- Review
- Open access
- Published:
Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review
BMC Nephrology volume 26, Article number: 144 (2025)
Abstract
Acute kidney injury (AKI) is a clinical challenge characterized by elevated morbidity and a substantial impact on individual health and socioeconomic factors. A comprehensive examination of the molecular pathways behind AKI is essential for its prevention and management. In recent years, vigorous research in the domain of AKI has concentrated on pathophysiological characteristics, early identification, and therapeutic approaches across many aetiologies and highlighted the principal themes of oxidative stress, inflammatory response, apoptosis, necrosis, and immunological response. This review comprehensively reviewed the molecular mechanisms underlying AKI, including oxidative stress, inflammatory pathways, immune cell-mediated injury, activation of the renin-angiotensin-aldosterone (RAAS) system, mitochondrial damage and autophagy, apoptosis, necrosis, etc. Inflammatory pathways are involved in the injuries in all four structural components of the kidney. We also summarized therapeutic techniques and pharmacological agents associated with the aforementioned molecular pathways. This work aims to clarify the molecular mechanisms of AKI thoroughly, offer novel insights for further investigations of AKI, and facilitate the formulation of efficient therapeutic methods to avert the progression of AKI.
Introduction
Acute kidney injury (AKI) refers to the sudden damage to the renal parenchyma or sharp decline in kidney function, resulting from a range of factors with diverse underlying mechanisms. AKI can be broadly categorized into pre-renal AKI, which is centered on decreased glomerular filtration rate (GFR) due to decreased renal perfusion, and post-renal AKI, which is centered on urinary tract obstruction. The rest are renal AKI in which the structures of the nephron, such as glomeruli, tubules, vasculature, and renal tubules are affected (Table 1) [1]. AKI is a heavy health burden worldwide, with a global incidence of approximately 21.6% and 33.7% in adults and children, respectively, and an overall mortality rate of 23.9% and 13.8% in adults and children in 2013 [2]. AKI is significantly associated with in-hospital adverse events and progression to chronic kidney disease (CKD) and greatly increases the risk of long-term morbidity and mortality [3,4,5].
Although the clinical significance of AKI is widely recognized, there is still a lack of specific therapeutic options for AKI so far. Diverse molecular mechanisms have been reported to contribute to the parenchymal damage of kidneys in AKI, including complement activation [6], oxidative stress [7], and inflammation [7, 8]. Delving into these molecular mechanisms underlying the pathological processes of AKI is an important approach to identify specific biomarkers and potential therapeutic targets.
Here in this review, we summarized the molecular mechanisms and corresponding interventions of AKI in the literature, aiming to comprehensively and thoroughly understand the pathogenic molecular mechanisms of AKI and to provide assistance for future research on specific prevention and treatment strategies for AKI. For ease of description, the molecular mechanisms are summarized based on the four main components of the kidney, the glomeruli, tubules, vasculature, and interstitium. Interventions for individual mechanisms are also presented.
Underlying molecular mechanisms of glomerular injury in AKI
Immune complex deposition
Glomeruli are susceptible to commonly affected by injury caused by deposition of immune complexes in systemic immune and autoimmune disorders [9, 10]. Deposited immune complexes stimulate circulating immune cells, which subsequently release cytokines and vasoactive substances, creating a pro-inflammatory milieu after the deposition of immune complexes [11]. In IgA nephropathy, stimulatory circulating immune complexes containing galactose-deficient Immunoglobulin A1 induced the production of transforming growth factor-beta (TGF-β), which stimulated the production of extracellular matrix (ECM) proteins and promoted the synthesis of laminin, leading to the proliferation of mesangial cells [12].
Immune complexes activate complement via the classical pathway and have been reported in various types of glomerulonephritis [13], leading to damage to glomerular capillary walls, glomerular basement membrane, and other structures and inflammatory cell recruitment [14]. Complement Component 3a (C3a) and Complement Component 5a (C5a) formed by complement activation have potent proinflammatory functions and create the membrane attack complex (MAC) that can induce cell activation or lysis [15]. Activation of the complement system forms the C5a-C5a receptor1 axis to promote podocyte injury and disrupt the glomerular filtration barrier by enhancing Dynamin-related protein 1-mediated mitochondrial fission [16].
Activation of renin-angiotensin-aldosterone system (RAAS)
Reduced renal blood flow, particularly in the presence of pre-renal AKI, results in a diminished GFR, prompting para-glomerular cells to secrete increased amounts of renin, activating the RAAS system. Prolonged activation of the RAAS results in renal hypoxia, vasoconstriction, glomerulosclerosis, and glomerular interstitial fibrosis [17]. Angiotensin II (AngII) induces glomerulosclerosis through the upregulation of TGF-β in rat renal cells, leading to enhanced production of ECM proteins [18]. AngII has also been shown to enhance the expression of toll-like receptor 4 (TLR4), which in turn activates nuclear factor-kappa B (NF-κB) in mesangial cells, amplifying the inflammatory response [19]. Moreover, hyperactivation of RAAS promotes oxidative stress to exacerbate kidney damage [20]. Research indicates that angiotensin-converting enzyme inhibitors and AngII receptor blockers are associated with improved survival outcomes in AKI, although they may also contribute to a heightened likelihood of complications, including acute renal failure and hyperkalemia [21]. Spironolactone, another RAAS system inhibitor, antagonizes aldosterone receptors but poses a danger of hyperkalemia [22]. However, it was reported that spironolactone treatment after 24 h of ischemic injury leading to AKI can improve renal inflammation and mitigate the progression from AKI to CKD [23].
