Ying Zhao’s research while affiliated with Wenzhou Medical University and other places

What is this page?


This page lists works of an author who doesn't have a ResearchGate profile or hasn't added the works to their profile yet. It is automatically generated from public (personal) data to further our legitimate goal of comprehensive and accurate scientific recordkeeping. If you are this author and want this page removed, please let us know.

Publications (12)


JOSD2 alleviates acute kidney injury through deubiquitinating SIRT7 and negativity regulating SIRT7-NF-κB inflammatory pathway in renal tubular epithelial cells
  • Article

April 2025

·

1 Read

Acta Pharmacologica Sinica

Ying Zhao

·

Qing-qing Zhao

·

Shi-jie Fan

·

[...]

·

Acute kidney injury (AKI), triggered by various stimuli including ischemia-reperfusion, nephrotoxic insult, and sepsis, is characterized by an abrupt deterioration in kidney function. Ubiquitination is a post-translational modification of proteins that plays a critical role in the pathogenesis and progression of AKI. In this study, we aimed to investigate the role and underlying mechanism of the deubiquitinating enzyme Josephin Domain-containing protein 2 (JOSD2) in AKI. We found that deficiency of JOSD2 exacerbated renal tubular injury and inflammation in AKI mice induced by cisplatin or ischemia-reperfusion injury. Conversely, the specific overexpression of JOSD2 in renal tubular epithelial cells effectively prevented renal tubular injury and inflammation induced in AKI mice. Mechanistically, we identified Sirtuin 7 (SIRT7) as a potential substrate of JOSD2 through mass spectrometry combined with co-immunoprecipitation analysis. JOSD2 removes the K63-linked ubiquitination of SIRT7 via its active site C24 and promotes P62-mediated autophagic degradation of SIRT7, which subsequently prevents the phosphorylation and nuclear translocation of P65 and reduces inflammatory responses in renal tubular epithelial cells. Our study reveals the role of the JOSD2-SIRT7 axis in regulating AKI-induced renal inflammation and highlights the potential of JOSD2 as a promising therapeutic target for AKI.


