Liming Lin’s research while affiliated with First Affiliated Hospital of China 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 (10)


Deubiquitinase USP13 alleviates doxorubicin-induced cardiotoxicity through promoting the autophagy-mediated degradation of STING
  • Article

April 2025

·

2 Reads

Acta Pharmaceutica Sinica B

Liming Lin

·

·

Diyun Xu

·

[...]

·


Figure 1. The expression of MYSM1 is up-regulated in myocardial I/R injury. A-B. HL-1 cells were transfected with expressing plasmids of JAMMs family (e.g., oeMYSM1) or empty vehicle (EV) before H/R injury (4 h hypoxia, 6 h reoxygenation; n = 6; *, vs. Ctrl; #, vs. EV+H/R; # P < 0.05, ## P < 0.01, *** P < 0.001). (A) Cell viability of each group. (B) Bar graph showing LDH levels. C. The expression levels of JAMMs family genes in mRNA sequencing of human heart tissue (GSE133054, HF=heart failure, NHF=non-heart failure, n = 5). D. Comprehensive combined RNA-seq dataset and function screens of JAMMs family for identification of MYSM1 as a regulator in I/R injury. E-F. MYSM1 mRNA levels in H/R-induced HL-1 cells (E) or myocardial I/R injury (F) were detected by RT-qPCR (n = 8). G-H. The protein levels of MYSM1 in H/R-induced HL-1 cells (G) or myocardial I/R injury (H) were examined by western blotting analysis (n = 6). I. Representative images of immunofluorescence staining for MYSM1(red), α-actin (green), vimentin (green), or F4/80 (green) in heart sections from mice with I/R injury. Colocalization regions are shown in yellow in merged images. J. MYSM1 mRNA of cardiomyocytes (CMs), fibroblasts (Fbs), macrophages (Macs), and endothelial cells (ECs) were detected by RT-qPCR in H/R injury (n = 8). (ns = no significance, * P < 0.05, ** P < 0.01, *** P < 0.001).
Figure 2. MYSM1 knockdown mice protect against myocardial I/R injury in vivo. A. The workflow of the Mysm1 − /+ mice subjected to acute I/R injury (30 min of ischemia and 4 h of reperfusion) or chronic I/R injury (30 min of ischemia and 2 weeks of reperfusion). B-D. The Myocardial infarction size determined by Evans blue (EB)/triphenyl tetrazolium chloride (TTC) double-staining and quantitative data for area at risk (AAR, C) and infarct size (INF, D, n = 7). E-G. The levels of serum lactic
Figure 4. The loss-of-function and gain-of-function of MYSM1 were evaluated in H/R-treated cardiomyocyte. A-D. MYSM1 knockdown (KD) HL-1 cells were constructed via shRNA (shMYSM1) before H/R injury (4 h of hypoxia and 6 h of reoxygenation). (A) Cell viability was assessed using the CCK8 assay (n = 6). (B) Bar graph showing the levels of LDH (n = 6). (C, D) The effect of MYSM1 knockdown on H/R-induced apoptosis in HL-1 determined by PI staining. Representative images for PI staining are shown (D), with quantitative columns for PI-positive cells (C, n = 6). E-H. MYSM1 was overexpressed by transfecting HL-1 cells with Flag-MYSM1 (oeMYSM1) before H/R injury. (E) Cell viability was assessed using the CCK8 assay (n = 6). (F) Bar graph showing the levels of LDH (n = 6). (G-H) The effect of oeMYSM1 on H/R-induced apoptosis in HL-1 determined by PI staining. Representative images for PI staining are shown (H), with quantitative columns for PI-positive cells (G, n = 6). I-L. MYSM1 was knocked down by siRNA transfection (siMYSM1) in NRPCs before H/R injury. (I) Cell viability was assessed using the CCK8 assay (n = 6). (J) Bar graph showing the levels of LDH (n = 6). (K-L) The effect of siMYSM1 on H/R-induced apoptosis in NRPCs was determined by PI staining. Representative images for PI staining are shown (L), with quantitative columns for PI-positive cells (K, n = 6). (* P < 0.05, ** P < 0.01, *** P < 0.001).
Figure 6. MYSM1 deubiquitinates STAT1 and maintains the stability of STAT1. A. NIH/3T3 cells were transfected with Flag-MYSM1 (1 or 2 μg) then analyzed by western blot for MYSM1, STAT1, p-STAT1 (Tyr701), and GAPDH (loading control). B. Representative RT-qPCR for Mysm1 and Stat1 in NIH/3T3 cells transfected with Flag-MYSM1 or EV plasmid (ns = no significance, *** P < 0.001, n = 7). C, D. NIH/3T3 cells were transfected with Flag-MYSM1 and EV plasmid, treated with CHX (25 μg/mL). (C) Western blot analysis for MYSM1, STAT1, and GAPDH (loading control). (D) Densitometric quantification of STAT1 (ns = no significance vs. Vector-0; *, vs. Vector-6; #, vs. Vector-9; ** P < 0.01, ## P < 0.01, n = 6). E. NIH/3T3 cells were co-transfected with Flag-MYSM1, Myc-STAT1, HA-Ub, HA-K48, and HA-K63, and treated with 10 μM MG132 for 4 h before collection. Cell lysates were treated with anti-Myc antibody. Ubiquitinated STAT1 was detected by western blot. F. Schematic of MYSM1 and its deletion mutants, including MYSM1(∆SANT), MYSM1(∆SWIRM), and MYSM1(∆MPN). G. NIH/3T3 cells were co-transfected with Myc-STAT1, Flag-MYSM1-WT or ∆MPN, ∆SWIRM and ∆SANT mutants, and treated with 10 μM MG132 for 4 h before collection. Cell lysates were treated with anti-Flag antibody to identify the binding region of MYSM1 regulating deubiquitination of STAT1. H. NIH/3T3 cells were co-transfected with Myc-STAT1, HA-Ub, Flag-MYSM1-WT or MPN mutants, and treated with 10 μM MG132 for 4 h before
Deubiquitinase MYSM1 drives myocardial ischemia/reperfusion injury by stabilizing STAT1 in cardiomyocytes
  • Article
  • Full-text available

