1
|
Yang P, Gu H, Wu X, Chen G, Liu H, Chen Z. Tumour protein p53-activated lncRNA PGM5-AS1 suppresses lung cancer growth and stemness by targeting R-spondin1 via microRNA-1247-5p. Arch Physiol Biochem 2025:1-13. [PMID: 40035308 DOI: 10.1080/13813455.2025.2459318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/06/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025]
Abstract
OBJECTIVE This study was to investigated the inhibitory role of the tumour protein p53 (TP53)-activated PGM5-AS1 in lung cancer (LC) cell proliferation, invasion, and CSC-like properties and its underlying mechanisms. METHODS The effect of PGM5-AS1 on LC cell development was determined. Stem cell markers, aldehyde dehydrogenase activity in cells were tested, as well as the ability of stem cells to form spheroids. The interaction of PGM5-AS1 and TP53 was determined. The binding link of PGM5-AS1, miR-1247-5p, and R-spondin1 (RSPO1) was verified. RESULTS PGM5-AS1 was elevated by a combination of TP53 and PGM5-AS1 promoters. PGM5-AS1 was a molecular sponge of miR-1247-5p in LC cells, and miR-1247-5p targeted RSPO1. Elevating PGM5-AS1 or repressing miR-1247-5p restrained LC cell growth and stemness, which were reversed by depression of RSPO1. CONCLUSION This study conveys that TP53-elevated PGM5-AS1 mediates miR-1247-5p to target RSPO1, thereby inhibiting LC growth and stemness, representing a novel avenue for LC therapy.
Collapse
Affiliation(s)
- Peng Yang
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Hong Gu
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Xuanqin Wu
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Geng Chen
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Heng Liu
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| | - Zhongliang Chen
- Department of Cardiothoracic and Vascular Surgery, Shenzhen Guangming District People's Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
2
|
Chen Y, Xue C. Cross-talk of renal cells through WNT signal transduction in the development of fibrotic kidneys. Front Cell Dev Biol 2025; 12:1517181. [PMID: 40012992 PMCID: PMC11860889 DOI: 10.3389/fcell.2024.1517181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/16/2024] [Indexed: 02/28/2025] Open
Abstract
Chronic kidney disease (CKD) is a progressive condition that can lead to chronic renal failure (CRF), affecting 8%-16% of adults globally and imposing a significant burden on healthcare systems. Renal fibrosis is a key pathological hallmark of CKD progression and is linked to poor prognosis. Multiple signaling pathways, including WNT/β-catenin.Aberrant activation of WNT/β-catenin is implicated in renal fibrosis. The roles of renal macrophages and fibroblasts are pivotal in fibrosis progression and prognosis.
Collapse
Affiliation(s)
| | - Chao Xue
- Department of Nephrology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
3
|
Cheng X, Liu J, Niu D, Zhang C, Lin Y, Li S, Yang J, Wen J. Prognostic prediction and diagnostic role of Rspondin 1 expression in esophageal squamous cell carcinoma. INDIAN J PATHOL MICR 2025; 68:11-16. [PMID: 38864442 DOI: 10.4103/ijpm.ijpm_452_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 12/14/2023] [Indexed: 06/13/2024] Open
Abstract
CONTEXT Rspondin 1 (Rspo1), a protein family member featuring secreted furin-like domains, plays a pivotal role in cancer development and exhibits a positive correlation with tumor progression. However, its expression in esophageal squamous cell carcinoma (ESCC) is still unknown. AIMS Here, we assessed the correlation between Rspo1 and clinicopathological features of ESCC patients, and further investigated the potential role of Rspo1 in ESCC development and clinical outcomes. SETTINGS AND DESIGN This was a pilot study. MATERIALS AND METHODS A total of 112 paraffin-embedded tumor samples from patients with ESCC, including 68 matched adjacent normal tissues, were collected post-surgery. Subsequently, tissue microarray (TMA) and immunohistochemistry (IHC) techniques were employed to assess the protein levels of Rspo1. STATISTICAL ANALYSIS All statistical analyses were performed with SPSS 20.0 (SPSS, Inc., Chicago, IL). RESULTS We found that Rspo1 expression was significantly higher in ESCC than in adjacent normal tissues ( P < 0.0001). Moreover, Rspo1 was highly expressed in ESCC tumor specimens and showed a significant correlation with the T classification of ESCC ( P < 0.05). Additionally, our findings indicate a positive relationship between Rspo1 and survival time in ESCC. Patients exhibiting moderate to high levels of Rspo1 expression demonstrated superior survival outcomes compared to those with low expression ( P = 0.0002). CONCLUSIONS Our investigation has demonstrated that Rspo1 is upregulated in ESCC and exhibits a positive correlation with disease progression. Furthermore, we have observed a significant association between Rspo1 overexpression and improved patient survival rates, indicating its potential as a prognostic marker and therapeutic target for ESCC treatment.