Identification of OTUD5 as a regulator of podocyte inflammation and injury
a A volcano plot analysis illustrating the differential expression of DUBs induced by HG/PA in podocytes. (n = 3 samples for each Ctrl group and HG/PA group; P values were determined by Wald test from DESeq2 software with Benjamini-Hochberg’s correction). b A table shows DUBs with significant differences in MPC5 cells treated with HG/PA. c The mRNA level of Otud5 in HG/PA-induced MPC5 cell lines. (n = 3 independent experiments; P values were determined by two-tailed unpaired t-test and data are presented as mean ± SD). d Representative western blot of OTUD5 expression in MPC5 cell lines after stimulation with HG/PA for different durations. (n = 3 independent experiments). e Representative immunofluorescence (IF) images of OTUD5 expression in human renal tissue from normal subjects (n = 3 samples) and patients with DKD (n = 3 samples). Scale bar, 50 μm. Representative western blot of OTUD5 expression in renal cortex of T2DM (f) mice and T1DM mice (g). (n = 6 samples). Representative western blot of OTUD5 expression in renal cortex of NOD (h) mice and db/db mice (i). (n = 6 samples). j, k Representative western blot of OTUD5 expression in primary podocytes of T2DM (j) and T1DM (k) mice. (n = 6 samples). l MPC5 cells transfected with Flag-OTUD5 were stimulated with HG/PA for 8 h. Real-time qPCR showed the mRNA levels of Il6 and Tnfα. (n = 3 independent experiments; P values were determined by one-way ANOVA with Bonferroni’s correction and data are presented as mean ± SD). Representative western blot of Cleaved Caspase3 (m) and Nephrin (n) expression in OTUD5-overexpression podocytes stimulated by HG/PA for 24 h. (n = 3 independent experiments).
Podocyte-specific Otud5 knockout aggravates podocyte injury and DKD in T2DM mice
a Schematic diagram of the strategy for the generation of podocyte-specific Otud5 knockout mice (OTUD5CKO). b Schematic diagram depicting the procedure of STZ/HFD-induced T2DM mice. c Weekly monitoring of blood glucose levels in mice. Data are presented as mean ± SD. The levels of serum creatinine (d), urea nitrogen (e), and urine albumin to creatinine ratio (f) were analyzed in mice. g, h Representative images of hematoxylin and eosin staining (H&E), periodic acid-Schiff (PAS), and transmission electron microscopy (TEM) in mice. Scale bar: black 20 μm, red 1 μm. (n = 6 samples). Quantification of glomerular basement membrane (GBM) thickness (i) and podocyte foot process numbers (j, k) in the glomeruli. l Representative immunofluorescence (IF) images of Nephrin expression in glomeruli from mice. Scale bar, 20 μm. (n = 6 samples). Real-time qPCR showing mRNA levels of Il6 (m) and Tnfα (n) in kidney tissues of each group. n = 6 for each group. For d–f, i–k, m, and n, P values were determined by one-way ANOVA with Bonferroni’s correction, and data are presented as mean ± SD.
OTUD5CKO exacerbates podocyte injury and DKD in T1DM mice
a Weekly monitoring of blood glucose levels in mice. Data are presented as mean ± SD. The levels of serum creatinine (b), urea nitrogen (c), and urine albumin to creatinine ratio (d) were analyzed in mice. e, f Representative images of H&E, PAS, and TEM in mice. Scale bar: black 20 μm, red 1 μm. (n = 6 samples). Quantification of GBM thickness (g) and podocyte foot process numbers (h, i) in the glomeruli. j Representative IF images of Nephrin expression in glomeruli from mice. Scale bar, 20 μm. (n = 6 samples). Real-time qPCR showing mRNA levels of Il6 (k) and Tnfα (l) in the kidney tissues of each group. n =6 for each group. For b–d, g–i, k, and l, P values were determined by one-way ANOVA with Bonferroni’s correction, and data are presented as mean ± SD.