Underlying molecular mechanisms of renal tubular injury in AKI
Activation of the complement system
The proximal tubules are the primary location of damage during renal ischemia-reperfusion injury (IRI), and complement activation on ischemic renal tubules significantly contributes to ischemic AKI [24]. Complement activation is mediated via three distinct pathways: the classical, lectin, and alternative pathways. Although these three pathways have different initiation mechanisms, they ultimately converge on C3 cleavage, leading to the activation of C3 and C5 and forming MAC, which induces cell activation or cleavage [15]. Data on complement mechanisms in AKI are mainly from animal models. The lectin complement pathway is activated in the renal tubules of mice with AKI in rhabdomyolysis, probably through an aberrant pattern of fucosyltransferase 2-dependent cytosolic fucosylation, recognized by the pattern-recognition molecule collectin-11 and in a C4-independent bypass manner [25]. Stimulation of TGF-β1 production in cultured mouse renal tubular cells after complement activation by C3a and C5a binding to C5a receptor and C3a receptor, and stimulation of proximal tubular epithelial cells by MAC leading to increased expression of type IV collagen mediate renal fibrosis resulting in the transition from AKI to CKD [26]. Furthermore, C3a mediates podocyte injury through TLR4/NF-κB-P65 signaling during IRI-AKI and post-injury fibrosis [27]. Ongoing complement-inhibiting drugs are currently used to treat a variety of kidney diseases causing AKI and exhibit promising efficacy (Table 2).
Mitochondrial impairment and autophagy pathway
Mitochondria serves as a principal source of cellular energy, with the proximal tubule being the primary locus of mitochondrial failure in AKI [28]. Mitochondrial damage seems to occur prior to the clinical manifestations of AKI following the onset of injury [29]. Electron microscopy revealed mitochondrial swelling, accompanied by minimal acute renal function impairment within 60 min of occluding a renal vessel [30]. Mitochondrial injury results in elevated reactive oxygen species (ROS), which subsequently exacerbates mitochondrial damage [31]. To reduce harm, the body activates mitochondrial autophagy to remove surplus or impaired mitochondria [32]. Augmented mitochondrial autophagy facilitates cell survival initially, while excessive activation of mitochondrial autophagy results in cell death and tissue injury [33]. Research indicates that the mitochondria-targeted antioxidant 10-(6’-plastoquinonyl) decylrhodamine 19 enhances renal blood flow and protects endothelial cells following renal ischemia-reperfusion [34].
Oxidative stress
The kidney utilizes substantial quantities of oxygen to provide energy for tubular reabsorption; consequently, renal tubules are particularly vulnerable to ischemia and hypoxic damage [35]. Ischemia and hypoxia in renal tubular cells elevate endogenous free radical production and disrupt mitochondrial oxidative phosphorylation, resulting in excessive ROS, while minimal ROS and free radicals can confer advantages to biological systems, their surplus induces oxidative stress, inflicting damage on various cellular structures, including membranes, proteins, lipids, and DNA [36, 37]. The oxidative stress processes of AKI primarily encompass ROS production, nitric oxide depletion, damage-associated molecular patterns (DAMPs) formation, toll-like receptors (TLRs) activation, autophagy, and microvascular dysfunction [38]. In IRI, DAMPs released from necrotic renal tubular cells activate TLRs on renal cells, initiating a cascade in inhibitor of NF-κB, resulting in its release and facilitating the nuclear translocation of NF-κB, within the nucleus, NF-κB drives the expression of inflammatory cytokines, oxidative bursts, and inflammatory responses, thereby exacerbating the injury process [39]. Prolonged exposure to oxidative stress stimuli in kidney tissue results in an initial inflammatory phase, subsequently leading to the excessive production of fibrous tissue, which compromises organ function and may result in CKD [40].
Antioxidant treatment shows promising results in AKI (Table 3). N-acetylcysteine (NAC) helps to mitigate contrast-induced AKI (CI-AKI), while Au NCs-NAC (ultra-small gold nanoclusters measuring 1–2 nm, capped with NAC) exhibit significantly enhanced therapeutic efficacy compared to NAC alone [41, 42]. Vitamin C sequesters reactive nitrogen species and ROS and replenishes endogenous glutathione levels, establishing its role as a vital antioxidant [43]. Preventive oral vitamin E in conjunction with a 0.9% saline infusion has been demonstrated to diminish the occurrence of CI-AKI in patients with CKD undergoing elective coronary angiography, as opposed to saline alone [44]. Lipoic acid decreases signs of oxidative stress and inflammatory response and has a preventative effect on the course of severe AKI [45]. Coenzyme Q10 possesses anti-peroxidative, anti-apoptotic, and anti-inflammatory properties and offers renoprotection after AKI [46]. Alongside topical treatments, the body possesses endogenous antioxidant systems that can alleviate damage from oxidative stress [47]. The clinical trials of antioxidants are currently being conducted vigorously and demonstrate favorable therapeutic outcomes (Table 4).
Inflammatory pathways
Damaged cells and immune cells release various cytokines (e.g., tumor necrosis factor (TNF), interleukin (IL)) and chemokines that attract neutrophils and macrophages from the bloodstream to the damaged site [48, 49]. Numerous studies indicate that pro-inflammatory cytokines and chemokines rely on NF-κB, binding to specific DNA regulatory elements, and triggering the expression of various inflammatory genes [50]. Tacrolimus can reduce the nuclear translocation of NF-κB, while glucocorticoids suppress the DNA-binding function of NF-κB [51]. In addition, glucocorticoids may be an effective therapeutic approach for septic AKI by mitigating mitochondrial damage, apoptosis, and anti-inflammatory [52].
Other inflammatory pathways are also involved in tubular injury in AKI. Research has demonstrated that NOD-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome has an important role in CI-AKI models [53]. Remdesivir can suppress the activation of the NLRP3 inflammasome, therefore mitigating AKI [54]. TLR4 activates the NLRP3 inflammasome and the mitogen-activated protein kinases (MAPKs) phosphorylation inflammatory pathway, hence facilitating inflammatory reactions, inhibition of TLR4 confers a protective effect against sepsis-induced AKI in mice and represents a potential therapeutic approach for AKI [55, 56]. Furthermore, the thioredoxin-interacting protein facilitates the endoplasmic reticulum (ER) stress-mediated formation of the NLRP3 inflammasome complex and associates oxidative stress with the activation of the NLRP3 inflammasome, instigating mitochondria stress-induced apoptosis and promoting inflammatory cell death, thioredoxin-interacting protein may be a potential therapeutic target for AKI [57, 58].
