Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 Aug 13.
Published in final edited form as: J Am Soc Nephrol. 2025 Jul 2;36(11):2259–2268. doi: 10.1681/ASN.0000000809

Molecular mechanisms of sepsis-associated acute kidney injury

Takashi Hato 1,2,3,*, Pierre C Dagher 1,*
PMCID: PMC12344547  NIHMSID: NIHMS2102361  PMID: 40601936

Abstract

Sepsis-associated acute kidney injury (AKI) is a complex pathological state driven by dynamic interactions between the host and microbes. The rapid progression and the absence of a molecular clock that stages the disease timeline make precise therapeutic interventions highly challenging. In this Review, we aim to refine the timeline of sepsis-associated AKI by dissecting key molecular events that drive disease progression and may inform therapeutic strategies. AKI, initiated by microbes or infection mimicry, involves the rapid and simultaneous activation of inflammatory and anti-inflammatory pathways. This energy-intensive response is further fueled by the loss of distinction between self and non-self, leading to excessive antiviral responses mediated by self-derived nucleic acids. The resulting metabolic burden overwhelms cellular functions, triggering the integrated stress response and profound translation shutdown. While this shutdown response may be necessary for energy preservation and for priming endogenous recovery mechanisms, prolonged inhibition of translation represents a maladaptive feature of septic AKI. Despite these challenges, the kidney exhibits remarkable resilience. Recovery relies on metabolic flexibility and stress-adaptive mechanisms, such as enhanced polyamine biosynthesis and RNA editing. Meanwhile, microbes also demonstrate metabolic adaptability, enabling them to evade host defenses and exploit the host environment. Understanding this dynamic interplay along the timeline of septic AKI is essential for developing rational therapeutic strategies.

Introduction

By 2050, infectious diseases are projected to be the world’s leading cause of death due to the emergence of antimicrobial resistance.1, 2 The combination of microbial invasion and overwhelming host immune responses often leads to sepsis syndrome and multi-organ failure. The kidney’s role in homeostasis depends on a rich blood supply and a dense network of trafficking immune cells that constantly communicate systemic cues.3 While these characteristics are vital for total body homeostasis, they also render the kidney susceptible to systemic infections.4, 5 Indeed, acute kidney injury (AKI) is a very common complication in patients with sepsis and is an independent risk factor for mortality.6

The underlying pathobiology of sepsis-associated AKI remains poorly understood due to complex host-microbial interactions occurring within tissue niches that are difficult to capture.7, 8 Since kidney biopsy is rarely performed during active infection, even basic histopathological characterization is largely absent from the literature. Although potential therapeutic targets have been identified and tested in clinical trials, none have proven effective.911 This underscores the complexity of sepsis pathobiology, in which the molecular arms race between the host and pathogen has blurred the boundaries between therapeutic advantages and disadvantages.1214 However, despite the current grim prospects of sepsis and sepsis-associated AKI, effective therapeutics may be on the horizon as our understanding of the molecular unfolding of the disease becomes more refined.

The human body can exhibit immense plasticity and resilience to overcome a variety of adverse conditions when stress responses are well-coordinated. Conversely, disease progression occurs when tissue responses become maladaptive, including the overproduction of defense molecules and the persistent shutdown of cellular metabolism. Identifying strategies to balance stress responses and elicit desired endogenous recovery mechanisms is a critical aspect of therapy development. In this Review, we highlight several key cellular and molecular pathways essential for controlling stress responses and promoting tissue recovery, with a particular emphasis on the timeline of disease progression.

Timeline of sepsis-associated AKI

Acute inflammatory phase

Sepsis-associated AKI is a complex pathological state that progresses at a fast pace.15 The rapid progression of the disease poses a major challenge to implementing stage-specific therapeutics at the bedside (Figure 1). For example, in an animal model of endotoxemia, the surge of classic NFκB-mediated inflammatory molecules is extremely short-lived (a few hours).16 While the duration of such outburst is likely longer in human sepsis, it is still estimated to occur mostly prior to the time the patient arrives for care.12 This fast and elusive early inflammatory phase may explain the failure of dozens of clinical trials targeting upstream receptors and cytokines.1721

Figure 1: Renal tissue responses in sepsis.

Figure 1:

Phases of tissue responses in the kidney following a systemic infection. Surviving sepsis is analogous to navigating all the steps without being ‘wiped out’ by a wave. The magnitude and duration of each phase vary depending on the type of infection, but the sequence of responses remains constant. These phases overlap, each involving a diverse array of molecular pathways and cell types.

The initial inflammatory response is accompanied by a marked increase in global transcription and translation rates (Figure 2).16 This process is energetically costly but necessary to meet the demand for defense molecule production and immune system activation. Even under normal conditions, maintaining transcription and translation is energetically expensive and involves multiple layers of regulation. For example, the transcription cycle consists of four distinct steps: initiation, promoter-proximal pausing, elongation, and termination. The majority of initiated transcription events (~80%) are prematurely terminated at the pausing step, particularly in genes from stress-responsive pathways.22, 23 While seemingly wasteful, this energy-demanding system is nevertheless essential for enabling a rapid response to stress and the large-scale production of defense molecules.

Figure 2. Molecular mechanisms of sepsis-associated AKI.

Figure 2.

Illustration of representative molecular pathways involved in each phase of sepsis-associated AKI. The inner circle depicts a cadre of mechanisms, while the middle and outer circles highlight select genes and systems active in each phase. The Red Queen card symbolizes concurrent microbial evolution and adaptation, which are not detailed in this diagram. Note that the size of the quadrants does not represent actual duration of each phase in sepsis.

TLRs, Toll-like receptors; SG, stress granules; Pol II CTD, RNA polymerase II C-terminal domain; uORF, upstream open reading frame; CDS, coding sequence

The rapid upregulation of defense systems comes at the cost of restricting normal metabolic function. Excessive production of proinflammatory cytokines and chemokines also causes collateral tissue damage through multiple mechanisms, including activation of the cell death pathways, hemodynamic collapse resulting in hypoxia, and en masse recruitment of immune cells, which disrupts normal tissue architecture. To mitigate self-inflicted damage, the host deploys multiple regulatory mechanisms simultaneously—rather than sequentially—from the earliest stages of infection.24, 25 These modulatory systems include the expression of inhibitory molecules (e.g., NFκB Inhibitor-α, which prevents nuclear localization of the NFκB complex, and TNF-α Induced Protein 3, which terminates NFκB activation via proteasomal degradation of cell death mediators) as well as the upregulation of mRNA decay machinery (e.g., AU-rich element binding proteins and endonucleases).16 Collectively, the early stages of sepsis are characterized by the vigorous and simultaneous activation of both proinflammatory and anti-inflammatory pathways. This “left foot braking” technique used by our genome enables the drastic overexpression of proinflammatory genes, followed by their rapid downregulation within a matter of hours. In an animal model of endotoxemia, the synthesis of classic NFκB-mediated inflammatory molecules returns to near baseline levels within 4 hours.16

Many other layers of regulatory systems are also required for maintaining cellular homeostasis and enabling rapid adaptation to various perturbations. For instance, transcription occurs in bursts, a phenomenon known as transcriptional bursting,26 which enables the brisk production of nascent transcripts. Concomitantly, multiple mechanisms counteract this pulsatile nature of gene expression. These mechanisms include transcript and protein compartmentalization (e.g., the formation of stress granules to sequester transcripts and the conversion of membrane-bound proteins into soluble decoy receptors to abort signaling), translational regulation (e.g., the use of upstream open reading frames and initiation blocks to modulate translation kinetics), and splicing (e.g., generation of stress-induced isoforms targeted for nonsense-mediated decay to shorten their half-lives), among other mechanisms.27 Each of these molecular mechanisms contributes to the cell’s ability to mount dynamic and regulated stress responses, and are potential targets for precision therapeutics in sepsis.