January 2025

·

1 Read

Theranostics

Rationale: Myocardial ischemia/reperfusion (I/R) injury leads to irreversible cardiomyocyte death and aggravates myocardial infarction. Deubiquitinating enzymes (DUBs) are essential for maintaining substrate protein stability and functionality, playing significant roles in cardiac pathophysiology. In this study, we aimed to clarify the regulatory role of a DUB, Myb-like, SWIRM, and MPN domains 1 protein (MYSM1), in myocardial I/R injury and explore the molecular mechanism behind. Methods and Results: Firstly, it was found that the expression of MYSM1 positively correlates with myocardial I/R injury. Genetic knockdown of MYSM1 significantly conferred protection against I/R injury in hearts. Correspondingly, AAV9-mediated cardiomyocyte-specific knockdown of MYSM1 had a therapeutic effect on myocardial I/R injury. Through a comprehensive proteome-wide quantitative analysis, we identified signal transducer and activator of transcription 1 (STAT1) as the direct substrate of MYSM1. Mechanistically, MYSM1 mediated the K63-linked deubiquitination and stabilization of STAT1 at position K379 via its MPN metalloprotease domain. Additionally, MYSM1 initiates the expression of necroptosis-related genes by promoting the transcription factor function of STAT1. Conclusion: This study illustrated a MYSM1-STAT1 axis in regulating myocardial I/R injury and identified MYSM1 as a pharmacological target for myocardial I/R injury.

