Diabetes affects a large population of people worldwide. The global diabetes prevalence in 20–79 year olds in 2021 was estimated to be 10.5% (536.6 million people).1–6 Diabetic kidney disease (DKD) is defined as persistent albuminuria and/or the decrease of estimated glomerular filtration rate attributed to diabetes.7 It develops in approximately 40% of patients who are diabetic and is the leading cause of chronic kidney disease worldwide. And it is one of the most common microvascular complications of diabetes, and also the main cause of end-stage renal disease (ESRD) and global renal failure, which seriously affects the quality of life of patients.8–10 With the continuous increase of the global prevalence of diabetes, the incidence rate of DKD is also increasing year by year, which has brought a heavy burden to patients and the society.11,12
Dynamic feedback mechanisms are biological control systems where outputs (eg, molecular signals) recursively modulate their own production. In healthy organisms, feedback regulation is a mechanism used by organisms to maintain homeostasis, which is the stable internal environment required for optimal function. Feedback regulation is divided into negative feedback and positive feedback. Negative feedback can counteract or reduce the impact of stimuli. Positive feedback amplifies changes rather than reversing them. In DKD, the latter is more common.
In DKD, after the homeostasis is disrupted, pathogenic factors continuously promote the disease process through forming pathological loops that amplify damage. While previous reviews have focused on isolated pathways (eg, hyperglycemia or fibrosis), this work uniquely integrates multi-scale feedback networks to reveal how cross-tier interactions create self-sustaining disease progression. Our synthesis provides the first hierarchical framework categorizing DKD feedback loops into three tiers: systemic (eg, hyperglycemia-hypertension crosstalk), cellular (eg, apoptosis-ferroptosis synergy), and molecular (eg, AGE/RAGE-TGF-β axis). This approach uncovers previously overlooked amplification nodes that synchronize damage across biological scales.
A deep understanding of the feedbacks in the occurrence and development of DKD is of great significance for revealing the disease process and finding new therapeutic targets. Crucially, the hierarchical nature of these feedback systems implies that effective interventions must simultaneously target multiple regulatory tiers. For instance, breaking the systemic triad (hyperglycemia-hypertension-proteinuria) may require combined glycemic control and RAAS blockade, while molecular-level therapies could disrupt the feedforward amplification between oxidative stress and epigenetic dysregulation. This paradigm shift toward multi-tiered targeting forms the foundation for the therapeutic strategies discussed in subsequent sections.
DKD and Feedback of the SystemIt is well-established that feedback regulation at systemic level plays a key role in the development and progress of DKD.13,14 Macroscopically, sustained hyperglycemia and changes in renal hemodynamics lead to an increase in renal microvascular pressure. If this elevation is not strictly controlled, it usually leads to a gradual decline in kidney function. As the pressure inside the glomerulus increases, the structural integrity of the glomerulus is compromised, resulting in damage to the glomerular capillary walls. This type of damage causes protein leak from the blood into the urine, resulting in proteinuria.15 As proteinuria progresses to a more serious form, such as proteinuria within the scope of nephrosis, it will lead to severe water and sodium retention, and damage the body’s fluid balance. Meanwhile, impaired kidney function may result in the inability to effectively eliminate excess sodium and water, leading to fluid accumulation in the body. This accumulation will increase the blood volume and pressure inside the blood vessels, thereby exacerbating hypertension.16,17 A vicious cycle has been formed, where elevated blood pressure further damages the kidneys, increases kidney pressure, and worsens the condition. Therefore, early intervention is crucial for breaking this cycle and preventing disease progression.
Feedback in Cell Death (Figure 1)In kidney injury, a complex interplay of various cell death mechanisms often coexist, disrupting distinct cellular functions and signaling pathways, thereby triggering cell death through diverse processes. Although multiple pathways may be involved, several predominant routes of cell death are typically observed. These primary pathways not only rely on the signals that initiate the cell death cascade but also on the specific cell types that respond to these signals, as well as individual genetic and environmental factors. Moreover, these different forms of cell death are interconnected, influencing each other in a complex network.18,19
Figure 1 Feedback in Cell Death. High blood glucose, as a triggering factor, triggers a series of reactions through different pathways. Apoptosis is induced through the mitochondrial pathway, endoplasmic reticulum stress pathway, and death receptor pathway, respectively. These pathways activate ROS generation. High blood sugar can also trigger other forms of cell death. Autophagy exhibits dual-phase regulation in DKD progression: serving a protective role during early stages while paradoxically promoting apoptotic processes in advanced disease stages. In addition, there are other types of cell death mechanisms, such as ferroptosis and inflammatory mediator mediated pyroptosis. All these processes are involved in the progression of the disease and form a positive feedback loop, leading to gradual deterioration of kidney function and ultimately potentially causing ESRD.
Abbreviations: ESRD, end-stage renal disease; DAMPs, damage-associated molecular patterns; ROS, reactive oxygen species.
During the early stage of DKD, without timely preventive interventions, a hyperglycemic environment can trigger an inflammatory response and lead to the release of inflammasomes and damage-associated molecular patterns (DAMPs). Under the control of inflammatory cytokines, such as interleukins and tumor necrosis factor-alpha (TNF-α), a form of programmed cell death known as pyroptosis occurs.20 As the disease progresses and cellular damage accumulates, inflammatory factors are continuously secreted, exacerbating the inflammatory milieu as a positive feedback.21
Simultaneously, persistent production of reactive oxygen species (ROS) leads to mitochondrial dysfunction, causing the release of cytochrome c and apoptosis-inducing factors into the cytoplasm of kidney cells, promoting mitochondrial-mediated apoptosis. This is accompanied by excessive endoplasmic reticulum (ER) stress and activation of the death receptor pathway mediated by TNF-α, further accelerating the apoptotic program of cells.22,23
Mitochondrial damage results in depletion of intracellular glutathione, leading to an accumulation of ROS. Increased expression of the transferrin receptor allows more ferric iron (Fe3+) to enter the cell. Ferric iron is then reduced to ferrous iron (Fe2+) by iron reductases, and through the Fenton reaction, a positive feedback loop is established that amplifies ROS production, disturbing iron homeostasis and triggering ferroptosis, a form of regulated cell death characterized by lipid peroxidation.24
In the early stage of DKD, the process of autophagy can help clear excess or damaged organelles and protein aggregates. However, with the positive feedback loop involving proximal tubular injury, mitochondrial dysfunction, and an inflammatory response, autophagy undergo a transition from a protective mechanism to one that promotes cell death, particularly apoptosis.25,26
As mentioned above, mechanisms of different programmed cell death (eg, apoptosis, pyroptosis, ferroptosis and autophagy) together form a large positive feedback regulation, continuously exacerbating cell death and worsening the patients’ condition. Many DKD patients do not receive effective intervention in the early stage, and with the amplification of positive feedback effects, they may even develop ESRD. Targeting the induction of specific cell death mode or inhibiting unnecessary cell death by modulating key feedback loops could represent promising therapeutic strategies for kidney diseases. By understanding and intervening in these complex pathways, it may be possible to develop novel treatments aimed at preserving renal function and preventing the progression of kidney disease.
