1
|
Mao L, Yin R, Yang L, Zhao D. Elucidating the function of clusterin in the progression of diabetic kidney disease. Front Pharmacol 2025; 16:1573654. [PMID: 40438587 PMCID: PMC12116493 DOI: 10.3389/fphar.2025.1573654] [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: 02/09/2025] [Accepted: 04/23/2025] [Indexed: 06/01/2025] Open
Abstract
Diabetic kidney disease (DKD) is a common microvascular complication and the main cause of death in diabetic patients. Metabolic disorders can accelerate the occurrence and development of DKD through a variety of ways, Recent studies have found that Clusterin (Clu) levels are associated with renal dysfunction and can be used as a biomarker of renal tubular injury, while preclinical studies reveal its renoprotective function. This article reviews the molecular mechanisms of Clu in the interaction between various cells in DKD. In addition, we discuss the latest research progress of Clu in the field of DKD. This review aims to explore Clu as a potential therapeutic target for DKD and provide some guidance for future clinical treatment.
Collapse
Affiliation(s)
| | | | - Longyan Yang
- Beijing Key Laboratory of Diabetes Prevention and Research, Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Dong Zhao
- Beijing Key Laboratory of Diabetes Prevention and Research, Center for Endocrine Metabolic and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Miyano T, Sera T, Sakamoto N. Pharmacological activation of TRPML1 enhances autophagy regulating hypertonicity and TGF-β-induced EMT in proximal tubular epithelial cells. Biochem Biophys Res Commun 2025; 750:151432. [PMID: 39893888 DOI: 10.1016/j.bbrc.2025.151432] [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: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Proximal tubular epithelial cells (PTECs) are central to maintaining kidney homeostasis. Under pathological conditions, such as ischemia or inflammation, PTECs promote profibrotic signals, including transforming growth factor (TGF)-β, and undergo epithelial-mesenchymal transition (EMT). EMT is characterized by decreased epithelial markers (e.g., E-cadherin) and increased mesenchymal markers (e.g., α-smooth muscle actin [α-SMA]), which promote myofibroblast activation and fibrosis progression. We previously demonstrated that hyperosmotic stress, characterized by elevated extracellular solute concentrations, induces EMT in PTECs. However, we observed that hyperosmotic stress simultaneously activates autophagy, a cellular process that has antagonistic effects on EMT, primarily mediated by transient receptor potential mucolipin 1 (TRPML1). However, the interplay between hyperosmotic stress-induced EMT and autophagy remains unclear. This study examined whether enhancing autophagy via TRPML1 activation could modulate EMT under hyperosmotic stress. Using the TRPML1 agonist ML-SA1, we observed a significantly increased autophagic flux, indicated by elevated LC3-II levels, without cytotoxic effects. Under hyperosmotic conditions, ML-SA1 further amplified autophagic flux in PTECs compared to hyperosmotic stress alone. Notably, this enhanced autophagy suppressed EMT by maintaining E-cadherin expression and reducing α-SMA levels. Furthermore, the ML-SA1-mediated autophagy enhancement attenuated EMT and profibrotic factor production in TGF-β-treated cells, suggesting a broader protective role beyond hyperosmotic stress. These findings reveal a novel interaction between hyperosmotic stress-induced autophagy and EMT, emphasizing TRPML1 activation's therapeutic potential to mitigate PTEC injury and fibrosis progression.
Collapse
Affiliation(s)
- Takashi Miyano
- Department of Medical and Robotic Engineering Design, Tokyo University of Science, Tokyo, Japan; Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan.
| | - Toshihiro Sera
- Department of Medical and Robotic Engineering Design, Tokyo University of Science, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Mechanical Systems Engineering, Graduate School of Systems Design, Tokyo Metropolitan University, Tokyo, Japan
| |
Collapse
|
3
|
Wu JY, Lee GL, Chueh YF, Kuo CC, Hsu YJ, Wu KK. 5-Methoxytryptophan Protects against Toll-Like Receptor 2-Mediated Renal Tissue Inflammation and Fibrosis in a Murine Unilateral Ureteral Obstruction Model. J Innate Immun 2025; 17:78-94. [PMID: 39773569 PMCID: PMC11801855 DOI: 10.1159/000543275] [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: 08/07/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION 5-Methoxytryptophan (5-MTP) is a cellular metabolite with anti-inflammatory properties. Several recent reports indicate that 5-MTP protects against post-injury tissue fibrosis. It was unclear how 5-MTP controls tissue fibrosis. We postulated that 5-MTP attenuates renal interstitial fibrosis by blocking toll-like receptor 2 (TLR2) and transforming growth factor β (TGFβ) signaling pathways. METHODS In vivo experiments were carried out in a well-established unilateral ureteral obstruction (UUO) model in wild-type (WT) and tlr2-/- mice. The effect of 5-MTP on renal fibrosis was evaluated by pretreatment of WT UUO mice with intraperitoneal administration of 5-MTP. To determine whether 5-MTP attenuates fibrosis by inhibiting TLR2 and TGFβ signaling pathways, we evaluated the effect of 5-MTP on TLR2-induced fibroblast phenotypic switch in NRK-49F fibroblasts and TLR2 and TGFβ signaling pathways in human proximal tubular epithelial cells (HPTECs) and RAW264.7 macrophages stimulated with Pam3CSK4 (Pam3) or TGFβ1. RESULTS UUO-induced renal fibrosis was abrogated in tlr2-/- mice consistent with a crucial role of TLR2 in UUO-induced renal fibrosis. UUO-induced macrophage infiltration and pro-fibrotic cytokine production in renal tissues were suppressed by tlr2 knockout. 5-MTP administration attenuated renal tissue fibrosis accompanied by reduction of macrophage infiltration and IL-6 and TGFβ levels. 5-MTP inhibits TLR2 upregulation and blocks TLR2-MyD88-TRAF6 signaling pathway in macrophages. Furthermore, 5-MTP blocked Pam3- and TGFβ1-induced phenotypic switch of NRK-49F to myofibroblasts and inhibited Pam3- and TGFβ1-induced signaling pathways in HPTECs and RAW264.7 cells. CONCLUSION 5-MTP is effective in protecting against UUO-induced renal interstitial fibrosis by blocking TLR2 and TGFβ signaling pathways. INTRODUCTION 5-Methoxytryptophan (5-MTP) is a cellular metabolite with anti-inflammatory properties. Several recent reports indicate that 5-MTP protects against post-injury tissue fibrosis. It was unclear how 5-MTP controls tissue fibrosis. We postulated that 5-MTP attenuates renal interstitial fibrosis by blocking toll-like receptor 2 (TLR2) and transforming growth factor β (TGFβ) signaling pathways. METHODS In vivo experiments were carried out in a well-established unilateral ureteral obstruction (UUO) model in wild-type (WT) and tlr2-/- mice. The effect of 5-MTP on renal fibrosis was evaluated by pretreatment of WT UUO mice with intraperitoneal administration of 5-MTP. To determine whether 5-MTP attenuates fibrosis by inhibiting TLR2 and TGFβ signaling pathways, we evaluated the effect of 5-MTP on TLR2-induced fibroblast phenotypic switch in NRK-49F fibroblasts and TLR2 and TGFβ signaling pathways in human proximal tubular epithelial cells (HPTECs) and RAW264.7 macrophages stimulated with Pam3CSK4 (Pam3) or TGFβ1. RESULTS UUO-induced renal fibrosis was abrogated in tlr2-/- mice consistent with a crucial role of TLR2 in UUO-induced renal fibrosis. UUO-induced macrophage infiltration and pro-fibrotic cytokine production in renal tissues were suppressed by tlr2 knockout. 5-MTP administration attenuated renal tissue fibrosis accompanied by reduction of macrophage infiltration and IL-6 and TGFβ levels. 5-MTP inhibits TLR2 upregulation and blocks TLR2-MyD88-TRAF6 signaling pathway in macrophages. Furthermore, 5-MTP blocked Pam3- and TGFβ1-induced phenotypic switch of NRK-49F to myofibroblasts and inhibited Pam3- and TGFβ1-induced signaling pathways in HPTECs and RAW264.7 cells. CONCLUSION 5-MTP is effective in protecting against UUO-induced renal interstitial fibrosis by blocking TLR2 and TGFβ signaling pathways.