Download

Deubiquitinase MYSM1 promotes doxorubicin-induced cardiotoxicity by mediating TRIM21-ferroptosis axis in cardiomyocytes

December 2024

·

9 Reads

·

2 Citations

Cell Communication and Signaling

Anthracycline antitumor drug doxorubicin (DOX) induces severe cardiotoxicity. Deubiquitinating enzymes (DUBs) are crucial for protein stability and function and play a significant role in cardiac pathophysiology. By comparing RNA sequencing datasets and conducting functional screening, we determined that Myb-like, SWIRM, and MPN domains 1 (MYSM1) is a key regulator of DOX-induced cardiotoxicity. In this study, we aimed to explore the function and regulatory mechanisms of MYSM1 in DOX-induced cardiotoxicity. Genetic knockdown of MYSM1 significantly mitigated DOX-induced cardiomyopathy. Correspondingly, cardiomyocyte-specific knockdown of MYSM1 by AAV9 protected the heart from DOX-induced cardiotoxicity. Gain- and loss-of-function analysis verified that MYSM1 mediated DOX-induced cardiomyocyte injury in vitro. Through a Co-IP combined with LC-MS/MS analysis, we discovered that MYSM1 directly interacted with tripartite motif-containing protein 21 (TRIM21). Mechanistic investigations revealed that MYSM1 regulates the deubiquitination and the stability of TRIM21 via its MPN domain. Furthermore, MYSM1 exacerbated DOX-induced cardiotoxicity by enhancing ferroptosis. This study identified MYSM1 as a potential therapeutic target for DOX-induced cardiotoxicity and illustrated a MYSM1-TRIM21-ferroptosis axis in regulating DOX-induced cardiotoxicity. Supplementary Information The online version contains supplementary material available at 10.1186/s12964-024-01955-6.


