Abstract Acute kidney injury (AKI) manifests a hallmark pathological feature of extensive and severe DNA damage in renal tubules, primarily induced by the excessive of toxic reactive oxygen species (ROS) from the mitochondrial electron transport chain. The kidney's complex intricate physiological architecture and the heterogeneous intracellular environment pose significant challenges for effective sequential and high‐resolution drug delivery—an urgent issue that remains unresolved. To address this, a hierarchical‐targeting antioxidant nanodrug has been developed with a folic acid moiety (HAND) designed for high‐resolution drug delivery in AKI treatment. For the first time, HAND enables sequential targeting from the kidney to the most severely damaged proximal tubular epithelial cells (PTECs), ultimately concentrating in the DNA‐rich mitochondria and nucleus. As a result, HAND effectively scavenges ROS in situ, protecting both mitochondria and nuclei along with their vital genetic material. This action restores mitochondrial function, mitigates DNA oxidation and fragmentation, reduces apoptosis, and inhibits cGAS/STING‐mediated sterile inflammation. Consequently, HAND demonstrates remarkable efficacy in safeguarding injured kidneys during AKI. Overall, this work pioneers a hierarchical, high‐resolution antioxidant strategy, providing innovative guidance for the development of AKI therapies. Keywords: AKI, antioxidative nanomedicine, mitochondria, nucleus, oxidative stress, subcellular organelle targeting __________________________________________________________________ This study developed a hierarchical‐targeting antioxidant nanodrug (HAND) for precise AKI treatment, achieving targeted delivery from kidney to severely damaged proximal tubular epithelial cells, and concentrating in DNA‐rich mitochondria and nucleus. HAND effectively scavenges ROS in situ, protecting vital genetic material and demonstrating significant efficacy in preserving kidney function during AKI. graphic file with name ADVS-12-2411254-g005.jpg 1. Introduction Acute kidney injury (AKI) refers to the sudden deterioration of kidney function, primarily due to acute dysfunction of proximal tubular epithelial cells (PTECs).^[ [48]^1 ^] Globally, AKI affects over 13 million individuals annually and is particularly prevalent among hospitalized patients, with incidence rates reaching 30%−60% in critical care settings.^[ [49]^2 ^] Unfortunately, no pharmacological interventions have been successfully demonstrated to provide renal protection in AKI. Supportive care and renal replacement therapy are currently the only clinical treatment options,^[ [50]^3 ^] but their effectiveness is limited, and they impose a significant burden on healthcare systems. If untreated or inadequately managed, AKI can lead to renal failure and progress toward kidney fibrosis, driven by sustained oxidative stress and chronic inflammation, ultimately resulting in severe end‐stage renal disease, multi‐organ failure, or death.^[ [51]^4 ^] With an unacceptably high mortality rate of 1.7 million deaths annually, AKI presents a significant clinical challenge and underscores the urgent need for novel drug development.^[ [52]^5 , [53]^6 ^] AKI is marked by a defining pathological feature: extensive and severe DNA damage in renal tubules.^[ [54]^7 , [55]^8 ^] DNA, the repository of genetic information, is located exclusively in the nucleus and mitochondria of human cells, orchestrating cellular structure, function, and responses to stimuli through gene expression regulation.^[ [56]^9 ^] Damage to DNA disrupts this regulatory function and collapses the cellular command center, directly resulting in cell death.^[ [57]^10 ^] Moreover, irreparable DNA fragments released into the cytoplasm or extracellular space act as critical signals that activate innate DNA sensing pathways, triggering inflammatory cascades and cytokine storms.^[ [58]^11 ^] Consequently, the loss of functional cells combined with persistent inflammation significantly impairs renal function, leading to acute manifestations such as electrolyte imbalances, fluid overload, and acidosis, as well as the malignant progression of fibrosis.^[ [59]^12 ^] The primary source of DNA damage in AKI is endogenous oxidative stress, particularly from the production of toxic reactive oxygen species (ROS) by the mitochondrial electron transport chain (ETC).^[ [60]^13 ^] The kidneys, as one of the most mitochondria‐rich organs, receive 20% of cardiac output and utilize 10% of the body's oxygen.^[ [61]^14 ^] To support the intense transport and reabsorption functions in PTECs, mitochondria, serving as cellular power factories, are densely concentrated within these cells.^[ [62]^15 ^] Stimulants of AKI, such as rhabdomyolysis, systemic inflammation, or ischemia/reperfusion, trigger electron leakage from the mitochondrial ETC in PTECs, leading to a surge in mitochondrial ROS (mtROS).^[ [63]^16 ^] Critically, the high mitochondrial density within PTECs allows mtROS to rapidly diffuse through the mitochondrial pool, creating a storm that engulfs neighboring mitochondria and the nucleus, the latter of which is closely and fully surrounded by these mitochondria.^[ [64]^17 ^] As highly nucleophilic species with unpaired electrons, ROS engage in intense chemical reactions with electrophilic biomolecules, including DNA.^[ [65]^18 ^] These interactions result in substantial oxidative double‐stranded DNA damage, which subsequently activate cell death pathways and inflammatory signaling mediated by the cGAS/STING axis.^[ [66]^19 ^] Thus, the damage inflicted by ROS on the two crucial DNA‐rich organelles in PTECs—mitochondria and the nucleus—propagates rapidly, akin to a cascading domino effect, ultimately compromising the entire kidney. Therefore, effectively neutralizing excessive toxic ROS represents a crucial therapeutic objective to mitigate the source of damage and protect PTECs, ultimately improving renal function. However, clinically utilized antioxidant molecules, such as amifostine and N‐acetylcysteine (NAC), offer limited protection against AKI due to their lack of targeting capability and poor bioavailability. In recent years, emerging nanomedicine has demonstrated a remarkable trajectory of advancement,^[ [67]^20 , [68]^21 ^] with several mitochondria‐specific targeting nanostrategies under active investigation.^[ [69]^22 , [70]^23 ^] Nonetheless, these approaches face significant challenges in achieving effective and precise treatment for AKI. First, the presence of positive charges and/or hydrophobic structures in mitochondria‐targeting compounds limits their stability and in vivo applicability, as negatively charged enzymes or proteins in circulation may interact with them, thereby diminishing the targeting efficacy.^[ [71]^24 ^] Additionally, complex encapsulation or modification can result in increased particle size, complicating passage through the glomerular filtration barrier (GFB) and raising safety concerns related to residual materials.