of astragalus polysaccharide intervention on inflammation in diabetes nephropathy. Traditional Chin Medicinal Res. (2020) 33:1–5. doi: 10.3969/j.issn.1001-6910.2020.08.01

Crossref Full Text | Google Scholar

83. Juan P and Shuangyan S. Therapeutic effect of telmisartan combined with Jinshuibao capsule on diabetes nephropathy. Chin J Geriatric Care. (2016) 14:40–1. doi: 10.3969/j.issn.1672-4860.2016.01.018

Crossref Full Text | Google Scholar

84. Aiyuan W, Ting Z, Weiping W, Wenjun T, Yu W, and Xiaodong H. Effect of Jinshuibao capsule combined with candesartan kamedoxomil Tables on early diabetes nephropathy. Contemp Med Forum. (2020) 18:58–9. doi: 10.3969/j.issn.2095-7629.2020.12.034

Crossref Full Text | Google Scholar

85. Xufeng Y and Niansong W. Research progress on the mechanism of sulodexide in the treatment of diabetes nephropathy. Chin J Integrated Traditional Western Nephrology. (2011) 12:365–7. doi: 10.3969/j.issn.2095-7629.2020.12.034

Crossref Full Text | Google Scholar

86. Guo JM, Lu YW, Shang EX, Li T, Liu Y, Duan JA, et al. Metabolite identification strategy of non-targeted metabolomics and its application for the identification of components in Chinese multicomponent medicine Abelmoschus manihot L. Phytomedicine. (2015) 22:579–87. doi: 10.1016/j.phymed.2015.02.002

PubMed Abstract | Crossref Full Text | Google Scholar

87. Diao Z, Yu H, Wu Y, Sun Y, Tang H, Wang M, et al. Identification of the main flavonoids of Abelmoschus manihot (L.) medik and their metabolites in the treatment of diabetic nephropathy. Front Pharmacol. (2023) 14:1290868. doi: 10.3389/fphar.2023.1290868

PubMed Abstract | Crossref Full Text | Google Scholar

88. Ziyuan J, Wei L, Tianlin Z, Cuiting G, Haijing S, and Weiting Z. Clinical efficacy analysis of huangkui capsule and dapagliflozin in the treatment of diabetes nephropathy. China Pract Med. (2024) 19:19–23. doi: 10.14163/j.cnki.11-5547/r.2024.15.005

Crossref Full Text | Google Scholar

89. Shan C, Jiangping H, and Xinyu S. Effect of huangkui capsule on blood lipid components in diabetes nephropathy. Chin J Exp Traditional Med Formulae. (2011) 17:229–30. doi: 10.3969/j.issn.1005-9903.2011.05.070

Crossref Full Text | Google Scholar

90. Disheng H and Yi C. Study on the clinical effect and related mechanism of huangkui capsule in the treatment of diabetes nephropathy. Modern J Integrated Traditional Chin Western Med. (2024) 33:941–4. doi: 10.3969/j.issn.1008-8849.2024.07.013

Crossref Full Text | Google Scholar

91. Dafeng L and Mingxia Z. Effect of huangkui capsule on blood glucose, blood lipid and renal function in patients with diabetes nephropathy. J Chin Medicinal Materials. (2014) 37:912–4. doi: 10.13863/j.issn1001-4454.2014.05.052

Crossref Full Text | Google Scholar

92. Jianhua F, Jianming Y, Yi Z, and Jinglong M. Effect of huangkui capsules on clinical diabetes nephropathy and its mechanism. World Chin Med. (2016) 11:1693–6. doi: 10.3969/j.issn.1673-7202.2016.09.011

Crossref Full Text | Google Scholar

93. Daoqi S, Defang H, Jun X, Anxing Z, and Dan Y. Application of Huangkui capsule combined with reduced glutathione in diabetes nephropathy. World Chin Med. (2018) 13:834–7. doi: 10.3969/j.issn.1673-7202.2018.04.012

Crossref Full Text | Google Scholar

94. Xiangyue W. Huangkui capsule combined with captopril and compound α - ketoacid Tables in the treatment of diabetes nephropathy. Henan Med Res. (2020) 29:1445–6. doi: 10.3969/j.issn.1004-437X.2020.08.051

Crossref Full Text | Google Scholar

95. Ali Z. Clinical effect of valsartan combined with Huangkui capsule in the treatment of diabetes nephropathy and its influence on inflammatory reaction, renal function and blood lipid level. Clin Res Practice. (2021) 6:137–9. doi: 10.19347/j.cnki.2096-1413.202123045

Crossref Full Text | Google Scholar

96. Xiaojun X, Qiaoling K, Yuping W, Xiangfeng Z, and Linxiang S. Clinical effect of Huangkui capsule combined with simvastatin on diabetes nephropathy and its influence on cytokines and biochemical indicators. Modern Chin Doctor. (2021) 59:101–5.