Self-amplification phase (Antiviral response)

Despite the multitude of fine-tuning regulatory mechanisms, severe infections and infection mimicry alike can quickly overwhelm the host metabolic system due to self-amplifying stress responses. In contrast to the early inflammatory phase, which is mediated by a variety of innate ligand-receptor interactions, the self-amplifying phase of sepsis follows a conserved pathway common to most pathogens, including bacteria and viruses (Figure 2).28 Critical factors in this pathway include non-self and self RNA and DNA, which can exacerbate sepsis pathobiology.29 Pathogen-derived nucleic acids and their unchecked amplification pose a major threat to the host. Consequently, non-self nucleic acids are continuously monitored by potent pattern recognition receptors, such as Protein Kinase R (PKR/eIF2ak2), RIG-I (DDX58), and Melanoma Differentiation-Associated Protein 5 (MDA5/IFIT1) for cytosolic RNA, as well as cyclic GMP-AMP synthase (cGAS) for cytosolic DNA sensing.30 Although distinct molecular signatures allow for the differentiation of host- and pathogen-derived nucleic acids (e.g., RIG-I preferentially recognizes 5’-triphosphorylated RNA, indicative of viral-derived RNA), these sensors primarily rely on broad, less discriminatory features such as nucleotide length, conformation (double-stranded or single-stranded), localization, and accessibility. For instance, the double-stranded RNA (dsRNA) sensors PKR and MDA5 preferentially bind to long stretches of dsRNA (longer than 33 bp and 40 bp, respectively), a feature commonly associated with viral replication.31 Conversely, the accessibility of host-derived RNA is partially masked by host RNA-binding proteins and nucleotide modifications (e.g., pseudouridine). However, these distinguishing features are not absolute, creating the risk of triggering self-amplifying inflammatory responses.

Indeed, due to the sheer volume of host nascent RNA synthesis during the early stages of infection, self-derived RNA stress, in particular self-derived dsRNA stress, does occur.32 This is because a significant portion of self-transcripts harbor repeat elements, particularly in the 3’ untranslated region, which serve as a source of dsRNA through inter- and intramolecular base pairing. Repeat elements such as long terminal repeats (LTRs) and non-LTR retrotransposons (SINEs and LINEs) are also commonly found in the intronic and intergenic regions. Under basal conditions, the activity of transposable elements in the intergenic regions is suppressed through multiple epigenetic mechanisms, including DNA methylation and DNA-binding molecules.33, 34 For example, heat-shock protein 90 forms a complex with the nuclear corepressor protein KRAB-associated protein 1, which binds to endogenous retrovirus loci and represses their expression. In contrast, under stress conditions, heat-shock protein 90 is relocated, leading to the reactivation of endogenous retroviruses.35 Finally, mitochondrially-derived transcripts, which increase during early sepsis, are also prone to forming dsRNA structures due to the bidirectional transcription of the mitochondrial genome. Under normal circumstances, such dsRNA is kept in check by the mitochondrial degradosome. However, if the degradation system is overwhelmed, mitochondrial dsRNA could translocate to the cytosol and contribute to dsRNA stress.36 Similarly, the activation of cytosolic DNA sensing pathway due to mitochondrial damage has been implicated in various disease conditions in the kidney.3747

These host-derived amplifying processes converge on the activation of the antiviral pathway, irrespective of the initiating microbes.48 The antiviral pathway is characterized by various interferon-mediated responses and culminate in the integrated stress response, marked by the phosphorylation of eIF2α (Eukaryotic Translation Initiation Factor 2 subunit α). For example, the dsRNA sensor PKR autoactivates upon binding to dsRNA. Once activated, PKR phosphorylates eIF2α. Phosphorylated eIF2α in turn stalls nearly all translation initiation events by inhibiting the GTP/GDP exchange reaction required for eIF2 recycling between successive rounds of translation initiation.49 As a potent translation-inhibitory signal, eIF2α phosphorylation limits pathogen replication, but leads to host global translation shutdown.50

Therefore, the antiviral phase in sepsis-associated AKI, which resulted from a massive upregulation of transcription and translation, is terminated by the activation of the integrated stress response, leading to translation shutdown. In an animal model of endotoxemia, this antiviral phase lasts between 8 to 16 hours, depending on the severity, cell type, and experimental model.51

Shutdown phase

Among the phases of sepsis-associated AKI, global translation shutdown is the most pronounced and consequential. This shutdown is preceded by a series of inflammatory and antiviral responses that occur in each cell type but are not fully synchronized (e.g., endothelial and stromal cells exhibit antiviral responses first, followed by renal epithelial cells).52 In contrast, translation shutdown occurs globally across all cell types at a distinct time point. This phase is characterized by the loss of cell-cell communication, dedifferentiation of cell identity, and downregulation of physiological functions, best exemplified in a reversible model of endotoxemia (Figure 2).

Despite the apparent organ paralysis, this shutdown phase is far from a dormant state. Rather, it represents a critical transition period where genome-wide reprogramming takes place to facilitate recovery.52 For example, SOX9, a key transcription factor involved in renal repair,5355 is induced during this period. Notably, a subset of genes are highly resistant to translation shutdown. Shutdown-resistant genes are enriched in pathways related to mRNA processing and RNA splicing.16 RNA splicing and the resulting isoforms enable rapid diversification of transcript repertoires, driving alterations in cellular phenotypes. Thus, RNA splicing may play a role in recovery programs, allowing cells to simultaneously downregulate stress-induced genes while upregulating genes essential for recovery.

Stress-induced metabolic shutdown is a critical inflection point. In the face of severe stress, transient inhibition of global metabolism is likely beneficial, as it reduces energy consumption and allows for resource reallocation. However, prolonged suppression of global protein synthesis is clearly detrimental.56 Indeed, both pharmacologic and genetic reversal of translation shutdown promotes renal recovery. For example, ISRIB, a small molecule that enhances GTP/GDP exchange reaction on the eIF2 complex,57 rescues translation and improves kidney function.16 Similarly, overexpression of the eIF2α phosphatase GADD34 (PPP1R15A) reverses translation shutdown and promotes recovery.58 These examples indicate that this shutdown phase is not fully adaptive but rather a component of AKI pathophysiology. The balance between excessive and appropriate shutdown is context-dependent, underscoring the need for strategies to quantitate and calibrate this critical phase.

Recovery

Finding ways to balance stress responses and elicit desired endogenous recovery mechanisms remains a major challenge. Experimentally, despite significant injury processes, the kidney can ultimately adopt a recovery phenotype, leading to the restoration of renal function and normalization of gene expression in most cell types (Figure 2).52 Similarly, even though sepsis-associated AKI is a serious clinical problem, a significant proportion of patients recover kidney function.59 This highlights the remarkable plasticity and resilience of biological systems.

Nature provides many examples of resilience that has been optimized by evolution. Resilience takes many forms and exists across biological scales, from whole organismal levels to specific molecular pathways. For example, metabolic flexibility is a form of resilience in which robust toggling between distinct metabolic routes (or quiescent vs. active states) enables rapid adaptation to environmental changes (e.g., arousal of hibernating bears and acute renal recovery).60, 61 In the context of infection, resilience can manifest as efficient microbial killing (disease resistance).62, 63 Alternatively, disease tolerance—mitigating self-inflicted cytokine damage while permitting microbial cohabitation—is another form of resilience (e.g., bats serving as viral reservoirs).64, 65 More generally, a wide range of work demonstrates the importance of regulated stress as a fitness strategy against otherwise lethal challenges (e.g., phenomenon of preconditioning).6669 Collectively, the natural world illuminates multiple paths to harnessing powerful protective pathways and serves as a blueprint for unlocking the potential of survival mechanisms.

To delineate the link between the initial inflammatory response and endogenous recovery mechanisms, here we highlight the role of polyamines in the kidney as a prime example. Polyamines, namely putrescine, spermidine and spermine, are involved in a variety of fundamental biological processes such as transcription, translation, differentiation, and DNA repair.7072 As a crucial metabolic pathway, altered polyamine metabolism has been reported as a unifying feature across more than 10 different kidney injury models in mice, as well as in the context of post-kidney transplantation in humans.7375

The rate-limiting step in polyamine biosynthesis is the conversion of the polyamine precursor ornithine to putrescine. Notably, Antizyme Inhibitor 1 (AZIN1), the master activator of this conversion process, can exist in two distinct protein isoforms.76 One is derived from a canonical AZIN1 transcript, and another is produced through post-transcriptional RNA editing of the canonical AZIN1, where adenosine is converted to inosine at a specific locus (A-to-I RNA editing). The edited AZIN1 exhibits a gain-of-function phenotype, thus promoting polyamine biosynthesis. This RNA editing has been shown to play a key role in enhancing stemness in cancer cells and promoting the differentiation of hematopoietic stem cells.7680

Under stress conditions in the kidney, AZIN1 A-to-I editing confers an advantage over the unedited state by upregulating the polyamine pathway and co-opting glycolysis and nicotinamide biosynthesis, culminating in a metabolically robust phenotype.32 Importantly, this AZIN1 A-to-I editing is mediated by RNA-specific Adenosine deaminase (ADAR), specifically in response to interferon signaling and dsRNA stress.81, 82 Therefore, the initial inflammatory stress not only causes translation shutdown but also primes the kidney to activate the subsequent endogenous recovery programs by boosting polyamine biosynthesis through A-to-I editing.