Feedback in Cell Cycle Disorder (Figure 2)In the normal adult kidney, most kidney cells are primarily in a quiescent state, maintaining a stable and balanced environment essential for proper renal function.27 When DKD occurs, hemodynamic changes, high blood glucose levels, inflammatory factors, and other stimuli impact virtually all types of kidney cells. Under mild stimulation, cell cycle regulation may provide negative feedback to aid in the repair of kidney damage. However, a prolonged hyperglycemic condition often leads to cell cycle dysregulation, exacerbating kidney damage through a positive feedback loop involving cellular injury and metabolic disturbances.28
Figure 2 Feedback in cell cycle disorder. In the process of DKD, various major cell types exhibit different pathological characteristics and perform different functions. Podocytes undergo mitotic catastrophe, leading to proteinuria, while paracrine VEGF mediates the differentiation process of GECs. In the early stage, it promotes neovascularization, and in the late stage, with VEGF secretion degradations, leading to thinning of the glomerular capillaries and renal fibrosis. Over proliferation of MCs leads to glomerulosclerosis. TECs undergo EMT and fibroblast activation, manifested as the appearance of myofibroblasts, which progress to renal fibrosis.
Abbreviations: DKD, diabetic kidney disease; MCs, mesangial cells; GECs, glomerular endothelial cells; VEGF, Vascular endothelial growth factor; TECs, tubular epithelial cells; EMT, epithelial-mesenchymal transition.
In the glomerulus, podocytes, which are critical for maintaining the filtration barrier, are often stimulated by hyperglycemia to re-enter the cell cycle, a process that can promote mitotic catastrophe.29 Persistent or severe podocyte damage leads to cell detachment, barrier dysfunction, and ultimately proteinuria.30 Podocyte loss disrupts the glomerular filtration barrier, allowing proteins to pass into the urine, which is a hallmark of early DKD.
Mesangial cells (MCs), another important component of the glomerulus, are also activated and undergo excessive proliferation. They produce extracellular matrix (ECM) components and continuously deposit them, contributing to glomerulosclerosis.31 This accumulation of ECM components alters the glomerular structure, leading to thickening of the basement membrane and reduced filtration capacity.
The migration, proliferation, and differentiation of glomerular endothelial cells (GECs) are tightly regulated by the vascular endothelial growth factor A (VEGF-A)/vascular endothelial growth factor receptor 2 (VEGFR-2) system. Vascular endothelial growth factor (VEGF) is secreted by podocytes and is initially upregulated in the early stage of DKD, promoting neovascularization.15 However, as the disease progresses and podocytes are lost, VEGF secretion decreases, leading to thinning of the glomerular capillaries and renal fibrosis.16 This reduction in VEGF secretion can contribute to the ischemia and hypoxia in the late stage of DKD.
In the renal tubules, tubular epithelial cells (TECs) may undergo epithelial-mesenchymal transition (EMT), a process where they lose their epithelial characteristics and acquire mesenchymal features. Meanwhile, fibroblasts are activated.32 This transition contributes to fibroblast activation and the deposition of ECM components. Alongside the release of profibrotic factors, such as transforming growth factor-beta (TGF-β), the development of renal fibrosis is accelerated. The accumulation of ECM components and the transformation of TECs and fibroblasts into myofibroblasts contribute to the progressive decline in renal function and the development of interstitial fibrosis.
Overall, changes in cell cycle regulation, cell behavior, and ECM deposition play a critical role in the pathogenesis of DKD, forming a positive feedback loop that continuously exacerbates cellular morphological and functional abnormalities, which highlights the need for targeted therapies aimed at restoring normal cell cycle dynamics and inhibiting fibrotic processes.
Feedbacks in Molecular Level Molecular Mechanism of Acute Phase (Figure 3)The acute phase of DKD mainly focuses on mechanisms related to acute cellular injury, emphasizing inflammation, stress, apoptosis, and other aspects. Feedback regulation in this case plays a crucial role in the onset and progression of the disease. Research has shown that under hyperglycemic conditions, protein kinase C (PKC) is activated,33,34 which not only directly participates in oxidative stress-induced cell damage, leading to the accumulation of intracellular ROS and advanced glycation end products (AGEs), but also regulates vascular function, increases vascular permeability, and affects the expression of important vasoactive substances.13
Figure 3 Molecular mechanism of acute phase. The binding of RAGE to various ligands (including AGEs, HMGB-1, S100, among which AGEs can also directly lead to the accumulation of sorbitol and fructose through the polyol pathway, consuming intracellular antioxidants) activates numerous downstream molecules. The most crucial one among them is ROS. Next, various signaling pathways are utilized, such as NF-κB, PI3K/Akt, MAPK/ERK, JAK/STAT, Nrf2, and p53/p52. By regulating with mainly positive feedbacks, the disease process is affected and downstream pro inflammatory factors, adhesion molecules, and profibrotic mediators are released, which participate in promoting fibrosis and inflammatory response, cell adhesion, angiogenesis, and thrombogenesis. These feedbacks may lead to worsening of the condition or even progression to ESRD.
Abbreviations: AGEs, advanced glycation end products; AR, aldose reductase; RAGE, receptor for advanced glycation end products; ROS, reactive oxygen species; GSH, Glutathione; NADPH, Nicotinamide Adenine Dinucleotide Phosphate; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; MAPK, mitogen-activated protein kinases; ERK, extracellular signal-regulated kinases; JAK, Janus kinase; STAT, signal transducer and activator of transcription; TNF-α, tumor necrosis factor-alpha; IL-6, interleukin-6; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; Ang II, angiotensin II; TGF-β, transforming growth factor-beta; VEGF, Vascular endothelial growth factor; CTGF, connective tissue growth factor; LOX, lysyl oxidase.