The MAPK kinase signaling pathway activated by ROS from oxidative stress promotes the activation of activator protein-1, which regulates the modulation of pro-inflammatory cytokines and chemokines [59, 60]. Metformin mitigates inflammation, oxidative stress, and ER stress in renal tubular cells by activating 5’ Adenosine monophosphate-activated Protein Kinase in preclinical AKI models [61]. Pioglitazone improved IRI-AKI by attenuating oxidative stress and NLRP3 inflammasome activation [62]. The inflammasome-IL-1α/IL-1β-IL-1Receptor system is a pivotal component of renal inflammation and its systemic repercussions, with anti-inflammatory treatment utilizing IL-1 receptor antagonists being a viable option [63, 64]. Mast cells cause AKI through the production of TNF, and the application of TNF blockers (e.g., infliximab) protects renal function [65, 66]. Anti-inflammatory drugs for the treatment of AKI are summarized in Table 5. The ongoing clinical trials of anti-inflammatory drugs are summarized in Table 6.
Endoplasmic reticulum (ER) stress
The ER serves as the principal location for protein synthesis and processing, lipid metabolism, and signal transduction in the cell [67]. The accumulation of newly synthesized unfolded proteins due to ischemia, hypoxia, and oxidative stress induces ER stress, if ER stress is severe or prolonged, it ultimately results in cellular apoptosis, protein degradation, and translational attenuation [68]. Overall, In AKI, mild to moderate ER stress confers a protective effect on the kidneys, but severe ER stress worsens AKI [69]. The toxicity of clinically significant nephrotoxic agents, such as cisplatin and gentamicin, is markedly diminished in cells preconditioned with ER stress inducers [70]. However, TUG891 activates the G protein-coupled receptor 120, mitigating ER stress and apoptosis in cisplatin-induced AKI, thereby preserving kidney integrity [71]. Furthermore, ER stress is closely related to autophagy, which has a protective effect on ER stress-induced renal cell apoptosis, rapamycin enhances autophagy activity by inhibiting the mammalian target of rapamycin complex 1 to protect renal cells from ER stress-induced apoptosis [72].
Apoptosis and necrosis pathways
Proximal tubular cells exhibit significant metabolic activity and are susceptible to hypoxia and ischemia, rendering them a frequent locus for apoptosis and necrosis in AKI [35]. AKI encompasses several forms of renal cell death, including apoptosis, necrosis, pyroptosis, apoptosis-like necrosis, and ferroptosis [73]. Exposure of renal tubular cells to cisplatin results in tubular cell damage and mortality [74]. Cellular apoptosis and necrosis depend on trauma severity. Apoptosis involves intrinsic and extrinsic pathways and their interactions. Stress triggers the intrinsic pathway, releasing cytochrome c via Bcl-2-associated X protein/Bcl-2 antagonist killer 1-induced mitochondrial permeabilization, which activates caspase-9 and caspase-3, initiating apoptosis, in contrast, the extrinsic pathway involves Fas ligands binding death receptors, activating caspase-8, which triggers caspase-3 and cleaves BH3-interacting domain death agonist (Bid) into truncated Bid, which translocates to mitochondria, amplifying the intrinsic pathway and apoptotic cascade [8]. Selective reduction of pro-apoptotic genes, including Bax and Bak, reduces renal tubular cell apoptosis and offers protection against ischemia AKI [75]. Modulating specific apoptotic pathways may be advantageous for renal protection, the apoptosis of myofibroblast populations aids in renal recovery following IRI-AKI [76]. Recent discoveries indicate that ferroptosis plays a significant role in AKI. Ferroptosis is an iron-dependent form of cell death marked by the depletion of membrane lipid peroxidation products and plasma membrane polyunsaturated fatty acids, distinguishing it from apoptosis and necrosis [77]. Inhibition of necrosis and ferroptosis has demonstrated beneficial effects in many models of AKI [78].
Autophagy pathways
The induction of autophagy in proximal tubules during acute tubulointerstitial nephritis demonstrated that autophagy serves a nephroprotective function in AKI, nevertheless, excessive autophagy ultimately results in cellular demise [79]. Fibroblast growth factor 10 (FGF10) has been demonstrated to mitigate renal IRI-AKI by preventing excessive autophagy [80]. Autophagy may significantly modulate the inflammatory response in AKI, making the autophagy-inflammation axis a promising therapeutic target for AKI [81].
Underlying molecular mechanisms of renal vascular injury in AKI
Vascular endothelial damage
Under typical circumstances, endothelial cells perform activities including anticoagulation, anti-inflammation, vascular tone modulation, and release of vasoactive cytokines [82]. Damage to endothelial cells results in heightened secretion of the vasoconstrictor endothelin and diminished secretion of the vasodilator nitric oxide, causing sustained renal vasoconstriction, impaired renal perfusion, and aggravated renal injury [83]. Research indicates that the endothelin system is upregulated in AKI patients, this upregulation can promote the progression from AKI to CKD, and the use of selective antagonists of the endothelin-a receptor or non-dihydropyridine calcium channel blockers to block the endothelin system can protect the kidneys [84]. Meta-analyses indicate that the administration of inducible nitric oxide correlates with a diminished risk of AKI in individuals undergoing cardiac surgery, but it markedly elevates the risk of AKI in patients suffering from acute respiratory distress syndrome [85]. Furthermore, in the presence of ischemia and hypoxia, endothelial cells elevate the synthesis of ROS, and elevated levels of ROS inflict additional harm on endothelial cells [86].
