Introduction
Historically, Lewis Thomas was responsible for a paradigm shift in our concept of sepsis by changing the focus from pathogens to the pathologic dysregulated host response that serves as the basis for the clinical expression of the systemic inflammatory response syndrome (1). In 1991, the American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference on sepsis and organ failure defined sepsis as infection-induced systemic inflammatory response syndrome (2). In 2016, with the Third International Census Definition for Sepsis and Septic Shock, sepsis was defined as a life-threatening organ dysfunction caused by a dysregulated host response to infection (3). Thus, the focus shifted from an emphasis on the hyperactive immune response or cytokine storm in which fluids and a vasopressor were necessary for hemodynamic support, to the importance of organ damage, a condition that complicates clinical management and increases mortality.
The standard management approaches in sepsis (fluid resuscitation, vasopressors, parental antibiotics, or implementing Surviving Sepsis Campaign Care bundles) are associated with a reduction of hospital mortality (4). The time of sepsis bundle protocol initiation is critical (5). When the 3-hour bundle of care (blood cultures, broad-spectrum antibiotic agents, and lactate measurement) was initiated within 6 hours after arrival in the emergency department and completed within 12 hours, the longer time to completion of the bundle was associated with higher mortality. Although early initiation of a care bundle was associated with reduced mortality, it is not clear whether implementing these management strategies reduces sepsis-associated AKI. A recent study of Surviving Sepsis Campaign Care bundles was disappointing in terms of reducing sepsis-associated AKI (6). Thus, a better understanding of the pathophysiology of sepsis-associated AKI and improved clinical trial design are needed.
Epidemiology
Sepsis accounts for about 50% of all patients with AKI in the intensive care unit (ICU) and is the leading cause of death in the ICU (7–9). Mortality from sepsis-associated AKI cannot be explained solely by loss of kidney function, rather AKI-induced multiorgan dysfunction, including lung, heart, brain, liver, and intestine, contributes to the overall mortality in sepsis-associated AKI (10). The mortality of patients with AKI in ICU and non-ICU settings increases with the number of failed organ systems (11). Compared with AKI with nonseptic etiologies, the pathophysiology of sepsis-associated AKI is complex because the AKI episode is also a leading cause of sepsis (12). When sepsis develops after AKI, the prognosis is poor, the hospital stay is longer, and mortality rates are higher (13). Although timely administration of antibiotics is one of the most effective interventions to reduce mortality in patients who are septic, its role in sepsis-associated AKI is unclear (6) because antibiotics are often associated with nephrotoxicity in sepsis-associated AKI (14–17). Furthermore, established approaches for septic shock (e.g., fluid management, antibiotics, and vasopressors) have failed to improve outcomes in sepsis-associated AKI (18).
Failure of clinical trials focusing on single targets such as inflammatory cytokines (TNF-α and IL-1β) (19–21) may partly be due to the failed recognition that sepsis is a heterogeneous disorder. Investigators are recognizing that subphenotypes exist (22) that could permit stratification and enrichment of populations to specific targeted interventions. In adult respiratory distress syndrome, interventions for the most part have proven ineffective. However, it is important to consider that adult respiratory distress is a heterogeneous syndrome that is characterized by subphenotypes. In two randomized clinical trials, systemic paralysis and heavy sedation (23) or prone position (24) resulted in favorable outcomes in a severe phenotype of adult respiratory distress syndrome on the basis of PaO2/FiO2 ratio (24). Recently, Chaudhary et al. used deep-learning techniques and identified three distinct subphenotypes of patients with sepsis-associated AKI by analyzing routinely measured laboratory tests and vital signs (25). These three clusters differed with regard to comorbidities, laboratory tests, vital signs, and mortality. Thus sepsis-associated AKI is a complex syndrome consisting of subphenotypes. Prognostic enrichment may reduce this heterogeneous population to more homogeneous populations of patients with sepsis-associated AKI to improve the efficacy of targeted therapeutic interventions.
The pathophysiology of sepsis-associated AKI has been reviewed recently (26–29); in this review, we discuss newly identified targets that further expand our understanding of the fundamental mechanisms of sepsis-associated AKI from the perspective of microcirculatory dysfunction, inflammation, and metabolic reprogramming (Table 1). Lastly, we describe the relationship between sepsis-associated AKI and coronavirus disease 2019 (COVID-19).
Table 1. -
Potential targets for diagnosis and treatment of sepsis-associated AKI found in preclinical studies
Target |
Disease |
Model |
Intervention |
Outcome |
Reference |
Microcirculatory dysfunction and pericyte loss
|
Neural glial 2+ pericytes |
Nonseptic AKI |
Rat, IRI |
— |
Pericyte density ↓, vascular congestion ↑ |
(55) |
FoxD1+ pericytes |
Nonseptic AKI |
Mouse, diphtheria toxin |
Pericyte ablation |
Kidney injury ↑ |
(56) |
Gli1+ pericytes |
Nonseptic AKI |
Mouse, diphtheria toxin |
Pericyte ablation |
Kidney injury ↑ |
(57) |
Friend leukemia virus integration 1 |
SA-AKI |
Mouse, CLP |
Inhibition |
Vascular leakage ↓, survival ↑ |
(58) |
miR-145a |
SA-AKI |
Mouse, CLP |
Inhibition |
Vascular leakage ↑, survival ↓ |
(59) |
PDGFRβ+ pericytes |
SA-AKI |
Swine, LPS |
— |
Pericyte-to-myofibroblast transdifferentiation ↑ |
(60) |
NLRP3 inflammasome
|
Sirtuin 3 |
SA-AKI |
Mouse, LPS |
Inhibition |
Vascular leakage ↑, survival ↓ |
(61) |
NLRP3 |
SA-AKI |
Mouse, CLP |
Inhibition |
Kidney injury ↓ |
(69) |
NLRP3 |
Nonseptic AKI |
Mouse, cisplatin |
Inhibition |
Kidney injury → |
(70) |
Panx1 |
SA-AKI |
Mouse, LPS |
Inhibition |
Kidney injury ↓ |
(71) |
P2X7 |
SA-AKI |
Rat, fecal peritonitis |
Inhibition |
Kidney IL-1β ↓ |
(72) |
miRNAs
|
miR-494 |
Nonseptic AKI |
Mouse, IRI |
Activation |
Kidney injury ↑ |
(98) |
miR-452 |
SA-AKI |
Mouse, LPS, CLP |
— |
miR-452 ↑ before kidney injury |
(92) |
miR-762, miR-144–3p |
SA-AKI |
Mouse, LPS |
— |
miR-762 ↑, miR-144–3p ↑ |
(99) |
miR-34–5b |
SA-AKI |
Human, LPS-induced HK-2 cells (in vitro) |
Inhibition/activation |
Inflammation and apoptosis ↓/↑ |
(93) |
Extracellular vesicles
|
Unknown |
SA-AKI |
Rat, CLP |
Brain-derived extracellular vesicles |
Kidney injury ↑ |
(109) |
Unknown |
Nonseptic AKI |
Mouse |
Extracellular vesicles secreted on thorax trauma |
Kidney injury ↑ |
(110) |
Thioredoxin-interacting protein |
SA-AKI |
Mouse, CLP |
M2 macrophage-derived extracellular vesicles |
Kidney injury ↓ |
(111) |
Lysine (K)-specific demethylase 6B |
SA-AKI |
Mouse, CLP |
Endothelial progenitor cell-derived extracellular vesicles |
Kidney injury ↓ |
(112) |
Unknown |
SA-AKI |
Mouse, CLP |
Extracellular vesicles from adipose tissue-derived mesenchymal stem cells |
Kidney injury ↓ |
(113) |
Autophagy and efferocytosis
|
LC3 |
Sepsis |
Mouse, LPS |
Inhibition |
Survival ↓ |
(117) |
LC3 |
Sepsis |
Mouse, CLP |
Activation |
Survival ↑, lung injury ↓ |
(118) |
Atg7 |
SA-AKI |
Mouse, LPS |
Inhibition |
Kidney injury ↑ |
(122) |
Sirtuin 6 |
SA-AKI |
Human, LPS-induced HK-2 cells (in vitro) |
Activation/inhibition |
Inflammation and apoptosis ↓/↑ |
(123) |
p53 |
SA-AKI |
Mouse, CLP |
Deacetylation |
Kidney injury ↓ |
(124) |
AIM |
Nonseptic AKI |
Mouse, IRI |
Inhibition/activation |
Kidney injury ↑/↓ |
(132) |
Cholinergic anti-inflammatory pathway
|
Vagus nerve |
Sepsis |
Mouse, LPS |
Activation |
Plasma TNF-α ↓ |
(144) |
Vagus nerve |
Nonseptic AKI |
Mouse, IRI |
Activation |
Kidney injury ↓ |
(144,145) |
Cholinergic anti-inflammatory pathway |
Nonseptic AKI |
Mouse, IRI |
Activation by pulsed ultrasound |
Kidney injury ↓ |
(146) |
Cholinergic anti-inflammatory pathway |
SA-AKI |
Mouse, CLP |
Activation by pulsed ultrasound |
Kidney injury ↓ |
(147) |
Vitamin D
|
Vitamin D receptor |
SA-AKI |
Mouse, LPS |
Inhibition/activation |
Kidney injury ↑/↓ |
(153) |
Vitamin D |
SA-AKI |
Mouse, LPS |
Deprivation |
Kidney injury ↑ |
(154) |
Vitamin D receptor |
Nonseptic AKI |
Mouse, cisplatin |
Activation/Inhibition |
Kidney injury ↓/↑ |
(158) |
Vitamin D receptor |
Nonseptic AKI |
Mouse, IRI |
Activation |
Kidney injury ↓ |
(159) |
Metabolic reprogramming and mitochondrial function
|
PPARγ coactivator-1α |
SA-AKI |
Mouse, LPS |
Inhibition |
Kidney injury ↑ |
(168) |
Drp1 |
Nonseptic AKI |
Mouse, IRI |
Inhibition |
Kidney injury ↓ |
(170) |
PINK1/PARK2 |
SA-AKI |
Mouse, LPS, CLP |
Inhibition |
Kidney injury ↑ |
(171) |
STING |
Nonseptic AKI |
Mouse, cisplatin |
Inhibition |
Kidney injury ↓ |
(172) |
cGAS |
SA-AKI |
Mouse, CLP, LPS |
Inhibition |
Kidney injury ↓ |
(173) |
Mitochondrial reactive oxygen species |
SA-AKI |
Mouse, CLP |
Antioxidation by SS-31 |
Kidney injury ↓ |
(174) |
Drp1 |
SA-AKI |
Mouse, LPS |
Inhibition by Mdivi-1 |
Kidney injury ↓ |
(76) |
IRI, ischemia-reperfusion injury; FoxD1, forkhead box D1; SA-AKI, sepsis-associated AKI; CLP, cecal ligation puncture; miRNA, microRNA; PDGFRβ, platelet-derived growth factor receptor β; NLRP3, NOD-like receptor protein 3; Panx1, pannexin-1; LC3, microtubule-associated protein 1 light chain 3; Atg7, autophagy-related gene 7; AIM, apoptosis inhibitor of macrophage; Drp1, dynamin-related protein 1; PINK1, PTEN-induced kinase 1; PARK2, Parkin RBR E3 ubiquitin protein ligase; STING, stimulator of interferon genes; cGAS, cyclic GMP-AMP synthase; SS-31, Szeto-Schiller-31.
Overview of Sepsis-Associated AKI
Activation and Suppression of the Immune System
An overview of the pathogenesis of sepsis-associated AKI is depicted in Figure 1. After bacterial infection, a hyperactive dysregulated innate immune response leads to a cascade of proinflammatory molecules activating the complement system, and cellular innate immunity that contributes to sepsis-associated AKI. Cell death pathways and immune cells are activated, leading to infiltration of T cells, macrophages, and neutrophils, and amplification of tissue injury (30–32). Early events trigger innate immunity through the host response to danger-associated molecular patterns (DAMPs; normally endogenous “hidden” intracellular molecules that are released by dying or damaged cells and activate the immune system) or pathogen-associated molecular patterns (small molecular motifs, including LPS, flagellin, double-stranded RNA, and CpG DNA) (33), which are ligands for pattern recognition receptors, such as Toll-like receptors (TLRs), NOD-like receptors (NLRs), and RIG-I–like receptors (34,35). Early deaths are a result of a hyperinflammatory “cytokine storm” response leading to circulatory collapse, lactic acidosis, and hypercatabolism. Both the initial degree of inflammatory response and early deaths depend on a number of factors, including patient comorbid conditions, genetic background, and inflammatory factors such as pathogen virulence and bacterial burden (36). Subsequently, there is mitochondrial dysfunction (37), apoptosis (38), and other forms of regulated cell-death pathways, such as ferroptosis, necroptosis, and pyroptosis (39).
Figure 1.: Overview of the pathophysiology of sepsis-associated AKI. Key factors that provide pro- or anti-inflammatory effects on the kidneys during sepsis contribute to the pathophysiology of sepsis-associated AKI. DAMPs, danger-associated molecular patterns; PAMPs, pathogen-associated molecular patterns.