^[ [72]^25 ^] Second, some nanomaterials that are appropriately sized for the GFB often fail to effectively target PTECs, the primary site of injury in AKI, leading to a rapid loss of therapeutic efficacy as they are swiftly excreted in the urine. The nucleus, serving as the cell's primary command center, is often overlooked by researchers, leading to inadequate protection. This neglect is particularly concerning, given the close spatial and functional interconnections between mitochondria and the nucleus,^[ [73]^26 ^] both of which are highly susceptible to ROS. Thus, developing a hierarchical targeting strategy for the precise delivery of therapeutic agents to kidney‐PTECs‐mitochondria and the nucleus—facilitating targeted ROS clearance at the tissue, cellular, and subcellular levels—holds significant promise for innovative and effective treatment of AKI. However, the unique physiological structure of the kidney and the complex, heterogeneous intracellular environment present formidable challenges to achieving such sequential and high‐precision targeting. As a result, no practical applications from this perspective have been reported to date. To address this challenge, a portable and elegant hierarchical‐targeting antioxidant nanodrug (HAND) derived from folic acid (FA) and curcumin (Scheme [74]1i) has been developed for precise delivery to the mitochondria and the nucleus in PTECs, which enabled the specific and highly efficient clearance of toxic mtROS and mitigation of DNA damage. Curcumin, a lipophilic polyphenol compound extracted from turmeric rhizomes, has demonstrated a wide range of pharmacological activities, including antioxidant, antimicrobial, and anti‐inflammatory properties. Moreover, curcumin was recognized as a “generally regarded as safe” compound by the U.S. Food and Drug Administration. However, its therapeutic efficacy in treating AKI is limited by poor bioavailability, rapid metabolism, and low water solubility when used alone. Fortunately, the chemical structure of curcumin, characterized by abundant sp2 and sp3 carbon atoms along with antioxidant functional groups such as phenolic hydroxyl and methoxy groups, makes it an ideal precursor for synthesizing carbon dots with antioxidant properties. In this study, we have integrated curcumin with the targeting molecule FA into integrated carbon dots──HAND, not only enhancing curcumin's bioavailability but also promoting its precise delivery to AKI lesions. The ultrasmall HAND is abundant in FA‐like groups and possesses a strong negative charge on its surface. Therefore, HAND was less susceptible to sequestration by the hepatic reticuloendothelial system, facilitating its accumulation in the kidneys and enabling it to penetrate the GFB to reach the renal tubular lumen. Furthermore, PTECs expressed high levels of FA receptors (FR) on their surface, granting them a high affinity for HAND and facilitating efficient endocytosis. Notably, we have identified, for the first time, that FA demonstrates a strong affinity for translocases of the outer and inner mitochondrial membrane (TOM complex and TIM23 complex) and FG nucleoporins (FG Nups) within nuclear pore complexes. This property is effectively inherited by HAND, which shows pronounced co‐localization with both the nucleus and mitochondria in human renal tubular epithelial cells (HK‐2 cells). Therefore, HAND can actively target PTECs and preferentially accumulate in DNA‐rich regions (Scheme [75]1ii). By leveraging its abundant antioxidant groups, HAND efficiently scavenged toxic mtROS, thereby protecting the mitochondria and nucleus, restoring mitochondrial function, mitigating apoptosis, and attenuating cGAS/STING‐mediated sterile inflammation (Scheme [76]1iii,iv). This multifaceted approach effectively safeguarded PTECs and facilitated the restoration of renal function in AKI mouse models. Moreover, HAND, derived from natural products (curcumin) and essential nutrients (FA), exhibited commendable biocompatibility and was effectively eliminated from the body through renal clearance within 72 h. In conclusion, this study introduces a high‐resolution, hierarchical targeting antioxidant therapeutic strategy, providing a new paradigm for the development of therapeutic agents for renal diseases, including but not limited to AKI. Scheme 1. Scheme 1 [77]Open in a new tab Hierarchical Targeting Nanodrug with Holistic DNA Protection for Effective Treatment of Acute Kidney Injury. i) HAND was synthesized using curcumin and FA as precursor compounds through a hydrothermal method. Infrared spectroscopy results indicated that HAND successfully inherited the surface groups of FA. ii) Upon intravenous injection, ultrasmall HAND could passively transverse the GFB and be actively taken up by PTECs through FR‐mediated endocytosis. Furthermore, HAND could localize in the nucleus and mitochondria due to the high affinity of FA for their surface membrane proteins. iii) HAND effectively protected mitochondrial structure and function via scavenging mtROS, subsequently inhibiting damage‐induced mitophagy and intrinsic apoptosis. iv) Through effectively scavenging mtROS, HAND could reduce the release of dsDNA, thereby suppressing sterile inflammation caused by cGAS/STING pathway activation. v) TEM image of HAND (a). Insert: Size distribution of HAND. XPS N1s spectrum of HAND (b). UV absorption spectra of O[2] ^·− reacting with different concentrations of HAND. 2. Results 2.1. Synthesis and Characterization of HAND In this study, we synthesized HAND and CND (without FA) by inducing a carbonization process using FA and curcumin as carbon source precursors at 240 °C (Scheme [78]1i–a). Transmission electron microscopy (TEM) confirmed that HAND and CND were monodisperse, spherical nanoparticles with an average diameter of ≈11 nm (Scheme [79]1v–a; Figure [80]S1, Supporting Information), well below the particle size threshold for GFB (≈14 nm). The surface charges of HAND and CND were −34.5 and −23.5 mV, respectively, supporting their stable dispersion in circulation (Figure [81]S2, Supporting Information). We subsequently used X‐ray photoelectron spectroscopy (XPS) to analyze the composition of HAND. The XPS spectrum of HAND showed three main peaks: C1s (285 eV), N1s (400 eV), and O1s (534 eV) (Figure [82]S3, Supporting Information), while CND exhibited only two peaks: C1s and O1s (Figure [83]S4, Supporting Information), indicating that HAND inherited nitrogen from FA. As illustrated in Scheme [84]1v–b, the high‐resolution XPS N1s spectrum of HAND identified three distinct peaks attributed to pyridinic nitrogen (400.5 eV), amino nitrogen (401.6 eV), and graphitic nitrogen (402.6 eV). Concurrently, the XPS C1s spectrum (Figure [85]S5, Supporting Information) of HAND displayed five distinct peaks corresponding to C─C, C═C, C─N/C─O, C═O, and π–π^* transitions, suggesting a nitrogen‐doped carbon framework with FA‐derived surface groups. Fourier‐transform infrared (FT–IR) spectroscopy further supported these findings, revealing N‐containing absorption peaks such as ─NH, C═N, and C─N, consistent with FA, alongside typical carbon dot peaks for ─OH, C─H, C═C, and C─O (Scheme [86]1i–b). In contrast, CND showed only characteristic carbon dot peaks for ─OH, C─H, C═O (1638 cm⁻¹), C═C, and C─O (Figure [87]S6, Supporting Information). UV–vis spectroscopy of HAND and CND revealed two major absorption bands at 277 and 300–400 nm, corresponding to π–π^* transitions of polycyclic aromatic chromophores (C═C) and n‐π^* transitions of C─N/C═O, respectively (Figure [88]S7, Supporting Information). These results confirmed the successful synthesis of HAND, which exhibited an N‐doped conjugated carbon framework with an abundant surface group derived from FA and curcumin as expected. Consequently, these