key: cord-0935938-v00l2foz authors: Yang, Chen; Zhang, Yu; Zeng, Xia; Chen, Huijing; Chen, Yuchen; Yang, Dong; Shen, Ziwei; Wang, Xiaomu; Liu, Xinran; Xiong, Mingrui; Chen, Hong; Huang, Kun title: Kidney injury molecule-1 is a potential receptor for SARS-CoV-2 date: 2021-01-25 journal: J Mol Cell Biol DOI: 10.1093/jmcb/mjab003 sha: 0921170aafa6073ffa28c1a74c6f99907f872132 doc_id: 935938 cord_uid: v00l2foz COVID-19 patients present high incidence of kidney abnormalities, which are associated with poor prognosis and mortality. The identification of SARS-CoV-2 in the kidney of COVID-19 patients suggests renal tropism of SARS-CoV-2. However, whether there is a specific target of SARS-CoV-2 in the kidney remains unclear. Herein, by using in silico simulation, co-immunoprecipitation, fluorescence resonance energy transfer, fluorescein isothiocyanate labelling, and rational design of antagonist peptides, we demonstrate that kidney injury molecule-1 (KIM1), a molecule dramatically upregulated upon kidney injury, binds with the receptor-binding domain of SARS-CoV-2 and facilitates its attachment to cell membrane, with the immunoglobulin variable Ig-like (Ig V) domain of KIM1 playing a key role in this recognition. The interaction between SARS-CoV-2 receptor-binding domain and KIM1 is potently blockaded by a rationally designed KIM1-derived polypeptide AP2. In addition, our results also suggest interactions between KIM1 Ig V domain and the receptor-binding domains of SARS-CoV and MERS-CoV, pathogens of two severe infectious respiratory diseases. Together, these findings suggest KIM1 as a novel receptor for SARS-CoV-2 and other coronaviruses. We propose that KIM1 may thus mediate and exacerbate the renal infection of SARS-CoV-2 in a ‘vicious cycle’, and KIM1 could be further explored as a therapeutic target. The World Health Organization has announced the coronavirus disease 2019 (COVID-19) as a pandemic (Guan et al., 2020) . Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the pathogen of COVID-19, belongs to the beta-coronavirus genus that also includes SARS-CoV and Middle East respiratory syndrome coronavirus (MERS-CoV) (Shahrajabian et al., 2020) . SARS-CoV-2, SARS-CoV, and MERS-CoV mainly target respiratory systems to primarily manifest with respiratory illness (Shahrajabian et al., 2020) . Notably, reports about renal involvement among patients infected, as well as identification of viral infection in the kidney suggested that these coronaviruses may directly infect the kidney (Ding et al., 2004; Eckerle et al., 2013; Braun et al., 2020; Su et al., 2020) . Kidney impairment in hospitalized COVID-19 patients is common, and we and others have reported its association with severe inflammation, poor clinical progress, and high in-hospital mortality (Pei et al., 2020; Hirsch et al., 2020; Chen et al., 2020; Yang et al., 2020) . High incidence of acute kidney injury (AKI) (56.9%) among patients with COVID-19 has been observed (Fisher et al., 2020) . Importantly, the presence of infective SARS-CoV-2 has been confirmed in the kidney, especially in renal epithelial cells; and a post-mortem study suggested the renal tropism of SARS-CoV-2, which was detected in the kidneys of 72% of COVID-19 patients with AKI (Braun et al., 2020) . Among multi-organ manifestations in COVID-19 patients, apart from the lung, the kidney is highly vulnerable to the virus, and renal dysfunctions are closely associated with high mortality, with the underlying molecular mechanisms remaining unclear. SARS-CoV-2 invasion initiates from binding with cellular membrane receptors via its spike protein (Shahrajabian et al., 2020) . Presently, angiotensin-converting enzyme 2 (ACE2), which is enriched in the kidney and also the target for SARS-CoV, is the only well-recognized receptor for SARS-CoV-2 (Shang et al., 2020) . Responsible for receptor recognition, SARS-CoV-2 spike protein (SARS-CoV-2-S) consists of subunits S1 and S2 ( Figure 1A ), and the receptor-binding domain (RBD) in S1 binds ACE2 to initiate the fusion of S2 with cell membrane and subsequent cell entry (Shang et al., 2020) . Although comparison on the level of ACE2 between SARS-CoV-2 