3,619
Views
4
CrossRef citations to date
0
Altmetric
Research Article

Targeted drug delivery strategy: a bridge to the therapy of diabetic kidney disease

, , , , &
Article: 2160518 | Received 27 Oct 2022, Accepted 12 Dec 2022, Published online: 28 Dec 2022

Abstract

Diabetic kidney disease (DKD) is the main complication in diabetes mellitus (DM) and the main cause of end-stage kidney disease worldwide. However, sodium glucose cotransporter 2 (SGLT2) inhibition, glucagon-like peptide-1 (GLP-1) receptor agonist, mineralocorticoid receptor antagonists and endothelin receptor A inhibition have yielded promising effects in DKD, a great part of patients inevitably continue to progress to uremia. Newly effective therapeutic options are urgently needed to postpone DKD progression. Recently, accumulating evidence suggests that targeted drug delivery strategies, such as macromolecular carriers, nanoparticles, liposomes and so on, can enhance the drug efficacy and reduce the undesired side effects, which will be a milestone treatment in the management of DKD. The aim of this article is to summarize the current knowledge of targeted drug delivery strategies and select the optimal renal targeting strategy to provide new therapies for DKD.

Introduction

Diabetic kidney disease (DKD) is the predominant complication of diabetes mellitus (DM). Approximately 21.3% of the DM patients accompany with chronic kidney disease (CKD) in China, and the incidence is increasing year by year (Zhang et al., Citation2016). DKD is the most common cause of renal failure all over the world, and about 50% of patients progressed to dialysis or transplant stage in the USA, which significantly elevates the risk for mortality (DeFronzo et al., Citation2021). Effective treatment options are in urgent need to postpone the disease progression. Except for the traditional agents, such as angiotensin-converting enzyme inhibitors (ACEI) and angiotensin II receptor blockers (ARB), sodium glucose cotransporter 2 (SGLT2) inhibition, glucagon-like peptide-1 (GLP-1) receptor agonist, mineralocorticoid receptor antagonists (MRAs) and endothelin receptor A inhibition have a beneficial impact on the control of the pathophysiological abnormalities (Michos and Tuttle, Citation2021; Zhou et al., Citation2021; Barrera-Chimal et al., Citation2022; Ravindran and Munusamy, Citation2022). In spite of this, the adverse reaction is unavoidable and the incidence of diabetic nephropathy has not declined, with the result that many patients still inevitably develop uremia. In this condition, novel treatment options with improved targetability, bioavailability, efficacy and safety are urgently needed.

Targeted drug delivery strategy is a rapidly developed method in which the specific bio-actives are transported with carrier system to the predetermined organ or cell. It enables the therapeutic agents specifically transfer and accumulate at the diseased sites and increases the local concentration of drugs and minimizes the side effects (Liang et al., Citation2021). In this process, three important components are included as follows: first, the specific drug moiety; second, the drug delivery carrier; third, the targeted cells, tissues or organs to be treated. There are two types of mechanisms for carrier-targeted drug delivery systems: slowing down the rate of drug release, or releasing in large quantities at the target site by adjusting for changes in pH, heat, or light (Ashique et al., Citation2021). The carrier, with the properties of nontoxicity, biodegradability, diminished immunogenicity and easy detection, is used to encapsulate the drug molecules for delivery to the target organs, while enhancing the solubility, permeability and bioavailability of the drug molecules (Su et al., Citation2015).

The kidney-targeted drug delivery systems were comprehensively raised by Haas in 2002 (Haas et al., Citation2002). In the following two decades, the related researches about targeted therapies for different kinds of kidney diseases and renal ultrastructure were published successively. Targeted drug delivery for DKD is one of the current research hotspots and may be a promising solution for the prevention and treatment of DKD. The purpose of this review is to comprehensively summarize the current knowledge of targeted drug delivery strategies for DKD from the perspective of the pathogenesis and the renal ultrastructure, in order to select the optimal targeting principles and potential drug carriers to provide new therapies for DKD.

Diabetic kidney disease

DKD is one of the most severe microvascular complications of DM, which is often associated with cardiovascular disease and increased the risk of mortality in diabetic patients (Samsu, Citation2021; Fox et al., Citation2012). The estimated number of global DM patients has reached 463 million and the prevalence of DKD has obviously increased in the recent two decades (Saeedi et al., Citation2019). About 35 to 40% of type 1 or 2 diabetes patients will eventually develop DKD, which has a profound implication for global public health and socioeconomics (Yang et al., Citation2018).

Pathogenesis of DKD

The pathogenesis of DKD is complicated and involves plenty of different pathways (). It is always regarded that DKD is a non-inflammatory disease caused by metabolism, but with the deepening of research, increasing evidence suggests that inflammation also participates in its pathogenesis. The metabolic pathways in DKD mainly include reactive oxygen species (Han et al., Citation2018), mitochondrial dysfunction (W Dai et al., Citation2021), transcription factors (Nrf2) (S Li et al., 2021), NADPH oxidase (NOX) (You et al., Citation2016), protein kinase C (PKC) (Yin et al., Citation2019), apoptosis-signaling kinase 1 (ASK1) (Tesch et al., Citation2015), advanced glycation (Sanajou et al., Citation2018), autophagy (Yang et al., Citation2021), fibrosis (Jha et al., Citation2022), Janus kinase-signal transducer and activator of transcription (JAK-STAT) (Brosius et al., Citation2016) and epigenetics (Kato and Natarajan, Citation2019). Increased expression of inflammatory cytokines, adhesion molecules, growth factors and chemokines, including IL-1, IL-6, IL-18, tumor necrosis factor, intercellular adhesion molecule 1, Vascular cell adhesion protein 1, endothelial cell-selective adhesion molecule, CCL2, CX3CL1, CCL5 and so on, are reported to be involved in the pathogenesis of DKD (Navarro-González et al., Citation2011). Immune cells, such as macrophages, T cells, B cells, dendritic cells, neutrophils and mast cells, are commonly observed in DKD patients (Moon et al., Citation2012; Klessens et al., Citation2017; Yang and Mou, Citation2017). These pathways are potential candidates for the DKD therapy. The deeper the research mechanism, the more precise the targeted therapy of diabetic nephropathy.

Figure 1. The potential pathogenesis of diabetic kidney disease (DKD). Nrf2, transcription factors; NOX, NADPH oxidase; PKC, protein kinase C; ASK1, apoptosis-signaling kinase 1; JAK-STAT, Janus kinase-signal transducer and activator of transcription; TNF, tumor necrosis factor.

Figure 1. The potential pathogenesis of diabetic kidney disease (DKD). Nrf2, transcription factors; NOX, NADPH oxidase; PKC, protein kinase C; ASK1, apoptosis-signaling kinase 1; JAK-STAT, Janus kinase-signal transducer and activator of transcription; TNF, tumor necrosis factor.

Pathological changes of DKD

DKD was traditionally diagnosed by the development of persistent albuminuria and the followed progressive GFR decline, which was defined as the typical phenotype of DKD (National Kidney Foundation, Citation2012). However, several studies have reported that some DM patients without albuminuria progress to renal insufficiency, described as normoalbuminuric diabetic kidney disease (NADKD) (Q Dai et al., Citation2021; Jia et al., Citation2022). In a retrospective study, 32 out of 36 deceased diabetic kidney donors, presenting with non-albuminuria and an estimated GFR > 60 ml/min/1.73m2, showed histopathological lesions consistent with DKD, which indicated a high proportion of undiagnosed DKD (Comai et al., Citation2019). Therefore, the renal biopsy is of great importance in the diagnosis of DKD.