Identification of TAK1 as a potential substrate protein of OTUD5
a Schematic illustration of a quantitative proteomic screen to identify proteins binding to OTUD5. b Mass spectrometry/mass spectrometry (MS/MS) spectrum of the peptide MITTSGPTSEK from TAK1. Co-immunoprecipitation (Co-IP) of OTUD5 and TAK1 in MPC5 cells (c) and kidney tissues (d). Endogenous OTUD5 was immunoprecipitated. (n = 3 independent experiments). e Co-IP of OTUD5 in NIH/3T3 co-transfected with Flag-OTUD5 and His-TAK1 plasmids. Exogenous OTUD5 was immunoprecipitated using an anti-Flag antibody. (n = 3 independent experiments). f His-TAK1 and Flag-OTUD5 were transfected into MPC5 cells with or without HG/PA treatment and then subjected to 10 μM MG132 for 6 h. Ubiquitinated TAK1 was detected by immunoblotting using an anti-ubiquitin antibody. (n = 3 independent experiments). g His-TAK1, HA-WT Ub, HA-K48 Ub, and HA-K63 Ub were transfected into NIH/3T3 together with Flag-OTUD5 and then subjected to 10 μM MG132 for 6 h. Ubiquitinated TAK1 was detected by immunoblotting using an anti-HA antibody. (n = 3 independent experiments). h His-TAK1 and HA-K63 Ub were transfected into NIH/3T3 together with Flag-OTUD5 (WT or C224S) and then subjected to 10 μM MG132 for 6 h. Ubiquitinated TAK1 was detected by immunoblotting using an anti-HA antibody. (n = 3 independent experiments). i Schematic illustration of the construct for mutating the ubiquitinated lysine residue of TAK1. j His-TAK1 (WT, K34R, K158R, K209R or K562R) and HA-WT Ub were transfected into NIH/3T3 together with Flag-OTUD5 and then subjected to 10 μM MG132 for 6 h. Ubiquitinated TAK1 was detected by immunoblotting using an anti-HA antibody. (n = 3 independent experiments).
OTUD5 negatively regulates TAK1 activation and inflammation in podocytes
MPC5 cells transfected with Flag-OTUD5 (a) or si-OTUD5 (b) were stimulated with HG/PA for 30 min. Representative western blot analysis of P-TAK1. (n = 3 independent experiments). c, d Representative western blot analysis of P-TAK1 in kidney tissues of each group. (n = 6 samples). MPC5 cells transfected with Flag-OTUD5 (e) or si-OTUD5 (f) were stimulated with HG/PA for 30 min. Representative western blot analysis of phosphorylated and total protein levels of ERK, P38, and JNK. (n = 3 independent experiments). MPC5 cells transfected with si-OTUD5 were pretreated with 10 μM Takinib (TAK1 inhibitor) for 1 h before exposure to HG/PA. g Levels of P-TAK1, P-ERK, P-P38, and P-JNK were detected by western blot. h Real-time qPCR showing mRNA levels of Il6 and Tnfα. (n = 3 independent experiments; P values were determined by one-way ANOVA with Bonferroni’s correction and data are presented as mean ± SD). i His-TAK1 was transfected into NIH/3T3 with or without Flag-OTUD5 (WT or C224A). Co-IP was performed with an anti-His antibody, followed by a western blot of TAK1 and TAB2. (n = 3 independent experiments). j MPC5 cells transfected with Flag-OTUD5 (WT or C224A) were stimulated with HG/PA for 30 min. Representative western blot analysis of phosphorylated and total protein levels of TAK1, ERK, P38, and JNK. (n = 3 independent experiments). k His-TAK1(WT or K158R) was transfected into NIH/3T3 with or without Flag-OTUD5. Co-IP was performed with an anti-His antibody, followed by a western blot of TAK1 and TAB2. (n = 3 independent experiments). l His-TAK1 (WT or K158R) and Flag-OTUD5 were transfected into MPC5 cells for 24 h and then stimulated by HG/PA for 30 min. Representative western blot analysis of phosphorylated and total protein levels of TAK1, ERK, P38, and JNK. (n = 3 independent experiments).

