Graphical abstract graphic file with name fx1.jpg [31]Open in a new tab Highlights * • High expression of ANP32E is associated with the progression of AKI * • The deficiency of ANP32E alleviates the damage to renal tubular epithelial cells * • Inhibiting ANP32E plays a protective role in AKI through the AMPK pathway __________________________________________________________________ Molecular biology; Cell biology Introduction Acute kidney injury (AKI) is a clinical syndrome characterized by a rapid decrease in kidney function (within 48 h) induced by injury to renal structure or function caused by a variety of factors, such as renal hypoperfusion, trauma, sepsis, and toxic drugs.[32]^1^,[33]^2^,[34]^3 Kidney ischemia-reperfusion (I/R) damage is a frequent cause of AKI[35]^4 and a primary trigger of kidney transplant dysfunction.[36]^5 Despite tremendous efforts in examining the mechanisms underlying the pathophysiology of AKI, there are no valid therapeutic measures for treating or preventing AKI.[37]^6 Therefore, identifying the mechanism of AKI is critical for pharmaceutical discovery and improving the quality of life of patients with acute kidney damage. I/R injury is a significant cause of AKI and is associated with increased morbidity, mortality, and extended hospitalization.[38]^7^,[39]^8 This condition is characterized by unusual regional tissue function after the abrupt interruption of the blood supply to an organ or a large area of the body, resulting in severe tissue injury after blood flow restoration and reoxygenation. Acute ischemia results in ATP depletion, tubular epithelial damage, and hypoxic cell death. Reperfusion further aggravates injury by increasing the generation of reactive oxygen species and inducing leukocyte activation, infiltration, and inflammation.[40]^9^,[41]^10^,[42]^11^,[43]^12 I/R-induced organ or tissue injury can occur in other medical conditions, such as myocardial infarction,[44]^13^,[45]^14 chemic stroke,[46]^15 and organ transplantation.[47]^16^,[48]^17^,[49]^18 Acidic nuclear phosphoprotein 32 family member E (ANP32E) is a member of the histone chaperone family with leucine-rich repeats that removes the histone variant H2A.Z from chromatin. These proteins play significant roles in numerous cellular processes, such as chromatin remodeling and modification, the regulation of gene expression, protein phosphorylation, and intracellular transport regulation.[50]^19^,[51]^20^,[52]^21 ANP32E was first identified in the cerebellum as a member of the ANP32E family of protein phosphatase 2 (PP2A) inhibitors,[53]^19 where it regulated cerebellar synaptogenesis by inhibiting PP2A activity at synapses. AMP-activated protein kinase (AMPK) activation can facilitate autophagy, whereas mTOR activation can repress autophagy. Since mTOR modulates autophagy negatively, downregulation of mTOR by AMPK is an important regulatory mechanism that maintains cell survival in the context of cellular energy depletion. Phosphorylated AMPK can directly suppress the phosphorylation of mTORC1 and promote autophagy. In this study, we investigated ANP32E accumulation in AKI and clarified its correlation with ANP32E. Alleviating apoptosis and inflammation by silencing ANP32E suppressed the progression of AKI by promoting the AMPK/autophagy pathway. Results Identification of DEGs in mice subjected to I/R and validation of ANP32E in different kidney cell types and ATN patient specimens Single-cell RNA sequencing data from healthy human organs in the Human Protein Atlas ([54]http://www.proteinatlas.org/) showed that ANP32E was present in all the tissues with low tissue specificity ([55]Figure 1A), and the single-cell resolution data indicated that the expression of ANP32E was higher in tubular cells than in other cells ([56]Figure 1B). The translating ribosome affinity purification sequencing (TRAP-seq) validation dataset [57]GSE192532 (adj: p < 0.05) confirmed the increased expression of ANP32E in kidney after I/R injury ([58]Figure 1C). We then collected renal biopsy tissues from patients with acute tubular necrosis (ATN). Paracancerous tissues from patients who underwent nephrectomy were used as controls. H&E staining revealed the loss of epithelial cell brush borders of tubules, necrosis, tubular dilatation, and cast deposition in the ATN group compared to the control group ([59]Figures 1D