1
|
Zheng BW, Guo W. Multi-omics analysis unveils the role of inflammatory cancer-associated fibroblasts in chordoma progression. J Pathol 2025; 265:69-83. [PMID: 39611243 DOI: 10.1002/path.6369] [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: 07/17/2024] [Revised: 09/25/2024] [Accepted: 10/13/2024] [Indexed: 11/30/2024]
Abstract
Cancer-associated fibroblasts (CAFs) constitute the primary cellular component of the stroma in chordomas, characterized by an abundance of mucinous stromal elements, potentially facilitating their initiation and progression; however, this inference has yet to be fully confirmed. In this study, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics (ST), bulk RNA-seq, multiplexed quantitative immunofluorescence (QIF), and in vivo and in vitro experiments were performed to determine the heterogeneity, spatial distribution, and clinical significance of CAFs in chordoma. ScRNA-seq was performed on 87,693 single cells derived from seven tumor samples and four control nucleus pulposus samples. A distinct CAF cluster distinguished by the upregulated expression of inflammatory genes and enriched functionality in activating inflammation-associated cells was identified. Pseudotime trajectory and cell communication analyses suggested that this inflammatory CAF (iCAF) subset originated from normal fibroblasts and interacted extensively with tumors and various other cell types. By integrating the scRNA-seq results with ST, the presence of iCAF in chordoma tissue was further confirmed, indicating their positioning at a distance from the tumor cells. Bulk RNA-seq data analysis from 126 patients revealed a correlation between iCAF signature scores, chordoma invasiveness, and poor prognosis. QIF validation involving an additional 116 patients found that although iCAFs were not in close proximity to tumor cells compared with other CAF subsets, their density correlated with malignant tumor phenotypes and adverse outcomes. In vivo and in vitro experiments further confirmed that iCAFs accelerate the malignant progression of chordomas. These findings could provide insights into the development of novel therapeutic strategies. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Bo-Wen Zheng
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| | - Wei Guo
- Department of Musculoskeletal Tumor, Peking University People's Hospital, Peking University, Beijing, PR China
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People's Hospital, Beijing, PR China
| |
Collapse
|
2
|
Wang P, Chen W, li B, Yang S, Li W, Zhao S, Ning J, Zhou X, Cheng F. Exosomes on the development and progression of renal fibrosis. Cell Prolif 2024; 57:e13677. [PMID: 38898750 PMCID: PMC11533081 DOI: 10.1111/cpr.13677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/09/2024] [Accepted: 05/14/2024] [Indexed: 06/21/2024] Open
Abstract
Renal fibrosis is a prevalent pathological alteration that occurs throughout the progression of primary and secondary renal disorders towards end-stage renal disease. As a complex and irreversible pathophysiological phenomenon, it includes a sequence of intricate regulatory processes at the molecular and cellular levels. Exosomes are a distinct category of extracellular vesicles that play a crucial role in facilitating intercellular communication. Multiple pathways are regulated by exosomes produced by various cell types, including tubular epithelial cells and mesenchymal stem cells, in the context of renal fibrosis. Furthermore, research has shown that exosomes present in bodily fluids, including urine and blood, may be indicators of renal fibrosis. However, the regulatory mechanism of exosomes in renal fibrosis has not been fully elucidated. This article reviewed and analysed the various mechanisms by which exosomes regulate renal fibrosis, which may provide new ideas for further study of the pathophysiological process of renal fibrosis and targeted treatment of renal fibrosis with exosomes.
Collapse
Affiliation(s)
- Peihan Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Wu Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Bojun li
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Songyuan Yang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Wei Li
- Department of AnesthesiologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Sheng Zhao
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Jinzhuo Ning
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Xiangjun Zhou
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| | - Fan Cheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiP.R. China
| |
Collapse
|
3
|
Kwon S, Cheon S, Kim KH, Seo A, Bae E, Lee JW, Cha RH, Hwang JH, Kim YC, Kim DK, Kim YS, Han D, Yang SH. Unveiling the role of transgelin as a prognostic and therapeutic target in kidney fibrosis via a proteomic approach. Exp Mol Med 2024; 56:2296-2308. [PMID: 39375532 PMCID: PMC11542076 DOI: 10.1038/s12276-024-01319-7] [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: 12/06/2023] [Revised: 06/20/2024] [Accepted: 07/11/2024] [Indexed: 10/09/2024] Open
Abstract
Chronic kidney disease (CKD) progression involves tubulointerstitial fibrosis, a process characterized by excessive extracellular matrix accumulation. To identify potential biomarkers for kidney fibrosis, we performed mass spectrometry-based proteomic profiling of human kidney tubular epithelial cells and kidney tissue from a 5/6 nephrectomy rat model. Multidisciplinary analysis across kidney fibrosis models revealed 351 differentially expressed proteins associated with kidney fibrosis, and they were enriched in processes related to the extracellular matrix, kidney aging, and mitochondrial functions. Network analysis of the selected proteins revealed five crucial proteins, of which transgelin emerged as a candidate protein that interacts with known fibrosis-related proteins. Concordantly, the gene expression of transgelin in the kidney tissue from the 5/6 nephrectomy model was elevated. Transgelin expression in kidney tissue gradually increased from intermediate to advanced fibrosis stages in 5/6 Nx rats and mice with unilateral ureteral obstruction. Subsequent validation in kidney tissue and urine samples from patients with CKD confirmed the upregulation of transgelin, particularly under advanced disease stages. Moreover, we investigated whether blocking TAGLN ameliorated kidney fibrosis and reduced reactive oxygen species levels in cellular models. In conclusion, our proteomic approach identified TAGLN as a potential noninvasive biomarker and therapeutic target for CKD-associated kidney fibrosis, suggesting its role in modulating mitochondrial dysfunction and oxidative stress responses.
Collapse
Affiliation(s)
- Soie Kwon
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
- Department of Clinical Medical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Seongmin Cheon
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, Republic of Korea
| | - Kyu-Hong Kim
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Areum Seo
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Eunjin Bae
- Department of Internal Medicine, Gyeongsang National University College of Medicine, Gyeongsang University Changwon Hospital, Gyeongsang, Republic of Korea
| | - Jae Wook Lee
- Nephrology Clinic, National Cancer Center of Korea, Seoul, Republic of Korea
| | - Ran-Hui Cha
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
| | - Jin Ho Hwang
- Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, College of Medicine, Chung-Ang University, Seoul, Republic of Korea
| | - Yong Chul Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Dong Ki Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Yon Su Kim
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Transdisciplinary Medicine, Seoul National University Hospital, Seoul, Republic of Korea.
- Department of Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Seung-Hee Yang
- Kidney Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
- Biomedical Research Institute, Seoul National University Hospital, Seoul, Republic of Korea.
| |
Collapse
|
4
|
Lahane GP, Dhar A, Bhat A. Therapeutic approaches and novel antifibrotic agents in renal fibrosis: A comprehensive review. J Biochem Mol Toxicol 2024; 38:e23795. [PMID: 39132761 DOI: 10.1002/jbt.23795] [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: 01/25/2024] [Revised: 06/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
Renal fibrosis (RF) is one of the underlying pathological conditions leading to progressive loss of renal function and end-stage renal disease (ESRD). Over the years, various therapeutic approaches have been explored to combat RF and prevent ESRD. Despite significant advances in understanding the underlying molecular mechanism(s), effective therapeutic interventions for RF are limited. Current therapeutic strategies primarily target these underlying mechanisms to halt or reverse fibrotic progression. Inhibition of transforming growth factor-β (TGF-β) signaling, a pivotal mediator of RF has emerged as a central strategy to manage RF. Small molecules, peptides, and monoclonal antibodies that target TGF-β receptors or downstream effectors have demonstrated potential in preclinical models. Modulating the renin-angiotensin system and targeting the endothelin system also provide established approaches for controlling fibrosis-related hemodynamic changes. Complementary to pharmacological strategies, lifestyle modifications, and dietary interventions contribute to holistic management. This comprehensive review aims to summarize the underlying mechanisms of RF and provide an overview of the therapeutic strategies and novel antifibrotic agents that hold promise in its treatment.