surface groups, including methoxy, phenolic hydroxyl, and amino groups inherited from FA and curcumin, endowed HAND with exceptional and broad‐spectrum ROS scavenging capacity. For example, HAND effectively neutralized O[2] ^·⁻ in vitro, counteracting the primary ROS generated by mitochondrial ETC electron leakage, thus potentially limiting mtROS propagation (Scheme [89]1v–c). As a reductive sacrificial agent, HAND also effectively detoxified H₂O₂, a longer‐lived ROS, and secondary radicals derived from O[2] ^·⁻, such as ·OH and ONOO^⁻ (Figure [90]S8, Supporting Information). Similarly, CND demonstrated comparable ROS scavenging activity due to the antioxidant groups inherited from curcumin (Figure [91]S9, Supporting Information). Thus, HAND's broad‐spectrum antioxidant activity as a reductive sacrificial agent was robustly validated. In conclusion, HAND was successfully synthesized and comprehensively characterized, confirming its expected structure and potent antioxidant properties. With its ultra‐small size and potential for active targeting to PTECs, HAND holds considerable promise for renal accumulation, which is evaluated in subsequent animal models. 2.2. Hierarchical Targeting of HAND: from Kidney to PTECs, and Ultimately to Mitochondria and Nucleus To investigate the hierarchical targeting properties of HAND, a rhabdomyolysis‐induced AKI (RM‐AKI) mouse model was established by injecting equal volumes of 50% glycerol into both hind limbs. RM‐AKI, commonly encountered in trauma and perioperative contexts, represents one of the most prevalent forms of AKI in clinical practice. Following intravenous administration of HAND to the AKI mice, TEM images of the glomerulus were captured to assess the passage of HAND through the GFB. As shown in Figure [92]1A, HAND could be directly observed in the glomerular capsule, confirming its successful traversal of the GFB. To further track their distribution in vivo, HAND and CND were labeled with fluorescein isothiocyanate to form FITC‐HAND and FITC‐CND, which showed comparable particle size and surface charge to HAND and CND (Figure [93]S10, Supporting Information). The labeling process involved the reaction of the isothiocyanate group of FITC with the amino and hydroxyl functional groups present in HAND and CND, forming stable covalent bonds (Figure [94]S11, Supporting Information). The resulting conjugates were then visualized through stereo‐fluorescence microscopy to assess their distribution and localization in biological systems. As shown in Figure [95]1B and Figures [96]S12,S13 (Supporting Information), FITC‐HAND and FITC‐CND demonstrated significant renal accumulation in both Control and AKI mice, with minimal distribution in the liver and lungs, and negligible presence in the heart and spleen. Such in vivo pharmacokinetic behavior of HAND and CND can be attributed to their negative charge, ultra‐small size, and excellent water solubility. We then further analyzed the temporal variation in renal concentrations of HAND and CND. Within 1 h post‐injection, a distinct green fluorescence was observed in the kidneys of the AKI+FITC‐HAND and AKI+FITC‐CND groups, with fluorescence intensity increasing over time, peaking ≈6 h before gradually declining due to drug excretion (Figure [97]1C; Figures [98]S14,S15, Supporting Information). Notably, the peak fluorescence intensity (6 h) and fluorescence duration (24 h) of FITC‐HAND in the kidneys were significantly greater than those of FITC‐CND (Figure [99]S16, Supporting Information). This sustained accumulation highlighted the superior of HAND in actively targeting the focus, ensuring more specific and prolonged ROS scavenging in PTECs, as opposed to merely retaining in the tubular lumen or being rapidly excreted in urine. The enhanced specific and sustained accumulation of HANDs in the kidneys compared with CNDs may be attributed to surface groups inherited from FA, which specifically bind to FR‐rich PTECs. Meanwhile, HAND was labeled with a Cy7 probe and administered to mice for in vivo imaging. The results showed that once in circulation, HAND rapidly accumulated in the kidneys, demonstrating time‐dependent uptake that peaked at 6 h before gradually declining. This accumulation pattern is well‐aligned with stereo fluorescence imaging results, providing strong evidence of the targeted renal distribution of HAND. Notably, the distribution of HAND in other organs, such as the liver, was relatively low, further supporting its kidney‐specific targeting capability (Figure [100]S17, Supporting Information). Figure 1. Figure 1 [101]Open in a new tab The hierarchical targeting property of HAND. A) TEM images of GFB in different groups. B) Representative fluorescence images of different organs in AKI mice 6 h after intravenous injection of HAND. C) Representative fluorescence images of kidneys in AKI mice at various time points (1‐72 h) after intravenous injection of HAND. D) Representative IF staining images of FITC‐HAND with glomerular (synaptopodin) and tubular (AQP1) markers in mice kidney tissues, with yellow circles indicating glomeruli. E) Representative IF staining image of FITC‐HAND with FR in mice kidney tissues. F,G) Representative fluorescence images (F) and quantitative fluorescence analysis (G) of HK‐2 cells incubated with FITC‐HAND or FITC‐CND in different groups. H) Colocalization fluorescence images and correlation coefficient analysis of FITC‐HAND with different organelles in HK‐2 cells. I–L) Molecular docking results of FA with TOM comple, TIM23, and FG Nups. Data are presented as mean ± SD. One‐way ANOVA followed by the Student–Newman–Keuls (SNK) test was used for analysis. n = 3, ^*** p < 0.001, ^**** p < 0.0001; ^## p < 0.01, ^### p < 0.001; ns, not significant (p > 0.05). Thereafter, we further delved into the influence of FA‐derived groups on the more microcosmic targeting properties of HANDs at the cellular and organellar levels. FA, an essential micronutrient, is primarily regulated by the kidneys to maintain systemic homeostasis.^[ [102]^27 ^] Upon entering circulation, FA is freely filtered through the glomerulus, with most being reabsorbed by PTECs to prevent excretion. The reabsorption process relies heavily on the FR located on the brush border of PTECs.^[ [103]^28 ^] Our immunofluorescence (IF) staining of human kidney samples confirmed that PTECs express high levels of FR, establishing a foundation for the cell‐specific targeting of FA‐functionalized HAND (Figure [104]S18, Supporting Information). As expected, the ultrasmall FITC‐HAND and FITC‐CND were able to cross the GFB, primarily distributing within renal tubules (marked by AQP1) rather than glomeruli (marked by synaptopodin) in mouse kidney tissues (Figure [105]1D; Figure [106]S19, Supporting Information). Importantly, FITC‐HAND derived from FA showed a high degree of colocalization with FR on the surface of PTECs, which was significantly greater than that of FITC‐CND, highlighting the active targeting capability of HAND toward FR (Figure [107]1E; Figure [108]S20, Supporting Information). We also verified HAND's FR‐targeting ability at the cellular level by comparing HAND uptake across various cell lines with differing FR expression. As shown in Figure [109]S21 (Supporting Information), FITC‐HAND was effectively and abundantly taken up by FR‐high‐expressing HK‐2 cells, while its uptake by FR‐low‐expressing cardiomyocytes (H9c2) and hepatic stellate cells (LX2) was lower under same conditions. In contrast, FITC‐CND exhibited no significant differences in uptake among these cell lines. Notably, pre‐treatment with FA significantly inhibited FITC‐HAND uptake in HK‐2 cells, evidenced by a marked decrease in intracellular fluorescence (Figure [110]1F,G). This competitive inhibition indicates that HAND internalization is primarily FR‐mediated. Conversely, under the same conditions, FITC‐CND exhibited markedly lower fluorescence intensity in HK‐2 cells compared to FITC‐HAND, and FA pre‐treatment did not affect its uptake. Together, these findings underscore the FR‐dependent targeting capability of HAND. Furthermore, FA is a pivotal component of intracellular one‐carbon metabolism, exhibiting a compartmentalized pattern of utilization, predominantly within the mitochondria and nucleus. Such compartmentalization likely drives the selective intracellular localization of HAND. Specifically, HAND exhibited a pronounced dual‐targeting specificity for the nucleus and mitochondria in HK‐2 cells, with Pearson's correlation coefficient of 0.85 and 0.68, respectively, corresponding to the primary compartments involved in FA‐mediated one‐carbon metabolism (Figure [111]1H). To further elucidate the targeting and adhesion mechanisms of HAND, we employed molecular docking to assess FA's affinity for mitochondrial and nuclear proteins. TOM and TIM are critical macromolecular complexes embedded in the outer and inner mitochondrial membranes, respectively, facilitating the localization and import of nuclear‐encoded proteins into mitochondria.^[ [112]^29 ^] The nuclear pore complex (NPC) serves as the principal gateway for macromolecular transport between the nucleus and cytoplasm. NPC is highly conserved and intricate, with FG Nups in its central channel mediating the interactions with cargo.^[ [113]^30 ^] Molecular docking results indicated that FA achieved high docking scores with TOM, TIM, and FG Nups due to its molecular flexibility, stable conformation, and effective non‐bonded interactions, with calculated free binding energies of −6.15, −7.10, and −6.76 kcal mol^−1, respectively (Figure [114]1I–L). To further validate the affinity of FA‐mediated HAND for TOM, TIM, and FG Nups, we pre‐treated HK‐2 cells with FA before incubation with FITC‐HAND and examined its the colocalization of FITC‐HAND with various organelles. As illustrated in Figure [115]S22 (Supporting Information), pre‐treatment with FA significantly reduced the distribution of HAND in the mitochondria and nucleus, with co‐localization coefficients decreasing to 0.37 and 0.42, respectively. In contrast, there were no significant changes in the co‐localization of HAND with the endoplasmic reticulum (ER), Golgi apparatus, and lysosomes (Lyso), confirming that the dual‐targeting capability of HAND is mediated by FA. These findings provided compelling validation and mechanistic insights into the distinct dual‐targeting affinity of HAND for both mitochondria and the nucleus. In summary, the above evidence strongly demonstrated the hierarchical targeting properties of HAND: sequentially from the kidney to PTECs, and ultimately to the mitochondria and nucleus, which is characterized by high and persistent accumulation in the kidneys, specific binding to PTECs, and subsequent precise localization targeting to mitochondria and nucleus. 2.3. HAND Protected the Kidneys of AKI Mice by Suppressing Oxidative Stress Given its hierarchical targeting (Figure [116]2A) and potent antioxidant properties, the in vivo therapeutic effect of HAND on AKI was further explored in RM‐AKI mice. Renal tubular injury scores and renal function indicators are standard criteria for assessing the extent of renal damage in AKI. The renal tubular injury score, ranging from 0 to 5, reflects the severity of renal damage based on renal tubule morphology, with higher scores indicating more severe injury. Renal function is evaluated through serum creatinine (CRE) and urea nitrogen (BUN) levels, both of which increase with declining kidney function. In addition, neutrophil gelatinase‐associated lipocalin (NGAL) is a sensitive biomarker for early kidney injury, providing important information about renal damage. We first optimized the dosage of HAND for the treatment of AKI. As shown in renal function indicators detection results (Figure [117]S23, Supporting Information), HAND could increase the therapeutic effects of AKI in a dose‐dependent manner, with the optimal efficacy observed at a dose of 2 mg kg^−1. We then compared the effects of HAND, CND, and the clinical antioxidant NAC on the treatment of AKI were compared at the same dosage. Renal cortex morphology was observed by HE staining and the degree of renal tubular injury was scored accordingly. As shown in Figure [118]2B,C, RM induced severe renal tubular injury in the AKI group, characterized by obvious dilation and deformation of the renal tubules, loose arrangement between tubules, deposition of substances in the tubular lumen (casts), loss of the brush border of PTECs, PTECs swelling, necrosis, and sloughing, with a renal injury score as high as 4.5. Treatment with 2 mg kg^−1 HANDs effectively alleviated the pathological damage of the kidneys, restoring tubule morphology and structure to near‐normal conditions. In contrast, the pathological damage remained severe in the CND group (lacking PTECs targeting) and the NAC group (Figure [119]2B,C), with renal injury scores of 3.2 and 3.8, respectively, which was primarily due to their lower targeting and accumulation at PTECs. Moreover, the serum CRE (Figure [120]2D) and BUN (Figure [121]2E) levels in the AKI group rose sharply, reaching 4.67 and 3.59 times those of the Control group, meeting the criteria for stage 3 AKI. Similarly, NGAL levels in serum and urine also surged (Figure [122]2F; Figure [123]S24, Supporting Information), confirming the severity of renal injury. HAND treatment provided superior renal protection compared to the CND and NAC groups, with CRE, BUN, and NGAL levels returning to normal. During the experimental period, mice in the AKI group exhibited significant weight loss, whereas those in the HAND group displayed normal growth patterns (Figure [124]S25, Supporting Information). HO‐1 and KIM‐1, specific biomarkers of renal injury, are minimally expressed in normal renal tissues but are markedly upregulated in PTECs during renal impairment. Compared with the AKI group, levels of HO‐1 (Figure [125]2G) and KIM‐1 (Figure [126]2H) in the HAND group returned to normal, demonstrating far superior therapeutic effects compared to the CND group, while the NAC group showed negligible therapeutic benefit. Figure 2. Figure 2 [127]Open in a new tab HAND protected the kidneys of AKI mice by suppressing oxidative stress. A) Schematic illustration of HAND crossing the GFB and actively targeting PTECs. B) Representative H&E‐stained images of kidney tissues from different treatment groups. Arrows indicate damaged PTECs, and asterisks indicate casts. C–E) Kidney injury scores (C), levels of serum CRE (D), and BUN (E) in different treatment groups. F–H) Levels of NGAL (F), HO‐1 (G), and KIM‐1 (H) in kidney tissues of different treatment groups. I) Representative DHE fluorescence images of kidney tissues in different treatment groups. J) IHC results of 8‐OHdG in mouse kidneys from different treatment groups. Data