Google Scholar

97. Xiaohua H, Jianxun R, Yanghui W, Dragon G, Bin Y, Xiaoxia D, et al. Qizhu granule inhibits early proteinuria in rats with syndrome of qi yin deficiency and phlegm turbidity blocking collaterals in diabetes nephropathy. World Sci Technology-Modernization Traditional Chin Med. (2015) 17:1392–7. doi: 10.11842/wst.2015.07.011

Crossref Full Text | Google Scholar

98. Wu W, Hu W, Han WB, Liu YL, Tu Y, Yang HM, et al. Inhibition of akt/mTOR/p70S6K signaling activity with huangkui capsule alleviates the early glomerular pathological changes in diabetic nephropathy. Front Pharmacol. (2018) 9:443. doi: 10.3389/fphar.2018.00443

PubMed Abstract | Crossref Full Text | Google Scholar

99. Ge J, Miao JJ, Sun XY, and Yu JY. Huangkui capsule, an extract from Abelmoschus manihot (L.) medic, improves diabetic nephropathy via activating peroxisome proliferator-activated receptor (PPAR)-α/γ and attenuating endoplasmic reticulum stress in rats. J Ethnopharmacol. (2016) 189:238–49. doi: 10.1016/j.jep.2016.05.033

PubMed Abstract | Crossref Full Text | Google Scholar

100. Camino AM, Atorrasagasti C, Maccio D, Prada F, Salvatierra E, Rizzo M, et al. Adenovirus-mediated inhibition of SPARC attenuates liver fibrosis in rats. J Gene Med. (2008) 10:993–1004. doi: 10.1002/jgm.1228

PubMed Abstract | Crossref Full Text | Google Scholar

101. Kos K, Wong S, Tan B, Gummesson A, Jernas M, Franck N, et al. Regulation of the fibrosis and angiogenesis promoter SPARC/osteonectin in human adipose tissue by weight change, leptin, insulin, and glucose. Diabetes. (2009) 58:1780–8. doi: 10.2337/db09-0211

PubMed Abstract | Crossref Full Text | Google Scholar

102. Mao ZM, Shen SM, Wan YG, Sun W, Chen HL, Huang MM, et al. Huangkui capsule attenuates renal fibrosis in diabetic nephropathy rats through regulating oxidative stress and p38MAPK/Akt pathways, compared to α-lipoic acid. J Ethnopharmacol. (2015) 173:256–65. doi: 10.1016/j.jep.2015.07.036

PubMed Abstract | Crossref Full Text | Google Scholar

103. Yang X, Luo M, Jiang Q, and Wang Y. Effects of huangkui capsule on the expression of SPARC in the kidney tissue of a rat model with diabetic nephropathy. Curr Gene Ther. (2019) 19:211–5. doi: 10.2174/1566523219666190925112249

PubMed Abstract | Crossref Full Text | Google Scholar

104. Gu LY, Yun S, Tang HT, and Xu ZX. Huangkui capsule in combination with metformin ameliorates diabetic nephropathy via the Klotho/TGF-β1/p38MAPK signaling pathway. J Ethnopharmacol. (2021) 281:113548. doi: 10.1016/j.jep.2020.113548

PubMed Abstract | Crossref Full Text | Google Scholar

105. Han W, Ma Q, Liu Y, Wu W, Tu Y, Huang L, et al. Huangkui capsule alleviates renal tubular epithelial-mesenchymal transition in diabetic nephropathy via inhibiting NLRP3 inflammasome activation and TLR4/NF-κB signaling. Phytomedicine. (2019) 57:203–14. doi: 10.1016/j.phymed.2018.12.021

PubMed Abstract | Crossref Full Text | Google Scholar

106. Yu H, Tang H, Wang M, Xu Q, Yu J, Ge H, et al. Effects of total flavones of Abelmoschus manihot (L.) on the treatment of diabetic nephropathy via the activation of solute carriers in renal tubular epithelial cells. BioMed Pharmacother. (2023) 169:115899. doi: 10.1016/j.biopha.2023.115899

PubMed Abstract | Crossref Full Text | Google Scholar

107. Miaomiao Z, Qing Z, Bing L, Wensheng Z, Peidong Z, and Xiaohan Z. Meta-analysis and sequential analysis of randomized controlled trial of Huangkui capsule combined with ACEI/ARB drugs in the treatment of early diabetes nephropathy. Eval Anal Drug-Use Hospitals China. (2023) 23:329–38. doi: 10.14009/j.issn.1672-2124.2023.03.017