Collapse
Affiliation(s)
- Ganesh Panditrao Lahane
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Arti Dhar
- Department of Pharmacy, Birla Institute of Technology and Sciences (BITS) Pilani, Hyderabad, Telangana, India
| | - Audesh Bhat
- Centre for Molecular Biology, Central University of Jammu, Samba, Jammu and Kashmir, India
| |
Collapse
|
5
|
Omer MH, Shafqat A, Ahmad O, Nadri J, AlKattan K, Yaqinuddin A. Urinary Biomarkers for Lupus Nephritis: A Systems Biology Approach. J Clin Med 2024; 13:2339. [PMID: 38673612 PMCID: PMC11051403 DOI: 10.3390/jcm13082339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/12/2024] [Accepted: 04/13/2024] [Indexed: 04/28/2024] Open
Abstract
Systemic lupus erythematosus (SLE) is the prototypical systemic autoimmune disorder. Kidney involvement, termed lupus nephritis (LN), is seen in 40-60% of patients with systemic lupus erythematosus (SLE). After the diagnosis, serial measurement of proteinuria is the most common method of monitoring treatment response and progression. However, present treatments for LN-corticosteroids and immunosuppressants-target inflammation, not proteinuria. Furthermore, subclinical renal inflammation can persist despite improving proteinuria. Serial kidney biopsies-the gold standard for disease monitoring-are also not feasible due to their inherent risk of complications. Biomarkers that reflect the underlying renal inflammatory process and better predict LN progression and treatment response are urgently needed. Urinary biomarkers are particularly relevant as they can be measured non-invasively and may better reflect the compartmentalized renal response in LN, unlike serum studies that are non-specific to the kidney. The past decade has overseen a boom in applying cutting-edge technologies to dissect the pathogenesis of diseases at the molecular and cellular levels. Using these technologies in LN is beginning to reveal novel disease biomarkers and therapeutic targets for LN, potentially improving patient outcomes if successfully translated to clinical practice.
Collapse
Affiliation(s)
- Mohamed H. Omer
- School of Medicine, Cardiff University, Cardiff CF14 4YS, UK;
| | - Areez Shafqat
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Omar Ahmad
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Juzer Nadri
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Khaled AlKattan
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| | - Ahmed Yaqinuddin
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (O.A.); (J.N.); (K.A.); (A.Y.)
| |
Collapse
|
6
|
Rajabi S, Saberi S, Najafipour H, Askaripour M, Rajizadeh MA, Shahraki S, Kazeminia S. Interaction of estradiol and renin-angiotensin system with microRNAs-21 and -29 in renal fibrosis: focus on TGF-β/smad signaling pathway. Mol Biol Rep 2024; 51:137. [PMID: 38236310 DOI: 10.1007/s11033-023-09127-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 12/06/2023] [Indexed: 01/19/2024]
Abstract
Kidney fibrosis is one of the complications of chronic kidney disease (CKD (and contributes to end-stage renal disease which requires dialysis and kidney transplantation. Several signaling pathways such as renin-angiotensin system (RAS), microRNAs (miRNAs) and transforming growth factor-β1 (TGF-β1)/Smad have a prominent role in pathophysiology and progression of renal fibrosis. Activation of classical RAS, the elevation of angiotensin II (Ang II) production and overexpression of AT1R, develop renal fibrosis via TGF-β/Smad pathway. While the non-classical RAS arm, Ang 1-7/AT2R, MasR reveals an anti-fibrotic effect via antagonizing Ang II. This review focused on studies illustrating the interaction of RAS with sexual female hormone estradiol and miRNAs in the progression of renal fibrosis with more emphasis on the TGF-β signaling pathway. MiRNAs, especially miRNA-21 and miRNA-29 showed regulatory effects in renal fibrosis. Also, 17β-estradiol (E2) is a renoprotective hormone that improved renal fibrosis. Beneficial effects of ACE inhibitors and ARBs are reported in the prevention of renal fibrosis in patients. Future studies are also merited to delineate the new therapy strategies such as miRNAs targeting, combination therapy of E2 or HRT, ACEis, and ARBs with miRNAs mimics and antagomirs in CKD to provide a new therapeutic approach for kidney patients.
Collapse
Affiliation(s)
- Soodeh Rajabi
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Shadan Saberi
- Department of Physiology and Pharmacology, Afzalipour Medical Faculty, Kerman University of Medical Sciences, Kerman, Iran
| | - Hamid Najafipour
- Cardiovascular Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Askaripour
- Department of Physiology, School of Medicine, Bam University of Medical Sciences, Bam, Iran.
| | - Mohammad Amin Rajizadeh
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Sarieh Shahraki
- Department of Physiology and Pharmacology, School of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Sara Kazeminia
- Division of Nephrology and Hypertension, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
7
|
Tang R, Lin W, Shen C, Hu X, Yu L, Meng T, Zhang L, Eggenhuizen PJ, Ooi JD, Jin P, Ding X, Xiao X, Zhong Y. Single-cell transcriptomics uncover hub genes and cell-cell crosstalk in patients with hypertensive nephropathy. Int Immunopharmacol 2023; 125:111104. [PMID: 37897949 DOI: 10.1016/j.intimp.2023.111104] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/15/2023] [Accepted: 10/19/2023] [Indexed: 10/30/2023]
Abstract
Hypertensive nephropathy (HTN) is one of the leading causes of end-stage renal disease, yet the molecular mechanisms are still unknown. To explore novel mechanisms and gene targets for HTN, the gene expression profiles of renal biopsy samples obtained from 2 healthy living donor controls and 5 HTN patients were determined by single-cell RNA sequencing. Key hub genes expression were validated by the Nephroseq v5 platform. The HTN endothelium upregulated cellular adhesion genes (ICAM2 and CEACAM1), inflammatory genes (ETS2 and IFI6) and apoptosis related genes (CNN3). Proximal tubules in HTN highly expressed hub genes including BBOX1, TPM1, TMSB10, SDC4, and NUP58, which might be potential novel targets for proximal tubular injury. The upregulated genes in tubules of HTN were mainly participating in inflammatory signatures including IFN-γ signature, NF-κB signaling, IL-12 signaling and Wnt signaling pathway. Receptor-ligand interaction analysis indicated potential cell-cell crosstalk between endothelial cells or mesangial cells with other renal resident cells in HTN. Together, our data identify a distinct cell-specific gene expression profile, pathogenic inflammatory signaling and potential cell-cell communications between endothelial cells or mesangial cells with other renal resident cells in HTN. These findings may provide a promising novel landscape for mechanisms and treatment of human HTN.
Collapse
Affiliation(s)
- Rong Tang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wei Lin
- Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Pathology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chanjuan Shen
- Department of Hematology, The Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, Zhuzhou, Hunan, China
| | - Xueling Hu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Leilin Yu
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Department of Nephrology, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, China
| | - Ting Meng
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Linlin Zhang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Peter J Eggenhuizen
- Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Joshua D Ooi
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Centre for Inflammatory Diseases, Monash University Department of Medicine, Monash Medical Centre, Clayton, VIC, Australia
| | - Peng Jin
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiang Ding
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiangcheng Xiao
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Yong Zhong
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, Hunan, China; Key Laboratory of Biological Nanotechnology of National Health Commission, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
8
|
Lu QB, Fu X, Liu Y, Wang ZC, Liu SY, Li YC, Sun HJ. Disrupted cardiac fibroblast BCAA catabolism contributes to diabetic cardiomyopathy via a periostin/NAP1L2/SIRT3 axis. Cell Mol Biol Lett 2023; 28:93. [PMID: 37993768 PMCID: PMC10666354 DOI: 10.1186/s11658-023-00510-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
BACKGROUND Periostin is an extracellular matrix protein that plays a critical role in cell fate determination and tissue remodeling, but the underlying role and mechanism of periostin in diabetic cardiomyopathy (DCM) are far from clear. Thus, we aimed to clarify the mechanistic participation of periostin in DCM. METHODS The expression of periostin was examined in DCM patients, diabetic mice and high glucose (HG)-exposed cardiac fibroblasts (CF). Gain- and loss-of-function experiments assessed the potential role of periostin in DCM pathogenesis. RNA sequencing was used to investigate the underlying mechanisms of periostin in DCM. RESULTS A mouse cytokine antibody array showed that the protein expression of periostin was most significantly upregulated in diabetic mouse heart, and this increase was also observed in patients with DCM or HG-incubated CF. Periostin-deficient mice were protected from diabetes-induced cardiac dysfunction and myocardial damage, while overexpression of periostin held the opposite effects. Hyperglycemia stimulated the expression of periostin in a TGF-β/Smad-dependent manner. RNA sequencing results showed that periostin upregulated the expression of nucleosome assembly protein 1-like 2 (NAP1L2) which recruited SIRT3 to deacetylate H3K27ac on the promoters of the branched-chain amino acid (BCAA) catabolism-related enzymes BCAT2 and PP2Cm, resulting in BCAA catabolism impairment. Additionally, CF-derived periostin induced hypertrophy, oxidative injury and inflammation in primary cardiomyocytes. Finally, we identified that glucosyringic acid (GA) specifically targeted and inhibited periostin to ameliorate DCM. CONCLUSION Overall, manipulating periostin expression may function as a promising strategy in the treatment of DCM.