Events later in sepsis-associated AKI include organ crosstalk and pronounced immunosuppression (10,36,40,41). Spleens and lungs, harvested 30–180 minutes after death from patients who had multiple organ failures (circulatory, hepatic, renal), and succumbed in the ICU, had immune cells with decreased production of pro- and anti-inflammatory cytokines (41,42). Reduced cytokine secretion including TNF-α, IL-6, and IL-10, and increased regulatory T cells, a feature of T cell exhaustion, were observed in the spleen (42). Lung epithelial cells also showed a severe increase in ligands or molecules for inhibitory receptors, such as programmed cell death ligand 1 (42); binding to its target inhibitory receptor, PD-1, dampens the immune response. These results characterize a condition of profound immunosuppression and evidence for T cell exhaustion in the late phase of sepsis (41,42) and provide the rationale for immunotherapy and the use of immune checkpoint inhibitors in patients with sepsis-associated immunosuppression (41). However, effective immunotherapeutic strategies regulating inflammation during sepsis have not been established in clinical use, but are underway (43).
Microcirculatory Dysfunction and Pericyte Loss
Previously, sepsis-associated AKI was thought to be due to a disorder of macrocirculation in which hypotension and decreased kidney blood flow (RBF) resulted in acute tubular necrosis (44). The predominant causal feature in sepsis-associated AKI was thought to be generalized arterial vasodilation associated with a decrease in systemic vascular resistance (45). In sepsis-associated AKI, decreased RBF is a consistent finding in rodent models, but there is now substantial evidence that global RBF is preserved or even increased during the development of kidney injury in large animals and humans (28,46,47). In systematic analysis, RBF was increased in three human studies of sepsis, but decreased in 62% of 159 animal studies or had either no change or increased in 38% of these animal studies (48). These observations emphasize mechanisms of injury despite maintained RBF incuding the importance of the microcirculation. Microvascular function is a major contributor to the microcirculation (26), and, despite maintenance of sustained RBF, altered microcirculatory flow may lead to altered kidney tissue hypoperfusion. Using intravital video microscopy, Wu et al. demonstrated that peritubular capillary flow was compromised in LPS-induced sepsis (49). Peritubular capillary flow decreased by 88%, intermittent flow increased by 400%, and no flow increased by approximately 300%. Using multiparametric photo-acoustic microscopy, a novel tool that can measure real-time changes in microhemodynamics and oxygen metabolism, our group showed that sepsis induced an acute and significant reduction in peritubular capillary oxygen saturation of hemoglobin, concomitant with a marked reduction in kidney ATP levels (50). These intravital studies demonstrate that sepsis altered microvascular flow, resulting in tissue hypoxia and compromised bioenergetics. The microvascular dysfunction is heterogeneous and relates to patchy areas of leukostasis, with associated changes in oxygen extraction (metabolic rate of oxygen) (49,50). These studies show local microvascular dysfunction and patchy areas of oxygen saturation, likely related to endothelial dysfunction and inflammatory and other oxidative factors (51).
Although much has been written about the inflamed local microenvironment and its contribution to microvascular dysfunction during sepsis (29,49,50,52) (Figure 2), an understanding of the contribution of pericytes to the pathophysiology of sepsis-associated AKI is still lacking. Pericytes, which intermittently surround capillary endothelial cells to maintain microvascular homeostasis including blood flow and vascular permeability, are considered the main source of myofibroblasts in CKD with progressive fibrosis (53,54). One study using a rat AKI model demonstrated that pericyte density in the kidney medulla decreased after ischemia-reperfusion, which induced vascular congestion (55). Using a genetic model to selectively ablate >90% of kidney pericytes but no other cell lineages, there was worsening AKI, lipid accumulation, and apoptosis of tubule epithelial cells (56). Detachment of Gli1+ cells, a major subset of pericytes, from endothelial cells after AKI was associated with endothelial cell damage and reduced capillary number (57). These results suggest pericytes could be implicated in the heterogeneity of microcirculatory abnormalities during the progression of sepsis-associated AKI (Figure 2), as supported by recently identified molecular mechanisms underlying sepsis-associated dysfunction of kidney pericytes. For instance, the transcriptional factor Friend leukemia virus integration 1 as a target of miR-145a promoted kidney pericyte loss and vascular leakage via activation of NF-κB signaling in a murine cecal ligation puncture model (58,59). In a swine model of LPS-induced AKI, pericyte-to-myofibroblast transdifferentiation was observed within 9 hours after LPS challenge and might be mediated by the secretion of LPS-binding protein and subsequent stimulation of TLR4 signaling in pericytes (60). Moreover, global sirtuin 3 (which controls key metabolic pathways by activating mitochondrial enzymes involved in the respiratory chain, in ATP production, and in both the citric acid and urea cycles) knockout mice exhibited increased vascular permeability in the lung, heart, kidney, and brain after LPS treatment (61). Angiopoietins/Tie-2 and hypoxia-inducible factor-2α/Notch3 signaling were identified as downstream pathways of sirtuin 3, leading to LPS-induced pericyte loss in the lung; the same pathways may function in the kidney. Further investigation of the relationship between pericyte-mediated regulation of microvasculature and GFR reduction during the development of sepsis is necessary to better understand sepsis-associated AKI.
Figure 2.: Microcirculatory dysfunction and pericyte loss. A possible model of microcirculatory dysfunction resulting from activation of adhesion molecules leading to leukostasis, degranulation of leukocytes, and release of chemokines, cytokines, and reactive oxygen species. Endothelial injury and pericyte loss develop during sepsis-associated AKI. Leukostasis leads to regions of hypoxia and further injury. During sepsis, detachment of pericytes and pericyte-to-myofibroblast transdifferentiation are promoted in the capillaries around the proximal tubules, which could further induce immune cell infiltration into the interstitial space, leukostasis, and hypoxia, even in the early phase of infection. These events might be a leading cause of microcirculatory abnormalities and subsequent kidney injury.
Inflammation
NOD-Like Receptor Protein 3 Inflammasome
Disruption of the innate immune system has profound effects on sepsis-induced organ dysfunction (27,30,62). In response to infection, pathogen-associated molecular patterns and DAMPs bind to TLRs on the cell surface and promote the release of proinflammatory cytokines (33,63–66). In addition to these factors, the cytoplasmic NOD-like receptor protein 3 (NLRP3) inflammasome is integral to the inflammatory cascade in sepsis-associated AKI (65–77) (Figure 3). The NLRP3 inflammasome activates caspase-1 facilitating the maturation of IL-1β and IL-18, which exacerbates inflammation (65,66,78,79). In a mouse model of sepsis, NLRP3 knockout reduced caspase-1 and IL-1β/IL-18 levels in the kidney and attenuated kidney injury (69). In contrast, a protective effect of NLRP3 knockout was not observed in a cisplatin-induced AKI model (70), suggesting that the activation of NLRP3 inflammasome during the development of kidney injury may be context dependent. Furthermore, recent investigations have shown that cellular energy metabolism, especially in ATP metabolism, is pivotal for regulating the NLRP3 inflammasome (71,72). Pannexin-1, a transmembrane channel for the transport of ATP and other small molecules, was highly expressed in the kidney tissue of LPS-treated mice and patients with sepsis-associated AKI, and its inhibition prevented the activation of the NLRP3 inflammasome (71). The administration of an ATP-gated P2X7 receptor antagonist (A-438079) also decreased kidney IL-1β in a rat model of sepsis (72). Because extracellular ATP released from pannexin-1 or from injured cells can stimulate P2X7 as a DAMP (66,67), pannexin-1 and P2X7 could play a role in the transduction of inflammatory signaling to neighboring cells. Modulators of the NLRP3 inflammasome (sirtuin 1, sirtuin 3, parkin, and dynamin-related protein 1) in experimental models of murine sepsis were mostly associated with mitochondrial function, the main source of ATP (73–77). In summary, the NLRP3 inflammasome is central to the inflammatory cascade in sepsis-activated innate immunity and should be considered as an important target for therapeutic intervention in sepsis-associated AKI.
Figure 3.: NOD-like receptor protein 3 (NLRP3) inflammasome. NLRP3 inflammasome plays a central role in the promotion of inflammatory responses during sepsis. Pathogen-associated molecular patterns (PAMPs) or danger-associated molecular patterns (DAMPs) activate the NLRP3 inflammasome, which might be mediated by the disturbance of mitochondrial homeostasis and metabolic reprogramming. The extracellular release of ATP through pannexin-1 (Panx1) and its binding to the P2X7 receptor also promotes the maturation of IL-1β in the kidney. TLR, toll-like receptor.
MicroRNAs
Emerging evidence suggests that microRNAs (miRNAs), small noncoding RNAs composed of about 22 nucleotides, are critical for the pathophysiology of both sepsis and AKI (80–84). miRNAs repress target protein expression at the post-transcriptional level, which controls cellular functions such as cell development, differentiation, metabolism, inflammatory responses, and cell death (80–84). The clinical importance of miRNAs is the emergence of their role in disease states including sepsis-associated AKI. Previously, clinical studies unsuccessfully targeted single molecules (e.g., TNF-α), but we now know that a single RNA makes more than one protein. Thus, one miRNA candidate has the ability to regulate entire pathogenic biologic pathways (85), potentially rendering the candidate miRNA more effective. Recent clinical studies have identified several miRNAs that are increased or decreased in patients with sepsis-associated AKI (86–96) (Table 2). Several miRNAs (miR-4321, -4270, -15a–5p, -15b–5p, and -16–5p) are involved in the phosphatidylinositol-3-kinase/Akt/mammalian target of rapamycin signaling pathway (87,94), emphasizing the importance of cellular metabolic processes in the development of sepsis-associated AKI. During kidney ischemia-reperfusion injury, miR-494 increased in the urine before the rise in creatinine and exacerbated inflammation by inhibiting activating transcription factor 3 (ATF3) (97,98). Liu et al. also showed increased miR-452, a target gene of NF-κB, in the serum and urine before kidney dysfunction in LPS- and cecal ligation puncture–induced AKI murine models (92). Tod et al. found changes in miRNAs in sepsis-associated AKI from miRNA microarray analysis of LPS-treated mice (99). In the early phase (at 1.5 and 6 hours after LPS injection), the most elevated miRNA in the kidney was miR-762, whereas the expression of miR-144–3p was significantly increased in the late phase (at 24 hours after LPS injection). Subsequent proteome analysis revealed that downstream targets of miR-762 and miR-144–3p might be the secretion-associated Ras-related GTPase 1B and aquaporin-1, respectively. Similarly, miR-34–5b, which was elevated in the serum of patients with sepsis-associated AKI, suppressed the expression of aquaporin-2 in tubular epithelial cells (93). These observations indicate that miR-494, miR-452, and miR-762 could be early predictors of sepsis-associated AKI pathogenesis, and regulation of aquaporin expression by miR-144–3p and miR-34–5b might be critical for the homeostasis of kidney microcirculation. Although miRNAs are not Food and Drug Administration approved for clinical intervention, miRNA candidate drugs are in clinical development in phase 1 and phase 2 trials (100).
Table 2. -
MicroRNAs in sepsis-associated AKI
MicroRNA |
Sample |
Control |
Target Pathway |
Reference |
Title of the Reference |
Increased
|
miR-107 |
CECs |
Healthy volunteers, nonseptic AKI, septic non-AKI |
DUSP7 |
(86) |
MiR-107 induces TNF-α secretion in endothelial cells causing tubular cell injury in patients with septic AKI |
miR-4321 |
Serum |
Healthy volunteers, septic non-AKI |
AKT1, mTOR, NOX5 |
(87) |
Differentially expressed miRNAs in sepsis-induced AKI target oxidative stress and mitochondrial dysfunction pathways |
miR-4270 |
Serum |
Healthy volunteers, septic non-AKI |
PPARGC1A, AKT3, NOX5, PIK3C3, WNT1 |
(87) |
Differentially expressed miRNAs in sepsis-induced AKI target oxidative stress and mitochondrial dysfunction pathways |
miR-29a |
Serum |
Septic non-AKI |
Unknown |
(88) |
Predictive value of miRNA-29a and miRNA-10a–5p for 28-day mortality in patients with sepsis-induced AKI |
miR-10a-5p |
Serum |
Septic non-AKI |
Unknown |
(88) |
Predictive value of miRNA-29a and miRNA-10a–5p for 28-day mortality in patients with sepsis-induced AKI |
miR-26b |
Urine |
Septic non-AKI |
Unknown |
(89) |
Urinary miR-26b as a potential biomarker for patients with sepsis-associated AKI: a Chinese population-based study |
miR-210 |
Plasma |
Healthy volunteers |
Unknown |
(90) |
Expression patterns and prognostic value of miR-210, miR-494, and miR-205 in middle-aged and old patients with sepsis-induced AKI |
miR-494 |
Plasma |
Healthy volunteers |
Unknown |
(90) |
Expression patterns and prognostic value of miR-210, miR-494, and miR-205 in middle-aged and old patients with sepsis-induced AKI |
miR-152–3p |
Serum |
Healthy volunteers |
ERRFI1, STAT3 |
(91) |
A novel role of the miR-152–3p/ERRFI1/STAT3 pathway modulates the apoptosis and inflammatory response after AKI |
miR-452 |
Serum, Urine |
Healthy volunteers, septic non-AKI |
Unknown |
(92) |
Discovery and validation of miR-452 as an effective biomarker for AKI in sepsis |
miR-34b–5p |
Serum |
Healthy volunteers |
AQP2 |
(93) |
miR-34b–5p promotes renal cell inflammation and apoptosis by inhibiting AQP2 in sepsis-induced AKI |
Decreased
|
miR-205 |
Plasma |
Healthy volunteers |
Unknown |
(90) |
Expression patterns and prognostic value of miR-210, miR-494, and miR-205 in middle-aged and old patients with sepsis-induced AKI |
miR-15a–5p |
Serum |
Healthy volunteers, septic non-AKI |
mTOR signaling |
(94) |
The miR-15a–5p-XIST-CUL3 regulatory axis is important for sepsis-induced AKI |
miR-15b–5p |
Serum |
Healthy volunteers, septic non-AKI |
mTOR signaling |
(94) |
The miR-15a–5p-XIST-CUL3 regulatory axis is important for sepsis-induced AKI |
miR-16–5p |
Serum |
Healthy volunteers, septic non-AKI |
PI3K/AKT/mTOR signaling |
(94) |
The miR-15a–5p-XIST-CUL3 regulatory axis is important for sepsis-induced AKI |
miR-376b |
Urine |
Healthy volunteers, septic non-AKI |
NFKBIZ |
(95) |
The negative feedback loop of NF-κB/miR-376b/NFKBIZ in septic AKI |
miR-150–5p |
Serum |
Healthy volunteers |
MEKK3/JNK pathway |
(96) |
MiR-150–5p protects against septic acute kidney injury via repressing the MEKK3/JNK pathway |
miRNA, microRNA; CECs, circulating endothelial cells; DUSP7, dual-specificity phosphatase 7; AKT1, AKT serine/threonine kinase 1; mTOR, mammalian target of rapamycin; NOX5, NADPH oxidase; PPARGC1A, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; PIK3C3, phosphatidylinositol 3-kinase catalytic subunit type 3; WNT1, WNT family member 1; ERRFI1, ERBB receptor feedback inhibitor 1; STAT3, signal transducer and activator of transcription 3; AQP2, aquaporin-2; PI3K, phosphatidylinositol-3-kinase; NFKBIZ, NF-κB inhibitor ζ; MEKK3, mitogen-activated protein kinase 3; JNK, c-Jun N-terminal kinase.