+2

Podocyte OTUD5 alleviates diabetic kidney disease through deubiquitinating TAK1 and reducing podocyte inflammation and injury
  • Article
  • Full-text available

June 2024

·

83 Reads

·

11 Citations

Recent studies have shown the crucial role of podocyte injury in the development of diabetic kidney disease (DKD). Deubiquitinating modification of proteins is widely involved in the occurrence and development of diseases. Here, we explore the role and regulating mechanism of a deubiquitinating enzyme, OTUD5, in podocyte injury and DKD. RNA-seq analysis indicates a significantly decreased expression of OTUD5 in HG/PA-stimulated podocytes. Podocyte-specific Otud5 knockout exacerbates podocyte injury and DKD in both type 1 and type 2 diabetic mice. Furthermore, AVV9-mediated OTUD5 overexpression in podocytes shows a therapeutic effect against DKD. Mass spectrometry and co-immunoprecipitation experiments reveal an inflammation-regulating protein, TAK1, as the substrate of OTUD5 in podocytes. Mechanistically, OTUD5 deubiquitinates K63-linked TAK1 at the K158 site through its active site C224, which subsequently prevents the phosphorylation of TAK1 and reduces downstream inflammatory responses in podocytes. Our findings show an OTUD5-TAK1 axis in podocyte inflammation and injury and highlight the potential of OTUD5 as a promising therapeutic target for DKD.

Download


OTUD6A in tubular epithelial cells mediates Angiotensin II-induced kidney injury by targeting STAT3

December 2023

·

13 Reads

·

4 Citations

AJP Cell Physiology

Kidney fibrosis is a prominent pathological feature of hypertensive kidney diseases (HKD). Recent studies have highlighted the role of ubiquitinating/deubiquitinating protein modification in kidney pathophysiology. Ovarian tumor Domain-Containing Protein 6A (OTUD6A) is a deubiquitinating enzyme involved in tumor progression. However, its role in kidney pathophysiology remains elusive. We aimed to investigate the role and underlying mechanism of OTUD6A during kidney fibrosis in HKD. The results revealed higher OTUD6A expression in kidney tissues of nephropathy patients and mice with chronic Ang II administration than that from the control ones. OTUD6A was mainly located in tubular epithelial cells. Moreover, OTUD6A deficiency significantly protected mice against Ang II-induced kidney dysfunction and fibrosis. Also, knocking OTUD6A down suppressed Ang II-induced fibrosis in cultured tubular epithelial cells, while overexpression of OTUD6A enhanced fibrogenic responses. Mechanistically, OTUD6A bounded to STAT3 and removed K63 linked- ubiquitin chains to promote STAT3 phosphorylation at tyrosine705 position and nuclear translocation, which then induced pro-fibrotic gene transcription in epithelial cells. These studies identified STAT3 as a direct substrate of OTUD6A and highlighted the pivotal role of OTUD6A in Ang II-induced kidney injury, indicating OTUD6A as a potential therapeutic target for HKD.