More broadly, these findings suggest a general model in which inflammation and resultant RNA editing function as an autonomous feedback system, protecting against sustained metabolic shutdown and signaling tissue recovery. Fundamentally, A-to-I editing represents a form of gene pool diversification under basal condition and in response to infections and various environmental factors. This mechanism is exploited by a variety of species to enhance survival fitness (e.g., temperature-dependent A-to-I editing in octopus).83

Excitingly, ADAR-based RNA-editing technology has advanced to clinical trials,84, 85 and the first positive results were announced recently (for alpha-1 antitrypsin deficiency).86 This modular technology, which harnesses endogenous ADAR, could be a promising strategy for treating septic AKI at specific timepoints. Additionally, A-to-I RNA editing at various gene loci has been linked to genetic traits of common inflammatory diseases.87 Therefore, identifying editable sites across the genome and understanding when and in what context editing occurs in the kidney could pave the way for novel RNA-editing therapeutic strategies.

Host-microbial interplay in the kidney

“It is not the strongest or the most intelligent species that survives, but the one most adaptable to change”

—Leon Megginson/Charles Darwin

From the species level down to individual cells, the ability to adapt to diverse stresses is essential for survival and fitness. For example, host cells dynamically adjust their bioenergetics in response to their environment by modulating the balance between glycolysis and oxidative phosphorylation. Neither glycolysis nor oxidative phosphorylation is inherently superior; the optimal pathway depends on the cellular context. Similarly, invading microbes exhibit metabolic flexibility as they adapt to host ecological niches through genetic drift, adaptive evolution, and transcriptional reprogramming.88, 89 In fact, lethal bloodstream infection and tissue destruction are evolutionary dead ends for the invading microbe. Therefore, beyond modulating virulence and immune evasion factors, microbes initiate a complex crosstalk with the host that is constantly at play.90, 91 This poses a formidable challenge in resolving the evolutionary conflict between species over their environmental responses and control.

In this final section, we discuss Staphylococcus aureus infection in the kidney, a common cause of AKI,9297 particularly Methicillin-resistant S. aureus (MRSA), to highlight the complexity of sepsis syndrome and unresolved questions.

Both as a commensal and as a pathogen, MRSA has been highly successful in prevailing in multiple hosts since it was first identified in the 1960s. MRSA can traverse diverse host environments and persist in tissue niches by employing a variety of metabolic adaptation strategies and subverting the host immune system.88 On the host side, neutrophils are the main defense against MRSA infections.89 Neutrophils launch various bactericidal arsenals such as degranulation and neutrophil extracellular traps. In turn, MRSA immediately counters by mounting multiple neutralizing molecules. On balance, the molecular arms race between the host and MRSA is currently very close, and the identification of a novel therapeutic strategy is urgently needed.

In murine models of MRSA infection, systemically administered MRSA localizes primarily to the liver during the early stages of infection. The focus of infection then shifts to the kidney later in the course, resulting in renal tissue damage and high mortality.98100 Even before the emergence of MRSA, the kidney was known as a major destination for bloodstream S. aureus infections in both humans and animals.101103 Human autopsies and animal experiments conducted before the era of molecular biology have clearly defined the aggressive nature of “descending” S. aureus infection in the kidney (i.e., hematologic dissemination).104 However, the molecular mechanisms leading to the spread of S. aureus in the kidney are unknown.

The difference between MRSA and MSSA (Methicillin-susceptible S. aureus) is not solely based on antibiotic resistance. The largest genomic region that sets MRSA apart from MSSA is the presence of the arginine catabolic mobile element (ACME) in MRSA—specifically, the ACME that contains spermidine acetyltransferase in the USA300 strain (Figure 3).88, 105, 106 USA300 is the dominant MRSA strain, accounting for almost all community-associated MRSA cases presenting to emergency rooms in the United States,107, 108 as well as healthcare settings.109 The ACME encodes arginine deiminase enzymes (arc Operon arcA/arcB/arcC) and spermidine acetyltransferase. The acquisition of ACME by USA300 MRSA coincided with its global spread,106, 107 suggesting that arginine catabolism (the conversion of arginine to ornithine) confers a significant survival advantage. The byproducts of arginine catabolism in S. aureus include ATP and ammonium, which provide energy and contribute to acid resistance.105 It is possible that the unique kidney microenvironment, such as the inner medulla, could serve as an MRSA reservoir due to its ability to resist acid, hypoxia, and high osmolality.

Figure 3. Staphylococcus aureus infection.

Figure 3.

Left: Genome structure of MRSA USA300. The region corresponding to the arginine catabolic mobile element (ACME) is magnified (blue). SCCmec: Staphylococcal Cassette Chromosome mec, which encodes methicillin resistance. speG: spermidine acetyltransferase. Middle: Representative clinical challenges encountered by nephrologists. Right: Illustration of hematogenous dissemination of S. aureus resulting in acute kidney injury (descending pyelonephritis).

Notably, S. aureus (both MRSA and MSSA) does not synthesize its own polyamines and instead relies solely on external sources. This is highly unusual among living organisms.105, 106, 110112 Polyamine biosynthesis was once believed to be conserved across the tree of life (still the case for nearly all archaea and eukaryotes, and the vast majority of bacteria).113, 114 USA300 MRSA encodes only a single polyamine catabolic gene, spermidine acetyltransferase, whereas MSSA lacks this gene and any other genes related to polyamine metabolism.110 Spermidine acetyltransferase neutralizes the amine groups on spermidine and spermine, thereby facilitating their export to the extracellular space. This results in differential use of polyamines between MRSA and MSSA. The implications of these characteristics and resulting susceptibility to polyamines, or the advantages polyamines offer to S. aureus in the kidney, remain largely unexplored. In response to S. aureus infection, host neutrophils and macrophages upregulate arginine catabolism via arginase 2 and arginase 1, respectively (unpublished observation). These innate immune cells also upregulate Spermidine/spermine N1-acetyltransferase 1, an enzyme functionally analogous to Spermidine acetyltransferase in MRSA. This suggests a parallel—competitive or exploitative—dynamic between host and pathogen along the axis of arginine and polyamine catabolism, underscoring the complex ecology of host-microbial interactions.

Beyond direct kidney infection, S. aureus remains a vexing problem across the spectrum of clinical practice, ranging from endocarditis and osteomyelitis to dialysis access complications.115, 116 In addition, the pathobiology of S. aureus-associated glomerulonephritis117, 118 and vancomycin nephrotoxicity119121 remains poorly understood, as these conditions are confounded by concomitant bacteremia. Further investigation into the host-microbe interplay will enhance our understanding of disease unfolding and the unique renal microenvironment impacting AKI outcomes in sepsis.

Outlook

“Now, here, you see, it takes all the running you can do, to keep in the same place. If you want to get somewhere else, you must run at least twice as fast as that”

—Lewis Carroll/Leigh Van Valen (The Red Queen hypothesis)

To survive sepsis and maintain vital physiological functions, the kidney must dynamically adapt and evolve throughout the course of infection. The constant battle between hosts and microbes renders any therapeutic option irrelevant or even detrimental if the timeline of sepsis is not carefully considered. While we have dissected several key molecular pathways involved in the progression and transition of septic AKI, many other equally important and active pathways remain beyond the scope of this Review.122158 In particular, we acknowledge that this Review places less emphasis on cell-specific effects, as we have prioritized defining the overall tissue phenotype and timeline. Although the intense competition between hosts and microbes is a major driver of sepsis, it is likely more than a simple zero-sum game. Complex interactions within and between species may even generate new resources and foster mutualism. A remarkable array of technologically advanced research tools is now available at scale. More than ever, we must re-examine the time-dependent molecular crosstalk between host and pathogens to identify novel therapeutic approaches for improving the outcome of septic AKI.

Acknowledgements

This work was supported by NIH grants R01-AI148282 and Veterans Affairs Merit (BX002901) to TH, R01-DK107623 and U54DK137328 to PCD

Support:

NIH R01-AI148282, Veterans Affairs Merit BX002901, NIH R01-DK107623, U01DK114923

Footnotes

Conflict of interest: The authors have declared that no conflict of interest exists.