are presented as mean ± SD. One‐way ANOVA followed by the SNK test was used for analysis. n = 6, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001; ns, not significant (p > 0.05). The pharmacological basis of HAND is its ability to inhibit oxidative stress. To investigate this, we conducted a thorough examination of HAND's in vivo antioxidant activity as a meticulously designed reductive scavenger. Dihydroethidium (DHE) probe staining (Figure [128]2I; Figure [129]S26, Supporting Information) revealed that the ROS level in the renal tissues of AKI mice was significantly elevated compared to the Control group. After treatment with HAND, the level of ROS markedly decreased to baseline, demonstrating its effectiveness in alleviating oxidative stress in the kidneys during AKI. In comparison, both CND and NAC exhibited weaker antioxidant activity at the same dosage, primarily due to their limited capacity for active targeting of PTECs. As highly destructive molecules, ROS inflict extensive cellular oxidative damage by aggressively attacking biomacromolecules, particularly DNA and membrane lipids in mitochondria and the nucleus.^[ [130]^31 ^] As shown in the IHC results of the DNA oxidation marker, 8‐OHdG, DNA was severely damaged and widely diffused throughout the PTECs, affecting both the nucleus and mitochondria in the AKI group (Figure [131]2J). Elisa's results further corroborated a significantly elevated level of 8‐OHdG in the kidney tissues of the AKI group (Figure [132]S27, Supporting Information). Thanks to the precise targeting of mitochondria and the nucleus, HAND offered comprehensive protection for the DNA by significantly reducing DNA oxidative damage, as evidenced by the decreased levels of 8‐OHdG in both IHC and Elisa results. In comparison, CND and NAC exhibited lower DNA protection at the same dosage. Moreover, the γ‐H2AX staining results provided additional confirmation of HAND's efficacy in mitigating DNA damage. In the AKI group, a marked increase in γ‐H2AX foci was observed in PTECs, indicating widespread DNA double‐strand breaks (DSBs) (Figure [133]S28, Supporting Information). However, treatment with HAND significantly reduced the number of γ‐H2AX‐positive cells. In contrast, CND and NAC treatments resulted in higher γ‐H2AX levels, underscoring their comparatively weaker protection against DSBs. These results collectively validate the superior capability of HAND in safeguarding DNA integrity under oxidative stress conditions in AKI. Moreover, the antioxidant effects of HAND are further underscored by the reduction in damage to lipids. As shown in Figure [134]S29 (Supporting Information), levels of the lipid peroxidation product products malondialdehyde (MDA) and thiobarbituric acid reactive substances (TBARS) in the kidneys of the AKI group were significantly elevated, reaching two to three times those of the Control group. In the HAND group, these levels returned to normal, indicating that the heightened oxidative stress in kidney tissues was effectively mitigated by HAND. However, the levels of lipid peroxidation products in the NAC group showed no significant reduction, and the antioxidant effect of CND was notably weaker compared to the HAND group. To sum up, owing to its high‐precision hierarchical targeting and outstanding antioxidant properties, HAND demonstrated superior therapeutic benefits over CND and NAC in RM‐AKI mice, which are underscored by its effective suppression of severe oxidative stress in AKI kidneys. 2.4. RNA Sequencing Excavated the Therapeutic Mechanism of HAND To further explore the pathological mechanisms of AKI and the pharmacological effects of HAND, we conducted RNA sequencing analysis (RNA‐Seq) on kidney samples from Control, AKI, and HAND groups. The quality and reliability of the data were evaluated through correlation analysis (Figure [135] 3A) and principal component analysis (Figure [136]S30, Supporting Information). These analyses revealed complete separation among samples from different groups, indicating significant intergroup differences and high intragroup correlation. Subsequently, we screened for differentially expressed genes (DEGs) using the criteria of |log[2]FC| > 1 and Q < 0.05. The Venn diagram (Figure [137]3B) and volcano plot (Figure [138]3C) revealed 1246 overlapping DEGs between the AKI group and the other two groups (Control group and HAND group). These DEGs underwent enrichment and clustering analyses, leveraging the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases to uncover gene functions and pathways pertinent to our study. The GO enrichment analysis indicated that the DEGs between the AKI and HAND groups were primarily associated with the regulation of cellular processes such as ROS metabolism, ROS response, inflammation response, DNA damage response, apoptosis process, mitophagy, and mitochondrial ETC activity (Figure [139]3D). KEGG pathway enrichment analysis further revealed that these genes were involved in the regulation of pathways related to ROS, oxidative phosphorylation, cell cycle, apoptosis, TNF signaling, NF‐κB signaling, mitophagy, cytokine‐cytokine receptor interactions, cytosolic DNA sensing, and the citric acid cycle (Figure [140]3E). These findings underscored the therapeutic potential of HAND in alleviating oxidative stress, protecting mitochondria and DNA, and suppressing excessive inflammation during AKI, which benefited from its precise targeting capabilities and broad‐spectrum antioxidant activity. We further illustrated gene expression variations across samples through visual clustering heatmaps. As shown in Figure [141]3F, the overall gene expression pattern of the HAND group closely resembled that of the Control group, whereas the AKI group showed significant divergence from the other two, suggesting that HAND effectively reversed the gene expression profile alterations induced by AKI. Additionally, we constructed clustering heatmaps for five key categories—oxidative stress, apoptosis, mitophagy, inflammation, and DNA sensing—highlighted from the above enrichment analysis. As depicted in Figure [142]3G–K, the gene expression patterns in the HAND group more closely aligned with those in the Control group, further confirming the regulatory efficacy of HAND on these critical gene categories, which will be further explored in detail in the following sections. Figure 3. Figure 3 [143]Open in a new tab RNA‐seq revealed the therapeutic mechanisms of HAND. A) Heatmap showing the correlation of gene expression from different groups in RNA‐seq of mouse kidney tissues. B) Venn diagram of DEGs in RNA‐seq of kidney tissues. C) Volcano plot of DEGs between the AKI group and the AKI+HAND group. D,E) GO enrichment chord diagram (D) and KEGG pathway enrichment bubble chart (E) for DEGs between the AKI and AKI+HAND groups. F–K) Cluster heatmap of gene expression across different groups (F) and for five functional categories: oxidative stress (G), apoptosis (H), mitophagy (I), inflammation (J), and DNA sensing (K). n = 3, |log[2]FC| > 1, Q < 0.05. 2.5. HAND Comprehensively Protected Mitochondria Mitochondria, as prime examples of endosymbiotic integration, coordinate critical cellular and organelle signaling pathways essential for adaptive responses and evolutionary processes, making them pivotal signaling hubs that determine cell fate.