OTUB1 inhibits breast cancer by non-canonically stabilizing CCN6

August 2023

·

21 Reads

·

13 Citations

Background: CCN6 is a matricellular protein that critically regulates the tumourigenesis and progression of breast cancer. Although the tumour-suppressive function of CCN6 has been extensively studied, molecular mechanisms regulating protein levels of CCN6 remain largely unclear. This study aims to investigate the regulation of CCN6 by ubiquitination and deubiquitinating enzymes (DUBs) in breast cancer. Methods: A screening assay was performed to identify OTUB1 as the DUB for CCN6. Various biochemical methods were applied to elucidate the molecular mechanism of OTUB1 in the regulation of CCN6. The role of OTUB1-CCN6 interaction in breast cancer was studied with cell experiments and the allograft model. The correlation of OTUB1 and CCN6 in human breast cancer was determined by immunohistochemistry and Western blot. Results: We found that CCN6 protein levels were controlled by the ubiquitin-proteasome system. The K48 ubiquitination and degradation of CCN6 was inhibited by OTUB1, which directly interacted with CCN6 through its linker domain. Furthermore, OTUB1 inhibited the ubiquitination of CCN6 in a non-canonical manner. Deletion of OTUB1, concomitant with reduced CCN6 abundance, increased the migration, proliferation and viability of breast cancer cells. Supplementation of CCN6 abolished the effect of OTUB1 deletion on breast cancer. Importantly, OTUB1 expression was downregulated in human breast cancer and positively correlated with CCN6 levels. Conclusion: This study identified OTUB1 as a novel regulator of CCN6 in breast cancer.


USP25 inhibits renal fibrosis by regulating TGFβ-SMAD signaling pathway in Ang II-induced hypertensive mice

April 2023

·

18 Reads

·

12 Citations

Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease

Renal fibrosis is a crucial pathological feature of hypertensive renal disease (HRD). In-depth analysis of the pathogenesis of fibrosis is of great significance for the development of new drugs for the treatment of HRD. USP25 is a deubiquitinase that can regulate the progression of many diseases, but its function in the kidney remains unclear. We found that USP25 was significantly increased in human and mice HRD kidney tissues. In the HRD model induced by Ang II, USP25-/- mice showed significant aggravation of renal dysfunction and fibrosis compared with the control mice. Consistently, AAV9-mediated overexpression of USP25 significantly improved renal dysfunction and fibrosis. Mechanistically, USP25 inhibited the TGF-β pathway by reducing SMAD4 K63-linked polyubiquitination, thereby suppressing SMAD2 nuclear translocation. In conclusion, this study demonstrates for the first time that the deubiquitinase USP25 plays an important regulatory role in HRD.


USP25 Ameliorates Pathological Cardiac Hypertrophy by Stabilizing SERCA2a in Cardiomyocytes

February 2023

·

60 Reads

·

79 Citations

Circulation Research

Background: Pathological cardiac hypertrophy can lead to heart failure and is one of the leading causes of death globally. Understanding the molecular mechanism of pathological cardiac hypertrophy will contribute to the treatment of heart failure. DUBs (deubiquitinating enzymes) are essential to cardiac pathophysiology by precisely controlling protein function, localization, and degradation. This study set out to investigate the role and molecular mechanism of a DUB, USP25 (ubiquitin-specific peptidase 25), in pathological cardiac hypertrophy. Methods: The role of USP25 in myocardial hypertrophy was evaluated in murine cardiomyocytes in response to Ang II (angiotensin II) and transverse aortic constriction stimulation and in hypertrophic myocardium tissues of heart failure patients. Liquid chromotography with mass spectrometry/mass spectrometry analysis combined with Co-IP was used to identify SERCA2a (sarcoplasmic/endoplasmic reticulum Ca2+-ATPase 2A), an antihypertrophy protein, as an interacting protein of USP25. To clarify the molecular mechanism of USP25 in the regulation of SERCA2a, we constructed a series of mutant plasmids of USP25. In addition, we overexpressed USP25 and SERCA2a in the heart with adenoassociated virus serotype 9 vectors to validate the biological function of USP25 and SERCA2a interaction. Results: We revealed increased protein level of USP25 in murine cardiomyocytes subject to Ang II and transverse aortic constriction stimulation and in hypertrophic myocardium tissues of patients with heart failure. USP25 deficiency aggravated cardiac hypertrophy and cardiac dysfunction under Ang II and transverse aortic constriction treatment. Mechanistically, USP25 bound to SERCA2a directly via its USP (ubiquitin-specific protease) domain and cysteine at position 178 of USP25 exerts deubiquitination to maintain the stability of the SERCA2a protein by removing the K48 ubiquitin chain and preventing proteasomal pathway degradation, thereby maintaining calcium handling in cardiomyocytes. Moreover, restoration of USP25 expression via adenoassociated virus serotype 9 vectors in USP25-/- mice attenuated Ang II-induced cardiac hypertrophy and cardiac dysfunction, whereas myocardial overexpression of SERCA2a could mimic the effect of USP25. Conclusions: We confirmed that USP25 inhibited cardiac hypertrophy by deubiquitinating and stabilizing SERCA2a.