Collapse
Affiliation(s)
- Xiaoxia Cheng
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Jiao Liu
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Danye Niu
- Department of Clinical Nutrition, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu, China
| | - Changsong Zhang
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Yuansheng Lin
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Shengjun Li
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Jiao Yang
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| | - Jiangtao Wen
- Suzhou Hospital, Affiliated Hospital of Medical School, Nanjing University, Suzhou, Jiangsu, China
| |
Collapse
|
4
|
Jensen M, Heinl ES, Federlein A, Schwartz U, Lund L, Madsen K, Jensen BL, Schweda F. Identification of natriuretic peptide receptor A-related gene expression signatures in podocytes in vivo reveals baseline control of protective pathways. Am J Physiol Renal Physiol 2024; 327:F806-F821. [PMID: 39298549 DOI: 10.1152/ajprenal.00394.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 08/26/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024] Open
Abstract
Natriuretic peptide receptor-A (NPR-A) is the principal receptor for the natriuretic peptides atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). Targeted deletion of NPR-A in mouse glomerular podocytes significantly enhances renal injury in vivo in the DOCA-salt experimental model. It was therefore hypothesized that natriuretic peptides exert a direct protective effect on glomerular barrier integrity through activation of NPR-A and modulation of gene expression patterns in podocytes. Green fluorescence-positive podocytes from mice with a conditional deletion of Npr1 encoding NPR-A were isolated by fluorescence-activated cell sorting (FACS). Differentially expressed genes (DEGs) in podocytes were identified by RNA sequencing of podocytes from wild-type and NPR-A-deleted mice. Enrichment analysis was performed on the DEGs using Gene Ontology (GO) terms. Identified transcripts were validated by real-time PCR and ELISA of cultured isolated human and mouse glomeruli. In addition, the effect of natriuretic peptides on podocyte migration was investigated by measuring the outgrowth of podocytes from cultured glomeruli. A total of 158 DEGs were identified with 81 downregulated DEGs and 77 upregulated DEGs in Npr1-deficient podocytes. Among the downregulated genes were protein S and semaphorin 3G, which are known to have protective effects in podocytes. Protein S was also expressed in and secreted from isolated human glomeruli. GO enrichment analysis revealed that the upregulated DEGs in NPR-A deficient podocytes were associated with cell migration and motility. In line, BNP significantly decreased podocyte outgrowth from cultured glomeruli. In conclusion, endogenous levels of natriuretic peptides in mice support baseline protective pathways at glomerular podocytes such as protein S and suppress podocyte migration.NEW & NOTEWORTHY A combination of fluorescence-activated cell sorting and RNA sequencing identified 158 changed gene products in adult mouse kidneys with and without podocyte-specific deletion of the natriuretic peptide receptor A. Downregulated products included protein S and semaphorin 3G, both with proven renoprotective impact, whereas upregulated products were related to mobility of podocytes. Protein S was produced and released from human and murine isolated glomeruli, and atrial natriuretic peptide (ANP) led to decreased migration of podocytes.