Renal artery thrombosis and renal artery thromboembolism
Renal artery thromboembolism is a rare yet significant etiology of AKI, the formation of a thrombus in one or both arteries supplying blood to the kidneys results in diminished renal perfusion, potentially causing AKI [87]. Intact vascular endothelial cells possess anticoagulant properties; but upon activation due to endothelial injury, they facilitate renal artery thrombosis by initiating the blood coagulation cascade through the synthesis of procoagulant factors that enhance the generation and spread of thrombin [88]. Notably, renal artery thrombosis may arise as a secondary complication of coronavirus infection [89].
Vascular thinning
Vascular thinning denotes the diminishment of capillaries or a decrease in density, and the diminished density of renal microvessels may be a significant characteristic of the progression from AKI to CKD [90]. Contemporary therapies for vascular atrophy concentrate on protecting and rehabilitating the capillary network, numerous studies have suggested that the severity of AKI may be mitigated and the prognosis enhanced by facilitating neovascularization and protecting existing arteries [91].
Underling molecular mechanisms of renal interstitial injury in AKI
Immune cells-mediated damage
The renal interstitium houses the majority of the renal immune system, predominantly including dendritic cells, along with macrophages and fibroblasts [92]. Dendritic cells collect and deliver antigens to initiate adaptive immune responses [10]. Antigen-presenting cells, including dendritic cells and macrophages, convey antigens to naive T lymphocytes, triggering their activation and differentiation, CD4 + T helper cells are essential for coordinating immune responses by aiding B cells and activating cytotoxic CD8 + T cells, B lymphocytes mature into plasma cells, which generate antibodies that combat infections and facilitate their eradication [93]. B lymphocytes also secrete chemokines C-C motif-ligand 7 (CCL7), which recruit neutrophils and monocytes to the kidney during AKI [94]. Neutrophils contribute to inflammation by secreting cytokines, ROS, proteases, and other substances into renal tissue [95]. Macrophages are classified into type 1 and type 2, with M1 macrophages generating pro-inflammatory cytokines to promote inflammation, and M2 macrophages exhibit anti-inflammatory and pro-fibrotic properties [96]. Furthermore, interferon-γ, secreted by natural killer cells, activates M1 macrophages within renal tissue [97]. Various inflammatory cells release several inflammatory factors, infiltrate the renal interstitium, and aggravate renal injury [10].
Renal interstitial fibrosis
In AKI, renal interstitial cells generate significant quantities of inflammatory mediators, initiating sustained inflammatory responses that result in renal interstitial fibrosis and subsequently impair the recovery of kidney function [98]. Renal interstitial fibrosis is an irreversible phenomenon marked by the abnormal buildup of ECM, influenced by many cellular and molecular pathways [99]. During AKI, transitory Wnt-β-catenin signaling facilitates repair and regeneration, while persistent upregulation promotes renal fibrosis [100]. Snail 1-mediated partial transformation of epithelial cells into mesenchymal cells facilitates renal fibrosis in mice [101]. Additionally, AngII facilitates renal fibrosis by activating Janus kinases and MAPKs, promoting intracellular calcium influx via Ca2+ channels and enhancing the signaling pathways of TGF-β [102]. FGF23 is increased in individuals with AKI, and enhancement of FGF23 activates TGF-β and Wnt/β-catenin, promoting renal fibrosis and contributing to the transition from AKI to CKD [103, 104]. Research indicates that pirfenidone is a suitable choice for placebo-controlled trials in individuals with advanced CKD [105]. Subsequent investigations indicate that long-acting sustained-release pirfenidone did not enhance the clinical progression of septic AKI, however, it is safe and does not cause adverse effects [106].
The underlying molecular mechanisms in the four components of the kidney are summarized in Fig. 1. Inflammatory pathways are involved in the injuries in all four components. The reported mechanisms are not independent of each other, but rather interlinked with each other (Fig. 2).
Interlinkages among mechanisms underlying AKI. Abbreviations: ROS: Reactive oxygen species; AngII: Angiotensin II; NF-κB: Nuclear factor-kappa B; IκB: Inhibitor of NF-Κb; TLR: Toll-like receptor; TXNIP: Thioredoxin-interacting protein; IL: Interleukin; C3a: Complement Components 3a; NLRP3: NOD-like receptor family, Pyrin domain containing 3; ER: Endoplasmic reticulum; AP-1: Activator protein-1; AKI: Acute kidney injury
Conclusions
AKI is a critical concern in nephrology, inflicting considerable harm on both physical and mental health, while its elevated incidence and fatality rates place a tremendous strain on the healthcare system. The clinical diagnosis of AKI relies on changes in serum creatinine levels and urine output, lacking definitive methodologies. It is imperative to investigate the molecular pathways to create highly specific and readily observable diagnostic tools and to develop therapeutic agents targeting these mechanisms.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- AKI:
-
Acute kidney injury
- AngII:
-
Angiotensin II
- Bid:
-
BH3-interacting domain death agonist
- CCL7:
-
Chemokines C-C motif-ligand 7
- CI-AKI:
-
Mitigate contrast-induced AKI
- CKD:
-
Chronic kidney disease
- C3a:
-
Complement Components 3a
- C5a:
-
Complement Components 5a
- DAMPs:
-
Damage-associated molecular patterns
- ECM:
-
Extracellular matrix
- ER:
-
Endoplasmic reticulum
- FGF10:
-
Fibroblast growth factor 10
- GFR:
-
Decreased glomerular filtration rate
- IL:
-
Interleukin
- IRI:
-
Ischemia-reperfusion injury
- MAC:
-
Membrane attack complex
- MAPKs:
-
Mitogen-activated protein kinases
- NAC:
-
N-acetylcysteine
- NF-κB:
-
Transcription factor-kappa B
- NLRP3:
-
NOD-like receptor family, pyrin domain containing 3
- RAAS:
-
Renin-angiotensin-aldosterone system
- ROS:
-
Reactive oxygen species
- TGF-β:
-
Transforming growth factor-β
- TLR4:
-
Toll-like receptor 4
- TNF:
-
Tumor necrosis factor
References
Sluman C, Gudka PM, McCormick K. Acute kidney injury: pre-renal, intra-renal and post-renal. Ren Med Clin Pharm. 2020;1:23–44.