Crossref Full Text | Google Scholar

108. Yunling G, Zhaocheng D, Ying W, Pingna Z, Jingyi T, Ping L, et al. Efficacy of huangkui capsules in the treatment of diabetic kidney disease: A systematic review and using network pharmacology. Integr Med Nephrol Andrology. (2023) 10:e00020. doi: 10.1097/IMNA-D-22-00020

Crossref Full Text | Google Scholar

109. Zhen S, Tao C, Shan A, Xiangyu L, Ranran G, Wei L, et al. Analysis of the mechanism of Yishen Huashi Granule in improving diabetes nephropathy based on metabonomics and transcriptomics. Chin J Exp Traditional Med Formulae. (2023) 29:109–17. doi: 10.13422/j.cnki.syfjx.20230565

Crossref Full Text | Google Scholar

110. Zhang S, Lu M, Shang W, Du H, Wang C, Wen Z, et al. Network pharmacology, molecular docking, and experimental verification reveal the mechanism of Yi-Shen-Hua-Shi granules treating acute kidney injury. J Ethnopharmacol. (2025) 343:119320. doi: 10.1016/j.jep.2025.119320

PubMed Abstract | Crossref Full Text | Google Scholar

111. Xu R, Zhang J, Hu X, Xu P, Huang S, Cui S, et al. Yi-shen-hua-shi granules modulate immune and inflammatory damage via the ALG3/PPARγ/NF-κB pathway in the treatment of immunoglobulin a nephropathy. J Ethnopharmacol. (2024) 319:117204. doi: 10.1016/j.jep.2023.117204

PubMed Abstract | Crossref Full Text | Google Scholar

112. Cuilan L, Wen Y, Shengjun L, Linlin W, Yanchun C, Xing P, et al. Clinical effect of Yishen Huashi Granule combined with calcitriol on patients with diabetes nephropathy. Chin Traditional Patent Med. (2023) 45:86–9. doi: 10.3969/j.issn.1001-1528.2023.01.016

Crossref Full Text | Google Scholar

113. Wang G, Si Q, Yang S, Jiao T, Zhu H, Tian P, et al. Lactic acid bacteria reduce diabetes symptoms in mice by alleviating gut microbiota dysbiosis and inflammation in different manners. Food Funct. (2020) 11:5898–914. doi: 10.1039/C9FO02761K

PubMed Abstract | Crossref Full Text | Google Scholar

114. Tao C, Cong H, Zhen S, Le Z, Xiangyu L, Shan A, et al. Mechanism of Yishen Huashi Granule in Improving diabetes Nephropathy Based on the Theory of "Intestine kidney Axis. Chin J Integrated Traditional Western Nephrol. (2022) 23:384–7+471-2.

Google Scholar

115. Han C, Shen Z, Cui T, Ai SS, Gao RR, Liu Y, et al. Yi-Shen-Hua-Shi granule ameliorates diabetic kidney disease by the "gut-kidney axis. J Ethnopharmacol. (2023) 307:116257. doi: 10.1016/j.jep.2023.116257

PubMed Abstract | Crossref Full Text | Google Scholar

116. Hongbo W, Yongli Z, Shuan Z, Weiping C, Yulin C, and Ling L. Effect of yishen huashi granule on renal function of type 2 diabetes nephropathy rats. Chin J Integrated Traditional Western Nephrol. (2022) 23:130–2+92. doi: 10.3969/j.issn.1009-587X.2022.02.009

Crossref Full Text | Google Scholar

117. Guohua L, Yudan L, Zhisheng D, Xue C, and Zhiwen Z. Treatment of 30 cases of diabetes nephropathy with Yishen Huashi Granule and Irbesartan. J Jiangxi Univ Traditional Chin Med. (2013) 25:25–7.

Google Scholar

118. Guosheng Z, Xiaojing H, and Guangling Z. Effect of yishen huashi granule and shuluodite soft capsule on diabetes nephropathy. Chin J Gerontology. (2014) 34:6910–2.

Google Scholar

119. Yanna C, Jing P, Anzhi, Yongbo H, and Xiaoli H. Effect of Qianliedil combined with Yishen Huashi Granule on blood glucose, blood lipid, renal function and urinary podocyte related protein in patients with diabetes nephropathy. Prog Modern Biomedicine. (2017) 17:4714–8. doi: 10.13241/j.cnki.pmb.2017.24.028

Crossref Full Text | Google Scholar

120. Yuyue H. Effect of sulodete combined with yishen huashi granule on oxidative stress and endothelial function in early senile diabetes nephropathy. Drug Evaluation. (2018) 15:28–30. doi: 10.3969/j.issn.1672-2809.2018.07.008

Crossref Full Text | Google Scholar

121. Tao C. Effect of Yishen Huashi Granule combined with Shulodide on early senile diabetes nephropathy and its influence on oxidative stress and endothelial function. Modern J Integrated Traditional Chin Western Med. (2018) 27:3495–8. doi: 10.3969/j.issn.1008-8849.2018.31