USP25 inhibits renal fibrosis by regulating TGFβ-SMAD signaling pathway in Ang II-induced hypertensive mice

July 2022

·

49 Reads

Renal fibrosis is a crucial pathological feature of hypertensive renal disease (HRD). In-depth analysis of the pathogenesis of fibrosis is of great significance for the development of new drugs for the treatment of HRD. USP25 is a deubiquitinase that can regulate the progression of many diseases, but its function in the kidney remains unclear. We found that USP25 was significantly increased in human and mouse HRD kidney tissues. In the HRD model induced by Ang II, USP25 −/− mice showed significant aggravation of renal dysfunction and fibrosis compared with the control mice. Consistently, AAV9-mediated overexpression of USP25 significantly improved renal dysfunction and fibrosis. Mechanistically, USP25 inhibited the TGF-β pathway by reducing SMAD4 k63-linked polyubiquitination, thereby suppressing SMAD2 nuclear translocation. In conclusion, this study demonstrates for the first time that the deubiquitinase USP25 plays an important regulatory role in HRD.


Fig. 2 FAK inhibition suppresses LPS-induced MAPK phosphorylation. A, B RAW 264.7 cells (RAW) were transfected with siRNA against FAK and then exposed to 0.5 μg/mL LPS for 30 min. Control cells were transfected with negative control siRNA (NC). Panel A shows immunoblots of p-FAK and FAK. Total FAK and GAPDH were used as controls. Quantification of p-FAK and FAK levels is shown in panel B [Mean ± SEM, 3 independent experiments; **P < 0.01 compared to Ctrl siRNA; ## P < 0.01 compared to Ctrl siRNA+LPS]. C, D Activation of the MAPK pathway was assessed by measuring phosphorylated ERK, p38, and JNK in panel C. Total ERK, p38, and JNK were used as control. Quantification of p-ERK, p-JNK and p-P38 levels is shown in panel D [Mean ± SEM, 3 independent experiments; **P < 0.01 and ***P < 0.001 compared to Ctrl siRNA; # P < 0.05 and ## P < 0.01 compared to Ctrl siRNA+LPS]. E-H Macrophages were pretreated with PND-1186 for 1 h prior to LPS exposure. Figure showing RAW line (E) and mouse peritoneal macrophages (MPMs; G). Concentration of PND-1186 and LPS are as indicated. Activation of the MAPK pathway was assessed by immunoblotting for phospho-proteins. Densitometric quantification of p-ERK, p-JNK and p-P38 were detected in RAW cells (F) and MPMs (H) [Mean ± SEM, 3 independent experiments; *P < 0.05, **P < 0.01 and ***P < 0.001 compared to Ctrl;
Fig. 4 FAK regulates inflammatory responses in macrophages through interacting with TAK1. A RAW 264.7 (RAW) cells were pretreated with 1 μM PND-1186 for 1 h and then stimulated with 0.5 μg/mL LPS for 30 min. Cell lysates were analyzed for p-TAK1 (Ser412) and p-IKKα/β (Ser176/180) levels. Total TAK1 and IKKβ were used as control. B RAW cells were treated with 0.5 μg/mL LPS for 10 min. Complexes of FAK-TAK1 were detected by immunoprecipitation. C RAW cells were treated with 0.5 μg/mL LPS for the indicated times. Complexes of FAK-TAK1 were detected by immunoprecipitation. D RAW cells were pretreated with 1 μM PND-1186 for 1 h and then exposed to 0.5 μg/mL LPS for 10 min. Complexes of FAK-TAK1 were detected by immunoprecipitation. E RAW cells were pretreated with 2 μM Takinib for 1 h and then exposed to 0.5 μg/mL LPS for 30 min. The phosphorylated ERK, p38 and JNK were examined by western blot assay. Total ERK, p38, and JNK were used as control (TAKi = Takinib). F RAW cells were pretreated with 2 μM Takinib for 1 h and then stimulated with 0.5 μg/mL LPS for 30 min. Cell lysates were analyzed for p-IKKα/β and IκBα levels. Total IKKβ and GAPDH were used as control. G RAW cells were pretreated with 2 μM Takinib for 1 h and then exposed to 0.5 μg/mL LPS for 24 h. IL-6 proteins in the culture medium were measured by ELISA. Data normalized to total proteins and presented as % LPS [Mean ± SEM, 3 independent experiments; ***P < 0.001 compared to LPS]. H RAW cells were pretreated with 2 μM Takinib for 1 h and then exposed to 0.5 μg/mL LPS for 8 h. mRNA levels of IL-6 were measured. Data normalized to β-actin and expressed as % Ctrl [Mean ± SEM, 3 independent experiments; *P < 0.05 compared to LPS]. I RAW cells were transfected with FAK-expressing plasmid. After 24 h, levels of p-FAK and p-TAK1 were detected. Total FAK, TAK1, and GAPDH were used as control. Control cells were transfected with negative control/empty vector (NC = negative control, O/E = overexpression). J 3T3 cells were transfected with FAK-WT-Flag/FAK-Y397F-Flag/ TAK1-Myc expressing plasmid, respectively. After 24 h, levels of p-TAK1 were detected using western blot, with total FAK, Flag, Myc, and GAPDH as controls. K, L Cell-free kinase assay showing rhFAK phosphorylates rhTAK1. rhTAK1 was incubated with rhFAK in the presence or absence of ATP (100 μM). The samples were separated by SDS-PAGE and western blotting was used to detect p-TAK1, TAK1, and FAK in panel K. Densitometric quantification of p-TAK1 levels was determined in panel L [Mean ± SEM, 3 independent experiments; **P < 0.01 compared to rhTAK1]. M, N FAK-expressing RAW cells were treated with 2 μM Takinib for 12 h. IL-6 (L) and TNF-α (M) proteins in the culture medium were measured by ELISA. Data normalized to total proteins and presented as fold difference compare to NC [TAKi = Takinib; Mean ± SEM, 3 independent experiments; **P < 0.01 and ***P < 0.001 compared to NC; # P < 0.05 and ## P < 0.01 compared to O/E].
Fig. 5 FAK inhibitor protects against LPS-induced inflammatory response in mice. A Representative H&E-stained sections of lung tissues harvested from mice following LPS challenge [scale bar = 100 μm]. B Lung injury scores were assessed from histological analyses of mouse lung tissues [Mean ± SEM, n = 8; *P < 0.05 compared to Ctrl; # P < 0.05 compared to LPS]. C Lung wet/dry ratio was determined at 6 h after LPS challenge. [Mean ± SEM, n = 8; *P < 0.05 compared to Ctrl; # P < 0.05 compared to LPS]. D BALF was collected 6 h after LPS challenge and the amounts of proteins were measured. [Mean ± SEM, n = 8; *P < 0.05 compared to Ctrl; # P < 0.05 compared to LPS]. E, F Levels of IL-6 (E) and TNF-α (F) in BALF samples. [Mean ± SEM, n = 8; **P < 0.01 and ***P < 0.001 compared to Ctrl; # P < 0.05 compared to LPS]. G, H Levels of IL-6 (G) and TNF-α (H) in serum samples of mice [Mean ± SEM. N = 8; ***P < 0.001 compared to Ctrl; # P < 0.05 compared to LPS]. I Mice were challenged with intratracheal LPS and treated with PND-1186. Lung tissues were stained for inflammation marker p-p65 (green). Slides were counterstained with DAPI (blue) [scale bar = 100 μm]. J Protein levels of p-FAK, p-TAK1 and IκBα in mouse lung tissues challenged with intratracheal LPS. Total FAK, TAK, and GAPDH were used as control. K Complexes of FAK-TAK1 in mouse lung tissues challenged with intratracheal LPS were detected by immunoprecipitation.
FAK mediates LPS-induced inflammatory lung injury through interacting TAK1 and activating TAK1-NFκB pathway