References

  • 1.Antimicrobial Resistance, C: Global burden of bacterial antimicrobial resistance in 2019: a systematic analysis. Lancet, 399: 629–655, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Collaborators, GBDAR: Global burden of bacterial antimicrobial resistance 1990–2021: a systematic analysis with forecasts to 2050. Lancet, 404: 1199–1226, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Stewart BJ, Ferdinand JR, Young MD, Mitchell TJ, Loudon KW, Riding AM, Richoz N, Frazer GL, Staniforth JUL, Vieira Braga FA, Botting RA, Popescu DM, Vento-Tormo R, Stephenson E, Cagan A, Farndon SJ, Polanski K, Efremova M, Green K, Del Castillo Velasco-Herrera M, Guzzo C, Collord G, Mamanova L, Aho T, Armitage JN, Riddick ACP, Mushtaq I, Farrell S, Rampling D, Nicholson J, Filby A, Burge J, Lisgo S, Lindsay S, Bajenoff M, Warren AY, Stewart GD, Sebire N, Coleman N, Haniffa M, Teichmann SA, Behjati S, Clatworthy MR: Spatiotemporal immune zonation of the human kidney. Science, 365: 1461–1466, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Zarbock A, Nadim MK, Pickkers P, Gomez H, Bell S, Joannidis M, Kashani K, Koyner JL, Pannu N, Meersch M, Reis T, Rimmele T, Bagshaw SM, Bellomo R, Cantaluppi V, Deep A, De Rosa S, Perez-Fernandez X, Husain-Syed F, Kane-Gill SL, Kelly Y, Mehta RL, Murray PT, Ostermann M, Prowle J, Ricci Z, See EJ, Schneider A, Soranno DE, Tolwani A, Villa G, Ronco C, Forni LG: Sepsis-associated acute kidney injury: consensus report of the 28th Acute Disease Quality Initiative workgroup. Nat Rev Nephrol, 19: 401–417, 2023. [DOI] [PubMed] [Google Scholar]
  • 5.Hato T, Dagher PC: How the Innate Immune System Senses Trouble and Causes Trouble. Clin J Am Soc Nephrol, 10: 1459–1469, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Bagshaw SM, George C, Bellomo R, Committee, ADM: Early acute kidney injury and sepsis: a multicentre evaluation. Crit Care, 12: R47, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Peerapornratana S, Manrique-Caballero CL, Gomez H, Kellum JA: Acute kidney injury from sepsis: current concepts, epidemiology, pathophysiology, prevention and treatment. Kidney Int, 96: 1083–1099, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Zarjou A, Agarwal A: Sepsis and acute kidney injury. J Am Soc Nephrol, 22: 999–1006, 2011. [DOI] [PubMed] [Google Scholar]
  • 9.Kuwabara S, Goggins E, Okusa MD: The Pathophysiology of Sepsis-Associated AKI. Clin J Am Soc Nephrol, 17: 1050–1069, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Legrand M, Bagshaw SM, Bhatraju PK, Bihorac A, Caniglia E, Khanna AK, Kellum JA, Koyner J, Harhay MO, Zampieri FG, Zarbock A, Chung K, Liu K, Mehta R, Pickkers P, Ryan A, Bernholz J, Dember L, Gallagher M, Rossignol P, Ostermann M: Sepsis-associated acute kidney injury: recent advances in enrichment strategies, sub-phenotyping and clinical trials. Crit Care, 28: 92, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Cavaillon JM, Singer M, Skirecki T: Sepsis therapies: learning from 30 years of failure of translational research to propose new leads. EMBO Mol Med, 12: e10128, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Deutschman CS, Tracey KJ: Sepsis: current dogma and new perspectives. Immunity, 40: 463–475, 2014. [DOI] [PubMed] [Google Scholar]
  • 13.Levy M, Thaiss CA, Zeevi D, Dohnalova L, Zilberman-Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, Pevsner-Fischer M, Shapiro H, Christ A, Harmelin A, Halpern Z, Latz E, Flavell RA, Amit I, Segal E, Elinav E: Microbiota-Modulated Metabolites Shape the Intestinal Microenvironment by Regulating NLRP6 Inflammasome Signaling. Cell, 163: 1428–1443, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Troha K, Ayres JS: Metabolic Adaptations to Infections at the Organismal Level. Trends Immunol, 41: 113–125, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Poston JT, Koyner JL: Sepsis associated acute kidney injury. BMJ, 364: k4891, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Hato T, Maier B, Syed F, Myslinski J, Zollman A, Plotkin Z, Eadon MT, Dagher PC: Bacterial sepsis triggers an antiviral response that causes translation shutdown. J Clin Invest, 129: 296–309, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Rice TW, Wheeler AP, Bernard GR, Vincent JL, Angus DC, Aikawa N, Demeyer I, Sainati S, Amlot N, Cao C, Ii M, Matsuda H, Mouri K, Cohen J: A randomized, double-blind, placebo-controlled trial of TAK-242 for the treatment of severe sepsis. Crit Care Med, 38: 1685–1694, 2010. [DOI] [PubMed] [Google Scholar]
  • 18.Tidswell M, Tillis W, Larosa SP, Lynn M, Wittek AE, Kao R, Wheeler J, Gogate J, Opal SM, Eritoran Sepsis Study, G: Phase 2 trial of eritoran tetrasodium (E5564), a toll-like receptor 4 antagonist, in patients with severe sepsis. Crit Care Med, 38: 72–83, 2010. [DOI] [PubMed] [Google Scholar]
  • 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, Group, AS: Effect of eritoran, an antagonist of MD2-TLR4, on mortality in patients with severe sepsis: the ACCESS randomized trial. JAMA, 309: 1154–1162, 2013. [DOI] [PubMed] [Google Scholar]
  • 20.Marshall JC: Sepsis: rethinking the approach to clinical research. J Leukoc Biol, 83: 471–482, 2008. [DOI] [PubMed] [Google Scholar]
  • 21.Dellinger RP, Bagshaw SM, Antonelli M, Foster DM, Klein DJ, Marshall JC, Palevsky PM, Weisberg LS, Schorr CA, Trzeciak S, Walker PM, Investigators, ET: Effect of Targeted Polymyxin B Hemoperfusion on 28-Day Mortality in Patients With Septic Shock and Elevated Endotoxin Level: The EUPHRATES Randomized Clinical Trial. JAMA, 320: 1455–1463, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Zimmer JT, Rosa-Mercado NA, Canzio D, Steitz JA, Simon MD: STL-seq reveals pause-release and termination kinetics for promoter-proximal paused RNA polymerase II transcripts. Mol Cell, 81: 4398–4412 e4397, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Liu X, Kraus WL, Bai X: Ready, pause, go: regulation of RNA polymerase II pausing and release by cellular signaling pathways. Trends Biochem Sci, 40: 516–525, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Hotchkiss RS, Monneret G, Payen D: Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol, 13: 862–874, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Xiao W, Mindrinos MN, Seok J, Cuschieri J, Cuenca AG, Gao H, Hayden DL, Hennessy L, Moore EE, Minei JP, Bankey PE, Johnson JL, Sperry J, Nathens AB, Billiar TR, West MA, Brownstein BH, Mason PH, Baker HV, Finnerty CC, Jeschke MG, Lopez MC, Klein MB, Gamelli RL, Gibran NS, Arnoldo B, Xu W, Zhang Y, Calvano SE, McDonald-Smith GP, Schoenfeld DA, Storey JD, Cobb JP, Warren HS, Moldawer LL, Herndon DN, Lowry SF, Maier RV, Davis RW, Tompkins RG, Inflammation, Host Response to Injury Large-Scale Collaborative Research, P: A genomic storm in critically injured humans. J Exp Med, 208: 2581–2590, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tunnacliffe E, Chubb JR: What Is a Transcriptional Burst? Trends Genet, 36: 288–297, 2020. [DOI] [PubMed] [Google Scholar]
  • 27.Carpenter S, Ricci EP, Mercier BC, Moore MJ, Fitzgerald KA: Post-transcriptional regulation of gene expression in innate immunity. Nat Rev Immunol, 14: 361–376, 2014. [DOI] [PubMed] [Google Scholar]