Figure 1. The expression of USP28 is increased in cardiac hypertrophy and is principally distributed in cardiomyocytes. (A) Heatmap showing the mRNA profile of DUB genes in Ang II-induced mouse hypertrophic hearts from our published transcriptome (n = 3; GSE221396). (B) The mRNA expression of Usp28 in Ang II-or TACinduced mouse cardiac hypertrophy (n = 6; ** P < 0.01, *** P < 0.001). (C) Western blot and quantitation of USP28 protein expression in mouse hypertrophic myocardium (n = 6; *** P < 0.001). (D) The Usp28 mRNA level in human hypertrophic myocardium (NCH = non-cardiac hypertrophy, CH = cardiac hypertrophy; n = 4; *** P < 0.001). (E) scRNA-seq was performed in TAC mice hearts (For each group, single-cell suspensions from 3-4 hearts were pooled as 1 sample). Left, the UMAP dimensional reduction showing 5 main cell types of heart, including cardiomyocytes (CM), fibroblasts (FB), macrophages (MP), endothelial cells (EC) and pericytes (PC), and their specific marker genes. Approximate 17000 single heart cells (CM: 4484, EC: 3666, FB: 4320, MP: 2691, PC: 2154) were analyzed. Right, Biaxial scatter plot showing the expression pattern of Usp28. (F) Immunofluorescence staining of USP28 (red) and α-actin (green) in cardiac sections treated with Ang II (upper) or TAC (below). (G) NRPCs were transfected with siRNAs of NC (negative control) or USP28 followed by Ang II (1μM, 24h). TRITC-labeled rhodamine phalloidin staining (left) and the quantitative analysis (right) showed the surface area of NRPCs (n = 6; *** P < 0.001).
Figure 2. Cardiomyocyte-specific USP28 knockout ameliorates TAC-induced myocardial dysfunction and hypertrophy. USP28CKO and USP28 fl/fl mice were subjected to TAC or sham operations for 4 weeks. (A) M-mode echocardiographic images of left ventricle (LV) were assessed by non-invasive transthoracic echocardiography. (B) LV ejection fraction (EF) and fractional shortening (FS). (C) Serum ANP of indicated mice was tested by a mouse ANP ELISA Kit (F10062, Westang). (D-E) The ratio of heart weight (HW, mg) to body weight (BW, g) and representative gross-heart. (F) Representative H&E stained images of heart sections. (G-H) Representative WGA-stained sections and corresponding quantitative analysis of cardiomyocyte cross-sectional area. (I-J) The mRNA levels of Myh7 and Nppa in heart tissues of indicated mice. (K-L) Representative Masson's trichome-stained sections and corresponding quantitative analysis of fibrotic area. (n = 7 for each group; ** P < 0.01, *** P < 0.001)
Figure 3. Cardiomyocyte-specific USP28 deletion counteracts Ang II-induced hypertrophic heart failure. USP28CKO and USP28 fl/fl mice were subcutaneously implanted with osmotic mini-pump (Saline or Ang II (1000 ng/kg/min)) for 4 weeks. (A-B) M-mode echocardiographic images, EF and FS of left ventricle. (C) Serum ANP of indicated mice. (D-E) The ratio of HW (mg) to BW (g) and representative gross-heart. (F) Representative H&E stained images of heart sections. (G-H) Representative WGA-stained sections and corresponding quantitative analysis of cardiomyocyte cross-sectional area. (I-J) The mRNA levels of Myh7 and Nppa in heart tissues of indicated mice. (K-L) Representative Masson's trichome-stained sections and corresponding quantitative analysis of fibrotic area. (n = 7 for each group; ** P < 0.01, *** P < 0.001)
Figure 5. USP28 negatively regulates antioxidant response via its substrate TRIM21. (A) GSEA enrichment analysis of transcriptome in Ang II (1μM, 24h)-induced HL-1 cells expressing Flag-USP28 (USP28 OE ) or EV (NES: normalized enrichment score; FDR: false discovery rate). B-F: NRPCs were transfected with siRNAs of NC (negative
Figure 6. Selective USP28 inhibitor Otilonium Bromide (OB) abrogates TAC-induced hypertrophic heart failure and oxidative stress. (A) Mice of indicated group were underwent TAC for 6 weeks. Selective USP28 inhibitor OB (5 or 10 mg/kg/day) was intragastric administrated at the end of 2 nd week after TAC. All groups were harvested 6 weeks after TAC. (B) Serum ANP of indicated mice. (C-E) M-mode echocardiographic images, EF and FS of left ventricle. (F) The ratio of HW (mg) to BW (g). (G-H) Representative WGA-stained sections and corresponding quantitative analysis of cardiomyocyte cross-sectional area. (I-J) Representative gross-heart and H&E stained images of heart sections. (K-L) The mRNA levels of Myh7 and Nppa in heart tissues of indicated mice. (M-N) The mRNA levels of Nrf2 and Nqo1. (n = 7 for each group; ** P < 0.01, *** P < 0.001, ns = no significant)
Cardiomyocyte-derived USP28 negatively regulates antioxidant response and promotes cardiac hypertrophy via deubiquitinating TRIM21

September 2024

·

17 Reads

·

8 Citations

Theranostics

Rationale: Cardiac hypertrophy is an important pathological basis for heart failure. Most physiological activities of cardiomyocytes are regulated by proteins and their post-translational modification. Deubiquitinating enzymes (DUBs) are involved in protein stability maintenance and closely related to myocardial hypertrophy. In this study, we aimed to clarify the regulatory role of a DUB, ubiquitin-specific peptidase 28 (USP28), in cardiac hypertrophy and explore the molecular mechanism behind. Methods: Transcriptome and single-cell mRNA sequencing was used to demonstrate the association of USP28 and cardiac hypertrophy. Cardiomyocyte-specific USP28 knockout mice (USP28CKO) were subjected to angiotensin II (Ang II) infusion or transverse aortic constriction (TAC) models. Coimmunoprecipitation combined mass spectrum analysis (Co-IP/MS) was applied to screen out the substrate of USP28. Results: We first showed the up-regulation of USP28 in cardiac hypertrophy, and its cellular localization of cardiomyocytes. USP28CKO protects mouse heart against Ang II- or TAC-induced cardiac dysfunction and hypertrophy. Mechanistically, we identified tripartite motif-containing protein 21 (TRIM21) as the potential substrate of USP28 by Co-IP/MS analysis. Cardiomyocyte USP28 deubiquitinates and stabilizes TRIM21 to negatively regulate nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant response, increasing oxidative stress in cardiomyocytes and promoting cardiac hypertrophy and injury. Finally, using a selective USP28 inhibitor Otilonium Bromide, we confirmed the therapeutic effect of pharmacological inhibition of USP28 against TAC-induced established hypertrophic heart failure. Conclusion: Our study illustrates a cardiomyocyte-specific USP28-TRIM21 axis in regulating hypertrophic cardiomyopathy and presents USP28 as a potential target for the treatment of cardiac hypertrophy.