Collapse
Affiliation(s)
- Jing-Yiing Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Guan-Lin Lee
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Fan Chueh
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Cheng-Chin Kuo
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
- Department of Bioscience Technology, Chung Yuan Christian University, Taoyuan, Taiwan
| | - Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Kenneth K. Wu
- Institute of Cellular and System Medicine, National Health Research Institutes, Zhunan, Taiwan
| |
Collapse
|
4
|
Lu C, Liu Y, Ren F, Zhang H, Hou Y, Zhang H, Chen Z, Du X. HO-1: An emerging target in fibrosis. J Cell Physiol 2025; 240:e31465. [PMID: 39420552 DOI: 10.1002/jcp.31465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/08/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024]
Abstract
Fibrosis, an aberrant reparative response to tissue injury, involves a disruption in the equilibrium between the synthesis and degradation of the extracellular matrix, leading to its excessive accumulation within normal tissues, and culminating in organ dysfunction. Manifesting in the terminal stages of nearly all chronic ailments, fibrosis carries a high mortality rate and poses a significant threat to human health. Heme oxygenase-1 (HO-1) emerges as an endogenous protective agent, mitigating tissue damage through its antioxidant, anti-inflammatory, and antiapoptotic properties. Numerous studies have corroborated HO-1's potential as a therapeutic target in anti-fibrosis treatment. This review delves into the structural and functional attributes, and the upstream and downstream pathways of HO-1. Additionally, the regulatory networks and mechanisms of HO-1 in cells associated with fibrosis are elucidated. The role of HO-1 in various fibrosis-related diseases is also explored. Collectively, this comprehensive information serves as a foundation for future research and augments the viability of HO-1 as a therapeutic target for fibrosis.
Collapse
Affiliation(s)
- Chenxi Lu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yuan Liu
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Feifei Ren
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Haoran Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Yafang Hou
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Hong Zhang
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Zhiyong Chen
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| | - Xia Du
- Institute of Traditional Chinese Medicine, Shaanxi Academy of Traditional Chinese Medicine, Xi'an, China
| |
Collapse
|
5
|
Zhang Y, Arzaghi H, Ma Z, Roye Y, Musah S. Epigenetics of Hypertensive Nephropathy. Biomedicines 2024; 12:2622. [PMID: 39595187 PMCID: PMC11591919 DOI: 10.3390/biomedicines12112622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/05/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Hypertensive nephropathy (HN) is a leading cause of chronic kidney disease (CKD) and end-stage renal disease (ESRD), contributing to significant morbidity, mortality, and rising healthcare costs. In this review article, we explore the role of epigenetic mechanisms in HN progression and their potential therapeutic implications. We begin by examining key epigenetic modifications-DNA methylation, histone modifications, and non-coding RNAs-observed in kidney disease. Next, we discuss the underlying pathophysiology of HN and highlight current in vitro and in vivo models used to study the condition. Finally, we compare various types of HN-induced renal injury and their associated epigenetic mechanisms with those observed in other kidney injury models, drawing inferences on potential epigenetic therapies for HN. The information gathered in this work indicate that epigenetic mechanisms can drive the progression of HN by regulating key molecular signaling pathways involved in renal damage and fibrosis. The limitations of Renin-Angiotensin-Aldosterone System (RAAS) inhibitors underscore the need for alternative treatments targeting epigenetic pathways. This review emphasizes the importance of further research into the epigenetic regulation of HN to develop more effective therapies and preventive strategies. Identifying novel epigenetic markers could provide new therapeutic opportunities for managing CKD and reducing the burden of ESRD.
Collapse
Affiliation(s)
- Yize Zhang
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Hamidreza Arzaghi
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Zhehan Ma
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Yasmin Roye
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
| | - Samira Musah
- Department of Biomedical Engineering, Pratt School of Engineering, Duke University, Durham, NC 27708, USA
- Center for Biomolecular and Tissue Engineering, Duke University, Durham, NC 27708, USA
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC 27710, USA
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
- Affiliate Faculty of the Developmental and Stem Cell Biology Program, Duke Regeneration Center, and Duke MEDx Initiative, Duke University, Durham, NC 27710, USA
| |
Collapse
|
6
|
Wei S, Huang X, Zhu Q, Chen T, Zhang Y, Tian J, Pan T, Zhang L, Xie T, Zhang Q, Kuang X, Lei E, Li Y. TRIM65 deficiency alleviates renal fibrosis through NUDT21-mediated alternative polyadenylation. Cell Death Differ 2024; 31:1422-1438. [PMID: 38951701 PMCID: PMC11519343 DOI: 10.1038/s41418-024-01336-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/19/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Chronic kidney disease (CKD) is a major global health concern and the third leading cause of premature death. Renal fibrosis is the primary process driving the progression of CKD, but the mechanisms behind it are not fully understood, making treatment options limited. Here, we find that the E3 ligase TRIM65 is a positive regulator of renal fibrosis. Deletion of TRIM65 results in a reduction of pathological lesions and renal fibrosis in mouse models of kidney fibrosis induced by unilateral ureteral obstruction (UUO)- and folic acid. Through screening with a yeast-hybrid system, we identify a new interactor of TRIM65, the mammalian cleavage factor I subunit CFIm25 (NUDT21), which plays a crucial role in fibrosis through alternative polyadenylation (APA). TRIM65 interacts with NUDT21 to induce K48-linked polyubiquitination of lysine 56 and proteasomal degradation, leading to the inhibition of TGF-β1-mediated SMAD and ERK1/2 signaling pathways. The degradation of NUDT21 subsequently altered the length and sequence content of the 3'UTR (3'UTR-APA) of several pro-fibrotic genes including Col1a1, Fn-1, Tgfbr1, Wnt5a, and Fzd2. Furthermore, reducing NUDT21 expression via hydrodynamic renal pelvis injection of adeno-associated virus 9 (AAV9) exacerbated UUO-induced renal fibrosis in the normal mouse kidneys and blocked the protective effect of TRIM65 deletion. These findings suggest that TRIM65 promotes renal fibrosis by regulating NUDT21-mediated APA and highlight TRIM65 as a potential target for reducing renal fibrosis in CKD patients.