Collapse
Affiliation(s)
- Mia Jensen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Elena-Sofia Heinl
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Anna Federlein
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Uwe Schwartz
- NGS Analysis Center, Biology and Pre-Clinical Medicine, University of Regensburg, Regensburg, Germany
| | - Lars Lund
- Department of Urology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kirsten Madsen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Boye L Jensen
- Unit of Cardiovascular and Renal Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Frank Schweda
- Institute of Physiology, University of Regensburg, Regensburg, Germany
| |
Collapse
|
5
|
Luo Q, Dai T, Dong Y, Liang J, Xu Z, Sun Z. N6-methyladenosine-modified TRIM37 augments sunitinib resistance by promoting the ubiquitin-degradation of SmARCC2 and activating the Wnt signaling pathway in renal cell carcinoma. Cell Death Discov 2024; 10:418. [PMID: 39349442 PMCID: PMC11442835 DOI: 10.1038/s41420-024-02187-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/05/2024] [Accepted: 09/19/2024] [Indexed: 10/02/2024] Open
Abstract
Tripartite motif-containing 37 (TRIM37) is reportedly a key member of the superfamily of TRIM proteins. Emerging evidence underscores the close association between dysregulated TRIM37 expression and the progression of various human malignancies. However, the precise biological functions and regulatory mechanisms of TRIM37 remain elusive. This study aimed to elucidate the impact of TRIM37 on the chemotherapy sensitivity of renal cell carcinoma (RCC) and uncover its specific molecular regulatory role. Using RT-qPCR and western blot assays, we assessed TRIM37 expression in both RCC patients and RCC cells. Through in vitro and in vivo experiments, we investigated the effects of TRIM37 silencing and overexpression on RCC cell proliferation, stemness capacity, and chemotherapy sensitivity using colony formation and sphere formation assays. Additionally, a co-immunoprecipitation (Co-IP) experiment was conducted to explore putative interacting proteins. Our results revealed elevated TRIM37 expression in both RCC patient tumor tissues and RCC cells. Functional experiments consistently demonstrated that TRIM37 silencing reduced proliferation and stemness capacity while enhancing chemotherapy sensitivity in RCC cells. Furthermore, we discovered that TRIM37 mediates the degradation of SMARCC2 via ubiquitin-proteasome pathways, thereby further activating the Wnt signaling pathway. In conclusion, this study not only sheds light on the biological role of TRIM37 in RCC progression but also identifies a potential molecular target for therapeutic intervention in RCC patients.
Collapse
Affiliation(s)
- Qiang Luo
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Ting Dai
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, 130000, Jilin, China
| | - Yihong Dong
- Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Jianpeng Liang
- The First Peoples Hospital of Foshan, Foshan, 528000, Guangdong, China
| | - Zhipeng Xu
- Third Xiangya Hospital of Central South University, Changsha, 410013, Hunan, China
| | - Zhixia Sun
- Department of Ultrasound, China-Japan Union Hospital of Jilin University, Changchun, 130000, Jilin, China.
| |
Collapse
|
6
|
Wang Y, Liu Y, Cai J, Zong T, Zhang Z, Xie T, Mu T, Wu M, Yang Q, Wang Y, Wang X, Yao Y. Differentially expressed genes in orbital adipose/connective tissue of thyroid-associated orbitopathy. PeerJ 2023; 11:e16569. [PMID: 38130930 PMCID: PMC10734407 DOI: 10.7717/peerj.16569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 11/13/2023] [Indexed: 12/23/2023] Open
Abstract
Background Thyroid-associated orbitopathy (TAO) is a disease associated with autoimmune thyroid disorders and it can lead to proptosis, diplopia, and vision-threatening compressive optic neuropathy. To comprehensively understand the molecular mechanisms underlying orbital adipogenesis in TAO, we characterize the intrinsic molecular properties of orbital adipose/connective tissue from patients with TAO and control individuals. Methods RNA sequencing analysis (RNA-seq) was performed to measure the gene expression of orbital adipose/connective tissues of TAO patients. Differentially expressed genes (DEGs) were detected and analyzed through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, and Gene Set Enrichment Analysis (GSEA). The protein-protein interaction (PPI) network was constructed using the STRING database, and hub genes were identified by the Cytoscape plug-in, cytoHubba. We validated several top DEGs through quantitative real-time polymerase chain reaction (qRT-PCR). Results We identified 183 DEGs in adipose tissue between TAO patients (n = 3) and control patients (n = 3) through RNA sequencing, including 114 upregulated genes and 69 downregulated genes. The PPI network of these DEGs had 202 nodes and 743 edges. PCR-based validation results of orbital adipose tissue showed multiple top-ranked genes in TAO patients (n = 4) are immune and inflammatory response genes compared with the control individual (n = 4). They include ceruloplasmin isoform x3 (CP), alkaline tissue-nonspecific isozyme isoform x1 (ALPL), and angiotensinogen (AGT), which were