Podocyte OTUD5 alleviates diabetic kidney disease through deubiquitinating TAK1 and reducing podocyte inflammation and injury

June 2024

·

76 Reads

·

8 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.





Tussilagone attenuates atherosclerosis through inhibiting MAPKs-mediated inflammation in macrophages

April 2023

·

15 Reads

·

3 Citations

International Immunopharmacology

Atherosclerosis is a common chronic inflammatory disease. Recent studies have highlighted the key role of macrophages and inflammation in process of atherosclerotic lesion formation. A natural product, tussilagone (TUS), has previously exhibited anti-inflammatory activities in other diseases. In this study, we explored the potential effects and mechanisms of TUS on the inflammatory atherosclerosis. Atherosclerosis was induced in ApoE-/- mice by feeding them with a high-fat diet (HFD) for 8 weeks, followed by administration of TUS (10, 20 mg ·kg-1·d-1, i.g.) for 8 weeks. We demonstrated that TUS alleviated inflammatory response and reduced atherosclerotic plaque areas in HFD-fed ApoE-/- mice. Pro-inflammatory factor and adhesion factors were inhibited by TUS treatment. In vitro, TUS suppressed foam cell formation and oxLDL-induced inflammatory response in MPMs. RNA-sequencing analysis indicated that MAPK pathway was related to the anti-inflammation and anti-atherosclerosis effects of TUS. We further confirmed that TUS inhibited MAPKs phosphorylation in plaque lesion of aortas and cultured macrophages. MAPK inhibition blocked oxLDL-induced inflammatory response and prevented the innately pharmacological effects of TUS. Our findings present a mechanistic explanation for the pharmacological effect of TUS against atherosclerosis and indicate TUS as a potentially therapeutic candidate for atherosclerosis.


Macrophage DCLK1 promotes atherosclerosis via binding to IKKβ and inducing inflammatory responses

March 2023

·

30 Reads

·

27 Citations

EMBO Molecular Medicine

Atherosclerosis is a chronic inflammatory disease with high morbidity and mortality rates worldwide. Doublecortin-like kinase 1 (DCLK1), a microtubule-associated protein kinase, is involved in neurogenesis and human cancers. However, the role of DCLK1 in atherosclerosis remains undefined. In this study, we identified upregulated DCLK1 in macrophages in atherosclerotic lesions of ApoE-/- mice fed an HFD and determined that macrophage-specific DCLK1 deletion attenuates atherosclerosis by reducing inflammation in mice. Mechanistically, RNA sequencing analysis indicated that DCLK1 mediates oxLDL-induced inflammation via NF-κB signaling pathway in primary macrophages. Coimmunoprecipitation followed by LC-MS/MS analysis identified IKKβ as a binding protein of DCLK1. We confirmed that DCLK1 directly interacts with IKKβ and phosphorylates IKKβ at S177/181, thereby facilitating subsequent NF-κB activation and inflammatory gene expression in macrophages. Finally, a pharmacological inhibitor of DCLK1 prevents atherosclerotic progression and inflammation both in vitro and in vivo. Our findings demonstrated that macrophage DCLK1 promotes inflammatory atherosclerosis by binding to IKKβ and activating IKKβ/NF-κB. This study reports DCLK1 as a new IKKβ regulator in inflammation and a potential therapeutic target for inflammatory atherosclerosis.