Glomerular basement membrane (GBM) thickening is considered as the earliest observed pathological feature in patients with DKD, which is appeared within 1–2 years after the onset of DM (Tervaert et al., Citation2010; Ponchiardi et al., Citation2013). Endothelial cells play an important role in the progression of DKD. With the development of DKD, the fenestrated ECs are decreased in diabetic patients, which correlates with albuminuria and the loss of GFR (Dou and Jourde-Chiche, Citation2019). Mesangial expansion, caused by Mesangial cells (MCs) enlargement and accumulation of glomerular matrix protein, is the most common renal pathological change in DKD (Reidy et al., Citation2014; Zhang et al., Citation2019). On the glomerular capillary side of MCs, without the surrounding of GBM and podocytes, drugs can be delivered to MCs for treating kidney diseases (Scindia et al., Citation2008). Podocytes are glomerular epithelial cells which contain 3 separate elements: cell body, extending processes and foot processes (Garg, Citation2018). Podocytes injury in DKD is induced by many compound factors, such as inflammatory reaction, mechanical stress, oxidative stress, renin angiotensin aldosterone system activation, TGF-β1 induction, and AGEs accumulation, and any part of the pathway is expected to be the target of DKD therapy (Kawanami et al., Citation2016). The renal tubules consist of the proximal tubules, collecting tubules and distal tubules. The morphological and functional changes of the renal tubules are involved in the pathogenesis and progression of DKD (Duan et al., Citation2021). Most renal tubular targeted systems are directed at the proximal tubules (Christensen et al., Citation2012).

Management of DKD

The management of DKD mainly contains modification of lifestyle (smoking cessation, proper exercise, low-salt and low-protein diet and weight reduction), glucose control, blood pressure control and lipid profiles control. Some novel glucose-lowering agents have both glucose control and renal protective effects. SGLT2 inhibitor can inhibit the glucose reabsorption in proximal tubular and promote urinary glucose excretion. Empagliflozin, an SGLT2 inhibitor, can significantly reduce the cardiovascular events in type2 DM and delay the progression of kidney disease (Zinman et al., Citation2015; Wanner et al., Citation2016). Canagliflozin and dapagliflozin demonstrated the similar renal protection (Neal et al., Citation2017; Wiviott et al., Citation2019). GLP-1 receptor agonist and dipeptidyl peptidase-4 (DPP4) inhibitor are incretin hormone, released from the gastrointestinal tract, which can stimulate insulin secretion and keep glucose in a stable level. Recent trials have further illustrated the kidney benefits of GLP1 receptor agonist in DKD (Gerstein et al., Citation2021). DPP4 inhibitor can reduce the albuminuria level, but the renoprotective effect needs further research (Mosenzon et al., Citation2017). Mineralocorticoid receptor antagonists (MRAs) are a prospective therapeutic option in DKD because of the activation of mineralocorticoid receptor in podocytes, mesangial cells, inflammatory cells, fibroblasts, and vascular cells (Barrera-Chimal et al., Citation2022). Spironolactone and eplerenone, kinds of MRAs, can decrease albuminuria and ameliorate renal injury in DKD (Makhlough et al., Citation2014; El Mokadem et al., Citation2020). Although these new hypoglycemic drugs have satisfying effects in the treatment of DKD, some side effects, just as the urinary tract infection of SGLT2 inhibitor, the gastrointestinal reaction of incretin hormone, the hyperkalemia of MRAs, limit their application in some patients. As the impaired glomerular filtration and tubular damage in DKD, the conventional dosage of drug does not ensure the required amount of it get into the targeted cell. Kidney targeted drug delivery strategy may help to solve the above problems.

Targeted drug delivery strategy

The development of new drugs usually fails in the final stage due to safety and efficacy, and the root causes are the high enrichment of off-target organs and the low enrichment of target organs, which lead to the dose-related toxicities and dose-limited efficacies. Targeted drug delivery strategy is designed to enhance the drug’s therapeutic efficacy through targeted distribution and to decrease the undesired toxicities and reduce the off-target effects. The three important parts in this process are drugs, delivery systems and the targeted organs or cells. The drug delivery system contains five different types: passive drug targeting, active drug targeting, cancer cell targeting, endothelial cell targeting and triggered drug release (Ozturk-Atar et al., Citation2018). Passive drug targeting systems rely on pharmacological or physicochemical factors to work and they do not have a specific targeting ligand to guide drugs or carriers to their target, so they need a prolonged circulation time in the blood in order to have enough drug amounts in target site (Hirsjarvi et al., Citation2011). Active drug targeting included both direct and indirect ways. The direct active targeting refers that the moieties on the drug or the drug carrier directly conjugate to the target cells or organs through a specific structure (Byrne et al., Citation2008). The indirect ways need a two-step strategy, just as the antibody-directed enzyme prodrug therapy or gene-directed enzyme prodrug therapy (Schellmann et al., Citation2010). Compared to the passive drug targeting, the active targeting can significantly increase the number of drugs that are delivered to the target cell (Ashique et al., Citation2021).

Targeted drug delivery strategy in DKD

The targeted treatments for DKD are the research hotspot at present. We mainly review the delivery system about the carriers which can transfer the effective drugs in the therapy of DKD ().

Figure 2. Targeted drug delivery strategies for diabetic kidney disease (DKD).

Figure 2. Targeted drug delivery strategies for diabetic kidney disease (DKD).

Nanoparticles

Nanoparticle is a kind of structure with the size vary from 10–1000 nm, with a solid core surrounded by suitable chemicals which may affect the size and polarity (Xu et al., Citation2018). The size of the nanoparticles has important effects on circulation half-life, cellular absorption and target cells, and the ones with the size of 100 nm have the longest half-life (Sharma et al., Citation2011). Many shapes have been constructed such as rod, spherical, cubic, helical and hexagonal, among them the rod one has shown to be more effective and the spherical one clears more slowly (Chauhan et al., Citation2011; Blanco et al., Citation2015). The drugs can be encapsulated, dissolved in the nanoparticle or attached to its matrix, which can increase the bioavailability, solubility, drug penetration and protect it from degradation (Qamar et al., Citation2019). It always combined with chitosan, poly(d,l-lactide-co-glycolide) (PLGA), poly(ɛ-caprolactone) (PCL) polymer in drug delivery area. (Merlin and Li, Citation2021) briefly summarized the nanocarriers which were used to carry drugs in the form of nanocomplex. Selenium nanoparticles (SeNPs), with the diameter ranging from 40–90 nm, can downregulate the levels of creatine, blood urea nitrogen, collagen and fibronectin and upregulate the albumin level in DKD rats, meanwhile SeNPs regulate the level of Bax and Bcl-2, and elevate the expression of SIRT1 and heat shock protein-70 (Kumar et al., Citation2014). Rhein (RH) is a kind of Chinese traditional herbal medicine with multi-therapy effects on DKD, but its application is limited by the low bioavailability, poor solubility, decreased renal distribution. RH-loaded polyethyleneglycol-co-polycaprolactoneco-polyethylenimine nanoparticles (PPP-RH-NPs) with the size of 75 ± 25 nm are designed for kidney-targeted drug delivery, which can suppress the expression of TGF-β1 and Smad2/3 phosphorylation and reduce the level of proteinuria, blood creatine, blood urea nitrogen in DKD rats (Chen et al., Citation2018). Advanced glycation end products (AGEs) and its receptor (RAGE) are potential key therapy targets in DKD. Xuemei Zhou et al have proposed a dual-target nanoparticle (ACEs inhibitors and RAGE inhibitors) with high efficiency and synergistic effect, which may have potential application in the therapy of DKD (Zhou et al., Citation2012). MET-HMSNs-CeO2, a multifunctional nanoparticle, was doped with ceria in the surface and loaded metformin (MET) in the pore of hollow mesoporous silica nanocomposite (HMSN), which could exhibit antioxidative and antiapoptotic function in DKD (Tong et al., Citation2020). We summarize the other kinds of nanoparticles in the treatment of DKD ().