and 1E). TUNEL staining showed that the cells underwent apoptosis ([60]Figures 1F and 1G). We used immunohistochemical staining to confirm the upregulation of ANP32E in renal tissue from patients with ATN compared to that in paracancerous renal tissue obtained from patients without other renal diseases who underwent nephrectomy ([61]Figures 1H and 1I). Figure 1. [62]Figure 1 [63]Open in a new tab Bioinformatics analysis of ANP32E expression in different renal cells and patients with ATN (A and B) The average expression level of ANP32E in human organs and single-cell type data of kidney samples in the Human Protein Atlas. (C) The expression level of ANP32E after renal I/R injury in the TRAP-seq validation dataset ([64]GSE192532 ). (D) Representative images of H&E staining. Bar, 50 μm. (E) Pathological score indicating tubular damage. (F) TUNEL staining of apoptosis-positive cells. Bar, 50 μm. (G) Quantification of TUNEL-positive cells in the two groups. (H) Immunohistochemical staining showing the expression of ANP32E in patients with ATN. Bar, 50 μm. (I) Statistical charts quantifying the immunohistochemical staining of ANP32E. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. ANP32E expression is upregulated and positively correlated with renal injury We constructed a bilateral renal I/R mouse model. Serum creatinine ([65]Figure 2A) and urea nitrogen ([66]Figure 2B) were used to assess the degree of kidney damage; renal structure was analyzed by H&E staining and revealed tubular deposition and tubular dilation after I/R ([67]Figures 2C and 2D). TUNEL staining indicated that apoptosis occurred in the model ([68]Figures 2E and 2F). We further confirmed the upregulation of ANP32E in the kidneys of mice with renal I/R by quantitative reverse-transcription PCR (RT-qPCR) ([69]Figure 2G), western blotting ([70]Figures 2H–2J), and immunohistochemical analysis ([71]Figures 2K and 2L). To determine the tubular segment specificity of ANP32E expression in the kidney, we performed dual immunostaining for ANP32E (red) and proximal tubular markers (green) in the kidney ([72]Figures 2M and 2N). ANP32E was expressed in proximal tubules after kidney I/R. Figure 2. [73]Figure 2 [74]Open in a new tab ANP32E is upregulated in the kidneys of mice with renal I/R and is positively correlated with kidney injury (A) Scr concentrations in the two groups. (B) BUN levels in the two groups. (C) Representative images of hematoxylin and eosin (H&E) staining. Bar, 50 μm. (D) Pathological score indicating tubular damage. (E and F) Representative images of TUNEL staining and the quantification of TUNEL-positive cells in the two groups. Bar, 50 μm. (G) The mRNA levels of ANP32E in the two groups. (H–J) Western blotting and statistical analysis of the expression of ANP32E and NGAL in the two groups. (K and L) Representative images and statistical analysis of immunohistochemical staining of ANP32E in the different groups. Bar, 50 μm. (M) Representative immunofluorescence staining of ANP32E in the renal proximal tubules in the sham group and the I/R group. Bar, 50 μm. See also [75]Figure S7. n = 6. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. ANP32E is upregulated in hypoxia/reoxygenation-stimulated HK-2 cells To further examine ANP32E expression in vitro, we established two models of AKI by treating HK-2 cells with hypoxia/reoxygenation and CoCl[2]. We first examined ANP32E protein expression in HK-2 cells exposed to different durations of reoxygenation following hypoxia ([76]Figures 3A–3C). RT-qPCR and immunofluorescence staining revealed that ANP32E expression levels were increased in the hypoxia/reoxygenation group compared to the control group ([77]Figures 3D and 3E). Subsequently, HK-2 cells were treated with increasing concentrations of CoCl[2] (100, 250, and 500 μM) for 24 h, and cells in the control group were not treated with any CoCl[2]. Western blotting ([78]Figures 3F–3H), RT-qPCR ([79]Figure 3I), and immunofluorescence analysis ([80]Figure 3J) demonstrated that the expression of ANP32E in mRNA level and protein level was increased compared with the control group. Overexpression of ANP32E ([81]Figures 3K and [82]S2A) upregulated NGAL in HK-2 cells ([83]Figures 3L and 3M). Overexpression of ANP32E increased the overall percentage of cell death and the number of apoptotic and dead cells, as determined by flow