1
|
Lei L, Xiang YX, Luo ML, Zhang ZY, Wu HW, Tang C, Cui TJ, Zhang XM, Wang XH, Delic D, Klein T, Liu Y, Krämer BK, Zheng ZH, Lu YP, Hocher B, Zhu T. Intercellular Communication Network of CellChat Uncovers Mechanisms of Kidney Fibrosis Based on Single-Cell RNA Sequencing. Kidney Blood Press Res 2025; 50:276-299. [PMID: 40112793 DOI: 10.1159/000545209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 03/03/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is a global health concern, with renal fibrosis being a major pathological feature. Empagliflozin (Empa), a sodium-glucose co-transporter-2 inhibitor, has shown promise in protecting the kidney. This study aimed to investigate the effects of Empa on renal fibrosis in a nondiabetic CKD model and to elucidate the underlying mechanisms. METHODS We established a CKD model using 5/6 nephrectomy (5/6 Nx) rats and divided them into three groups: placebo-treated sham surgery rats, placebo-treated 5/6 Nx rats, and Empa-treated 5/6 Nx rats. Kidney function was assessed by measuring blood urea nitrogen, serum creatinine, and urinary albumin-to-creatinine ratio. Renal fibrosis was evaluated histologically. Single-cell RNA sequencing (scRNA-seq) was performed to analyze intercellular communication networks and identify alterations in ligand-receptor pairs and signaling pathways involved in fibrosis. RESULTS Empa treatment significantly improved kidney function and reduced renal interstitial fibrosis in 5/6 Nx rats. scRNA-seq revealed that Empa modulated the TGF-β signaling pathway, inhibited intercellular communication, and reduced the expression of fibrotic genes such as COLLAGEN, FN1, THBS, and LAMININ. Furthermore, Empa downregulated GRN gene expression, weakened signal transmission in the MIF pathway, consequently reduced the interaction between M2 macrophages and other cell types, such as endothelial cells, fibroblasts, and mesangial cells. CONCLUSION This study elucidates the potential mechanisms by which Empa slows the progression of renal fibrosis in nondiabetic CKD. By reducing the number of M2 macrophages and inhibiting signal transduction in both pro-inflammatory and fibrotic pathways, Empa modulates the intercellular communication network in renal cells, offering a promising therapeutic strategy for CKD management.
Collapse
Affiliation(s)
- Lei Lei
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yun-Xiu Xiang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Mao-Lin Luo
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Department of Endocrinology and Metabolism, People's Hospital of Liwan District, Guangzhou, China
| | - Ze-Yu Zhang
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Hong-Wei Wu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Chun Tang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Tian-Jiao Cui
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xue-Mei Zhang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Xiao-Hua Wang
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Denis Delic
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Thomas Klein
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Yvonne Liu
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany,
- Medical Faculty, Charité Universitätsmedizin Berlin, Berlin, Germany,
| | - Bernhard K Krämer
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Zhi-Hua Zheng
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yong-Ping Lu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
- Department of Nephrology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Berthold Hocher
- Fifth Department of Medicine (Nephrology/Endocrinology/Rheumatology), University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
- Institute of Medical Diagnostics, IMD, Berlin, Germany
- Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
- School of Medicine, Central South University, Changsha, China
| | - Ting Zhu
- Department of Nephrology, Center of Kidney and Urology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
2
|
Zhao J, Wang X, Wu Y, Zhao C. Krüppel-like factor 4 modulates the miR-101/COL10A1 axis to inhibit renal fibrosis after AKI by regulating epithelial-mesenchymal transition. Ren Fail 2024; 46:2316259. [PMID: 38345033 PMCID: PMC10863509 DOI: 10.1080/0886022x.2024.2316259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 02/03/2024] [Indexed: 02/15/2024] Open
Abstract
Acute kidney injury (AKI) can progress to renal fibrosis and chronic kidney disease (CKD), which reduces quality of life and increases the economic burden on patients. However, the molecular mechanisms underlying renal fibrosis following AKI remain unclear. This study tested the hypothesis that the Krüppel-like factor 4 (KLF4)/miR-101/Collagen alpha-1X (COL10A1) axis could inhibit epithelial-mesenchymal transition (EMT) and renal fibrosis after AKI in a mouse model of ischemia-reperfusion (I/R)-induced renal fibrosis and HK-2 cells by gene silencing, overexpression, immunofluorescence, immunohistochemistry, real-time quantitative PCR, Western blotting, dual-luciferase reporter assay, fluorescence in situ hybridization (FISH) and ELISA. Compared with the Sham group, I/R induced renal tubular and glomerular injury and fibrosis, and increased the levels of BUN, serum Scr and neutrophil gelatinase-associated lipocalin (NGAL), Col10a1 and Vimentin expression, but decreased E-cadherin expression in the kidney tissues of mice at 42 days post-I/R. Similarly, hypoxia promoted fibroblastic morphological changes in HK-2 cells and enhanced NGAL, COL10A1, Vimentin, and α-SMA expression, but reduced E-cadherin expression in HK-2 cells. These pathological changes were significantly mitigated in COL10A1-silenced renal tissues and HK-2 cells. KLF4 induces miR-101 transcription. More importantly, hypoxia upregulated Vimentin and COL10A1 expression, but decreased miR-101, KLF4, and E-cadherin expression in HK-2 cells. These hypoxic effects were significantly mitigated or abrogated by KLF4 over-expression in the HK-2 cells. Our data indicate that KLF4 up-regulates miR-101 expression, leading to the downregulation of COL10A1 expression, inhibition of EMT and renal fibrosis during the pathogenic process of I/R-related renal fibrosis.
Collapse
Affiliation(s)
- Jingying Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Xiuli Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yubin Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Chengguang Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
3
|
Bracken CP, Goodall GJ, Gregory PA. RNA regulatory mechanisms controlling TGF-β signaling and EMT in cancer. Semin Cancer Biol 2024; 102-103:4-16. [PMID: 38917876 DOI: 10.1016/j.semcancer.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/05/2024] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
Epithelial-mesenchymal transition (EMT) is a major contributor to metastatic progression and is prominently regulated by TGF-β signalling. Both EMT and TGF-β pathway components are tightly controlled by non-coding RNAs - including microRNAs (miRNAs), long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs) - that collectively have major impacts on gene expression and resulting cellular states. While miRNAs are the best characterised regulators of EMT and TGF-β signaling and the miR-200-ZEB1/2 feedback loop plays a central role, important functions for lncRNAs and circRNAs are also now emerging. This review will summarise our current understanding of the roles of non-coding RNAs in EMT and TGF-β signaling with a focus on their functions in cancer progression.
Collapse
Affiliation(s)
- Cameron P Bracken
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Gregory J Goodall
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia; School of Biological Sciences, Faculty of Sciences, Engineering and Technology, The University of Adelaide, Adelaide, SA 5000, Australia.
| | - Philip A Gregory
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
4
|
Shelke V, Kale A, Sankrityayan H, Anders HJ, Gaikwad AB. Long non-coding RNAs as emerging regulators of miRNAs and epigenetics in diabetes-related chronic kidney disease. Arch Physiol Biochem 2024; 130:230-241. [PMID: 34986074 DOI: 10.1080/13813455.2021.2023580] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/22/2021] [Indexed: 01/19/2023]
Abstract
Diabetes is one of the major cause of chronic kidney disease (CKD), including "diabetic nephropathy," and is an increasingly prevalent accelerator of the progression of non-diabetic forms of CKD. The long non-coding RNAs (lncRNAs) have come into the limelight in the past few years as one of the emerging weapons against CKD in diabetes. Available data over the past few years demonstrate the interaction of lncRNAs with miRNAs and epigenetic machinery. Interestingly, the evolving data suggest that lncRNAs play a vital role in diabetes-associated CKD by regulation of epigenetic enzymes such as DNA methyltransferase, histone deacetylases, and histone methyltransferases. LncRNAs are also engaged in the regulation of several miRNAs in diabetic nephropathy. Hence this review will elaborate on the association between lncRNAs and their interaction with epigenetic regulators involved in different aspects and thus the progression of CKD in diabetes.
Collapse
Affiliation(s)
- Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani, India
| | - Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani, India
| | - Himanshu Sankrityayan
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, University Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani, India
| |
Collapse
|
5
|
Lee SR, Lee HE, Yoo JY, An EJ, Song SJ, Han KH, Cha DR, Bae YS. Nox4-SH3YL1 complex is involved in diabetic nephropathy. iScience 2024; 27:108868. [PMID: 38318360 PMCID: PMC10839645 DOI: 10.1016/j.isci.2024.108868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/25/2023] [Accepted: 01/08/2024] [Indexed: 02/07/2024] Open
Abstract
Nox4-derived H2O2 generation plays an important role in the pathogenesis of chronic kidney diseases (CKDs) such as diabetic nephropathy (DN). Here, we showed that SH3 domain-containing Ysc84-like 1 (SH3YL1), a Nox4 cytosolic activator, regulated DN. Streptozotocin (STZ)-induced type Ⅰ diabetic models in SH3YL1 whole-body knockout (KO) mice and podocyte-specific SH3YL1 conditional KO (Nphs2-Cre/SH3YL1fl/fl) mice were established to investigate the function of SH3YL1 in DN. The expression of fibrosis markers and inflammatory cytokines, the generation of oxidative stress, and the loss of podocytes were suppressed in diabetic SH3YL1 KO and Nphs2-Cre/SH3YL1fl/fl mice, compared to diabetic control mice. To extrapolate the observations derived from diabetic mice to clinical implication, we measured the protein level of SH3YL1 in patients DN. In fact, the SH3YL1 level was increased in patients DN. Overall, the SH3YL1-Nox4 complex was identified to play an important role in renal inflammation and fibrosis, resulting in the development of DN.
Collapse
Affiliation(s)
- Sae Rom Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Hye Eun Lee
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Jung-Yeon Yoo
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Eun Jung An
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| | - Soo-Jin Song
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Ki-Hwan Han
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul 07804, Korea
| | - Dae Ryong Cha
- Department of Internal Medicine, Division of Nephrology, Korea University Ansan Hospital, 516 Kojan-Dong, Ansan City, Kyungki-Do 15355, Korea
| | - Yun Soo Bae
- Department of Life Sciences, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
6
|
Liebisch M, Wolf G. Role of Epigenetic Changes in the Pathophysiology of Diabetic Kidney Disease. GLOMERULAR DISEASES 2024; 4:211-226. [PMID: 39649441 PMCID: PMC11623970 DOI: 10.1159/000541923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 10/08/2024] [Indexed: 12/10/2024]
Abstract
Background Diabetic kidney disease (DKD) is a global health issue. Epigenetic changes play an important role in the pathogenesis of this disease. Summary DKD is currently the leading cause of kidney failure worldwide. Although much is known about the pathophysiology of DKD, the research field of epigenetics is relatively new. Several recent studies have demonstrated that diabetes-induced dysregulation of epigenetic mechanisms alters the expression of pathological genes in kidney cells. If these changes persist for a long time, the so-called "metabolic memory" could be established. In this review, we highlight diabetes-induced epigenetic modifications associated with DKD. While there is a substantial amount of literature on epigenetic changes, only a few studies describe the underlying molecular mechanisms. Detailed analyses have shown that epigenetic changes play an important role in known pathological features of DKD, such as podocyte injury, fibrosis, accumulation of extracellular matrix, or oxidative injury, all of which contribute to the pathophysiology of disease. The transforming growth factor-β plays a key role as it is involved in all-mentioned epigenetic types of regulation. Key Messages Epigenetic is crucial for the development and progression of DKD, but the detailed molecular mechanisms have to be further analyzed more in detail.