infected and normal kidneys has not been reported, decreased level of ACE2 upon coronavirus infection in lung has been observed , therefore, the renal tropism of SARS-CoV-2 and associated kidney injury seem unlikely associate with the level of ACE2. In addition, we recently reported that the administration of ACE2 inhibitors showed no association with clinical outcomes among COVID-19 patients . Given that viral cell entry may involve multiple transmembrane receptors (Lan et al., 2020) , we speculate that additional receptors may mediate the renal infection of SARS-CoV-2. Kidney injury molecule-1 (KIM1) is primary expressed in kidney and drastically up-regulated in injured kidney proximal tubule upon injury, and plays crucial roles in inflammation infiltration and immune responses (Rong et al., 2011) . Structurally, KIM1 consists of immunoglobulin variable Ig-like (Ig V) domain, mucin domain, transmembrane domain and cytosolic domain. Among them, Ig V domain is required for virus binding and internalization, such as the entry of Ebola and Dengue viruses (Yuan et al., 2015; Dejarnac et al., 2018) . Here, we investigated whether KIM1 is a binding target of SARS-CoV-2 that mediates its kidney invasion. To elucidate KIM1 and ACE2 enrichment in tissues, the transcriptome and histology-based protein expression data from the Tissue Atlas of Human Protein Atlas were collected. The top 10 tissues with mRNA and protein abundance of KIM1 and ACE are listed in Supplementary Figure S1A-F. Notably, KIM1 and ACE2 co-expressed in the kidney, colon, rectum, testis, and gallbladder (Supplementary Figure S1C and F), which are all among the target organs of SARS-CoV-2 (Cha et al., 2020) , implicating a close correlation of KIM1 with COVID-19 manifestations. The primary sequences of SARS-CoV-RBD and SARS-CoV-2-RBD share high similarity (62.93%), with 9 of 14 ACE2-contacting residues conserved in both RBDs ( Figure 1B ). In comparison, MERS-CoV-RBD, which recognizes DPP4 (Li et al., 2020c) , shows low similarity with SARS-CoV-RBD and SARS-CoV-2-RBD (17.07% and 14.86%, respectively; Figure 1B ). Kim1 is drastically upregulated in the kidneys of ischemia-reperfusion (I/R)-or cisplatin-injured mice, while only mild changes of Ace2 were observed ( Figure 1C ). Among four domains of KIM1 ( Figure 1D and E), Ig V domain is responsible for virus binding (Yuan et al., 2015; Dejarnac et al., 2018) , and molecular dynamic docking was thus conducted to investigate its binding with SARS-CoV-2-RBD. Docking and structural information of SARS-CoV-2-RBD and KIM1 Ig V complex are provided in Supplementary Figures S1G-I and S2. Phe338, Val367, Ser371, Phe374, and Trp436 of SARS-CoV-2-RBD contact Leu54, Phe55, Gln58, Trp112, and Phe113 of KIM1 Ig V ( Figure 2A) and lead to a combined binding free energy of -35.64 kcal/mol (Table 1; Supplementary Table S1) , which is lower than that of SARS-CoV-2-RBD and ACE2 (-50.60 kcal/mol) but comparable to that of SARS-CoV-RBD and ACE2 (-38.3 kcal/mol) (Li et al., 2020c) . Since ACE2 is recognized as a key receptor for SARS-CoV-RBD (Li et al., 2020c) , a strong interaction between SARS-CoV-2-RBD and KIM1 is suggested (Table 1; Supplementary Table S1) . Notably, the different binding regions of SARS-CoV-2-RBD to KIM1 and to ACE2 indicated by our data suggest that KIM1 and ACE2 may synergistically mediate SARS-CoV-2 invasion ( Figure 2B ). Clinically, mutations in SARS-CoV-2-S have been identified (Supplementary Figure S3 and Table S2 ), and COVID-19 cases carrying V367F mutation in SARS-CoV-2-S, which contacting KIM1, have been reported (http://giorgilab.dyndns.org/coronapp/, summarized in Supplementary Figure S3B and C) (Mercatelli et al., 2020) . Molecular mechanics generalized born surface area (MM-GBSA) analysis suggests that V367F mutation leads to enhanced binding with KIM1 (Supplementary Table S1 ), which may associate with clinical findings that V367F leads to enhanced infectivity of SARS-CoV-2 (Starr et al., 2020; Li et al., 2020a) ; further investigations on these clinical mutations will be important. Microarray data showed increased Kim1 expression in SARS patients-derived peripheral blood mononuclear cells