Collapse
Affiliation(s)
- Qing-Bo Lu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
- Department of Endocrine, Affiliated Hospital of Jiangnan University, Jiangnan University, Wuxi, 214125, China
| | - Xiao Fu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yao Liu
- Department of Cardiac Ultrasound, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, 210000, Jiangsu, China
| | - Zi-Chao Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China
| | - Shi-Yi Liu
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Yu-Chao Li
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China
| | - Hai-Jian Sun
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, 214122, China.
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 24 Tongjia Lane, Nanjing, 210009, China.
| |
Collapse
|
9
|
Wójtowicz A, Molcan T, Lukasik K, Żebrowska E, Pawlina-Tyszko K, Gurgul A, Szmatoła T, Bugno-Poniewierska M, Ferreira-Dias G, Skarzynski DJ, Szóstek-Mioduchowska A. The potential role of miRNAs and regulation of their expression in the development of mare endometrial fibrosis. Sci Rep 2023; 13:15938. [PMID: 37743390 PMCID: PMC10518347 DOI: 10.1038/s41598-023-42149-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 09/06/2023] [Indexed: 09/26/2023] Open
Abstract
Mare endometrial fibrosis (endometrosis), is one of the main causes of equine infertility. Despite the high prevalence, both ethology, pathogenesis and the nature of its progression remain poorly understood. Recent studies have shown that microRNAs (miRNAs) are important regulators in multiple cellular processes and functions under physiological and pathological circumstances. In this article, we reported changes in miRNA expression at different stages of endometrosis and the effect of transforming growth factor (TGF)-β1 on the expression of the most dysregulated miRNAs. We identified 1, 26, and 5 differentially expressed miRNAs (DEmiRs), in categories IIA (mild fibrosis), IIB (moderate fibrosis), and III (severe fibrosis) groups compared to category I (no fibrosis) endometria group, respectively (Padjusted < 0.05, log2FC ≥ 1.0/log2FC ≤ - 1.0). This study indicated the potential involvement of miRNAs in the regulation of the process associated to the development and progression of endometrosis. The functional enrichment analysis revealed, that DEmiRs target genes involved in the mitogen-activated protein kinases, Hippo, and phosphoinositide-3-kinase (PI3K)-Akt signalling pathways, focal adhesion, and extracellular matrix-receptor interaction. Moreover, we demonstrated that the most potent profibrotic cytokine-TGF-β1-downregulated novel-eca-miR-42 (P < 0.05) expression in fibroblasts derived from endometria at early-stage endometrosis (category IIA).
Collapse
Affiliation(s)
- Anna Wójtowicz
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Tomasz Molcan
- Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Karolina Lukasik
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Ewelina Żebrowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland
| | - Klaudia Pawlina-Tyszko
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
| | - Artur Gurgul
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Tomasz Szmatoła
- Department of Animal Molecular Biology, National Research Institute of Animal Production, Cracow, Poland
- Center for Experimental and Innovative Medicine, University of Agriculture in Krakow, Cracow, Poland
| | - Monika Bugno-Poniewierska
- Department of Animal Reproduction, Anatomy and Genomics, The University of Agriculture in Krakow, Cracow, Poland
| | - Graca Ferreira-Dias
- Faculty of Veterinary Medicine, CIISA - Center for Interdisciplinary Research in Animal Health, University of Lisbon, Lisbon, Portugal
- Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), Lisbon, Portugal
| | - Dariusz J Skarzynski
- Department of Reproduction and Clinic of Farm Animals, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Anna Szóstek-Mioduchowska
- Department of Reproductive Immunology and Pathology, Institute of Animal Reproduction and Food Research of Polish Academy of Sciences, Tuwima 10, 10-748, Olsztyn, Poland.
| |
Collapse
|
10
|
Basalova N, Illarionova M, Skryabina M, Vigovskiy M, Tolstoluzhinskaya A, Primak A, Chechekhina E, Chechekhin V, Karagyaur M, Efimenko A. Advances and Obstacles in Using CRISPR/Cas9 Technology for Non-Coding RNA Gene Knockout in Human Mesenchymal Stromal Cells. Noncoding RNA 2023; 9:49. [PMID: 37736895 PMCID: PMC10514828 DOI: 10.3390/ncrna9050049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/17/2023] [Accepted: 08/22/2023] [Indexed: 09/23/2023] Open
Abstract
Non-coding RNA (ncRNAs) genes have attracted increasing attention in recent years due to their widespread involvement in physiological and pathological processes and regulatory networks. The study of the function and molecular partners of ncRNAs opens up opportunities for the early diagnosis and treatment of previously incurable diseases. However, the classical "loss-of-function" approach in ncRNA function analysis is challenged due to some specific issues. Here, we have studied the potency of two CRISPR/Cas9 variants, wild-type (SpCas9wt) and nickase (SpCas9D10A) programmable nucleases, for the editing of extended DNA sequences in human mesenchymal stromal cells (MSCs). Editing the genes of fibrosis-related hsa-miR-21-5p and hsa-miR-29c-3p, we have shown that a pair of SpCas9D10A molecules can effectively disrupt miRNA genes within the genomes of MSCs. This leads not only to a decrease in the level of knockout miRNA in MSCs and MSC-produced extracellular vesicles, but also to a change in cell physiology and the antifibrotic properties of the cell secretome. These changes correlate well with previously published data for the knockdown of certain miRNAs. The proposed approach can be used to knock out ncRNA genes within the genomes of MSCs or similar cell types in order to study their function in biological processes.
Collapse
Affiliation(s)
- Nataliya Basalova
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.B.); (M.V.); (A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Maria Illarionova
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Mariya Skryabina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Maksim Vigovskiy
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.B.); (M.V.); (A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Anastasia Tolstoluzhinskaya
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.B.); (M.V.); (A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Alexandra Primak
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Elizaveta Chechekhina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Vadim Chechekhin
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Maxim Karagyaur
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.B.); (M.V.); (A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| | - Anastasia Efimenko
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.B.); (M.V.); (A.T.)
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (M.I.); (M.S.); (A.P.); (E.C.); (V.C.)
| |
Collapse
|
11
|
Ali N, Xavier J, Engur M, Pv M, Bernardino de la Serna J. The impact of e-cigarette exposure on different organ systems: A review of recent evidence and future perspectives. JOURNAL OF HAZARDOUS MATERIALS 2023; 457:131828. [PMID: 37320902 DOI: 10.1016/j.jhazmat.2023.131828] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/22/2023] [Accepted: 06/08/2023] [Indexed: 06/17/2023]
Abstract
The use of electronic cigarettes (e-cigs) is rapidly increasing worldwide and is promoted as a smoking cessation tool. The impact of traditional cigs on human health has been well-defined in both animal and human studies. In contrast, little is known about the adverse effects of e-cigs exposure on human health. This review summarizes the impact of e-cigs exposure on different organ systems based on the rapidly expanding recent evidence from experimental and human studies. A number of growing studies have shown the adverse effects of e-cigs exposure on various organ systems. The summarized data in this review indicate that while e-cigs use causes less adverse effects on different organs compared to traditional cigs, its long-term exposure may lead to serious health effects. Data on short-term organ effects are limited and there is no sufficient evidence on long-term organ effects. Moreover, the adverse effects of secondhand and third hand e-cigs vapour exposure have not been thoroughly investigated in previous studies. Although some studies demonstrated e-cigs used as a smoking cessation tool, there is a lack of strong evidence to support it. While some researchers suggested e-cigs as a safer alternative to tobacco smoking, their long-term exposure health effects remain largely unknown. Therefore, more epidemiological and prospective studies including mechanistic studies are needed to address the potential adverse health effects of e-cigs to draw a firm conclusion about their safe use. A wide variation in e-cigs products and the lack of standardized testing methods are the major barriers to evaluating the existing data. Specific regulatory guidelines for both e-cigs components and the manufacturing process may be effective to protect consumer health.