July 2022

·

171 Reads

·

40 Citations

Cell Death and Disease

Acute lung injury (ALI), characterized by inflammatory damage, is a major clinical challenge. Developing specific treatment options for ALI requires the identification of novel targetable signaling pathways. Recent studies reported that endotoxin lipopolysaccharide (LPS) induced a TLR4-dependent activation of focal adhesion kinase (FAK) in colorectal adenocarcinoma cells, suggesting that FAK may be involved in LPS-induced inflammatory responses. Here, we investigated the involvement and mechanism of FAK in mediating LPS-induced inflammation and ALI. We show that LPS phosphorylates FAK in macrophages. Either FAK inhibitor, site-directly mutation, or siRNA knockdown of FAK significantly suppresses LPS-induced inflammatory cytokine production in macrophages. FAK inhibition also blocked LPS-induced activation of MAPKs and NFκB. Mechanistically, we demonstrate that activated FAK directly interacts with transforming growth factor-β-activated kinase-1 (TAK1), an upstream kinase of MAPKs and NFκB, and then phosphorylates TAK1 at Ser412. In a mouse model of LPS-induced ALI, pharmacological inhibition of FAK suppressed FAK/TAK activation and inflammatory response in lung tissues. These activities resulted in the preservation of lung tissues in LPS-challenged mice and increased survival during LPS-induced septic shock. Collectively, our results illustrate a novel FAK-TAK1-NFκB signaling axis in LPS-induced inflammation and ALI, and support FAK as a potential target for the treatment of ALI.


Design, synthesis and anticancer evaluation of 6,7-disubstituted-4-phenoxyquinoline derivatives bearing 1,8-naphthyridine-3-carboxamide moiety as novel multi-target TKIs

February 2022

·

31 Reads

·

6 Citations

Bioorganic Chemistry

Giving the fact that the disorders of multiple receptor tyrosine kinases (RTKs) are characteristics of various cancers, we assumed that developing novel multi-target drugs might have an advantage in treating the complex cancers. Taking the multi-target c-Met inhibitor Foretinib as the leading compound, we discovered a novel series of 6,7-disubstituted-4-phenoxyquinoline derivatives bearing 1,8-naphthyridine-3-carboxamide moiety with the help of molecular docking. Among them, the most promising compound 33 showed a prominent activity against Hela (IC50 = 0.21 µM), A549 (IC50 = 0.39 µM), and MCF-7 (IC50 = 0.33 µM), which were 3.28-4.82 times more active than that of Foretinib. Additionally, compound 33 dose dependently induced apoptosis by arresting A549 cells at G1 phase. Enzymatic assays and docking analyses were further confirmed that compound 33 was a multi-target inhibitor with the strong potencies against c-Met (IC50 = 11.77 nM), MEK1 (IC50 = 10.71 nM), and Flt-3 (IC50 = 22.36 nM). In the A549 cells mediated xenograft mouse model, compound 33 inhibited the tumor growth (TGI = 64%) without obvious toxicity, establishing compound 33 as a promising candidate for cancer therapy.


Citations (8)


... Podocytes and their foot processes constitute critical components of the renal filtration barrier, governing glomerular permeability. Podocyte injury is widely recognized as a central pathological mechanism in various kidney diseases, particularly dN and primary tubular disorders (37,38). Emerging evidence indicates that podocytes utilize lactate as an energy substrate and possess intrinsic regulatory systems to maintain lactate homeostasis. ...

Reference:

Lactate and lactylation in the kidneys: Current advances and prospects (Review)
Podocyte OTUD5 alleviates diabetic kidney disease through deubiquitinating TAK1 and reducing podocyte inflammation and injury

... OTUB1 demonstrates elevated expression levels across multiple tumor types [24,25] and participates in cell cycle control and apoptotic regulation through protein degradation pathways. Furthermore, OTUB1 promotes hypoxia-induced glycolytic reprogramming and enhances DNA damage, ultimately influencing tumor progression and metastasis through multiple molecular mechanisms [26][27][28]. Notably, increased OTUB1 expression in colorectal cancer is associated with metastatic potential and unfavorable clinical outcomes. Mechanistically, OTUB1 facilitates cancer cell migration and invasion by regulating epithelial-mesenchymal transition (EMT) markers [29]. ...

OTUB1 inhibits breast cancer by non-canonically stabilizing CCN6

... Increased expression of USP25 in AKI affects PKM2 (M2 isoform of pyruvate kinase, muscle), a crucial enzyme for glycolysis, which in turn causes kidney damage in AKI [105]. In contrast, in a hypertensive nephropathy model, USP25 inhibited the TGF-β signaling pathway by suppressing the ubiquitination process of Smad4, delaying renal fibrosis and protecting renal function [106]. ...