Extracellular Vesicles
There is a growing interest in extracellular vesicles as potential biomarkers and bioregulators of kidney diseases (101–106) (Figure 4). Extracellular vesicles are lipid bilayer-enclosed particles that can be released from most cell types and contain a variety of molecules, such as DNAs, RNAs, miRNAs, proteins, lipids, and other cytosolic components. They can be classified into three types according to their biogenesis and size: exosomes (30–100 nm), microvesicles (100–1000 nm), and apoptotic bodies (50–5000 nm). The biologic characteristics of extracellular vesicles vary across the cell types of origins, thus extracellular vesicles can have not only diagnostic value, but also functional (e.g., proinflammatory) and therapeutic (e.g., anti-inflammatory) effects on neighboring cells or distant organs. Changes in extracellular vesicle counts and their components have been detected during sepsis, AKI (103–106), and sepsis-associated AKI. For example, compared with healthy volunteers and patients who are septic without AKI, patients with sepsis-associated AKI showed higher levels of urinary extracellular vesicles containing ATF3 in the early phase of sepsis (107). This increase is not consistent with the fact that ATF3 expression is repressed by miR-494, and urinary miR-494 was elevated during AKI (97,98); however, the finding that miRNAs contribute to the determination of extracellular vesicle release and intracellular retention (108) may be the key to clarifying the relationship between miR-494 and exosomal ATF3. Lin et al. demonstrated the functional effect of brain-derived extracellular vesicles (mainly from neurons and astrocytes) on kidney tissue in sepsis; treatment with brain-derived extracellular vesicles exacerbated sepsis-induced injury and apoptosis in the kidney of rats (109). Moreover, Seibold et al. suggested that extracellular vesicles secreted on thorax trauma are strongly associated with sepsis and biochemical markers of AKI (110). The authors further showed the thorax trauma–responding extracellular vesicles might originate from endothelial cells and contain endothelial activators, such as vascular cell adhesion molecule 1, E‐selectin, and cytokines (110). In contrast, there is some evidence for therapeutic roles of extracellular vesicles during sepsis-associated AKI. In a murine sepsis model, miR‐93–5p–expressing extracellular vesicles derived from M2 (anti-inflammatory) macrophages and endothelial progenitor cells protected the kidney against injury (111,112). In addition, extracellular vesicles from adipose tissue–derived mesenchymal stem cells reduced sepsis-induced kidney cell injury, but the principal component of extracellular vesicles responsible for the protection of the kidneys has not been identified (113).
Figure 4.: Extracellular vesicles. Shown here are examples of sepsis-associated AKI-associated extracellular vesicles that are identified from recent studies. The origins of extracellular vesicles are heterogeneous. Extracellular vesicles provide diagnostic, functional, and therapeutic roles in sepsis-associated AKI. Urinary exosomes (smaller extracellular vesicles) containing activating transcription factor 3 (ATF3) are upregulated in patients with sepsis-associated AKI and are an example of an extracellular vesicle biomarker in AKI. Brain- and plasma-derived (on thorax trauma) extracellular vesicles promote kidney inflammation, an example of extracellular vesicle functional role. In addition, extracellular vesicles from M2 macrophages, endothelial progenitor cells, and adipose tissue-derived mesenchymal stem cells induce an anti-inflammatory effect on the kidney. The latter also indicates a regenerative or therapeutic role. VCAM-1, vascular cell adhesion molecule 1.
Cell-based therapies for clinical disorders have led the way to extracellular vesicle research and the potential for the use of extracellular vesicles therapeutically. Extracellular vesicles therapies offer a number of advantages over cell-based therapies, including less immunogenicity, longer shelf life, lack of replication, and reduced tumor risk (114). Despite these advantages, there are still gaps in our knowledge that require continued investigations; for example, we need to better understand precise cell targeting properties and targeting uptake mechanisms to deliver to specific intracellular sites. Furthermore, because extracellular vesicles are a heterogeneous population, extracellular vesicle subphenotypes could offer distinct therapeutic advantages (114).
Autophagy and Efferocytosis
Autophagy is a conserved process by which damaged internal organelles and cytoplasmic components, such as mitochondria, are processed by lysosomal degradative pathways to facilitate cell survival; a successful and limited autophagy response triggers an anti-inflammatory response and tissue repair (115,116) (Figure 5). The autophagosome complex consists of serine/threonine protein kinases ULK1, ULK2, the class III phosphatidylinositol-3–kinase complex, two ubiquitin-like conjugation systems, the ATG12-ATG5-ATG16L system, and the microtubule-associated protein 1 light chain 3 (MAP1LC3 [LC3]) system (115,116). Mice that are deficient in the autophagy LC3 gene are more susceptible to sepsis-induced mortality (117). LC3 overexpression improved survival and reduced lung injury through increased autophagosome clearance (118). Autophagy is activated during sepsis and AKI (119,120) and plays a protective role in sepsis-associated AKI (121). Mei et al., using both pharmacologic inhibitors and mouse models, demonstrated a critical role of autophagy in sepsis-associated AKI (122). Chloroquine, an inhibitor of autophagy, worsened LPS-induced AKI. Furthermore, proximal tubule-specific autophagy-related gene 7 knockout mice displayed more severe kidney damage 24 hours after LPS treatment compared with wild-type mice. In addition, overexpression of the stress-responsive histone deacetylase, sirtuin 6, induces autophagy, inhibits apoptosis, and has a protective effect in kidney tubule cells after LPS treatment (123). Pretreatment with the autophagy agonist, rapamycin, attenuated kidney tubule damage in a mouse sepsis model, whereas administration of the autophagy inhibitors, 3-MA and chloroquine, significantly worsened kidney injury (124). Additionally, Sun et al. clarified the role of p53, which has been previously shown to play a role in various forms of AKI during sepsis-associated AKI (124,125). Although the expression of p53 protein did not change in HK-2 cells (a human cell line derived from normal proximal tubular epithelial cells) or in the kidney cortex, there was an increase in p53 translocation from the nucleus to the cytoplasm, along with an increase in p53 acetylation in sepsis models (124). Deacetylation of p53 by sirtuin 1 promoted autophagy and alleviated kidney damage in septic mice (124).
Figure 5.: Autophagy and efferocytosis. Boosting autophagy or efferocytosis has the potential to protect kidney cells against septic damage. Autophagic flux consists of a variety of proteins such as ATG12-ATG5-ATG16L system and the microtubule-associated protein 1 light chain 3 (MAP1LC3 [LC3]) system. Among these factors, LC3 and autophagy-related gene 7 (Atg7), which are involved in the maturation of an autophagosome, are suggested to ameliorate lung and kidney injury during sepsis, respectively. Sirtuin 6 and deacetylated p53 by sirtuin 1 facilitate the elongation of a phagophore, and their activation also leads to kidney protection. As well as autophagy, efferocytosis can provide anti-inflammatory effects via the removal of dying cells. During AKI, apoptosis inhibitor of macrophage (AIM) dissociated from IgM is filtered through the glomerulus followed by the binding with dying cells. The complex of AIM and dying cells then interact with kidney injury molecule–1 (KIM-1) expressed on the tubular cells. Cells engulf these complexes, which attenuates sepsis-induced damage. BCR, B cell receptor; PI3K, phosphatidylinositol 3-kinase; mTOR, mammalian target of rapamycin; ULK1, unc-51 like autophagy activating kinase; MAPK, mitogen-activated protein kinase; ERK, extracellular signal–regulated kinase.
Efferocytosis refers to the clearance of dead or dying cells by professional or nonprofessional phagocytes (126–128) (Figure 5). Apoptosis, necroptosis, pyroptosis, or ferroptosis are types of programmed cell death, depending on specific conditions; once activated, cellular clearance is necessary. Dying cells need to be removed quickly to prevent the release of DAMPs, which exacerbate inflammation resulting in worsening tissue damage (126–128). For apoptotic cells, exposure of phosphatidylserine on cell membranes leads to engulfment by phagocytic cells. Inflammatory mediators released by dying cells during AKI (primarily proximal tubule cells) attract infiltrating immune cells and exacerbate tissue injury. Specific molecules that are involved in the recognition of necrotic cells by phagocytes assist in resolution of AKI. Apoptosis inhibitor of macrophage (AIM) is a member of the scavenger receptor cysteine-rich domain superfamily. Most circulating AIM is bound to IgM pentamers, which prevents its kidney excretion (129–131). However, during AKI, AIM dissociates from IgM, through unknown mechanisms, and is filtered through the glomerulus (132). AIM then accumulates on necrotic cell debris within the kidney proximal tubules and binds to kidney injury molecule–1 (KIM-1), a well-known urinary biomarker of tubular injury (132–134). This interaction enhances KIM-1–mediated phagocytic removal of the debris. In AIM knockout mice, there is a decrease in necrotic cell clearance and persistent kidney inflammation after AKI, leading to higher mortality due to progressive kidney dysfunction. Recombinant AIM resulted in removal of debris, with improved kidney pathology after AKI (132). These results suggest the potential therapeutic benefit of enhancing efferocytosis of dying cells to improve outcomes in sepsis-associated AKI (135).
Inflammatory Reflex Pathway
Neuroimmunomodulation plays a key role during inflammation (136,137). Inflammatory signals in peripheral tissues are transmitted by afferent fibers of vagus nerve toward the central nervous system, which activates the cholinergic anti-inflammatory pathway through vagus efferent fibers (138–140) (Figure 6). Various studies have demonstrated that an anti-inflammatory effect of that pathway is mediated by splenic nerve, CD4+ T cells expressing choline acetyltransferase, and α7 nicotinic acetylcholine receptor–positive macrophages in the spleen (136–143). Recently, Inoue et al. showed that electrical vagus nerve stimulation significantly reduced plasma TNF-α levels elicited by LPS administration and protected the kidney from ischemia-reperfusion injury (144). Furthermore, the precise neural circuits involved in vagus nerve stimulation–induced kidney protection were identified; efferent or afferent stimulation was adequate to protect the kidney, and afferent vagus nerve stimulation activated the C1 neurons (in the medulla oblongata)–sympathetic nervous system–splenic nerve–spleen–kidney axis (145). We also found that pulsed ultrasound activates the cholinergic anti-inflammatory pathway and ameliorates mouse kidney damage in both an ischemia-reperfusion injury model and a sepsis model of AKI (146,147). These findings indicate that strategies for the activation of the inflammatory reflex pathway have great potential to attenuate ischemic AKI and sepsis-associated AKI. There are ongoing clinical trials evaluating the protective effect of vagus nerve stimulation on inflammatory diseases (e.g., rheumatoid arthritis [148] and inflammatory bowel diseases [149]) and the approved clinical usage of ultrasound devices. Pulsed ultrasound using Food and Drug Administration–approved parameters is portable and nonpharmacologic, thus avoiding off-target side effects.
Figure 6.: Inflammatory reflex pathway. The original description of the inflammatory reflex consisted of afferent vagus nerve fibers transmitting danger signals to the nucleus tractus solitarius (NTS) from the periphery, efferent vagus nerve arising in the dorsal motor nucleus of the vagus (DMV), splenic nerve, norepinephrine (NE) release in the spleen, activation of choline acetyltransferase (ChAT)–positive T cells expressing
β2-adrenergic receptors, and activation of
α7 nicotinic acetylcholine receptors (
α7nAChRs) on macrophages, leading to suppression of proinflammatory cytokine release and reduced inflammation and/or tissue injury. Recent studies have shown that the reflex is much more complicated than previously thought. For example, in addition to activation of the cholinergic anti-inflammatory pathway (CAP) by a vagal preganglionic efferent pathway, stimulation of brainstem C1 neurons, which protects kidneys from ischemia-reperfusion injury (IRI), elicits activation of the CAP
via a sympathetic efferent pathway (
191) that involves known C1 projections to sympathetic preganglionic neurons. These innervate sympathetic ganglia, such as the celiac and suprarenal ganglia, or
via other C1 projections in the brain that would stimulate a sympathetic pathway (
145,
191). Either electrical vagus nerve stimulation (
144,
145) or pulsed ultrasound (
146,
147) is thought to activate the inflammatory reflex pathway and attenuate inflammation and AKI and sepsis-associated AKI. Dashed lines represent unconfirmed pathways, and solid lines represent confirmed pathways.