Collapse
Affiliation(s)
- Sisi Wei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Xuan Huang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Qing Zhu
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Tao Chen
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yan Zhang
- Department of Biological Sciences, College of Sciences and Arts, Michigan Technological University, Houghton, MI, 49931-1295, USA
| | - Juan Tian
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tingyu Pan
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Lv Zhang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Tao Xie
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Qi Zhang
- The National Engineering Research Center for Bioengineering Drugs and the Technologies; Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Xian Kuang
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Enjun Lei
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Yong Li
- Department of Anesthesiology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
7
|
Zhang C, Wang K, Chen X, Li Y. Mechanistic study on lncRNA XIST/miR-124-3p/ITGB1 axis in renal fibrosis in obstructive nephropathy. Exp Cell Res 2024; 442:114194. [PMID: 39127440 DOI: 10.1016/j.yexcr.2024.114194] [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: 03/14/2024] [Revised: 07/04/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
OBJECTIVE The purpose of this study was to investigate the role and possible mechanism of lncRNA XIST in renal fibrosis and to provide potential endogenous targets for renal fibrosis in obstructive nephropathy (ON). METHODS The study included 50 cases of ON with renal fibrosis (samples taken from patients undergoing nephrectomy due to ON) and 50 cases of normal renal tissue (samples taken from patients undergoing total or partial nephrectomy due to accidental injury, congenital malformations, and benign tumors). Treatment of human proximal renal tubular epithelium (HK-2) cells with TGF-β1 simulated renal fibrosis in vitro. Cell viability and proliferation were measured by CCK-8 and EdU, and cell migration was measured by transwell. XIST, miR-124-3p, ITGB1, and epithelial-mesenchymal transition (EMT)-related proteins (E-cadherin, α-SMA, and fibronectin) were detected by PCR and immunoblot. The targeting relationship between miR-124-3p and XIST or ITGB1 was verified by starBase and dual luciferase reporter gene experiments. In addition, The left ureter was ligated in mice as a model of unilateral ureteral obstruction (UUO), and the renal histopathology was observed by HE staining and Masson staining. RESULTS ON patients with renal fibrosis had elevated XIST and ITGB1 levels and reduced miR-124-3p levels. The administration of TGF-β1 exhibited a dose-dependent promotion of HK-2 cell viability, proliferation, migration, and EMT. Conversely, depleting XIST or enhancing miR-124-3p hindered HK-2 cell viability, proliferation, migration, and EMT in TGF-β1-damaged HK-2 cells HK-2 cells. XIST functioned as a miR-124-3p sponge. Additionally, miR-124-3p negatively regulated ITGB1 expression. Elevating ITGB1 weakened the impact of XIST depletion on TGF-β1-damaged HK-2 cells. Down-regulating XIST improved renal fibrosis in UUO mice. CONCLUSION XIST promotes renal fibrosis in ON by elevating miR-124-3p and reducing ITGB1 expressions.
Collapse
Affiliation(s)
- ChiTeng Zhang
- Department of Urology Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, 421001, China
| | - KangNing Wang
- Department of Urology Surgery, Xiangya Hospital Central South University, Changsha City, Hunan Province, 410000, China
| | - Xiang Chen
- Department of Urology Surgery, Xiangya Hospital Central South University, Changsha City, Hunan Province, 410000, China
| | - Yong Li
- Department of Urology Surgery, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang City, Hunan Province, 421001, China.
| |
Collapse
|
8
|
Ma D, Zhang J, Du L, Shi J, Liu Z, Qin J, Chen X, Guo M. Colquhounia root tablet improves diabetic kidney disease by regulating epithelial-mesenchymal transition via the PTEN/PI3K/AKT pathway. Front Pharmacol 2024; 15:1418588. [PMID: 39130629 PMCID: PMC11310013 DOI: 10.3389/fphar.2024.1418588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024] Open
Abstract
Background Diabetic kidney disease (DKD) is a severe microvascular complication of diabetes mellitus that can lead to end-stage renal disease. Colquhounia root tablet (CRT) has shown therapeutic potential in treating DKD, but its efficacy and underlying mechanisms remain to be elucidated. Methods A randomized controlled clinical trial was conducted on 61 DKD patients. The treatment group received CRT in addition to standard therapy, while the control group received standard therapy alone. Treatment efficacy and adverse events were evaluated after 3 months. Additionally, in vitro experiments using human renal tubular epithelial cells (HK-2) were performed to investigate the effect of CRT on high glucose (HG)-induced epithelial-mesenchymal transition (EMT) and the involvement of the PTEN/PI3K/AKT signaling pathway. Results CRT treatment significantly improved proteinuria and increased the effective treatment rate in DKD patients compared to the control group, with no significant difference in adverse events. Moreover, CRT reversed HG-induced EMT in HK-2 cells, as evidenced by the downregulation of α-SMA and upregulation of E-cadherin at both mRNA and protein levels. Mechanistically, CRT increased PTEN expression and inhibited the PI3K/AKT pathway, similar to the effects of the PI3K inhibitor LY29400. The combination of CRT and LY29400 further enhanced PTEN mRNA expression under HG conditions. Conclusion CRT effectively improves proteinuria in DKD patients and ameliorates HG-induced EMT in HK-2 cells. The underlying mechanism may involve the upregulation of PTEN and subsequent inhibition of the PI3K/AKT signaling pathway. These findings provide new insights into the therapeutic potential of CRT for DKD treatment.
Collapse
Affiliation(s)
- Donghong Ma
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
- Xinxiang Key Laboratory of Precise Therapy for Diabetic Kidney Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Jiao Zhang
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Lu Du
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Jingjing Shi
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Zhaoyan Liu
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
- Xinxiang Key Laboratory of Precise Therapy for Diabetic Kidney Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Jilin Qin
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
- Xinxiang Key Laboratory of Precise Therapy for Diabetic Kidney Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Xiaoxiao Chen
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
- Xinxiang Key Laboratory of Precise Therapy for Diabetic Kidney Disease, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| | - Minghao Guo
- Department of Nephrology, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan Province, China
| |
Collapse
|
9
|
Deng Z, Fan T, Xiao C, Tian H, Zheng Y, Li C, He J. TGF-β signaling in health, disease, and therapeutics. Signal Transduct Target Ther 2024; 9:61. [PMID: 38514615 PMCID: PMC10958066 DOI: 10.1038/s41392-024-01764-w] [Citation(s) in RCA: 217] [Impact Index Per Article: 217.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 08/31/2023] [Accepted: 01/31/2024] [Indexed: 03/23/2024] Open
Abstract
Transforming growth factor (TGF)-β is a multifunctional cytokine expressed by almost every tissue and cell type. The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue homeostasis, and immune homeostasis in health. The dysfunction of TGF-β can play key roles in many diseases, and numerous targeted therapies have been developed to rectify its pathogenic activity. In the past decades, a large number of studies on TGF-β signaling have been carried out, covering a broad spectrum of topics in health, disease, and therapeutics. Thus, a comprehensive overview of TGF-β signaling is required for a general picture of the studies in this field. In this review, we retrace the research history of TGF-β and introduce the molecular mechanisms regarding its biosynthesis, activation, and signal transduction. We also provide deep insights into the functions of TGF-β signaling in physiological conditions as well as in pathological processes. TGF-β-targeting therapies which have brought fresh hope to the treatment of relevant diseases are highlighted. Through the summary of previous knowledge and recent updates, this review aims to provide a systematic understanding of TGF-β signaling and to attract more attention and interest to this research area.
Collapse
Affiliation(s)
- Ziqin Deng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - He Tian
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yujia Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
10
|
Markasz L, Mobini-Far H, Sindelar R. Collagen type IV alpha 1 chain (COL4A1) expression in the developing human lung. BMC Pulm Med 2024; 24:75. [PMID: 38331745 PMCID: PMC10851591 DOI: 10.1186/s12890-024-02875-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
BACKGROUND Collagen type IV alpha 1 chain (COL4A1) in the basement membrane is an important component during lung development, as suggested from animal models where COL4A1 has been shown to regulate alveolarization and angiogenesis. Less is known about its role in human lung development. Our aim was to study COL4A1 expression in preterm infants with different lung maturational and clinical features. METHODS COL4A1 expression in 115 lung samples from newborn infants (21-41 weeks' gestational age; 0-228 days' postnatal age [PNA]) was studied by immunohistochemistry combined with digital image analysis. Cluster analysis was performed to find subgroups according to immunohistologic and clinical data. RESULTS Patients were automatically categorized into 4 Groups depending on their COL4A1 expression. Expression of COL4A1 was mainly extracellular in Group 1, low in Group 2, intracellular in Group 3, and both extra- and intracellular in Group 4. Intracellular/extracellular ratio of COL4A1 expression related to PNA showed a distinctive postnatal maturational pattern on days 1-7, where intracellular expression of COL4A1 was overrepresented in extremely preterm infants. CONCLUSIONS COL4A1 expression seems to be highly dynamic during the postnatal life due to a possible rapid remodeling of the basement membrane. Intracellular accumulation of COL4A1 in the lungs of extremely premature infants occurs more frequently between 1 and 7 postnatal days than during the first 24 hours. In view of the lung arrest described in extremely preterm infants, the pathological and/or developmental role of postnatally increased intracellular COL4A1 as marker for basement membrane turnover, needs to be further investigated.