USP25 inhibits renal fibrosis by regulating TGFβ-SMAD signaling pathway in Ang II-induced hypertensive mice
  • Citing Article
  • April 2023

Biochimica et Biophysica Acta (BBA) - Molecular Basis of Disease

... The ubiquitin-proteasome system (UPS) is responsible for degrading the majority of intracellular proteins, maintaining a delicate equilibrium between ubiquitination catalyzed by E3 ubiquitin ligases and deubiquitination facilitated by deubiquitinating enzymes (DUBs) [6,7]. The human genome encodes approximately 100 DUBs, which play pivotal roles in stabilizing targeted proteins by removing ubiquitin molecules [8,9]. DUBs are further categorized into seven subclasses: ubiquitin-specific proteases (USPs), ubiquitin C-terminal hydrolases, ovarian tumor proteases (OTUs), Machado-Josephin domain proteases, a motif interacting with the ubiquitincontaining DUB family, zinc finger proteins with a UFM1-specific peptidase domain/zinc finger containing Ub peptidase 1, and JAB1/MPN/Mov34 metalloenzyme [10,11]. ...

USP25 Ameliorates Pathological Cardiac Hypertrophy by Stabilizing SERCA2a in Cardiomyocytes
  • Citing Article
  • February 2023

Circulation Research

... As expected, both in vivo (immunofluorescence staining) and in vitro experiments demonstrated that fisetin could limit the intracellular S. typhimurium load. Recent advance demonstrated that FAK is activated in macrophages to promote intracellular survival in response to Salmonella treatment [37]. Overall, these findings suggest that fisetin can regulate the microbiota and restrict intracellular S. typhimurium replication by inactivating FAK signaling. ...

FAK mediates LPS-induced inflammatory lung injury through interacting TAK1 and activating TAK1-NFκB pathway

Cell Death and Disease

... A 25% of active pharmaceutical ingredient contains amide scaffold due to its broad spectrum activity (Ghose et al., 1999). The benzamide moiety (A) was usually incorporated in hybrid molecules due to glucokinase activators (Mao et al., 2012), anticancer (Chandregowda et al., 2009;El-Hashash et al., 2018), central nervous systems of a 5-HT receptor (Dumuis et al., 1989), antiviral (Wu et al., 2017), anti-inflammatory (Tumer et al., 2017), 11β-HSD1 inhibitors (Rew et al., 2009), antiprion (Fiorino et al., 2012), antioxidant (Perin et al., 2018), histone deacetylase inhibitors (Lu et al., 2011), antitumor (Zhang et al., 2022), antifungal (Zhang et al., 2016), and antimicrobial activity (Ibrahim et al., 2016). Moreover, the hybrid compound is an artificial combination of two or more pharmacophores for triggering the desired activity (Alkhzem et al., 2022) such artificial scaffolds would able to act as an antibiotic with the ability to overcome drug resistance, intensify activity and increase binding affinity. ...

Synthesis, bioevaluation and molecular dynamics of pyrrolo-pyridine benzamide derivatives as potential antitumor agents in vitro and in vivo
  • Citing Article
  • February 2022

European Journal of Medicinal Chemistry

... With the development of cytobiology and molecular biology, the essential principles of tumorigenesis, invasion, migration, and metastasis induced by quinoline derivatives have been further explained. Antitumor mechanisms of quinoline derivatives include alkylating DNA [14], inhibiting c-Met kinase [15], epidermal growth factor receptor (EGFR) [16], and vascular endothelial growth factor (VEGF) [16][17][18]. Some were also proven to inhibit Pglycoprotein [19]. ...

Design, synthesis and anticancer evaluation of 6,7-disubstituted-4-phenoxyquinoline derivatives bearing 1,8-naphthyridine-3-carboxamide moiety as novel multi-target TKIs
  • Citing Article
  • February 2022

Bioorganic Chemistry

... Tis selective prooxidant cytotoxicity ofers a promising therapeutic strategy to target malignant cells while sparing healthy tissue (88)(89)(90). Examples include curcumin, which acts as an Nrf2 activator and NF-κB inhibitor in normal cells but induces ROS-mediated mitochondrial apoptosis in tumor cells via glutathione peroxidase inhibition (91,92). Resveratrol similarly protects normal cells but promotes apoptosis in cancer cells by impairing mitochondrial function and increasing ROS [93,94]. ...

Celastrol induces ROS-mediated apoptosis via directly targeting peroxiredoxin-2 in gastric cancer cells

Theranostics