Collapse
Affiliation(s)
- Marita Liebisch
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| | - Gunter Wolf
- Department of Internal Medicine III, University Hospital Jena, Jena, Germany
| |
Collapse
|
7
|
Swaminathan SM, Rao IR, Shenoy SV, Prabhu AR, Mohan PB, Rangaswamy D, Bhojaraja MV, Nagri SK, Nagaraju SP. Novel biomarkers for prognosticating diabetic kidney disease progression. Int Urol Nephrol 2023; 55:913-928. [PMID: 36271990 PMCID: PMC10030535 DOI: 10.1007/s11255-022-03354-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 08/21/2022] [Indexed: 10/24/2022]
Abstract
The global burden of diabetic kidney disease (DKD) is escalating, and it remains as a predominant cause of the end-stage renal disease (ESRD). DKD is associated with increased cardiovascular disease and morbidity in all types of diabetes. Prediction of progression with albuminuria and eGFR is challenging in DKD, especially in non-proteinuric DKD patients. The pathogenesis of DKD is multifactorial characterized by injury to all components of the nephron, whereas albuminuria is an indicator of only glomerular injury. The limits in the diagnostic and prognostic value of urine albumin demonstrate the need for alternative and clinically significant early biomarkers, allowing more targeted and effective diabetic treatment, to reduce the burden of DKD and ESRD. Identification of biomarkers, based on multifactorial pathogenesis of DKD can be the crucial paradigm in the treatment algorithm of DKD patients. This review focuses on the potential biomarkers linked to DKD pathogenesis, particularly with the hope of broadening the diagnostic window to identify patients with different stages of DKD progression.
Collapse
Affiliation(s)
- Shilna Muttickal Swaminathan
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Indu Ramachandra Rao
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Srinivas Vinayak Shenoy
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Attur Ravindra Prabhu
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Pooja Basthi Mohan
- Department of Gastroenterology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Dharshan Rangaswamy
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Mohan V Bhojaraja
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Shivashankara Kaniyoor Nagri
- Department of Medicine, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India
| | - Shankar Prasad Nagaraju
- Department of Nephrology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Manipal, India.
| |
Collapse
|
8
|
Xia S, Vila Ellis L, Winkley K, Menden H, Mabry SM, Venkatraman A, Louiselle D, Gibson M, Grundberg E, Chen J, Sampath V. Neonatal hyperoxia induces activated pulmonary cellular states and sex-dependent transcriptomic changes in a model of experimental bronchopulmonary dysplasia. Am J Physiol Lung Cell Mol Physiol 2023; 324:L123-L140. [PMID: 36537711 PMCID: PMC9902224 DOI: 10.1152/ajplung.00252.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 11/08/2022] [Accepted: 11/17/2022] [Indexed: 12/24/2022] Open
Abstract
Hyperoxia disrupts lung development in mice and causes bronchopulmonary dysplasia (BPD) in neonates. To investigate sex-dependent molecular and cellular programming involved in hyperoxia, we surveyed the mouse lung using single cell RNA sequencing (scRNA-seq), and validated our findings in human neonatal lung cells in vitro. Hyperoxia-induced inflammation in alveolar type (AT) 2 cells gave rise to damage-associated transient progenitors (DATPs). It also induced a new subpopulation of AT1 cells with reduced expression of growth factors normally secreted by AT1 cells, but increased mitochondrial gene expression. Female alveolar epithelial cells had less EMT and pulmonary fibrosis signaling in hyperoxia. In the endothelium, expansion of Car4+ EC (Cap2) was seen in hyperoxia along with an emergent subpopulation of Cap2 with repressed VEGF signaling. This regenerative response was increased in females exposed to hyperoxia. Mesenchymal cells had inflammatory signatures in hyperoxia, with a new distal interstitial fibroblast subcluster characterized by repressed lipid biosynthesis and a transcriptomic signature resembling myofibroblasts. Hyperoxia-induced gene expression signatures in human neonatal fibroblasts and alveolar epithelial cells in vitro resembled mouse scRNA-seq data. These findings suggest that neonatal exposure to hyperoxia programs distinct sex-specific stem cell progenitor and cellular reparative responses that underpin lung remodeling in BPD.
Collapse
Affiliation(s)
- Sheng Xia
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Lisandra Vila Ellis
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Konner Winkley
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Heather Menden
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Sherry M Mabry
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Aparna Venkatraman
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
| | - Daniel Louiselle
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Margaret Gibson
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
| | - Elin Grundberg
- Genomic Medicine Center, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| | - Jichao Chen
- Department of Pulmonary Medicine, University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | - Venkatesh Sampath
- Department of Pediatrics, Children's Mercy Hospital, Kansas City, Missouri
- Children's Mercy Research Institute, Kansas City, Missouri
| |
Collapse
|
9
|
Li H, Liu P, Li D, Wang Z, Ding Z, Zhou M, Chen X, Miao M, Ding J, Lin W, Liu Y, Zha X. STAT3/miR-130b-3p/MBNL1 feedback loop regulated by mTORC1 signaling promotes angiogenesis and tumor growth. J Exp Clin Cancer Res 2022; 41:297. [PMID: 36217202 PMCID: PMC9552455 DOI: 10.1186/s13046-022-02513-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Aberrantly activated mammalian target of rapamycin complex 1 (mTORC1) plays a vital role in tumor angiogenesis, but its precise mechanisms are still unclear. METHODS Micro-RNA-130b-3p (miR-130b-3p) expression in mTORC1-activated and control cells was examined by quantitative real-time PCR (qRT-PCR). MiR-130b-3p levels and their correlation with mTORC1 activity were evaluated by analyzing publicly available databases and in-house head and neck squamous cell carcinoma (HNSCC) tissues. The role of miR-130b-3p in mTORC1-mediated angiogenesis and tumor growth was examined using tube formation assay, chicken chorioallantoic membrane assay, cell line - derived xenograft models, and an HNSCC patient-derived xenograft (PDX) model. The regulatory mechanisms among signal transducer and activator of transcription 3 (STAT3), miR-130b-3p, and muscleblind-like protein 1 (MBNL1) were investigated via bioinformatics analyses, qRT-PCR, western blot, RNA immunoprecipitation, immunofluorescence, luciferase reporter assay, and chromatin immunoprecipitation assay. RESULTS Elevated miR-130b-3p enhanced the angiogenic and tumorigenic abilities of mTORC1-activated cells both in vitro and in vivo. STAT3, a downstream effector of mTORC1, transactivated miR-130b-3p by direct binding promoter of the miR-130b gene. MBNL1 was identified as a direct target of miR-130b-3p. MBNL1 depletion rescued the compromised angiogenesis and tumor growth caused by miR-130b-3p inhibition. MiR-130b-3p levels were significantly upregulated and positively correlated with mTORC1 signaling in multiple cancers. MiR-130b-3p inhibition attenuated tumor angiogenesis and growth in an HNSCC PDX model. MBNL1 feedback inhibited STAT3 activation in mTORC1-activated cells. CONCLUSIONS The STAT3/miR-130b-3p/MBNL1 feedback loop plays a vital role in mTORC1-mediated angiogenesis and tumor progression. This pathway could be targeted for therapeutic intervention of mTORC1-related cancers.
Collapse
Affiliation(s)
- Hongwu Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- Anhui Public Health Clinical Center, Hefei, 230032, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Ping Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
- Anhui Public Health Clinical Center, Hefei, 230032, China
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Dapeng Li
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Zixi Wang
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Zhao Ding
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Meng Zhou
- Department of Pharmacy, Genertec Universal Medical Maanshan Shiqiye Hospital, Maanshan, 243000, Anhui Province, China
| | - Xu Chen
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Manli Miao
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China
| | - Junli Ding
- Department of Pediatrics, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Wei Lin
- Department of Stomatology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Yehai Liu
- Department of Otorhinolaryngology, Head & Neck Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
10
|
dos Santos Bronel BA, Anauate AC, Maquigussa E, Boim MA, da Silva Novaes A. Determination of reference genes as a quantitative standard for gene expression analysis in mouse mesangial cells stimulated with TGF-β. Sci Rep 2022; 12:15626. [PMID: 36115882 PMCID: PMC9482652 DOI: 10.1038/s41598-022-19548-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 08/30/2022] [Indexed: 11/29/2022] Open
Abstract
Reverse transcription-quantitative polymerase chain reaction (RT-PCR) is the gold standard technique for gene expression analysis, but the choice of quantitative reference genes (housekeeping genes, HKG) remains challenging. Identify the best HKG is essential for estimating the expression level of target genes. Therefore, the aim of this study was to determine the best HKG for an in vitro model with mouse mesangial cells (MMCs) stimulated with 5 ng/mL of TGF-β. Five candidates HKG were selected: Actb, Hprt, Gapdh, 18S and Ppia. After quantitative expression, the best combination of these genes was analyzed in silico using six software programs. To validate the results, the best genes were used to normalize the expression levels of fibronectin, vimentin and α-SMA. In silico analysis revealed that Ppia, Gapdh and 18S were the most stable genes between the groups. GenEX software and Spearman's correlation determined Ppia and Gapdh as the best HKG pair, and validation of the HKG by normalizing fibronectin, vimentin and α-SMA were consistent with results from the literature. Our results established the combination of Ppia and Gapdh as the best HKG pair for gene expression analysis by RT-PCR in this in vitro model using MMCs treated with TGF-β.
Collapse
|
11
|
Morgan S, Lee LH, Halu A, Nicolau JS, Higashi H, Ha AH, Wen JR, Daugherty A, Libby P, Cameron SJ, Mix D, Aikawa E, Owens AP, Singh SA, Aikawa M. Identifying novel mechanisms of abdominal aortic aneurysm via unbiased proteomics and systems biology. Front Cardiovasc Med 2022; 9:889994. [PMID: 35990960 PMCID: PMC9382335 DOI: 10.3389/fcvm.2022.889994] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022] Open
Abstract
Background Abdominal aortic aneurysm (AAA), characterized by a continued expansion of the aorta, leads to rupture if not surgically repaired. Mice aid the study of disease progression and its underlying mechanisms since sequential studies of aneurysm development are not feasible in humans. The present study used unbiased proteomics and systems biology to understand the molecular relationship between the mouse models of AAA and the human disease. Methods and results Aortic tissues of developing and established aneurysms produced by either angiotensin II (AngII) infusion in Apoe -/- and Ldlr -/- mice or intraluminal elastase incubation in wildtype C57BL/6J mice were examined. Aortas were dissected free and separated into eight anatomical segments for proteomics in comparison to their appropriate controls. High-dimensional proteome cluster analyses identified site-specific protein signatures in the suprarenal segment for AngII-infused mice (159 for Apoe -/- and 158 for Ldlr -/-) and the infrarenal segment for elastase-incubated mice (173). Network analysis revealed a predominance of inflammatory and coagulation factors in developing aneurysms, and a predominance of fibrosis-related pathways in established aneurysms for both models. To further substantiate our discovery platform, proteomics was performed on human infrarenal aortic aneurysm tissues as well as aortic tissue collected from age-matched controls. Protein processing and inflammatory pathways, particularly neutrophil-associated inflammation, dominated the proteome of the human aneurysm abdominal tissue. Aneurysmal tissue from both mouse and human had inflammation, coagulation, and protein processing signatures, but differed in the prevalence of neutrophil-associated pathways, and erythrocyte and oxidative stress-dominated networks in the human aneurysms. Conclusions Identifying changes unique to each mouse model will help to contextualize model-specific findings. Focusing on shared proteins between mouse experimental models or between mouse and human tissues may help to better understand the mechanisms for AAA and establish molecular bases for novel therapies.