Collapse
Affiliation(s)
- Laszlo Markasz
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden.
| | - Hamid Mobini-Far
- Department of Pathology, Uppsala University Hospital, Uppsala, Sweden
| | - Richard Sindelar
- Department of Women's and Children's Health, Uppsala University, Uppsala, SE-751 85, Sweden
| |
Collapse
|
11
|
Yeh TH, Tu KC, Wang HY, Chen JY. From Acute to Chronic: Unraveling the Pathophysiological Mechanisms of the Progression from Acute Kidney Injury to Acute Kidney Disease to Chronic Kidney Disease. Int J Mol Sci 2024; 25:1755. [PMID: 38339031 PMCID: PMC10855633 DOI: 10.3390/ijms25031755] [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: 01/05/2024] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
This article provides a thorough overview of the biomarkers, pathophysiology, and molecular pathways involved in the transition from acute kidney injury (AKI) and acute kidney disease (AKD) to chronic kidney disease (CKD). It categorizes the biomarkers of AKI into stress, damage, and functional markers, highlighting their importance in early detection, prognosis, and clinical applications. This review also highlights the links between renal injury and the pathophysiological mechanisms underlying AKI and AKD, including renal hypoperfusion, sepsis, nephrotoxicity, and immune responses. In addition, various molecules play pivotal roles in inflammation and hypoxia, triggering maladaptive repair, mitochondrial dysfunction, immune system reactions, and the cellular senescence of renal cells. Key signaling pathways, such as Wnt/β-catenin, TGF-β/SMAD, and Hippo/YAP/TAZ, promote fibrosis and impact renal function. The renin-angiotensin-aldosterone system (RAAS) triggers a cascade leading to renal fibrosis, with aldosterone exacerbating the oxidative stress and cellular changes that promote fibrosis. The clinical evidence suggests that RAS inhibitors may protect against CKD progression, especially post-AKI, though more extensive trials are needed to confirm their full impact.
Collapse
Affiliation(s)
- Tzu-Hsuan Yeh
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan 71004, Taiwan; (T.-H.Y.); (H.-Y.W.)
| | - Kuan-Chieh Tu
- Division of Cardiology, Department of Internal Medicine, Chi Mei Medical Center, Tainan 71004, Taiwan;
| | - Hsien-Yi Wang
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan 71004, Taiwan; (T.-H.Y.); (H.-Y.W.)
- Department of Sport Management, College of Leisure and Recreation Management, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| | - Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan 71004, Taiwan; (T.-H.Y.); (H.-Y.W.)
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan 71710, Taiwan
| |
Collapse
|
12
|
Li L, Ye H, Chen Q, Wei L. COL28 promotes proliferation, migration, and EMT of renal tubular epithelial cells. Ren Fail 2023; 45:2187236. [PMID: 36883360 PMCID: PMC10013395 DOI: 10.1080/0886022x.2023.2187236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Abstract
Type XXVIII collagen (COL28) is involved in cancer and lung fibrosis. COL28 polymorphisms and mutations might be involved in kidney fibrosis, but the exact role of COL28 in renal fibrosis is unknown. This study explored the function of COL28 in renal tubular cells by examining the expression of COL28 mRNA and the effects of COL28 overexpression in human tubular cells. COL28 mRNA expression and localization were observed in normal and fibrotic kidney tissues from humans and mice using real-time PCR, western blot, immunofluorescence, and immunohistochemistry. The consequences of COL28 overexpression on cell proliferation, migration, cell polarity, and epithelial-to-mesenchymal transition (EMT) induced by TGF-β1 were examined in human tubular HK-2 cells. COL28 expression was low in human normal renal tissues, mainly observed in the renal tubular epithelial cells and especially in proximal renal tubules. COL28 protein expression in human and mouse obstructive kidney disease was higher than in normal tissues (p < 0.05) and more significant in the UUO2-Week than the UUO1-Week group. The overexpression of COL28 promoted HK-2 cell proliferation and enhanced their migration ability (all p < 0.05). TGF-β1 (10 ng/ml) induced COL28 mRNA expression in HK-2 cells, decreased E-cadherin and increased α-SMA in the COL28-overexpression group compared with controls (p < 0.05). ZO-1 expression decreased while COL6 increased in the COL28-overexpression group compared with controls (p < 0.05). In conclusion, COL28 overexpression promotes the migration and proliferation of renal tubular epithelial cells. The EMT could also be involved. COL28 could be a therapeutic target against renal- fibrotic diseases.
Collapse
Affiliation(s)
- Linlin Li
- Department of nephrology, Fujian Medical University, Union Hospital, Fuzhou, Fujian, China
| | - Hong Ye
- Department of nephrology, Fujian Medical University, Union Hospital, Fuzhou, Fujian, China
| | - Qiaoling Chen
- Department of nephrology, Fujian Medical University, Union Hospital, Fuzhou, Fujian, China
| | - Lixin Wei
- Department of nephrology, Fujian Medical University, Union Hospital, Fuzhou, Fujian, China
| |
Collapse
|
13
|
Akhurst RJ. From shape-shifting embryonic cells to oncology: The fascinating history of epithelial mesenchymal transition. Semin Cancer Biol 2023; 96:100-114. [PMID: 37852342 PMCID: PMC10883734 DOI: 10.1016/j.semcancer.2023.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 08/29/2023] [Accepted: 10/09/2023] [Indexed: 10/20/2023]
Abstract
Epithelial-to-mesenchymal transition or transformation (EMT) is a cell shape-changing process that is utilized repeatedly throughout embryogenesis and is critical to the attainment of a precise body plan. In the adult, EMT is observed under both normal and pathological conditions, such as during normal wounding healing, during development of certain fibrotic states and vascular anomalies, as well as in some cancers when malignant cells progress to become more aggressive, invasive, and metastatic. Epithelia derived from any of the three embryonic germ layers can undergo EMT, including those derived from mesoderm, such as endothelial cells (sometimes termed Endo-MT) and those derived from endoderm such as fetal liver stroma. At the cellular level, EMT is defined as the transformation of epithelial cells towards a mesenchymal phenotype and is marked by attenuation of expression of epithelial markers and de novo expression of mesenchymal markers. This process is induced by extracellular factors and can be reversible, resulting in mesenchymal-to-epithelial transformation (MET). It is now clear that a cell can simultaneously express properties of both epithelia and mesenchyme, and that such transitional cell-types drive tumor cell heterogeneity, an important aspect of cancer progression, development of a stem-like cell state, and drug resistance. Here we review some of the earliest studies demonstrating the existence of EMT during embryogenesis and discuss the discovery of the extracellular factors and intracellular signaling pathways that contribute to this process, with components of the TGFβ signaling superfamily playing a prominent role. We mention early controversies surrounding in vivo EMT during embryonic development and in adult diseased states, and the maturation of the field to a stage wherein targeting EMT to control disease states is an aspirational goal.
Collapse
Affiliation(s)
- Rosemary J Akhurst
- Department of Anatomy and UCSF Helen Diller Family Comprehensive Cancer Center, USA
| |
Collapse
|
14
|
Naas S, Schiffer M, Schödel J. Hypoxia and renal fibrosis. Am J Physiol Cell Physiol 2023; 325:C999-C1016. [PMID: 37661918 DOI: 10.1152/ajpcell.00201.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 09/05/2023]
Abstract
Renal fibrosis is the final stage of most progressive kidney diseases. Chronic kidney disease (CKD) is associated with high comorbidity and mortality. Thus, preventing fibrosis and thereby preserving kidney function increases the quality of life and prolongs the survival of patients with CKD. Many processes such as inflammation or metabolic stress modulate the progression of kidney fibrosis. Hypoxia has also been implicated in the pathogenesis of renal fibrosis, and oxygen sensing in the kidney is of outstanding importance for the body. The dysregulation of oxygen sensing in the diseased kidney is best exemplified by the loss of stimulation of erythropoietin production from interstitial cells in the fibrotic kidney despite anemia. Furthermore, hypoxia is present in acute or chronic kidney diseases and may affect all cell types present in the kidney including tubular and glomerular cells as well as resident immune cells. Pro- and antifibrotic effects of the transcription factors hypoxia-inducible factors 1 and 2 have been described in a plethora of animal models of acute and chronic kidney diseases, but recent advances in sequencing technologies now allow for novel and deeper insights into the role of hypoxia and its cell type-specific effects on the progression of renal fibrosis, especially in humans. Here, we review existing literature on how hypoxia impacts the development and progression of renal fibrosis.