The polyol pathway, a minor pathway in normal glucose metabolism, becomes overactive during sustained hyperglycemia. Glucose is reduced to sorbitol by aldose reductase (AR), and this sorbitol is further converted to fructose by sorbitol dehydrogenase. During chronic hyperglycemia, the accumulation of sorbitol and fructose leads to osmotic stress within cells and can undergo non-enzymatic glycation reactions with proteins, forming AGEs through a positive feedback mechanism. This process consumes large amounts of antioxidants and increases oxidative stress, further exacerbating the damaging effects of hyperglycemia.35,36
The latest evidence suggests that the receptor for advanced glycation end products (RAGE) plays a central role in the occurrence of DKD. In patients with DKD, high blood glucose levels stimulate the formation of AGEs, which are primarily metabolized by the kidneys and tend to deposit in renal tissue. Concurrently, the expression of RAGE is upregulated,13,14 and the downstream signaling pathways mediated by the AGE/RAGE interaction are activated. These include the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB),37 phosphoinositide 3-kinase/protein kinase B (PI3K/Akt),38 mitogen-activated protein kinases/extracellular signal-regulated kinases (MAPK/ERK),39 and Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathways.36,40,41
These signaling cascades activate a variety of pro-inflammatory factors (inducible nitric oxide synthase,41 TNF-α,42 interleukin-6 [IL-6],43 NOD-like receptor family pyrin domain containing 3 inflammasome [NLRP3]44,45), adhesion molecules (intercellular adhesion molecule-1 [ICAM-1], vascular cell adhesion molecule-1 [VCAM-1]),46,47 and profibrotic mediators (angiotensin II [Ang II],48 connective tissue growth factor [CTGF],49 VEGF,50 TGF-β,51 lysyl oxidase [LOX])52 Additionally, the expression of cell cycle regulatory proteins (p53,53 p2154,55) is altered, contributing to cellular senescence and dysfunction.
At the same time, these pathways interact with each other, inducing oxidative stress, inflammatory reactions, cellular senescence, and further increasing the formation of AGEs.56,57 Throughout this process, there is also a protective factor, nuclear factor erythroid 2-related factor 2 (Nrf2), which has negative feedback mechanism and regulates the disease progression of DKD by activating antioxidant response elements (ARE), inhibiting inflammatory responses, and regulating fibrosis processes.58,59
As the disease progresses, symptoms such as thickening of the glomerular basement membrane, accumulation of ECM, glomerulosclerosis, vascular damage, and interstitial fibrosis worsen. The feedback loop composed of the AGE/RAGE axis and its downstream signaling pathways continues to exacerbate the condition of DKD, forming a vicious cycle that may ultimately lead to ESRD.60,61
This complex interplay of signaling pathways and feedback mechanisms underscores the importance of targeting these processes in the development of novel therapeutic strategies for DKD.
Molecular Mechanism of Chronic Phase (Figure 4)As previously discussed, factors such as hyperglycemia and hypertension damage the intrinsic cells of the kidney, including TECs, podocytes, and MCs. The acute phase features are mainly manifested in inflammation, stress, apoptosis and other aspects. The molecular mechanisms that lead to sustained disease progression also include chronic phase, mainly manifested as abnormal tissue repair, such as fibrosis, etc. This damage leads to the release of profibrotic factors, cytokines, and growth factors thereby initiating the process of chronic fibrosis. The process involves the activation and interaction of multiple signaling pathways, primarily including TGF-β,51,62 MAPK,39 Wnt/β-catenin,63,64 PI3K/Akt,38 JAK/STAT,22 and Notch pathways.65 While the pro-fibrotic role of TGF-β considered as a key mediator in renal fibrosis and induces renal scarring. Studies from gene knockout mice demonstrate that TGF-β1 acts by stimulating its downstream Smads to diversely regulate kidney injury. In the context of renal fibrosis and inflammation, Smad3 is pathogenic,66 while Smad2 and Smad7 are protective.67,68
Figure 4 Molecular mechanism of chronic phase. During the chronic phase, it mainly involves fibrosis of the kidneys. Activation of multiple pathways such as TGF-β, MAPK, Wnt/β-catenin, PI3K/Akt, JAK/STAT, and Notch pathways through TECs, podocytes, and MCs initiates the process of chronic fibrosis. During this process, the cell phenotype gradually undergoes a transformation. Renal tubular epithelial cells undergo EMT, endothelial cells undergo EndoMT, and fibroblasts and pericytes are activated. These cells are transformed into myofibroblasts, and ECM is deposited in large quantities to promote the fibrosis process, which aggravates the damage of diabetes nephropathy.
Abbreviations: TGF-β, transforming growth factor-beta; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; JAK, Janus kinase; STAT, signal transducer and activator of transcription; IL-6, interleukin-6; EMT, epithelial-mesenchymal transition; EndoMT, endothelial-mesenchymal transition; NICD, Notch intracellular domain; ECM, extracellular matrix; ESRD, end-stage renal disease.
Gradually, as kidney cell damage intensifies, cell phenotype transformations occur. Renal tubular epithelial cells undergo EMT,32,69 a process where they lose their epithelial characteristics and acquire mesenchymal features, such as increased migratory ability and the ability to secrete ECM components. Similarly, endothelial cells undergo endothelial-mesenchymal transition (EndoMT),53 which is a special EMT subset that occurs in endothelial cells and is like EMT, further contributing to the accumulation of ECM components. Fibroblasts and pericytes are also activated, participating in the fibrotic process.32,69
These pathological processes accelerate the irreversible formation of myofibroblasts, specialized cells that secrete large amounts of ECM components, including collagen and fibronectin. As ECM synthesis increases and degradation decreases, excessive ECM accumulates in the renal interstitium, leading to tissue hardening and structural remodeling. Accumulation of ECM components alters the normal architecture of the kidney, impairing its function and contributing to the development of fibrosis.