Figure 3. Schematic illustration of nanoparticles (Merlin and Li, Citation2021).

Figure 3. Schematic illustration of nanoparticles (Merlin and Li, Citation2021).

Table 1. Nanoparticles in the treatment of DKD.

Macromolecular carriers

Polymers

The application of polymers has played a significant role in the targeted drug delivery system. Polymeric micelles, polymersomes and nanohydrogels are polymer nanosystems, assembled by polymers with different compositions, structures and molecular weights, and each type of them has its particular identity to attach or encapsulate therapeutic agents (Joglekar and Trewyn, Citation2013). They can minimize the adverse effects of drugs by the site-specific therapy. Polymeric micelles, with the dimension of 5–100 nm, can prolong the blood circulation time by reducing the clearance of the reticuloendothelial system and decreasing the drug side effects by lower the opsonization (Joglekar and Trewyn, Citation2013). Polymersome are polymer-based vesicles with amphiphilic copolymers and it can be stimulated by a certain wide wavelength of light in order to increase the drug efficacy and decrease adverse effects (Hernandez Becerra et al., Citation2022). Nanohydrogel, possessed the characteristic of hydrogel and nanoparticle, is a three-dimensional and cross-linked hydrophilic polymer network, with the diameter of 1–100 nm (Gonçalves et al., Citation2010). The nanohydrogel can encapsulate the drug through physical attachment, self-assembly and covalent conjugation (Dalwadi and Patel, Citation2015). Chen L et al. reported a new one-dimensional Cu (II) coordination polymer by using a NNO tridentate (NNO) Schiff base ligand4-fluoro-2-(((2-(methylamino)ethyl)amino)methyl)phenol, which can downregulate the serum amyloid A and TNF-α in STZ induced DKD rats (Pan-Pan Lin and Chen, Citation2021).

Chitosan

Chitosan is a kind of polysaccharide derived from natural chitin which exists in the crustacean shells, fungal cell walls, arthropods and insects, with the properties of low toxicity, bioactivity, biocompatibility, biodegradability and structural variability (Kean and Thanou, Citation2010; Motiei et al., Citation2017).Chitosan can be degraded by chitosanase and lysozyme to form oligosaccharides and monosaccharides, and then absorbed by the body (Woraphatphadung et al., Citation2018). Chitosan stabilized nanoparticles (Ch-SeNPs) can downregulate the expression of TNF-α, IL-6, and IL-1β in type 2 DM rats, and Ch-SeNPs combine with metformin can reduce the creatine, proteinuria and urea (Khater et al., Citation2021). Researchers reported that chitosan oligosaccharides can ameliorate the proteinuria, reverse the kidney pathological changes, reduce the expression of TGF-β1 and fibronectin and the activity of urine N-acetyl-β-D-glucosaminidase (NAG) in STZ-T2DM rat model (Zhang et al., Citation2018).

Liposomes

Liposomes are spherical vesicles with an aqueous nucleus surrounded by concentric phospholipid bilayers (Guimaraes et al., Citation2021). Liposomes are widely used in targeted drug delivery system on account of their nontoxic and biodegradable. The targeting strategies of liposome can be divided into passive and active targeting, while the passive one always relies on the vasculature and the active targeting depends on the binding of the receptor-specific ligands on the liposome surface (Lehner et al., Citation2013). Quercetin is a sort of bioflavonoid with the features of anti-inflammatory, anti-diabetes, anti-allergic, anti-cancer and reduce aldose reductase (Shi et al., Citation2019). The poor water solubility of quercetin restrains its application; hence, quercetin liposomes are designed to improve its solubility. Quercetin liposomes significantly downregulate the expression of creatine, urea, TNF-α, IL-1β, AGEs and alleviate the renal histopathological changes (Tang et al., Citation2020). Oxidative stress plays an important role in the pathogenesis of DKD (Vodosek Hojs et al., Citation2020). Glutathione is a kind of antioxidant substance, but it is unstable and cannot easily penetrate the cell membrane. Huajuan Shen et al reported glutathione liposomes can improve the stability, bioavailability and antioxidant ability of glutathione, which can target kidney and reduce the proteinuria, creatine, urea, alleviate the kidney fibrosis in DKD rats (Shen and Wang, Citation2021).

Exosomes

Exosomes, with a diameters of 30–150 nm, are extracellular vesicles composed of bilayer membranes, and take part in the process of immune response, inflammation, coagulation, cell proliferation and cell migration (Simons and Raposo, Citation2009). Exosomes can reduce immunogenicity, enhanced tissue penetration and cross biological barriers, such as the blood–brain barrier and the gastrointestinal tract (Shao et al., Citation2020). A wide range of molecules, such as drugs, nucleic acid and protein, can be delivered by exosomes in terms of incubation, electroporation or sonication (Gutierrez-Millan et al., Citation2021). The exosomes can fuze with the targeted cells and release the contents into them. Mesenchymal stem cell (MSC)-derived exosomes downregulate serum creatinine, blood urea nitrogen and proteinuria, decrease mesangial expansion and collagen fibers around glomerular capillaries and the tubules, with obvious upregulating of LC3 and Beclin-1 and downregulating of mTOR, indicating that autophagy induced by exosomes may attenuate the clinical and pathological manifestation of DKD in the STZ-DM rats (Ebrahim et al., Citation2018). Exosomes, derived from MSC-conditioned medium, are administrated into unilateral kidney subcapsular space of STZ-induced DM rats, which can reduce renal tubules dilatation, tubular epithelial atrophy and inflammatory cells infiltration (Nagaishi et al., Citation2016). Juan Jin et al reported that adipose-derived stem cells-exosome (ADSCs-Exo) can alleviate the levels of proteinuria, serum creatinine, blood urea nitrogen and podocyte apoptosis in DKD rats, and the underlying mechanism could be the upregulated expression of miR-486 induce the inhibition of the Smad1/mTOR signaling pathway (Jin et al., Citation2019).

Peptide-based carriers

Therapeutic peptides are sometimes limited by their poor stability and poor cellular uptake, in this case, the development of cell-penetrating peptide-based drug delivery system can overcome the limitation to some extent. The cell penetrating peptide (CPP), are short peptides with positive charge, contains synthetic and natural CPP, protein transduction domains (PTDs) and membrane-translocating sequences (Wadia and Dowdy, Citation2002). These CPPs and the drugs they transport are internalized by cells through the endocytic pathway (Deshayes et al., Citation2005). Therapeutic peptides in DKD mainly include insulin, GLP-1 and analogs, which are administrated parenterally. Delivery of these peptides by oral route has been a research hotspot. The binding of peptides to CPPs is a potential route for the oral administration of these macromolecular drugs (Kristensen et al., Citation2016). PLGA-GLP-1 nanoparticles conjugated to CPP, then encapsulated with HPMC-polymer and loaded with iDPP4, which formed GLP-1/iDPP4 delivery multifunctional composite system, and the system was reported to have prolonged hypoglycemic effects in DM rats (Araujo et al., Citation2016). Thiolated polymer is a kind of delivery system of noninvasive peptides and proteins. Thiolated-chitosan-6-mercaptonicotinic acid for oral insulin delivery system can downregulate the serum glucose in rats (Millotti et al., Citation2014). Peptide can be delivered by microencapsulation which can protect them from being digested by the gastric acid. Oral β-cyclodextrin insulin microparticles had a promising effect in decreasing the blood glucose in DM rats and the hypoglycemic effect was similar to that of subcutaneous injection of insulin (D’Souza et al., Citation2015). Nanoparticles, liposome and micelles were also reported to be the promising oral peptides delivery system (Sharma et al., Citation2015; Ismail and Csoka, Citation2017).