Crossref Full Text | Google Scholar

122. Yi F, Bangming C, Xin L, Lin W, Zhaodong W, Yi F, et al. Effect of yishen huashi granule and tripterygium wilfordii polysaccharide tables on diabetes nephropathy. Chin Traditional Herbal Drugs. (2020) 51:6045–9. doi: 10.7501/j.issn.0253-2670.2020.23.017

Crossref Full Text | Google Scholar

123. Yi F, Ying Z, Bangming C, Xin L, Liqin W, Jing S, et al. Effect of yishen huashi granule and ligustrazine hydrochloride on blood lipids and renal function in patients with diabetes nephropathy. Lishizhen Med Materia Med Res. (2021) 32:911–3. doi: 10.3969/j.issn.1008-0805.2021.04.40

Crossref Full Text | Google Scholar

124. Sitong L, Zhiyuan G, Yue Z, Security CP, Shuai Z, Yan W, et al. Effects and mechanisms of Shenmai Injection in regulating copper death in myocardial fibrosis in rats. China J Chin Materia Med. (2024) 50(6):1601–9. doi: 10.19540/j.cnki.cjcmm.20240930.401

PubMed Abstract | Crossref Full Text | Google Scholar

125. He Y, Hu C, Liu S, Xu M, Liang G, Du D, et al. Anti-inflammatory effects and molecular mechanisms of shenmai injection in treating acute pancreatitis: network pharmacology analysis and experimental verification. Drug Des Devel Ther. (2022) 16:2479–95. doi: 10.2147/DDDT.S364352

PubMed Abstract | Crossref Full Text | Google Scholar

126. Wu J, Li Z, Dong X, Liu J, and Wang L. Shenmai Injection enhances short-term outcomes in ischemic stroke patients after thrombolysis via AMPKα1. Front Pharmacol. (2025) 16:1552493. doi: 10.3389/fphar.2025.1552493

PubMed Abstract | PubMed Abstract | Crossref Full Text | Google Scholar

127. Yang Q, Qin J, Li Z, He Y, and Zhou Y. Efficacy and safety of Shenmai injection in the treatment of viral myocarditis: a systematic review and meta-analysis. Front Pharmacol. (2024) 15:1453946. doi: 10.3389/fphar.2024.1453946

PubMed Abstract | Crossref Full Text | Google Scholar

128. Qiufang C. Shenmai injection combined with alprostadil treatment QXXY diabetic nephropathy of randomized controlled study. J Pract Traditional Chin Internal Med. (2016) 30:64–6. doi: 10.13729/j.issn.1671-7813.2016.11.26

Crossref Full Text | Google Scholar

129. Yanli W, Menghan S, and Jianhua D. Qiyao xiaoke capsule in the treatment of 41 cases of early diabetes nephropathy. Modern Traditional Chin Med. (2013) 33:32–4. doi: 10.13424/j.cnki.mtcm.2013.04.014

Crossref Full Text | Google Scholar

130. Mountain J, Zhengxiang Z, Baoli L, Xin Z, Jing C, Long Z, et al. Effect of soft capsules of compound oil of jujube, arboruitae and gardeniaon type 2 diabetes nephropathy rats. Chin J Gerontology. (2019) 39:3269–73. doi: 10.3969/j.issn.1005-9202.2019.13.057

Crossref Full Text | Google Scholar

131. Ze W, Qiuhong W, Xiaowen L, Fenghui S, and Lan L. The research progress of 'Liu wei di huang wan' Treatment to diabetic nephropathy. Jiangsu J Traditional Chin Med. (2019) 51:86–9. doi: 10.3969/j.issn.1672-397X.2019.01.031

Crossref Full Text | Google Scholar

132. Yan Z, Zhihai F, Rui C, Xianhui D, and Li L. A systematic review on the Keluoxin capsules plus conventional Western medicine in the treatment of diabetic kidney disease. Clin J Chin Med. (2022) 14:130–6. doi: 10.3969/j.issn.1674-7860.2022.26.040

Crossref Full Text | Google Scholar

133. Ruiting L, Cong M, and Ruiqian L. Efficacy analysis of irbesartan combined with compound Danshen dropping pills on diabetic nephropathy. Chin J Modern Drug Application. (2023) 17:136–8. doi: 10.14164/j.cnki.cn11-5581/r.2023.07.041

Crossref Full Text | Google Scholar

134. Liucui W. Effect, safety analysis and mechanism study of Shenqi Jiangtang Granule combined with Irbesartan Tables in the treatment of early diabetes nephropathy. Strait Pharm J. (2016) 28:108–10. doi: 10.3969/j.issn.1006-3765.2016.10.049

Crossref Full Text | Google Scholar

135. Xianyu T, Liu H, Jiali H, Yu Y, Lu S, and Peng Z. Clinical effect of tian qi tang capsule combined with western medicine in treatment of diabetic nephropathy and its effect on blood lipid metabolism. J Hubei Coll Traditional Chin Med. (2016) 18:63–5.