Intriguingly, certain self-limiting mechanisms may transiently mitigate damage. For instance, stress-induced tubular cell senescence may paradoxically suppress TGF-β1-mediated fibrosis in early-stage DKD through cell cycle arrest, though this protective adaptation is ultimately overwhelmed by persistent metabolic insults. Such similar negative feedback mechanisms may serve as potential protective factors for renal function.
On the other hand, the loss of functional nephrons leads to a positive feedback loop of symptoms such as cellular autophagy, inflammatory response, and proteinuria, further exacerbating kidney damage.70 Proteinuria indicates significant damage to the glomerular filtration barrier and is a hallmark of DKD progression. Eventually, as kidney function gradually declines, the disease enters the irreversible stage of renal fibrosis, leading to ESRD.
TreatmentAs mentioned above, there are many positive feedback mechanisms whereas negative feedback modes are limited in DKD, and there are intervention measures correspondingly based on the pathogenic effects of these regulatory mechanisms. It can be divided into treatments targeting AGEs/RAGE axis and treatments targeting fibrosis.
Targeting AGEs/RAGE AxisIn the context of DKD and aging kidneys, the aim is to block or alleviate oxidative stress, inflammatory response,71 and tissue damage caused by the interaction between AGEs and RAGE.
The Most Important Goal Is to Inhibit the Formation and Accumulation of AGEsAdopting a low-AGEs diet by reducing intake of high-sugar and high-temperature processed foods can effectively decrease exogenous AGEs intake. This is because ingested AGEs can accumulate in the gastrointestinal tract, thereby altering the gut microbiota, regulating immune signaling, and reducing antioxidant enzyme activity to induce inflammation.72,73 Natural anti-glycation agents derived from botanical sources, including Eucommia ulmoides Oliv74 and diphlorethohydroxycarmalol (DPHC),75 have shown efficacy in inhibiting AGEs formation. They inhibit the formation of AGEs and RAGE expression, reducing the damage to cells. Feeding mice with rapid peptide Maillard reaction products can optimize the abundance and diversity of the gut microbiota, inhibit pathogenic bacteria while increasing beneficial bacteria, reduce oxidative stress, and delay organ aging.76
In the Process of RAGE Signal Transduction, Timely Artificial Regulation Should Be Carried OutDeveloping specific RAGE antagonists or neutralizing antibodies that directly block AGEs-RAGE binding represents a promising strategy to mitigate downstream inflammatory and oxidative stress responses. Alternatively, small molecule inhibitors or other drugs can be used to reduce the transmission of downstream harmful signals. Drugs such as FPS-ZM1 are designed to selectively inhibit RAGE, which can protect the glomerular filtration barrier and improve the renal function of diabetic rats, and when combined with angiotensin receptor blockers (such as valsartan), they can more effectively reduce the activation of RAGE and its downstream NF-κB.77,78 Hepatic β-Hydroxy-β–methyl-glutaryl-Co-A (HMG-CoA) reductase inhibitors and ezetimibe may exert their effects via targeting intracellular ROS, NRP-1 function, and RAGE related genes (ie NF-κB, TGF-β, and MMP-2) to treat oxidative stress and tissue damage induced by AGEs in DKD.79 Fluorofenidone and osthole inhibit the PKC/NOX pathway80 and JAK2-STAT1/STAT3 signaling transduction,81 respectively.
Activate Protective Signaling PathwaysBy using compounds that can activate Nrf2/ARE, such as certain plant extracts and drugs, the antioxidant capacity of cells can be enhanced, oxidative stress caused by AGEs can be counteracted and their accumulation can be reduced, thus delaying the progression of DKD.82,83
Targeting FibrosisRenal fibrosis is a chronic pathological process involving multiple factors and pathways, which ultimately leads to tissue restructuring and functional decline through the interaction and feedback regulation of multiple signaling pathways.84,85 Accurately regulating these pathways and their interactions is beneficial for reversing the fibrosis process:
Use Renal Protective DrugsAngiotensin converting enzyme inhibitors or angiotensin receptor blockers partially interfere with Ang II mediated TGF-β expression, delay hyperglycemia-induced renal cell fibrosis, reduce proteinuria, and delay renal function decline.86 Metformin not only has the effect of lowering blood glucose levels, but also inhibits mammalian target of rapamycin, PI3K/AKT, and TGF-β pathways by activating AMP-activated kinase, regulating renal cell autophagy, restoring cell repair mechanisms, and reducing ER stress.87 Metformin also has great potential as a renal protective drug by reducing the expression of HIF-1α and improving renal cell hypoxia by lowering ATP levels and renal oxygen consumption.88
Use Signal Pathway InhibitorsDevelop specific inhibitors targeting signaling pathways such as TGF-β, MAPK, Wnt/β-catenin, PI3K/Akt, JAK/STAT, and Notch that promote fibrosis. Pirfenidone antagonizes the TGF-β and MAPK pathways to alleviate EMT and renal fibrosis.89 Triptolide, an inhibitor of the PI3K/Akt signaling pathway, can reduce the production of CTGF, fibronectin, and collagen through the interaction between miR-188-5p and phosphatase and tensin homolog, and slow down renal EMT.90 Oral administration of selective inhibitor Baricitinib can mitigate albuminuria, inhibit JAK1/JAK2, and treat DKD.91 TGF-β antibodies and Wnt/β-catenin signaling pathway inhibitors are used to reduce the production of fibrosis related proteins, decrease ECM deposition, and alleviate renal interstitial fibrosis.64,92–94
ProspectUnderstanding these feedback mechanisms is crucial for developing targeted therapies and improving the management of DKD. For example, interventions aimed at suppressing hypertension and proteinuria circulation can significantly slow down the progression of kidney injury. At the cellular level, targeting specific pathways involved in cell death, such as pyroptosis and ferroptosis, or alleviating autophagic dysfunction, and at the molecular level, inhibiting or activating specific feedback pathways may provide new therapeutic opportunities.
Looking ahead, it is necessary to continue research to elucidate the precise mechanisms behind these feedback loops, understand whether drugs targeting different feedback pathways interact, and identify new biomarkers that can predict disease progression. As exploration deepens, it is believed that new targets and therapeutic drugs will continue to emerge. With the advancement of genomics and proteomics, the development of personalized medical methods is expected to tailor treatment methods according to the needs of individual patients.