compared to healthy controls (GSE1739, Supplementary Figure S4A ; Reghunathan et al., 2005) . Considering the facts that SARS-CoV-RBD and SARS-CoV-2-RBD both invade the kidney (Ding et al., 2004; Braun et al., 2020) and share high homology ( Figure 1B) , we evaluated the binding potential of SARS-CoV-RBD with KIM1 ( Supplementary Figures S4 and S5 ). Sharing the same binding pocket within KIM1 (contacting surface shown in Figure 2C ), SARS-CoV-RBD binds to KIM1 Ig V at a combined free energy of -21.59 kcal/mol (Supplementary Tables S1 and S3), suggesting a relatively lower affinity to KIM1 than that of SARS-CoV-2-RBD (-35.64 kcal/mol), whereas an even weak interaction was found between MERS-COV-RBD and Ig V (-10.12 kcal/mol, Supplementary Table S1 ). Therefore, SARS-CoV-2-RBD showed the highest binding affinity to KIM1; moreover, SARS-CoV-RBD and SARS-CoV-2-RBD share the same binding pocket on the Ig V domain ( Figure 2C ). To confirm the binding between SARS-CoV-2-RBD and KIM1, endogenous and exogenous co-immunoprecipitation (co-IP) assays were performed ( Figure Since KIM Ig V is crucial in mediating viral receptor binding (Yuan et al., 2015; Niu et al., 2018) , plasmids overexpressing full-length KIM1, the Ig V domain of KIM1, or truncated KIM1 without Ig V domain (ΔIg V) were respectively co-transfected with SARS-CoV-2-RBD into a stable KIM1 knockout HK-2 cell line or HEK293T cells ( Figure 3C and D; Supplemental Figure S6B and C). Knocking out KIM1 or deletion of Ig V domain abolished the binding between KIM1 and SARS-CoV-2-RBD ( Figure 3C and D). The interaction between KIM1 Ig V and SARS-CoV-2-RBD was also verified by FRET-based assays, and no obvious FRET signal was observed in cells co-transfected with KIM1 ΔIg V-CFP and SARS-CoV-2-RBD-YFP ( Figure 3I and J). These results together suggest that Ig V domain is crucial in mediating the interaction between KIM1 and SARS-CoV-2. We next used fluorescein isothiocyanate (FITC) labelling to track SARS-CoV-2-RBD in human cells. For each indicated group, at least 100 cells from 5 fields under high power objective lens were included in assessment. We observed less binding signal of FITC-SARS-CoV-2-RBD on the surface of human renal cells when KIM1 was knocked out, while more intense signal when KIM1 was overexpressed ( Figure 4A and B). In KIM1 knockout HK-2 cells, restoring full-length KIM1 and overexpressing Ig V both rescued binding signals of SARS-CoV-2-RBD on cell surface ( Figure 4A ), demonstrating the importance of KIM1 Ig V in mediating viral attachment. Moreover, knockout of KIM1 attenuated the cytotoxicity induced by SARS-CoV-2-RBD (Supplementary Figure S6D) . Together, these results further confirm the crucial role of Ig V domain in mediating SARS-CoV-2 attachment to renal cells. To competitively bind with SARS-CoV-2-RBD and inhibit its interaction with KIM1, we rationally designed two antagonist peptides based on SARS-CoV-2-contacting motifs in KIM1 (motif 1: Leu54, Phe55, Gln58; motif 2: Trp112, Phe113; Figure 5A ). Peptide 1 (AP1) mimics motif 1, while peptide 2 (AP2) covers both motifs, with three glycine used as a flexible linker ( Figure 5A ). The binding free energy, which indicates binding between peptides and SARS-CoV-2-RBD, was provided in Supplementary Table S1 . Both peptides did not show distinct cytotoxicity, and AP2 reduced SARS-CoV-2-RBD attachment to cell surface and protected against its cytotoxicity ( Figure 5B-D) . Moreover, AP2 significantly inhibited the interaction between KIM1 and SARS-CoV-2-RBD, indicated by the abolished FRET signal between KIM1 and SARS-CoV-2-RBD upon AP2 treatment ( Figure 5E and F). Enhanced SARS-CoV-2-RBD binding and prolonged half-life are undergoing by optimizing the sequences or modifications of AP2 with the approaches we recently described (Wang et al., 2020) . Since KIM1 is protective against AKI (Yang et al., 2015) , our strategy is unlikely to interfere with the beneficial effects of KIM1 in vivo. To fight against COVID-19 pandemic, a deep understanding of how SARS-CoV-2 invades human cells is warranted. Studies have indicated direct infection of SARS-CoV-2 in the kidney in addition to the lung (Braun