Google Scholar

136. Guangming H. Effect of Tianqi Jiangtang Capsule on early diabetes nephropathy and blood lipid metabolism. Chin J Integr Med Cardio-/Cerebrovascular Disease. (2017) 15:3221–3. doi: 10.3969/j.issn.1672-1349.2017.24.045

Crossref Full Text | Google Scholar

137. Wang X, Zhou W, Wang Q, Zhang Y, Ling Y, Zhao T, et al. A novel and comprehensive strategy for quality control in complex Chinese medicine formula using UHPLC-Q-Orbitrap HRMS and UHPLC-MS/MS combined with network pharmacology analysis: Take Tangshen formula as an example. J Chromatogr B Analyt Technol BioMed Life Sci. (2021) 1183:122889. doi: 10.1016/j.jchromb.2021.122889

PubMed Abstract | Crossref Full Text | Google Scholar

138. Kong Q, Zhang H, Zhao T, Zhang W, Yan M, Dong X, et al. Tangshen formula attenuates hepatic steatosis by inhibiting hepatic lipogenesis and augmenting fatty acid oxidation in db/db mice. Int J Mol Med. (2016) 38:1715–26. doi: 10.3892/ijmm.2016.2799

PubMed Abstract | Crossref Full Text | Google Scholar

139. Zhao T, Sun S, Zhang H, Huang X, Yan M, Dong X, et al. Therapeutic effects of tangshen formula on diabetic nephropathy in rats. PloS One. (2016) 11:e0147693. doi: 10.1371/journal.pone.0147693

PubMed Abstract | Crossref Full Text | Google Scholar

140. Li N, Zhao T, Cao Y, Zhang H, Peng L, Wang Y, et al. Tangshen formula attenuates diabetic kidney injury by imparting anti-pyroptotic effects via the TXNIP-NLRP3-GSDMD axis. Front Pharmacol. (2020) 11:623489. doi: 10.3389/fphar.2020.623489

PubMed Abstract | Crossref Full Text | Google Scholar

141. Liu P, Peng L, Zhang H, Tang PM, Zhao T, Yan M, et al. Tangshen formula attenuates diabetic nephropathy by promoting ABCA1-mediated renal cholesterol efflux in db/db mice. Front Physiol. (2018) 9:343. doi: 10.3389/fphys.2018.00343

PubMed Abstract | Crossref Full Text | Google Scholar

142. Li P, Chen Y, Liu J, Hong J, Deng Y, Yang F, et al. Efficacy and safety of tangshen formula on patients with type 2 diabet ic kidney disease: a multicenter double-blinded randomized placebo-con trolled trial. PloS One. (2015) 10:e0126027. doi: 10.12114/j.issn.1007-9572.2021.00.563

PubMed Abstract | Crossref Full Text | Google Scholar

143. Liying C, Mengxi W, Chong Z, and Fengmin S. Clinical efficacy of dangguibuxue decoction as an adjuvant therapy for diabetic nephropathy:a meta-analysis. Chin Gen Practice. (2021) 24:3477–83.

Google Scholar

144. Yufang L, Yang G, and Liqin C. Effect of danggui buxue decoction and jinkui shenqi pill on the treatment of diabetes nephropathy and the level of serum scr and BUN. Chin J Integrated Traditional Western Nephrology. (2020) 21:169–71.

Google Scholar

145. Xin S, Siquan Z, Yingwen Z, Haoliang K, and Shuaiyu. Effect of Danggui Buxue Decoction on PERK pathway in diabetes nephropathy rats. J Tianjin Univ Traditional Chin Med. (2018) 37:131–6. doi: 10.11656/j.issn.1673-9043.2018.02.11

Crossref Full Text | Google Scholar

146. Xiuping W and Yingwen Z. Effect of astragals and angelica mixture on the high density lipoprotein and urinary microalbuminuria in diabetic nephropathy rats. Chin J Integrated Traditional Western Nephrology. (2015) 16:1044–7.