  • 28.McNab F, Mayer-Barber K, Sher A, Wack A, O’Garra A: Type I interferons in infectious disease. Nat Rev Immunol, 15: 87–103, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Gebhardt A, Laudenbach BT, Pichlmair A: Discrimination of Self and Non-Self Ribonucleic Acids. J Interferon Cytokine Res, 37: 184–197, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Dalskov L, Gad HH, Hartmann R: Viral recognition and the antiviral interferon response. EMBO J, 42: e112907, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Hur S: Double-Stranded RNA Sensors and Modulators in Innate Immunity. Annu Rev Immunol, 37: 349–375, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Heruye SH, Myslinski J, Zeng C, Zollman A, Makino S, Nanamatsu A, Mir Q, Janga SC, Doud EH, Eadon MT, Maier B, Hamada M, Tran TM, Dagher PC, Hato T: Inflammation primes the murine kidney for recovery by activating AZIN1 adenosine-to-inosine editing. J Clin Invest, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stoye JP: Studies of endogenous retroviruses reveal a continuing evolutionary saga. Nat Rev Microbiol, 10: 395–406, 2012. [DOI] [PubMed] [Google Scholar]
  • 34.Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, Makarov V, Budhu S, Slamon DJ, Wolchok JD, Pardoll DM, Beckmann MW, Zahnow CA, Merghoub T, Chan TA, Baylin SB, Strick R: Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell, 169: 361, 2017. [DOI] [PubMed] [Google Scholar]
  • 35.Hummel B, Hansen EC, Yoveva A, Aprile-Garcia F, Hussong R, Sawarkar R: The evolutionary capacitor HSP90 buffers the regulatory effects of mammalian endogenous retroviruses. Nat Struct Mol Biol, 24: 234–242, 2017. [DOI] [PubMed] [Google Scholar]
  • 36.Dhir A, Dhir S, Borowski LS, Jimenez L, Teitell M, Rotig A, Crow YJ, Rice GI, Duffy D, Tamby C, Nojima T, Munnich A, Schiff M, de Almeida CR, Rehwinkel J, Dziembowski A, Szczesny RJ, Proudfoot NJ: Mitochondrial double-stranded RNA triggers antiviral signalling in humans. Nature, 560: 238–242, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Chung KW, Dhillon P, Huang S, Sheng X, Shrestha R, Qiu C, Kaufman BA, Park J, Pei L, Baur J, Palmer M, Susztak K: Mitochondrial Damage and Activation of the STING Pathway Lead to Renal Inflammation and Fibrosis. Cell Metab, 30: 784–799 e785, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Khedr S, Dissanayake LV, Alsheikh AJ, Zietara A, Spires DR, Kerketta R, Mathison AJ, Urrutia R, Palygin O, Staruschenko A: Role of cGAS/STING pathway in aging and sexual dimorphism in diabetic kidney disease. JCI Insight, 10, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Liang X, Liu H, Hu H, Ha E, Zhou J, Abedini A, Sanchez-Navarro A, Klotzer KA, Susztak K: TET2 germline variants promote kidney disease by impairing DNA repair and activating cytosolic nucleotide sensors. Nat Commun, 15: 9621, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.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 e1266, 2019. [DOI] [PubMed] [Google Scholar]
  • 41.Tsai YC, Hsieh TH, Liao YR, Tsai MT, Lin TP, Lee DY, Park J, Kim D, Susztak K, Yang SF, Lin CC, Li SY: METTL3-Mediated N 6 -Methyladenosine mRNA Modification and cGAS-STING Pathway Activity in Kidney Fibrosis. J Am Soc Nephrol, 35: 1312–1329, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Zang N, Cui C, Guo X, Song J, Hu H, Yang M, Xu M, Wang L, Hou X, He Q, Sun Z, Wang C, Chen L: cGAS-STING activation contributes to podocyte injury in diabetic kidney disease. iScience, 25: 105145, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Tsuji N, Tsuji T, Yamashita T, Hayase N, Hu X, Yuen PS, Star RA: BAM15 treats mouse sepsis and kidney injury, linking mortality, mitochondrial DNA, tubule damage, and neutrophils. J Clin Invest, 133, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.van der Slikke EC, Star BS, van Meurs M, Henning RH, Moser J, Bouma HR: Sepsis is associated with mitochondrial DNA damage and a reduced mitochondrial mass in the kidney of patients with sepsis-AKI. Crit Care, 25: 36, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhang X, Agborbesong E, Li X: The Role of Mitochondria in Acute Kidney Injury and Chronic Kidney Disease and Its Therapeutic Potential. Int J Mol Sci, 22, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Parikh SM, Yang Y, He L, Tang C, Zhan M, Dong Z: Mitochondrial function and disturbances in the septic kidney. Semin Nephrol, 35: 108–119, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Chevrier N, Mertins P, Artyomov MN, Shalek AK, Iannacone M, Ciaccio MF, Gat-Viks I, Tonti E, DeGrace MM, Clauser KR, Garber M, Eisenhaure TM, Yosef N, Robinson J, Sutton A, Andersen MS, Root DE, von Andrian U, Jones RB, Park H, Carr SA, Regev A, Amit I, Hacohen N: Systematic discovery of TLR signaling components delineates viral-sensing circuits. Cell, 147: 853–867, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Costa-Mattioli M, Walter P: The integrated stress response: From mechanism to disease. Science, 368, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li MM, MacDonald MR, Rice CM: To translate, or not to translate: viral and host mRNA regulation by interferon-stimulated genes. Trends Cell Biol, 25: 320–329, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Doi K, Leelahavanichkul A, Yuen PS, Star RA: Animal models of sepsis and sepsis-induced kidney injury. J Clin Invest, 119: 2868–2878, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Janosevic D, Myslinski J, McCarthy TW, Zollman A, Syed F, Xuei X, Gao H, Liu YL, Collins KS, Cheng YH, Winfree S, El-Achkar TM, Maier B, Melo Ferreira R, Eadon MT, Hato T, Dagher PC: The orchestrated cellular and molecular responses of the kidney to endotoxin define a precise sepsis timeline. Elife, 10, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Aggarwal S, Wang Z, Rincon Fernandez Pacheco D, Rinaldi A, Rajewski A, Callemeyn J, Van Loon E, Lamarthee B, Covarrubias AE, Hou J, Yamashita M, Akiyama H, Karumanchi SA, Svendsen CN, Noble PW, Jordan SC, Breunig JJ, Naesens M, Cippa PE, Kumar S: SOX9 switch links regeneration to fibrosis at the single-cell level in mammalian kidneys. Science, 383: eadd6371, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Kumar S, Liu J, Pang P, Krautzberger AM, Reginensi A, Akiyama H, Schedl A, Humphreys BD, McMahon AP: Sox9 Activation Highlights a Cellular Pathway of Renal Repair in the Acutely Injured Mammalian Kidney. Cell Rep, 12: 1325–1338, 2015. [DOI] [PubMed] [Google Scholar]
  • 55.Kang HM, Huang S, Reidy K, Han SH, Chinga F, Susztak K: Sox9-Positive Progenitor Cells Play a Key Role in Renal Tubule Epithelial Regeneration in Mice. Cell Rep, 14: 861–871, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Harding HP, Zhang Y, Scheuner D, Chen JJ, Kaufman RJ, Ron D: Ppp1r15 gene knockout reveals an essential role for translation initiation factor 2 alpha (eIF2alpha) dephosphorylation in mammalian development. Proc Natl Acad Sci U S A, 106: 1832–1837, 2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Sidrauski C, McGeachy AM, Ingolia NT, Walter P: The small molecule ISRIB reverses the effects of eIF2alpha phosphorylation on translation and stress granule assembly. Elife, 4, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Kidwell A, Yadav SPS, Maier B, Zollman A, Ni K, Halim A, Janosevic D, Myslinski J, Syed F, Zeng L, Waffo AB, Banno K, Xuei X, Doud EH, Dagher PC, Hato T: Translation Rescue by Targeting Ppp1r15a through Its Upstream Open Reading Frame in Sepsis-Induced Acute Kidney Injury in a Murine Model. J Am Soc Nephrol, 34: 220–240, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Manrique-Caballero CL, Del Rio-Pertuz G, Gomez H: Sepsis-Associated Acute Kidney Injury. Crit Care Clin, 37: 279–301, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Stenvinkel P, Jani AH, Johnson RJ: Hibernating bears (Ursidae): metabolic magicians of definite interest for the nephrologist. Kidney Int, 83: 207–212, 2013. [DOI] [PubMed] [Google Scholar]