Small-molecule prodrugs

The definition of prodrug raised by Albert can best summarize its features, that is ‘Prodrugs are chemicals with little or no pharmacological activity, undergoing biotransformation to a therapeutically active metabolite (Albert, Citation1958)’. There are two kinds of prodrugs, the bioprecursors and the carrier-linked prodrugs. Bioprecursors, including quinones, nitroarenes, N-oxides and amidoximes, do not have carriers and they can be activated by hydration, while the carrier-linked prodrugs are always esters activated by enzymatic hydrolysis (Testa, Citation2009). Gluconeogenesis is an important cause of hyperglycemia in type 2 DM. Researchers synthesized a small molecular 4-[2-(1H-indol-3-yl) vinyl] quinoline (IVQ) as a prodrug, which can suppress hepatic gluconeogenesis and ameliorate hyperglycemia without the cardiovascular side effects and genotoxicity in type 2 DM rats (Zhou et al., Citation2019). Bardoxolone methyl (CDDO-Me) exhibited a promising therapy for diabetic nephropathy in clinical trials, but the cardiac toxicity restrained its application. Therefore, a series of prodrugs, just as Cathepsin B (CTSB)-α-cyano-α, β-unsaturated ketone (CUK)-polyethylene glycol (PEG), were formed, making CDDO-Me without the active CUK part exposure in order to reduce the side effects of the treatment (Liu et al., Citation2022).

Antibody-modified carriers

Antibody, a complex protein-based molecule, is a crucial component in the indirect ways of active targeting. Antibodies vary in molecular weight, and those with molecular weight greater than 150 kDa cannot pass through the glomerular filtration barrier. Antibodies with the molecular weight lower than 50 kDa may be promising moieties in the kidney-targeted delivery system as they pass through the glomerular filtration barrier and can be reabsorbed by the proximal tubular cells (Chen et al., Citation2020a). Bovine albumin-based NPs with the diameters of 10 nm can selectively target the Fc receptor (FcRn) on the surface of podocytes (Wu et al., Citation2017). Anti-vascular cell adhesion protein 1 (VCAM-1)-rapamycin-SAINT-O-Somes can delivery rapamycin to the kidney and have little effect on cellular viability in order to reduce the side effect (Visweswaran et al., Citation2015). E-selectin is overexpressed in highly inflammatory renal environments and is also used as an antibody target for NP kidney targeting (Asgeirsdottir et al., Citation2008).

Targeted drug delivery strategy for MCs

Nanoparticles

Glomerular MCs is the potential drug delivery target cell in DKD. Nanoparticles pass through the fenestrated endothelium and get close to the MCs, in this case, the MCs-targeted nanoparticle should be designed to be smaller than the diameter of the endothelium pore. The diameter of glomerular endothelium pore is reported to be 80–100 nm (Luft et al., Citation1982). PEGylated gold nanoparticles with the size of 75 ± 25 nm can specifically target the MCs in mice (Choi et al., Citation2011).Chitosan improve the protection efficiency for siRNA and Emine Salva et al construct siPDGF-B- and siPDGFR-β-containing chitosan nanoparticles which can target the MCs to knock down PDGF-B and its receptor in order to decrease the MCs’ proliferation (Salva et al., Citation2017). Glucose transporter 1(GUT1) is a glucose transporter enriched in the MCs of DKD patients and the expression of GUT1 in MCs is positively correlated with oxidative stress to promote the progression of DKD (Zhang et al., Citation2010). Astaxanthin (AST) is a natural nontoxic lutein carotenoid with powerful antioxidant capacity (Sila et al., Citation2015). As the low stability and solubility of AST restrict the antioxidant capacity, AST-GLU-LIP is constructed to overcome the shortcomings and the results demonstrate that AST-GLU-LIP can improve the bioavailability and antioxidant capacity of AST and ameliorate the kidney pathological lesion in DKD rats (Chen et al., Citation2020b). A kind of glucose ligand modified neutrophil-like cell membrane coated melanin nanoparticle (Melanin @Glc-NCM) was prepared and applied in DKD of gestational rats, which can promote the apoptosis of glomerular MCs and restrain the hyperproliferation of MCs (J Li et al., 2021).

Liposome

Thy 1.1 antigen is specifically expressed in MCs. OX7-coupled liposome was equipped by connecting them with the Fab fragments of OX7 monoclonal antibody to thy1.1 antigen, and the OX7-LIP exhibits the specific targeting to MCs in rats (Tuffin et al., Citation2005). Anti-α8 integrin immunoliposome was a specific delivery to the MCs as the α8 integrin was expressed exclusively in MCs (Scindia et al., Citation2008). Triptolide (TP) suppresses the proliferation of MCs in DKD via the inhibition of PDK1/Akt/mTOR Pathway (Han et al., Citation2017). The side effects of TP, such as bone marrow suppression, liver function damage, and genital system toxicity, limit its application. TP was loaded into the TRX-20 modified liposomes and modified with PEG5000, forming PEG-TRX-TP-LP and specific binding to MCs, which displayed anti-inflammatory actions (Yuan et al., Citation2017). Puerarin is an anti-hyperglycemic and anti-oxidative active ingredient, but the aqueous solubility and poor oral availability restrain its clinical use. Puerarin liposome with the concentration of 50–100 µM was the most effective one to lower the MCs proliferation in rat MCs line under high glucose environment (Barro et al., Citation2021). Wei B et al reported that quercetin liposome (Q-PEGL) could improve the renal related biochemistry and pathological changes in the STZ induced DKD rats (Tang et al., Citation2020).

Targeted drug delivery strategy for podocytes

Nanoparticles

Podocytes locate on the outer side of the glomerular capillaries and they are reachable for the drug delivery carriers. Celastrol (CLT) is one of the active ingredients in tripterygium wilfordii, which have therapeutic effect in kidney disease but with organ toxicity. Wu et al designed a kind of peptides coupled CLT-phospholipid lipid nanoparticles (PC-PLNs) to deliver CLT to the podocytes and they can alleviate inflammation and reduce the CLT toxicity in chronic kidney disease (Wu et al., Citation2022). HDAC4 involves in the process of podocytes injury in DKD and HDAC4 siRNA has revealed a good prospect. DTsiANp/HDAC4 is designed to delivery HDAC4 siRNA to the podocytes in DKD rat model, after four weeks treatment, the glucose level, urinary albumin excretion ratio (UAER), MCs proliferation and glomerular sclerosis are all reduced significantly (Raval et al., Citation2019). Gold nanoparticles with the diameter of 50 nm can reduce the 24 h urinary albumin excretion rate, blood glucose, GBM thickness and foot process width in DKD rat models (Alomari et al., Citation2020). CoenzymeQ10 (CoQ10) is an antioxidant and may be a promising treatment for early-stage DKD. However, the low water solubility and nonspecific distribution of coenzyme Q10 limit its clinical application. In this condition, liposomes containing CoQ10 (CoQ10-lip) are equipped and combined with ultrasound microbubbles in DKD rat models, with a result of improved proteinuria and oxidative stress indexes (Yue et al., Citation2017).

Peptide

Inflammation involves in the pathogenesis of DKD. NF-κB is a bridge linking inflammation and hyperglycemia. A Cell-permeable peptide, designing with the inhibitor of kappa B kinase γ (IKKγ)/NF-κB essential modulator (NEMO)-binding domain (NBD), alleviates the albuminuria, kidney damage, podocyte loss and GBM thickness in DKD rat models (Opazo-Rios et al., Citation2020).

Polymers

miRNA30a, which is primarily responsible for podocyte homeostasis, is suppressed by hyperglycemic in DKD, leading the apoptosis of podocyte. Therefore, delivery exogenous miRNA30a to podocyte may ameliorate the podocyte injury. Cyclo (RGDfC)-gated polymeric-nanoplexes with dendrimer templates are can transfer the miRNA30a to the podocytes and alleviate the injury of podocytes in DKD (Raval et al., Citation2021).