Google Scholar

147. Sun L, Yang Z, Zhao W, Chen Q, Bai H, Wang S, et al. Integrated lipidomics, transcriptomics and network pharmacology analysis to reveal the mechanisms of Danggui Buxue Decoction in the treatment of diabetic nephropathy in type 2 diabetes mellitus. J Ethnopharmacol. (2022) 283:114699. doi: 10.1016/j.jep.2021.114699

PubMed Abstract | Crossref Full Text | Google Scholar

148. Dan Z, Qiming L, Yuqing H, Juan W, Mengxue W, and Xianjing Z. The influence of chinese angelica blood-supplementing decoction on diabetes nephropathy with qi deficiency and blood stasis syndrome and Renal fibrosis. Henan Traditional Chin Med. (2022) 42:1720–3. doi: 10.16367/j.issn.1003-5028.2022.11.0363

Crossref Full Text | Google Scholar

149. Song Haixiang LQ and Chunxiao Y. Effect of tangshenkang on urinary interleukin-6 in patients with diabetes nephropathy. J Shandong Univ Traditional Chin Med. (1999) 04):32–4. doi: 10.16294/j.cnki.1007-659x.1999.04.013

Crossref Full Text | Google Scholar

150. Song Haixiang WZ, Qi Li, and Zhensu Z. Effect of Tangshenkang on urinary transforming growth factor - β 1 in patients with diabetes nephropathy. Chin J Integrated Traditional Western Nephrol. (2004) 09):519–21. doi: 10.3969/j.issn.1009-587X.2004.09.008

Crossref Full Text | Google Scholar

151. Dandan W, Na Z, Lijun Y, Chunyan L, Dazhong C, and Jian M. Study on protective effect and mechanism of tangshenkang concentrated pill on diabetes nephropathy rats. J Liaoning Univ Traditional Chin Med. (2018) 20:40–3. doi: 10.13194/j.issn.1673-842x.2018.09.010

Crossref Full Text | Google Scholar

152. Xiaowei S, Dinghua Z, Xiangxia L, Yi L, Wenli P, Dongpeng Z, et al. Effect of kidney tonifying qi resolving turbidity tongluo method on renal extracellular matrix in diabetes nephropathy mice. Chin J Exp Traditional Med Formulae. (2018) 24:114–20. doi: 10.13422/j.cnki.syfjx.20180792

Crossref Full Text | Google Scholar

153. Qi W, Dandan W, Lijun Y, and Dazhong C. Effects of tangshenkang water pill on blood lipids and renal parameters in diabetes nephropathy model rats. Heilongjiang J Traditional Chin Med. (2020) 49:180–1.

Google Scholar

154. Yuning L, Lizhong G, Lihong W, Linqi Z, and Wei D. Experimental study on the prevention and treatment of diabetes nephropathy with the method of supplementing qi, nourishing yin, removing blood stasis and lowering turbidity. Chin Arch Traditional Chin Med. (2008) 08):1711–3. doi: 10.13193/j.archtcm.2008.08.112.liuyn.026

Crossref Full Text | Google Scholar

155. Xingkun Z, Li Z, Cuilan C, and Yashen L. Clinical study on intervention of tangshenkang capsule on serum ET-1 of phase III kidney deficiency and blood stasis syndrome in diabetes nephropathy. Liaoning J Traditional Chin Med. (2016) 43:347–9. doi: 10.13192/j.issn.1000-1719.2016.02.049

Crossref Full Text | Google Scholar

156. Wenli P, Xiaowei S, Yuan L, Dinghua Z, Dongpeng Z, Yufen D, et al. The clinical efficacy of Tangshenkang in the treatment of type 2 diabetes nephropathy and its regulatory effect on the disorder of lipid metabolism. Chin Arch Traditional Chin Med. (2021) 39:142–5. doi: 10.13193/j.issn.1673-7717.2021.10.034

Crossref Full Text | Google Scholar

157. Chen Y, Rui R, Wang L, Wang H, Zhu B, and Cao A. Huangqi decoction ameliorates kidney injury in db/db mice by regulatin g the BMP/Smad signaling pathway. BMC Complement Med Ther. (2023) 23:209. doi: 10.1186/s12906-023-04029-1

PubMed Abstract | Crossref Full Text | Google Scholar

158. Ying C, Yang L, and Aili C. The potential of huangqi decoction for treating diabetic kidney disease. Integr Med Nephrol Andrology. (2024) 11:e00020. doi: 10.1097/IMNA-D-23-00020

Crossref Full Text | Google Scholar

159. Ze H, Ri PC, Xi C, and Expedition S. Clinical study on 35 cases of diabetes nephropathy treated by detoxification, tongluo and kidney preservation. China Pract Med. (2011) 6:165–7. doi: 10.14163/j.cnki.11-5547/r.2011.06.012

Crossref Full Text | Google Scholar

160. Jinying C, Wenhang L, Haotian Q, Weichen N, Mijia, and Expedition S. Study on the mechanism of xiaoke shen'an decoction in treating diabetes nephropathy based on network pharmacology and molecular docking. Chin J Gerontology. (2022) 42:5711–7. doi: 10.3969/j.issn.1005-9202.2022.23.012