et al., 2020; Farkash et al., 2020) . However, ACE2 remains the only well-recognized receptor that may mediate this invasion. Furthermore, the renal tropism of SARS-CoV-2 and associated kidney injury seem unexplainable by the relatively decreased level of ACE2 upon viral invasion . Here, our study suggests that KIM1, a drastically upregulated biomarker for kidney injury (Yang et al., 2015) , mediates SARS-CoV-2 kidney invasion as a receptor. We also found that SARS-CoV-2-RBD binds to KIM1 with a higher affinity than that of SARS-CoV-RBD and MERS-COV-RBD, which probably underlies the stronger contagion of SARS-CoV-2 (Rabaan et al., 2020) ; therefore, the renal infection and the roles of KIM1 in these severe respiratory diseases worth revisiting. Notably, our results suggest distinct binding sites of KIM1 and ACE2 on viral RBD, thus it is worth investigating whether and how KIM1 and ACE2 co-mediate SARS-CoV-2 invasion in these organs. In addition, since KIM1 is endocytosed via clathrin-dependent pathways (Zhao et al., 2016) , it would also be interesting to further explore the KIM1-dependent process after viral attachment to cell membrane. ACE2 is the most well-studied receptor for SARS-CoV-2, yet it is not an ideal therapeutic target for COVID-19, since it is widely expressed in multiple organs and plays crucial roles in regulating blood pressure and preventing heart/kidney injury Li et al., 2020d) . In contrast, KIM1 has stronger association with kidney function and is highly expressed only after renal injury (Kondratowicz et al., 2011; Yuan et al., 2015; Costafreda et al., 2018) , which makes it a more specific and maybe also safer therapeutic target for COVID-19 patients with kidney diseases. In summary, our data suggest a crucial role of KIM1 in SARS-CoV-2 renal tropism as a potential receptor for SARS-CoV-2. Here, we propose a model of a 'vicious cycle' co-mediated by KIM1 and ACE2 ( Figure 5G ), which may explain the renal tropism of SARS-CoV-2 in COVID-19 patients. During the initial stage of SARS-CoV-2 invasion, the higher physiological level (Supplementary Figure S1 ) and binding affinity (Supplementary Table S1 ) make ACE2 the primary target, which is not kidney-specific. However, after onset of virus-induced acute kidney injury, the resulting drastically upregulated KIM1 rapidly promotes a secondary viral infection co-mediated by KIM1 and ACE2, which is more kidney-specific, and consequently exacerbates kidney damage in a vicious cycle ( Figure 5G ). Approaches that can break the interaction between SARS-CoV-2 and KIM1, including anti-KIM1 antibodies, small-molecule inhibitors, and KIM1-derived antagonist peptides, may shed light on COVID-19 treatment. Recombinant SARS-CoV-2-RBD (T80302) was obtained from Genscript. Antagonist peptide 1 (AP1, SCSLFTCQNGIV, purity >95%) and antagonist peptide 2 (AP2, SCSLFTCQNGGGWF, purity >95%) were chemically synthesized by Genscript. Anti-mouse-IgG antibody (p/n 18-8816-33) and anti-rabbit-IgG antibody (p/n 18-8817-33) were obtained from RockLand. IgG with SureBeads TM Protein G magnetic beads (J2112LB-02) was purchased from Bio-Rad. DAPI (D9542) was from Sigma. Alex Flour 594 labelled phalloidin (C2205S) was from Beyotimes. Antibodies against KIM1 (NBP1-76701, Novus Biologicals), Flag (F1804, Sigma), HA (H6908, Sigma), and ACE2 (21115-1-AP, Proteintech) were used. To obtain the comprehensive transcriptome and protein profiles of KIM1 and ACE2 for human tissues, we collected and analyzed the transcriptome data and immunohistochemistry-based protein profiles from Human Protein Atlas (HPA, https://www.proteinatlas.org), which showed the expression and localization of human proteins across tissues and organs, based on deep sequencing of RNA (RNA-seq) from 37 normal tissue and immunohistochemistry on tissue microarrays containing 44 tissue types (Uhlen et al., 2015) . HPA RNA-seq tissue of the protein-coding gene was recorded as mean protein-coding transcripts per million (pTPM), corresponding to mean values of samples from each tissue. Histology-based protein expression levels were analyzed manually into 4 levels (not detected, low, medium, and high). In Supplementary