ConclusionIn conclusion, DKD progression is fundamentally driven by self-reinforcing feedback networks, which is a paradigm shift from traditional single-pathway models. This review provides three key advances over previous studies: (1) We establish the first hierarchical framework integrating systemic, cellular and molecular feedback loops in DKD pathogenesis; (2) We reveal how cross-tier amplification between acute inflammatory drivers (eg, AGE/RAGE axis) and chronic fibrotic processes creates irreversible disease momentum; (3) We propose time-sensitive therapeutic strategies targeting these networked feedback systems. For instance, early intervention combining RAGE antagonists with Nrf2 activators could break the acute-phase oxidative stress-inflammation cycle, while precision modulation of fibrotic pathways using TGF-β/JAK-STAT dual inhibitors may attenuate established fibrosis.
Our analysis identifies previously unrecognized therapeutic nodes, which enable two transformative clinical approaches: First, specific interventions for different stages - targeting AGEs/RAGE axis to suppressing the acute phase feedbacks before fibrosis establishment, then switching to inhibitors targeting signaling pathways such as TGF-β and JAK/STAT that promote fibrosis. Second, precision medicine map dominant feedback loops in individual patients, enabling tailored combinations like RAGE antagonists for predominant glycation stress or JAK inhibitors for inflammatory phenotypes.
Future research should focus on three frontiers: (1) Developing feedback-aware therapeutic indices that quantify network disruption while preserving physiological repair mechanisms; (2) Creating computational models simulating feedback loop interactions to predict treatment outcomes; (3) Validating novel combination therapies through clinical trials stratified by feedback pathway activation patterns. By redefining DKD as a network disorder of maladaptive feedbacks, this framework opens new avenues for intercepting disease progression through temporally and spatially coordinated interventions.
AbbreviationsDKD, diabetic kidney disease; ESRD, end-stage renal disease; DAMPs, damage-associated molecular patterns; TNF-α, tumor necrosis factor-alpha; ROS, reactive oxygen species; ER, endoplasmic reticulum; MCs, mesangial cells; ECM, extracellular matrix; GECs, glomerular endothelial cells; VEGF, Vascular endothelial growth factor; TECs, tubular epithelial cells; EMT, epithelial-mesenchymal transition; TGF-β, transforming growth factor-beta; PKC, protein kinase C; AGEs, advanced glycation end products; AR, aldose reductase; RAGE, receptor for advanced glycation end products; GSH, Glutathione; NADPH, Nicotinamide Adenine Dinucleotide Phosphate; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PI3K, phosphoinositide 3-kinase; Akt, protein kinase B; MAPK, mitogen-activated protein kinases; ERK, extracellular signal-regulated kinases; JAK, Janus kinase; STAT, signal transducer and activator of transcription; IL-6, interleukin-6; NLRP3, NOD-like receptor family pyrin domain containing 3 inflammasome; ICAM-1, intercellular adhesion molecule-1; VCAM-1, vascular cell adhesion molecule-1; Ang II, angiotensin II; CTGF, connective tissue growth factor; LOX, lysyl oxidase; Nrf2, nuclear factor erythroid 2-related factor 2; ARE, antioxidant response elements; EndoMT, endothelial-mesenchymal transition; NICD, Notch intracellular domain; DPHC, diphlorethohydroxycarmalol; HMG-CoA, Hepatic β-Hydroxy-β–methyl-glutaryl-Co-A.
Consent for PublicationAll authors agreed to publish the review.
Author ContributionsAll authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, visualization, project administration, or in all these areas. All authors took part in drafting, revising or critically reviewing the article. All authors gave final approval of the version to be published. All authors have agreed on the journal to which the article has been submitted. All authors agree to be accountable for all aspects of the work.
FundingThis work is funded by grants from Changjiang Scholar Program, Ministry of Education, China (PDJH202204).
DisclosureThe authors report no conflicts of interest in this work.
References1. Saeedi P, Petersohn I, Salpea P, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabet Res Clin Pract. 2019;157:107843. doi:10.1016/j.diabres.2019.107843
2. Sun H, Saeedi P, Karuranga S, et al. IDF Diabetes Atlas: global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabet Res Clin Pract. 2022;183:109119. doi:10.1016/j.diabres.2021.109119
3. Zelnick LR, Weiss NS, Kestenbaum BR, et al. Diabetes and CKD in the United States Population, 2009–2014. Clin J Am Soc Nephrol. 2017;12:1984–1990. doi:10.2215/CJN.03700417
4. Ma RCW. Epidemiology of diabetes and diabetic complications in China. Diabetologia. 2018;61:1249–1260. doi:10.1007/s00125-018-4557-7
5. Unnikrishnan R, Anjana RM, Mohan V. Diabetes mellitus and its complications in India. Nat Rev Endocrinol. 2016;12:357–370. doi:10.1038/nrendo.2016.53
6. Mbanya JCN, Motala AA, Sobngwi E, et al. Diabetes in Sub-Saharan Africa. Lancet. 2010;375:2254–2266. doi:10.1016/S0140-6736(10)60550-8
7. Thomas MC, Brownlee M, Susztak K, et al. Diabetic kidney disease. Nat Rev Dis Primers. 2015;1:15018. doi:10.1038/nrdp.2015.18