Crossref Full Text | Google Scholar

161. Gao Y, Su X, Xue T, and Zhang N. The beneficial effects of astragaloside IV on ameliorating diabetic kidney disease. BioMed Pharmacother. (2023) 163:114598. doi: 10.1016/j.biopha.2023.114598

PubMed Abstract | Crossref Full Text | Google Scholar

162. Liu J, Ren J, Zhou L, Tan K, Du D, Xu L, et al. Proteomic and lipidomic analysis of the mechanism underlying astragalo side IV in mitigating ferroptosis through hypoxia-inducible factor 1α/ heme oxygenase 1 pathway in renal tubular epithelial cells in diabetic kidney disease. J Ethnopharmacol. (2024) 334:118517. doi: 10.1016/j.jep.2024.118517

PubMed Abstract | Crossref Full Text | Google Scholar

163. Su J, Gao C, Xie L, Fan Y, Shen Y, Huang Q, et al. Astragaloside II ameliorated podocyte injury and mitochondrial dysfunction in streptozotocin-induced diabetic rats. Front Pharmacol. (2021) 12:638422. doi: 10.3389/fphar.2021.638422

PubMed Abstract | Crossref Full Text | Google Scholar

164. Xue R, Zhai R, Xie L, Zheng Z, Jian G, Chen T, et al. Xuesaitong protects podocytes from apoptosis in diabetic rats through modulating PTEN-PDK1-akt-mTOR pathway. J Diabetes Res. (2020) 2020:9309768. doi: 10.1155/2020/9309768

PubMed Abstract | Crossref Full Text | Google Scholar

165. Shen Y, Chen W, Lin K, Zhang H, Guo X, An X, et al. Notoginsenoside Fc, a novel renoprotective agent, ameliorates glomerular endothelial cells pyroptosis and mitochondrial dysfunction in diabetic nephropathy through regulating HMGCS2 pathway. Phytomedicine. (2024) 126:155445. doi: 10.1016/j.phymed.2024.155445

PubMed Abstract | Crossref Full Text | Google Scholar

166. Tao A, Zhang Y, Gan Z, Yin C, Tian Y, Zhang L, et al. Isolation, structural features, and bioactivities of polysaccharides from Panax notoginseng: A review. Int J Biol Macromol. (2024) 280:135765. doi: 10.1016/j.ijbiomac.2024.135765

PubMed Abstract | Crossref Full Text | Google Scholar

167. Yi L and Xi Y. Experimental Study on the Effects of Panax notoginseng Polysaccharide on Inflammatory Response and Lipid Metabolism in diabetes Nephropathy Rats. Chin J Traditional Med Sci Technology. (2018) 25:43–7.

Google Scholar

168. Cai L, Chen Y, Xue H, Yang Y, Wang Y, Xu J, et al. Effect and pharmacological mechanism of Salvia miltiorrhiza and its characteristic extracts on diabetic nephropathy. J Ethnopharmacol. (2024) 319:117354. doi: 10.1016/j.jep.2023.117354

PubMed Abstract | Crossref Full Text | Google Scholar

169. Yang XY, Sun L, Xu P, Gong LL, Qiang GF, Zhang L, et al. Effects of salvianolic scid A on plantar microcirculation and peripheral nerve function in diabetic rats. Eur J Pharmacol. (2011) 665:40–6. doi: 10.1016/j.ejphar.2011.03.054

PubMed Abstract | Crossref Full Text | Google Scholar

170. Huang MQ, Zhou CJ, Zhang YP, Zhang XQ, Xu W, Lin J, et al. Salvianolic Acid B Ameliorates Hyperglycemia and Dyslipidemia in db/db Mice through the AMPK Pathway. Cell Physiol Biochem. (2016) 40:933–43. doi: 10.1159/000453151

PubMed Abstract | Crossref Full Text | Google Scholar

171. Jun C, Haizhou Z, Yulin W, and Qinan W. Effect of salvia miltiorrhiza polyphenolate combined with losartan potassium on related factors of senile diabetes nephropathy. Med J West China. (2018) 30:1451–5+61. doi: 10.3969/j.issn.1672-3511.2018.10.012

Crossref Full Text | Google Scholar

172. Wanxia H and Guannan Z. Effects of high flux hemodialysis combined with salvia miltiorrhiza polyphenolate injection on oxidative stress and micro inflammation in patients with diabetes nephropathy. Modern J Integrated Traditional Chin Western Med. (2019) 28:1207–10. doi: 10.3969/j.issn.1008-8849.2019.11.018

Crossref Full Text | Google Scholar

173. Han W, Baoyuan G, Xuetuan Z, and Da F. Effect of salvia miltiorrhiza polyphenolate in adjuvant treatment of diabetes nephropathy and its influence on the levels of inflammatory factors and serum oxidative stress indicators. Chin J Public Health Eng. (2021) 20:1027–8+31. doi: 10.19937/j.issn.1671-4199.2021.06.055