Collapse
Affiliation(s)
- Nurshad Ali
- National Heart and Lung Institute, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, UK; Department of Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh.
| | - Joseph Xavier
- National Heart and Lung Institute, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, UK; Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India.
| | - Melih Engur
- National Heart and Lung Institute, Imperial College London, Sir Alexander Fleming Building, London SW7 2AZ, UK
| | - Mohanan Pv
- Toxicology Division, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences and Technology (Govt. of India), Poojapura, Trivandrum 695012, Kerala, India.
| | | |
Collapse
|
12
|
Macak N, Jovanovic I, Zivkovic M, Mitrovic K, Cvetkovic M, Kostic M, Stankovic A. Downregulation of fibrosis related hsa-miR-29c-3p in human CAKUT. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2023; 42:945-958. [PMID: 37291879 DOI: 10.1080/15257770.2023.2218430] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 05/20/2023] [Indexed: 06/10/2023]
Abstract
Congenital anomalies of the kidney and urinary tract (CAKUT) represent structural and functional urinary system malformations and take place as one of the most common congenital malformations with an incidence of 1:500. Ureteral obstruction-induced hydronephrosis is associated with renal fibrosis and chronic kidney diseases in the pediatric CAKUT. We aimed to construct interaction network of previously bioinformatically associated miRNAs with CAKUT differentially expressed genes in order to prioritize those associated with fibrotic process and to experimentally validate the expression of selected miRNAs in CAKUT patients compared to control group. We constructed interaction network of hsa-miR-101-3p, hsa-miR-101-5p and hsa-miR-29c-3p that showed significant association with fibrosis. The top enriched molecular pathway was extracellular matrix-receptor interaction (adjusted p = .0000263). We experimentally confirmed expression of three miRNAs (hsa-miR-29c-3p, hsa-miR-101-3p and hsa-miR-101-5p) in obstructed ureters (ureteropelvic junction obstruction and primary obstructive megaureter) and vesicoureteral reflux. The hsa-miR-29c-3p was shown to have lower expression in both patient groups compared to controls. Relative levels of hsa-miR-101-5p and hsa-miR-101-3p showed significant positive correlations in both groups of patients. Statistically significant correlation was observed between hsa-miR-101 (-3p and -5p) and hsa-miR-29c-3p only in the obstructed group. The significant downregulation of anti-fibrotic hsa-miR-29c-3p in obstructive CAKUT could explain activation of genes involved in fibrotic processes. As miRNAs are promising candidates in therapeutic approaches our results need further measurement of fibrotic markers or assessment of extent of fibrosis and functional evaluation of hsa-miR-29c.
Collapse
Affiliation(s)
- Natasa Macak
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ivan Jovanovic
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Maja Zivkovic
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Kristina Mitrovic
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Mirjana Cvetkovic
- Nephrology and Urology Departments, University Children's Hospital, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Mirjana Kostic
- Nephrology and Urology Departments, University Children's Hospital, Belgrade, Serbia
- Medical Faculty, University of Belgrade, Belgrade, Serbia
| | - Aleksandra Stankovic
- Laboratory for Radiobiology and Molecular Genetics, Vinča Institute of Nuclear Sciences - National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
13
|
Zhang H, Zhou Y, Wen D, Wang J. Noncoding RNAs: Master Regulator of Fibroblast to Myofibroblast Transition in Fibrosis. Int J Mol Sci 2023; 24:1801. [PMID: 36675315 PMCID: PMC9861037 DOI: 10.3390/ijms24021801] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/11/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Myofibroblasts escape apoptosis and proliferate abnormally under pathological conditions, especially fibrosis; they synthesize and secrete a large amount of extracellular matrix (ECM), such as α-SMA and collagen, which leads to the distortion of organ parenchyma structure, an imbalance in collagen deposition and degradation, and the replacement of parenchymal cells by fibrous connective tissues. Fibroblast to myofibroblast transition (FMT) is considered to be the main source of myofibroblasts. Therefore, it is crucial to explore the influencing factors regulating the process of FMT for the prevention, treatment, and diagnosis of FMT-related diseases. In recent years, non-coding RNAs, including microRNA, long non-coding RNAs, and circular RNAs, have attracted extensive attention from scientists due to their powerful regulatory functions, and they have been found to play a vital role in regulating FMT. In this review, we summarized ncRNAs which regulate FMT during fibrosis and found that they mainly regulated signaling pathways, including TGF-β/Smad, MAPK/P38/ERK/JNK, PI3K/AKT, and WNT/β-catenin. Furthermore, the expression of downstream transcription factors can be promoted or inhibited, indicating that ncRNAs have the potential to be a new therapeutic target for FMT-related diseases.
Collapse
Affiliation(s)
| | | | | | - Jie Wang
- Department of Immunology, Xiangya School of Medicine, Central South University, Xiangya Road, Changsha 410000, China
| |
Collapse
|
14
|
Piao SG, Ding J, Lin XJ, Nan QY, Xuan MY, Jiang YJ, Zheng HL, Jin JZ, Li C. Inhibition of RIP1-RIP3-mediated necroptosis attenuates renal fibrosis via Wnt3α/β-catenin/GSK-3β signaling in unilateral ureteral obstruction. PLoS One 2022; 17:e0274116. [PMID: 36223414 PMCID: PMC9555645 DOI: 10.1371/journal.pone.0274116] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 08/22/2022] [Indexed: 11/07/2022] Open
Abstract
Renal fibrosis represents the final common outcome of chronic kidney disease of virtually any etiology. However, the mechanism underlying the evolution of renal fibrosis remains to be addressed. This study sought to clarify whether RIP1-RIP3-mediated necroptosis is involved in renal fibrosis via Wnt3α/β-catenin/GSK-3β signaling in vitro and in a rat model of unilateral ureteral obstruction (UUO). Rats with UUO were administered RIP inhibitors (necrostatin-1 or GSK872) or β-catenin/TCF inhibitor ICG-001 daily for 7 consecutive days. UUO caused significant renal tubular necrosis and overexpression of RIP1-RIP3-MLKL axis proteins, and was accompanied by activation of the NLRP3 inflammasome and renal fibrosis. Oxidative stress caused by UUO was closely associated with endoplasmic reticulum stress and mitochondrial dysfunction, which resulted in apoptotic cell death via Wnt3α/β-catenin/GSK-3β signaling. All of these effects were abolished by an RIP inhibitor (necrostatin-1 or GSK872) or ICG-001. In H2O2-treated HK-2 cells, both RIP inhibitor and ICG-001 decreased intracellular reactive oxygen species production and apoptotic cells, but increased cell viability. Activated Wnt3α/β-catenin/GSK-3β signaling was decreased by either RIP inhibitor or ICG-001. Our findings suggest that RIP1-RIP3-mediated necroptosis contributes to the development of renal fibrosis via Wnt3α/β-catenin/GSK-3β signaling in UUO and may be a therapeutic target for protection against renal scarring of other origins.