α7nAChR, alpha 7 nicotinic acetylcholine receptors; ACh, acetylcholine.
Vitamin D
The main circulating metabolite of vitamin D is 25-hydroxyvitamin D, which serves as a marker for evaluation of vitamin D status and is metabolized in the kidney by the enzyme 25-hydroxyvitamin D-1α–hydroxylase to its active form, 1,25-dihydroxyvitamin D (150,151). Accumulating evidence suggests an association between vitamin D actions and anti-inflammation in the kidney (152–154). In humans, vitamin D deficiency is highly prevalent in patients who are critically ill and has been associated with increased rates of sepsis, an observation that provides the rationale for its use in sepsis-associated AKI (155). In preclinical studies, vitamin D and its receptor are also considered to be a therapeutic target for kidney injury (156,157). In a murine LPS-induced AKI model, genetic deletion of vitamin D receptor induced higher levels of serum creatinine, proinflammatory cytokines, such as TNF-α and IL-1β in the cortex, and tubular cell apoptosis than littermate controls (153). These alterations during LPS-induced AKI have also been observed in mice fed a vitamin D–deficient diet (154). Furthermore, pretreatment with vitamin D analogs (paricalcitol, calcitriol, and cholecalciferol) before exposure to various causes of AKI including LPS, cisplatin, and ischemia-reperfusion protects kidneys against injury in these experimental preclinical models and could serve as a potentially promising therapeutic approach (153,154,158,159). Despite the results of animal studies, a randomized clinical trial in patients who are critically ill with sepsis demonstrated that a single administration of calcitriol to patients who were septic failed to decrease plasma proinflammatory cytokines and urinary neutrophil gelatinase-associated lipocalin and KIM-1 within 48 hours compared with patients who were placebo treated (160). Although it is essential to take into account the fact that both low and high doses of vitamin D can be a risk factor for AKI (161), vitamin D administration in the early stage of sepsis might be important to prevent sepsis-associated AKI. Additional studies are necessary to determine the role of vitamin D in sepsis-associated AKI.
Metabolic Reprogramming and Mitochondrial Function
Metabolic reprogramming during sepsis-associated AKI can have a major effect on outcomes, but this process remains incompletely understood (162,163). As a key player in energy production, mitochondria contributes importantly to the pathophysiology of sepsis-associated AKI (164) (Figure 7). Under physiologic conditions, proximal tubule cells use aerobic respiration for ATP production; however, early in sepsis, cells switch to using glycolysis (162,163,165,166). This shift can serve a few functions. Aerobic glycolysis can provide sufficient energy for vital functions to avoid cell death, while also allowing for the reallocation of energy to critical processes including macromolecule synthesis. Additionally, this can reduce mitochondrial damage caused by reactive oxygen species production during oxidative phosphorylation. This early metabolic switch is followed by a late anti-inflammatory catabolic phase in which tubule cells return to using oxidative phosphorylation. Using a sepsis model in mice, mitochondrial oxygen consumption in proximal tubules 4 hours after sepsis was decreased, whereas mitochondrial content and biogenesis markers were increased (167). After 24 hours, these markers were decreased, and mitochondria displayed high respiratory capacity. Moreover, the enhancement of mitochondrial biogenesis through PPARγ coactivator-1α may be essential during recovery from sepsis-associated AKI (168). These differing metabolic adaptations in early and late sepsis-associated AKI are crucial and can determine the extent of damage, including organ dysfunction and progression to CKD. Recent studies have identified potential mechanisms of mitochondrial damage during sepsis-associated AKI (169–173). There appears to be an increase in mitochondrial fragmentation, mediated by an upregulation of the mitochondrial fission gene, dynamin-related protein 1, during sepsis (169). In support of this finding, proximal tubule-specific deletion of dynamin-related protein 1 attenuated kidney ischemia-reperfusion injury, inflammation, and programmed cell death and promoted epithelial recovery; loss of dynamin-related protein 1 preserved mitochondrial structure and reduced oxidative stress in injured kidneys (170). Dysfunctional mitochondria are normally removed by selective mitochondrial autophagy. Activation of the PTEN-induced kinase 1/Parkin RBR E3 ubiquitin protein ligase pathway of mitophagy is protective in experimental models of sepsis-associated AKI (171). Mitochondrial damage also activates the cyclic GMP-AMP synthase–stimulator of interferon genes pathway (172), and the severity of sepsis-induced AKI was lessened in cyclic GMP-AMP synthase–deficient mice (173). Furthermore, the Szeto-Schiller-31 peptide, an antioxidant, improves mitochondrial function and ameliorates AKI. In a sepsis model, Szeto-Schiller-31 restored creatinine and blood urea nitrogen and reversed the sepsis-induced increase in reactive oxygen species and decrease in ATP levels (174). Another approach has focused on targeting mitochondrial dynamics. In an LPS-induced sepsis-associated AKI model, the administration of Mdivi-1, a dynamin-related protein 1 inhibitor, improved mitochondrial function and reduced NLRP3 inflammasome-mediated pyroptosis in tubular epithelial cells (76). Together, metabolic reprogramming associated with the disturbance of mitochondrial homeostasis is clearly an important feature during sepsis-associated AKI, and strategies targeting mitochondrial function may hold therapeutic promise in sepsis-associated AKI.
Figure 7.: Overview of metabolic reprogramming and mitochondrial function in sepsis-associated AKI. The principal pathway of energy production in kidney cells switches from oxidative phosphorylation (OXPHOS) to glycolysis in the early stage of sepsis. A functional changes in mitochondria in this phase may determine the extent of kidney injury in the later stage. Mitochondrial biogenesis through PPARγ coactivator-1α (PGC-1α) and the removal of damaged mitochondrial via mitophagy mediated by the PTEN-induced kinase 1 (PINK1)/Parkin RBR E3 ubiquitin protein ligase (PARK2) may allow cells to return to using OXPHOS, followed by the recovery from injury. In contrast, abnormal mitochondrial dynamics, such as an increase in dynamin-related protein 1 (Drp1), exacerbate sepsis-associated AKI. In response to the mitochondrial damage, the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is also activated, which accelerates inflammation in kidney cells. Lastly, Szeto-Schiller-31 (SS-31; antioxidant) and Mdivi-1 (Drp1 inhibitor) have potential as therapeutic drugs for sepsis-associated AKI via the attenuation of mitochondrial damage.
Relationship between Sepsis-Associated AKI and COVID-19
In January 2020, the World Health Organization declared COVID-19 a public health emergency. Kidney involvement is a common manifestation of the disease, and >25% of patients hospitalized for COVID-19 develop AKI (175,176). Studies have shown similarities between sepsis-associated AKI and COVID-19–associated AKI (177,178). For example, in both diseases, only modest tubular and glomerular damage is seen, despite profound changes in kidney function. A recent study compared morphologic and molecular characteristics from postmortem samples of patients with sepsis-associated AKI and COVID-19–associated AKI (179). The transcriptional analysis demonstrated that both had enrichment of pathways involved in inflammation compared with nonseptic AKI. Morphologic characteristics were also similar. However, important distinctions exist. Although a cytokine storm has been described in COVID-19, a meta-analysis found that patients with sepsis had 27 times higher mean IL-6 concentrations than patients with COVID-19 (180). Another study found that levels of TNF-α, IL-8, and IL-6 were lower in patients with COVID-19 than in patients with sepsis (181). Additionally, thrombi and intravascular coagulation are features of COVID-19–associated AKI not commonly seen in sepsis-associated AKI, and concentrations of D-dimer (a fibrin degradation product after blood clot degradation) are five times higher in patients who have COVID-19 and are critically ill (180). Despite these differences, it is likely that what we have learned regarding the management of sepsis-associated AKI can be applied to COVID-19–associated AKI.
Clinical Implications
Translating preclinical research into clinical care has been a major hurdle in improving treatment for AKI (182). Although we recognize the importance of timely administration of care bundles (5), the effect on AKI requires consideration of additional approaches and factors. First, we need to better understand the pathophysiology of sepsis-associated AKI. In addition to traditional targets such as TNF-α and IL-1β, development of novel pharmacologic agents and nonpharmacologic approaches (i.e., activating the inflammatory reflex pathway [146,147]) and targeting unique pathways will be important (Figure 1). Second, it is important to realize that patients with sepsis may have AKI unrelated to sepsis. For example, antibiotics are universally administered to patients who are septic, resulting in nephrotoxic AKI. Third, the pathogenesis is complex and, although preclinical studies utilizing various animal models may be useful, it also introduces additional complexities. When examining activated gene sets from a mouse model of prerenal and intrinsic AKI, functionally unrelated signal transduction pathways were identified and were localized in different cell types in the kidney (183,184). Compared with AKI with nonseptic etiologies, the pathophysiology of sepsis-associated AKI is complex; microarray studies using various rodent models of AKI showed that the LPS-induced sepsis-associated AKI had the largest number of uniquely altered genes compared with nonseptic AKI (185). Most of these genes were associated with mitochondria, whereas unique genes in an ischemia-reperfusion–induced AKI model were enriched with oxidative stress and that in a cisplatin-induced model were ribosomal genes (185). This study underscores the complexity of the pathogenesis of AKI and the difficulty in its study, as different models affect unique gene expression patterns. Last, it is important to identify relevant human molecular targets, evaluate their efficacy in relevant animal models (186), and validate results in well-designed clinical trials (187). The National Institutes of Diabetes and Digestive and Kidney Diseases (NIDDK) Kidney Precision Medicine Project focuses in part on obtaining kidney biopsies safely in patients with AKI and on interrogating molecular targets and endophenotypes (188). Enrichment approaches through molecular and clinical phenotypes may lead to improved therapeutic precision and identify those groups with a greater likelihood of responding to treatment (22,189,190) (Figure 8).
Figure 8.: Precision approach to therapies in sepsis-associated AKI. Ineffective therapies in sepsis-associated AKI are due to the complexity of pathogenesis and the heterogeneity of the critically ill population. (A) Traditionally, clinical trials randomly assign this heterogeneous group to a therapeutic intervention with limited success. (B) Enrichment approaches through genomics, RNA sequencing, proteomics, metabolomics, and biomarkers will identify molecular endophenotypes, and clinical and laboratory parameters may identify subphenotypes that are responsive to specific targeted interventions (theoretical Drug X, Y, and Z) yielding greater likelihood of treatment response. The National Institutes of Diabetes and Digestive and Kidney Diseases (NIDDK) Kidney Precision Medicine Project (KPMP) focuses in part on obtaining kidney biopsies safely in patients with AKI to interrogate molecular targets and identify endophenotypes.
Disclosures
E. Goggins reports receiving research funding from the University of Virginia Medical Scientist Training Program. M.D. Okusa reports consultancy agreements with HemoShear and Janssen; receiving research funding from AM Pharma/Pfizer and an National Institutes of Health (NIH) research grant; receiving honoraria from UpToDate; having patents or royalties with University of Virginia Patent Office; serving in an advisory or leadership role for NIDDH/NIDDK DSMB; and having other interests or relationships with the John Bower Foundation. The remaining author has nothing to disclose.
Funding
This work was supported by NIH/NIDDK grants R01 DK085259 and R01 DK123248 (to M. Okusa) and Uehara Memorial Foundation Research Fellowship (to S. Kuwabara, 202030018), and the Medical Scientist Training Program (NIH/NIGMS grant T32 GM007267 support for E. Goggins).
Acknowledgments
The authors would like to acknowledge the careful editing by Drs. Diane Rosin and Uta Erdbrüegger (Division of Nephrology) and advice from members of the Okusa laboratory. Some of the illustrations were created with BioRender.com.
Author Contributions
M. Okusa conceptualized the study, was responsible for the funding acquisition, and provided supervision; S. Kuwabara, E. Goggins, and M. Okusa wrote the original draft and reviewed and edited the manuscript.