cytometry ([84]Figures S2B and S2C). In addition, we found that the hypoxia/reoxygenation-induced increase in the levels of proinflammatory mediators was enhanced by the overexpression of ANP32E in HK-2 cells ([85]Figures S2D and S2E). Taken together, these results suggest that ANP32E may play an important role in facilitating AKI. Figure 3. [86]Figure 3 [87]Open in a new tab ANP32E is upregulated in hypoxia/reoxygenation-stimulated HK-2 cells and promotes damage (A–C) Western blotting and statistical analysis of the expression of ANP32E and NGAL in response to different reoxygenation durations after hypoxia and in the control group. (D) The mRNA levels of ANP32E in the four groups. (E) Representative immunofluorescence staining of ANP32E in HK-2 cells in the four groups. Bar, 50 μm. (F–H) Western blotting and statistical analysis of the expression of ANP32E and NGAL in cells treated with increasing concentrations of CoCl[2] for 24 h and in the control group. (I) The mRNA levels of ANP32E in the four groups. (J) Representative immunofluorescence staining of ANP32E in HK-2 cells in the four groups. Bar, 50 μm. (K) Western blotting analysis of ANP32E in HK-2 cells transfected with the empty vector or lentivirus plasmids. (L and M) Western blotting and densitometric analysis of NGAL in HK-2 cells transfected with the empty vector or lentivirus plasmids. See also [88]Figure S2. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Knockdown of ANP32E modulates apoptosis and inflammatory responses in I/R-induced AKI We examined whether knockdown of ANP32E protected against AKI induced by I/R. The experimental design is shown in [89]Figure 4A. ANP32E expression was inhibited by the lentivirus, and the efficiency of ANP32E inhibition was examined by western blotting ([90]Figures S1A and S1B). Next, we tested whether the inhibition of ANP32E could protect tubule cells from injury and restore some of the function of the kidney. As shown by H&E staining, there was no significant difference in morphology in the sham group. The renal tubular epithelial cells showed vacuolar degeneration and cell tube type formation and inflammatory cell infiltration when the mice undergo I/R. However, inhibiting ANP32E reduced tubular damage and reduced tube structure ([91]Figures 4D and 4E). Immunohistochemical staining of KIM-1, a tubular cell damage marker, showed that inhibiting ANP32E alleviated I/R-induced impairment of kidney function in AKI mice ([92]Figures 4F and 4G). Inhibition of ANP32E also alleviated the increase in serum creatinine and urea nitrogen concentrations ([93]Figures 4B and 4C). We next examined cellular apoptosis. Consistent with our hypothesis, in I/R-induced mice, LV-Anp32e abrogated the increase in the expression of cleaved caspase-3, which is a key marker of apoptosis ([94]Figures 4H–4J). TUNEL staining also showed that LV-ANP32E blocked apoptosis ([95]Figures 4K and 4L). Furthermore, RT-qPCR was used to measure tumor necrosis factor alpha (TNF-α) and interleukin (IL)-1β levels ([96]Figures 4M and 4N), and the results indicated that LV-Anp32e reduced the inflammatory response in the kidneys of mice subjected to I/R. Figure 4. [97]Figure 4 [98]Open in a new tab Knockdown of ANP32E ameliorates renal I/R injury in mice (A) A schematic of the experimental design. (B and C) Scr and BUN levels in the four groups. (D) Representative images of hematoxylin and eosin (H&E) staining. Bar, 50 μm. (E) Pathological score indicating tubular damage. (F and G) Representative images and statistical analysis of immunohistochemical staining of KIM-1 in the different groups. Bar, 50 μm. (H–J) Western blotting and statistical analysis of the expression of cleaved caspase-3 and NGAL in the four groups. (K and L) Representative images of TUNEL staining and quantification of TUNEL-positive cells. Bar, 50 μm. (M and N) The mRNA levels of TNF-α and IL-1β in the four groups. See also [99]Figures S1 and [100]S7. n = 6. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Silencing ANP32E ameliorates apoptosis and inflammatory responses in vitro In vitro, we successfully constructed ANP32E-targeted siRNA sequences ([101]Figures 5A–5C). Silencing ANP32E reduced the protein levels of cleaved caspase-3, which is a major target of apoptosis ([102]Figures 5D and 5E). ANP32E protein and mRNA expression levels were decreased after treatment with ANP32E-targeted siRNA sequences ([103]Figures 5F and 5G). The results also indicated that the overall percentage of cell death and the number of apoptotic and dead HK-2 cells were reduced by ANP32E silencing under hypoxia/reoxygenation conditions, as determined by flow cytometry ([104]Figures 5H and 5I). Silencing ANP32E inhibited the hypoxia/reoxygenation-induced increases in the levels of proinflammatory mediators ([105]Figures 5J and 5K). Figure 5. [106]Figure 5 [107]Open in a new tab Silencing ANP32E alleviates disease progression by affecting hypoxia/reoxygenation-induced inflammation and apoptosis in HK-2 cells (A–C) Western blotting, statistical analysis, and RT-qPCR were used to examine the silencing efficiency of ANP32E. (D–F) The protein levels of ANP32E and cleaved caspase-3 in the six groups. (G) The mRNA levels of ANP32E in the six groups. (H and I) Flow cytometric analysis of cell apoptosis and death. (J and K) The influence of ANP32E on the mRNA levels of proinflammatory mediators in HK-2 cells exposed to hypoxia/reoxygenation conditions. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Knockdown of ANP32E promotes autophagy during I/R-induced AKI We then examined how ANP32E controls AKI progression. The results of our analysis are summarized in volcano plots, which included 547 genes that were upregulated and 323 genes that were downregulated following ANP32E silencing in patients with AKI ([108]Figure 6A). Gene Ontology analysis of the differentially expressed genes (DEGs) revealed functions, bioprocesses, and expression levels ([109]Figure 6B). KEGG (Kyoto Encyclopedia of Genes and Genomes) and heatmap enrichment analysis revealed that autophagy and AMPK pathway were enriched in the siRNA-ANP32E + AKI group compared to the NC + AKI group ([110]Figures 6C and 6D). Western blotting analysis revealed that knockdown of ANP32E increased the protein levels of Beclin 1 and decreased the expression of P62 in vivo ([111]Figures 6E–6G), which was further confirmed by immunofluorescence and immunohistochemical analyses of LC3 ([112]Figures 6H–6J). Figure 6. [113]Figure 6 [114]Open in a new tab Knockdown of ANP32E promotes autophagy in AKI induced by I/R (A) Volcano plots showing the DEGs (DEGs: |log2FC|>1, adjusted p < 0.05). (B) Loopcircos of the DEGs. (C) Heatmap of molecules related to autophagy. (D) KEGG enrichment analysis of the AMPK signaling pathway. (E–G) Western blotting and statistical analysis of the expression of Beclin 1 and P62 in the four groups. (H and I) Representative images and statistical analysis of immunohistochemical staining of LC3 in the different groups. Bar, 50 μm. (J) Representative immunofluorescence staining of LC3 in vivo. Bar, 50 μm. See also [115]Figures S4 and [116]S7. n = 6. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Silencing ANP32E promotes AMPK pathway activity After confirming that ANP32E affected the progression of AKI by inhibiting autophagy in vivo, we examined whether this effect also occurred in vitro. Western blotting analysis revealed decreased protein levels of Beclin 1 and decreased expression of P62 in HK-2 cells ([117]Figures 7A–7C), which was further confirmed by immunofluorescence analyses of LC3 ([118]Figure 7D). The RNA sequencing (RNA-seq) data indicated that the AMPK pathway was affected by ANP32E silencing in the AKI cell model. Western blotting analysis revealed a marked change of key molecules of the AMPK pathway, such as P-AMPK and P-mTOR ([119]Figures 7E–7G). Then, we tested AMPK pathway-related molecules in vivo; the results showed that the expression of P-AMPK was increased after ANP32E knockdown in the I/R group, while the expression of P-mTOR was further reduced ([120]Figures S4A–S4C), indicating that ANP32E knockdown could exert a renal protective effect by activating the AMPK signaling pathway. Pathway activation was decreased after the AMPK inhibitor compound C (CC) was used to inhibit AMPK in the AKI model ([121]Figures 7H and 7I). The autophagic flux showed an increase of RFP in hypoxia/reoxygenation-induced HK-2 cells. The signal intensity was increased in HK-2 cells transfected with si-ANP32E compared to the hypoxia/reoxygenation group, while exposure to the AMPK inhibitor CC reduced autophagy ([122]Figure S5A). These results indicate that silencing ANP32E promotes AMPK pathway activity. Subsequently, we