Citations (5)


... Besides cancer, the regulation of ubiquitination on ferroptosis also plays a role in the treatment of other diseases. As a deubiquitinating enzyme, mysm1 regulates trim 21 stability and mediates doxorubicin (DOX) -induced ferroptosis in cardiomyocytes, suggesting that cardiomyocyte specific knockdown of mysm1 may be an attractive therapeutic strategy for this disease (82). The latest research found that Hyperoside, a bioactive compound extracted from traditional Chinese medicine, promotes the translocation of Nrf2 to the nucleus by inhibiting ubiquitin mediated degradation of Nrf2, resulting in the up regulation of the expression of anti ferroptosis genes, and inhibiting ferroptosis in the treatment of sepsis induced acute lung injury (83). ...

Reference:

Research progress of ferroptosis pathway and its related molecular ubiquitination modification in liver cancer
Deubiquitinase MYSM1 promotes doxorubicin-induced cardiotoxicity by mediating TRIM21-ferroptosis axis in cardiomyocytes

Cell Communication and Signaling

... USP25 is elevated in hypertrophic myocardium, where it stabilizes SERCA2a by removing K48-linked ubiquitin chains, thereby inhibiting cardiac hypertrophy [44]. Additionally, USP28 and UCHL1 expression increases in hypertrophic and failing hearts [47,48]. OTUD7B has been reported to regulate antiviral immunity [16], inflammation [49], and cancer cell proliferation [14]. ...

Cardiomyocyte-derived USP28 negatively regulates antioxidant response and promotes cardiac hypertrophy via deubiquitinating TRIM21

Theranostics

... As a post-translational modification, ubiquitination/ deubiquitination critically regulates protein stability and function, and is involved in the pathophysiology of many diseases [12]. Deubiquitinases (DUBs) counterbalance ubiquitination by removing ubiquitin moieties from target proteins, thereby modulating their function and stability [13]. Ubiquitin C-terminal hydrolase (UCH) L3 (UCHL3), a member of the UCH family of DUBs, has substrates, including Forkhead box protein M1 (FOXM1), Lactate dehydrogenase A (LDHA), and TNF receptor-associated factor 2 (TRAF2), which it deubiquitinates to modulate their activity [14]. ...

Podocyte OTUD5 alleviates diabetic kidney disease through deubiquitinating TAK1 and reducing podocyte inflammation and injury

... 5,6 The GSDMD-N protein undergoes oligomerization on the cell membrane, creating pores that facilitate the release of inflammatory cytokines from the cytoplasm, thereby intensifying the inflammatory response. 5,6 Our previous studies showed that the key role of GSDMD in a variety of CVDs, including septic myocardial dysfunction, 7 doxorubicin-induced cardiotoxicity, 8 cardiac hypertrophy, 9,10 vascular remodelling, 11 abdominal aortic aneurysm 12 and arteriosclerosis. 13 Numerous studies have reported that GSDMD is involved in the formation of atherosclerotic plaque. ...

Macrophage-derived GSDMD promotes abdominal aortic aneurysm and aortic smooth muscle cells pyroptosis
  • Citing Article
  • January 2024

International Immunopharmacology

... Macrophages determine the development of atherosclerotic lesions [5,44,45]. As reported by Luo et al. 's findings, quercetin inhibits senescence of plaque macrophages driven by ox-LDL and alleviates AS through suppressing p38 MAPK/p16 signaling pathway [31]. ...

Macrophage DCLK1 promotes atherosclerosis via binding to IKKβ and inducing inflammatory responses
  • Citing Article
  • March 2023

EMBO Molecular Medicine