References
1. Thomas L: Germs. N Engl J Med 287: 553–555, 1972 10.1056/NEJM197209142871109
2. Bone RC, Sibbald WJ, Sprung CL: The ACCP-SCCM consensus conference on sepsis and organ failure. Chest 101: 1481–1483, 1992 10.1378/chest.101.6.1481
3. Schepp W, Classen M: Omeprazole in the acute treatment of gastric ulcer. Scand J Gastroenterol Suppl 166: 58–62, discussion 74–75, 1989 10.3109/00365528909091246
4. Levy MM, Dellinger RP, Townsend SR, Linde-Zwirble WT, Marshall JC, Bion J, Schorr C, Artigas A, Ramsay G, Beale R, Parker MM, Gerlach H, Reinhart K, Silva E, Harvey M, Regan S, Angus DC: The Surviving Sepsis Campaign: Results of an international guideline-based performance improvement program targeting severe sepsis. Intensive Care Med 36: 222–231, 2010 10.1007/s00134-009-1738-3
5. Seymour CW, Gesten F, Prescott HC, Friedrich ME, Iwashyna TJ, Phillips GS, Lemeshow S, Osborn T, Terry KM, Levy MM: Time to treatment and mortality during mandated emergency care for sepsis. N Engl J Med 376: 2235–2244, 2017 10.1056/NEJMoa1703058
6. Pérez-Fernández X, Sabater-Riera J, Ballus-Noguera J, Cárdenas-Campos P, Moreno-González G, Alonso-Juste V, Corral-Velez V, Gutierrez-Arámbula D, Gumucio-Sanguino V, Betbesé-Roig A, Liu KD: No impact of surviving sepsis campaign care bundles in reducing sepsis-associated acute kidney injury. Clin Nephrol 88: 105–111, 2017 10.5414/CN109134
7. Uchino S, Kellum JA, Bellomo R, Doig GS, Morimatsu H, Morgera S, Schetz M, Tan I, Bouman C, Macedo E, Gibney N, Tolwani A, Ronco C; Beginning and Ending Supportive Therapy for the Kidney (BEST Kidney) Investigators: Acute renal failure in critically ill patients: A multinational, multicenter study. JAMA 294: 813–818, 2005 10.1001/jama.294.7.813
8. Hutchins NA, Unsinger J, Hotchkiss RS, Ayala A: The new normal: Immunomodulatory agents against sepsis immune suppression. Trends Mol Med 20: 224–233, 2014 10.1016/j.molmed.2014.01.002
9. Angus DC, van der Poll T: Severe sepsis and septic shock. N Engl J Med 369: 840–851, 2013 10.1056/NEJMra1208623
10. Lee SA, Cozzi M, Bush EL, Rabb H: Distant organ dysfunction in acute kidney injury: A review. Am J Kidney Dis 72: 846–856, 2018 10.1053/j.ajkd.2018.03.028
11. Star RA: Treatment of acute renal failure. Kidney Int 54: 1817–1831, 1998 10.1046/j.1523-1755.1998.00210.x
12. Matejovic M, Chvojka J, Radej J, Ledvinova L, Karvunidis T, Krouzecky A, Novak I: Sepsis and acute kidney injury are bidirectional. Contrib Nephrol 174: 78–88, 2011 10.1159/000329239
13. Mehta RL, Bouchard J, Soroko SB, Ikizler TA, Paganini EP, Chertow GM, Himmelfarb J; Program to Improve Care in Acute Renal Disease (PICARD) Study Group: Sepsis as a cause and consequence of acute kidney injury: Program to improve care in acute renal disease. Intensive Care Med 37: 241–248, 2011 10.1007/s00134-010-2089-9
14. Morales-Alvarez MC: Nephrotoxicity of antimicrobials and antibiotics. Adv Chronic Kidney Dis 27: 31–37, 2020 10.1053/j.ackd.2019.08.001
15. Appel GB, Neu HC: The nephrotoxicity of antimicrobial agents (second of three parts). N Engl J Med 296: 722–728, 1977 10.1056/NEJM197703312961305
16. Appel GB, Neu HC: The nephrotoxicity of antimicrobial agents (first of three parts). N Engl J Med 296: 663–670, 1977 10.1056/NEJM197703242961205
17. Appel GB, Neu HC: The nephrotoxicity of antimicrobial agents (third of three parts). N Engl J Med 296: 784–787, 1977 10.1056/NEJM197704072961404
18. Angus DC, van der Poll T: Severe sepsis and septic shock. N Engl J Med 369: 2063, 2013 10.1056/NEJMc1312359
19. Opal SM, Laterre PF, Francois B, LaRosa SP, Angus DC, Mira JP, Wittebole X, Dugernier T, Perrotin D, Tidswell M, Jauregui L, Krell K, Pachl J, Takahashi T, Peckelsen C, Cordasco E, Chang CS, Oeyen S, Aikawa N, Maruyama T, Schein R, Kalil AC, Van Nuffelen M, Lynn M, Rossignol DP, Gogate J, Roberts MB, Wheeler JL, Vincent JL; ACCESS Study Group: Effect of eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: The ACCESS randomized trial. JAMA 309: 1154–1162, 2013 10.1001/jama.2013.2194
20. Osuchowski MF, Welch K, Siddiqui J, Remick DG: Circulating cytokine/inhibitor profiles reshape the understanding of the SIRS/CARS continuum in sepsis and predict mortality. J Immunol 177: 1967–1974, 2006
21. Abraham E, Glauser MP, Butler T, Garbino J, Gelmont D, Laterre PF, Kudsk K, Bruining HA, Otto C, Tobin E, Zwingelstein C, Lesslauer W, Leighton A: p55 Tumor necrosis factor receptor fusion protein in the treatment of patients with severe sepsis and septic shock. A randomized controlled multicenter trial. Ro 45-2081 Study Group. JAMA 277: 1531–1538, 1997
22. Moledina DG, Parikh CR: Phenotyping of acute kidney injury: Beyond serum creatinine. Semin Nephrol 38: 3–11, 2018 10.1016/j.semnephrol.2017.09.002
23. Papazian L, Forel JM, Gacouin A, Penot-Ragon C, Perrin G, Loundou A, Jaber S, Arnal JM, Perez D, Seghboyan JM, Constantin JM, Courant P, Lefrant JY, Guérin C, Prat G, Morange S, Roch A; ACURASYS Study Investigators: Neuromuscular blockers in early acute respiratory distress syndrome. N Engl J Med 363: 1107–1116, 2010 10.1056/NEJMoa1005372
24. Guérin C, Reignier J, Richard JC, Beuret P, Gacouin A, Boulain T, Mercier E, Badet M, Mercat A, Baudin O, Clavel M, Chatellier D, Jaber S, Rosselli S, Mancebo J, Sirodot M, Hilbert G, Bengler C, Richecoeur J, Gainnier M, Bayle F, Bourdin G, Leray V, Girard R, Baboi L, Ayzac L; PROSEVA Study Group: Prone positioning in severe acute respiratory distress syndrome. N Engl J Med 368: 2159–2168, 2013 10.1056/NEJMoa1214103
25. Chaudhary K, Vaid A, Duffy Á, Paranjpe I, Jaladanki S, Paranjpe M, Johnson K, Gokhale A, Pattharanitima P, Chauhan K, O’Hagan R, Van Vleck T, Coca SG, Cooper R, Glicksberg B, Bottinger EP, Chan L, Nadkarni GN: Utilization of deep learning for subphenotype identification in sepsis-associated acute kidney injury. Clin J Am Soc Nephrol 15: 1557–1565, 2020 10.2215/CJN.09330819
26. Molema G, Zijlstra JG, van Meurs M, Kamps J: Renal microvascular endothelial cell responses in sepsis-induced acute kidney injury. Nat Rev Nephrol 18: 95–112, 2022 10.1038/s41581-021-00489-1
27. Peerapornratana S, Manrique-Caballero CL, Gómez H, Kellum JA: Acute kidney injury from sepsis: Current concepts, epidemiology, pathophysiology, prevention and treatment. Kidney Int 96: 1083–1099, 2019 10.1016/j.kint.2019.05.026
28. Lankadeva YR, Okazaki N, Evans RG, Bellomo R, May CN: Renal medullary hypoxia: A new therapeutic target for septic acute kidney injury? Semin Nephrol 39: 543–553, 2019 10.1016/j.semnephrol.2019.10.004
29. Ma S, Evans RG, Iguchi N, Tare M, Parkington HC, Bellomo R, May CN, Lankadeva YR: Sepsis-induced acute kidney injury: A disease of the microcirculation. Microcirculation 26: e12483, 2019 10.1111/micc.12483
30. Rabb H, Griffin MD, McKay DB, Swaminathan S, Pickkers P, Rosner MH, Kellum JA, Ronco C; Acute Dialysis Quality Initiative Consensus XIII Work Group: Inflammation in AKI: Current understanding, key questions, and knowledge gaps. J Am Soc Nephrol 27: 371–379, 2016 10.1681/ASN.2015030261
31. Li L, Okusa MD: Macrophages, dendritic cells, and kidney ischemia-reperfusion injury. Semin Nephrol 30: 268–277, 2010 10.1016/j.semnephrol.2010.03.005
32. Kinsey GR, Li L, Okusa MD: Inflammation in acute kidney injury. Nephron Exp Nephrol 109: e102–107, 2008 10.1159/000142934
33. Rosin DL, Okusa MD: Dangers within: DAMP responses to damage and cell death in kidney disease. J Am Soc Nephrol 22: 416–425, 2011 10.1681/ASN.2010040430
34. Uematsu S, Akira S: Toll-like receptors and innate immunity. J Mol Med (Berl) 84: 712–725, 2006 10.1007/s00109-006-0084-y
35. Creagh EM, O’Neill LA: TLRs, NLRs and RLRs: A trinity of pathogen sensors that co-operate in innate immunity. Trends Immunol 27: 352–357, 2006 10.1016/j.it.2006.06.003
36. Hotchkiss RS, Karl IE: The pathophysiology and treatment of sepsis. N Engl J Med 348: 138–150, 2003 10.1056/NEJMra021333
37. Tang C, Cai J, Yin XM, Weinberg JM, Venkatachalam MA, Dong Z: Mitochondrial quality control in kidney injury and repair. Nat Rev Nephrol 17: 299–318, 2021 10.1038/s41581-020-00369-0
38. Hotchkiss RS, Nicholson DW: Apoptosis and caspases regulate death and inflammation in sepsis. Nat Rev Immunol 6: 813–822, 2006 10.1038/nri1943
39. Linkermann A, Chen G, Dong G, Kunzendorf U, Krautwald S, Dong Z: Regulated cell death in AKI. J Am Soc Nephrol 25: 2689–2701, 2014 10.1681/ASN.2014030262
40. Li X, Hassoun HT, Santora R, Rabb H: Organ crosstalk: The role of the kidney. Curr Opin Crit Care 15: 481–487, 2009 10.1097/MCC.0b013e328332f69e
41. Hotchkiss RS, Monneret G, Payen D: Sepsis-induced immunosuppression: From cellular dysfunctions to immunotherapy. Nat Rev Immunol 13: 862–874, 2013 10.1038/nri3552
42. Boomer JS, To K, Chang KC, Takasu O, Osborne DF, Walton AH, Bricker TL, Jarman SD 2nd, Kreisel D, Krupnick AS, Srivastava A, Swanson PE, Green JM, Hotchkiss RS: Immunosuppression in patients who die of sepsis and multiple organ failure. JAMA 306: 2594–2605, 2011 10.1001/jama.2011.1829
43. Hotchkiss RS, Colston E, Yende S, Angus DC, Moldawer LL, Crouser ED, Martin GS, Coopersmith CM, Brakenridge S, Mayr FB, Park PK, Ye J, Catlett IM, Girgis IG, Grasela DM: Immune checkpoint inhibition in sepsis: A phase 1b randomized, placebo-controlled, single ascending dose study of antiprogrammed cell death-ligand 1 antibody (BMS-936559). Crit Care Med 47: 632–642, 2019 10.1097/CCM.0000000000003685
44. Schrier RW, Wang W: Acute renal failure and sepsis. N Engl J Med 351: 159–169, 2004 10.1056/NEJMra032401351/2/159
45. Landry DW, Oliver JA: The pathogenesis of vasodilatory shock. N Engl J Med 345: 588–595, 2001 10.1056/NEJMra002709
46. Wan L, Langenberg C, Bellomo R, May CN: Angiotensin II in experimental hyperdynamic sepsis. Crit Care 13: R190, 2009 10.1186/cc8185
47. Langenberg C, Wan L, Egi M, May CN, Bellomo R: Renal blood flow in experimental septic acute renal failure. Kidney Int 69: 1996–2002, 2006 10.1038/sj.ki.5000440
48. Langenberg C, Bellomo R, May C, Wan L, Egi M, Morgera S: Renal blood flow in sepsis. Crit Care 9: R363–374, 2005 10.1186/cc3540
49. Wu L, Tiwari MM, Messer KJ, Holthoff JH, Gokden N, Brock RW, Mayeux PR: Peritubular capillary dysfunction and renal tubular epithelial cell stress following lipopolysaccharide administration in mice. Am J Physiol Renal Physiol 292: F261–F268, 2007 10.1152/ajprenal.00263.2006
50. Sun N, Zheng S, Rosin DL, Poudel N, Yao J, Perry HM, Cao R, Okusa MD, Hu S: Development of a photoacoustic microscopy technique to assess peritubular capillary function and oxygen metabolism in the mouse kidney. Kidney Int 100: 613–620, 2021 10.1016/j.kint.2021.06.018
51. Zafrani L, Payen D, Azoulay E, Ince C: The microcirculation of the septic kidney. Semin Nephrol 35: 75–84, 2015 10.1016/j.semnephrol.2015.01.008