8. Groop P-H, Thomas MC, Moran JL, et al. The presence and severity of chronic kidney disease predicts all-cause mortality in type 1 diabetes. Diabetes. 2009;58:1651–1658. doi:10.2337/db08-1543
9. Orchard TJ, Secrest AM, Miller RG, Costacou T. In the absence of renal disease, 20 year mortality risk in type 1 diabetes is comparable to that of the general population: a report from the Pittsburgh Epidemiology of Diabetes Complications Study. Diabetologia. 2010;53:2312–2319. doi:10.1007/s00125-010-1860-3
10. Zhang J, Long M, Sun Z, et al. Association between Thymosin beta-4, acute kidney injury, and mortality in patients with sepsis: an observational cohort study. Int Immunopharmacol. 2021;101(Pt A):108167. doi:10.1016/j.intimp.2021.108167
11. Bommer C, Sagalova V, Heesemann E, et al. Global economic burden of diabetes in adults: projections from 2015 to 2030. Diabetes Care. 2018;41:963–970. doi:10.2337/dc17-1962
12. Einarson TR, Acs A, Ludwig C, Panton UH. Economic burden of cardiovascular disease in type 2 diabetes: a systematic review. Value Health. 2018;21:881–890. doi:10.1016/j.jval.2017.12.019
13. Taguchi K, Fukami K. RAGE signaling regulates the progression of diabetic complications. Front Pharmacol. 2023;14:1128872. doi:10.3389/fphar.2023.1128872
14. Wu XQ, Zhang DD, Wang YN, et al. AGE/RAGE in diabetic kidney disease and ageing kidney. Free Radic Biol Med. 2021;171:260–271. doi:10.1016/j.freeradbiomed.2021.05.025
15. D’Amico G, Bazzi C. Pathophysiology of proteinuria. Kidney Int. 2003;63:809–825. doi:10.1046/j.1523-1755.2003.00840.x
16. Passarella P, Kiseleva TA, Valeeva FV, Gosmanov AR. Hypertension management in diabetes: 2018 update. Diabetes Spectr. 2018;31:218–224. doi:10.2337/ds17-0085
17. Fraley EE, Feldman BH. Renal hypertension. N Engl J Med. 1972;287:550–552. doi:10.1056/NEJM197209142871107
18. Yang C, Zhang Z, Liu J, et al. Research progress on multiple cell death pathways of podocytes in diabetic kidney disease. Mol Med. 2023;29:135. doi:10.1186/s10020-023-00732-4
19. Zhou X, Xu C, Dong J, Liao L. Role of renal tubular programed cell death in diabetic kidney disease. Diabetes Metab Res Rev. 2023;39:e3596. doi:10.1002/dmrr.3596
20. Liu P, Zhang Z, Li Y. Relevance of the pyroptosis-related inflammasome pathway in the pathogenesis of diabetic kidney disease. Front Immunol. 2021;12:603416. doi:10.3389/fimmu.2021.603416
21. Fan Q, Li R, Wei H, et al. Research progress of pyroptosis in diabetic kidney disease. Int J Mol Sci. 2024;25:7130. doi:10.3390/ijms25137130
22. Shen S, Ji C, Wei K. Cellular senescence and regulated cell death of tubular epithelial cells in diabetic kidney disease. Front Endocrinol. 2022;13:924299. doi:10.3389/fendo.2022.924299
23. Wang Y, Jin M, Cheng CK, Li Q. Tubular injury in diabetic kidney disease: molecular mechanisms and potential therapeutic perspectives. Front Endocrinol. 2023;14. doi:10.3389/fendo.2023.1238927
24. Wang J, Liu Y, Wang Y, Sun L. The cross-link between ferroptosis and kidney diseases. Oxid Med Cell Longev. 2021;2021:6654887. doi:10.1155/2021/6654887
25. Ding Y, Choi ME. Autophagy in diabetic nephropathy. J Endocrinol. 2015;224:R15–R30. doi:10.1530/JOE-14-0437
26. Tseng C-H, Shah KM, Chiu I-J, Hsiao -L-L. The role of autophagy in type 2 diabetic kidney disease management. Cells. 2023;12:2691. doi:10.3390/cells12232691
27. Nadasdy T, Laszik Z, Blick KE, et al. Proliferative activity of intrinsic cell populations in the normal human kidney. J Am Soc Nephrol. 1994;4:2032. doi:10.1681/ASN.V4122032
28. Deng B, Song A, Zhang C. Cell-cycle dysregulation in the pathogenesis of diabetic kidney disease: an update. Int J Mol Sci. 2023;24:2133. doi:10.3390/ijms24032133
29. Hara M, Oohara K, Dai D-F, Liapis H. Mitotic catastrophe causes podocyte loss in the urine of human diabetics. Am J Pathol. 2019;189:248–257. doi:10.1016/j.ajpath.2018.10.016
30. Nagata M. Podocyte injury and its consequences. Kidney Int. 2016;89:1221–1230. doi:10.1016/j.kint.2016.01.012
31. Boi R, Ebefors K, Nyström J. The role of the mesangium in glomerular function. Acta Physiol. 2023;239:e14045. doi:10.1111/apha.14045
32. Hung P-H, Hsu Y-C, Chen T-H, Lin C-L. Recent advances in diabetic kidney diseases: from kidney injury to kidney fibrosis. Int J Mol Sci. 2021;22:11857. doi:10.3390/ijms222111857
33. Pan D, Xu L, Guo M. The role of protein kinase C in diabetic microvascular complications. Front Endocrinol. 2022;13:973058. doi:10.3389/fendo.2022.973058
34. Xiao Q, Wang D, Li D, et al. Protein kinase C: a potential therapeutic target for endothelial dysfunction in diabetes. J Diabetes Complications. 2023;37:108565. doi:10.1016/j.jdiacomp.2023.108565
35. Garg SS, Gupta J. Polyol pathway and redox balance in diabetes. Pharmacol Res. 2022;182:106326. doi:10.1016/j.phrs.2022.106326
36. Wang N, Zhang C. Oxidative stress: a culprit in the progression of diabetic kidney disease. Antioxidants. 2024;13:455. doi:10.3390/antiox13040455
37. Hong J, Wang X, Zhang N, et al. D-ribose induces nephropathy through RAGE-dependent NF-κB inflammation. Arch Pharm Res. 2018;41:838–847. doi:10.1007/s12272-018-1061-z
38. Yao L, Liang X, Liu Y, et al. Non-steroidal mineralocorticoid receptor antagonist finerenone ameliorates mitochondrial dysfunction via PI3K/Akt/eNOS signaling pathway in diabetic tubulopathy. Redox Biol. 2023;68:102946. doi:10.1016/j.redox.2023.102946