Collapse
Affiliation(s)
- Stephanie Morgan
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Lang Ho Lee
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Arda Halu
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jessica S. Nicolau
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Hideyuki Higashi
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Anna H. Ha
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Jennifer R. Wen
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Alan Daugherty
- Department of Physiology, Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY, United States
| | - Peter Libby
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Scott J. Cameron
- Department of Cardiovascular Medicine, Section of Vascular Medicine, Heart Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Doran Mix
- Division of Vascular Surgery, Department of Surgery, University of Rochester School of Medicine, Rochester, NY, United States
| | - Elena Aikawa
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - A. Phillip Owens
- Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Sasha A. Singh
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Masanori Aikawa
- Cardiovascular Division, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
12
|
Kumar V, Ramnarayanan K, Sundar R, Padmanabhan N, Srivastava S, Koiwa M, Yasuda T, Koh V, Huang KK, Tay ST, Ho SWT, Tan ALK, Ishimoto T, Kim G, Shabbir A, Chen Q, Zhang B, Xu S, Lam KP, Lum HYJ, Teh M, Yong WP, So JBY, Tan P. Single-Cell Atlas of Lineage States, Tumor Microenvironment, and Subtype-Specific Expression Programs in Gastric Cancer. Cancer Discov 2022; 12:670-691. [PMID: 34642171 PMCID: PMC9394383 DOI: 10.1158/2159-8290.cd-21-0683] [Citation(s) in RCA: 277] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 09/27/2021] [Accepted: 10/07/2021] [Indexed: 01/07/2023]
Abstract
Gastric cancer heterogeneity represents a barrier to disease management. We generated a comprehensive single-cell atlas of gastric cancer (>200,000 cells) comprising 48 samples from 31 patients across clinical stages and histologic subtypes. We identified 34 distinct cell-lineage states including novel rare cell populations. Many lineage states exhibited distinct cancer-associated expression profiles, individually contributing to a combined tumor-wide molecular collage. We observed increased plasma cell proportions in diffuse-type tumors associated with epithelial-resident KLF2 and stage-wise accrual of cancer-associated fibroblast subpopulations marked by high INHBA and FAP coexpression. Single-cell comparisons between patient-derived organoids (PDO) and primary tumors highlighted inter- and intralineage similarities and differences, demarcating molecular boundaries of PDOs as experimental models. We complemented these findings by spatial transcriptomics, orthogonal validation in independent bulk RNA-sequencing cohorts, and functional demonstration using in vitro and in vivo models. Our results provide a high-resolution molecular resource of intra- and interpatient lineage states across distinct gastric cancer subtypes. SIGNIFICANCE We profiled gastric malignancies at single-cell resolution and identified increased plasma cell proportions as a novel feature of diffuse-type tumors. We also uncovered distinct cancer-associated fibroblast subtypes with INHBA-FAP-high cell populations as predictors of poor clinical prognosis. Our findings highlight potential origins of deregulated cell states in the gastric tumor ecosystem. This article is highlighted in the In This Issue feature, p. 587.
Collapse
Affiliation(s)
- Vikrant Kumar
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | | | - Raghav Sundar
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore.,Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,The N.1 Institute for Health, National University of Singapore, Singapore.,Singapore Gastric Cancer Consortium, Singapore
| | - Nisha Padmanabhan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | | | - Mayu Koiwa
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Tadahito Yasuda
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Vivien Koh
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kie Kyon Huang
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Su Ting Tay
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Shamaine Wei Ting Ho
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Angie Lay Keng Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore
| | - Takatsugu Ishimoto
- Gastrointestinal Cancer Biology, International Research Center for Medical Sciences (IRCMS), Kumamoto University, Kumamoto, Japan
| | - Guowei Kim
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Asim Shabbir
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Department of Surgery, University Surgical Cluster, National University Health System, Singapore
| | - Qingfeng Chen
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore.,Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Singapore
| | - Biyan Zhang
- Singapore Immunology Network (SIgN), A*STAR, Singapore
| | - Shengli Xu
- Singapore Immunology Network (SIgN), A*STAR, Singapore.,Department of Physiology, National University of Singapore, Singapore
| | - Kong-Peng Lam
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, Singapore.,Singapore Immunology Network (SIgN), A*STAR, Singapore.,School of Biological Sciences, Nanyang Technological University, Singapore
| | | | - Ming Teh
- Department of Pathology, National University Health System, Singapore
| | - Wei Peng Yong
- Department of Haematology-Oncology, National University Cancer Institute, National University Health System, Singapore.,Singapore Gastric Cancer Consortium, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Jimmy Bok Yan So
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore.,Singapore Gastric Cancer Consortium, Singapore.,Department of Surgery, University Surgical Cluster, National University Health System, Singapore.,Division of Surgical Oncology, National University Cancer Institute, Singapore
| | - Patrick Tan
- Cancer and Stem Cell Biology Program, Duke-NUS Medical School, Singapore.,Singapore Gastric Cancer Consortium, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore.,Department of Physiology, National University of Singapore, Singapore.,Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.,SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore.,Corresponding Author: Patrick Tan, Cancer and Stem Cell Biology Program, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore. Phone: 65-6516-1783; Fax: 65-6221-2402; E-mail:
| |
Collapse
|
13
|
Recent Advances in Diabetic Kidney Diseases: From Kidney Injury to Kidney Fibrosis. Int J Mol Sci 2021; 22:ijms222111857. [PMID: 34769288 PMCID: PMC8584225 DOI: 10.3390/ijms222111857] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 10/08/2021] [Accepted: 10/30/2021] [Indexed: 12/14/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of chronic kidney disease and end-stage renal disease. The natural history of DKD includes glomerular hyperfiltration, progressive albuminuria, declining estimated glomerular filtration rate, and, ultimately, kidney failure. It is known that DKD is associated with metabolic changes caused by hyperglycemia, resulting in glomerular hypertrophy, glomerulosclerosis, and tubulointerstitial inflammation and fibrosis. Hyperglycemia is also known to cause programmed epigenetic modification. However, the detailed mechanisms involved in the onset and progression of DKD remain elusive. In this review, we discuss recent advances regarding the pathogenic mechanisms involved in DKD.
Collapse
|
14
|
Farzamikia N, Baradaran B, Mostafavi S, Ahmadian E, Hosseiniyan Khatibi SM, Zununi Vahed S, Ardalan M. Podocyte-derived microparticles in IgA nephropathy. Biomed Pharmacother 2021; 141:111891. [PMID: 34237594 DOI: 10.1016/j.biopha.2021.111891] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/06/2021] [Accepted: 06/28/2021] [Indexed: 12/21/2022] Open
Abstract
Microparticles are a general term for different types of cell plasma membrane-originated vesicles that are released into the extracellular environment. The paracrine action of these nano-sized vesicles is crucial for intercellular communications through the transfer of diverse lipids, cytosolic proteins, RNA as well as microRNAs. The progression of different diseases influences the composition, occurrence, and functions of these cell-derived particles. Podocyte injury has been shown to have an important role in the pathophysiology of many glomerular diseases including IgA nephropathy (IgAN). This review would focus on the possible potential of podocyte-derived microparticles detected in urine to be used as a diagnostic tool in IgAN.
Collapse
Affiliation(s)
- Negin Farzamikia
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soroush Mostafavi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | | |
Collapse
|
15
|
Effect of UVA1 on hypertrophic scarring in the rabbit ear model. Biosci Rep 2021; 40:221742. [PMID: 31894858 PMCID: PMC6974420 DOI: 10.1042/bsr20190007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/28/2019] [Accepted: 12/19/2019] [Indexed: 01/30/2023] Open
Abstract
Hypertrophic scars (HTSs) are common and cause functional and psychological morbidity. UVA1 (340-400 nm) phototherapy has been previously shown to be effective in the treatment of localized scleroderma, systemic sclerosis, and POEMS syndrome with minimal side effects, all of which are presented as collagen fibrils hyperplasia that is common with scarring in skin histology. In the present study, we aimed to investigate the impact of UVA1 on the protein expression of TGF-β signal pathway and myofibroblasts in a rabbit model of cutaneous scarring. Full-thickness skin wounds (2 cm × 5 cm in diameter) were made in New Zealand white rabbits to establish the hypertrophic scarring model. New Zealand white rabbits were divided into two treatment groups (n=30 wounds per group with an equal number of controls): medium-dose of UVA1 phototherapy group: 60 J/cm2; high-dose of UVA1 phototherapy group: 110 J/cm2. Left ears were used for treatment and the right ones were used for control. Treatment was administered five times weekly for 6 weeks. Treated and untreated control wounds were harvested at various time points and examined by histologic examination, immunohistochemical assessment, and ultrastructural evaluation. The results showed that UVA1 phototherapy caused a significant reduction in dermal thickness by histological features, whereas the scar index was descended significantly in both medium- and high-dose UVA1 groups compared with the control group. Examination of immunohistochemistry also revealed a marked suppression of tissue growth factor-β (TGF-β) (both medium- and high-dose), α smooth muscle actin (α-SMA) (only high-dose), and tissue inhibitor of metalloproteinase 1 (TIMP-1) (only high-dose), and apparent increase in matrix metalloproteinases (MMP-1) (both medium- and high-dose) compared with the control. The ultrastructural evaluation showed the collagen fibers' diameter had shrunk, and that fibroblastic cytoplasm was not affluent and in a quiescent stage. These findings of the present study suggested that administration of UVA1 irradiation is effective to improve the experimental HTS model and raises a possibility of the therapeutic approach of UVA1 in the scar. Although not directly examined in the present study, MMP inhibition is hypothesized to be responsible for this effect. However, early UVA1 treatment could not prevent the formation of scar model.
Collapse
|
16
|
Chang CJ, Taniguchi A. Establishment of a Nanopatterned Renal Disease Model by Mimicking the Physical and Chemical Cues of a Diseased Mesangial Cell Microenvironment. ACS APPLIED BIO MATERIALS 2021; 4:1573-1583. [PMID: 35014506 DOI: 10.1021/acsabm.0c01406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Modulation of mesangial cell (MC) response by in vitro disease models offers therapeutic strategies for the treatment of several glomerular diseases. However, traditional cell culture models lack the nanostructured extracellular matrix (ECM), which has unique physical and chemical properties, so they poorly reflect the complexities of the native microenvironment. Therefore, a cell disease model with ECM nanostructures is required to better mimic the in vivo diseased nanoenvironment. To establish a renal disease model, we used a titanium dioxide-based disease-mimic nanopattern as the physical cues and transforming growth factor-beta 1 (TGF-β1) as a chemical cue. The effects of this renal disease model on proliferation and mesangial matrix (MM) component changes in the SV40MES13 (MES13) mouse mesangial cell line were evaluated. Our results showed that both the presence of the disease-mimic nanopattern and TGF-β1 intensified proliferation and resulted in increased type I collagen and fibronectin and decreased type IV collagen expressions in MES13 cells. These effects could be involved in increased TGF-β type I receptor expression in MES13 cells. The intracellular reactive oxygen species (ROS) level as a biomarker of this renal disease model indicated that the cells were in a diseased state. A small molecule A83-01 and known drug dexamethasone markedly attenuated the intracellular ROS production in MES13 that was induced by the disease-mimic nanopattern and TGF-β1. These results highlight the significant effects of physical and chemical cues in facilitating disease-like behavior in MES13 cells, providing an important theoretical basis for developing a drug screening platform for glomerular diseases.