52. Ince C: The central role of renal microcirculatory dysfunction in the pathogenesis of acute kidney injury. Nephron Clin Pract 127: 124–128, 2014 10.1159/000363203
53. Kramann R, Humphreys BD: Kidney pericytes: Roles in regeneration and fibrosis. Semin Nephrol 34: 374–383, 2014 10.1016/j.semnephrol.2014.06.004
54. Humphreys BD, Lin SL, Kobayashi A, Hudson TE, Nowlin BT, Bonventre JV, Valerius MT, McMahon AP, Duffield JS: Fate tracing reveals the pericyte and not epithelial origin of myofibroblasts in kidney fibrosis. Am J Pathol 176: 85–97, 2010 10.2353/ajpath.2010.090517
55. Crislip GR, O’Connor PM, Wei Q, Sullivan JC: Vasa recta pericyte density is negatively associated with vascular congestion in the renal medulla following ischemia reperfusion in rats. Am J Physiol Renal Physiol 313: F1097–F1105, 2017 10.1152/ajprenal.00261.2017
56. Lemos DR, Marsh G, Huang A, Campanholle G, Aburatani T, Dang L, Gomez I, Fisher K, Ligresti G, Peti-Peterdi J, Duffield JS: Maintenance of vascular integrity by pericytes is essential for normal kidney function. Am J Physiol Renal Physiol 311: F1230–F1242, 2016 10.1152/ajprenal.00030.2016
57. Kramann R, Wongboonsin J, Chang-Panesso M, Machado FG, Humphreys BD: Gli1
+ pericyte loss induces capillary rarefaction and proximal tubular injury. J Am Soc Nephrol 28: 776–784, 2017 10.1681/ASN.2016030297
58. Li P, Zhou Y, Goodwin AJ, Cook JA, Halushka PV, Zhang XK, Wilson CL, Schnapp LM, Zingarelli B, Fan H: Fli-1 governs pericyte dysfunction in a murine model of sepsis. J Infect Dis 218: 1995–2005, 2018 10.1093/infdis/jiy451
59. Wu Y, Li P, Goodwin AJ, Cook JA, Halushka PV, Zingarelli B, Fan H: miR-145a regulation of pericyte dysfunction in a murine model of sepsis. J Infect Dis 222: 1037–1045, 2020 10.1093/infdis/jiaa184
60. Castellano G, Stasi A, Franzin R, Sallustio F, Divella C, Spinelli A, Netti GS, Fiaccadori E, Cantaluppi V, Crovace A, Staffieri F, Lacitignola L, Grandaliano G, Simone S, Pertosa GB, Gesualdo L: LPS-binding protein modulates acute renal fibrosis by inducing pericyte-to-myofibroblast trans-differentiation through TLR-4 signaling. Int J Mol Sci 20: E3682, 2019 10.3390/ijms20153682
61. Zeng H, He X, Tuo QH, Liao DF, Zhang GQ, Chen JX: LPS causes pericyte loss and microvascular dysfunction via disruption of Sirt3/angiopoietins/Tie-2 and HIF-2α/Notch3 pathways. Sci Rep 6: 20931, 2016 10.1038/srep20931
62. Swaminathan S, Rosner MH, Okusa MD: Emerging therapeutic targets of sepsis-associated acute kidney injury. Semin Nephrol 35: 38–54, 2015 10.1016/j.semnephrol.2015.01.005
63. Ludes PO, de Roquetaillade C, Chousterman BG, Pottecher J, Mebazaa A: Role of damage-associated molecular patterns in septic acute kidney injury, from injury to recovery. Front Immunol 12: 606622, 2021 10.3389/fimmu.2021.606622
64. Dagher PC, Basile DP: An expanding role of Toll-like receptors in sepsis-induced acute kidney injury. Am J Physiol Renal Physiol 294: F1048–F1049, 2008 10.1152/ajprenal.00125.2008
65. Martinon F, Burns K, Tschopp J: The inflammasome: A molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell 10: 417–426, 2002 10.1016/s1097-2765(02)00599-3
66. Matsuda A, Jacob A, Wu R, Aziz M, Yang WL, Matsutani T, Suzuki H, Furukawa K, Uchida E, Wang P: Novel therapeutic targets for sepsis: regulation of exaggerated inflammatory responses. J Nippon Med Sch 79: 4–18, 2012
67. Roh JS, Sohn DH: Damage-associated molecular patterns in inflammatory diseases. Immune Netw 18: e27, 2018 10.4110/in.2018.18.e27
68. Gonçalves GM, Zamboni DS, Câmara NO: The role of innate immunity in septic acute kidney injuries. Shock 34[Suppl 1]: 22–26, 2010 10.1097/SHK.0b013e3181e7e69e
69. Cao Y, Fei D, Chen M, Sun M, Xu J, Kang K, Jiang L, Zhao M: Role of the nucleotide-binding domain-like receptor protein 3 inflammasome in acute kidney injury. FEBS J 282: 3799–3807, 2015 10.1111/febs.13379
70. Kim HJ, Lee DW, Ravichandran K, O Keys D, Akcay A, Nguyen Q, He Z, Jani A, Ljubanovic D, Edelstein CL: NLRP3 inflammasome knockout mice are protected against ischemic but not cisplatin-induced acute kidney injury. J Pharmacol Exp Ther 346: 465–472, 2013 10.1124/jpet.113.205732
71. Huang G, Bao J, Shao X, Zhou W, Wu B, Ni Z, Wang L: Inhibiting pannexin-1 alleviates sepsis-induced acute kidney injury via decreasing NLRP3 inflammasome activation and cell apoptosis. Life Sci 254: 117791, 2020 10.1016/j.lfs.2020.117791
72. Arulkumaran N, Sixma ML, Pollen S, Ceravola E, Jentho E, Prendecki M, Bass PS, Tam FWK, Unwin RJ, Singer M: P2X
7 receptor antagonism ameliorates renal dysfunction in a rat model of sepsis. Physiol Rep 6: e13622, 2018 10.14814/phy2.13622
73. Gao Q, Zhu H: The overexpression of sirtuin1 (SIRT1) alleviated lipopolysaccharide (LPS)-induced acute kidney injury (AKI) via inhibiting the activation of nucleotide-binding oligomerization domain-like receptors (NLR) family pyrin domain containing 3 (NLRP3) inflammasome. Med Sci Monit 25: 2718–2726, 2019 10.12659/MSM.913146
74. Zhao WY, Zhang L, Sui MX, Zhu YH, Zeng L: Protective effects of sirtuin 3 in a murine model of sepsis-induced acute kidney injury. Sci Rep 6: 33201, 2016 10.1038/srep33201
75. Gao Y, Dai X, Li Y, Li G, Lin X, Ai C, Cao Y, Li T, Lin B: Role of Parkin-mediated mitophagy in the protective effect of polydatin in sepsis-induced acute kidney injury. J Transl Med 18: 114, 2020 10.1186/s12967-020-02283-2
76. Liu R, Wang SC, Li M, Ma XH, Jia XN, Bu Y, Sun L, Yu KJ: An inhibitor of DRP1 (Mdivi-1) alleviates LPS-induced septic AKI by inhibiting NLRP3 inflammasome activation. BioMed Res Int 2020: 2398420, 2020 10.1155/2020/2398420
77. Liu JX, Yang C, Zhang WH, Su HY, Liu ZJ, Pan Q, Liu HF: Disturbance of mitochondrial dynamics and mitophagy in sepsis-induced acute kidney injury. Life Sci 235: 116828, 2019 10.1016/j.lfs.2019.116828
78. Leemans JC, Kors L, Anders HJ, Florquin S: Pattern recognition receptors and the inflammasome in kidney disease. Nat Rev Nephrol 10: 398–414, 2014 10.1038/nrneph.2014.91
79. Chen GY, Nuñez G: Sterile inflammation: Sensing and reacting to damage. Nat Rev Immunol 10: 826–837, 2010 10.1038/nri2873
80. Lee RC, Feinbaum RL, Ambros V: The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to lin-14. Cell 75: 843–854, 1993 10.1016/0092-8674(93)90529-y
81. Petejova N, Martinek A, Zadrazil J, Kanova M, Klementa V, Sigutova R, Kacirova I, Hrabovsky V, Svagera Z, Stejskal D: Acute kidney injury in septic patients treated by selected nephrotoxic antibiotic agents-pathophysiology and biomarkers-a review. Int J Mol Sci 21: E7115, 2020 10.3390/ijms21197115
82. Giza DE, Fuentes-Mattei E, Bullock MD, Tudor S, Goblirsch MJ, Fabbri M, Lupu F, Yeung SJ, Vasilescu C, Calin GA: Cellular and viral microRNAs in sepsis: Mechanisms of action and clinical applications. Cell Death Differ 23: 1906–1918, 2016 10.1038/cdd.2016.94
83. Fan PC, Chen CC, Chen YC, Chang YS, Chu PH: MicroRNAs in acute kidney injury. Hum Genomics 10: 29, 2016 10.1186/s40246-016-0085-z
84. Benz F, Roy S, Trautwein C, Roderburg C, Luedde T: Circulating microRNAs as biomarkers for sepsis. Int J Mol Sci 17: E78, 2016 10.3390/ijms17010078
85. Bartel DP: MicroRNAs: Target recognition and regulatory functions. Cell 136: 215–233, 2009 10.1016/j.cell.2009.01.002
86. Wang S, Zhang Z, Wang J, Miao H: MiR-107 induces TNF-α secretion in endothelial cells causing tubular cell injury in patients with septic acute kidney injury. Biochem Biophys Res Commun 483: 45–51, 2017 10.1016/j.bbrc.2017.01.013
87. Ge QM, Huang CM, Zhu XY, Bian F, Pan SM: Differentially expressed miRNAs in sepsis-induced acute kidney injury target oxidative stress and mitochondrial dysfunction pathways. PLoS One 12: e0173292, 2017 10.1371/journal.pone.0173292
88. Huo R, Dai M, Fan Y, Zhou JZ, Li L, Zu J: [Predictive value of miRNA-29a and miRNA-10a-5p for 28-day mortality in patients with sepsis-induced acute kidney injury]. Nan Fang Yi Ke Da Xue Xue Bao 37: 646–651, 2017
89. Zhang J, Wang CJ, Tang XM, Wei YK: Urinary miR-26b as a potential biomarker for patients with sepsis-associated acute kidney injury: A Chinese population-based study. Eur Rev Med Pharmacol Sci 22: 4604–4610, 2018 10.26355/eurrev_201807_15518
90. Lin Y, Ding Y, Song S, Li M, Wang T, Guo F: Expression patterns and prognostic value of miR-210, miR-494, and miR-205 in middle-aged and old patients with sepsis-induced acute kidney injury. Bosn J Basic Med Sci 19: 249–256, 2019 10.17305/bjbms.2019.4131
91. Ma P, Zhang C, Huo P, Li Y, Yang H: A novel role of the miR-152-3p/ERRFI1/STAT3 pathway modulates the apoptosis and inflammatory response after acute kidney injury [published online ahead of print]. J Biochem Mol Toxicol 10.1002/jbt.22540
92. Liu Z, Yang D, Gao J, Xiang X, Hu X, Li S, Wu W, Cai J, Tang C, Zhang D, Dong Z: Discovery and validation of miR-452 as an effective biomarker for acute kidney injury in sepsis. Theranostics 10: 11963–11975, 2020 10.7150/thno.50093
93. Zheng C, Wu D, Shi S, Wang L: miR-34b-5p promotes renal cell inflammation and apoptosis by inhibiting aquaporin-2 in sepsis-induced acute kidney injury. Ren Fail 43: 291–301, 2021 10.1080/0886022X.2021.1871922
94. Xu G, Mo L, Wu C, Shen X, Dong H, Yu L, Pan P, Pan K: The
miR-15a-5p-XIST-CUL3 regulatory axis is important for sepsis-induced acute kidney injury. Ren Fail 41: 955–966, 2019 10.1080/0886022X.2019.1669460
95. Liu Z, Tang C, He L, Yang D, Cai J, Zhu J, Shu S, Liu Y, Yin L, Chen G, Liu Y, Zhang D, Dong Z: The negative feedback loop of NF-κB/miR-376b/NFKBIZ in septic acute kidney injury. JCI Insight 5: 142272, 2020 10.1172/jci.insight.142272
96. Shi L, Zhang Y, Xia Y, Li C, Song Z, Zhu J: MiR-150-5p protects against septic acute kidney injury via repressing the MEKK3/JNK pathway. Cell Signal 86: 110101, 2021 10.1016/j.cellsig.2021.110101
97. Wu YL, Li HF, Chen HH, Lin H: MicroRNAs as biomarkers and therapeutic targets in inflammation- and ischemia-reperfusion-related acute renal injury. Int J Mol Sci 21: E6738, 2020 10.3390/ijms21186738
98. Lan YF, Chen HH, Lai PF, Cheng CF, Huang YT, Lee YC, Chen TW, Lin H: MicroRNA-494 reduces ATF3 expression and promotes AKI. J Am Soc Nephrol 23: 2012–2023, 2012 10.1681/ASN.2012050438
99. Tod P, Róka B, Kaucsár T, Szatmári K, Vizovišek M, Vidmar R, Fonovič M, Szénási G, Hamar P: Time-dependent miRNA profile during septic acute kidney injury in mice. Int J Mol Sci 21: E5316, 2020 10.3390/ijms21155316
100. Hanna J, Hossain GS, Kocerha J: The potential for microRNA therapeutics and clinical research. Front Genet 10: 478, 2019 10.3389/fgene.2019.00478
101. Wolf P: The nature and significance of platelet products in human plasma. Br J Haematol 13: 269–288, 1967 10.1111/j.1365-2141.1967.tb08741.x