Susantitaphong P, Cruz DN, Cerda J, Abulfaraj M, Alqahtani F, Koulouridis I, et al. World incidence of AKI: A meta-analysis. Clin J Am Soc Nephrol. 2013;8(9):1482–93.
Darmon M, Clec’h C, Adrie C, Argaud L, Allaouchiche B, Azoulay E, et al. Acute respiratory distress syndrome and risk of AKI among critically ill patients. Clin J Am Soc Nephrol. 2014;9(8):1347–53.
Gambardella I, Gaudino M, Ronco C, Lau C, Ivascu N, Girardi LN. Congestive kidney failure in cardiac surgery: The relationship between central venous pressure and acute kidney injury. Interact Cardiovasc Thorac Surg. 2016;23(5):800–5.
See EJ, Jayasinghe K, Glassford N, Bailey M, Johnson DW, Polkinghorne KR, et al. Long-term risk of adverse outcomes after acute kidney injury: A systematic review and meta-analysis of cohort studies using consensus definitions of exposure. Kidney Int. 2019;95(1):160–72.
McCullough JW, Renner B, Thurman JM. The role of the complement system in acute kidney injury. Semin Nephrol. 2013;33(6):543–56.
Andrianova NV, Zorov DB, Plotnikov EY. Targeting inflammation and oxidative stress as a therapy for ischemic kidney injury. Biochem (Mosc). 2020;85(12):1591–602.
Han SJ, Lee HT. Mechanisms and therapeutic targets of ischemic acute kidney injury. Kidney Res Clin Pract. 2019;38(4):427–40.
Anders HJ, Saxena R, Zhao MH, Parodis I, Salmon JE, Mohan C. Lupus nephritis. Nat Rev Dis Primers. 2020;6(1):7.
Tecklenborg J, Clayton D, Siebert S, Coley SM. The role of the immune system in kidney disease. Clin Exp Immunol. 2018;192(2):142–50.
Couser WG. Basic and translational concepts of immune-mediated glomerular diseases. J Am Soc Nephrol. 2012;23(3):381–99.
Novak J, Raskova Kafkova L, Suzuki H, Tomana M, Matousovic K, Brown R, et al. IgA1 immune complexes from pediatric patients with IgA nephropathy activate cultured human mesangial cells. Nephrol Dial Transpl. 2011;26(11):3451–7.
Zwirner J, Felber E, Herzog V, Riethmüller G, Feucht HE. Classical pathway of complement activation in normal and diseased human glomeruli. Kidney Int. 1989;36(6):1069–77.
Anguiano L, Kain R, Anders HJ. The glomerular crescent: Triggers, evolution, resolution, and implications for therapy. Curr Opin Nephrol Hypertens. 2020;29(3):302–9.
Petr V, Thurman JM. The role of complement in kidney disease. Nat Rev Nephrol. 2023;19(12):771–87.
Ye B, Chen B, Guo C, Xiong N, Huang Y, Li M, et al. C5a-C5aR1 axis controls mitochondrial fission to promote podocyte injury in lupus nephritis. Mol Ther. 2024;32(5):1540–60.
Chou YH, Chu TS, Lin SL. Role of renin-angiotensin system in acute kidney injury-chronic kidney disease transition. Nephrol (Carlton). 2018;23(Suppl 4):121–5.
Kagami S, Border WA, Miller DE, Noble NA. Angiotensin II stimulates extracellular matrix protein synthesis through induction of transforming growth factor-beta expression in rat glomerular mesangial cells. J Clin Invest. 1994;93(6):2431–7.
Wolf G, Bohlender J, Bondeva T, Roger T, Thaiss F, Wenzel UO. Angiotensin II upregulates toll-like receptor 4 on mesangial cells. J Am Soc Nephrol. 2006;17(6):1585–93.
Ma K, Gao W, Xu H, Liang W, Ma G. Role and mechanism of the Renin-Angiotensin-Aldosterone system in the onset and development of cardiorenal syndrome. J Renin Angiotensin Aldosterone Syst. 2022;2022:3239057.
Brar S, Ye F, James MT, Hemmelgarn B, Klarenbach S, Pannu N. Association of angiotensin-converting enzyme inhibitor or angiotensin receptor blocker use with outcomes after acute kidney injury. JAMA Intern Med. 2018;178(12):1681–90.
Lindhardt JL, Nielsen PM, Hansen ESS, Qi H, Tougaard RS, Mariager C, et al. The hemodynamic and metabolic effects of spironolactone treatment in acute kidney injury assessed by hyperpolarized MRI. NMR Biomed. 2020;33(10):e4371.
Barrera-Chimal J, Rocha L, Amador-MartÃnez I, Pérez-Villalva R, González R, Cortés-González C, et al. Delayed spironolactone administration prevents the transition from acute kidney injury to chronic kidney disease through improving renal inflammation. Nephrol Dial Transpl. 2019;34(5):794–801.
Thurman JM, Ljubanovic D, Edelstein CL, Gilkeson GS, Holers VM. Lack of a functional alternative complement pathway ameliorates ischemic acute renal failure in mice. J Immunol. 2003;170(3):1517–23.
Boudhabhay I, Poillerat V, Grunenwald A, Torset C, Leon J, Daugan MV, et al. Complement activation is a crucial driver of acute kidney injury in rhabdomyolysis. Kidney Int. 2021;99(3):581–97.