^[ [144]^32 , [145]^33 ^] During AKI, electron leakage from the mitochondrial ETC turns mitochondria into a primary source of ROS. Simultaneously, these mitochondria become the main victims of the resulting toxic ROS, which leads to severe mitochondrial dysfunction, disrupted energy metabolism, and heightened ROS production, ultimately triggering a ROS storm that engulfs the entire PTECs.^[ [146]^34 , [147]^35 ^] Consequently, mitochondrial health emerges as a crucial target in AKI therapy, serving as a direct indicator of the effectiveness of therapeutic interventions, a theme we have examined comprehensively in this section. HAND exhibited precise mitochondrial targeting and sustained antioxidant activity, enabling it to effectively eliminate mtROS in situ and protect mitochondria from oxidative damage and its detrimental consequences (Figure [148]4A). We conducted a series of experiments to validate the comprehensive protective effects of HAND on mitochondrial structure, function, oxidative stress status, and homeostatic regulation. First, we analyzed the mitochondrial morphology of PTECs across different groups using TEM. In contrast to the intact mitochondria observed in the control group, those in the AKI group exhibited significant structural damage, including swelling, rupture, and cristae fragmentation (Figure [149]4B). Following HAND treatment, mitochondrial morphology in the AKI mice showed notable restoration, while the CND group continued to display morphological abnormalities. Next, we investigated the mitochondrial protective mechanisms of HAND in HK‐2 cells. DCFH‐DA (ROS probe) detection results showed that HAND significantly alleviated the overall increase in intracellular ROS levels induced by H[2]O[2] (Figure [150]4C,D). Additionally, mitoSOX (mtROS‐specific probe with red fluorescence) fluorescence imaging and flow cytometry analysis further revealed the potent mtROS clearance of HAND under H[2]O[2] stimulation, underscoring its targeted mitochondrial protection (Figure [151]4E,F; Figure [152]S31, Supporting Information). Furthermore, mitochondria maintain an electrochemical gradient across their inner membrane via proton pumps in the ETC, which drives ATP synthesis. Thus, mitochondrial membrane potential (MMP) and ATP levels are key indicators of mitochondrial function. We evaluated the effect of HAND on MMP under H[2]O[2] stimulation using the JC‐1 fluorescent dye. As shown in Figure [153]4G,H; Figure [154]S32 (Supporting Information), H[2]O[2] administration led to a significant decrease in MMP, which was indicated by the predominant green fluorescence corresponding to JC‐1 monomers in the mitochondria. Co‐incubation with HAND effectively mitigated the loss of MMP, resulting in the formation of JC‐1 aggregates that emitted red fluorescence in mitochondria. Consequently, the restoration of MMP ensured the efficiency and integrity of the mitochondrial ETC, leading to a notable recovery of ATP production in the HAND group compared to the H[2]O[2] group (Figure [155]4I). Moreover, mitophagy is another indicator of mitochondrial damage, aiding in the removal of impaired mitochondria to prevent further damage.^[ [156]^36 ^] Mitophagy is tightly regulated by various signaling pathways and proteins, with the PINK1‐Parkin pathway being the most important.^[ [157]^37 ^] PINK1, which contains a mitochondrial targeting sequence at its N‐terminus, is normally located in mitochondria, where it is cleaved and degraded by the proteasome. However, disruptions such as MMP loss disrupt this process, leading to PINK1 accumulation on the outer mitochondrial membrane. Activated PINK1 recruits Parkin from the cytosol to the mitochondria, where Parkin, an E3 ubiquitin ligase, ubiquitinates outer mitochondrial membrane proteins, attracting autophagic vesicle proteins like LC3B II. During this process, P62/SQSTM1 acts as a bridging protein, linking LC3B II with ubiquitinated proteins on the mitochondrial surface, promoting the formation of autophagosome1^[ [158]^12 ^] (Figure [159]4A). TEM images showed that during AKI, high levels of mtROS caused severe mitochondrial damage and activated mitophagy, resulting in the presence of many autophagosomes in PTECs (Figure [160]4J). Western blot (WB) analysis further revealed that the expression levels of mitophagy‐related proteins, including PINK1, Parkin, P62, and LC3BII/I, were significantly elevated in AKI kidneys, indicating high mitophagy activation. HAND treatment reduced the number of autophagosomes in PTECs and downregulated the expression of mitophagy‐related proteins compared to the AKI group, suggesting reduced mitochondrial damage (Figure [161]4K; Figure [162]S33, Supporting Information). In summary, HAND provided exceptional, comprehensive protection for mitochondria due to its high‐resolution mitochondrial localization and remarkable antioxidant capacity, thereby preserving the most critical organelles in PTECs during AKI. Figure 4. Figure 4 [163]Open in a new tab Comprehensive mitochondrial protection of HAND. A) Schematic illustration of the mitochondrial protecting ability of HAND. B) TEM images of mitochondria in PTECs from different treatment groups of mice. C,D) Representative DCFH‐DA fluorescence images (C) and corresponding fluorescence intensity (D) of HK‐2 cells in different treatment groups. E,F) Mito‐SOX fluorescence staining images and corresponding fluorescence intensity (F) of HK‐2 cells from different treatment groups. G,H) JC‐1 fluorescence staining images (G) and corresponding fluorescence intensity (H) of HK‐2 cells from different treatment groups. I) ATP production of HK‐2 cells from different treatment groups. J) TEM images of mitophagy in PTECs from different treatment groups of mice. K) The grayscale analysis of PINK1, Parkin, P62, and LC3BII/I of Western blot results from kidney tissues in different treatment groups. Data are presented as mean ± SD. One‐way ANOVA followed by the SNK test was used for analysis. n = 3, ^* p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001. 2.6. HAND Effectively Inhibited Apoptosis of PTECs As central arbiters of cellular function and integrity, mitochondria are crucial in mediating cell death signaling, particularly in intrinsic apoptosis (also known as the mitochondrial pathway of apoptosis)^[ [164]^38 ^] (Figure [165]5A). In the pathological state of AKI, mtROS damages mitochondrial lipids and proteins, leading to compromised membrane integrity and inducing mitochondrial outer membrane permeabilization (MOMP). MOMP, the initiating event of intrinsic apoptosis, is primarily mediated by pro‐apoptotic members of the Bcl‐2 family, such as Bax. Under oxidative stress, Bax accumulates on the mitochondrial outer membrane, forming pores that enable the release of essential apoptotic factors, such as cytochrome c (Cyt c), into the cytosol. Once released, Cyt c promptly activates the caspase cascade, with Caspase‐3 serving as the principal executor. Upon activation through cleavage, it degrades various cellular components, thereby driving apoptosis.