overrepresented by 2.27- to 6.40-fold. Meanwhile, protein mab-21-like 1 (MAB21L1), phosphoinositide 3-kinase gamma-subunit (PIK3C2G), and clavesin-2 (CLVS2) decreased by 2.6% to 32.8%. R-spondin 1 (RSPO1), which is related to oogonia differentiation and developmental angiogenesis, was significantly downregulated in the orbital muscle tissues of patients with TAO compared with the control groups (P = 0.024). Conclusions Our results suggest that there are genetic differences in orbital adipose-connective tissues derived from TAO patients. The upregulation of the inflammatory response in orbital fat of TAO may be consistent with the clinical phenotype like eyelid edema, exophthalmos, and excess tearing. Downregulation of MAB21L1, PIK3C2G, and CLVS2 in TAO tissue demonstrates dysregulation of differentiation, oxidative stress, and developmental pathways.
Collapse
Affiliation(s)
- Yan Wang
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yanqiu Liu
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Jiping Cai
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Tianyi Zong
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Ziyin Zhang
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Tianhua Xie
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Tong Mu
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Meili Wu
- Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Qian Yang
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
- Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yangningzhi Wang
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Xiaolu Wang
- Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Yong Yao
- Department of Ophthalmology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| |
Collapse
|
7
|
Hu L, Ding M, He W. Emerging Therapeutic Strategies for Attenuating Tubular EMT and Kidney Fibrosis by Targeting Wnt/β-Catenin Signaling. Front Pharmacol 2022; 12:830340. [PMID: 35082683 PMCID: PMC8784548 DOI: 10.3389/fphar.2021.830340] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022] Open
Abstract
Epithelial-mesenchymal transition (EMT) is defined as a process in which differentiated epithelial cells undergo phenotypic transformation into myofibroblasts capable of producing extracellular matrix, and is generally regarded as an integral part of fibrogenesis after tissue injury. Although there is evidence that the complete EMT of tubular epithelial cells (TECs) is not a major contributor to interstitial myofibroblasts in kidney fibrosis, the partial EMT, a status that damaged TECs remain inside tubules, and co-express both epithelial and mesenchymal markers, has been demonstrated to be a crucial stage for intensifying fibrogenesis in the interstitium. The process of tubular EMT is governed by multiple intracellular pathways, among which Wnt/β-catenin signaling is considered to be essential mainly because it controls the transcriptome associated with EMT, making it a potential therapeutic target against kidney fibrosis. A growing body of data suggest that reducing the hyperactivity of Wnt/β-catenin by natural compounds, specific inhibitors, or manipulation of genes expression attenuates tubular EMT, and interstitial fibrogenesis in the TECs cultured under profibrotic environments and in animal models of kidney fibrosis. These emerging therapeutic strategies in basic researches may provide beneficial ideas for clinical prevention and treatment of chronic kidney disease.
Collapse
Affiliation(s)
- Lichao Hu
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Mengyuan Ding
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| | - Weichun He
- Center for Kidney Disease, Second Affiliated Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
8
|
Yang L, Wang J, Gong X, Fan Q, Yang X, Cui Y, Gao X, Li L, Sun X, Li Y, Wang Y. Emerging Roles for LGR4 in Organ Development, Energy Metabolism and Carcinogenesis. Front Genet 2022; 12:728827. [PMID: 35140734 PMCID: PMC8819683 DOI: 10.3389/fgene.2021.728827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 12/30/2021] [Indexed: 11/26/2022] Open
Abstract
The leucine-rich repeats containing G protein-coupled receptor 4 (LGR4) belonging to G protein-coupled receptors (GPCRs) family, had various regulatory roles at multiple cellular types and numerous targeting sites, and aberrant LGR4 signaling played crucial roles in diseases and carcinogenesis. On the basis of these facts, LGR4 may become an appealing therapeutic target for the treatment of diseases and tumors. However, a comprehensive investigation of its functions and applications was still lacking. Hence, this paper provided an overview of the molecular characteristics and signaling mechanisms of LGR4, its involvement in multiple organ development and participation in the modulation of immunology related diseases, metabolic diseases, and oxidative stress damage along with cancer progression. Given that GPCRs accounted for almost a third of current clinical drug targets, the in-depth understanding of the sophisticated connections of LGR4 and its ligands would not only enrich their regulatory networks, but also shed new light on designing novel molecular targeted drugs and small molecule blockers for revolutionizing the treatment of various diseases and tumors.