Collapse
Affiliation(s)
- Shang Guo Piao
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Jun Ding
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Xue Jing Lin
- Department of Nephrology, Yanbian University Hospital, Yanji, China
- Department of Radionuclide Medicine, Yanbian University Hospital, Yanji, China
| | - Qi Yan Nan
- Department of Nephrology, Yanbian University Hospital, Yanji, China
- Department of Intensive Care Unit, Yanbian University Hospital, Yanji, China
| | - Mei Ying Xuan
- Department of Nephrology, Yanbian University Hospital, Yanji, China
- Department of Health Examination Central, Yanbian University Hospital, Yanji, China
| | - Yu Ji Jiang
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Hai Lan Zheng
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Ji Zhe Jin
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| | - Can Li
- Department of Nephrology, Yanbian University Hospital, Yanji, China
| |
Collapse
|
15
|
He M, Feng L, Chen Y, Gao B, Du Y, Zhou L, Li F, Liu H. Polydatin attenuates tubulointerstitial fibrosis in diabetic kidney disease by inhibiting YAP expression and nuclear translocation. Front Physiol 2022; 13:927794. [PMID: 36277194 PMCID: PMC9585250 DOI: 10.3389/fphys.2022.927794] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 09/20/2022] [Indexed: 11/25/2022] Open
Abstract
The activation of Yes-associated protein (YAP) pathway is mutually causal with the increase of extracellular matrix (ECM) stiffness. Polydatin (PD) has been proved to have anti-fibrosis effect in diabetic kidney disease (DKD), but it is still a mystery whether PD participates in YAP-related mechano-transduction. Therefore, this study intends to solve the following two problems: 1) To construct an in vitro system of polyacrylamide hydrogels (PA gels) based on the true stiffness of kidneys in healthy and DKD rats, and observe the effect of PD on pathological matrix stiffness-induced YAP expression in renal fibroblasts; 2) Compared with verteporfin (VP), a pharmacological inhibitor of YAP, to explore whether the therapeutic effect of PD on DKD in vivo model is related to the regulation of YAP. In this study, the in vitro system of PA gels with 3 kPa, 12 kPa and 30 kPa stiffness was constructed and determined for the first time to simulate the kidney stiffness of healthy rats, rats with DKD for 8 weeks and 16 weeks, respectively. Compared with the PA gels with 3 kPa stiffness, the PA gels with 12 kPa and 30 kPa stiffness significantly increased the expression of YAP, α-smooth muscle actin (α-SMA) and collagen I, and the production of reactive oxygen species (ROS) in renal fibroblasts, and the PA gels with 30 kPa stiffness were the highest. PD significantly inhibited the above-mentioned changes of fibroblasts induced by pathological matrix stiffness, suggesting that the inhibition of PD on fibroblast-to-myofibroblast transformation and ECM production was at least partially associated with regulating YAP-related mechano-transduction pathway. Importantly, the inhibitory effect of PD on YAP expression and nuclear translocation in kidneys of DKD rats is similar to that of VP, but PD is superior to VP in reducing urinary protein, blood glucose, blood urea nitrogen and serum creatinine, as well as decreasing the expression of α-SMA and collagen I, ROS overproduction and renal fibrosis. Our results prove for the first time from the biomechanical point of view that PD is a potential therapeutic strategy for delaying the progression of renal fibrosis by inhibiting YAP expression and nuclear translocation.
Collapse
Affiliation(s)
- Manlin He
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Lan Feng
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Yang Chen
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Bin Gao
- Department of Endocrinology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Yiwei Du
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Lu Zhou
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
| | - Fei Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Hongbao Liu, ; Fei Li,
| | - Hongbao Liu
- Department of Nephrology, Tangdu Hospital, Air Force Military Medical University (Fourth Military Medical University), Xi’an, China
- *Correspondence: Hongbao Liu, ; Fei Li,
| |
Collapse
|
16
|
Feng W, Xie H, Li J, Yan X, Zhu S, Sun S. miR-29c Inhibits Renal Interstitial Fibrotic Proliferative Properties through PI3K-AKT Pathway. Appl Bionics Biomech 2022; 2022:6382323. [PMID: 36051820 PMCID: PMC9427319 DOI: 10.1155/2022/6382323] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022] Open
Abstract
Renal fibrosis, in particular tubulointerstitial fibrosis, which is characterized by an increased extracellular matrix (ECM) formation and development in the interstitium, is the common end pathway for nearly all progressive kidney disorders. One of the sources for this matrix is the epithelial to mesenchymal transition (EMT) from the tabular epithelium. The driving force behind it is some profibrotic growth factors such as transforming growth factor-β (TGF-β) which is responsible for the formation of collagen in renal fibrosis. miR-29c, which is an antifibrotic microRNA, downregulates renal interstitial fibrosis by downregulating the TGF-β and collagen. However, it is not known whether miR-29c mediates the TGF-β1-driven PI3K-Akt pathway and Col-1 triggering within NRK-52E cultures. The main objective of this investigation was to examine the influence of miR-29c on the downregulation of the TGF-β1-driven PI3K-Akt pathway and Col-1 triggering in NRK-52E cultures. This study revealed that miR-29c inhibited TGF-β1 expression in NRK-52E cell cultures. Overexpression of miR-29c significantly inhibits NRK-52E culture proliferation mediated by TGF-β1. miR-29c inhibited the expression of Col-1 and decreased PI3K/Akt phosphorylation. These findings revealed a novel mechanism by which miR29c inhibits the proliferation of renal interstitial fibrotic cultures by downregulating the PI3k-Akt pathway, which is controlled by TGF-β1.
Collapse
Affiliation(s)
- Weifeng Feng
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Huijun Xie
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Jiong Li
- Department of Anatomy, College of Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xianxin Yan
- College of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Shiping Zhu
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Shengyun Sun
- Department of Traditional Chinese Medicine, First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Raja A, Zelikoff JT, Jaimes EA. A contemporary review of nephrotoxicity and e-cigarette use. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
18
|
Chen Y, Zou H, Lu H, Xiang H, Chen S. Research progress of endothelial-mesenchymal transition in diabetic kidney disease. J Cell Mol Med 2022; 26:3313-3322. [PMID: 35560773 PMCID: PMC9189345 DOI: 10.1111/jcmm.17356] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 02/22/2022] [Accepted: 03/28/2022] [Indexed: 12/25/2022] Open
Abstract
Renal fibrosis is an important pathological feature of diabetic kidney disease (DKD), manifested as tubular interstitial fibrosis, tubular atrophy, glomerulosclerosis and damage to the normal structure of the kidney. Renal fibrosis can eventually develop into renal failure. A better understanding of renal fibrosis in DKD is needed due to clinical limitations of current anti‐fibrotic drugs in terms of effectiveness, cost‐effectiveness and side effects. Fibrosis is characterized by local excessive deposition of extracellular matrix, which is derived from activated myofibroblasts to increase its production or specific tissue inhibitors of metalloproteinases to reduce its degradation. In recent years, endothelial‐mesenchymal transition (EndMT) has gradually integrated into the pathogenesis of fibrosis. In animal models of diabetic kidney disease, it has been found that EndMT is involved in the formation of renal fibrosis and multiple signalling pathways such as TGF‐β signalling pathway, Wnt signalling pathway and non‐coding RNA network participate in the regulation of EndMT during fibrosis. Here, we mainly review EndMT regulation and targeted therapy of renal fibrosis in DKD.
Collapse
Affiliation(s)
- Ying Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Hang Zou
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Shuhua Chen
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, Changsha, China
| |
Collapse
|
19
|
Zhou M, Xue C, Wu Z, Wu X, Li M. Genome-Wide Association Study Identifies New Risk Loci for Progression of Schistosomiasis Among the Chinese Population. Front Cell Infect Microbiol 2022; 12:871545. [PMID: 35493725 PMCID: PMC9039613 DOI: 10.3389/fcimb.2022.871545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 03/18/2022] [Indexed: 11/13/2022] Open
Abstract
Schistosoma japonicum infections, which lead to local inflammatory responses to schistosome eggs trapped in host tissues, can result in long-term, severe complications. The development of schistosomiasis may result from a complex interaction between the pathogenic, environmental, and host genetic components. Notably, the genetic factors that influence the development of schistosomiasis complications are poorly understood. Here we performed a genome-wide association study on multiple schistosomiasis-related phenotypes of 637 unrelated schistosomiasis patients in the Chinese population. Among three indicators of liver damage, we identified two novel, genome-wide significant single-nucleotide polymorphisms (SNPs) rs34486793 (P = 1.415 × 10-8) and rs2008259 (P = 6.78 × 10-8) at locus 14q32.2 as well as a gene, PMEPA1, at 20q13.31 (index rs62205791, P = 6.52 × 10-7). These were significantly associated with serum levels of hyaluronic acid (HA). In addition, RASIP1 and MAMSTR at 19q13.33 (index rs62132778, P = 1.72 × 10-7) were significantly associated with serum levels of aspartate aminotransferase (AST), and TPM1 at 15q22.2 (index rs12442303, P = 4.39 × 10-7) was significantly associated with serum levels of albumin. In schistosomiasis clinical signs, ITIH4 at 3p21.1 (index rs2239548) was associated with portal vein diameter (PVD) class, an indicator of portal hypertension, and OGDHL at 10q11.23 (index rs1258172) was related to ascites grade. We also detected an increased expression of these six genes in livers of mice with severe schistosomiasis. Summary data-based Mendelian randomization analyses indicated that ITIH4, PMEPA1 and MAMSTR were pleiotropically associated with PVD class, HA and AST, respectively.