Collapse
Affiliation(s)
- Chia-Jung Chang
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.,Department of Nanoscience and Nanoengineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| | - Akiyoshi Taniguchi
- Research Center for Functional Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan.,Department of Nanoscience and Nanoengineering, Graduate School of Advanced Science and Engineering, Waseda University, 3-4-1 Okubo, Shinjuku-ku, Tokyo 169-8555, Japan
| |
Collapse
|
17
|
Yu X, Su Q, Geng J, Liu H, Liu Y, Liu J, Shi Y, Zou Y. Ginkgo biloba leaf extract prevents diabetic nephropathy through the suppression of tissue transglutaminase. Exp Ther Med 2021; 21:333. [PMID: 33732306 PMCID: PMC7903480 DOI: 10.3892/etm.2021.9764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 01/18/2021] [Indexed: 12/28/2022] Open
Abstract
The present study aimed to investigate the preventive effects of Ginkgo biloba leaf extract (GBE) against extracellular matrix (ECM) accumulation in a streptozotocin (STZ)-induced rat model of diabetic nephropathy (DN), and to determine its underlying molecular mechanism. In vivo, a rat model of DN was established by intraperitoneal injection of STZ, and the rats were subsequently administered GBE. The results demonstrated that GBE significantly decreased blood glucose, the urine protein excretion rate and ECM accumulation in DN rats. In addition, the development of DN significantly induced tissue transglutaminase (tTG) protein expression, which was detected by immunohistochemistry, western blotting and PCR analyses, while GBE administration decreased tTG expression in the diabetic kidney. In vitro, rat glomerular mesangial cells (HBZY-1 cells) cultured with high glucose were also treated with GBE. The concentrations of tTG, fibronectin, type IV collagen, transforming growth factor (TGF)-β and connective tissue growth factor (CTGF) were detected via ELISA. The results demonstrated that GBE notably decreased the concentration of these proteins, and tTG expression was positively associated with TGF-β. GBE also suppressed tTG expression of high glucose-treated HBZY-1 cells in a concentration-dependent manner. Furthermore, tTG protein expression was detected in high glucose-treated HBZY-1 cells transfected with small interfering RNA (siRNA) oligonucleotides against TGF-β and CTGF to investigate a possible mechanism of GBE-mediated inhibition of tTG. The results demonstrated that the tTG levels remained unchanged in CTGF siRNA-transfected cells, but were decreased in the GBE + CTGF siRNA group compared with the control siRNA group, suggesting that tTG may not be regulated by CTGF, and the inhibitory effect of GBE on tTG may not be associated with the direct inhibition of CTGF. However, tTG expression was decreased following the transfection with TGF-β siRNA, in which levels of tTG were similar compared with both the GBE group and GBE + TGF-β siRNA group, indicating that tTG may be regulated by TGF-β, and that the GBE-induced repression of tTG expression may be associated with the downregulation of TGF-β. Taken together, the results of the present study suggest that GBE prevented ECM accumulation by suppressing tTG expression in DN, which was predominantly mediated by TGF-β.
Collapse
Affiliation(s)
- Xiaoyan Yu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qing Su
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jianan Geng
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hui Liu
- Department of Anatomy, College of Basic Medical Science, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yumeng Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jinming Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yan Shi
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yinggang Zou
- Department of Obstetrics and Gynecology, The Second Hospital, Jilin University, Changchun, Jilin 130041, P.R. China
| |
Collapse
|
18
|
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
|
19
|
Xu S, He L, Ding K, Zhang L, Xu X, Wang S, Qian X. Tanshinone IIA Ameliorates Streptozotocin-Induced Diabetic Nephropathy, Partly by Attenuating PERK Pathway-Induced Fibrosis. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:5773-5782. [PMID: 33408464 PMCID: PMC7780857 DOI: 10.2147/dddt.s257734] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
Purpose Tanshinone IIA (Tan IIA), a compound extracted from Salvia miltiorrhiza, can improve type II diabetes, while the molecular mechanisms underlying Tan IIA-mediated protective effects in diabetic nephropathy are unclear. This study explored the protective actions of Tan IIA on renal tissues in streptozotocin (STZ)-induced diabetic nephropathy. Materials and Methods Tan IIA (2, 4, 8 mg/kg/day) was daily administered to STZ-treated rats by intraperitoneal injection for 42 days. The morphologic pathology was evaluated by hematoxylin-eosin and Masson’s trichrome staining, and transmission electron microscopy. The protein expression levels in renal tissues were evaluated by Western blotting and immunohistochemistry; the mRNA expression level was determined by quantitative real-time PCR. Results Tan IIA at 2 and 4 mg/kg attenuated the increase in the levels of uric acid and blood urea nitrogen and restored the reduction in the superoxide dismutase activity in the serum of the diabetic rats. Tan IIA at 2 and 4 mg/kg, but not 8 mg/kg, ameliorated the thickening of renal tubule in the diabetic rats; Tan IIA at 2 and 4 and 8 mg/kg attenuated the thickening of glomerular basement membrane and the collagen deposition in the renal tissues of the diabetic rats. Tan IIA treatment at 2, 4, 8 mg/kg decreased the expression levels of transforming growth factor-beta1, TSP-1, Grp78 and CHOP in the diabetic rats. Tan IIA at 2 and 4 and 8 mg/kg attenuated the increase in the protein levels of p-PERK, p-elf2α and ATF-4 from the renal tissues of diabetic rats, while the protein level of AFT-6 and the mRNA expression levels of XBP-1t, XBP-1s and p58IPK in the renal tissues were not affected by STZ or Tan IIA treatment. Conclusion Tan IIA-mediated protective effects on the STZ-induced diabetic nephropathy may be associated with the reduced endoplasmic reticulum stress via attenuating PERK signaling activities.
Collapse
Affiliation(s)
- Shujuan Xu
- Department of Surgery, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Lianjun He
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Keke Ding
- Department of Urology, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Lingling Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, People's Republic of China
| | - Xinhui Xu
- School of Pharmacy, Wannan Medical College, Wuhu, People's Republic of China
| | - Sheng Wang
- Department of Pharmacy, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| | - Xueyi Qian
- Precision Medicine Centre, Yijishan Hospital of Wannan Medical College, Wuhu, People's Republic of China
| |
Collapse
|
20
|
He X, Zhang J, Luo L, Shi J, Hu D. New Progress of Adipose-derived Stem Cells in the Therapy of Hypertrophic Scars. Curr Stem Cell Res Ther 2020; 15:77-85. [PMID: 31483236 DOI: 10.2174/1574888x14666190904125800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 11/01/2018] [Accepted: 07/10/2019] [Indexed: 12/30/2022]
Abstract
Burns are a global public health issue of great concern. The formation of scars after burns and physical dysfunction of patients remain major challenges in the treatment of scars. Regenerative medicine based on cell therapy has become a hot topic in this century. Adipose-derived stem cells (ADSCs) play an important role in cellular therapy and have become a promising source of regenerative medicine and wound repair transplantation. However, the anti-scarring mechanism of ADSCs is still unclear yet. With the widespread application of ADSCs in medical, we firmly believe that it will bring great benefits to patients with hypertrophic scars.
Collapse
Affiliation(s)
- Xiang He
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, Shaanxi, China
| | - Julei Zhang
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, Shaanxi, China
| | - Liang Luo
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, Shaanxi, China
| | - Jihong Shi
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, Shaanxi, China
| | - Dahai Hu
- Department of Burns and Cutaneous Surgery, Xijing Hospital, the Fourth Military Medical University, Xi' an, Shaanxi, China
| |
Collapse
|
21
|
Cai J, Liu B, Guo T, Zhang Y, Wu X, Leng J, Zhu N, Guo J, Zhou Y. Effects of thromboxane prostanoid receptor deficiency on diabetic nephropathy induced by high fat diet and streptozotocin in mice. Eur J Pharmacol 2020; 882:173254. [PMID: 32553735 DOI: 10.1016/j.ejphar.2020.173254] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 05/14/2020] [Accepted: 06/05/2020] [Indexed: 02/05/2023]
Abstract
Diabetic nephropathy (DN), one of the main causes of end-stage renal disease, still remains as a challenge of clinical management. This study aimed to determine whether deficiency of the thromboxane (TX) prostanoid receptor (TP), which mediates the contractile activities of all prostanoids, alleviates the development of DN and if so, to examine the underlying mechanism(s). Diabetes was induced by high fat diet and streptozotocin injection in wild-type (WT) mice and those with TP deficiency (TP-/-). Here we show that WT and TP-/- mice developed diabetes with a similar blood glucose level; however, signs of renal functional impairments and pathologies occurred to a lesser extent in TP-/- than in WT mice. Also, the extent of an increase in the expression level of transforming growth factor-β1 (TGF-β1), a common pathological mediator of DN, in diabetic renal cortexes of TP-/- mice was lower than that of WT counterparts. Moreover, we noted that expression levels of cyclooxygenase (COX)-2 and calcium-dependent phospholipase A2 (cPLA2) as well as levels of prostaglandin E2 and TXA2 in diabetic renal cortexes were increased as compared to those of non-diabetic conditions. These results thus demonstrate that possibly due to up-regulated cPLA2 and COX-2 that lead to increased prostanoid syntheses in diabetic renal cortexes, TP-/- alleviates DN development. In addition, our results suggest that such an effect of TP-/- might be related to the suppression of TGF-β1 up-regulation that is commonly associated with the disease condition.
Collapse
Affiliation(s)
- Juyu Cai
- Department of Medicine, Medical College of Jiaying University, Meizhou, China; Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Bin Liu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Tingting Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingzhan Zhang
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Xiangzhong Wu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jing Leng
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Ningxia Zhu
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Jinwei Guo
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China
| | - Yingbi Zhou
- Cardiovascular Research Center, Shantou University Medical College, Shantou, China.
| |
Collapse
|
22
|
Pereira BM, Thieme K, de Araújo L, Rodrigues AC. Lack of adiponectin in mice accelerates high-fat diet-induced progression of chronic kidney disease. Life Sci 2020; 257:118061. [PMID: 32652137 DOI: 10.1016/j.lfs.2020.118061] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/27/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022]
|
23
|
Ren ZX, Zou YL, Tan X, Chen Z, Li L, Wang ZX. Modified autologous adipose transplantation in the treatment of depressed scars: an experimental study. Am J Transl Res 2020; 12:708-717. [PMID: 32194917 PMCID: PMC7061831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 02/01/2020] [Indexed: 06/10/2023]
Abstract
OBJECTIVE Complex depressed scars can cause tissue adhesion, resulting in serious joint dysfunction. In recent years, autologous adipose and adipose-derived stem cells have been widely used to treat depressed scars, but there are still limitations in these treatment that should be resolved. This study aimed to investigate the therapeutic effects of adipose tissues collected with modified technique on the depressed scars in animals. METHODS The adipose tissues were collected with a forward technique, and tissue viability in vitro and the survival of transplanted tissues in in nude mice were further assessed. Furthermore, the therapeutic effects of adipose tissues collected with new technique and traditional technique on the depressed scars were explored in an animal model of bleomycin induced scar formation. RESULTS The adipose tissues collected with the new technique had a higher glucose transport (P<0.01); after transplantation into the nude mice, the amount of residual tissues and the survival rate in the modified group were higher than in the traditional group (P<0.05); electron microscopy showed the intercellular space was covered with reticular structure, in which there was a large amount of microvessel structure in the adipose tissue of the modified group; immunohistochemistry showed that the microvessel density (MVD) in the modified group increased significantly (P<0.01). At 28 d after transplantation into the scar animals, the dermal collagen fibers became thicker and showed regular arrangement, the myofibroblasts became regenerative and inflammation was improved as compared to blank control group. In the untreated scar group, the collagen fibers were loose and irregular, and a large amount of inflammatory cells was observed. In addition, the dermal expression of α-SMA and TGF-β1 in the transplantation group reduced significantly as compared to scar group (P<0.05). CONCLUSION The autologous adipose tissues collected with the new technique possess higher activity ad contain more. In scar animals, transplantation of these adipose tissues may improve the scar structure and inhibit the scar formation which may be related to the suppressed expression of α-SMA and TGF-β1.