Franzin R, Stasi A, Fiorentino M, Stallone G, Cantaluppi V, Gesualdo L, et al. Inflammaging and complement system: A link between acute kidney injury and chronic graft damage. Front Immunol. 2020;11:734.
Chen Y, Lin L, Rao S, Tao X, Cui J, Wan J. Complement C3 mediates podocyte injury through TLR4/NFΚB-P65 signaling during ischemia-reperfusion acute kidney injury and post-injury fibrosis. Eur J Med Res. 2023;28(1):135.
Emma F, Montini G, Parikh SM, Salviati L. Mitochondrial dysfunction in inherited renal disease and acute kidney injury. Nat Rev Nephrol. 2016;12(5):267–80.
Ralto KM, Parikh SM. Mitochondria in acute kidney injury. Semin Nephrol. 2016;36(1):8–16.
Parekh DJ, Weinberg JM, Ercole B, Torkko KC, Hilton W, Bennett M, et al. Tolerance of the human kidney to isolated controlled ischemia. J Am Soc Nephrol. 2013;24(3):506–17.
Giorgi C, Marchi S, Simoes ICM, Ren Z, Morciano G, Perrone M, et al. Mitochondria and reactive oxygen species in aging and age-related diseases. Int Rev Cell Mol Biol. 2018;340:209–344.
Harper JW, Ordureau A, Heo JM. Building and decoding ubiquitin chains for mitophagy. Nat Rev Mol Cell Biol. 2018;19(2):93–108.
Aggarwal S, Mannam P, Zhang J. Differential regulation of autophagy and mitophagy in pulmonary diseases. Am J Physiol Lung Cell Mol Physiol. 2016;311(2):L433–52.
Jankauskas SS, Andrianova NV, Alieva IB, Prusov AN, Matsievsky DD, Zorova LD, et al. Dysfunction of kidney endothelium after ischemia/reperfusion and its prevention by mitochondria-targeted antioxidant. Biochem (Mosc). 2016;81(12):1538–48.
Rahbar Saadat Y, Hosseiniyan Khatibi SM, Sani A, Zununi Vahed S, Ardalan M. Ischemic tubular injury: Oxygen-sensitive signals and metabolic reprogramming. Inflammopharmacology. 2023;31(4):1657–69.
Pizzino G, Irrera N, Cucinotta M, Pallio G, Mannino F, Arcoraci V, et al. Oxidative stress: Harms and benefits for human health. Oxid Med Cell Longev. 2017;2017:8416763.
Halliwell B. Biochemistry of oxidative stress. Biochem Soc Trans. 2007;35(Pt 5):1147–50.
Pavlakou P, Liakopoulos V, Eleftheriadis T, Mitsis M, Dounousi E. Oxidative stress and acute kidney injury in critical illness: Pathophysiologic mechanisms-biomarkers-interventions, and future perspectives. Oxid Med Cell Longev. 2017;2017:6193694.
Radi ZA. Immunopathogenesis of acute kidney injury. Toxicol Pathol. 2018;46(8):930–43.
Su H, Wan C, Song A, Qiu Y, Xiong W, Zhang C. Oxidative stress and renal fibrosis: Mechanisms and therapies. Adv Exp Med Biol. 2019;1165:585–604.
Tepel M, van der Giet M, Schwarzfeld C, Laufer U, Liermann D, Zidek W. Prevention of radiographic-contrast-agent-induced reductions in renal function by acetylcysteine. N Engl J Med. 2000;343(3):180–4.
Zhang DY, Tu T, Younis MR, Zhu KS, Liu H, Lei S, et al. Clinically translatable gold nanozymes with broad spectrum antioxidant and anti-inflammatory activity for alleviating acute kidney injury. Theranostics. 2021;11(20):9904–17.
Kaur KK, Allahbadia G, Singh M. An update in the utilization of N-acetyl cysteine & Vitamin C for tackling the oxidative stress in acute kidney injury secondary to robust sepsis-A systematic review. J Clin Nephrol. 2022;6(1):001–18.
Rezaei Y, Khademvatani K, Rahimi B, Khoshfetrat M, Arjmand N, Seyyed-Mohammadzad MH. Short-term high-dose Vitamin E to prevent contrast medium-induced acute kidney injury in patients with chronic kidney disease undergoing elective coronary angiography: A randomized Placebo-Controlled trial. J Am Heart Assoc. 2016;5(3):e002919.
Ali RM, Khairy MA, Mansour DY. Effect of alpha-lipoic acid on acute lung injury and acute kidney injury in major postpartum hemorrhage. Ain Shams J Anesthesiology. 2016;9(2).
Zhao S, Wu W, Liao J, Zhang X, Shen M, Li X, et al. Molecular mechanisms underlying the renal protective effects of coenzyme Q10 in acute kidney injury. Cell Mol Biol Lett. 2022;27(1):57.
Day BJ. Antioxidant therapeutics: Pandora’s box. Free Radic Biol Med. 2014;66:58–64.
Ernandez T, Mayadas TN. The changing landscape of renal inflammation. Trends Mol Med. 2016;22(2):151–63.
Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, et al. Inflammation in AKI: Current understanding, key questions, and knowledge gaps. J Am Soc Nephrol. 2016;27(2):371–9.
Oeckinghaus A, Ghosh S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb Perspect Biol. 2009;1(4):a000034.
Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther. 2017;2:17023.
Choi HM, Jo SK, Kim SH, Lee JW, Cho E, Hyun YY, et al. Glucocorticoids attenuate septic acute kidney injury. Biochem Biophys Res Commun. 2013;435(4):678–84.
Shen J, Wang L, Jiang N, Mou S, Zhang M, Gu L, et al. NLRP3 inflammasome mediates contrast media-induced acute kidney injury by regulating cell apoptosis. Sci Rep. 2016;6:34682.