  • 61.Srivastava A, Kumar Sarsani V, Fiddes I, Sheehan SM, Seger RL, Barter ME, Neptune-Bear S, Lindqvist C, Korstanje R: Genome assembly and gene expression in the American black bear provides new insights into the renal response to hibernation. DNA Res, 26: 37–44, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Schneider DS, Ayres JS: Two ways to survive infection: what resistance and tolerance can teach us about treating infectious diseases. Nat Rev Immunol, 8: 889–895, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Medzhitov R, Schneider DS, Soares MP: Disease tolerance as a defense strategy. Science, 335: 936–941, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Irving AT, Ahn M, Goh G, Anderson DE, Wang LF: Lessons from the host defences of bats, a unique viral reservoir. Nature, 589: 363–370, 2021. [DOI] [PubMed] [Google Scholar]
  • 65.Colaco HG, Barros A, Neves-Costa A, Seixas E, Pedroso D, Velho T, Willmann KL, Faisca P, Grabmann G, Yi HS, Shong M, Benes V, Weis S, Kocher T, Moita LF: Tetracycline Antibiotics Induce Host-Dependent Disease Tolerance to Infection. Immunity, 54: 53–67 e57, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Hato T, Zollman A, Plotkin Z, El-Achkar TM, Maier BF, Pay SL, Dube S, Cabral P, Yoshimoto M, McClintick J, Dagher PC: Endotoxin Preconditioning Reprograms S1 Tubules and Macrophages to Protect the Kidney. J Am Soc Nephrol, 29: 104–117, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hato T, Winfree S, Kalakeche R, Dube S, Kumar R, Yoshimoto M, Plotkin Z, Dagher PC: The macrophage mediates the renoprotective effects of endotoxin preconditioning. J Am Soc Nephrol, 26: 1347–1362, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Tomai F, Crea F, Chiariello L, Gioffre PA: Ischemic preconditioning in humans: models, mediators, and clinical relevance. Circulation, 100: 559–563, 1999. [DOI] [PubMed] [Google Scholar]
  • 69.Biswas SK, Lopez-Collazo E: Endotoxin tolerance: new mechanisms, molecules and clinical significance. Trends Immunol, 30: 475–487, 2009. [DOI] [PubMed] [Google Scholar]
  • 70.Miller-Fleming L, Olin-Sandoval V, Campbell K, Ralser M: Remaining Mysteries of Molecular Biology: The Role of Polyamines in the Cell. J Mol Biol, 427: 3389–3406, 2015. [DOI] [PubMed] [Google Scholar]
  • 71.Madeo F, Eisenberg T, Pietrocola F, Kroemer G: Spermidine in health and disease. Science, 359, 2018. [DOI] [PubMed] [Google Scholar]
  • 72.Pegg AE: Functions of Polyamines in Mammals. J Biol Chem, 291: 14904–14912, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sieckmann T, Schley G, Ogel N, Kelterborn S, Boivin FJ, Fahling M, Ashraf MI, Reichel M, Vigolo E, Hartner A, Lichtenberger FB, Breiderhoff T, Knauf F, Rosenberger C, Aigner F, Schmidt-Ott K, Scholz H, Kirschner KM: Strikingly conserved gene expression changes of polyamine regulating enzymes among various forms of acute and chronic kidney injury. Kidney Int, 104: 90–107, 2023. [DOI] [PubMed] [Google Scholar]
  • 74.Evans RG: Maybe the various forms of kidney disease are not so mechanistically different? Kidney Int, 104: 31–33, 2023. [DOI] [PubMed] [Google Scholar]
  • 75.Zahedi K, Barone S, Soleimani M: Polyamines and Their Metabolism: From the Maintenance of Physiological Homeostasis to the Mediation of Disease. Med Sci (Basel), 10, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Chen L, Li Y, Lin CH, Chan TH, Chow RK, Song Y, Liu M, Yuan YF, Fu L, Kong KL, Qi L, Li Y, Zhang N, Tong AH, Kwong DL, Man K, Lo CM, Lok S, Tenen DG, Guan XY: Recoding RNA editing of AZIN1 predisposes to hepatocellular carcinoma. Nat Med, 19: 209–216, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Shigeyasu K, Okugawa Y, Toden S, Miyoshi J, Toiyama Y, Nagasaka T, Takahashi N, Kusunoki M, Takayama T, Yamada Y, Fujiwara T, Chen L, Goel A: AZIN1 RNA editing confers cancer stemness and enhances oncogenic potential in colorectal cancer. JCI Insight, 3, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ghalali A, Wang L, Stopsack KH, Rice JM, Wu S, Wu CL, Zetter BR, Rogers MS: AZIN1 RNA editing alters protein interactions, leading to nuclear translocation and worse outcomes in prostate cancer. Exp Mol Med, 54: 1713–1726, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Qin YR, Qiao JJ, Chan TH, Zhu YH, Li FF, Liu H, Fei J, Li Y, Guan XY, Chen L: Adenosine-to-inosine RNA editing mediated by ADARs in esophageal squamous cell carcinoma. Cancer Res, 74: 840–851, 2014. [DOI] [PubMed] [Google Scholar]
  • 80.Wang F, He J, Liu S, Gao A, Yang L, Sun G, Ding W, Li CY, Gou F, He M, Wang F, Wang X, Zhao X, Zhu P, Hao S, Ma Y, Cheng H, Yu J, Cheng T: A comprehensive RNA editome reveals that edited Azin1 partners with DDX1 to enable hematopoietic stem cell differentiation. Blood, 138: 1939–1952, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Sun T, Yu Y, Wu X, Acevedo A, Luo JD, Wang J, Schneider WM, Hurwitz B, Rosenberg BR, Chung H, Rice CM: Decoupling expression and editing preferences of ADAR1 p150 and p110 isoforms. Proc Natl Acad Sci U S A, 118, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Xing Y, Nakahama T, Wu Y, Inoue M, Kim JI, Todo H, Shibuya T, Kato Y, Kawahara Y: RNA editing of AZIN1 coding sites is catalyzed by ADAR1 p150 after splicing. J Biol Chem, 299: 104840, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Birk MA, Liscovitch-Brauer N, Dominguez MJ, McNeme S, Yue Y, Hoff JD, Twersky I, Verhey KJ, Sutton RB, Eisenberg E, Rosenthal JJC: Temperature-dependent RNA editing in octopus extensively recodes the neural proteome. Cell, 186: 2544–2555 e2513, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Lenharo M: Move over, CRISPR: RNA-editing therapies pick up steam. Nature, 626: 933–934, 2024. [DOI] [PubMed] [Google Scholar]
  • 85.Booth BJ, Nourreddine S, Katrekar D, Savva Y, Bose D, Long TJ, Huss DJ, Mali P: RNA editing: Expanding the potential of RNA therapeutics. Mol Ther, 31: 1533–1549, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.First proof-of-mechanism for RNA editing in humans. Nat Biotechnol, 42: 1627, 2024. [DOI] [PubMed] [Google Scholar]
  • 87.Li Q, Gloudemans MJ, Geisinger JM, Fan B, Aguet F, Sun T, Ramaswami G, Li YI, Ma JB, Pritchard JK, Montgomery SB, Li JB: RNA editing underlies genetic risk of common inflammatory diseases. Nature, 608: 569–577, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Turner NA, Sharma-Kuinkel BK, Maskarinec SA, Eichenberger EM, Shah PP, Carugati M, Holland TL, Fowler VG Jr.: Methicillin-resistant Staphylococcus aureus: an overview of basic and clinical research. Nat Rev Microbiol, 17: 203–218, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Howden BP, Giulieri SG, Wong Fok Lung T, Baines SL, Sharkey LK, Lee JYH, Hachani A, Monk IR, Stinear TP: Staphylococcus aureus host interactions and adaptation. Nat Rev Microbiol, 21: 380–395, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sonnert ND, Rosen CE, Ghazi AR, Franzosa EA, Duncan-Lowey B, Gonzalez-Hernandez JA, Huck JD, Yang Y, Dai Y, Rice TA, Nguyen MT, Song D, Cao Y, Martin AL, Bielecka AA, Fischer S, Guan C, Oh J, Huttenhower C, Ring AM, Palm NW: A host-microbiota interactome reveals extensive transkingdom connectivity. Nature, 628: 171–179, 2024. [DOI] [PubMed] [Google Scholar]
  • 91.Wong Fok Lung T, Charytonowicz D, Beaumont KG, Shah SS, Sridhar SH, Gorrie CL, Mu A, Hofstaedter CE, Varisco D, McConville TH, Drikic M, Fowler B, Urso A, Shi W, Fucich D, Annavajhala MK, Khan IN, Oussenko I, Francoeur N, Smith ML, Stockwell BR, Lewis IA, Hachani A, Upadhyay Baskota S, Uhlemann AC, Ahn D, Ernst RK, Howden BP, Sebra R, Prince A: Klebsiella pneumoniae induces host metabolic stress that promotes tolerance to pulmonary infection. Cell Metab, 34: 761–774 e769, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Westgeest AC, Schippers EF, Delfos NM, Visser LG, de Fijter JW, de Boer MGJ, Lambregts MMC: Acute kidney injury in Staphylococcus aureus bacteremia. Eur J Clin Microbiol Infect Dis, 41: 431–437, 2022. [DOI] [PubMed] [Google Scholar]