Collapse
Affiliation(s)
- Miao Zhou
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Chao Xue
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Zhongdao Wu
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Provincial Engineering Technology Research Center for Biological Vector Control, Sun Yat-sen University, Guangzhou, China
| | - Xiaoying Wu
- Department of Gastroenterology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- School of Public Health, Fudan University, Shanghai, China
- *Correspondence: Xiaoying Wu, ; Miaoxin Li,
| | - Miaoxin Li
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Tropical Disease Control, Ministry of Education, Sun Yat-Sen University, Guangzhou, China
- Center for Precision Medicine, Sun Yat-Sen University, Guangzhou, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- *Correspondence: Xiaoying Wu, ; Miaoxin Li,
| |
Collapse
|
20
|
Li W, Cheng B. Knockdown of LncRNA NEAT1 inhibits myofibroblast activity in oral submucous fibrosis through miR-760/TPM1 axis. J Dent Sci 2021; 17:707-717. [PMID: 35756787 PMCID: PMC9201657 DOI: 10.1016/j.jds.2021.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 11/23/2022] Open
Abstract
Background/purpose Oral submucous fibrosis (OSF) is a fibrotic disease with high transformation of malignant disorders. Aberrant expression of lncRNA nuclear enriched abundant transcript 1 (NEAT1) was engaged with various fibrosis models, but its mechanism in OSF remained elusive. Materials and methods Fibrous buccal mucosa fibroblasts (fBMFs) were from OSF specimens. Myofibroblast activities including the alpha smooth muscle actin (α-SMA) distribution and invasion capacities were determined by Immunocytochemistry and Transwell assays. Gene and protein were identified by quantitative real time polymerase chain reaction or western blotting. Binding relationship was analyzed via Starbase and dual-luciferase reporter or RNA immunoprecipitation assays. Results NEAT1 and Tropomyosin-1 (TPM1) were significantly increased in OSF specimens, but miR-760 was decreased. NEAT1 knockdown repressed myofibroblast activities and reduced the fibrosis and Wnt/β-catenin pathway via miR-760/TPM1 axis. MiR-760 inhibition could reverse the regulation of NEAT1 knockdown via TPM1 in fBMFs. Conclusion NEAT1 knockdown inhibited myofibroblast activities and Wnt/β-catenin pathway via miR-760/TPM1 axis in fBMFs. NEAT1 could be the target for inhibiting myofibroblast activities in fBMFs for OSF treatment.
Collapse
|
21
|
Yang PY, Ho DCY, Chen SH, Hsieh PL, Liao YW, Tsai LL, Yu CC, Fang CY. Down-regulation of miR-29c promotes the progression of oral submucous fibrosis through targeting tropomyosin-1. J Formos Med Assoc 2021; 121:1117-1122. [PMID: 34696938 DOI: 10.1016/j.jfma.2021.10.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 09/29/2021] [Accepted: 10/06/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND/PURPOSE Various microRNAs (miRs) have been found to be associated with the development of the precancerous condition of the oral cavity, oral submucous fibrosis (OSF). The expression of miR-29c is dysregulated in oral cancer, but its role in OSF has not been investigated. The purpose of the study is to investigate the functional role of miR-29c and its target in OSF. METHODS The expression levels of miR-29c in OSF tissues and fibrotic buccal mucosal fibroblasts (fBMFs) were assessed using next-generation sequencing and real-time Polymerase Chain Reaction (PCR) analysis. MiR-29c mimic and inhibitors were employed to examine its functional role of myofibroblast transdifferentiation. In addition, several myofibroblast phenotypes, such as collagen gel contraction and migration were tested, and a luciferase reporter assay was conducted to confirm the relationship between miR-29c and its predicted target, tropomyosin-1 (TPM1). RESULTS We observed that miR-29c expression was downregulated in fBMFs. fBMFs transfected with miR-29c mimics exhibited reduced migration ability and collagen gel contractility, whereas inhibition of miR-29c in normal BMFs induced the myofibroblast phenotypes. Results from the luciferase reporter assay showed that TPM1 was a direct target of miR-29c and the expression of TPM1 was suppressed in the fBMFs transfected with miR-29c mimics. Besides, we confirmed that the expression of miR-29c was indeed downregulated in OSF specimens. CONCLUSION MiR-29c seems to exert an inhibitory effect on myofibroblast activation, such as collagen gel contractility and migration ability, via suppressing TPM1. These results suggested that approaches to upregulate miR-29c may be able to ameliorate the progression of OSF.
Collapse
Affiliation(s)
- Po-Yu Yang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Dennis Chun-Yu Ho
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei, Taiwan
| | - Szu-Han Chen
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Department of Anatomy, School of Medicine, China Medical University, Taichung, Taiwan
| | - Yi-Wen Liao
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Lo-Lin Tsai
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei, Taiwan
| | - Cheng-Chia Yu
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan; Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan; Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan.
| | - Chih-Yuan Fang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan; Division of Oral and Maxillofacial Surgery, Department of Dentistry, Wan Fang Hospital, Taipei, Taiwan.
| |
Collapse
|
22
|
Human Liver Stem Cell Derived Extracellular Vesicles Alleviate Kidney Fibrosis by Interfering with the β-Catenin Pathway through miR29b. Int J Mol Sci 2021; 22:ijms221910780. [PMID: 34639119 PMCID: PMC8509541 DOI: 10.3390/ijms221910780] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/29/2021] [Accepted: 10/02/2021] [Indexed: 02/07/2023] Open
Abstract
Human liver stem-cell-derived extracellular vesicles (HLSC-EVs) exhibit therapeutic properties in various pre-clinical models of kidney injury. We previously reported an overall improvement in kidney function following treatment with HLSC-EVs in a model of aristolochic acid nephropathy (AAN). Here, we provide evidence that HLSC-EVs exert anti-fibrotic effects by interfering with β-catenin signalling. A mouse model of AAN and an in vitro pro-fibrotic model were used. The β-catenin mRNA and protein expression, together with the pro-fibrotic markers α-SMA and collagen 1, were evaluated in vivo and in vitro following treatment with HLSC-EVs. Expression and functional analysis of miR29b was performed in vitro following HLSC-EV treatments through loss-of-function experiments. Results showed that expression of β-catenin was amplified both in vivo and in vitro, and β-catenin gene silencing in fibroblasts prevented AA-induced up-regulation of pro-fibrotic genes, revealing that β-catenin is an important factor in fibroblast activation. Treatment with HLSC-EVs caused increased expression of miR29b, which was significantly inhibited in the presence of α-amanitin. The suppression of the miR29b function with a selective inhibitor abolished the anti-fibrotic effects of HLSC-EVs, resulting in the up-regulation of β-catenin and pro-fibrotic α-Sma and collagen type 1 genes. Together, these data suggest a novel HLSC-EV-dependent regulatory mechanism in which β-catenin is down regulated by HLSC-EVs-induced miR29b expression.
Collapse
|
23
|
Xue T, Qiu X, Liu H, Gan C, Tan Z, Xie Y, Wang Y, Ye T. Epigenetic regulation in fibrosis progress. Pharmacol Res 2021; 173:105910. [PMID: 34562602 DOI: 10.1016/j.phrs.2021.105910] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/23/2021] [Accepted: 09/15/2021] [Indexed: 02/08/2023]
Abstract
Fibrosis, a common process of chronic inflammatory diseases, is defined as a repair response disorder when organs undergo continuous damage, ultimately leading to scar formation and functional failure. Around the world, fibrotic diseases cause high mortality, unfortunately, with limited treatment means in clinical practice. With the development and application of deep sequencing technology, comprehensively exploring the epigenetic mechanism in fibrosis has been allowed. Extensive remodeling of epigenetics controlling various cells phenotype and molecular mechanisms involved in fibrogenesis was subsequently verified. In this review, we summarize the regulatory mechanisms of DNA methylation, histone modification, noncoding RNAs (ncRNAs) and N6-methyladenosine (m6A) modification in organ fibrosis, focusing on heart, liver, lung and kidney. Additionally, we emphasize the diversity of epigenetics in the cellular and molecular mechanisms related to fibrosis. Finally, the potential and prospect of targeted therapy for fibrosis based on epigenetic is discussed.