Collapse
Affiliation(s)
- Zhang-Xia Ren
- Department of Burn and Plastic Surgery, Guang’an People’s HospitalGuang’an 638001, Sichuan, China
| | - Yan-Long Zou
- Department of Plastic Surgery, Southwest Hospital of Army Medical UniversityChongqing 400038, China
| | - Xi Tan
- Department of Plastic Surgery, Southwest Hospital of Army Medical UniversityChongqing 400038, China
| | - Zhuo Chen
- Department of Plastic Surgery, Southwest Hospital of Army Medical UniversityChongqing 400038, China
| | - Le Li
- Department of Plastic Surgery, Southwest Hospital of Army Medical UniversityChongqing 400038, China
| | - Zhen-Xiang Wang
- Department of Plastic Surgery, Wanjiayan Plastic Surgery HospitalChongqing 400084, China
| |
Collapse
|
24
|
Miguel V, Lamas S. Redox distress in organ fibrosis: The role of noncoding RNAs. OXIDATIVE STRESS 2020:779-820. [DOI: 10.1016/b978-0-12-818606-0.00037-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
25
|
Singh R, Alape D, de Lima A, Ascanio J, Majid A, Gangadharan SP. Regulatory T Cells in Respiratory Health and Diseases. Pulm Med 2019; 2019:1907807. [PMID: 31827925 PMCID: PMC6886321 DOI: 10.1155/2019/1907807] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 09/05/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Respiratory diseases compromise the health of millions of people all over the world and are strongly linked to the immune dysfunction. CD4+FOXP3+ T regulatory cells, also known as Tregs, have a central role maintaining tissue homeostasis during immune responses. Their activity and clinical impact have been widely studied in different clinical conditions including autoimmune diseases, inflammatory conditions, and cancer, amongst others. Tregs express transcription factor forkhead box P3 (FOXP3), which allows regulation of the immune response through anti-inflammatory cytokines such as IL-10 or transforming growth factor beta (TGF-β) and direct cell-to-cell interaction. Maintenance of immune tolerance is achieved via modulation of effector CD4+ T helper 1, 2 or 17 (Th1, Th2, Th17) cells by Tregs. This review highlights the recent progress in the understanding of Tregs in different disorders of the respiratory system.
Collapse
Affiliation(s)
- Rani Singh
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Daniel Alape
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Andrés de Lima
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Juan Ascanio
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Adnan Majid
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sidhu P. Gangadharan
- Division of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Wu X, Zhu J, Wei Y, Guan X, Zhang Y, Chen W, Gao B. MicroRNA-663 participates in myocardial fibrosis through interaction with TGF-β1. Exp Ther Med 2019; 18:3172-3176. [PMID: 31572558 DOI: 10.3892/etm.2019.7902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 05/31/2019] [Indexed: 12/11/2022] Open
Abstract
MicroRNA-663 (miRNA-663) regulates the expression of transforming growth factor β1 (TGF-β1), which participates in the pathogenesis of myocardial fibrosis. Therefore, microRNA-663 may also serve a role in myocardial fibrosis. The present study aimed to determine whether miRNA-663 participates in myocardial fibrosis via interaction with TGF-β1. In the present study, the expression of miRNA-663 was significantly downregulated, whereas that of TGF-β1 was significantly upregulated in the endomyocardial biopsies of patients with myocardial fibrosis compared with those in control necropsies. Pearson's correlation analysis revealed that the expression levels of miRNA-663 were negatively correlated with those of TGF-β1 in patients with myocardial fibrosis, but not in the controls. Receiver operating characteristic curve analysis demonstrated that the downregulation of miRNA-663 distinguished patients with myocardial fibrosis from controls. In the AC16 human cardiomyocyte cell line, miRNA-663 overexpression resulted in downregulated TGF-β1 expression, whereas exogenous TGF-β1 treatment exhibited no significant effects on miRNA-663 expression. These results indicate that miRNA-663 may participate in myocardial fibrosis, possibly through interaction with TGF-β1.
Collapse
Affiliation(s)
- Xiangyang Wu
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Jie Zhu
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yalin Wei
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Xinqiang Guan
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Yanchun Zhang
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Wensheng Chen
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Bingren Gao
- Department of Cardiac Surgery, The Second Hospital of Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
27
|
Liu Y, Yang Y, Wang Q, Kahaer A, Zhang J, Liao J, Abudureyimu M, Yahefu R, Qi J, Zhao L, Zhu J. Regulatory Effect of 1,25(OH)2D3 on TGF- β1 and miR-130b Expression in Streptozotocin-Induced Diabetic Nephropathy in Rats. Int J Endocrinol 2019; 2019:1231346. [PMID: 31781203 PMCID: PMC6875179 DOI: 10.1155/2019/1231346] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 09/17/2019] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE To investigate the role of microRNA-130b in 1,25(OH)2D3 mediated improvement of renal fibrosis via transforming growth factor-beta 1 in a rat model of diabetic nephropathy (DN). METHODS DN was induced in 30 rats by intraperitoneal injection of streptozotocin. These rats were randomly allocated to the DN group, TGF-β1 overexpression group (in situ injection of TGF-β1 lentivirus to kidney tissues), and TGF-β1 siRNA group (in situ injection of TGF-β1 siRNA lentivirus to kidney tissues). Rats with different expression levels of TGF-β1 were administered 1,25(OH)2D3 (0.03 μg/kg/d) or peanut oil as control. DN rats were treated only with peanut oil. All rats were randomly divided into five groups (n = 6 per group): TGF-β1 overexpression + oil, TGF-β1 overexpression + 1,25(OH)2D3, TGF-β1 siRNA + oil, TGF-β1 siRNA + 1,25(OH)2D3, and DN + oil groups. After 37 days, kidney samples were collected and the expression of TGF-β1 and miR-130b was determined by real-time PCR, western blotting, and immunohistochemistry. Hematoxylin and eosin staining and Masson staining were used to evaluate kidney morphological and fibrogenic changes. Differences were determined using ANOVA and Student's t-test. RESULTS RT-PCR, western blotting, and immunohistochemistry revealed that interference of TGF-β1 significantly decreased mRNA and protein levels of TGF-β1 in renal tissues of DN rats compared to those in renal tissues of rats overexpressing TGF-β1 (p < 0.05). Histological analysis showed that upregulated TGF-β1 led to disorganized kidney structure and severe kidney fibrosis. The expression of miR-130b was significantly lowered upon lentivirus-mediated overexpression of TGF-β1 than upon downregulation of TGF-β1 (p < 0.05). Treatment with 1,25(OH)2D3 led to a significant reduction of TGF-β1 at the mRNA and protein levels (both p < 0.05), improvement of renal structure and fibrosis, and an increase in miR-130b expression (p < 0.05). CONCLUSION TGF-β1 can decrease the expression of miR-130b in kidney tissues of DN rats. Moreover, miR-130b may be involved in the protective effect of 1,25(OH)2D3 on renal fibrosis via TGF-β1.
Collapse
Affiliation(s)
- Yuetong Liu
- Department of Endocrinology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, Xinjiang, China
| | - Ye Yang
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Qin Wang
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Apaer Kahaer
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jiyun Zhang
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jing Liao
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Mairemugu Abudureyimu
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Reyila Yahefu
- Department of No. 1 Cadres, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jing Qi
- Department of Chu Medical, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Lei Zhao
- Department of Chu Medical, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi 830063, Xinjiang, China
| | - Jun Zhu
- Department of Endocrinology, People's Hospital of Shenzhen Baoan District, The Second School of Clinical Medicine, Southern Medical University, Shenzhen, Guangdong 518101, China
| |
Collapse
|
28
|
Tang J, Yao D, Yan H, Chen X, Wang L, Zhan H. The Role of MicroRNAs in the Pathogenesis of Diabetic Nephropathy. Int J Endocrinol 2019; 2019:8719060. [PMID: 31885563 PMCID: PMC6914872 DOI: 10.1155/2019/8719060] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/07/2019] [Accepted: 10/22/2019] [Indexed: 02/08/2023] Open
Abstract
Diabetic nephropathy (DN) is one of the most common microvascular complications in diabetic patients; it is also an important cause of renal dysfunction, renal fibrosis, and end-stage renal disease. Unfortunately, the pathogenesis of DN is complex and has not yet been fully elucidated; hence, the pathogenesis of DN to determine effective treatments of crucial importance is deeply explored. Early DN research focuses on hemodynamic changes and metabolic disorders, and recent studies have shown the regulatory role of microRNAs (miRNAs) in genes, which may be a new diagnostic marker and therapeutic target for diabetic nephropathy. In this review, we summarize the recent advances in the clinical value and molecular mechanisms of miRNAs in DN, providing new ideas for the diagnosis and treatment of DN.
Collapse
Affiliation(s)
- Jian Tang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Deyi Yao
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Haiying Yan
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Xing Chen
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Linjia Wang
- Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| | - Huakui Zhan
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610075, Sichuan, China
| |
Collapse
|
29
|
Wang B, Ji G, Naeem H, Wang J, Kantharidis P, Powell D, Ricardo SD. The Use of Targeted Next Generation Sequencing to Explore Candidate Regulators of TGF-β1's Impact on Kidney Cells. Front Physiol 2018; 9:1755. [PMID: 30618784 PMCID: PMC6295563 DOI: 10.3389/fphys.2018.01755] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 11/20/2018] [Indexed: 12/26/2022] Open
Abstract
Aims/Hypothesis: Transforming growth factor-beta (TGF-β1) plays an important regulatory role in the progression of chronic kidney failure. Further, damage to kidney glomerular mesangial cells is central to the progression of diabetic nephropathy. The aim of this study was to explore the genetic associations between mRNA, microRNA, and epigenetics in mesangial cells in response to TGF-β1. Methods: The regulatory effects of TGF-β1 on mesangial cells were investigated at different molecular levels by treating mesangial cells with TGF-β1 for 3 days followed by genome-wide miRNA, RNA, DNA methylation, and H3K27me3 expression profiling using next generation sequencing (NGS). Results: Our results provide the first comprehensive, computationally integrated report of RNA-Seq, miRNA-Seq, and epigenomic analyses across all genetic variations, confirming the occurrence of DNA methylation and H3K27me3 in response to TGF-β1. Our findings show that the expression of KLF7 and Gja4 are involved in TGF-β1 regulated DNA methylation. Our data also provide evidence of the association between epigenetic changes and the expression of genes closely related to TGF-β1 regulation. Conclusion: This study has advanced our current knowledge of mechanisms that contribute to the expression of TGF-β1-regulated genes involved in the pathogenesis of kidney disease. The molecular underpinnings of TGF-β1 stimulation of kidney cells was determined, thereby providing a robust platform for further target exploration.
Collapse
Affiliation(s)
- Bo Wang
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Guanyu Ji
- Shenzhen E-GENE Tech Co., Ltd., Shenzhen, China
| | - Haroon Naeem
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia.,Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Junwen Wang
- Shenzhen E-GENE Tech Co., Ltd., Shenzhen, China
| | | | - David Powell
- Monash Bioinformatics Platform, Monash University, Clayton, VIC, Australia.,Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Sharon D Ricardo
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| |
Collapse
|
30
|
Oba S, Ayuzawa N, Nishimoto M, Kawarazaki W, Ueda K, Hirohama D, Kawakami-Mori F, Shimosawa T, Marumo T, Fujita T. Aberrant DNA methylation of Tgfb1 in diabetic kidney mesangial cells. Sci Rep 2018; 8:16338. [PMID: 30397232 PMCID: PMC6218490 DOI: 10.1038/s41598-018-34612-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 10/22/2018] [Indexed: 01/19/2023] Open
Abstract
Epigenetic modulation may underlie the progression of diabetic nephropathy (DN). Involvement of TGFB1 in mesangial fibrosis of DN led us to hypothesize that Tgfb1 DNA demethylation contributes to progression of DN. In primary mesangial cells from diabetic (db/db) mouse kidneys, demethylation of Tgfb1 DNA and upregulation of Tgfb1 mRNA progressed simultaneously. USF1 binding site in Tgfb1 promoter region were demethylated, and binding of USF1 increased, with decreased binding of DNMT1 in db/db compared with control. Given downregulation of Tgfb1 expression by folic acid, antioxidant Tempol reversed DNA demethylation, with increased and decreased recruitment of DNMT1 and USF1 to the promoter, resulting in decreased Tgfb1 expression in db/db mice. Addition of H2O2 to mesangial cells induced DNA demethylation and upregulated Tgfb1 expression. Finally, Tempol attenuated mesangial fibrosis in db/db mice. We conclude that aberrant DNA methylation of Tgfb1 due to ROS overproduction play a key to mesangial fibrosis during DN progression.