Targeted drug delivery strategy for renal tubular epithelial cells (RTECs)

Nanoparticles

RETCs are vital target cells in the kidney targeted drug delivery systems for preventing renal fibrosis of DKD. PLGA-Gypenoside (Gyp) XLIX nanoparticles can effectively reduce collagen deposition and inhibit renal fibrosis in RETCs (HK2) cells stimulated by TGF-β (Q Liu et al., Citation2021). FITC labeled renal tubular-targeting peptide modified PLGA-PEG nanoparticles are used to transfer asiatic acid, which exhibits renal protective and anti-fibrosis effects (He et al., Citation2020). Pax2 gene, associating with interstitial fibrosis, is expressed in RTECs, and polyethyleneimine nanoparticles are employed to deliver Pax2-siRNA to RTECs, which can ameliorate the tubular damage and interstitial fibrosis (Li et al., Citation2012). Autophagy has been reported to be involved in the albuminuria caused renal tubular injury, and Fe3O4 magnetic nanoparticles are related to the autophagy. Fe3O4 magnetic albumin nanoparticles can alleviate renal tubular injury and delay the development of tubulointerstitial fibrosis (L Liu et al., Citation2021).

Liposome

Heat shock protein 72 (HSP72) is a stress-inducible protein capable of protecting cells from ischemic injury. Liposomes containing HSP72 are transferred in the renal tubular cells (LLC-PK1), which prevent the activation and translocation of NF-κB, and the apoptosis of renal tubular cells (Meldrum et al., Citation2003).

Discussion and conclusion

We conclude the advantages and disadvantages of different kinds of targeted drug delivery systems in . At present, researches on targeted drug delivery systems for DKD are almost in the early stages, and most of them have significant therapeutic effects and reduced side effects in animal models or cell series. But there are still several limitations. The first is biosecurity, as the toxicity of most non-biodegradable carriers is unclear. The second is the clinical efficacy. Most of the research data is obtained from animals and cells, which is different from the actual patients. The third is the stability of the transporter. Producing large-scale stable high-quality NPs and liposomes remains difficult. In this circumstance, targeted drug delivery systems with nontoxic, biodegradable and non-immunogenic characteristics may be facilitate by the transition from laboratory to clinical application.

Table 2. The advantages and disadvantages of different kinds of targeted drug delivery systems.

Authors’ contributions

LH conceived of the topic for this review. XC wrote the manuscript. All authors provided intellectual input to the editorial.

Ethical standard statement

This review was approved by the Ethics committee of the Second Xiangya Hospital, Central South University.

Disclosure statement

No potential conflict of interest was reported by the authors.

Additional information

Funding

This work was supported by Hunan Provincial Natural Science Foundation for Outstanding Youth (No. 2022JJ10093), the Scientific Research Fund of Hunan Provincial Health Commission (B202303056777), Natural Science Foundation of China (81870500), S&T Program of Hebei (21377753 D) and Hebei Provincial Medical Science Project Research (20221078).