102. Johnstone RM, Adam M, Hammond JR, Orr L, Turbide C: Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem 262: 9412–9420, 1987
103. Hu Q, Lyon CJ, Fletcher JK, Tang W, Wan M, Hu TY: Extracellular vesicle activities regulating macrophage- and tissue-mediated injury and repair responses. Acta Pharm Sin B 11: 1493–1512, 2021 10.1016/j.apsb.2020.12.014
104. Behrens F, Holle J, Kuebler WM, Simmons S: Extracellular vesicles as regulators of kidney function and disease. Intensive Care Med Exp 8[Suppl 1]: 22, 2020 10.1186/s40635-020-00306-2
105. Erdbrügger U, Le TH: Extracellular vesicles in renal diseases: More than novel biomarkers? J Am Soc Nephrol 27: 12–26, 2016 10.1681/ASN.2015010074
106. Souza AC, Yuen PS, Star RA: Microparticles: Markers and mediators of sepsis-induced microvascular dysfunction, immunosuppression, and AKI. Kidney Int 87: 1100–1108, 2015 10.1038/ki.2015.26
107. Panich T, Chancharoenthana W, Somparn P, Issara-Amphorn J, Hirankarn N, Leelahavanichkul A: Urinary exosomal activating transcriptional factor 3 as the early diagnostic biomarker for sepsis-induced acute kidney injury. BMC Nephrol 18: 10, 2017 10.1186/s12882-016-0415-3
108. Garcia-Martin R, Wang G, Brandao BB, Zanotto TM, Shah S, Kumar Patel S, Schilling B, Kahn CR: MicroRNA sequence codes for small extracellular vesicle release and cellular retention. Nature 601: 446–451, 2022, 10.1038/s41586-021-04234-3
109. Lin H, Chen H, Qi B, Jiang Y, Lian N, Zhuang X, Yu Y: Brain-derived extracellular vesicles mediated coagulopathy, inflammation and apoptosis after sepsis. Thromb Res 207: 85–95, 2021 10.1016/j.thromres.2021.09.014
110. Seibold T, Schönfelder J, Weeber F, Lechel A, Armacki M, Waldenmaier M, Wille C, Palmer A, Halbgebauer R, Karasu E, Huber-Lang M, Kalbitz M, Radermacher P, Paschke S, Seufferlein T, Eiseler T: Small extracellular vesicles propagate the inflammatory response after trauma. Adv Sci (Weinh) 8: e2102381, 2021 10.1002/advs.202102381
111. Juan CX, Mao Y, Cao Q, Chen Y, Zhou LB, Li S, Chen H, Chen JH, Zhou GP, Jin R: Exosome-mediated pyroptosis of miR-93-TXNIP-NLRP3 leads to functional difference between M1 and M2 macrophages in sepsis-induced acute kidney injury. J Cell Mol Med 25: 4786–4799, 2021 10.1111/jcmm.16449
112. He Z, Wang H, Yue L: Endothelial progenitor cells-secreted extracellular vesicles containing microRNA-93-5p confer protection against sepsis-induced acute kidney injury via the KDM6B/H3K27me3/TNF-α axis. Exp Cell Res 395: 112173, 2020 10.1016/j.yexcr.2020.112173
113. Gao F, Zuo B, Wang Y, Li S, Yang J, Sun D: Protective function of exosomes from adipose tissue-derived mesenchymal stem cells in acute kidney injury through SIRT1 pathway. Life Sci 255: 117719, 2020 10.1016/j.lfs.2020.117719
114. Murphy DE, de Jong OG, Brouwer M, Wood MJ, Lavieu G, Schiffelers RM, Vader P: Extracellular vesicle-based therapeutics: Natural versus engineered targeting and trafficking. Exp Mol Med 51: 1–12, 2019 10.1038/s12276-019-0223-5
115. Yang Z, Klionsky DJ: Eaten alive: A history of macroautophagy. Nat Cell Biol 12: 814–822, 2010 10.1038/ncb0910-814
116. Tang C, Livingston MJ, Liu Z, Dong Z: Autophagy in kidney homeostasis and disease. Nat Rev Nephrol 16: 489–508, 2020 10.1038/s41581-020-0309-2
117. Nakahira K, Haspel JA, Rathinam VA, Lee SJ, Dolinay T, Lam HC, Englert JA, Rabinovitch M, Cernadas M, Kim HP, Fitzgerald KA, Ryter SW, Choi AM: Autophagy proteins regulate innate immune responses by inhibiting the release of mitochondrial DNA mediated by the NALP3 inflammasome. Nat Immunol 12: 222–230, 2011 10.1038/ni.1980
118. Lo S, Yuan SS, Hsu C, Cheng YJ, Chang YF, Hsueh HW, Lee PH, Hsieh YC: Lc3 over-expression improves survival and attenuates lung injury through increasing autophagosomal clearance in septic mice. Ann Surg 257: 352–363, 2013 10.1097/SLA.0b013e318269d0e2
119. Kaushal GP, Shah SV: Autophagy in acute kidney injury. Kidney Int 89: 779–791, 2016 10.1016/j.kint.2015.11.021
120. Ho J, Yu J, Wong SH, Zhang L, Liu X, Wong WT, Leung CC, Choi G, Wang MH, Gin T, Chan MT, Wu WK: Autophagy in sepsis: Degradation into exhaustion? Autophagy 12: 1073–1082, 2016 10.1080/15548627.2016.1179410
121. Yin X, Xin H, Mao S, Wu G, Guo L: The role of autophagy in sepsis: Protection and injury to organs. Front Physiol 10: 1071, 2019 10.3389/fphys.2019.01071
122. Mei S, Livingston M, Hao J, Li L, Mei C, Dong Z: Autophagy is activated to protect against endotoxic acute kidney injury. Sci Rep 6: 22171, 2016 10.1038/srep22171
123. Zhang Y, Wang L, Meng L, Cao G, Wu Y: Sirtuin 6 overexpression relieves sepsis-induced acute kidney injury by promoting autophagy. Cell Cycle 18: 425–436, 2019 10.1080/15384101.2019.1568746
124. Sun M, Li J, Mao L, Wu J, Deng Z, He M, An S, Zeng Z, Huang Q, Chen Z: p53 deacetylation alleviates sepsis-induced acute kidney injury by promoting autophagy. Front Immunol 12: 685523, 2021 10.3389/fimmu.2021.685523
125. Molitoris BA, Dagher PC, Sandoval RM, Campos SB, Ashush H, Fridman E, Brafman A, Faerman A, Atkinson SJ, Thompson JD, Kalinski H, Skaliter R, Erlich S, Feinstein E: siRNA targeted to p53 attenuates ischemic and cisplatin-induced acute kidney injury. J Am Soc Nephrol 20: 1754–1764, 2009 10.1681/ASN.2008111204
126. Morioka S, Maueröder C, Ravichandran KS: Living on the edge: Efferocytosis at the interface of homeostasis and pathology. Immunity 50: 1149–1162, 2019 10.1016/j.immuni.2019.04.018
127. Doran AC, Yurdagul Jr A, Tabas I: Efferocytosis in health and disease. Nat Rev Immunol 20: 254–267, 2020 10.1038/s41577-019-0240-6
128. Boada-Romero E, Martinez J, Heckmann BL, Green DR: The clearance of dead cells by efferocytosis. Nat Rev Mol Cell Biol 21: 398–414, 2020 10.1038/s41580-020-0232-1
129. Miyazaki T, Hirokami Y, Matsuhashi N, Takatsuka H, Naito M: Increased susceptibility of thymocytes to apoptosis in mice lacking AIM, a novel murine macrophage-derived soluble factor belonging to the scavenger receptor cysteine-rich domain superfamily. J Exp Med 189: 413–422, 1999 10.1084/jem.189.2.413
130. Miyazaki T, Yamazaki T, Sugisawa R, Gershwin ME, Arai S: AIM associated with the IgM pentamer: Attackers on stand-by at aircraft carrier. Cell Mol Immunol 15: 563–574, 2018 10.1038/cmi.2017.141
131. Hiramoto E, Tsutsumi A, Suzuki R, Matsuoka S, Arai S, Kikkawa M, Miyazaki T: The IgM pentamer is an asymmetric pentagon with an open groove that binds the AIM protein. Sci Adv 4: eaau1199, 2018 10.1126/sciadv.aau1199
132. Arai S, Kitada K, Yamazaki T, Takai R, Zhang X, Tsugawa Y, Sugisawa R, Matsumoto A, Mori M, Yoshihara Y, Doi K, Maehara N, Kusunoki S, Takahata A, Noiri E, Suzuki Y, Yahagi N, Nishiyama A, Gunaratnam L, Takano T, Miyazaki T: Apoptosis inhibitor of macrophage protein enhances intraluminal debris clearance and ameliorates acute kidney injury in mice. Nat Med 22: 183–193, 2016 10.1038/nm.4012
133. Song L, Xue L, Yu J, Zhao J, Zhang W, Fu Y: Kidney injury molecule-1 expression is closely associated with renal allograft damage. Bosn J Basic Med Sci 13: 170–174, 2013 10.17305/bjbms.2013.2357
134. Han WK, Bailly V, Abichandani R, Thadhani R, Bonventre JV: Kidney injury molecule-1 (KIM-1): A novel biomarker for human renal proximal tubule injury. Kidney Int 62: 237–244, 2002
135. Arai Y, Yamaoka Y, Morioka S: Sweeping up dying cells during tissue injury. Nephron 146: 249–252, 2022, 10.1159/000517731
136. Okusa MD, Rosin DL, Tracey KJ: Targeting neural reflex circuits in immunity to treat kidney disease. Nat Rev Nephrol 13: 669–680, 2017 10.1038/nrneph.2017.132
137. Tracey KJ: Reflex control of immunity. Nat Rev Immunol 9: 418–428, 2009 10.1038/nri2566
138. Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, Wang H, Abumrad N, Eaton JW, Tracey KJ: Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature 405: 458–462, 2000 10.1038/35013070
139. Tracey KJ: The inflammatory reflex. Nature 420: 853–859, 2002 10.1038/nature01321
140. Kuwabara S, Goggins E, Tanaka S: Neuroimmune circuits activated by vagus nerve stimulation. Nephron 146: 286–290, 2022, 10.1159/000518176
141. Rosas-Ballina M, Ochani M, Parrish WR, Ochani K, Harris YT, Huston JM, Chavan S, Tracey KJ: Splenic nerve is required for cholinergic anti-inflammatory pathway control of TNF in endotoxemia. Proc Natl Acad Sci U S A 105: 11008–11013, 2008 10.1073/pnas.0803237105
142. Rosas-Ballina M, Olofsson PS, Ochani M, Valdés-Ferrer SI, Levine YA, Reardon C, Tusche MW, Pavlov VA, Andersson U, Chavan S, Mak TW, Tracey KJ: Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science 334: 98–101, 2011 10.1126/science.1209985
143. Wang H, Yu M, Ochani M, Amella CA, Tanovic M, Susarla S, Li JH, Wang H, Yang H, Ulloa L, Al-Abed Y, Czura CJ, Tracey KJ: Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature 421: 384–388, 2003 10.1038/nature01339
144. Inoue T, Abe C, Sung SS, Moscalu S, Jankowski J, Huang L, Ye H, Rosin DL, Guyenet PG, Okusa MD: Vagus nerve stimulation mediates protection from kidney ischemia-reperfusion injury through α7nAChR+ splenocytes. J Clin Invest 126: 1939–1952, 2016 10.1172/JCI83658
145. Tanaka S, Abe C, Abbott SBG, Zheng S, Yamaoka Y, Lipsey JE, Skrypnyk NI, Yao J, Inoue T, Nash WT, Stornetta DS, Rosin DL, Stornetta RL, Guyenet PG, Okusa MD: Vagus nerve stimulation activates two distinct neuroimmune circuits converging in the spleen to protect mice from kidney injury. Proc Natl Acad Sci U S A 118: e2021758118, 2021 10.1073/pnas.2021758118
146. Gigliotti JC, Huang L, Ye H, Bajwa A, Chattrabhuti K, Lee S, Klibanov AL, Kalantari K, Rosin DL, Okusa MD: Ultrasound prevents renal ischemia-reperfusion injury by stimulating the splenic cholinergic anti-inflammatory pathway. J Am Soc Nephrol 24: 1451–1460, 2013 10.1681/ASN.2013010084
147. Gigliotti JC, Huang L, Bajwa A, Ye H, Mace EH, Hossack JA, Kalantari K, Inoue T, Rosin DL, Okusa MD: Ultrasound modulates the splenic neuroimmune axis in attenuating AKI. J Am Soc Nephrol 26: 2470–2481, 2015 10.1681/ASN.2014080769
148. Koopman FA, Schuurman PR, Vervoordeldonk MJ, Tak PP: Vagus nerve stimulation: A new bioelectronics approach to treat rheumatoid arthritis? Best Pract Res Clin Rheumatol 28: 625–635, 2014 10.1016/j.berh.2014.10.015
149. Matteoli G, Boeckxstaens GE: The vagal innervation of the gut and immune homeostasis. Gut 62: 1214–1222, 2013 10.1136/gutjnl-2012-302550
150. Holick MF: Resurrection of vitamin D deficiency and rickets. J Clin Invest 116: 2062–2072, 2006 10.1172/JCI29449
151. DeLuca HF: Overview of general physiologic features and functions of vitamin D. Am J Clin Nutr 80[Suppl]: 1689S–1696S, 2004 10.1093/ajcn/80.6.1689S
152. Li N, Wang Y, Wang X, Sun N, Gong YH: Pathway network of pyroptosis and its potential inhibitors in acute kidney injury. Pharmacol Res 175: 106033, 2022 10.1016/j.phrs.2021.106033