Collapse
Affiliation(s)
- Shigeyoshi Oba
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| | - Nobuhiro Ayuzawa
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Mitsuhiro Nishimoto
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Wakako Kawarazaki
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kohei Ueda
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Daigoro Hirohama
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | | | - Tatsuo Shimosawa
- Department of Clinical Laboratory, International University of Health and Welfare, School of Medicine, Mita Hospital IUHW, Tokyo, Japan
| | - Takeshi Marumo
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshiro Fujita
- Division of Clinical Epigenetics, Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
31
|
Fan B, Luk AOY, Chan JCN, Ma RCW. MicroRNA and Diabetic Complications: A Clinical Perspective. Antioxid Redox Signal 2018; 29:1041-1063. [PMID: 28950710 DOI: 10.1089/ars.2017.7318] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
SIGNIFICANCE The rising global prevalence of diabetes and its debilitating complications give rise to significant disability and premature mortality. Due to the silent nature of diabetes and its vascular complications, and limitations in current methods for detection, there is a need for novel biomarkers for early detection and prognosis. Recent Advances: Metabolic memory and epigenetic factors are important in the pathogenesis of diabetic complications and interact with genetic variants, metabolic factors, and clinical risk factors. Micro(mi)RNAs interact with epigenetic mechanisms and pleiotropically mediate the effects of hyperglycemia on the vasculature. Utilizing mature profiling techniques and platforms, an increasing number of miRNA signatures and interaction networks have been identified for diabetes and its related cardiorenal complications. As a result, these short, single-stranded molecules are emerging as potential diagnostic and predictive tools in human studies, and may function as disease biomarkers, as well as treatment targets. CRITICAL ISSUES However, there is complex interaction between the genome and epigenome. The regulation of miRNAs may differ across species and tissues. Most profiling studies to date lack validation, often requiring large, well-characterized cohorts and reliable normalization strategies. Furthermore, the incremental benefits of miRNAs as biomarkers, beyond prediction provided by traditional risk factors, are critical issues to consider, yet often neglected in published studies. FUTURE DIRECTIONS All in all, the future for miRNA-based diagnostics and therapeutics for diabetic complications appears promising. Improved understanding of the complex mechanisms underlying miRNA dysregulation, and more well-designed studies utilizing prospective samples would facilitate the translation to clinical use.
Collapse
Affiliation(s)
- Baoqi Fan
- 1 Department of Medicine and Therapeutics, The Chinese University of Hong Kong , Shatin, China
| | - Andrea On Yan Luk
- 1 Department of Medicine and Therapeutics, The Chinese University of Hong Kong , Shatin, China .,2 Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong , Shatin, China
| | - Juliana Chung Ngor Chan
- 1 Department of Medicine and Therapeutics, The Chinese University of Hong Kong , Shatin, China .,2 Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong , Shatin, China .,3 Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Prince of Wales Hospital, Shatin, China .,4 The Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine , Shatin, China
| | - Ronald Ching Wan Ma
- 1 Department of Medicine and Therapeutics, The Chinese University of Hong Kong , Shatin, China .,2 Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong , Shatin, China .,3 Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong , Prince of Wales Hospital, Shatin, China .,4 The Chinese University of Hong Kong-Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine , Shatin, China
| |
Collapse
|
32
|
Kato M. Noncoding RNAs as therapeutic targets in early stage diabetic kidney disease. Kidney Res Clin Pract 2018; 37:197-209. [PMID: 30254844 PMCID: PMC6147183 DOI: 10.23876/j.krcp.2018.37.3.197] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 02/01/2023] Open
Abstract
Diabetic kidney disease (DKD) is a major renal complication of diabetes that leads to renal dysfunction and end-stage renal disease (ESRD). Major features of DKD include accumulation of extracellular matrix proteins and glomerular hypertrophy, especially in early stage. Transforming growth factor-β plays key roles in regulation of profibrotic genes and signal transducers such as Akt kinase and MAPK as well as endoplasmic reticulum stress, oxidant stress, and autophagy related to hypertrophy in diabetes. Many drugs targeting the pathogenic signaling in DKD (mostly through protein-coding genes) are under development. However, because of the limited number of protein-coding genes, noncoding RNAs (ncRNAs) including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) are attracting more attention as potential new drug targets for human diseases. Some miRNAs and lncRNAs regulate each other (by hosting, enhancing transcription from the neighbor, hybridizing each other, and changing chromatin modifications) and create circuits and cascades enhancing the pathogenic signaling in DKD. In this short and focused review, the functional significance of ncRNAs (miRNAs and lncRNAs) in the early stages of DKD and their therapeutic potential are discussed.
Collapse
Affiliation(s)
- Mitsuo Kato
- Beckman Research Institute of City of Hope, Duarte, CA, USA
| |
Collapse
|
33
|
Dewanjee S, Bhattacharjee N. MicroRNA: A new generation therapeutic target in diabetic nephropathy. Biochem Pharmacol 2018; 155:32-47. [DOI: 10.1016/j.bcp.2018.06.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
|
34
|
Roles of the TGF-β⁻VEGF-C Pathway in Fibrosis-Related Lymphangiogenesis. Int J Mol Sci 2018; 19:ijms19092487. [PMID: 30142879 PMCID: PMC6163754 DOI: 10.3390/ijms19092487] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 08/16/2018] [Accepted: 08/18/2018] [Indexed: 12/11/2022] Open
Abstract
Lymphatic vessels drain excess tissue fluids to maintain the interstitial environment. Lymphatic capillaries develop during the progression of tissue fibrosis in various clinical and pathological situations, such as chronic kidney disease, peritoneal injury during peritoneal dialysis, tissue inflammation, and tumor progression. The role of fibrosis-related lymphangiogenesis appears to vary based on organ specificity and etiology. Signaling via vascular endothelial growth factor (VEGF)-C, VEGF-D, and VEGF receptor (VEGFR)-3 is a central molecular mechanism for lymphangiogenesis. Transforming growth factor-β (TGF-β) is a key player in tissue fibrosis. TGF-β induces peritoneal fibrosis in association with peritoneal dialysis, and also induces peritoneal neoangiogenesis through interaction with VEGF-A. On the other hand, TGF-β has a direct inhibitory effect on lymphatic endothelial cell growth. We proposed a possible mechanism of the TGF-β–VEGF-C pathway in which TGF-β promotes VEGF-C production in tubular epithelial cells, macrophages, and mesothelial cells, leading to lymphangiogenesis in renal and peritoneal fibrosis. Connective tissue growth factor (CTGF) is also involved in fibrosis-associated renal lymphangiogenesis through interaction with VEGF-C, in part by mediating TGF-β signaling. Further clarification of the mechanism might lead to the development of new therapeutic strategies to treat fibrotic diseases.
Collapse
|
35
|
Huang YQ, Huang C, Li J, Zhang B, Feng YQ. The association of miR-29a with proteinuria in essential hypertension. J Hum Hypertens 2018; 32:775-780. [PMID: 30127486 DOI: 10.1038/s41371-018-0097-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2018] [Revised: 07/12/2018] [Accepted: 07/31/2018] [Indexed: 12/22/2022]
Abstract
Recently, miRNAs have emerged as new indirect markers of inflammation that are associated with adverse outcomes in cardiovascular disease. The aim of the study was to evaluate the relationship between miR29a and proteinuria in hypertension. Fifty patients with normal albuminuria, fifty patients with micro-albuminuria, and fifty patients with macro-albuminuria were enrolled. The highest levels of miR-29a and transforming growth factor-β1 (TGF-β1) were observed in the macro-albuminuria group, followed by the micro-albuminuria and the normal albuminuria groups. The level of miR-29a was negatively correlated with the glomerular filtration rate, but was positively correlated with C-reactive protein, TGF-β1, and the urinary albumin to creatinine ratio (UACR). Circulating miR-29a was found to be significantly and independently associated with proteinuria. Our findings showed that miR-29a reflects the pathogenesis of hypertensive nephropathy and may serve as a potential non-invasive marker for detecting early stages of hypertensive nephropathy.
Collapse
Affiliation(s)
- Yu-Qing Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Cheng Huang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Jie Li
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China
| | - Bin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China.
| | - Ying-Qing Feng
- Department of Cardiology, Guangdong Cardiovascular Institute, Hypertension Research Laboratory, Guangdong General Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, 510080, Guangzhou, China.
| |
Collapse
|
36
|
Wu MY, Chen CS, Yiang GT, Cheng PW, Chen YL, Chiu HC, Liu KH, Lee WC, Li CJ. The Emerging Role of Pathogenesis of IgA Nephropathy. J Clin Med 2018; 7:225. [PMID: 30127305 PMCID: PMC6112037 DOI: 10.3390/jcm7080225] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 08/12/2018] [Accepted: 08/16/2018] [Indexed: 12/20/2022] Open
Abstract
IgA nephropathy is an autoimmune disease induced by fthe ormation of galactose-deficient IgA1 and anti-glycans autoantibody. A multi-hit hypothesis was promoted to explain full expression of IgA nephropathy. The deposition of immune complex resulted in activation of the complement, increasing oxidative stress, promoting inflammatory cascade, and inducing cell apoptosis via mesangio-podocytic-tubular crosstalk. The interlinked signaling pathways of immune-complex-mediated inflammation can offer a novel target for therapeutic approaches. Treatments of IgA nephropathy are also summarized in our review article. In this article, we provide an overview of the recent basic and clinical studies in cell molecular regulation of IgAN for further treatment interventions.
Collapse
Affiliation(s)
- Meng-Yu Wu
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Chien-Sheng Chen
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Giou-Teng Yiang
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Pei-Wen Cheng
- Yuh-Ing Junior College of Health Care & Management, Kaohsiung 807, Taiwan.
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan.
| | - Yu-Long Chen
- Department of Emergency Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Emergency Medicine, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Hsiao-Chen Chiu
- Department of Obstetrics and Gynecology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 231, Taiwan.
- Department of Obstetrics and Gynecology, School of Medicine, Tzu Chi University, Hualien 970, Taiwan.
| | - Kuan-Hung Liu
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine National Cheng Kung University, Tainan 704, Taiwan.
| | - Wen-Chin Lee
- Division of Nephrology, Department of Internal Medicine, Chang Bing Show Chwan Memorial Hospital, Changhua 505, Taiwan.
| | - Chia-Jung Li
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
| |
Collapse
|
37
|
Allinovi M, De Chiara L, Angelotti ML, Becherucci F, Romagnani P. Anti-fibrotic treatments: A review of clinical evidence. Matrix Biol 2018; 68-69:333-354. [DOI: 10.1016/j.matbio.2018.02.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 02/06/2023]
|
38
|
NaoXinTong Capsules inhibit the development of diabetic nephropathy in db/db mice. Sci Rep 2018; 8:9158. [PMID: 29904053 PMCID: PMC6002396 DOI: 10.1038/s41598-018-26746-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 05/04/2018] [Indexed: 12/17/2022] Open
Abstract
NaoXinTong Capsule (NXT), a Chinese medicine, is currently used to treat patients with cardiovascular and cerebrovascular diseases. Clinical observations indicate its anti-diabetic functions with unclear mechanisms. Herein, we report the effect of NXT on diabetic nephropathy (DN). Type 2 diabetic db/db mice were treated with NXT for 14 weeks. In the course of treatment, NXT reduced diabetes-increased glucose levels and improved renal functions. At the end of treatment, we found that NXT ameliorated serum lipid profiles and other biochemical parameters. In the kidney, NXT inhibited mesangial matrix expansion, expression of vascular endothelial growth factor A, fibronectin, advanced glycation end product and its receptor. Meanwhile, it reduced the diabetes-induced podocyte injury by increasing WT1 and nephrin expression. In addition, NXT inhibited accumulation of extracellular matrix proteins by increasing MMP2/9 expression through inactivation of TGFβ/Smad pathway and CTGF expression. Mechanically, NXT activated insulin signaling pathway by increasing expression of INSR, IRS and FGF21, phosphorylation of Akt and AMPKα in the liver, INSR phosphorylation in the kidney, and FGF21 and GLUT4 expression in adipose tissue and skeletal muscle. Taken together, our study demonstrates that NXT inhibits DN by ameliorating glucose/lipid metabolism, maintaining tissue structure integrity, and correcting diabetes-induced renal dysfunctions.