References

  • Ahangarpour A, Oroojan AA, Khorsandi L, et al. (2019). Antioxidant, anti-apoptotic, and protective effects of myricitrin and its solid lipid nanoparticle on streptozotocin-nicotinamide-induced diabetic nephropathy in type 2 diabetic male mice. Iran J Basic Med Sci 22:1–11.
  • Albert A. (1958). Chemical aspects of selective toxicity. Nature 182:421–2.
  • Alomari G, Al-Trad B, Hamdan S, et al. (2020). Gold nanoparticles attenuate albuminuria by inhibiting podocyte injury in a rat model of diabetic nephropathy. Drug Deliv Transl Res 10:216–26.
  • Araujo F, Shrestha N, Gomes MJ, et al. (2016). In vivo dual-delivery of glucagon like peptide-1 (GLP-1) and dipeptidyl peptidase-4 (DPP4) inhibitor through composites prepared by microfluidics for diabetes therapy. Nanoscale 8:10706–13.
  • Asgeirsdottir SA, Zwiers PJ, Morselt HW, et al. (2008). Inhibition of proinflammatory genes in anti-GBM glomerulonephritis by targeted dexamethasone-loaded AbEsel liposomes. Am J Physiol Renal Physiol 294:F554–561.
  • Ashique S, Sandhu NK, Chawla V, Chawla PA. (2021). Targeted drug delivery: trends and perspectives. Curr Drug Deliv 18:1435–55.
  • Barrera-Chimal J, Lima-Posada I, Bakris GL, Jaisser F. (2022). Mineralocorticoid receptor antagonists in diabetic kidney disease - mechanistic and therapeutic effects. Nat Rev Nephrol 18:56–70.
  • Barro L, Hsiao JT, Chen CY, et al. (2021). Cytoprotective effect of liposomal puerarin on high glucose-induced injury in rat mesangial cells. Antioxidants (Basel) 10:1177.
  • Berillo D, Yeskendir A, Zharkinbekov Z, et al. (2021). Peptide-based drug delivery systems. Medicina (Kaunas) 57:1209.
  • Blanco E, Shen H, Ferrari M. (2015). Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol 33:941–51.
  • Brosius FC, Tuttle KR, Kretzler M. (2016). JAK inhibition in the treatment of diabetic kidney disease. Diabetologia 59:1624–7.
  • Byrne JD, Betancourt T, Brannon-Peppas L. (2008). Active targeting schemes for nanoparticle systems in cancer therapeutics. Adv Drug Deliv Rev 60:1615–26.
  • Chauhan VP, Popovic Z, Chen O, et al. (2011). Fluorescent nanorods and nanospheres for real-time in vivo probing of nanoparticle shape-dependent tumor penetration. Angew Chem Int Ed Engl 50:11417–20.
  • Chen D, Han S, Zhu Y, et al. (2018). Kidney-targeted drug delivery via rhein-loaded polyethyleneglycol-co-polycaprolactone-co-polyethylenimine nanoparticles for diabetic nephropathy therapy. Int J Nanomed 13:3507–27.
  • Chen Z, Peng H, Zhang C. (2020a). Advances in kidney-targeted drug delivery systems. Int J Pharm 587:119679.
  • Chen Z, Li W, Shi L, et al. (2020b). Kidney-targeted astaxanthin natural antioxidant nanosystem for diabetic nephropathy therapy. Eur J Pharm Biopharm 156:143–54.
  • Choi CH, Zuckerman JE, Webster P, Davis ME. (2011). Targeting kidney mesangium by nanoparticles of defined size. Proc Natl Acad Sci U S A 108:6656–61.
  • Christensen EI, Birn H, Storm T, et al. (2012). Endocytic receptors in the renal proximal tubule. Physiology (Bethesda) 27:223–36.
  • Comai G, Malvi D, Angeletti A, et al. (2019). Histological evidence of diabetic kidney disease precede clinical diagnosis. Am J Nephrol 50:29–36.
  • Dai Q, Chen N, Zeng L, et al. (2021). Clinical features of and risk factors for normoalbuminuric diabetic kidney disease in hospitalized patients with type 2 diabetes mellitus: a retrospective cross-sectional study. BMC Endocr Disord 21:104.
  • Dai W, Lu H, Chen Y, et al. (2021). The loss of mitochondrial quality control in diabetic kidney disease. Front Cell Dev Biol 9:706832.
  • Dalwadi C, Patel G. (2015). Application of nanohydrogels in drug delivery systems: recent patents review. Recent Pat Nanotechnol 9:17–25.
  • DeFronzo RA, Reeves WB, Awad AS. (2021). Pathophysiology of diabetic kidney disease: impact of SGLT2 inhibitors. Nat Rev Nephrol 17:319–34.
  • Deshayes S, Morris MC, Divita G, Heitz F. (2005). Cell-penetrating peptides: tools for intracellular delivery of therapeutics. Cell Mol Life Sci 62:1839–49.
  • Dou L, Jourde-Chiche N. (2019). Endothelial toxicity of high glucose and its by-products in diabetic kidney disease. Toxins (Basel) 11:578.
  • D’Souza B, Bhowmik T, Uddin MN, et al. (2015). Development of beta-cyclodextrin-based sustained release microparticles for oral insulin delivery. Drug Dev Ind Pharm 41:1288–93.
  • Duan S, Lu F, Song D, et al. (2021). Current challenges and future perspectives of renal tubular dysfunction in diabetic kidney disease. Front Endocrinol (Lausanne) 12:661185.
  • Ebrahim N, Ahmed IA, Hussien NI, et al. (2018). Mesenchymal stem cell-derived exosomes ameliorated diabetic nephropathy by autophagy induction through the mTOR signaling pathway. Cells 7:226.
  • El Mokadem M, Abd El Hady Y, Aziz A. (2020). A prospective single-blind randomized trial of ramipril, eplerenone and their combination in type 2 diabetic nephropathy. Cardiorenal Med 10:392–401.
  • Farshbaf M, Davaran S, Zarebkohan A, et al. (2018). Significant role of cationic polymers in drug delivery systems. Artif Cells Nanomed Biotechnol 46:1872–91.
  • Fleischmann D, Harloff M, Figueroa SM, et al. (2021). Targeted delivery of soluble guanylate cyclase (sGC) activator cinaciguat to renal mesangial cells via virus-mimetic nanoparticles potentiates anti-fibrotic effects by cGMP-mediated suppression of the TGF-beta pathway. Int J Mol Sci 22(5):2557.
  • Fox CS, Matsushita K, Woodward M, Chronic Kidney Disease Prognosis Consortium, et al. (2012). Associations of kidney disease measures with mortality and end-stage renal disease in individuals with and without diabetes: a meta-analysis. Lancet 380:1662–73.
  • Garg P. (2018). A review of podocyte biology. Am J Nephrol 47:3–13.
  • Gerstein HC, Sattar N, Rosenstock J, AMPLITUDE-O Trial Investigators, et al. (2021). Cardiovascular and renal outcomes with efpeglenatide in type 2 diabetes. N Engl J Med 385:896–907.
  • Gonçalves C, Pereira P, Gama M. (2010). Self-assembled hydrogel nanoparticles for drug delivery applications. Materials 3:1420–60.
  • Gregoriadis G, Perrie Y. (2010). Liposomes. In: Encyclopedia of life sciences. Chicheste: John Wiley & Sons, Ltd, 2656.
  • Guimaraes D, Cavaco-Paulo A, Nogueira E. (2021). Design of liposomes as drug delivery system for therapeutic applications. Int J Pharm 601:120571.
  • Gutierrez-Millan C, Calvo Díaz C, Lanao JM, Colino CI. (2021). Advances in exosomes-based drug delivery systems. Macromol Biosci 21:e2000269.
  • Haas M, Moolenaar F, Meijer DK, de Zeeuw D. (2002). Specific drug delivery to the kidney. Cardiovasc Drugs Ther 16:489–96.
  • Han F, Xue M, Chang Y, et al. (2017). Triptolide suppresses glomerular mesangial cell proliferation in diabetic nephropathy is associated with inhibition of PDK1/Akt/mTOR pathway. Int J Biol Sci 13:1266–75.
  • Han Y, Xu X, Tang C, et al. (2018). Reactive oxygen species promote tubular injury in diabetic nephropathy: the role of the mitochondrial ros-txnip-nlrp3 biological axis. Redox Biol 16:32–46.
  • He J, Chen H, Zhou W, et al. (2020). Kidney targeted delivery of asiatic acid using a FITC labeled renal tubular-targeting peptide modified PLGA-PEG system. Int J Pharm 584:119455.
  • Hernandez Becerra E, Quinchia J, Castro C, Orozco J. (2022). Light-triggered polymersome-based anticancer therapeutics delivery. Nanomaterials (Basel) 12:836.
  • Hirsjarvi S, Passirani C, Benoit JP. (2011). Passive and active tumour targeting with nanocarriers. Curr Drug Discov Technol 8:188–96.
  • Ismail R, Csoka I. (2017). Novel strategies in the oral delivery of antidiabetic peptide drugs - insulin, GLP 1 and its analogs. Eur J Pharm Biopharm 115:257–67.
  • Jha JC, Dai A, Garzarella J, Charlton A, et al. (2022). Independent of Renox, NOX5 promotes renal inflammation and fibrosis in diabetes by activating ROS-sensitive pathways. Diabetes 71:1282–98.