Collapse
Affiliation(s)
- Linlin Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Jing Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaodi Gong
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Qiong Fan
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoming Yang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yunxia Cui
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiaoyan Gao
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Lijuan Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Xiao Sun
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
| | - Yuhong Li
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| | - Yudong Wang
- Department of Gynecological Oncology, The International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Municipal Key Clinical Specialty, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Disease, Shanghai, China
- *Correspondence: Yuhong Li, ; Yudong Wang,
| |
Collapse
|
9
|
Zhu B, Liu Y, Qi D, Zhao L, Yang X, Su E, Zhu Z, Zhang Y, Gao C. Renal interstitial fibrosis is reduced in high-fat diet-induced obese pigs following renal denervation from the intima and adventitia of the renal artery. Kidney Blood Press Res 2021; 47:135-146. [PMID: 34852339 DOI: 10.1159/000521100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/22/2021] [Indexed: 11/19/2022] Open
Abstract
Background This study aims to compare whether two different routes of Renal denervation (RDN) from the intima and adventitia of the renal artery can reduce renal fibrosis in a pig model of hypertension induced by a high-fat diet and to explore possible molecular mechanisms. Methods Twenty-four Bama miniature pigs were randomly divided into a control group (normal diet, n=6) or a hypertension model group (high-fat diet, n=18). The model group was randomly divided into the intima-RDN group (n=6), the adventitia-RDN group (n=6), or the renal arteriography only group (sham group, n=6). All animals were fed separately. The model group was fed a high-fat diet after the operation, and the control group was fed conventionally for 6 months. After 6 months, renal artery angiography was performed again to observe the condition of the renal arteries, after which all animals were euthanized. The blood pressure (BP) and blood biochemical results of each group were evaluated 6 months after the operation; kidney tissue morphology and collagen fiber content were examined by hematoxylin-eosin (HE) staining and Masson staining; Superoxide dismutase (SOD) activity and the malondialdehyde (MDA) content of kidney tissue were assessed by a biochemical enzyme method; the protein expression level of transforming growth factor-β 1 (TGF-β1), α smooth muscle actin (αSMA) and Smad3 were assessed by Western blot; and electron microscopy was used to examine changes in kidney microstructure. Results After 6 months of a high-fat diet, the blood lipid levels of the model group were significantly higher compared to baseline and to that of the control group during the same period (all showed P<0.05); the blood lipid levels of the control group did not change significantly from baseline (P>0.05). The degree of glomerular damage caused by hyperlipidemia in the intima-RDN group and the adventitia-RDN group was significantly lower than that of the sham and control groups, and the renal fibrosis area percentage was also significantly lower (P<0.05). Electron microscopy showed that both the intima-RDN group and the adventitia-RDN group had a more even distribution of chromosomes and less mitochondrial swelling compared with the sham group. Conclusion RDN from the adventitia of the renal artery and RDN from the intima of the renal artery have the similar advantages of delaying high fat-induced renal fibrosis. The anti-fibrotic effect of RDN may be related to inhibition of the TGF-β1/smad3 pathway.
Collapse
Affiliation(s)
- Binbin Zhu
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Yahui Liu
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Datun Qi
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Linwei Zhao
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Xiaohang Yang
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Enyong Su
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Zhongyu Zhu
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - You Zhang
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| | - Chuanyu Gao
- Department of Cardiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Cardiology, Henan Provincial Key Lab for Control of Coronary Heart Disease, Central China Fuwai Hospital, Zhengzhou, China
| |
Collapse
|