^[ [166]^39 ^] As shown in the WB results (Figure [167]5B–G), expression levels of Bax and Cytosolic‐Cyt c (C‐Cyt c) were significantly upregulated in the kidneys of the AKI group, while the expression of the anti‐apoptotic protein Bcl‐2 was downregulated, indicating the activation of intrinsic apoptosis. Furthermore, the caspase cascade was effectively triggered, with Caspase‐3 being cleaved and downregulated, which corresponded to a significant increase in Cleaved‐Caspase‐3 levels, confirming the execution of intrinsic apoptosis. Importantly, following HAND treatment, the expression of mitochondrial pro‐apoptotic proteins, including Bax, C‐Cyt c, and Cleaved‐Caspase‐3, was significantly reduced in the kidneys, while Bcl‐2 levels were markedly increased compared to the AKI group. Moreover, DNA fragmentation induced by Caspase‐activated DNase or ROS is a critical hallmark of apoptosis, leading to chromatin fragmentation and condensation, ultimately resulting in irreversible cell death. We then validated the anti‐apoptotic effect of HAND through TUNEL staining, which identifies and labels DNA strand breaks. As shown in Figure [168]5H,I, the number of TUNEL‐positive cells in the renal tubules of the AKI group was significantly increased compared to the Control group, with an apoptotic cell rate of 37%. Notably, HAND treatment significantly reduced the proportion of TUNEL‐positive cells, demonstrating superior anti‐apoptotic efficacy compared to CND without PTEC targeting. Subsequently, the anti‐apoptotic effect of HAND was further validated in HK‐2 cells through Annexin V‐FITC/PI flow cytometry. Annexin V specifically binds to phosphatidylserines, which translocate to the outer leaflet of the cell membrane during early apoptosis, while propidium iodide (PI) penetrates only late apoptotic cells with compromised membranes to bind to DNA. Under H[2]O[2] stimulation, HK‐2 cells exhibited a high apoptosis rate, with the proportion of early and late apoptotic cells reaching 49.2%, which was significantly reduced to 14.9% following HAND treatment (Figure [169]5J; Figure [170]S34, Supporting Information). Additionally, WB analysis confirmed that HAND effectively down‐regulated the expression of pro‐apoptotic proteins in HK‐2 cells induced by H[2]O[2] (Figure [171]S35, Supporting Information). Collectively, these findings demonstrated the superior anti‐apoptotic protective effects of HAND in both in vivo and in vitro models. Figure 5. Figure 5 [172]Open in a new tab HAND effectively inhibited intrinsic apoptosis. A) Schematic illustration of HAND inhibiting intrinsic apoptosis. B–G) Western blot analysis of apoptosis‐related protein expression levels in mouse kidney tissues from different treatment groups and grayscale analysis of Bax (C), Bcl‐2 (D), Caspase‐3 (E), Cleaved‐Caspase 3 (F), and C‐Cyt c (G). (H‐I) TUNEL fluorescence staining results (H) and quantification of TUNEL‐positive cells (I) in mouse kidneys from different treatment groups. J) Annexin‐V/PI flow cytometry results in HK‐2 cells from different treatment groups. Data are presented as mean ± SD. One‐way ANOVA followed by the SNK test was used for analysis. n = 3, ^* p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001. 2.7. HAND‐Inhibited cGAS‐STING‐Mediated Sterile Inflammation Excessive and unresolved sterile inflammation during AKI exacerbates tissue damage and accelerates the progression of renal fibrosis.^[ [173]^40 ^] Because in the absence of pathogens, the indiscriminate attacks by immune cells are mainly directed against endogenous tissues.^[ [174]^41 ^] Notably, mitochondria and the nucleus, beyond their central role in mediating cell death during AKI, also serve as key regulators of the inflammatory response, significantly influencing inflammatory signaling through the release of damaged DNA. The precise targeting capability of HAND to mitochondria and the nucleus, along with its potent mtROS‐scavenging efficacy, offers HAND a significant advantage in modulating inflammation during AKI (Figure [175]6A). The activation of the cGAS‐STING DNA‐sensing pathway has been increasingly recognized as a contributor to AKI‐related sterile inflammation, initiating a cascade of biological effects by activating innate immune responses. cGAS, a cytosolic DNA sensor, detects double‐stranded DNA (dsDNA) and catalyzes the synthesis of the second messenger cGAMP. Thus, the aberrant presence of cytosolic dsDNA is amplified by cGAMP and transmitted to the ER membrane receptor STING. This interaction induces conformational changes in STING, triggering downstream signaling cascades, including the activation of IRF3 and the phosphorylation and nuclear translocation of NF‐κB.^[ [176]^11 ^] In eukaryotic cells, the nucleus and mitochondria are the exclusive compartments for DNA, both of which undergo intense oxidative stress during AKI, leading to the production of substantial oxidative‐DNA fragments. Figure 6. Figure 6 [177]Open in a new tab HAND inhibited cGAS‐STING mediated sterile inflammation. A) Schematic illustration of HAND inhibiting cGAS‐STING mediated sterile inflammation. B) Fluorescence staining results of dsDNA (green), TOM20 (red), and DAPI (blue) in HK‐2 cells from different treatment groups. C) WB analysis of cGAS‐STING related protein expression in HK‐2 cells. D) Fluorescence staining results of dsDNA (green), TOM20 (red), and DAPI (blue) in kidneys from different treatment groups. E) Levels of 2′‐3′cGAMP in kidneys from different treatment groups. F) IHC results of cGAS and STING in kidneys from different treatment groups. G–J) Levels of chemokines CXCL1 (G), CCL2 (H), and inflammatory cytokines TNF‐α (I), and IL‐6 (J) in kidneys from different treatment groups. K) IHC results of F4/80 and Ly‐6G in kidneys from different treatment groups. Data are presented as mean ± SD. One‐way ANOVA followed by the SNK test was used for analysis. n = 3, ^* p < 0.05, ^** p < 0.01, ^*** p < 0.001, ^**** p < 0.0001. As shown in Figure [178]6B, H[2]O[2] stimulation significantly reduced the colocalization of dsDNA with the nucleus and mitochondria in HK‐2 cells. Further quantitative results showed that the level of mtDNA in the cytoplasm of the H[2]O[2] group increased significantly, while the mtDNA level in mitochondria decreased, which proved that the contents were released due to membrane rupture during mitochondrial injury (Figure [179]S36, Supporting Information). The released dsDNA then strongly sensitized the cGAS/STING pathway, evidenced by the significant upregulation of cGAS and STING in HK‐2 cells, as well as the activation of downstream IRF3 and phosphorylation of NF‐κB (Figure [180]6C). Notably, HAND treatment effectively mitigated dsDNA leakage and subsequent cGAS‐STING activation by mitigating DNA oxidative damage (Figure [181]6B,C; Figures [182]S36,S37, Supporting Information). We further validated the modulation of HAND on cGAS‐STING‐mediated inflammatory response in AKI mice. PTECs play a central role in AKI‐induced damage, serving as the initial site of injury that propagates signals to induce an inflammatory response. As shown in Figure [183]6D, the colocalization of dsDNA with TOM20 and DAPI in the renal cortex was significantly reduced in the AKI group, with dsDNA dispersed into the extracellular space. cGAS was then highly sensitized, generating a large amount of second messenger to activate STING, as evidenced by the high levels of 2′‐3′ cGAMP in renal tissue (Figure [184]6E) and the elevated expression of cGAS and STING in damaged proximal tubules (Figure [185]6F). Correspondingly, downstream pro‐inflammatory signals of the cGAS/STING pathway were activated (Figure [186]S38, Supporting Information), mediating the expression and secretion of various inflammatory cytokines (such as TNF‐α, IL‐1β, IL‐6) and chemokines (CXCL1, CXCL2, CCL2) (Figure [187]6G–J; Figure [188]S39, Supporting Information). Among these, CXCL1 and CXCL2 primarily recruit neutrophils, while CCL2 mainly recruits macrophages and other monocytes. As a result, in contrast to the small number of resident immune cells in the Control group, the AKI group displayed extensive infiltration of neutrophils and macrophages in the renal tubular interstitium. These recruited immune cells further released inflammatory mediators such as cytokines, chemokines, ROS, and proteases, perpetuating a vicious cycle that fosters chronic inflammation. Such persistent and extensive damage to PTECs would ultimately deplete the functional epithelium, resulting in the formation of scar tissue formation and irreversible fibrosis. Fortunately, the potent ROS‐scavenging ability of HAND effectively disrupted the vicious cycle of oxidative stress, inflammation, and tissue damage, as evidenced by the significantly reduced levels of inflammatory mediators and decreased immune cell infiltration in the HAND group, demonstrating superior immunomodulatory effects compared to CND (Figure [189]6K). Furthermore, to substantiate these findings, we employed a conditioned medium model to investigate the interactions between injured PTECs and immune cells under oxidative stress. HK‐2 cells were first treated with H[2]O[2] to induce oxidative damage and activation of the cGAS‐STING pathway, after which the conditioned medium from these HK‐2 cells was collected and used to stimulate RAW 264.7 macrophages (Figure [190]S40 (A), Supporting Information). In this system, macrophages exhibited significant activation, as evidenced by the upregulation of COX‐2 and iNOS, along with elevated levels of pro‐inflammatory cytokines (TNF‐α, IL‐1β, IL‐6), which further exacerbated inflammation. However, HAND treatment to H[2]O[2]‐stimulated HK‐2 cells resulted in a marked reduction in COX‐2 and iNOS expression in RAW 264.7 macrophages, as well as significantly lower levels of pro‐inflammatory cytokines compared to the CND group (Figure [191]S40 (B), Supporting Information). The enhanced anti‐inflammatory effect is primarily attributed to HAND's ability to protect HK‐2 cells from oxidative damage, underscoring the therapeutic advantage of HAND in mitigating oxidative stress‐induced inflammation by preserving cellular integrity and minimizing damage‐associated signals that drive excessive immune responses. Taken together, the above findings suggested that HAND could successfully halt the pathological progression of AKI by precisely eliminating the source of ROS and inhibiting the cGAS‐STING‐mediated sterile inflammation. 2.8. HAND Exhibited Excellent Biocompatibility Finally, we evaluated the biocompatibility of HAND in vivo (Figure [192]7A,B). Following the administration of a high dosage (10 mg kg^−1, 5 times the therapeutic dose) for 24 h, the lungs, liver, heart, spleen, and kidneys were harvested for HE staining. Compared with the PBS group, HAND did not cause any damage to these vital organs (Figure [193]7C). Additionally, hematological parameters, including white blood cells, red blood cells, and platelets, showed no significant differences compared to the PBS group (Figure [194]7D; Figure [195]S41, Supporting Information). Liver function indicators (AST, ALT) (Figure [196]7E), kidney function markers (BUN, CRE) (Figure [197]7F), and inflammatory cytokine levels (TNF‐α, IL‐6, IL‐1β) (Figure [198]7G) also remained stable, indicating that HAND did not adversely affect liver or kidney function and did not provoke an inflammatory response. Moreover, during long‐term administration of HAND in mice (therapeutic dosage once a week for 4 weeks), no significant changes were observed in major organ histology (Figure [199]7C), hematological parameters (Figure [200]7H; Figure [201]S42, Supporting Information), liver and kidney function (Figure [202]7I,J), or inflammatory markers (Figure [203]7K). These results collectively demonstrated the excellent biosafety of the naturally derived HAND. Figure 7. Figure 7 [204]Open in a new tab HAND exhibited excellent biocompatibility. A,B) Schematic illustration of short‐term and long‐term treatment protocol for biosafety evaluation of HAND. C) Representative H&E‐stained images of major organs from KM mice after short‐term (left) and long‐term (right) administration of HAND or 1× PBS. D–G) Hematological parameters (D), liver function indicators (E), kidney function indicators (F), and inflammatory cytokine levels (G) of short‐term treated mice in different treatment groups. H–K) Hematological parameters (H), liver function indicators (I), kidney function indicators (J), and inflammatory cytokine levels (K) of long‐term treated mice in different treatment groups. Data are presented as mean ± SD. n = 6. 3. Discussion Over the past two decades, the proportion of deaths attributed to kidney disease has steadily increased, with its global prevalence surpassing that of any other major non‐communicable diseases, including cardiovascular diseases, cancer, chronic respiratory diseases, and diabetes.^[ [205]^42 ^] Patients with kidney disease are at high risk for comorbidities that contribute not only to elevated mortality rates but also to a significant symptom burden and substantial treatment costs.^[ [206]^43 ^] Given the urgent clinical demand, the development of therapeutics for kidney diseases holds significant promise but also faces substantial challenges. Recent advancements in genetics, bioinformatics, and pharmacodynamics have facilitated deeper exploration into g the pathophysiology of AKI and the development of new drugs. However, the diverse etiologies and heterogeneous pathological processes of AKI complicate efforts to create effective treatments targeting gene regulation or specific downstream molecules, which may fail to rapidly halt acute damage progression. Because the oxidative stress‐PTECs injury‐inflammation vicious cycle is a key factor in almost all types of AKI, which interferes with numerous cellular processes and spreads rapidly like a snowball to cause a swift decline in renal function.^[ [207]^16 ^] This cycle begins with the excessive production of ROS in response to various stressors, leading to oxidative stress that directly damages PTECs. Once injured, PTECs release pro‐inflammatory cytokines that intensify the inflammatory response and attract immune cells to the injury site. The influx of these inflammatory cells, in turn, generates additional ROS, creating a feedback loop that perpetuates cellular damage and inflammation. As the cycle continues, impairs vital cellular functions, including mitochondrial activity and signaling pathways, ultimately resulting in rapid renal function decline. Moreover, the increasing emphasis on precision medicine underscores the need for more organized, rational, and high‐resolution targeting strategies.^[ [208]^44 ^] Non‐selective drug distribution often necessitates higher therapeutic doses and leads to unavoidable off‐target toxic side effects.^[ [209]^45 ^] These challenges significantly complicate the development of AKI therapies, highlighting the urgent need for more precise and efficient treatment strategies. In this context, we specifically target the primary source of injury in AKI—excessive toxic ROS—and designed an FA‐optimized nanomedicine, HAND, that aligns with molecular preferences for targeted tissues