  • Jia X, Zang L, Pang P, et al. (2022). A study on the status of normoalbuminuric renal insufficiency among type 2 diabetes mellitus patients: a multicenter study based on a Chinese population. J Diabetes 14:15–25.
  • Jin J, Shi Y, Gong J, et al. (2019). Exosome secreted from adipose-derived stem cells attenuates diabetic nephropathy by promoting autophagy flux and inhibiting apoptosis in podocyte. Stem Cell Res Ther 10:95.
  • Joglekar M, Trewyn BG. (2013). Polymer-based stimuli-responsive nanosystems for biomedical applications. Biotechnol J 8:931–45.
  • Kato M, Natarajan R. (2019). Epigenetics and epigenomics in ­diabetic kidney disease and metabolic memory. Nat Rev Nephrol 15:327–45.
  • Kawanami D, Matoba K, Utsunomiya K. (2016). Signaling pathways in diabetic nephropathy. Histol Histopathol 31:1059–67.
  • Kean T, Thanou M. (2010). Biodegradation, biodistribution and toxicity of chitosan. Adv Drug Deliv Rev 62:3–11.
  • Khater SI, Mohamed AA, Arisha AH, Ebraheim LLM, et al. (2021). Stabilized-chitosan selenium nanoparticles efficiently reduce renal tissue injury and regulate the expression pattern of aldose reductase in the diabetic-nephropathy rat model. Life Sci 279:119674.
  • Klessens CQF, Zandbergen M, Wolterbeek R, et al. (2017). DHT IJ: macrophages in diabetic nephropathy in patients with type 2 diabetes. Nephrol Dial Transplant 32:1322–9.
  • Kristensen M, Birch D, Morck Nielsen H. (2016). Applications and challenges for use of cell-penetrating peptides as delivery vectors for peptide and protein cargos. Int J Mol Sci 17(2):185.
  • Kumar GS, Kulkarni A, Khurana A, et al. (2014). Selenium nanoparticles involve HSP-70 and SIRT1 in preventing the progression of type 1 diabetic nephropathy. Chem Biol Interact 223:125–33.
  • Lehner R, Wang X, Marsch S, Hunziker P. (2013). Intelligent nanomaterials for medicine: carrier platforms and targeting strategies in the context of clinical application. Nanomedicine 9:742–57.
  • Li G, Sun B, Li Y, et al. (2021). Small-molecule prodrug nanoassemblies: an emerging nanoplatform for anticancer drug delivery. Small 17:e2101460.
  • Li J, Zhang J, Lu Y, Zhang C. (2021). Effect of neutrophil-like melanin biomimic photothermal nanoparticles on glomerular mesangial cells in rats with gestational diabetic nephropathy. Colloid Interface Sci Commun 43:100458. (
  • Li L, Wu Y, Wang C, Zhang W. (2012). Inhibition of PAX2 gene expression by siRNA (polyethylenimine) in experimental model of obstructive nephropathy. Ren Fail 34:1288–96.
  • Li S, Zheng L, Zhang J, et al. (2021). Inhibition of ferroptosis by up-regulating Nrf2 delayed the progression of diabetic nephropathy. Free Radic Biol Med 162:435–49.
  • Liang Y, Duan L, Lu J, Xia J. (2021). Engineering exosomes for targeted drug delivery. Theranostics 11:3183–95.
  • Liu L, Pan X, Xie F, et al. (2022). Design, synthesis and biological activity evaluation of a series of bardoxolone methyl prodrugs. Bioorg Chem 124:105831.
  • Liu L, Xu Q, Zhang L, et al. (2021). Fe3O4 magnetic nanoparticles ameliorate albumin-induced tubulointerstitial fibrosis by autophagy related to Rab7. Colloids Surf B Biointerfaces 198:111470.
  • Liu Q, Chen X, Kan M, et al. (2021). Gypenoside XLIX loaded nanoparticles targeting therapy for renal fibrosis and its mechanism. Eur J Pharmacol 910:174501.
  • Luft FC, Aronoff GR, Evan AP, et al. (1982). Effects of moxalactam and cefotaxime on rabbit renal tissue. Antimicrob Agents Chemother 21:830–5.
  • Makhlough A, Kashi Z, Akha O, et al. (2014). Effect of spironolactone on diabetic nephropathy compared to the combination of spironolactone and losartan. Nephrourol Mon 6:e12148.
  • Manna K, Mishra S, Saha M, et al. (2019). Amelioration of diabetic nephropathy using pomegranate peel extract-stabilized gold nanoparticles: assessment of NF-kappaB and Nrf2 signaling system. Int J Nanomed 14:1753–77.
  • Meldrum KK, Burnett AL, Meng X, et al. (2003). Liposomal delivery of heat shock protein 72 into renal tubular cells blocks nuclear factor-kappaB activation, tumor necrosis factor-alpha production, and subsequent ischemia-induced apoptosis. Circ Res 92:293–9.
  • Merlin JPJ, Li X. (2021). Role of nanotechnology and their perspectives in the treatment of kidney diseases. Front Genet 12:817974.
  • Michos ED, Tuttle KR. (2021). GLP-1 receptor agonists in diabetic kidney disease. Clin J Am Soc Nephrol 16:1578–80.
  • Millotti G, Laffleur F, Perera G, et al. (2014). In vivo evaluation of thiolated chitosan tablets for oral insulin delivery. J Pharm Sci 103:3165–70.
  • Moon JY, Jeong KH, Lee TW, et al. (2012). Aberrant recruitment and activation of T cells in diabetic nephropathy. Am J Nephrol 35:164–74.
  • Mosenzon O, Leibowitz G, Bhatt DL, et al. (2017). Effect of Saxagliptin on Renal Outcomes in the SAVOR-TIMI 53 Trial. Diabetes Care 40:69–76.
  • Motiei M, Kashanian S, Lucia LA, Khazaei M. (2017). Intrinsic parameters for the synthesis and tuned properties of amphiphilic chitosan drug delivery nanocarriers. J Control Release 260:213–25.
  • Nagaishi K, Mizue Y, Chikenji T, et al. (2016). Mesenchymal stem cell therapy ameliorates diabetic nephropathy via the paracrine effect of renal trophic factors including exosomes. Sci Rep 6:34842.
  • National Kidney Foundation. (2012). KDOQI clinical practice guideline for diabetes and CKD: 2012 update. Am J Kidney Dis 60:850–86.
  • Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, García-Pérez J. (2011). Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol 7:327–40.
  • Neal B, Perkovic V, Mahaffey KW, CANVAS Program Collaborative Group, et al. (2017). Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med 377:644–57.
  • Opazo-Rios L, Plaza A, Sanchez Matus Y, et al. (2020). Targeting NF-kappaB by the cell-permeable nemo-binding domain peptide improves albuminuria and renal lesions in an experimental model of type 2 diabetic nephropathy. Int J Mol Sci 21(12):4225.
  • Ozturk-Atar K, Eroglu H, Calis S. (2018). Novel advances in targeted drug delivery. J Drug Target 26:633–42.
  • Pan-Pan Lin Q-LX, Chen L-W. (2021). A new one-dimensional copper(II) coordination polymer: crystal structure and treatment activity on diabetic nephropathy. In: Inorganic and nano-metal chemistry. Taylor & Francis.
  • Park W, Na K. (2015). Advances in the synthesis and application of nanoparticles for drug delivery. Wiley Interdiscip Rev Nanomed Nanobiotechnol 7:494–508.
  • Ponchiardi C, Mauer M, Najafian B. (2013). Temporal profile of diabetic nephropathy pathologic changes. Curr Diab Rep 13:592–9.
  • Qamar Z, Qizilbash FF, Iqubal MK, et al. (2019). Nano-based drug delivery system: recent strategies for the treatment of ocular disease and future perspective. Recent Pat Drug Deliv Formul 13:246–54.
  • Raval N, Gondaliya P, Tambe V, et al. (2021). Engineered nanoplex mediated targeted miRNA delivery to rescue dying podocytes in diabetic nephropathy. Int J Pharm 605:120842.
  • Raval N, Jogi H, Gondaliya P, et al. (2019). Method and its Composition for encapsulation, stabilization, and delivery of siRNA in Anionic polymeric nanoplex: an in vitro- in vivo assessment. Sci Rep 9:16047.
  • Ravindran S, Munusamy S. (2022). Renoprotective mechanisms of sodium-glucose co-transporter 2 (SGLT2) inhibitors against the progression of diabetic kidney disease. J Cell Physiol 237:1182–205.
  • Reidy K, Kang HM, Hostetter T, Susztak K. (2014). Molecular mechanisms of diabetic kidney disease. J Clin Invest 124:2333–40.
  • Saeedi P, Petersohn I, Salpea P, IDF Diabetes Atlas Committee, et al. (2019). Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract 157:107843.
  • Salva E, Turan SÖ, Akbuğa J. (2017). Inhibition of glomerular mesangial cell proliferation by siPDGF-B- and siPDGFR-beta-containing chitosan nanoplexes. AAPS PharmSciTech 18:1031–42.