Yin L, Zhao H, Zhang H, Li Y, Dong Y, Ju H, et al. Remdesivir alleviates acute kidney injury by inhibiting the activation of NLRP3 inflammasome. Front Immunol. 2021;12:652446.
Yue L, Liu X, Wu C, Lai J, Wang J, Zhong H, et al. Toll-like receptor 4 promotes the inflammatory response in septic acute kidney injury by promoting p38 mitogen-activated protein kinase phosphorylation. J Bioenerg Biomembr. 2023;55(5):353–63.
Jha AK, Gairola S, Kundu S, Doye P, Syed AM, Ram C, et al. Toll-like receptor 4: An attractive therapeutic target for acute kidney injury. Life Sci. 2021;271:119155.
Zhou R, Tardivel A, Thorens B, Choi I, Tschopp J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat Immunol. 2010;11(2):136–40.
Choi EH, Park SJ. TXNIP: A key protein in the cellular stress response pathway and a potential therapeutic target. Exp Mol Med. 2023;55(7):1348–56.
McCubrey JA, Lahair MM, Franklin RA. Reactive oxygen species-induced activation of the MAP kinase signaling pathways. Antioxid Redox Signal. 2006;8(9–10):1775–89.
Kyriakis JM, Avruch J. Mammalian mitogen-activated protein kinase signal transduction pathways activated by stress and inflammation. Physiol Rev. 2001;81(2):807–69.
Song A, Zhang C, Meng X. Mechanism and application of metformin in kidney diseases: An update. Biomed Pharmacother. 2021;138:111454.
Ye Z, Zhang J, Xu Z, Li Z, Huang G, Tong B, et al. Pioglitazone ameliorates ischemia/reperfusion-induced acute kidney injury via oxidative stress attenuation and NLRP3 inflammasome. Hum Cell. 2024;37(4):959–71.
Anders HJ. Of inflammasomes and alarmins: IL-1β and IL-1α in kidney disease. J Am Soc Nephrol. 2016;27(9):2564–75.
Dinarello CA, Simon A, van der Meer JW. Treating inflammation by blocking interleukin-1 in a broad spectrum of diseases. Nat Rev Drug Discov. 2012;11(8):633–52.
Summers SA, Chan J, Gan PY, Dewage L, Nozaki Y, Steinmetz OM, et al. Mast cells mediate acute kidney injury through the production of TNF. J Am Soc Nephrol. 2011;22(12):2226–36.
Netrebenko AS, Gureev VV, Pokrovskii MV, Gureeva AV, Tsuverkalova YM, Rozhkov IS. Assessment of the nephroprotective properties of the erythropoietin mimetic peptide and infliximab in kidney ischemia-reperfusion injury in rats. Arch Razi Inst. 2021;76(4):995–1004.
Almanza A, Carlesso A, Chintha C, Creedican S, Doultsinos D, Leuzzi B, et al. Endoplasmic reticulum stress signalling - from basic mechanisms to clinical applications. Febs J. 2019;286(2):241–78.
Inagi R. Endoplasmic reticulum stress in the kidney as a novel mediator of kidney injury. Nephron Exp Nephrol. 2009;112(1):e1–9.
Ni L, Yuan C, Wu X. Endoplasmic reticulum stress in diabetic nephrology: Regulation, pathological role, and therapeutic potential. Oxid Med Cell Longev. 2021;2021:7277966.
Peyrou M, Cribb AE. Effect of endoplasmic reticulum stress preconditioning on cytotoxicity of clinically relevant nephrotoxins in renal cell lines. Toxicol Vitro. 2007;21(5):878–86.
Huang Z, Guo F, Xia Z, Liang Y, Lei S, Tan Z, et al. Activation of GPR120 by TUG891 ameliorated cisplatin-induced acute kidney injury via repressing ER stress and apoptosis. Biomed Pharmacother. 2020;126:110056.
Dong G, Liu Y, Zhang L, Huang S, Ding HF, Dong Z. mTOR contributes to ER stress and associated apoptosis in renal tubular cells. Am J Physiol Ren Physiol. 2015;308(3):F267–74.
Linkermann A. Nonapoptotic cell death in acute kidney injury and transplantation. Kidney Int. 2016;89(1):46–57.
Pabla N, Dong Z. Cisplatin nephrotoxicity: Mechanisms and renoprotective strategies. Kidney Int. 2008;73(9):994–1007.
Wei Q, Dong G, Chen JK, Ramesh G, Dong Z. Bax and bak have critical roles in ischemic acute kidney injury in global and proximal tubule-specific knockout mouse models. Kidney Int. 2013;84(1):138–48.
Chou YH, Pan SY, Shao YH, Shih HM, Wei SY, Lai CF, et al. Methylation in pericytes after acute injury promotes chronic kidney disease. J Clin Invest. 2020;130(9):4845–57.
Hu Z, Zhang H, Yang SK, Wu X, He D, Cao K, et al. Emerging role of ferroptosis in acute kidney injury. Oxid Med Cell Longev. 2019;2019:8010614.
Sanz AB, Sanchez-Niño MD, Ramos AM, Ortiz A. Regulated cell death pathways in kidney disease. Nat Rev Nephrol. 2023;19(5):281–99.
Zeng Y, Li S, Wu J, Chen W, Sun H, Peng W, et al. Autophagy inhibitors promoted aristolochic acid I induced renal tubular epithelial cell apoptosis via mitochondrial pathway but alleviated nonapoptotic cell death in mouse acute aritolochic acid nephropathy model. Apoptosis. 2014;19(8):1215–24.
Tan X, Zhu H, Tao Q, Guo L, Jiang T, Xu L, et al. FGF10 protects against renal ischemia/reperfusion injury by regulating autophagy and inflammatory signaling. Front Genet. 2018;9:556.