  • 93.Buis DTP, van der Vaart TW, Mohan A, Prins JM, van der Meer JTM, Bonten MJM, Jakulj L, van Werkhoven CH, Sigaloff KCE: Acute kidney injury in Staphylococcus aureus bacteremia: a recurrent events analysis. Clin Microbiol Infect, 2024. [DOI] [PubMed] [Google Scholar]
  • 94.Collaborators, GBDAR: Global mortality associated with 33 bacterial pathogens in 2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet, 400: 2221–2248, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Schuler F, Barth PJ, Niemann S, Schaumburg F: A Narrative Review on the Role of Staphylococcus aureus Bacteriuria in S. aureus Bacteremia. Open Forum Infect Dis, 8: ofab158, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Li T, Liu JY, Liu JF, Duan M, Li A: The Correlation Between the Types of Initial Bacterial Infection and Clinical Prognosis in Patients With Septic AKI. Front Med (Lausanne), 8: 800532, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Pant P, Chihara S, Krishnamoorthy V, Treggiari MM, Messina JA, Privratsky JR, Raghunathan K, Ohnuma T: Association of Causative Pathogens With Acute Kidney Injury in Adult Patients With Community-Onset Sepsis. Crit Care Explor, 7: e1219, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Surewaard BG, Deniset JF, Zemp FJ, Amrein M, Otto M, Conly J, Omri A, Yates RM, Kubes P: Identification and treatment of the Staphylococcus aureus reservoir in vivo. J Exp Med, 213: 1141–1151, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Pollitt EJG, Szkuta PT, Burns N, Foster SJ: Staphylococcus aureus infection dynamics. PLoS Pathog, 14: e1007112, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Groma M, Horst SA, Das S, Huettel B, Klepsch M, Rudel T, Medina E, Fraunholz M: Identification of a Novel LysR-Type Transcriptional Regulator in Staphylococcus aureus That Is Crucial for Secondary Tissue Colonization during Metastatic Bloodstream Infection. mBio, 11, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nesbit R: Acute staphylococcal infections of the kidney. JAMA, 98: 709–714, 1932. [Google Scholar]
  • 102.OM WJS: Pathology of so-called acute pyelitis in infants. American Journal of Diseases of Children, 38: 227–240, 1929. [Google Scholar]
  • 103.Gorrill RH: The establishment of staphylococcal abscesses in the mouse kidney. Br J Exp Pathol, 39: 203–212, 1958. [PMC free article] [PubMed] [Google Scholar]
  • 104.De NS: Experimental pyelonephritis in the rabbit produced by staphylococcal infection. J Pathol Bacteriol, 62: 429–436, 1950. [DOI] [PubMed] [Google Scholar]
  • 105.Thurlow LR, Joshi GS, Clark JR, Spontak JS, Neely CJ, Maile R, Richardson AR: Functional modularity of the arginine catabolic mobile element contributes to the success of USA300 methicillin-resistant Staphylococcus aureus. Cell Host Microbe, 13: 100–107, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Planet PJ, LaRussa SJ, Dana A, Smith H, Xu A, Ryan C, Uhlemann AC, Boundy S, Goldberg J, Narechania A, Kulkarni R, Ratner AJ, Geoghegan JA, Kolokotronis SO, Prince A: Emergence of the epidemic methicillin-resistant Staphylococcus aureus strain USA300 coincides with horizontal transfer of the arginine catabolic mobile element and speG-mediated adaptations for survival on skin. mBio, 4: e00889–00813, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Glaser P, Martins-Simoes P, Villain A, Barbier M, Tristan A, Bouchier C, Ma L, Bes M, Laurent F, Guillemot D, Wirth T, Vandenesch F: Demography and Intercontinental Spread of the USA300 Community-Acquired Methicillin-Resistant Staphylococcus aureus Lineage. mBio, 7: e02183–02115, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Lee AS, de Lencastre H, Garau J, Kluytmans J, Malhotra-Kumar S, Peschel A, Harbarth S: Methicillin-resistant Staphylococcus aureus. Nat Rev Dis Primers, 4: 18033, 2018. [DOI] [PubMed] [Google Scholar]
  • 109.Seybold U, Kourbatova EV, Johnson JG, Halvosa SJ, Wang YF, King MD, Ray SM, Blumberg HM: Emergence of community-associated methicillin-resistant Staphylococcus aureus USA300 genotype as a major cause of health care-associated blood stream infections. Clin Infect Dis, 42: 647–656, 2006. [DOI] [PubMed] [Google Scholar]
  • 110.Joshi GS, Spontak JS, Klapper DG, Richardson AR: Arginine catabolic mobile element encoded speG abrogates the unique hypersensitivity of Staphylococcus aureus to exogenous polyamines. Mol Microbiol, 82: 9–20, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Campbell K, Kowalski CH, Kohler KM, Barber MF: Evolution of polyamine resistance in Staphylococcus aureus through modulation of potassium transport. bioRxiv, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Krysenko S, Wohlleben W: Polyamine and Ethanolamine Metabolism in Bacteria as an Important Component of Nitrogen Assimilation for Survival and Pathogenicity. Med Sci (Basel), 10, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Tabor CW, Tabor H: Polyamines in microorganisms. Microbiol Rev, 49: 81–99, 1985. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Michael AJ: Polyamines in Eukaryotes, Bacteria, and Archaea. J Biol Chem, 291: 14896–14903, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Wilson WR, Bower TC, Creager MA, Amin-Hanjani S, O’Gara PT, Lockhart PB, Darouiche RO, Ramlawi B, Derdeyn CP, Bolger AF, Levison ME, Taubert KA, Baltimore RS, Baddour LM, American Heart Association Committee on Rheumatic Fever, E, Kawasaki Disease of the Council on Cardiovascular Disease in the, Y, Council on, C, Stroke, N, Council on Cardiovascular, R, Intervention, Council on Cardiovascular, S, Anesthesia, Council on Peripheral Vascular, D, Stroke, C: Vascular Graft Infections, Mycotic Aneurysms, and Endovascular Infections: A Scientific Statement From the American Heart Association. Circulation, 134: e412–e460, 2016. [DOI] [PubMed] [Google Scholar]
  • 116.Boils CL, Nasr SH, Walker PD, Couser WG, Larsen CP: Update on endocarditis-associated glomerulonephritis. Kidney Int, 87: 1241–1249, 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rana P, Aljabban J, Prarat M, Pancholi P, Balada-Llasat JM, Stephens J, Webb A, Chen L, Brodsky SV, Nadasdy T, Zhang Y, Parikh SV, Wozniak DJ, Wang SH, Olson M, Satoskar AA: Genomic Study on Blood Culture Isolates From Patients With Staphylococcus Infection-associated Glomerulonephritis. Kidney Int Rep, 7: 2264–2278, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Takayasu M, Hirayama K, Shimohata H, Kobayashi M, Koyama A: Staphylococcus aureus Infection-Related Glomerulonephritis with Dominant IgA Deposition. Int J Mol Sci, 23, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Luque Y, Louis K, Jouanneau C, Placier S, Esteve E, Bazin D, Rondeau E, Letavernier E, Wolfromm A, Gosset C, Boueilh A, Burbach M, Frere P, Verpont MC, Vandermeersch S, Langui D, Daudon M, Frochot V, Mesnard L: Vancomycin-Associated Cast Nephropathy. J Am Soc Nephrol, 28: 1723–1728, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Stokes MB, Stevens JS: Vancomycin-Associated Cast Nephropathy: Reality or Fantasy? Kidney360, 3: 372–375, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Ramos S, Carlos AR, Sundaram B, Jeney V, Ribeiro A, Gozzelino R, Bank C, Gjini E, Braza F, Martins R, Ademolue TW, Blankenhaus B, Gouveia Z, Faisca P, Trujillo D, Cardoso S, Rebelo S, Del Barrio L, Zarjou A, Bolisetty S, Agarwal A, Soares MP: Renal control of disease tolerance to malaria. Proc Natl Acad Sci U S A, 116: 5681–5686, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.