Figure S1 , top 10 tissular transcriptional levels and histology-based protein expression levels of KIM1 and ACE2 are listed, respectively, and the overlapped expression profile of KIM1 and ACE2 are summarized. RMSD was utilized to estimate the average change in displacement of a selection of atoms for a particular frame as described (Li et al., 2011) . RMSF was conducted to study the displacement changes in the protein chain (Li et al., 2011) . I/R injury was performed on C57BL/6 mice as we previously described (Chen et al., 2015 (Chen et al., , 2017 . For cisplatin-induced acute kidney injury, 30 mg/kg bodyweight cisplatin was injected intraperitoneally into 8-week-old male mice, and mice were sacrificed 3 days later. Blood and kidney samples were collected for further analysis, with n = 4 for each experimental animal group. Total RNA was isolated from kidneys by RNA iso Plus (TaKaRa) and reverse-transcribed into cDNA using the M-MLV first-strand synthesis system (Invitrogen). The abundance of specific gene transcripts was assessed by qPCR. Primers used in the study are provided (Supplementary Table S4 ). Human kidney tubular cell line HK-2 (obtained from China Center for Type Culture Collection) was cultured in DMEM/F12 media (Hyclone) containing 17.5 mM glucose and 10% fetal bovine serum. To evaluate the impact of SARS-CoV-2 on cells, HK-2 cells were transfected with SARS-CoV-2-S and SARS-CoV-2-RBD plasmids, and then collected for further detection. Indicated HK-2/HEK293T cells (1×10 7 ) were lysed in 1 ml pre-lysis buffer (25 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1% NP-40, 1 mM EDTA, 5% glycerol), which is formulated for pulldown and IP assays and as a wash buffer for beads. For IP, cell lysate was immunoprecipitated with the indicated antibody or respective IgG with SureBeads TM Protein G magnetic beads overnight at 4°C. After washing with pre-lysis buffer containing 500 mM NaCl, the beads were boiled in loading buffer and subjected to immunoblotting (Wan et al., 2017) . FRET-based assay (Karpova et al., 2006) (Li et al., 2020b) . Co-transfection of unconjugated CFP and YFP was included as a negative control as described (Karpova and McNally, 2006) . Interaction between KIM1 and its ligand TIM4 was detected by FRET assay as a positive control (Rong et al., 2011) . The CRISPR-Cas9-based protocols for genome engineering were used as described (Zhang et al., 2017) . Guide RNA target sequences for KIM1 are provided (Supplementary Table S4 ). FITC label was performed as we previously described Li et al., 2020b) . Briefly, SARS-CoV-2-RBD was co-incubated with FITC (molar ratio 1:5) overnight, and then 5 mM NH 4 Cl was added to stop the reaction and quench the un-reacted FITC. The solution was dialyzed twice and lyophilized for further use. HEK293T cells (5 × 10 6 ) or HK-2 cells (1 × 10 7 ) were incubated with free FITC or FITC-SARS-CoV-2-RBD (100 μg/ml) for 2 . For peptide-based internalization assays, AP1 or AP2 (50 μM) was co-added with FITC-SARS-CoV-2-RBD (100 μg/ml). After fixing with 4% (w/v) formaldehyde, cell membranes were stained with Alex Flour 594 labelled phalloidin (2 μg/ml) and the nuclei were stained by DAPI (1 μg/ml), and then imaged with a Leica TCS SP8 confocal microscope. For each group, at least 100 cells from 5 fields under high power objective lens (64×) were included in the assessment. Representative images were presented. Quantification of images was conducted by Image J 1.8.0. Cells were plated at 3000-4000 cells per well in 96-well plates. At 80% confluence, cells incubated with SARS-CoV-2-RBD (100 μg/ml) were treated with or without AP1 or AP2 (10, 50, 100 μM). After that, 10 μl MTT (5 mg/ml) was added to each well for 4 h, medium was removed, and DMSO was added. Absorbance measured at 490 nm was normalized to the respective control group. Data were expressed as mean ± SD. Significant differences were assessed by two-tailed Student's test. Two-sided P-value <0.05 was considered statistically significant. Analyses were performed with EXCEL 2017 and GraphPad Prism 8.0. Top 10 ranked residues involved in the binding of SARS-CoV-2-RBD and KIM1 Ig V are listed. After 36 