Collapse
Affiliation(s)
- Stephanie Naas
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mario Schiffer
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Johannes Schödel
- Department of Nephrology and Hypertension, Uniklinikum Erlangen und Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
15
|
Guo W, Li H, Li Y, Kong W. Renal intrinsic cells remodeling in diabetic kidney disease and the regulatory effects of SGLT2 Inhibitors. Biomed Pharmacother 2023; 165:115025. [PMID: 37385209 DOI: 10.1016/j.biopha.2023.115025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/11/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023] Open
Abstract
Diabetic kidney disease (DKD) is a prevalent complication of diabetes and a major secondary factor leading to end-stage renal disease. The kidney, a vital organ, is composed of a heterogeneous group of intrinsic cells, including glomerular endothelial cells, podocytes, mesangial cells, tubular epithelial cells, and interstitial fibroblasts. In the context of DKD, hyperglycemia elicits direct or indirect injury to these intrinsic cells, leading to their structural and functional changes, such as cell proliferation, apoptosis, and transdifferentiation. The dynamic remodeling of intrinsic cells represents an adaptive response to stimulus during the pathogenesis of diabetic kidney disease. However, the persistent stimulus may trigger an irreversible remodeling, leading to fibrosis and functional deterioration of the kidney. Sodium-glucose cotransporter 2 (SGLT2) inhibitors, a new class of hypoglycemic drugs, exhibit efficacy in reducing blood glucose levels by curtailing renal tubular glucose reabsorption. Furthermore, SGLT2 inhibitors have been shown to modulate intrinsic cell remodeling in the kidney, ameliorate kidney structure and function, and decelerate DKD progression. This review will elaborate on the intrinsic cell remodeling in DKD and the underlying mechanism of SGLT2 inhibitors in modulating it from the perspective of the renal intrinsic cell, providing insights into the pathogenesis of DKD and the renal protective action of SGLT2 inhibitors.
Collapse
Affiliation(s)
- Wenwen Guo
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Han Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Yixuan Li
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Diabetes and Metabolic Disease Clinical Research Center of Hubei Province, Wuhan, Hubei 430022, China; Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Wuhan, Hubei 430022, China; Hubei Branch of National Center for Clinical Medical Research of Metabolic Diseases, Wuhan, Hubei 430022, China.
| |
Collapse
|
16
|
Yu H, Zaveri S, Sattar Z, Schaible M, Perez Gandara B, Uddin A, McGarvey LR, Ohlmeyer M, Geraghty P. Protein Phosphatase 2A as a Therapeutic Target in Pulmonary Diseases. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1552. [PMID: 37763671 PMCID: PMC10535831 DOI: 10.3390/medicina59091552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 08/24/2023] [Indexed: 09/29/2023]
Abstract
New disease targets and medicinal chemistry approaches are urgently needed to develop novel therapeutic strategies for treating pulmonary diseases. Emerging evidence suggests that reduced activity of protein phosphatase 2A (PP2A), a complex heterotrimeric enzyme that regulates dephosphorylation of serine and threonine residues from many proteins, is observed in multiple pulmonary diseases, including lung cancer, smoke-induced chronic obstructive pulmonary disease, alpha-1 antitrypsin deficiency, asthma, and idiopathic pulmonary fibrosis. Loss of PP2A responses is linked to many mechanisms associated with disease progressions, such as senescence, proliferation, inflammation, corticosteroid resistance, enhanced protease responses, and mRNA stability. Therefore, chemical restoration of PP2A may represent a novel treatment for these diseases. This review outlines the potential impact of reduced PP2A activity in pulmonary diseases, endogenous and exogenous inhibitors of PP2A, details the possible PP2A-dependent mechanisms observed in these conditions, and outlines potential therapeutic strategies for treatment. Substantial medicinal chemistry efforts are underway to develop therapeutics targeting PP2A activity. The development of specific activators of PP2A that selectively target PP2A holoenzymes could improve our understanding of the function of PP2A in pulmonary diseases. This may lead to the development of therapeutics for restoring normal PP2A responses within the lung.
Collapse
Affiliation(s)
- Howard Yu
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Sahil Zaveri
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Zeeshan Sattar
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Michael Schaible
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Brais Perez Gandara
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Anwar Uddin
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | - Lucas R. McGarvey
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| | | | - Patrick Geraghty
- Department of Medicine, State University of New York Downstate Health Sciences University, 450 Clarkson Avenue, Brooklyn, NY 11203, USA; (H.Y.); (S.Z.); (Z.S.); (M.S.); (B.P.G.); (A.U.); (L.R.M.)
| |
Collapse
|
17
|
Gluba-Sagr A, Franczyk B, Rysz-Górzyńska M, Ławiński J, Rysz J. The Role of miRNA in Renal Fibrosis Leading to Chronic Kidney Disease. Biomedicines 2023; 11:2358. [PMID: 37760798 PMCID: PMC10525803 DOI: 10.3390/biomedicines11092358] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/29/2023] Open
Abstract
Chronic kidney disease (CKD) is an important health concern that is expected to be the fifth most widespread cause of death worldwide by 2040. The presence of chronic inflammation, oxidative stress, ischemia, etc., stimulates the development and progression of CKD. Tubulointerstitial fibrosis is a common pathomechanism of renal dysfunction, irrespective of the primary origin of renal injury. With time, fibrosis leads to end-stage renal disease (ESRD). Many studies have demonstrated that microRNAs (miRNAs, miRs) are involved in the onset and development of fibrosis and CKD. miRNAs are vital regulators of some pathophysiological processes; therefore, their utility as therapeutic agents in various diseases has been suggested. Several miRNAs were demonstrated to participate in the development and progression of kidney disease. Since renal fibrosis is an important problem in chronic kidney disease, many scientists have focused on the determination of miRNAs associated with kidney fibrosis. In this review, we present the role of several miRNAs in renal fibrosis and the potential pathways involved. However, as well as those mentioned above, other miRs have also been suggested to play a role in this process in CKD. The reports concerning the impact of some miRNAs on fibrosis are conflicting, probably because the expression and regulation of miRNAs occur in a tissue- and even cell-dependent manner. Moreover, different assessment modes and populations have been used. There is a need for large studies and clinical trials to confirm the role of miRs in a clinical setting. miRNAs have great potential; thus, their analysis may improve diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Anna Gluba-Sagr
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| | - Magdalena Rysz-Górzyńska
- Department of Ophthalmology and Visual Rehabilitation, Medical University of Lodz, 90-549 Lodz, Poland
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, College of Medical Sciences, University of Rzeszow, 35-055 Rzeszow, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 90-549 Lodz, Poland
| |
Collapse
|
18
|
Huang CW, Lee SY, Du CX, Ku HC. Soluble dipeptidyl peptidase-4 induces epithelial-mesenchymal transition through tumor growth factor-β receptor. Pharmacol Rep 2023:10.1007/s43440-023-00496-y. [PMID: 37233949 DOI: 10.1007/s43440-023-00496-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Kidney fibrosis is the final manifestation of chronic kidney disease, a condition mainly caused by diabetic nephropathy. Persistent tissue damage leads to chronic inflammation and excessive deposition of extracellular matrix (ECM) proteins. Epithelial-mesenchymal transition (EMT) is involved in a variety of tissue fibrosis and is a process during which epithelial cells transform into mesenchymal-like cells and lose their epithelial functionality and characteristics Dipeptidyl peptidase-4 (DPP4) is widely expressed in tissues, especially those of the kidney and small intestine. DPP4 exists in two forms: a plasma membrane-bound and a soluble form. Serum-soluble DPP4 (sDPP4) levels are altered in many pathophysiological conditions. Elevated circulating sDPP4 is correlated with metabolic syndrome. Because the role of sDPP4 in EMT remains unclear, we examined the effect of sDPP4 on renal epithelial cells. METHODS The influences of sDPP4 on renal epithelial cells were demonstrated by measuring the expression of EMT markers and ECM proteins. RESULTS sDPP4 upregulated the EMT markers ACTA2 and COL1A1 and increased total collagen content. sDPP4 activated SMAD signaling in renal epithelial cells. Using genetic and pharmacological methods to target TGFBR, we observed that sDPP4 activated SMAD signaling through TGFBR in epithelial cells, whereas genetic ablation and treatment with TGFBR antagonist prevented SMAD signaling and EMT. Linagliptin, a clinically available DPP4 inhibitor, abrogated sDPP4-induced EMT. CONCLUSIONS This study indicated that sDPP4/TGFBR/SMAD axis leads to EMT in renal epithelial cells. Elevated circulating sDPP4 levels may contribute to mediators that induce renal fibrosis.