Collapse
|
39
|
Motawi TK, Shehata NI, ElNokeety MM, El-Emady YF. Potential serum biomarkers for early detection of diabetic nephropathy. Diabetes Res Clin Pract 2018; 136:150-158. [PMID: 29253627 DOI: 10.1016/j.diabres.2017.12.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 10/28/2017] [Accepted: 12/12/2017] [Indexed: 12/09/2022]
Abstract
AIM Diabetic nephropathy (DN) is considered as one of the diabetic complications affecting up to 40% of patients with type 1 or type 2 diabetes. In clinical practice, the frequently used markers of renal disease and progression are serum creatinine, estimated glomerular filtration rate (eGFR) and albuminuria. The aim of this study is to determine new biomarkers in human serum which are promising for early detection of DN. METHODS This study included 50 patients with type 2 diabetes mellitus (T2DM) and 25 clinically healthy individuals. The patients were divided into two groups; group I included 25 T2DM patients with normoalbuminuria, and group II consisted of 25 T2DM patients with microalbuminuria. In all groups, neutrophil gelatinase-associated lipocalin (NGAL), β-trace protein (βTP) and microRNA- 130b (miR-130b) were estimated. RESULTS The serum levels of NGAL and βTP were significantly elevated in T2DM patients with microalbuminuria (group II) compared with T2DM patients with normoalbuminuria (group I) and control subjects but there was no significant difference between group I and control subjects. Serum miR-130b level was significantly decreased in patients with T2DM (groups I and II) compared with healthy control subjects, with a higher decrease in their levels in group II compared with group I. CONCLUSION Our results suggest that serum NGAL and βTP as tubular and glomerular markers respectively, together with serum miR-130b may be independent and reliable biomarkers for early detection of DN in patients with T2DM.
Collapse
Affiliation(s)
- Tarek Kamal Motawi
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Nagwa Ibrahim Shehata
- Biochemistry Department, Faculty of Pharmacy, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Mahmoud Mohamed ElNokeety
- Department of Internal Medicine, Faculty of Medicine, Cairo University, Kasr Al Ainy, St. Cairo 11562, Egypt
| | - Yasmin Farid El-Emady
- The Holding Company for Biological Products & Vaccines (VACSERA), 51 Wezaret ElZeraa St., Agouza, Giza 12622, Egypt.
| |
Collapse
|
40
|
Tacrolimus-induced nephrotoxicity in mice is associated with microRNA deregulation. Arch Toxicol 2018; 92:1539-1550. [DOI: 10.1007/s00204-018-2158-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/17/2018] [Indexed: 01/16/2023]
|
41
|
Sauriasari R, Pratiwi MY. Urinary TGF-β1 was not independently associated with renal function in diabetes mellitus. Diabetes Metab Syndr Obes 2018; 11:597-602. [PMID: 30323640 PMCID: PMC6181110 DOI: 10.2147/dmso.s172057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Several clinical studies have shown increased level of urinary TGF-β1 in diabetic nephropathy patients and its correlation with urine albumin-to-creatinine ratio (UACR), but other studies showed different results. Because of this contradiction, this study aims to analyze the correlation between urinary TGF-β1 concentration and UACR, and also estimated glomerular filtration rate (eGFR) in type 2 diabetes mellitus (DM) patients by controlling some confounding factors. METHODS This was a cross-sectional study, and the samples were obtained using consecutive sampling technique. The study was performed on 99 subjects (62 DM normoalbuminuria patients, 27 DM albuminuria patients, and 10 non-DM patients as controls) at Pasar Minggu Community Health Center. Urinary TGF-β1 concentration was measured by ELISA, and UACR was measured using immunoturbidimetry and an enzymatic colorimetric method. The eGFR value was calculated based on serum creatinine using Chronic Kidney Disease Epidemiology Collaboration equation. The results were then subjected to be analyzed statistically. RESULTS There was an increase of urinary TGF-β1 in albuminuria (326.49±48.98) as compared to normoalbuminuria (290.54±30.71) and non-DM subjects (229.83±31.90), but the values did not differ statistically (P=0.790). In addition, no correlation was observed between urinary TGF-β1 and UACR (r=-0.084, P=0.410) and eGFR (r=0.155, P=0.125), but a correlation was found with SBP (r=-0.224, P=0.026). Linear regression analysis showed that urinary TGF-β1 and HbA1c could predict UACR, but only HbA1c could be considered as a significant predictor of UACR. CONCLUSION There is an increase of urinary TGF-β1 concentrations in albuminuria patients clinically, but not statistically. The concentration of TGF-β1 was not correlated with UACR and eGFR, but correlated with SBP. Since TGF-β1 could be interfered by many factors, including hypertension and its medication, urinary TGF-β1 might not be independently associated with renal function in diabetes.
Collapse
Affiliation(s)
- Rani Sauriasari
- Faculty of Pharmacy, Universitas Indonesia, Depok, Indonesia,
| | | |
Collapse
|
42
|
Barutta F, Bellini S, Mastrocola R, Bruno G, Gruden G. MicroRNA and Microvascular Complications of Diabetes. Int J Endocrinol 2018; 2018:6890501. [PMID: 29707000 PMCID: PMC5863305 DOI: 10.1155/2018/6890501] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 12/28/2017] [Accepted: 01/11/2018] [Indexed: 12/11/2022] Open
Abstract
In the last decade, miRNAs have received substantial attention as potential players of diabetes microvascular complications, affecting the kidney, the retina, and the peripheral neurons. Compelling evidence indicates that abnormally expressed miRNAs have pivotal roles in key pathogenic processes of microvascular complications, such as fibrosis, apoptosis, inflammation, and angiogenesis. Moreover, clinical research into innovative both diagnostic and prognostic tools suggests circulating miRNAs as possible novel noninvasive markers of diabetes microvascular complications. In this review, we summarize current knowledge and understanding of the role of miRNAs in the injury to the microvascular bed in diabetes and discuss the potential of miRNAs as clinical biomarkers of diabetes microvascular complications.
Collapse
Affiliation(s)
- F. Barutta
- Laboratory of Diabetic Nephropathy, Department of Medical Sciences, University of Turin, Turin, Italy
| | - S. Bellini
- Laboratory of Diabetic Nephropathy, Department of Medical Sciences, University of Turin, Turin, Italy
| | - R. Mastrocola
- Department of Clinical and Biological Sciences, University of Turin, Turin, Italy
| | - G. Bruno
- Laboratory of Diabetic Nephropathy, Department of Medical Sciences, University of Turin, Turin, Italy
| | - G. Gruden
- Laboratory of Diabetic Nephropathy, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
43
|
Yan Y, Wang C, Lu Y, Gong H, Wu Z, Ma X, Li H, Wang B, Zhang X. Mineralocorticoid receptor antagonism protects the aorta from vascular smooth muscle cell proliferation and collagen deposition in a rat model of adrenal aldosterone-producing adenoma. J Physiol Biochem 2017; 74:17-24. [PMID: 29164516 DOI: 10.1007/s13105-017-0600-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
The number of patients with adrenal aldosterone-producing adenomas (APAs) has gradually increased. However, even after adenoma resection, some patients still suffer from high systolic blood pressure (SBP), which is possibly due to great arterial remodeling. Moreover, mineralocorticoid receptors (MRs) were found to be expressed in vascular smooth muscle cells (VSMCs). This study aims to determine whether MR antagonism protects the aorta from aldosterone-induced aortic remolding. Male rats were subcutaneously implanted with an osmotic minipumps and randomly divided into four groups: control; aldosterone (1 μg/h); aldosterone plus a specific MR antagonist, eplerenone (100 mg/kg/day); and aldosterone plus a vasodilator, hydralazine (25 mg/kg/day). After 8 weeks of infusion, aortic smooth muscle cell proliferation and collagen deposition, as well as the MDM2 and TGF-β1 expression levels in the aorta, were examined. Model rats with APAs were successfully constructed. Compared with the control rats, the model rats exhibited (1) marked SBP elevation, (2) no significant alteration in aortic morphology, (3) increased VSMC proliferation and MDM2 expression in the aorta, and (4) enhanced total collagen and collagen III depositions in the aorta, accompanied with up-regulated expression of TGF-β1. These effects were significantly inhibited by co-administration with eplerenone but not with hydralazine. These findings suggested that specific MR antagonism protects the aorta from aldosterone-induced VSMC proliferation and collagen deposition.
Collapse
Affiliation(s)
- Yongji Yan
- Department of Urology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China.,Department of Urology, East Section of Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Chao Wang
- Department of Urology, First People's Hospital of Jining City, Jining, Shandong Province, People's Republic of China
| | - Yiqin Lu
- Department of Gynecology, First People's Hospital of Yunnan Province, Kunming, Yunnan Province, People's Republic of China
| | - Huijie Gong
- Department of Urology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China.,Department of Urology, East Section of Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, People's Republic of China
| | - Zhun Wu
- Department of Urology, First Affiliated Hospital of Xiamen University, Xiamen, Fujian Province, People's Republic of China
| | - Xin Ma
- Department of Urology, Chinese People's Liberation Army General Hospital, Military Postgraduate Medical College, 28 Fu Xing Road, Hai Dian District, Beijing, 100853, People's Republic of China
| | - Hongzhao Li
- Department of Urology, Chinese People's Liberation Army General Hospital, Military Postgraduate Medical College, 28 Fu Xing Road, Hai Dian District, Beijing, 100853, People's Republic of China
| | - Baojun Wang
- Department of Urology, Chinese People's Liberation Army General Hospital, Military Postgraduate Medical College, 28 Fu Xing Road, Hai Dian District, Beijing, 100853, People's Republic of China
| | - Xu Zhang
- Department of Urology, Chinese People's Liberation Army General Hospital, Military Postgraduate Medical College, 28 Fu Xing Road, Hai Dian District, Beijing, 100853, People's Republic of China.
| |
Collapse
|
44
|
Apoptosis signal-regulating kinase 1 inhibition attenuates cardiac hypertrophy and cardiorenal fibrosis induced by uremic toxins: Implications for cardiorenal syndrome. PLoS One 2017; 12:e0187459. [PMID: 29107962 PMCID: PMC5673193 DOI: 10.1371/journal.pone.0187459] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/22/2017] [Indexed: 01/16/2023] Open
Abstract
Intracellular accumulation of protein-bound uremic toxins in the setting of cardiorenal syndrome leads to adverse effects on cardiorenal cellular functions, where cardiac hypertrophy and cardiorenal fibrosis are the hallmarks. In this study, we sought to determine if Apoptosis Signal-Regulated Kinase 1 (ASK1), an upstream regulator of cellular stress response, mediates cardiac hypertrophy and cardiorenal fibrosis induced by indoxyl sulfate (IS) and p-cresol sulfate (PCS) in vitro, and whether ASK1 inhibition is beneficial to ameliorate these cellular effects. PCS augmented cardiac myocyte hypertrophy and fibroblast collagen synthesis (as determined by 3H-leucine and 3H-proline incorporation, respectively), similar to our previous finding with IS. IS and PCS also increased collagen synthesis of proximal tubular cells and renal mesangial cells. Pro-hypertrophic (α-skeletal muscle actin and β-MHC) and pro-fibrotic genes (TGF-β1 and ctgf) were induced by both IS and PCS. Western blot analyses revealed the activation of ASK1 and downstream mitogen activated protein kinases (MAPKs) (p38MAPK and ERK1/2) as well as nuclear factor-kappa B (NF-κB) by IS and PCS. ASK1, OAT1/3, ERK1/2 and p38MAPK inhibitors suppressed all these effects. In summary, IS and PCS exhibit pro-hypertrophic and pro-fibrotic properties, at least in part, via the activation of ASK1 and its downstream pathways. ASK1 inhibitor is an effective therapeutic agent to alleviate protein-bound uremic toxin-induced cardiac hypertrophy and cardiorenal fibrosis in vitro, and may be translated further for cardiorenal syndrome therapy.