39. Haneda M, Araki S, Togawa M, et al. Mitogen-activated protein kinase cascade is activated in glomeruli of diabetic rats and glomerular mesangial cells cultured under high glucose conditions. Diabetes. 1997;46:847–853. doi:10.2337/diab.46.5.847
40. Liu Y, Wang W, Zhang J, et al. JAK/STAT signaling in diabetic kidney disease. Front Cell Dev Biol. 2023;11:1233259. doi:10.3389/fcell.2023.1233259
41. Jha JC, Banal C, Chow BSM, et al. Diabetes and kidney disease: role of oxidative stress. Antioxid Redox Signal. 2016;25:657–684. doi:10.1089/ars.2016.6664
42. Nishad R, Mukhi D, Kethavath S, et al. Podocyte derived TNF-α mediates monocyte differentiation and contributes to glomerular injury. FASEB J. 2022;36:e22622. doi:10.1096/fj.202200923R
43. Donate-Correa J, Ferri CM, Sánchez-Quintana F, et al. Inflammatory cytokines in diabetic kidney disease: pathophysiologic and therapeutic implications. Front Med. 2020;7:628289. doi:10.3389/fmed.2020.628289
44. Wan J, Liu D, Pan S, et al. NLRP3-mediated pyroptosis in diabetic nephropathy. Front Pharmacol. 2022;13:998574. doi:10.3389/fphar.2022.998574
45. Wada J, Makino H. Innate immunity in diabetes and diabetic nephropathy. Nat Rev Nephrol. 2016;12:13–26. doi:10.1038/nrneph.2015.175
46. Navarro-González JF, Mora-Fernández C, Muros de Fuentes M, García-Pérez J. Inflammatory molecules and pathways in the pathogenesis of diabetic nephropathy. Nat Rev Nephrol. 2011;7:327–340. doi:10.1038/nrneph.2011.51
47. Wada J, Makino H. Inflammation and the pathogenesis of diabetic nephropathy. Clin Sci. 2013;124:139–152. doi:10.1042/CS20120198
48. Barrera-Chimal J, Lima-Posada I, Bakris GL, Jaisser F. Mineralocorticoid receptor antagonists in diabetic kidney disease - mechanistic and therapeutic effects. Nat Rev Nephrol. 2022;18:56–70. doi:10.1038/s41581-021-00490-8
49. Adler SG, Schwartz S, Williams ME, et al. Phase 1 study of anti-CTGF monoclonal antibody in patients with diabetes and microalbuminuria. Clin J Am Soc Nephrol. 2010;5:1420–1428. doi:10.2215/CJN.09321209
50. Falkevall A, Mehlem A, Palombo I, et al. Reducing VEGF-B signaling ameliorates renal lipotoxicity and protects against diabetic kidney disease. Cell Metab. 2017;25:713–726. doi:10.1016/j.cmet.2017.01.004
51. Hong Q, Zhang L, Fu J, et al. LRG1 promotes diabetic kidney disease progression by enhancing TGF-β-induced angiogenesis. J Am Soc Nephrol. 2019;30:546–562. doi:10.1681/ASN.2018060599
52. Adamopoulos C, Piperi C, Gargalionis AN, et al. Advanced glycation end products upregulate lysyl oxidase and endothelin-1 in human aortic endothelial cells via parallel activation of ERK1/2-NF-κB and JNK-AP-1 signaling pathways. Cell Mol Life Sci. 2016;73:1685–1698. doi:10.1007/s00018-015-2091-z
53. Ma Z, Li L, Livingston MJ, et al. p53/microRNA-214/ULK1 axis impairs renal tubular autophagy in diabetic kidney disease. J Clin Invest. 2020;130:5011–5026. doi:10.1172/JCI135536
54. Al-Dabet MM, Shahzad K, Elwakiel A, et al. Reversal of the renal hyperglycemic memory in diabetic kidney disease by targeting sustained tubular p21 expression. Nat Commun. 2022;13:5062. doi:10.1038/s41467-022-32477-9
55. Liu J, Huang K, Cai G-Y, et al. Receptor for advanced glycation end-products promotes premature senescence of proximal tubular epithelial cells via activation of endoplasmic reticulum stress-dependent p21 signaling. Cell Signal. 2014;26:110–121. doi:10.1016/j.cellsig.2013.10.002
56. Jung C-Y, Yoo T-H. Pathophysiologic mechanisms and potential biomarkers in diabetic kidney disease. Diabetes Metab J. 2022;46:181–197. doi:10.4093/dmj.2021.0329
57. Mohandes S, Doke T, Hu H, et al. Molecular pathways that drive diabetic kidney disease. J Clin Invest. 2023;133:e165654. doi:10.1172/JCI165654
58. Tanase DM, Gosav EM, Anton MI, et al. Oxidative stress and NRF2/KEAP1/ARE Pathway in Diabetic Kidney Disease (DKD): new perspectives. Biomolecules. 2022;12:1227. doi:10.3390/biom12091227
59. Hasan IH, Shaheen SY, Alhusaini AM, Mahmoud AM. Simvastatin mitigates diabetic nephropathy by upregulating farnesoid X receptor and Nrf2/HO-1 signaling and attenuating oxidative stress and inflammation in rats. Life Sci. 2024;340:122445. doi:10.1016/j.lfs.2024.122445
60. Watanabe K, Sato E, Mishima E, et al. What’s new in the molecular mechanisms of diabetic kidney disease: recent advances. Int J Mol Sci. 2022;24:570. doi:10.3390/ijms24010570
61. Gnudi L, Coward RJM, Long DA. Diabetic nephropathy: perspective on novel molecular mechanisms. Trends Endocrinol Metab. 2016;27:820–830. doi:10.1016/j.tem.2016.07.002
62. Zhao L, Zou Y, Liu F. Transforming growth factor-beta1 in diabetic kidney disease. Front Cell Dev Biol. 2020;8:187. doi:10.3389/fcell.2020.00187
63. Speer T, Schunk SJ. Klotho in diabetic kidney disease: more than dust in the Wnt. Kidney Int. 2022;102:469–471. doi:10.1016/j.kint.2022.05.016
64. Chen X, Tan H, Xu J, et al. Klotho-derived peptide 6 ameliorates diabetic kidney disease by targeting Wnt/β-catenin signaling. Kidney Int. 2022;102:506–520. doi:10.1016/j.kint.2022.04.028
65. Sweetwyne MT, Gruenwald A, Niranjan T, et al. Notch1 and Notch2 in podocytes play differential roles during diabetic nephropathy development. Diabetes. 2015;64:4099–4111. doi:10.2337/db15-0260