Collapse
Affiliation(s)
- Cheng-Wei Huang
- Department of Life Science, Fu Jen Catholic University, No.510, Zhongzheng Road, Xinzhuang District, New Taipei City, 242, Taiwan
| | - Shih-Yi Lee
- Division of Pulmonary and Critical Care Medicine, MacKay Memorial Hospital, Taipei, Taiwan
- MacKay Junior College of Medicine, Nursing and Management, Taipei, Taiwan
| | - Chen-Xuan Du
- Department of Life Science, Fu Jen Catholic University, No.510, Zhongzheng Road, Xinzhuang District, New Taipei City, 242, Taiwan
| | - Hui-Chun Ku
- Department of Life Science, Fu Jen Catholic University, No.510, Zhongzheng Road, Xinzhuang District, New Taipei City, 242, Taiwan.
| |
Collapse
|
19
|
Gao Y, Su X, Xue T, Zhang N. The beneficial effects of astragaloside IV on ameliorating diabetic kidney disease. Biomed Pharmacother 2023; 163:114598. [PMID: 37150034 DOI: 10.1016/j.biopha.2023.114598] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 02/22/2023] [Accepted: 03/23/2023] [Indexed: 05/09/2023] Open
Abstract
Diabetic kidney disease (DKD) has become the major cause of chronic kidney disease or end-stage renal disease. There is still a need for innovative treatment strategies for preventing, arresting, treating, and reversing DKD, and a plethora of scientific evidence has revealed that Chinese herbal monomers can attenuate DKD in multiple ways. Astragaloside IV (AS-IV) is one of the active ingredients of Astragalus membranaceus and was selected as a chemical marker in the Chinese Pharmacopeia for quality control purposes. An increasing amount of studies indicate that AS-IV is a promising novel drug for the treatment of DKD. AS-IV has been shown to improve DKD by combating oxidative stress, attenuating endoplasmic reticulum stress, regulating calcium homeostasis, alleviating inflammation, improving vascular function, improving epithelial to mesenchymal transition and so on. This review briefly summarizes the pathogenesis of DKD, systematically reviews the mechanisms by which AS-IV improves DKD, and aims to facilitate related pharmacological research and development to promote the utilization of Chinese herbal monomers in DKD.
Collapse
Affiliation(s)
- Yiwei Gao
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Xin Su
- Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Taiqi Xue
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China
| | - Ning Zhang
- Department of Nephrology and Endocrinology, Wangjing Hospital of China Academy of Chinese Medical Sciences, Beijing 100102, China.
| |
Collapse
|
20
|
O’Driscoll E, Hughes E, Irnaten M, Kuehn M, Wallace D, O’Brien C. Role of Epithelial-to-Mesenchymal Transition of Retinal Pigment Epithelial Cells in Glaucoma Cupping. J Clin Med 2023; 12:2737. [PMID: 37048820 PMCID: PMC10095336 DOI: 10.3390/jcm12072737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/29/2023] [Accepted: 04/01/2023] [Indexed: 04/08/2023] Open
Abstract
Optic nerve head (ONH) cupping is a clinical feature of glaucoma associated with extracellular matrix (ECM) remodelling and lamina cribrosa (LC) fibrosis. Peripapillary atrophy (PPA) occurs commonly in glaucoma, and is characterised by the loss of retinal pigment epithelium (RPE) adjacent to the ONH. Under pro-fibrotic conditions, epithelial cells throughout the body can differentiate into fibroblast-like cells through epithelial-to-mesenchymal transition (EMT) and contribute to ECM fibrosis. This is investigated here in the context of glaucoma and PPA. Human-donor ONH sections were assessed for the presence of the RPE cell-specific marker RPE65 using immunofluorescence. We examined the EMT response of ARPE-19 cells to the following glaucoma-related stimuli: cyclic mechanical stretch, mechanical stiffness, transforming growth factor beta (TGFβ), and tumour necrosis factor alpha (TNFα). The gene expression was measured using the PCR of the epithelial tight junction marker zona occludens 1 (ZO-1) and the mesenchymal markers alpha smooth muscle actin (αSMA) and vimentin. A scratch assay was used to assess the ARPE-19 migration. Significant RPE-65 staining was demonstrated in the glaucomatous ONH. The cyclic stretching and substrate stiffness of the ARPE-19 cells caused a significant decrease in ZO-1 (p = 0.04), and an increase in αSMA (p = 0.04). The scratch assays demonstrated increased migration of ARPE19 in the presence of TNFα (p = 0.02). Furthermore, ARPE-19 cells undergo an EMT-like transition (gain of αSMA, loss of ZO-1 and increased migration) in response to glaucomatous stimuli. This suggests that during PPA, RPE cells have the potential to migrate into the ONH and differentiate into fibroblast-like cells, contributing to glaucomatous ONH cupping.
Collapse
Affiliation(s)
- Eabha O’Driscoll
- Department of Ophthalmology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland (M.I.)
| | - Emily Hughes
- Department of Ophthalmology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland (M.I.)
| | - Mustapha Irnaten
- Department of Ophthalmology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland (M.I.)
| | - Markus Kuehn
- Department of Ophthalmology and Visual Sciences, University of Iowa Carver College of Medicine, University of Iowa, Iowa City, IA 52240, USA
| | - Deborah Wallace
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| | - Colm O’Brien
- Department of Ophthalmology, Mater Misericordiae University Hospital, D07 R2WY Dublin, Ireland (M.I.)
- School of Medicine, University College Dublin, D04 V1W8 Dublin, Ireland
| |
Collapse
|
21
|
Yang SA, Rhee KH, Yoo HJ, Pyo MC, Lee KW. Ochratoxin A induces endoplasmic reticulum stress and fibrosis in the kidney via the HIF-1α/miR-155-5p link. Toxicol Rep 2023; 10:133-145. [PMID: 36714464 PMCID: PMC9879730 DOI: 10.1016/j.toxrep.2023.01.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 01/19/2023] Open
Abstract
Ochratoxin A (OTA) is a ubiquitous fungal toxin found in agricultural products and foods that is toxic to both humans and animals. OTA mainly affects kidney, but the mechanisms underlying OTA-induced nephrotoxicity remain not fully understood. MicroRNA (miRNA) is involved in key cellular processes. The toxic mechanism and regulatory effects of miRNAs on OTA toxicity in kidney, and particularly the role of HIFα-1/miR-155-5p on OTA-caused ER stress and fibrosis, were investigated in this study. OTA induced hypoxia-like conditions such as ER stress and fibrosis in HK-2 cells and renal tissues via modulating HIF-1α, which was followed by regulation of ER stress-related proteins (GRP78 and ATF-4), as well as fibrosis-related markers (fibronectin, α-SMA, and E-cadherin). Notably, a total of 62 miRNAs showed significant differential expression in kidney of OTA-treated mice. Under OTA exposure, HIF-1α enhanced miR-155-5p expression, causing ER stress and fibrosis in HK-2 cells. HIF-1α knockdown decreased OTA-induced miR-155-5p expression as well as ER stress and fibrotic responses, whereas miR-155-5p overexpression restored this. Our data suggest that OTA enhances ER stress and fibrosis in the kidney through upregulating the HIF-1α/miR-155-5p link.