153. Du J, Jiang S, Hu Z, Tang S, Sun Y, He J, Li Z, Yi B, Wang J, Zhang H, Li YC: Vitamin D receptor activation protects against lipopolysaccharide-induced acute kidney injury through suppression of tubular cell apoptosis. Am J Physiol Renal Physiol 316: F1068–F1077, 2019 10.1152/ajprenal.00332.2018
154. He J, Du J, Yi B, Wang J, Zhang H, Li YC, Sun J: MicroRNA-122 contributes to lipopolysaccharide-induced acute kidney injury via down-regulating the vitamin D receptor in the kidney. Eur J Clin Invest 51: e13547, 2021 10.1111/eci.13547
155. Cameron LK, Lei K, Smith S, Doyle NL, Doyle JF, Flynn K, Purchase N, Smith J, Chan K, Kamara F, Kidane NG, Forni LG, Harrington D, Hampson G, Ostermann M: Vitamin D levels in critically ill patients with acute kidney injury: A protocol for a prospective cohort study (VID-AKI). BMJ Open 7: e016486, 2017 10.1136/bmjopen-2017-016486
156. Lin R: Crosstalk between vitamin D metabolism, VDR signalling, and innate immunity. BioMed Res Int 1375858: 2016, 2016 10.1155/2016/1375858
157. LaFavers K: Disruption of kidney-immune system crosstalk in sepsis with acute kidney injury: Lessons learned from animal models and their application to human health. Int J Mol Sci 23: 1702, 2022 10.3390/ijms23031702
158. Hu Z, Zhang H, Yi B, Yang S, Liu J, Hu J, Wang J, Cao K, Zhang W: VDR activation attenuate cisplatin induced AKI by inhibiting ferroptosis. Cell Death Dis 11: 73, 2020 10.1038/s41419-020-2256-z
159. Li J, Xu S, Zhu JB, Song J, Luo B, Song YP, Zhang ZH, Chen YH, Zhang ZQ, Xie DD, Yu DX, Xu DX: Pretreatment with cholecalciferol alleviates renal cellular stress response during ischemia/reperfusion-induced acute kidney injury. Oxid Med Cell Longev 2019: 1897316, 2019 10.1155/2019/1897316
160. Leaf DE, Raed A, Donnino MW, Ginde AA, Waikar SS: Randomized controlled trial of calcitriol in severe sepsis. Am J Respir Crit Care Med 190: 533–541, 2014 10.1164/rccm.201405-0988OC
161. Graidis S, Papavramidis TS, Papaioannou M: Vitamin D and acute kidney injury: A two-way causality relation and a predictive, prognostic, and therapeutic role of vitamin D. Front Nutr 7: 630951, 2021 10.3389/fnut.2020.630951
162. Gómez H, Kellum JA, Ronco C:
Metabolic reprogramming and tolerance during sepsis-induced AKI. Nat Rev Nephrol 13: 143–151, 2017 10.1038/nrneph.2016.186
163. Toro J, Manrique-Caballero CL, Gómez H:
Metabolic reprogramming and host tolerance: A novel concept to understand sepsis-associated AKI. J Clin Med 10: 4184, 2021 10.3390/jcm10184184
164. Sun J, Zhang J, Tian J, Virzì GM, Digvijay K, Cueto L, Yin Y, Rosner MH, Ronco C: Mitochondria in sepsis-induced AKI. J Am Soc Nephrol 30: 1151–1161, 2019 10.1681/ASN.2018111126
165. Smith JA, Stallons LJ, Schnellmann RG: Renal cortical hexokinase and pentose phosphate pathway activation through the EGFR/Akt signaling pathway in endotoxin-induced acute kidney injury. Am J Physiol Renal Physiol 307: F435–F444, 2014 10.1152/ajprenal.00271.2014
166. Waltz P, Carchman E, Gomez H, Zuckerbraun B: Sepsis results in an altered renal metabolic and osmolyte profile. J Surg Res 202: 8–12, 2016 10.1016/j.jss.2015.12.011
167. Li Y, Nourbakhsh N, Pham H, Tham R, Zuckerman JE, Singh P: Evolution of altered tubular metabolism and mitochondrial function in sepsis-associated acute kidney injury. Am J Physiol Renal Physiol 319: F229–F244, 2020 10.1152/ajprenal.00390.2019
168. Tran M, Tam D, Bardia A, Bhasin M, Rowe GC, Kher A, Zsengeller ZK, Akhavan-Sharif MR, Khankin EV, Saintgeniez M, David S, Burstein D, Karumanchi SA, Stillman IE, Arany Z, Parikh SM: PGC-1α promotes recovery after acute kidney injury during systemic inflammation in mice. J Clin Invest 121: 4003–4014, 2011 10.1172/JCI58662
169. Gonzalez AS, Elguero ME, Finocchietto P, Holod S, Romorini L, Miriuka SG, Peralta JG, Poderoso JJ, Carreras MC: Abnormal mitochondrial fusion-fission balance contributes to the progression of experimental sepsis. Free Radic Res 48: 769–783, 2014 10.3109/10715762.2014.906592
170. Perry HM, Huang L, Wilson RJ, Bajwa A, Sesaki H, Yan Z, Rosin DL, Kashatus DF, Okusa MD: Dynamin-related protein 1 deficiency promotes recovery from AKI. J Am Soc Nephrol 29: 194–206, 2018 10.1681/ASN.2017060659
171. Wang Y, Zhu J, Liu Z, Shu S, Fu Y, Liu Y, Cai J, Tang C, Liu Y, Yin X, Dong Z: The PINK1/PARK2/optineurin pathway of mitophagy is activated for protection in septic acute kidney injury. Redox Biol 38: 101767, 2021 10.1016/j.redox.2020.101767
172. Maekawa H, Inoue T, Ouchi H, Jao TM, Inoue R, Nishi H, Fujii R, Ishidate F, Tanaka T, Tanaka Y, Hirokawa N, Nangaku M, Inagi R: Mitochondrial damage causes inflammation via cGAS-STING signaling in acute kidney injury. Cell Rep 29: 1261–1273.e6, 2019 10.1016/j.celrep.2019.09.050
173. Visitchanakun P, Kaewduangduen W, Chareonsappakit A, Susantitaphong P, Pisitkun P, Ritprajak P, Townamchai N, Leelahavanichkul A: Interference on cytosolic DNA activation attenuates sepsis severity: Experiments on cyclic GMP-AMP Ssnthase (cGAS) deficient mice. Int J Mol Sci 22: 11450, 2021 10.3390/ijms222111450
174. Li G, Wu J, Li R, Yuan D, Fan Y, Yang J, Ji M, Zhu S: Protective effects of antioxidant peptide ss-31 against multiple organ dysfunctions during endotoxemia. Inflammation 39: 54–64, 2016 10.1007/s10753-015-0222-1
175. Legrand M, Bell S, Forni L, Joannidis M, Koyner JL, Liu K, Cantaluppi V: Pathophysiology of COVID-19-associated acute kidney injury. Nat Rev Nephrol 17: 751–764, 2021 10.1038/s41581-021-00452-0
176. Nadim MK, Forni LG, Mehta RL, Connor Jr MJ, Liu KD, Ostermann M, Rimmelé T, Zarbock A, Bell S, Bihorac A, Cantaluppi V, Hoste E, Husain-Syed F, Germain MJ, Goldstein SL, Gupta S, Joannidis M, Kashani K, Koyner JL, Legrand M, Lumlertgul N, Mohan S, Pannu N, Peng Z, Perez-Fernandez XL, Pickkers P, Prowle J, Reis T, Srisawat N, Tolwani A, Vijayan A, Villa G, Yang L, Ronco C, Kellum JA: COVID-19-associated acute kidney injury: Consensus report of the 25th Acute Disease Quality Initiative (ADQI) Workgroup. Nat Rev Nephrol 16: 747–764, 2020 10.1038/s41581-020-00356-5
177. Kellum JA, Nadim MK, Forni LG: Sepsis-associated acute kidney injury: Is COVID-19 different? Kidney Int 98: 1370–1372, 2020 10.1016/j.kint.2020.08.009
178. Golmai P, Larsen CP, DeVita MV, Wahl SJ, Weins A, Rennke HG, Bijol V, Rosenstock JL: Histopathologic and ultrastructural findings in postmortem kidney biopsy material in 12 patients with AKI and COVID-19. J Am Soc Nephrol 31: 1944–1947, 2020 10.1681/ASN.2020050683
179. Alexander MP, Mangalaparthi KK, Madugundu AK, Moyer AM, Adam BA, Mengel M, Singh S, Herrmann SM, Rule AD, Cheek EH, Herrera Hernandez LP, Graham RP, Aleksandar D, Aubry MC, Roden AC, Hagen CE, Quinton RA, Bois MC, Lin PT, Maleszewski JJ, Cornell LD, Sethi S, Pavelko KD, Charlesworth J, Narasimhan R, Larsen CP, Rizza SA, Nasr SH, Grande JP, McKee TD, Badley AD, Pandey A, Taner T: Acute kidney injury in severe COVID-19 has similarities to sepsis-associated kidney injury: A multi-omics study. Mayo Clin Proc 96: 2561–2575, 2021 10.1016/j.mayocp.2021.07.001
180. Leisman DE, Ronner L, Pinotti R, Taylor MD, Sinha P, Calfee CS, Hirayama AV, Mastroiani F, Turtle CJ, Harhay MO, Legrand M, Deutschman CS: Cytokine elevation in severe and critical COVID-19: A rapid systematic review, meta-analysis, and comparison with other inflammatory syndromes. Lancet Respir Med 8: 1233–1244, 2020 10.1016/S2213-2600(20)30404-5
181. Kox M, Waalders NJB, Kooistra EJ, Gerretsen J, Pickkers P: Cytokine levels in critically ill patients with COVID-19 and other conditions. JAMA 324: 1565–1567, 2020 10.1001/jama.2020.17052
182. Okusa MD, Rosner MH, Kellum JA, Ronco C; Acute Dialysis Quality Initiative XIII Workgroup: Therapeutic targets of human AKI: Harmonizing human and animal AKI. J Am Soc Nephrol 27: 44–48, 2016 10.1681/ASN.2015030233
183. Shen TH, Stauber J, Xu K, Jacunski A, Paragas N, Callahan M, Banlengchit R, Levitman AD, Desanti De Oliveira B, Beenken A, Grau MS, Mathieu E, Zhang Q, Li Y, Gopal T, Askanase N, Arumugam S, Mohan S, Good PI, Stevens JS, Lin F, Sia SK, Lin CS, D’Agati V, Kiryluk K, Tatonetti NP, Barasch J: Snapshots of nascent RNA reveal cell- and stimulus-specific responses to acute kidney injury. JCI Insight 7: e146374, 2022 10.1172/jci.insight.146374
184. Xu K, Rosenstiel P, Paragas N, Hinze C, Gao X, Huai Shen T, Werth M, Forster C, Deng R, Bruck E, Boles RW, Tornato A, Gopal T, Jones M, Konig J, Stauber J, D’Agati V, Erdjument-Bromage H, Saggi S, Wagener G, Schmidt-Ott KM, Tatonetti N, Tempst P, Oliver JA, Guarnieri P, Barasch J: Unique transcriptional programs identify subtypes of AKI. J Am Soc Nephrol 28: 1729–1740, 2017 10.1681/ASN.2016090974
185. Hultström M, Becirovic-Agic M, Jönsson S: Comparison of acute kidney injury of different etiology reveals in-common mechanisms of tissue damage. Physiol Genomics 50: 127–141, 2018 10.1152/physiolgenomics.00037.2017
186. Perrin S: Preclinical research: Make mouse studies work. Nature 507: 423–425, 2014 10.1038/507423a
187. Zuk A, Palevsky PM, Fried L, Harrell Jr FE, Khan S, McKay DB, Devey L, Chawla L, de Caestecker M, Kaufman JS, Thompson BT, Agarwal A, Greene T, Okusa MD, Bonventre JV, Dember LM, Liu KD, Humphreys BD, Gossett D, Xie Y, Norton JM, Kimmel PL, Star RA: Overcoming translational barriers in acute kidney injury: A report from an NIDDK workshop. Clin J Am Soc Nephrol 13: 1113–1123, 2018 10.2215/CJN.06820617
188. Scholz H, Boivin FJ, Schmidt-Ott KM, Bachmann S, Eckardt KU, Scholl UI, Persson PB: Kidney physiology and susceptibility to acute kidney injury: Implications for renoprotection. Nat Rev Nephrol 17: 335–349, 2021 10.1038/s41581-021-00394-7
189. Soussi S, Sharma D, Jüni P, Lebovic G, Brochard L, Marshall JC, Lawler PR, Herridge M, Ferguson N, Del Sorbo L, Feliot E, Mebazaa A, Acton E, Kennedy JN, Xu W, Gayat E, Dos Santos CC; FROG-ICU; CCCTBG trans-trial group study for InFACT - the International Forum for Acute Care Trialists: Identifying clinical subtypes in sepsis-survivors with different one-year outcomes: A secondary latent class analysis of the FROG-ICU cohort. Crit Care 26: 114, 2022 10.1186/s13054-022-03972-8
190. Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M; Sepsis Definitions Task Force: Developing a new definition and assessing new clinical criteria for septic shock: For the third international consensus definitions for sepsis and septic shock (Sepsis-3). JAMA 315: 775–787, 2016 10.1001/jama.2016.0289
191. Abe C, Inoue T, Inglis MA, Viar KE, Huang L, Ye H, Rosin DL, Stornetta RL, Okusa MD, Guyenet PG: C1 neurons mediate a stress-induced anti-inflammatory reflex in mice. Nat Neurosci 20: 700–707, 2017 10.1038/nn.4526