Collapse
Affiliation(s)
- Taixiong Xue
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingyu Qiu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongyao Liu
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Cailing Gan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zui Tan
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuting Xie
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuxi Wang
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Targeted Tracer Research and Development Laboratory, Institute of Respiratory Health, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China; Precision Medicine Key Laboratory of Sichuan Province & Precision Medicine Research Center, West China Hospital, Sichuan University, Chengdu, 610041 Sichuan, China.
| | - Tinghong Ye
- Sichuan University-University of Oxford Huaxi Joint Centre for Gastrointestinal Cancer, Department of Gastroenterology and Hepatology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
24
|
Li M, Chen J, Liu S, Sun X, Xu H, Gao Q, Chen X, Xi C, Huang D, Deng Y, Zhang F, Gao S, Qiu S, Tao X, Zhai J, Wei H, Yao H, Chen W. Spermine-Related DNA Hypermethylation and Elevated Expression of Genes for Collagen Formation are Susceptible Factors for Chemotherapy-Induced Hand-Foot Syndrome in Chinese Colorectal Cancer Patients. Front Pharmacol 2021; 12:746910. [PMID: 34539419 PMCID: PMC8440935 DOI: 10.3389/fphar.2021.746910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 08/13/2021] [Indexed: 01/06/2023] Open
Abstract
Hand-foot syndrome (HFS) is a common capecitabine-based chemotherapy-related adverse event (CRAE) in patients with colorectal cancer (CRC). It is of great significance to comprehensively identify susceptible factors for HFS, and further to elucidate the biomolecular mechanism of HFS susceptibility. We performed an untargeted multi-omics analysis integrating DNA methylation, transcriptome, and metabolome data of 63 Chinese CRC patients who had complete CRAE records during capecitabine-based chemotherapy. We found that the metabolome changes for each of matched plasma, urine, and normal colorectal tissue (CRT) in relation to HFS were characterized by chronic tissue damage, which was indicated by reduced nucleotide salvage, elevated spermine level, and increased production of endogenous cytotoxic metabolites. HFS-related transcriptome changes of CRT showed an overall suppressed inflammation profile but increased M2 macrophage polarization. HFS-related DNA methylation of CRT presented gene-specific hypermethylation on genes mainly for collagen formation. The hypermethylation was accumulated in the opensea and shore regions, which elicited a positive effect on gene expression. Additionally, we developed and validated models combining relevant biomarkers showing reasonably good discrimination performance with the area under the receiver operating characteristic curve values from 0.833 to 0.955. Our results demonstrated that the multi-omics variations associated with a profibrotic phenotype were closely related to HFS susceptibility. HFS-related biomolecular variations in CRT contributed more to the relevant biomolecular mechanism of HFS than in plasma and urine. Spermine-related DNA hypermethylation and elevated expression of genes for collagen formation were closely associated with HFS susceptibility. These findings provided new insights into the susceptible factors for chemotherapy-induced HFS, which can promote the implementation of individualized treatment against HFS.
Collapse
Affiliation(s)
- Mingming Li
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiani Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shaoqun Liu
- Department of Gastric Intestinal Surgery, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaomeng Sun
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, China
| | - Huilin Xu
- Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qianmin Gao
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xintao Chen
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Chaowen Xi
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, China
| | - Doudou Huang
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yi Deng
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Feng Zhang
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shouhong Gao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Shi Qiu
- Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xia Tao
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jingwen Zhai
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Hua Wei
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China.,Department of Pharmacy, 905th Hospital of PLA Navy, Naval Medical University, Shanghai, China
| | - Houshan Yao
- Department of General Surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Wansheng Chen
- Department of Pharmacy, Second Affiliated Hospital of Naval Medical University, Shanghai, China.,Traditional Chinese Medicine Resource and Technology Center, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
25
|
Zhao H, Feng YL, Liu T, Wang JJ, Yu J. MicroRNAs in organ fibrosis: From molecular mechanisms to potential therapeutic targets. Pathol Res Pract 2021; 225:153588. [PMID: 34419718 DOI: 10.1016/j.prp.2021.153588] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 12/20/2022]
Abstract
Fibrosis is caused by chronic tissue injury and characterized by the excessive deposition of extracellular matrix (ECM) that ultimately results in organ failure and death. Owing to lacking of effective treatment against tissue fibrosis, it causes a high morbidity and mortality worldwide. Thus, it is of great importance to find an effective therapy strategy for the treatment of fibrosis. MicroRNAs (miRNAs) play vital roles in many biological processes by targeting downstream genes. Numerous studies demonstrated that miRNAs served as biomarkers of various diseases, suggesting the potential therapeutic targets for diseases. It was recently reported that miRNAs played an important role in the development of organ fibrosis, which showed a promising prospect against fibrosis by targeting intervention. Here, we summarize the roles of miRNAs in the process of organ fibrosis, including liver, lung, heart and kidney, and highlight miRNAs being novel therapeutic targets for organ fibrosis.
Collapse
Affiliation(s)
- Hui Zhao
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China
| | - Ya-Long Feng
- School of Chemistry and Chemical Engineering, Xianyang Normal University, Xianyang, Shaanxi, 712000, China
| | - Tian Liu
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China
| | - Jing-Jing Wang
- Weinan Linwei District Maternal and Child Health Family Planning Service Center, No.144 Dongfeng Road Weinan, Shannxi 714000, China
| | - Jun Yu
- Clinical Experimental Center, Xi'an International Medical Center Hospital, No. 777 Xitai Road Xi'an, Shaanxi 710100, China; Xi'an Engineering Technology Research Center for Cardiovascular Active Peptids, No. 777 Xitai Road Xi'an, Shaanxi 710100, China.
| |
Collapse
|
26
|
Li SS, Sun Q, Hua MR, Suo P, Chen JR, Yu XY, Zhao YY. Targeting the Wnt/β-Catenin Signaling Pathway as a Potential Therapeutic Strategy in Renal Tubulointerstitial Fibrosis. Front Pharmacol 2021; 12:719880. [PMID: 34483931 PMCID: PMC8415231 DOI: 10.3389/fphar.2021.719880] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/03/2021] [Indexed: 12/15/2022] Open
Abstract
The Wnt/β-catenin signaling pathway plays important roles in embryonic development and tissue homeostasis. Wnt signaling is induced, and β-catenin is activated, associated with the development and progression of renal fibrosis. Wnt/β-catenin controls the expression of various downstream mediators such as snail1, twist, matrix metalloproteinase-7, plasminogen activator inhibitor-1, transient receptor potential canonical 6, and renin-angiotensin system components in epithelial cells, fibroblast, and macrophages. In addition, Wnt/β-catenin is usually intertwined with other signaling pathways to promote renal interstitial fibrosis. Actually, given the crucial of Wnt/β-catenin signaling in renal fibrogenesis, blocking this signaling may benefit renal interstitial fibrosis. There are several antagonists of Wnt signaling that negatively control Wnt activation, and these include soluble Fzd-related proteins, the family of Dickkopf 1 proteins, Klotho and Wnt inhibitory factor-1. Furthermore, numerous emerging small-molecule β-catenin inhibitors cannot be ignored to prevent and treat renal fibrosis. Moreover, we reviewed the knowledge focusing on anti-fibrotic effects of natural products commonly used in kidney disease by inhibiting the Wnt/β-catenin signaling pathway. Therefore, in this review, we summarize recent advances in the regulation, downstream targets, role, and mechanisms of Wnt/β-catenin signaling in renal fibrosis pathogenesis. We also discuss the therapeutic potential of targeting this pathway to treat renal fibrosis; this may shed new insights into effective treatment strategies to prevent and treat renal fibrosis.