Collapse
Affiliation(s)
- Seon Ah Yang
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
| | - Kyu Hyun Rhee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
| | - Hee Joon Yoo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
| | - Min Cheol Pyo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, 02841 Seoul, the Republic of Korea
| |
Collapse
|
22
|
Wen Y, Zhang X, Wei L, Wu M, Cheng Y, Zheng H, Shen A, Fu C, Ali F, Long L, Lu Y, Li J, Peng J. Gastrodin attenuates renal injury and collagen deposition via suppression of the TGF-β1/Smad2/3 signaling pathway based on network pharmacology analysis. Front Pharmacol 2023; 14:1082281. [PMID: 36733505 PMCID: PMC9887022 DOI: 10.3389/fphar.2023.1082281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 01/06/2023] [Indexed: 01/18/2023] Open
Abstract
Background: Gastrodin has been widely used clinically in China as an antihypertensive drug. However, its effect on hypertensive renal injury is yet to be elucidated. The current study aimed to investigate the effects of gastrodin on hypertensive renal injury and its underlying mechanisms by network pharmacology analysis and validation in vivo and in vitro. Methods: A total of 10 spontaneously hypertensive rats (SHRs) were randomly categorized into the following two groups: SHR and SHR + Gastrodin groups. Wistar Kyoto (WKY) rats were used as the control group (n = 5). The SHR + Gastrodin group was intragastrically administered gastrodin (3.5 mg/kg/day), and the rats in both WKY and SHR groups were intragastrically administered an equal amount of double-distilled water for 10 weeks. Hematoxylin-eosin, Masson's trichrome, and Sirius red staining were used to detect the pathological changes and collagen content in the renal tissues. Network pharmacology analysis was performed to explore its potential targets and related pathways. In vitro, the CCK-8 assay was used to determine the cell viability. Immunohistochemistry and western-blotting analyses were employed to assess the protein expression associated with renal fibrosis and transforming growth factor-β1 (TGF-β1) pathway-related proteins in the renal tissues or in TGF-β1-stimulated rat kidney fibroblast cell lines (NRK-49F). Results: Gastrodin treatment attenuates renal injury and pathological alterations in SHRs, including glomerular sclerosis and atrophy, epithelial cell atrophy, and tubular dilation. Gastrodin also reduced the accumulation of collagen in the renal tissues of SHRs, which were confirmed by downregulation of α-SMA, collagen I, collagen III protein expression. Network pharmacology analysis identified TGFB1 and SMAD2 as two of lead candidate targets of gastrodin on against hypertensive renal injury. Consistently, gastrodin treatment downregulated the increase of the protein expression of TGF-β1, and ratios of both p-Smad2/Smad2 and p-Samd3/Smad3 in renal tissues of SHRs. In vitro, gastrodin (25-100 μM) treatment significantly reversed the upregulation of α-SMA, fibronectin, collagen I, as well as p-Smad2 and p-Smad3 protein expressions without affecting the cell viability of TGF-β1 stimulated NRK-49F cells. Conclusion: Gastrodin treatment significantly attenuates hypertensive renal injury and renal fibrosis and suppresses TGF-β1/Smad2/3 signaling in vivo and in vitro.
Collapse
Affiliation(s)
- Ying Wen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China,Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Huifang Zheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China,Innovation and Transformation Center, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Changgeng Fu
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Farman Ali
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Linzi Long
- National Clinical Research Center for Chinese Medicine Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China,Department of Geriatrics, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yao Lu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China
| | - Jiapeng Li
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China,*Correspondence: Jiapeng Li, ; Jun Peng,
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Key Laboratory of Integrative Medicine in Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China,Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, China,*Correspondence: Jiapeng Li, ; Jun Peng,
| |
Collapse
|
23
|
Gharbia FZ, Abouhashem AS, Moqidem YA, Elbaz AA, Abdellatif A, Singh K, Sen CK, Azzazy HME. Adult skin fibroblast state change in murine wound healing. Sci Rep 2023; 13:886. [PMID: 36650180 PMCID: PMC9845335 DOI: 10.1038/s41598-022-27152-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/27/2022] [Indexed: 01/18/2023] Open
Abstract
Wound healing is a well-organized dynamic process involving coordinated consecutive phases: homeostasis, inflammation, proliferation and resolution. Fibroblasts play major roles in skin wound healing such as in wound contraction and release of growth factors which are of importance in angiogenesis and tissue remodeling. Abnormal fibroblast phenotypes have been identified in patients with chronic wounds. In this work, we analyzed scRNA-seq datasets of normal and wounded skin from mice at day 4 post-wound to investigate fibroblast heterogeneity during the proliferative phase of wound healing. Compositional analysis revealed a specific subset of fibroblast (cluster 3) that primarily increased in wounded skin (14%) compared to normal skin (3.9%). This subset was characterized by a gene signature marked by the plasma membrane proteins Sfrp2 + Sfrp4 + Sfrp1 + and the transcription factors Ebf1 + Prrx1 + Maged1 + . Differential gene expression and enrichment analysis identified epithelial to mesenchymal transition (EMT) and angiogenesis to be upregulated in the emerging subset of fibroblasts of the wounded skin. Using two other datasets for murine wounded skin confirmed the increase in cluster 3-like fibroblasts at days 2, 7 and 14 post-wounding with a peak at day 7. By performing a similarity check between the differential gene expression profile between wounded and normal skin for this emerging fibroblast subset with drug signature from the ConnectivityMap database, we identified drugs capable of mimicking the observed gene expression change in fibroblasts during wound healing. TTNPB, verteprofin and nicotinic acid were identified as candidate drugs capable of inducing fibroblast gene expression profile necessary for wound healing. On the other hand, methocarbamol, ifosfamide and penbutolol were recognized to antagonize the identified fibroblast differential expression profile during wound healing which might cause delay in wound healing. Taken together, analysis of murine transcriptomic skin wound healing datasets suggested a subset of fibroblasts capable of inducing EMT and further inferred drugs that might be tested as potential candidates to induce wound closure.
Collapse
Affiliation(s)
- Fatma Z Gharbia
- Graduate Nanotechnology Program, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Ann Arbor, MI, 48105, USA
| | - Ahmed S Abouhashem
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- Sharkia Clinical Research Department, Ministry of Health & Population, Zagazig, 44511, Sharkia, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Yomna A Moqidem
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Ahmed A Elbaz
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
- CytoTalk LLC, Cheyenne, WY, 82001, USA
| | - Ahmed Abdellatif
- Department of Biology, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt
| | - Kanhaiya Singh
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Hassan M E Azzazy
- Department of Chemistry, School of Sciences & Engineering, The American University in Cairo (AUC), AUC Avenue, P.O. Box 74, New Cairo, 11835, Egypt.