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-1alpha promotes recovery after acute kidney injury during systemic inflammation in mice. J Clin Invest, 121: 4003–4014, 2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.McCullough K, Bolisetty S: Iron Homeostasis and Ferritin in Sepsis-Associated Kidney Injury. Nephron, 144: 616–620, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Smith JA, Mayeux PR, Schnellmann RG: Delayed Mitogen-Activated Protein Kinase/Extracellular Signal-Regulated Kinase Inhibition by Trametinib Attenuates Systemic Inflammatory Responses and Multiple Organ Injury in Murine Sepsis. Crit Care Med, 44: e711–720, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Takasu O, Gaut JP, Watanabe E, To K, Fagley RE, Sato B, Jarman S, Efimov IR, Janks DL, Srivastava A, Bhayani SB, Drewry A, Swanson PE, Hotchkiss RS: Mechanisms of cardiac and renal dysfunction in patients dying of sepsis. Am J Respir Crit Care Med, 187: 509–517, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Agarwal A, Dong Z, Harris R, Murray P, Parikh SM, Rosner MH, Kellum JA, Ronco C, Acute Dialysis Quality Initiative, XWG: Cellular and Molecular Mechanisms of AKI. J Am Soc Nephrol, 27: 1288–1299, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Wang A, Huen SC, Luan HH, Yu S, Zhang C, Gallezot JD, Booth CJ, Medzhitov R: Opposing Effects of Fasting Metabolism on Tissue Tolerance in Bacterial and Viral Inflammation. Cell, 166: 1512–1525 e1512, 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Ferre S, Deng Y, Huen SC, Lu CY, Scherer PE, Igarashi P, Moe OW: Renal tubular cell spliced X-box binding protein 1 (Xbp1s) has a unique role in sepsis-induced acute kidney injury and inflammation. Kidney Int, 96: 1359–1373, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zhou L, Pereiro MT, Li Y, Derigs M, Kuenne C, Hielscher T, Huang W, Kranzlin B, Tian G, Kobayashi K, Lu GN, Roedl K, Schmidt C, Gunther S, Looso M, Huber J, Xu Y, Wiech T, Sperhake JP, Wichmann D, Grone HJ, Worzfeld T: Glucocorticoids induce a maladaptive epithelial stress response to aggravate acute kidney injury. Sci Transl Med, 16: eadk5005, 2024. [DOI] [PubMed] [Google Scholar]
  • 131.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Holthoff JH, Wang Z, Seely KA, Gokden N, Mayeux PR: Resveratrol improves renal microcirculation, protects the tubular epithelium, and prolongs survival in a mouse model of sepsis-induced acute kidney injury. Kidney Int, 81: 370–378, 2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Steenvoorden TS, Rood JAJ, Bemelman FJ, Armstrong R Jr., Leuvenink HGD, van der Heijden JW, Vogt L: Alkaline phosphatase treatment of acute kidney injury-an update. Nephrol Dial Transplant, 39: 1239–1247, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Hato T, El-Achkar TM, Dagher PC: Sisters in arms: myeloid and tubular epithelial cells shape renal innate immunity. Am J Physiol Renal Physiol, 304: F1243–1251, 2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Jayaraman P, Rajagopal M, Paranjpe I, Suarez-Farinas M, Liharska L, Thompson R, Del Valle DM, Beckmann N, Lund AN, Gownivaripally P, Oh W, Gulamali FF, Kauffman J, Gonzalez-Kozlova E, Dellepiane S, Vasquez-Rios G, Vaid A, Jiang J, Fox B, Sakhuja A, Chen S, Kenigsberg E, He JC, Coca SG, Chan L, Merad M, Kim-Schulze S, Gnjatic S, Tsalik E, Langley R, Charney AW, Nadkarni GN: Peripheral Transcriptomics in Acute and Long-Term Kidney Dysfunction in SARS-CoV2 Infection. Kidney360, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Higgins SJ, De Ceunynck K, Kellum JA, Chen X, Gu X, Chaudhry SA, Schulman S, Libermann TA, Lu S, Shapiro NI, Christiani DC, Flaumenhaft R, Parikh SM: Tie2 protects the vasculature against thrombus formation in systemic inflammation. J Clin Invest, 128: 1471–1484, 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Seufert AL, Hickman JW, Traxler SK, Peterson RM, Waugh TA, Lashley SJ, Shulzhenko N, Napier RJ, Napier BA: Enriched dietary saturated fatty acids induce trained immunity via ceramide production that enhances severity of endotoxemia and clearance of infection. Elife, 11, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Yasuda H, Leelahavanichkul A, Tsunoda S, Dear JW, Takahashi Y, Ito S, Hu X, Zhou H, Doi K, Childs R, Klinman DM, Yuen PS, Star RA: Chloroquine and inhibition of Toll-like receptor 9 protect from sepsis-induced acute kidney injury. Am J Physiol Renal Physiol, 294: F1050–1058, 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Suzuki T, Loyde E, Chen S, Etzrodt V, Idowu TO, Clark AJ, Saade MC, Flores BM, Lu S, Birrane G, Vemireddy V, Seeliger B, David S, Parikh SM: Cathepsin K cleavage of Angiopoietin-2 creates detrimental Tie2 antagonist fragments in sepsis. J Clin Invest, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.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 Synthase (cGAS) Deficient Mice. Int J Mol Sci, 22, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Privratsky JR, Ide S, Chen Y, Kitai H, Ren J, Fradin H, Lu X, Souma T, Crowley SD: A macrophage-endothelial immunoregulatory axis ameliorates septic acute kidney injury. Kidney Int, 103: 514–528, 2023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Janosevic D, De Luca T, Melo Ferreira R, Gisch DL, Cheng YH, Hato T, Luo J, Yang Y, Hodgin JB, Phillips CL, Dagher PC, Kidney Precision Medicine, P, Eadon MT: miRNA and mRNA Signatures in Human Acute Kidney Injury Tissue. Am J Pathol, 195: 102–114, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.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. [DOI] [PubMed] [Google Scholar]
  • 150.Rayes J, Lax S, Wichaiyo S, Watson SK, Di Y, Lombard S, Grygielska B, Smith SW, Skordilis K, Watson SP: The podoplanin-CLEC-2 axis inhibits inflammation in sepsis. Nat Commun, 8: 2239, 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Nakazawa D, Masuda S, Nishibata Y, Watanabe-Kusunoki K, Tomaru U, Ishizu A: Neutrophils and NETs in kidney disease. Nat Rev Nephrol, 2025. [DOI] [PubMed] [Google Scholar]
  • 152.de Roquetaillade C, Durand M, Beaucote V, Guillemin J, Chadjichristos CE, Roquilly A, Chousterman BG: Progression of Kidney Fibrosis after Sepsis: The Underestimated Role of Resident Macrophages and Recruited Monocytes. J Am Soc Nephrol, 2025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Uchida M, Maier B, Waghwani HK, Selivanovitch E, Pay SL, Avera J, Yun E, Sandoval RM, Molitoris BA, Zollman A, Douglas T, Hato T: The archaeal Dps nanocage targets kidney proximal tubules via glomerular filtration. J Clin Invest, 129: 3941–3951, 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Chang YM, Chou YT, Kan WC, Shiao CC: Sepsis and Acute Kidney Injury: A Review Focusing on the Bidirectional Interplay. Int J Mol Sci, 23, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Kounatidis D, Tzivaki I, Daskalopoulou S, Daskou A, Adamou A, Rigatou A, Sdogkos E, Karampela I, Dalamaga M, Vallianou NG: Sepsis-Associated Acute Kidney Injury: What’s New Regarding Its Diagnostics and Therapeutics? Diagnostics (Basel), 14, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.He FF, Wang YM, Chen YY, Huang W, Li ZQ, Zhang C: Sepsis-induced AKI: From pathogenesis to therapeutic approaches. Front Pharmacol, 13: 981578, 2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Sun S, Chen R, Dou X, Dai M, Long J, Wu Y, Lin Y: Immunoregulatory mechanism of acute kidney injury in sepsis: A Narrative Review. Biomed Pharmacother, 159: 114202, 2023. [DOI] [PubMed] [Google Scholar]
  • 158.Rubio I, Osuchowski MF, Shankar-Hari M, Skirecki T, Winkler MS, Lachmann G, La Rosee P, Monneret G, Venet F, Bauer M, Brunkhorst FM, Kox M, Cavaillon JM, Uhle F, Weigand MA, Flohe SB, Wiersinga WJ, Martin-Fernandez M, Almansa R, Martin-Loeches I, Torres A, Giamarellos-Bourboulis EJ, Girardis M, Cossarizza A, Netea MG, van der Poll T, Scherag A, Meisel C, Schefold JC, Bermejo-Martin JF: Current gaps in sepsis immunology: new opportunities for translational research. Lancet Infect Dis, 19: e422–e436, 2019. [DOI] [PubMed] [Google Scholar]

RESOURCES