Collapse
|
45
|
Wang G, Wu L, Chen Z, Sun J. Identification of crucial miRNAs and the targets in renal cortex of hypertensive patients by expression profiles. Ren Fail 2017; 39:92-99. [PMID: 27802793 PMCID: PMC6014400 DOI: 10.1080/0886022x.2016.1244083] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 09/04/2016] [Accepted: 09/16/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUNDS Defect in kidney is one major reason of hypertension. The study aimed ao uncovering the regulatory mechanisms of miRNAs and the targets in hypertensive kidney. METHODS Gene expression profile of GSE28345 and miRNA expression profile of GSE28283 were downloaded from GEO database. After data preprocessing, differently expressed genes (DEGs) and miRNAs (DE-miRs) were identified using limma package. Then targets of miRNAs were predicted according to information in relevant databases. Function and pathway enrichment analyses were performed for DEGs using DAVID software. Furthermore, protein-protein interaction (PPI) networks were constructed for up- and down-regulated genes, respectively, using the Cytoscape. Additionally, for down-regulated DEGs, the integrated regulatory network was established combining PPI network with the miRNA-mRNA interactions. RESULTS As a result, 285 DEGs were identified, including 177 up-regulated and 108 down-regulated genes. Combined with the predicted targets of miRNAs, 22 up-regulated DE-miRs were identified. In the integrated network for down-regulated DEGs, three crucial nodes were identified as ASPN, COL12A1, and SCN2A. ASPN was predicted as target of miR-21 and miR-374b, and COL12A1 was the target of miR-30e, miR-21, and miR-195, while SCN2A was the target of miR-30e, miR-374b, and miR-195. Notably, COL12A1 and ASPN were linked with each other in the network. CONCLUSION Three crucial genes were identified in hypertensive kidney, such as COL12A1, ASPN, and SCN2A. ASPN might co-function with COL12A1, and they both might be the targets of miR-21. SCN2A might be a novel target of miR-30e and miR-374b. However, more experiments are needed to validate these results.
Collapse
Affiliation(s)
- Guohua Wang
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Lan Wu
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| | - Zhi Chen
- Department of Nephrology, The First Hospital of Jilin University, Changchun, China
| | - Jinghui Sun
- Department of Pediatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
46
|
Yang F, Li B, Dong X, Cui W, Luo P. The beneficial effects of zinc on diabetes-induced kidney damage in murine rodent model of type 1 diabetes mellitus. J Trace Elem Med Biol 2017; 42:1-10. [PMID: 28595780 DOI: 10.1016/j.jtemb.2017.03.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 03/07/2017] [Accepted: 03/08/2017] [Indexed: 02/06/2023]
Abstract
Diabetes mellitus is a chronic multi-factorial metabolic disorder resulting from impaired glucose homeostasis. Zinc is a key co-factor for the correct functioning of anti-oxidant enzymes. Zinc deficiency therefore, impairs their synthesis, leading to increased oxidative stress within cells. Zinc deficiency occurs commonly in diabetic patients. The aim of this study is to investigate the effects of varying concentrations of zinc on diabetic nephropathy (DN) and the underlying mechanisms involved. FVB male mice aged 8 weeks were injected intraperitoneally with multiple low-dose streptozotocin at a concentration of 50mg/kg body weight daily for 5 days. Diabetic and age-matched control mice were treated with special diets supplemented with zinc at varying concentrations (0.85mg/kg, 30mg/kg, 150mg/kg) for 3 months. The mice were fed with zinc diets to mimic the process of oral administration of zinc in human. Zinc deficiency to some extent aggravated the damage of diabetic kidney. Feeding with normal (30mg/kg zinc/kg diet) and especially high (150mg/kg zinc/kg diet) concentration zinc could protect the kidney against diabetes-induced damage. The beneficial effects of zinc on DN are achieved most likely due to the upregulation of Nrf2 and its downstream factors NQO1, SOD1, SOD2. Zinc upregulated the expression of Akt phosphorylation and GSK-3β phosphorylation, resulting in a reduction in Fyn nuclear translocation and export of Nrf2 to the cytosol. Thus, regular monitoring and maintaining of adequate levels of zinc are recommended in diabetic individuals in order to delay the development of DN.
Collapse
Affiliation(s)
- Fan Yang
- The Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Bing Li
- The Department of Nephropathy, Jilin Province People's Hospital, Changchun China
| | - Xiaoming Dong
- The Department of Orthopaedics, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Wenpeng Cui
- The Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China
| | - Ping Luo
- The Department of Nephropathy, The Second Hospital of Jilin University, 218 Ziqiang Street, Changchun 130041, China.
| |
Collapse
|
47
|
Campion CG, Sanchez-Ferras O, Batchu SN. Potential Role of Serum and Urinary Biomarkers in Diagnosis and Prognosis of Diabetic Nephropathy. Can J Kidney Health Dis 2017; 4:2054358117705371. [PMID: 28616250 PMCID: PMC5461910 DOI: 10.1177/2054358117705371] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 02/17/2017] [Indexed: 12/11/2022] Open
Abstract
PURPOSE OF REVIEW Diabetic nephropathy (DN) is a progressive kidney disease caused by alterations in kidney architecture and function, and constitutes one of the leading causes of end-stage renal disease (ESRD). The purpose of this review is to summarize the state of the art of the DN-biomarker field with a focus on the new strategies that enhance the sensitivity of biomarkers to predict patients who will develop DN or are at risk of progressing to ESRD. OBJECTIVE In this review, we provide a description of the pathophysiology of DN and propose a panel of novel putative biomarkers associated with DN pathophysiology that have been increasingly investigated for diagnosis, to predict disease progression or to provide efficient personal treatment. METHODS We performed a review of the literature with PubMed and Google Scholar to collect baseline data about the pathophysiology of DN and biomarkers associated. We focused our research on new and emerging biomarkers of DN. KEY FINDINGS In this review, we summarized the critical signaling pathways and biological processes involved in DN and highlighted the pathogenic mediators of this disease. We next proposed a large review of the major advances that have been made in identifying new biomarkers which are more sensitive and reliable compared with currently used biomarkers. This includes information about emergent biomarkers such as functional noncoding RNAs, microRNAs, long noncoding RNAs, exosomes, and microparticles. LIMITATIONS Despite intensive strategies and constant investigation, no current single treatment has been able to reverse or at least mitigate the progression of DN, or reduce the morbidity and mortality associated with this disease. Major difficulties probably come from the renal disease being heterogeneous among the patients. IMPLICATIONS Expanding the proteomics screening, including oxidative stress and inflammatory markers, along with metabolomics approaches may further improve the prognostic value and help in identifying the patients with diabetes who are at high risk of developing kidney diseases.
Collapse
Affiliation(s)
- Carole G. Campion
- Centre de recherche, Centre Hospitalier de l’Université de Montréal (CRCHUM), Québec, Canada
| | - Oraly Sanchez-Ferras
- Department of Biochemistry, Goodman Cancer Research Centre, McGill University, Montreal, Québec, Canada
| | - Sri N. Batchu
- St. Michael’s Hospital, University of Toronto, Ontario, Canada
| |
Collapse
|
48
|
Kota SK, Kota SB. Noncoding RNA and epigenetic gene regulation in renal diseases. Drug Discov Today 2017; 22:1112-1122. [PMID: 28487070 DOI: 10.1016/j.drudis.2017.04.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 04/18/2017] [Accepted: 04/28/2017] [Indexed: 02/07/2023]
Abstract
Kidneys have a major role in normal physiology and metabolic homeostasis. Loss or impairment of kidney function is a common occurrence in several metabolic disorders, including hypertension and diabetes. Chronic kidney disease (CKD) affect nearly 10% of the population worldwide; ranks 18th in the list of causes of death; and contributes to a significant proportion of healthcare costs. The tissue repair and regenerative potential of kidneys are limited and they decline during aging. Recent studies have demonstrated a key role for epigenetic processes and players, such as DNA methylation, histone modifications, noncoding (nc)RNA, and so on, in both kidney development and disease. In this review, we highlight these recent findings with an emphasis on aberrant epigenetic changes that accompany renal diseases, key targets, and their therapeutic value.
Collapse
Affiliation(s)
- Satya K Kota
- Harvard School of Dental Medicine, Boston, MA, USA.
| | - Savithri B Kota
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
49
|
Vitamin C in Stem Cell Biology: Impact on Extracellular Matrix Homeostasis and Epigenetics. Stem Cells Int 2017; 2017:8936156. [PMID: 28512473 PMCID: PMC5415867 DOI: 10.1155/2017/8936156] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 03/05/2017] [Indexed: 12/30/2022] Open
Abstract
Transcription factors and signaling molecules are well-known regulators of stem cell identity and behavior; however, increasing evidence indicates that environmental cues contribute to this complex network of stimuli, acting as crucial determinants of stem cell fate. l-Ascorbic acid (vitamin C (VitC)) has gained growing interest for its multiple functions and mechanisms of action, contributing to the homeostasis of normal tissues and organs as well as to tissue regeneration. Here, we review the main functions of VitC and its effects on stem cells, focusing on its activity as cofactor of Fe+2/αKG dioxygenases, which regulate the epigenetic signatures, the redox status, and the extracellular matrix (ECM) composition, depending on the enzymes' subcellular localization. Acting as cofactor of collagen prolyl hydroxylases in the endoplasmic reticulum, VitC regulates ECM/collagen homeostasis and plays a key role in the differentiation of mesenchymal stem cells towards osteoblasts, chondrocytes, and tendons. In the nucleus, VitC enhances the activity of DNA and histone demethylases, improving somatic cell reprogramming and pushing embryonic stem cell towards the naive pluripotent state. The broad spectrum of actions of VitC highlights its relevance for stem cell biology in both physiology and disease.
Collapse
|
50
|
Cyclosporine A alters expression of renal microRNAs: New insights into calcineurin inhibitor nephrotoxicity. PLoS One 2017; 12:e0175242. [PMID: 28414804 PMCID: PMC5393575 DOI: 10.1371/journal.pone.0175242] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 03/22/2017] [Indexed: 12/21/2022] Open
Abstract
Calcineurin inhibitors are powerful immunosuppressants that revolutionized organ transplantation. However, non-immune effects of the calcineurin inhibitor, such as cyclosporine A (CsA), have significantly hindered their use. Specifically, nephrotoxicity, which is associated with tubulointerstitial fibrosis, inflammation, and podocyte damage, affects up to half of all transplant patients. Calcineurin is involved in many aspects of kidney development and function; therefore, mechanisms of CsA-induced nephrotoxicity are complex and not yet fully understood. MicroRNAs are short non-coding RNAs that regulate protein-coding RNA expression through post-translational repression of target messenger RNAs. MicroRNA dysregulation is known to be involved in kidney diseases including fibrosis. In this study, we compared the renal microRNA expression profiles between mice that received CsA (20 mg/kg) or vehicle daily for six weeks. The results demonstrate that CsA induces significant changes in renal microRNA expression profile. We used combined criteria of False Discovery Rate (≤0.1), fold change (≥2) and median signal strength (≥50) and identified 76 differencially expressed microRNAs. This approach identified microRNAs previously linked to renal fibrosis that includes let-7d, miR-21, miR-29, miR-30, miR-130, miR-192, and miR-200 as well as microRNAs that have not been reported to be related to nephrotoxicity or immunosuppression. Pathway analysis of microRNA/mRNA changes highlights the Wnt, TGF-β, mTOR, and VEGF pathways. The mRNA expression profiles were compared in the same samples. The change of mRNA and microRNA profiles showed close correlations. To validate that the observed microRNA and mRNA expression level changes in mice kidney tissue were directly related to CsA treatment, the expression change induced by CsA treatment of three microRNAs (miR-21, miR-186, and miR-709) and three mRNAs (BMPR1a, SMURF1 and SMAD7) were compared in HEK293 cell line. A similar trend of expression level change was induced by CsA treatment in all selected microRNAs and mRNAs in the in vitro cell model. These data provide a roadmap for future work to study the role of the known and novel candidate microRNAs in the mechanism of nephrotoxicity and their further therapeutic potential.
Collapse
|