  • Samsu N. (2021). Diabetic nephropathy: challenges in pathogenesis, diagnosis, and treatment. Biomed Res Int 2021:1497449.
  • Sanajou D, Ghorbani Haghjo A, Argani H, Aslani S. (2018). AGE-RAGE axis blockade in diabetic nephropathy: current status and future directions. Eur J Pharmacol 833:158–64.
  • Schellmann N, Deckert PM, Bachran D, et al. (2010). Targeted enzyme prodrug therapies. Mini Rev Med Chem 10:887–904.
  • Scindia Y, Deshmukh U, Thimmalapura PR, Bagavant H. (2008). Anti-alpha8 integrin immunoliposomes in glomeruli of lupus-susceptible mice: a novel system for delivery of therapeutic agents to the renal glomerulus in systemic lupus erythematosus. Arthritis Rheum 58:3884–91.
  • Shao J, Zaro J, Shen Y. (2020). Advances in exosome-based drug delivery and tumor targeting: from tissue distribution to intracellular fate. Int J Nanomed 15:9355–71.
  • Sharma G, Sharma AR, Nam JS, et al. (2015). Nanoparticle based insulin delivery system: the next generation efficient therapy for Type 1 diabetes. J Nanobiotechnol 13:74.
  • Sharma P, Blackburn RC, Parke CL, et al. (2011). Angiotensin-converting enzyme inhibitors and angiotensin receptor blockers for adults with early (stage 1 to 3) non-diabetic chronic kidney disease. Cochrane Database Syst Rev (10):CD007751.
  • Shen H, Wang W. (2021). Effect of glutathione liposomes on diabetic nephropathy based on oxidative stress and polyol pathway mechanism. J Liposome Res 31:317–25.
  • Shi GJ, Li Y, Cao QH, et al. (2019). In vitro and in vivo evidence that quercetin protects against diabetes and its complications: a systematic review of the literature. Biomed Pharmacother 109:1085–99.
  • Sila A, Ghlissi Z, Kamoun Z, et al. (2015). Astaxanthin from shrimp by-products ameliorates nephropathy in diabetic rats. Eur J Nutr 54:301–7.
  • Simons M, Raposo G. (2009). Exosomes–vesicular carriers for intercellular communication. Curr Opin Cell Biol 21:575–81.
  • Su Y, Xie Z, Kim GB, et al. (2015). Design strategies and applications of circulating cell-mediated drug delivery systems. ACS Biomater Sci Eng 1:201–17.
  • Tang L, Li K, Zhang Y, et al. (2020). Quercetin liposomes ameliorate streptozotocin-induced diabetic nephropathy in diabetic rats. Sci Rep 10:2440.
  • Tervaert TW, Mooyaart AL, Amann K, Renal Pathology Society, et al. (2010). Pathologic classification of diabetic nephropathy. J Am Soc Nephrol 21:556–63.
  • Tesch GH, Ma FY, Han Y, et al. (2015). ASK1 inhibitor halts progression of diabetic nephropathy in Nos3-deficient mice. Diabetes 64:3903–13.
  • Testa B. (2009). Prodrugs: bridging pharmacodynamic/pharmacokinetic gaps. Curr Opin Chem Biol 13:338–44.
  • Tong F, Liu S, Yan B, et al. (2017). Quercetin nanoparticle complex attenuated diabetic nephropathy via regulating the expression level of ICAM-1 on endothelium. Int J Nanomed 12:7799–813.
  • Tong Y, Zhang L, Gong R, et al. (2020). A ROS-scavenging multifunctional nanoparticle for combinational therapy of diabetic nephropathy. Nanoscale 12:23607–19.
  • Tuffin G, Waelti E, Huwyler J, et al. (2005). Immunoliposome targeting to mesangial cells: a promising strategy for specific drug delivery to the kidney. J Am Soc Nephrol 16:3295–305.
  • Visweswaran GR, Gholizadeh S, Ruiters MH, et al. (2015). Targeting rapamycin to podocytes using a vascular cell adhesion molecule-1 (VCAM-1)-harnessed SAINT-based lipid carrier system. PLoS One 10:e0138870.
  • Vodosek Hojs N, Bevc S, Ekart R, Hojs R. (2020). Oxidative stress markers in chronic kidney disease with emphasis on diabetic nephropathy. Antioxidants (Basel) 9:925.
  • Wadia JS, Dowdy SF. (2002). Protein transduction technology. Curr Opin Biotechnol 13:52–6.
  • Wang G, Li Q, Chen D, et al. (2019). Kidney-targeted rhein-loaded liponanoparticles for diabetic nephropathy therapy via size control and enhancement of renal cellular uptake. Theranostics 9:6191–208.
  • Wanner C, Inzucchi SE, Lachin JM, EMPA-REG OUTCOME Investigators, et al. (2016). Empagliflozin and progression of kidney disease in type 2 diabetes. N Engl J Med 375:323–34.
  • Wardani G, Nugraha J, Mustafa MR, Sudjarwo SA. (2022). Antioxidative stress and anti-inflammatory activity of fucoidan nanoparticles against nephropathy of streptozotocin-induced diabetes in rats. Evid Based Complement Alternat Med 2022:3405871.
  • Wiviott SD, Raz I, Bonaca MP, DECLARE–TIMI 58 Investigators, et al. (2019). Dapagliflozin and cardiovascular outcomes in type 2 diabetes. N Engl J Med 380:347–57.
  • Woraphatphadung T, Sajomsang W, Rojanarata T, et al. (2018). Development of chitosan-based pH-sensitive polymeric micelles containing curcumin for colon-targeted drug delivery. AAPS PharmSciTech 19:991–1000.
  • Wu L, Chen M, Mao H, et al. (2017). Albumin-based nanoparticles as methylprednisolone carriers for targeted delivery towards the neonatal Fc receptor in glomerular podocytes. Int J Mol Med 39:851–60.
  • Wu Q, Wang J, Wang Y, et al. (2022). Targeted delivery of celastrol to glomerular endothelium and podocytes for chronic kidney disease treatment. Nano Res 15:3556–68.
  • Xu L, Liang HW, Yang Y, Yu SH. (2018). Stability and reactivity: positive and negative aspects for nanoparticle processing. Chem Rev 118:3209–50.
  • Yang C, Chen XC, Li ZH, et al. (2021). SMAD3 promotes autophagy dysregulation by triggering lysosome depletion in tubular epithelial cells in diabetic nephropathy. Autophagy 17:2325–44.
  • Yang D, Livingston MJ, Liu Z, et al. (2018). Autophagy in diabetic kidney disease: regulation, pathological role and therapeutic potential. Cell Mol Life Sci 75:669–88.
  • Yang X, Mou S. (2017). Role of immune cells in diabetic kidney disease. Curr Gene Ther 17:424–33.
  • Yin W, Jiang Y, Xu S, et al. (2019). Protein kinase C and protein kinase A are involved in the protection of recombinant human glucagon-like peptide-1 on glomeruli and tubules in diabetic rats. J Diabetes Investig 10:613–25.
  • You YH, Quach T, Saito R, et al. (2016). Metabolomics reveals a key role for fumarate in mediating the effects of NADPH Oxidase 4 in diabetic kidney disease. J Am Soc Nephrol 27:466–81.
  • Yuan ZX, Jia L, Lim LY, et al. (2017). Renal-targeted delivery of triptolide by entrapment in pegylated TRX-20-modified liposomes. Int J Nanomed 12:5673–86.
  • Yue T, Xu HL, Chen PP, et al. (2017). Combination of coenzyme Q10-loaded liposomes with ultrasound targeted microbubbles destruction (UTMD) for early theranostics of diabetic nephropathy. Int J Pharm 528:664–74.
  • Zhang D, Gava AL, Van Krieken R, et al. (2019). The caveolin-1 regulated protein follistatin protects against diabetic kidney disease. Kidney Int 96:1134–49.
  • Zhang H, Schin M, Saha J, et al. (2010). Podocyte-specific overexpression of GLUT1 surprisingly reduces mesangial matrix expansion in diabetic nephropathy in mice. Am J Physiol Renal Physiol 299:F91–98.
  • Zhang H, Zhang S, Wang L, et al. (2018). Chitooligosaccharide guanidine inhibits high glucose-induced activation of DAG/PKC pathway by regulating expression of GLUT2 in type 2 diabetic nephropathy rats. J Funct Foods 41:41–7.
  • Zhang L, Long J, Jiang W, et al. (2016). Trends in chronic kidney disease in China. N Engl J Med 375:905–6.
  • Zhou TT, Zhao T, Ma F, et al. (2019). Small molecule IVQ, as a prodrug of gluconeogenesis inhibitor QVO, efficiently ameliorates glucose homeostasis in type 2 diabetic mice. Acta Pharmacol Sin 40:1193–204.
  • Zhou X, Wang B, Zhu L, Hao S. (2012). A novel improved therapy strategy for diabetic nephropathy: targeting AGEs. Organogenesis 8:18–21.
  • Zhou Y, Chi J, Huang Y, et al. (2021). Efficacy and safety of endothelin receptor antagonists in type 2 diabetic kidney disease: a systematic review and meta-analysis of randomized controlled trials. Diabet Med 38:e14411.
  • Zinman B, Wanner C, Lachin JM, EMPA-REG OUTCOME Investigators, et al. (2015). Empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med 373:2117–28.