used mTOR inhibition to stimulate HK-2 cells after the overexpression of ANP32E. The results indicated that the addition of rapamycin reduces the expression level of the NGAL ([123]Figures S6A and S6B). This suggests that the highly expression of ANP32E can partly damage renal tubular epithelial cells through mTOR. Figure 7. [124]Figure 7 [125]Open in a new tab Silencing ANP32E promotes HK-2 cell AMPK activity and autophagy in vitro (A–C) The protein levels and statistical analysis of Beclin 1 and P62 in the four groups. (D) Representative images and statistical analysis of immunofluorescence staining of LC3. Bar, 50 μm. (E–G) Western blotting images and corresponding quantification about the molecules of the AMPK pathway. (H) Western blotting images and corresponding quantification in HK-2 cells treated with an AMPK inhibitor. See also [126]Figures S3, [127]S5, and [128]S6. ∗p < 0.05, ∗∗p < 0.01, ∗∗∗p < 0.001, and ∗∗∗∗p < 0.0001. Briefly, our study has established how ANP32E affects AKI. Downregulation of ANP32E can activate AMPK according to the RNA-seq. When AMPK is activated, it can inhibit the phosphorylation of mTOR, leading to the lipidation of LC3 and activation of Beclin 1.[129]^22 Autophagy suppresses apoptosis by reducing the abundance of pro-apoptotic proteins in the cytoplasm and regulates NLRP3 inflammasome activation as well as IL-1β and IL-18 secretion negatively[130]^23 through the Toll-like receptor and Nod-like receptor signaling pathway ([131]Figure 8). Figure 8. [132]Figure 8 [133]Open in a new tab The mechanism described in this study In this study, I/R increased the expression of ANP32E in renal tubular epithelial cells, and ANP32E mediated the development of kidney inflammation and promoted cell apoptosis, which was associated with the regulation of the AMPK/autophagy pathway. Discussion Among innate renal cells and infiltrating cells, renal tubular epithelial cells are critical contributors to the development of AKI and are the most susceptible to kidney ischemia.[134]^24 Pathological analysis reveals the loss of tubular epithelial cells by apoptosis and necrosis, and renal tubular epithelial cell death and injury are significant causes of kidney dysfunction.[135]^25 DNA damage has been reported to be involved in I/R injury in a variety of vital organs, including the testis, brain, liver, and heart.[136]^26^,[137]^27^,[138]^28^,[139]^29 During renal I/R injury, DNA fragmentation and damage occurs as early as 12 h after reperfusion in the renal tubules and increases within 24 h.[140]^30 The present study used in vivo and in vitro models of ischemic injury and examined the underlying molecular mechanism. Autophagy is an evolutionarily conserved multistep process involving the degradation of intracellular organelles, proteins, and other macromolecules by lysosomal hydrolases.[141]^31^,[142]^32^,[143]^33 It has many features, including the regulation of programmed cell death, inflammation, and other biological processes.[144]^34 Pharmacological and genetic inhibition studies have indicated that autophagy plays a nephroprotective role in AKI.[145]^35 The occurrence of cellular autophagy requires the involvement of multiple proteins. These protein interactions regulate the occurrence of autophagy. In this process, the post-translational modification behaviors of proteins such as phosphorylation, acetylation, ubiquitylation, and succinylation play an important role in regulating the process of autophagy.[146]^36 A study of histone lactate showed that histone lactation promotes the transcription of rubicon-like autophagy enhancer, which promotes autophagosome maturation through interaction with Beclin 1. Inhibition of histone milk acidification has a therapeutic efficiency comparable to that of autophagy inhibitors.[147]^37 Studies have shown that methylation of histone H3K4 regulates the expression and activity of two key transcription factors HLH-30 and PHA-4,[148]^38 while the two molecules can regulate autophagy by controlling the expression of many autophagy-related genes.[149]^39^,[150]^40 Moreover, histone acetylation also affects a variety of cellular and physiological processes, including transcription, neural function, autophagy, and differentiation.[151]^41 However, autophagy is a double-edged sword. In general, it can maintain normal cellular homeostasis. However, when the stimulation is too strong, autophagy can play the opposite role.