h, cells were lysed and subjected to co-IP followed by immunoblotting with indicated antibodies. Anti-rabbit light chain-specific IgG was used to avoid interference of IgG heavy chain. KIM1/ACE2 mediate the initial kidney infection, and the resulting acute kidney injury drastically upregulates KIM1, which in turn promotes infection and consequently exacerbates the kidney injury. SARS-CoV-2 renal tropism associates with acute kidney injury Gastrointestinal and hepatic manifestations of COVID-19: a comprehensive review Apelin protects against acute renal injury by inhibiting TGF-β1. BBA-Mol ELABELA and an ELABELA Fragment Protect against AKI Clinical Characteristics and Outcomes of Patients With Diabetes and COVID-19 in Association With Glucose-Lowering Medication HAVCR1 (CD365) and Its Mouse Ortholog Are Functional Hepatitis A Virus (HAV) Cellular Receptors That Mediate HAV Infection TIM-1 Ubiquitination Mediates Dengue Virus Entry Organ distribution of severe acute respiratory syndrome (SARS) associated coronavirus (SARS-CoV) in SARS patients: implications for pathogenesis and virus transmission pathways In-vitro renal epithelial cell infection reveals a viral kidney tropism as a potential mechanism for acute renal failure during Middle East Respiratory Syndrome (MERS) Coronavirus infection Ultrastructural Evidence for Direct Renal Infection with SARS-CoV-2 AKI in Hospitalized Patients with and without COVID-19: A Comparison Study Clinical Characteristics of Coronavirus Disease 2019 in China Acute kidney injury in patients hospitalized with COVID-19 Angiotensin-converting enzyme 2 protects from severe acute lung failure Detecting protein-protein interactions with CFP-YFP FRET by acceptor photobleaching T-cell immunoglobulin and mucin domain 1 (TIM-1) is a receptor for Zaire Ebolavirus and Lake Victoria Marburgvirus A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor Molecular dynamics studies of the 3D structure and planar ligand binding of a quadruplex dimer The Impact of Mutations in SARS-CoV-2 Spike on Viral Infectivity and Antigenicity Copper and iron ions accelerate the prion-like propagation of α-synuclein: A vicious cycle in Parkinson's disease The MERS-CoV Receptor DPP4 as a Candidate Binding Target of the SARS-CoV-2 Spike Physiological and pathological regulation of ACE2, the SARS-CoV-2 receptor Coronapp: a web application to annotate and monitor SARS-CoV-2 mutations Binding-affinity predictions of HSP90 in the D3R Grand Challenge 2015 with docking, MM/GBSA, QM/MM, and free-energy simulations TIM-1 Promotes Japanese Encephalitis Virus Entry and Infection Renal Involvement and Early Prognosis in Patients with COVID-19 Pneumonia SARS-CoV-2, SARS-CoV, and MERS-COV: a comparative overview Expression profile of immune response genes in patients with Severe Acute Respiratory Syndrome The TIM-1:TIM-4 pathway enhances renal ischemia-reperfusion injury Renal histopathological analysis of 26 postmortem fi ndings of patients with COVID-19 in China Product of natural evolution (SARS, MERS, and SARS-CoV-2); deadly diseases, from SARS to SARS-CoV-2 Structural basis of receptor recognition by SARS-CoV-2 Deep Mutational Scanning of SARS-CoV-2 Receptor Binding Domain Reveals Constraints on Folding and ACE2 Binding Proteomics. Tissue-based map of the human proteome PEGylated and Acylated Elabela Analogues Show Enhanced Receptor Binding, Prolonged Stability, and Remedy of Acute Kidney Injury MacroH2A1.1 cooperates with EZH2 to promote adipogenesis by regulating Wnt signaling COVID-19 and chronic renal disease: clinical characteristics & prognosis KIM-1-mediated phagocytosis reduces acute injury to the kidney TIM-1 acts a dual-attachment receptor for Ebolavirus by interacting directly with viral GP and the PS on the viral envelope ANGPTL8 negatively regulates NF-κB activation by facilitating selective autophagic degradation of IKKγ Histone methyltransferase G9a protects against acute liver injury through GSTP1 Kidney Injury Molecule-1 Enhances Endocytosis of Albumin in Renal Proximal Tubular Cells The work was technically supported by the Analytical and Testing Center of Huazhong University of Science and Technology.