Collapse
Affiliation(s)
- Shan-Shan Li
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi’an, China
- The First School of Clinical Medicine, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Qian Sun
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi’an, China
- The First School of Clinical Medicine, Shaanxi University of Traditional Chinese Medicine, Xianyang, China
| | - Meng-Ru Hua
- Faculty of Life Science and Medicine, Northwest University, Xi’an, China
| | - Ping Suo
- Faculty of Life Science and Medicine, Northwest University, Xi’an, China
| | - Jia-Rong Chen
- Department of Clinical Pharmacy, Affiliated Hospital of Chengdu University, Chengdu, China
| | - Xiao-Yong Yu
- Department of Nephrology, Shaanxi Traditional Chinese Medicine Hospital, Xi’an, China
| | - Ying-Yong Zhao
- Faculty of Life Science and Medicine, Northwest University, Xi’an, China
| |
Collapse
|
27
|
Yiu WH, Li Y, Lok SWY, Chan KW, Chan LYY, Leung JCK, Lai KN, Tsu JHL, Chao J, Huang XR, Lan HY, Tang SCW. Protective role of kallistatin in renal fibrosis via modulation of Wnt/β-catenin signaling. Clin Sci (Lond) 2021; 135:429-446. [PMID: 33458750 DOI: 10.1042/cs20201161] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 12/31/2022]
Abstract
Kallistatin is a multiple functional serine protease inhibitor that protects against vascular injury, organ damage and tumor progression. Kallistatin treatment reduces inflammation and fibrosis in the progression of chronic kidney disease (CKD), but the molecular mechanisms underlying this protective process and whether kallistatin plays an endogenous role are incompletely understood. In the present study, we observed that renal kallistatin levels were significantly lower in patients with CKD. It was also positively correlated with estimated glomerular filtration rate (eGFR) and negatively correlated with serum creatinine level. Unilateral ureteral obstruction (UUO) in animals also led to down-regulation of kallistatin protein in the kidney, and depletion of endogenous kallistatin by antibody injection resulted in aggravated renal fibrosis, which was accompanied by enhanced Wnt/β-catenin activation. Conversely, overexpression of kallistatin attenuated renal inflammation, interstitial fibroblast activation and tubular injury in UUO mice. The protective effect of kallistatin was due to the suppression of TGF-β and β-catenin signaling pathways and subsequent inhibition of epithelial-to-mesenchymal transition (EMT) in cultured tubular cells. In addition, kallistatin could inhibit TGF-β-mediated fibroblast activation via modulation of Wnt4/β-catenin signaling pathway. Therefore, endogenous kallistatin protects against renal fibrosis by modulating Wnt/β-catenin-mediated EMT and fibroblast activation. Down-regulation of kallistatin in the progression of renal fibrosis underlies its potential as a valuable clinical biomarker and therapeutic target in CKD.
Collapse
Affiliation(s)
- Wai Han Yiu
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Ye Li
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Sarah W Y Lok
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kam Wa Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Loretta Y Y Chan
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Joseph C K Leung
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Kar Neng Lai
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - James H L Tsu
- Department of Surgery, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| | - Julie Chao
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, U.S.A
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Hui Yao Lan
- Department of Medicine and Therapeutics, and Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong
| | - Sydney C W Tang
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong
| |
Collapse
|
28
|
Gu YY, Lu FH, Huang XR, Zhang L, Mao W, Yu XQ, Liu XS, Lan HY. Non-Coding RNAs as Biomarkers and Therapeutic Targets for Diabetic Kidney Disease. Front Pharmacol 2021; 11:583528. [PMID: 33574750 PMCID: PMC7870688 DOI: 10.3389/fphar.2020.583528] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic kidney disease (DKD) is the most common diabetic complication and is a leading cause of end-stage kidney disease. Increasing evidence shows that DKD is regulated not only by many classical signaling pathways but also by epigenetic mechanisms involving chromatin histone modifications, DNA methylation, and non-coding RNA (ncRNAs). In this review, we focus on our current understanding of the role and mechanisms of ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in the pathogenesis of DKD. Of them, the regulatory role of TGF-β/Smad3-dependent miRNAs and lncRNAs in DKD is highlighted. Importantly, miRNAs and lncRNAs as biomarkers and therapeutic targets for DKD are also described, and the perspective of ncRNAs as a novel therapeutic approach for combating diabetic nephropathy is also discussed.
Collapse
Affiliation(s)
- Yue-Yu Gu
- Department of Nephrology and State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Fu-Hua Lu
- Department of Nephrology and State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao-Ru Huang
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong, China
| | - Lei Zhang
- Department of Nephrology and State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Mao
- Department of Nephrology and State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xue-Qing Yu
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
| | - Xu-Sheng Liu
- Department of Nephrology and State Key Laboratory of Dampness Syndrome of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui-Yao Lan
- Department of Medicine and Therapeutics, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Diseases, Guangdong Academy of Medical Sciences, Guangdong Provincial People’s Hospital, Guangzhou, China
- Guangdong-Hong Kong Joint Laboratory for Immunological and Genetic Kidney Diseases, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
29
|
Liu L, Chen G, Chen T, Shi W, Hu H, Song K, Huang R, Cai H, He Y. si-SNHG5-FOXF2 inhibits TGF-β1-induced fibrosis in human primary endometrial stromal cells by the Wnt/β-catenin signalling pathway. Stem Cell Res Ther 2020; 11:479. [PMID: 33176855 PMCID: PMC7656702 DOI: 10.1186/s13287-020-01990-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 10/21/2020] [Indexed: 11/10/2022] Open
Abstract
Background Intrauterine adhesions (IUAs) are manifestations of endometrial fibrosis characterized by inflammation and fibrinogen aggregation in the extracellular matrix (ECM). The available therapeutic interventions for IUA are insufficiently effective in the clinical setting for postoperative adhesion recurrence and infertility problems. In this study, we investigated whether si-SNHG5-FOXF2 can serve as a molecular mechanism for the inhibition of IUA fibrosis ex vivo. Methods FOXF2, TGF-β1 and collagen expression levels were measured by microarray sequencing analysis in three normal endometrium groups and six IUA patients. We induced primary human endometrial stromal cells (HESCs) into myofibroblasts (MFs) to develop an IUA cell model with various concentrations of TGF-β1 at various times. Downstream target genes of FOXF2 were screened by chromatin immunoprecipitation combined with whole-genome high-throughput sequencing (ChIP-seq). We investigated ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs with FOXF2 downregulation by quantitative reverse transcription-polymerase chain reaction (qRT-PCR), western blotting (WB), immunohistochemistry (IHC), flow cytometry, ethylenediurea (EdU) and CCK8 assays. We identified long noncoding RNAs (lncRNA) SNHG5 as the upstream regulatory gene of FOXF2 through RNA immunoprecipitation (RIP), RNA pulldown and fluorescence in situ hybridization (FISH). Finally, we examined FOXF2 expression, ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related proteins in primary HESCs upon FOXF2 downregulation. Results FOXF2 was highly expressed in the endometrium of patients with IUA. Treatment of primary HESCs with 10 ng/ml TGF-β1 for 72 h was found to be most effective for developing an IUA cell model. FOXF2 regulated multiple downstream target genes, including collagen, vimentin (VIM) and cyclin D2/DK4, by ChIP-seq and ChIP-PCR. FOXF2 downregulation inhibited TGF-β1-mediated primary HESC fibrosis, including ECM formation, cell proliferation and Wnt/β-catenin signalling pathway-related protein expression. We identified lncRNA SNHG5 as an upstream gene that directly regulates FOXF2 by RIP-seq, qRT-PCR, WB and FISH. SNHG5 downregulation suppressed FOXF2 expression in the IUA cell model, resulting in synergistic repression of the Wnt/β-catenin pathway, thereby altering TGF-β1-mediated ECM aggregation in endometrial stromal cells ex vivo. Conclusions Regulation of the Wnt/β-catenin signalling pathway and ECM formation by si-SNHG5-FOXF2 effectively inhibited the profibrotic effect of TGF-β1 on primary HESCs. This finding can provide a molecular basis for antagonizing TGF-β1-mediated fibrosis in primary HESCs.
Collapse
Affiliation(s)
- Limin Liu
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Guobin Chen
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Taoliang Chen
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Wenjuan Shi
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Haiyan Hu
- Department of Obstetrics and Gynecology, Affiliated Shenzhen Maternity & Child Healthcare Hospital, Southern Medical University, Shenzhen, China
| | - Kaijing Song
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ruichun Huang
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huihua Cai
- Department of Obstetrics and Gynecology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Yuanli He
- Department of Obstetrics and Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|