Gong L, Pan Q, Yang N, Corrigendum: Autophagy and inflammation regulation in acute kidney injury. Front Physiol. 2021;11:639938.
Krüger-Genge A, Blocki A, Franke RP, Jung F. Vascular endothelial cell biology: An update. Int J Mol Sci. 2019;20(18).
Molema G, Zijlstra JG, van Meurs M, Kamps J. Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol. 2022;18(2):95–112.
Czopek A, Moorhouse R, Gallacher PJ, Pugh D, Ivy JR, Farrah TE, et al. Endothelin blockade prevents the long-term cardiovascular and renal sequelae of acute kidney injury in mice. Sci Transl Med. 2022;14(675):eabf5074.
Wang J, Cong X, Miao M, Yang Y, Zhang J. Inhaled nitric oxide and acute kidney injury risk: A meta-analysis of randomized controlled trials. Ren Fail. 2021;43(1):281–90.
Alhayaza R, Haque E, Karbasiafshar C, Sellke FW, Abid MR. The relationship between reactive oxygen species and endothelial cell metabolism. Front Chem. 2020;8:592688.
Koivuviita N, Tertti R, Heiro M, Manner I, Metsärinne K. Thromboembolism as a cause of renal artery occlusion and acute kidney injury: The recovery of kidney function after two weeks. Case Rep Nephrol Urol. 2014;4(1):82–7.
Yau JW, Teoh H, Verma S. Endothelial cell control of thrombosis. BMC Cardiovasc Disord. 2015;15:130.
Acharya S, Anwar S, Siddiqui FS, Shabih S, Manchandani U, Dalezman S. Renal artery thrombosis in COVID-19. IDCases. 2020;22:e00968.
de Bragança AC, Volpini RA, Mehrotra P, Andrade L, Basile DP. Vitamin D deficiency contributes to vascular damage in sustained ischemic acute kidney injury. Physiol Rep. 2016;4(13).
Steegh F, Keijbeck AA, de Hoogt PA, Rademakers T, Houben A, Reesink KD, et al. Capillary rarefaction: A missing link in renal and cardiovascular disease? Angiogenesis. 2024;27(1):23–35.
Zeisberg M, Kalluri R. Physiology of the renal interstitium. Clin J Am Soc Nephrol. 2015;10(10):1831–40.
Foresto-Neto O, Menezes-Silva L, Leite JA, Andrade-Silva M, Câmara NOS. Immunology of kidney disease. Annu Rev Immunol. 2024;42(1):207–33.
Inaba A, Tuong ZK, Riding AM, Mathews RJ, Martin JL, Saeb-Parsy K, et al. B Lymphocyte-Derived CCL7 augments neutrophil and monocyte recruitment, exacerbating acute kidney injury. J Immunol. 2020;205(5):1376–84.
Gupta S, Kaplan MJ. The role of neutrophils and NETosis in autoimmune and renal diseases. Nat Rev Nephrol. 2016;12(7):402–13.
Rogers NM, Ferenbach DA, Isenberg JS, Thomson AW, Hughes J. Dendritic cells and macrophages in the kidney: A spectrum of good and evil. Nat Rev Nephrol. 2014;10(11):625–43.
Law BMP, Wilkinson R, Wang X, Kildey K, Lindner M, Rist MJ, et al. Interferon-γ production by tubulointerstitial human CD56(bright) natural killer cells contributes to renal fibrosis and chronic kidney disease progression. Kidney Int. 2017;92(1):79–88.
Neumann K, Tiegs G. Immune regulation in renal inflammation. Cell Tissue Res. 2021;385(2):305–22.
Liang L, Mi Y, Zhou S, Yang A, Wei C, Dai E. Advances in the study of key cells and signaling pathways in renal fibrosis and the interventional role of Chinese medicines. Front Pharmacol. 2024;15:1403227.
Schunk SJ, Floege J, Fliser D, Speer T. WNT-β-catenin signalling - a versatile player in kidney injury and repair. Nat Rev Nephrol. 2021;17(3):172–84.
Grande RodrÃguez M, López Blau C, Sánchez Laorden B, Frutos CAd, Boutet A, Grant Rowe R, et al. Snail1 factor behaves as a therapeutic target in renal fibrosis. 2015.
AlQudah M, Hale TM, Czubryt MP. Targeting the renin-angiotensin-aldosterone system in fibrosis. Matrix Biol. 2020;91–92:92–108.
Christov M, Waikar SS, Pereira RC, Havasi A, Leaf DE, Goltzman D, et al. Plasma FGF23 levels increase rapidly after acute kidney injury. Kidney Int. 2013;84(4):776–85.
Lu Y, Xu S, Tang R, Han C, Zheng C. A potential link between fibroblast growth factor-23 and the progression of AKI to CKD. BMC Nephrol. 2023;24(1):87.
Cho ME, Smith DC, Branton MH, Penzak SR, Kopp JB. Pirfenidone slows renal function decline in patients with focal segmental glomerulosclerosis. Clin J Am Soc Nephrol. 2007;2(5):906–13.
Chávez-Iñiguez JS, Poo JL, Ibarra-Estrada M, GarcÃa-Benavides L, Navarro-Blackaller G, Cervantes-Sánchez C, et al. Effect of Prolonged-Release Pirfenidone on renal function in septic acute kidney injury patients: A Double-Blind Placebo-Controlled clinical trial. Int J Nephrol. 2021;2021:8833278.
Acknowledgements
None.
Funding
Not applicable.
Author information
Authors and Affiliations
Contributions
JH did literature research, extracted the data, and drafted the manuscript. YF designed the research and reviewed the manuscript. All authors reviewed the manuscript. All authors read and approved the final manuscript.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
Not applicable.
Clinical trial number
Not applicable.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
He, J., Chen, Y., Li, Y. et al. Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review. BMC Nephrol 26, 144 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12882-025-04077-4
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12882-025-04077-4