[152]^42 AMPK is an energy-sensitive kinase that is involved in regulating bioenergetic metabolism.[153]^43 Under conditions of AKI, the expression of phosphorylated AMPK is significantly increased in kidney tissue, and activated AMPK can promote autophagy by inhibiting the mTOR signaling pathway.[154]^44^,[155]^45 In our study, we found that silencing ANP32E inhibited inflammation and apoptosis during AKI progression by further activating the AMPK/autophagy pathway. ANP32E is a protein with leucine-rich repeats that functions as a chaperone of H2A.Z histone and regulates the expression of downstream genes[156]^46 and other cellular processes, including chromatin modification and remodeling, gene regulation, protein phosphorylation, and cell apoptosis.[157]^47 It has been reported that ANP32E is not only essential for cerebellar development and synaptogenesis but is also involved in tumorigenesis. In triple-negative breast cancer, ANP32E induces tumorigenesis by increasing the expression of the transcription factor E2F1.[158]^48 ANP32E activates AKT/mTOR/HK2 signaling and glycolysis in thyroid cancer.[159]^49 In gastric cancer, overexpression of ANP32E can cause abnormal NUF2 expression, which promotes cancer progression.[160]^50 We assessed ANP32E expression in the GEO dataset and found increased expression of ANP32E in I/R-induced AKI. Our results suggested that the ANP32E protein was upregulated in the renal tubular cells of patients with ATN and I/R mice and in cultured proximal tubular cells exposed to hypoxia/reoxygenation. We examined the function and mechanism of ANP32E by conducting a series of in vivo and in vitro experiments. The results indicated that ANP32E knockdown alleviated the inflammatory response and apoptosis in I/R-induced mice and hypoxia/reoxygenation-stimulated HK-2 cells. KEGG pathway enrichment analysis was used to screen the AMPK signaling pathway and showed that ANP32E could participate in AKI progression by regulating autophagy through the AMPK signaling pathway. Studies have shown that knockdown of ANP32E can reduce AKI damage by further activating the AMPK signaling pathway. However, the specific mechanism by which ANP32E regulates the AMPK signaling pathway remains unclear. Previous reports have suggested the colocalization of ANP32E with the catalytic subunit of PP2A and the inhibition of PP2A activity.[161]^51 PP2A plays an important role in protecting against renal injury,[162]^52 and the involvement of PP2A in renal I/R injury has been demonstrated.[163]^53 Renal tubular epithelial cells have extremely high energy requirements, and AMPK is a key regulator of energy production. AMPK activity can be regulated by PP2A.[164]^54 These theories suggest that ANP32E can regulate the AMPK pathway by regulating the localization of PP2A in the cytoplasm. However, this hypothesis requires further research. In summary, we found that silencing ANP32E further activated autophagy through the AMPK pathway and ameliorated I/R-induced AKI. These findings will provide new ideas and targets for the treatment of AKI. Limitations of the study Although we found that ANP32E participates in the progression of AKI, our study still has many limitations. The progression of injury in mice is very similar to the progression of human AKI injury, but the mouse model cannot represent the complexity of human kidney fully. Moreover, the pathogenesis of I/R-induced AKI is complex, involving the involvement and crosstalk of multiple links such as apoptosis, inflammation, and autophagy, and this study needs to further explore the molecular mechanism interaction of the aforementioned links. Resource availability Lead contact Further information and requests for resources and reagents should be directed to the lead contact, Xiangdong Yang (yxd@email.sdu.edu.cn). Materials availability This study did not generate new unique reagents. Antibodies, reagents, and cell lines used were obtained from commercial or other sources as outlined in the [165]key resources table. Data and code availability * • Data from this study can be provided by the [166]lead contact upon request. * • No code was generated or analyzed during the current study. * • Any additional information is available from the [167]lead contact upon request. Acknowledgments