66. Inazaki K, Kanamaru Y, Kojima Y, et al. Smad3 deficiency attenuates renal fibrosis, inflammation, and apoptosis after unilateral ureteral obstruction. Kidney Int. 2004;66:597–604. doi:10.1111/j.1523-1755.2004.00779.x
67. Schwalm S, Beyer S, Frey H, et al. Sphingosine kinase-2 deficiency ameliorates kidney fibrosis by up-regulating Smad7 in a mouse model of unilateral ureteral obstruction. Am J Pathol. 2017;187:2413–2429. doi:10.1016/j.ajpath.2017.06.017
68. Lan HY. Diverse roles of TGF-β/Smads in renal fibrosis and inflammation. Int J Biol Sci. 2011;7:1056–1067. doi:10.7150/ijbs.7.1056
69. Zhang Y, Jin D, Kang X, et al. Signaling pathways involved in diabetic renal fibrosis. Front Cell Dev Biol. 2021;9:696542. doi:10.3389/fcell.2021.696542
70. Adeva-Andany MM, Carneiro-Freire N. Biochemical composition of the glomerular extracellular matrix in patients with diabetic kidney disease. World J Diabetes. 2022;13:498–520. doi:10.4239/wjd.v13.i7.498
71. Rayego-Mateos S, Rodrigues-Diez RR, Fernandez-Fernandez B, et al. Targeting inflammation to treat diabetic kidney disease: the road to 2030. Kidney Int. 2023;103:282–296. doi:10.1016/j.kint.2022.10.030
72. Kellow NJ, Coughlan MT. Effect of diet-derived advanced glycation end products on inflammation. Nutr Rev. 2015;73:737–759. doi:10.1093/nutrit/nuv030
73. Van Puyvelde K, Mets T, Njemini R, et al. Effect of advanced glycation end product intake on inflammation and aging: a systematic review. Nutr Rev. 2014;72:638–650. doi:10.1111/nure.12141
74. Do MH, Hur J, Choi J, et al. Eucommia ulmoides ameliorates glucotoxicity by suppressing advanced glycation end-products in diabetic mice kidney. Nutrients. 2018;10:265. doi:10.3390/nu10030265
75. Cha S-H, Hwang Y, Heo S-J, Jun H-S. Diphlorethohydroxycarmalol attenuates methylglyoxal-induced oxidative stress and advanced glycation end product formation in human kidney cells. Oxid Med Cell Longev. 2018;2018:3654095. doi:10.1155/2018/3654095
76. He S, Zhang Z, Sun H, et al. Potential effects of rapeseed peptide Maillard reaction products on aging-related disorder attenuation and gut microbiota modulation in d-galactose induced aging mice. Food Funct. 2019;10:4291–4303. doi:10.1039/c9fo00791a
77. Sanajou D, Ghorbani Haghjo A, Argani H, et al. Reduction of renal tubular injury with a RAGE inhibitor FPS-ZM1, valsartan and their combination in streptozotocin-induced diabetes in the rat. Eur J Pharmacol. 2019;842:40–48. doi:10.1016/j.ejphar.2018.10.035
78. Sanajou D, Ghorbani Haghjo A, Argani H, et al. FPS-ZM1 and valsartan combination protects better against glomerular filtration barrier damage in streptozotocin-induced diabetic rats. J Physiol Biochem. 2018;74:467–478. doi:10.1007/s13105-018-0640-2
79. Nabi R, Alvi SS, Shah A, et al. Modulatory role of HMG-CoA reductase inhibitors and ezetimibe on LDL-AGEs-induced ROS generation and RAGE-associated signalling in HEK-293 Cells. Life Sci. 2019;235:116823. doi:10.1016/j.lfs.2019.116823
80. Qin J, Peng Z, Yuan Q, et al. AKF-PD alleviates diabetic nephropathy via blocking the RAGE/AGEs/NOX and PKC/NOX Pathways. Sci Rep. 2019;9:4407. doi:10.1038/s41598-018-36344-w
81. Kan W-C, Hwang J-Y, Chuang L-Y, et al. Effect of osthole on advanced glycation end products-induced renal tubular hypertrophy and role of klotho in its mechanism of action. Phytomedicine. 2019;53:205–212. doi:10.1016/j.phymed.2018.09.030
82. Huang K, Huang J, Xie X, et al. Sirt1 resists advanced glycation end products-induced expressions of fibronectin and TGF-β1 by activating the Nrf2/ARE pathway in glomerular mesangial cells. Free Radic Biol Med. 2013;65:528–540. doi:10.1016/j.freeradbiomed.2013.07.029
83. Gong W, Li J, Chen Z, et al. Polydatin promotes Nrf2-ARE anti-oxidative pathway through activating CKIP-1 to resist HG-induced up-regulation of FN and ICAM-1 in GMCs and diabetic mice kidneys. Free Radic Biol Med. 2017;106:393–405. doi:10.1016/j.freeradbiomed.2017.03.003
84. Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012;18:1028–1040. doi:10.1038/nm.2807
85. Tampe D, Zeisberg M. Potential approaches to reverse or repair renal fibrosis. Nat Rev Nephrol. 2014;10:226–237. doi:10.1038/nrneph.2014.14
86. Tan W-Q, Fang -Q-Q, Shen XZ, et al. Angiotensin-converting enzyme inhibitor works as a scar formation inhibitor by down-regulating Smad and TGF-β-activated kinase 1 (TAK1) pathways in mice. Br J Pharmacol. 2018;175:4239–4252. doi:10.1111/bph.14489
87. Ravindran S, Kuruvilla V, Wilbur K, Munusamy S. Nephroprotective Effects of Metformin in Diabetic Nephropathy. J Cell Physiol. 2017;232:731–742. doi:10.1002/jcp.25598
88. Takiyama Y, Harumi T, Watanabe J, et al. Tubular injury in a rat model of type 2 diabetes is prevented by metformin: a possible role of HIF-1α expression and oxygen metabolism. Diabetes. 2011;60:981–992. doi:10.2337/db10-0655
89. Li Z, Liu X, Wang B, et al. Pirfenidone suppresses MAPK signalling pathway to reverse epithelial-mesenchymal transition and renal fibrosis. Nephrology. 2017;22:589–597. doi:10.1111/nep.12831
90. Xue M, Cheng Y, Han F, et al. Triptolide attenuates renal tubular epithelial-mesenchymal transition via the MiR-188-5p-mediated PI3K/AKT pathway in diabetic kidney disease. Int J Biol Sci. 2018;14:1545–1557. doi:10.7150/ijbs.2403
Comments (0)