Crossref Full Text | Google Scholar

174. Jia LQ, Zhang N, Xu Y, Chen WN, Zhu ML, Song N, et al. Tanshinone IIA affects the HDL subfractions distribution not serum lipid levels: Involving in intake and efflux of cholesterol. Arch Biochem Biophys. (2016) 592:50–9. doi: 10.1016/j.abb.2016.01.001

PubMed Abstract | Crossref Full Text | Google Scholar

175. Xu Z, Cai K, Su SL, Zhu Y, Liu F, and Duan JA. Salvianolic acid B and tanshinone IIA synergistically improve early diabetic nephropathy through regulating PI3K/Akt/NF-κB signaling pathway. J Ethnopharmacol. (2024) 319:117356. doi: 10.1016/j.jep.2023.117356

PubMed Abstract | Crossref Full Text | Google Scholar

176. Fan X, Xinlong L, Ye L, Rainforest F, and Feixia D. Meta analysis of the efficacy of sodium tanshinone II A sulfonate injection in the treatment of diabetes nephropathy. Chin J Modern Appl Pharmacy. (2020) 37:2371–7. doi: 10.13748/j.cnki.issn1007-7693.2020.19.013

Crossref Full Text | Google Scholar

177. Zhiting H, Jing Z, Ling F, Xiaoli X, and Wei L. The effect of tanshinone II A injection on the treatment of type 2 diabetes nephropathy patients with maintenance hemodialysis and the influence of IGF-1 and plasma protein C levels. Hainan Med J. (2021) 32:2861–4. doi: 10.3969/j.issn.1003-6350.2021.22.002

Crossref Full Text | Google Scholar

178. Wu Q, Guan YB, Zhang KJ, Li L, and Zhou Y. Tanshinone IIA mediates protection from diabetes kidney disease by inhibiting oxidative stress induced pyroptosis. J Ethnopharmacol. (2023) 316:116667. doi: 10.1016/j.jep.2023.116667

PubMed Abstract | Crossref Full Text | Google Scholar

179. Jie Y, Wenyi W, Caifang J, Xuelan Z, and Qinqing L. Study on the mechanism of Ligustrum lucidum in the treatment of diabetes nephropathy based on network pharmacology. Chin Med Modern Distance Educ China. (2024) 22:126–9. doi: 10.3969/j.issn.1672-2779.2024.05.041

Crossref Full Text | Google Scholar

180. Luan R, Zhao P, Zhang X, Li Q, Chen X, and Wang L. Pharmacodynamics, pharmacokinetics, and kidney distribution of raw and wine-steamed ligustri lucidi fructus extracts in diabetic nephropathy rats. Molecules. (2023) 28:791. doi: 10.3390/molecules28020791

PubMed Abstract | PubMed Abstract | Crossref Full Text | Google Scholar

181. Tian F, Yi X, Yang F, Chen Y, Zhu W, Liu P, et al. Research progress on the treatment of diabetic nephropathy with leech and its active ingredients. Front Endocrinol (Lausanne). (2024) 15:1296843. doi: 10.3389/fendo.2024.1296843

PubMed Abstract | Crossref Full Text | Google Scholar

182. Fan Y, Yachun L, Shuai G, and Zhiqiang C. Interventional effect of leech freeze-dried powder on related indexes of blood stasis syndrome in diabetes nephropathy rats. Chin Traditional Patent Med. (2022) 44:3017–22. doi: 10.3969/j.issn.1001-1528.2022.09.051

Crossref Full Text | Google Scholar

Keywords: diabetic kidney disease, lipid metabolism disorder, traditional Chinese patent medicines, Chinese medicine compound, single Chinese medicine, integrated traditional Chinese and Western medicine

Citation: Sheng L, Cao Z, Wang L, Xu Y and Gui D (2025) Research progress in the treatment of lipid metabolism disorder in patients with diabetic kidney disease by the integrated traditional Chinese and Western medicine. Front. Endocrinol. 16:1631312. doi: 10.3389/fendo.2025.1631312

Received: 19 May 2025; Accepted: 07 July 2025;
Published: 30 July 2025.

Edited by:

Tingting Zhao, University of Macau, China

Reviewed by:

Qida He, Xiamen University, China
Song Hu, Shanghai Pudong New Area People’s Hospital, China
Haimin Ye, The Second Affiliated Hospital of Hunan University of Chinese Medicine, China

Copyright © 2025 Sheng, Cao, Wang, Xu and Gui. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Youhua Xu, eWh4dUBtdXN0LmVkdS5tbw==; Dingkun Gui, ZGluZ2t1bmd1aUBhbHUuZnVkYW4uZWR1LmNu

These authors have contributed equally to this work

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.