- Department of Nanobiophotonics, Leibniz Institute for Photonic Technology, Albert Einstein Str. 9, 07745, Jena, Germany.
| |
Collapse
|
24
|
Tepus M, Tonoli E, Verderio EAM. Molecular profiling of urinary extracellular vesicles in chronic kidney disease and renal fibrosis. Front Pharmacol 2023; 13:1041327. [PMID: 36712680 PMCID: PMC9877239 DOI: 10.3389/fphar.2022.1041327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 12/21/2022] [Indexed: 01/13/2023] Open
Abstract
Chronic kidney disease (CKD) is a long-term kidney damage caused by gradual loss of essential kidney functions. A global health issue, CKD affects up to 16% of the population worldwide. Symptoms are often not apparent in the early stages, and if left untreated, CKD can progress to end-stage kidney disease (ESKD), also known as kidney failure, when the only possible treatments are dialysis and kidney transplantation. The end point of nearly all forms of CKD is kidney fibrosis, a process of unsuccessful wound-healing of kidney tissue. Detection of kidney fibrosis, therefore, often means detection of CKD. Renal biopsy remains the best test for renal scarring, despite being intrinsically limited by its invasiveness and sampling bias. Urine is a desirable source of fibrosis biomarkers as it can be easily obtained in a non-invasive way and in large volumes. Besides, urine contains biomolecules filtered through the glomeruli, mirroring the pathological state. There is, however, a problem of highly abundant urinary proteins that can mask rare disease biomarkers. Urinary extracellular vesicles (uEVs), which originate from renal cells and carry proteins, nucleic acids, and lipids, are an attractive source of potential rare CKD biomarkers. Their cargo consists of low-abundant proteins but highly concentrated in a nanosize-volume, as well as molecules too large to be filtered from plasma. Combining molecular profiling data (protein and miRNAs) of uEVs, isolated from patients affected by various forms of CKD, this review considers the possible diagnostic and prognostic value of uEVs biomarkers and their potential application in the translation of new experimental antifibrotic therapeutics.
Collapse
Affiliation(s)
- Melanie Tepus
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Elisa Tonoli
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| | - Elisabetta A. M. Verderio
- Centre for Health, Ageing and the Understanding of Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
- Department of Biological, Geological, and Environmental Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
25
|
Glucosidase inhibitor, Nimbidiol ameliorates renal fibrosis and dysfunction in type-1 diabetes. Sci Rep 2022; 12:21707. [PMID: 36522378 PMCID: PMC9755213 DOI: 10.1038/s41598-022-25848-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 12/06/2022] [Indexed: 12/23/2022] Open
Abstract
Diabetic nephropathy is characterized by excessive accumulation of extracellular matrix (ECM) leading to renal fibrosis, progressive deterioration of renal function, and eventually to end stage renal disease. Matrix metalloproteinases (MMPs) are known to regulate synthesis and degradation of the ECM. Earlier, we demonstrated that imbalanced MMPs promote adverse ECM remodeling leading to renal fibrosis in type-1 diabetes. Moreover, elevated macrophage infiltration, pro-inflammatory cytokines and epithelial‒mesenchymal transition (EMT) are known to contribute to the renal fibrosis. Various bioactive compounds derived from the medicinal plant, Azadirachta indica (neem) are shown to regulate inflammation and ECM proteins in different diseases. Nimbidiol is a neem-derived diterpenoid that is considered as a potential anti-diabetic compound due to its glucosidase inhibitory properties. We investigated whether Nimbidiol mitigates adverse ECM accumulation and renal fibrosis to improve kidney function in type-1 diabetes and the underlying mechanism. Wild-type (C57BL/6J) and type-1 diabetic (C57BL/6-Ins2Akita/J) mice were treated either with saline or with Nimbidiol (0.40 mg kg-1 d-1) for eight weeks. Diabetic kidney showed increased accumulation of M1 macrophages, elevated pro-inflammatory cytokines and EMT. In addition, upregulated MMP-9 and MMP-13, excessive collagen deposition in the glomerular and tubulointerstitial regions, and degradation of vascular elastin resulted to renal fibrosis in the Akita mice. These pathological changes in the diabetic mice were associated with functional impairments that include elevated resistive index and reduced blood flow in the renal cortex, and decreased glomerular filtration rate. Furthermore, TGF-β1, p-Smad2/3, p-P38, p-ERK1/2 and p-JNK were upregulated in diabetic kidney compared to WT mice. Treatment with Nimbidiol reversed the changes to alleviate inflammation, ECM accumulation and fibrosis and thus, improved renal function in Akita mice. Together, our results suggest that Nimbidiol attenuates inflammation and ECM accumulation and thereby, protects kidney from fibrosis and dysfunction possibly by inhibiting TGF-β/Smad and MAPK signaling pathways in type-1 diabetes.
Collapse
|
26
|
Yuan M, Briscese K, Hong TS, Brunetti L. Natural products for the prevention of antibiotic-associated kidney injury. CURRENT OPINION IN TOXICOLOGY 2022; 32:100363. [PMID: 38884043 PMCID: PMC11178348 DOI: 10.1016/j.cotox.2022.100363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Drug-induced acute kidney injury (AKI), especially from exposure to antibiotics, has a high prevalence secondary to their frequent prescription. Typically, drug-induced AKI results from acute tubular necrosis or acute interstitial nephritis. While some risk factors for the development of AKI in individuals treated with antibiotics are modifiable, others such as concomitant drug therapies to treat comorbidities, age, and pre-existing chronic kidney disease are not modifiable. As such, there is an urgent need to identify strategies to reduce the risk of AKI in individuals requiring antibiotic therapy. Natural products, especially those rich in active constituents possessing antioxidant properties are an attractive option to mitigate AKI risk. Given that mitochondrial dysfunction precedes AKI and natural products can restore mitochondrial health and counter the oxidative stress secondary to mitochondrial damage investigating their utility warrants further attention. The following review summarizes the available preclinical and clinical evidence that provides a foundation for future study.
Collapse
Affiliation(s)
- Marshall Yuan
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Kelsey Briscese
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Thomas S Hong
- Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Luigi Brunetti
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
27
|
Sun Y, Jin D, Zhang Z, Jin D, Xue J, Duan L, Zhang Y, Kang X, Lian F. The critical role of the Hippo signaling pathway in kidney diseases. Front Pharmacol 2022; 13:988175. [PMID: 36483738 PMCID: PMC9723352 DOI: 10.3389/fphar.2022.988175] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 11/03/2022] [Indexed: 09/14/2023] Open
Abstract
The Hippo signaling pathway is involved in cell growth, proliferation, and apoptosis, and it plays a key role in regulating organ size, tissue regeneration, and tumor development. The Hippo signaling pathway also participates in the occurrence and development of various human diseases. Recently, many studies have shown that the Hippo pathway is closely related to renal diseases, including renal cancer, cystic kidney disease, diabetic nephropathy, and renal fibrosis, and it promotes the transformation of acute kidney disease to chronic kidney disease (CKD). The present paper summarizes and analyzes the research status of the Hippo signaling pathway in different kidney diseases, and it also summarizes the expression of Hippo signaling pathway components in pathological tissues of kidney diseases. In addition, the present paper discusses the positive therapeutic significance of traditional Chinese medicine (TCM) in regulating the Hippo signaling pathway for treating kidney diseases. This article introduces new targets and ideas for drug development, clinical diagnosis, and treatment of kidney diseases.
Collapse
Affiliation(s)
- Yuting Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Ziwei Zhang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - Di Jin
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - JiaoJiao Xue
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| | - LiYun Duan
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - YuQing Zhang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - XiaoMin Kang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - FengMei Lian
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- College of Chinese Medicine, Changchun University of Chinese Medicine, Jilin, China
| |
Collapse
|
28
|
Gelat B, Rathaur P, Malaviya P, Patel B, Trivedi K, Johar K, Gelat R. The intervention of epithelial-mesenchymal transition in homeostasis of human retinal pigment epithelial cells: a review. J Histotechnol 2022; 45:148-160. [DOI: 10.1080/01478885.2022.2137665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Brijesh Gelat
- Department of Zoology, BMTC and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| | - Pooja Rathaur
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India
| | - Pooja Malaviya
- Department of Cell and Molecular Biology, Iladevi Cataract and IOL Research Centre, Ahmedabad, Gujarat, India
| | - Binita Patel
- Department of Life Science, School of Sciences, Gujarat University, Ahmedabad, India
| | - Krupali Trivedi
- Department of Zoology, BMTC and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| | - Kaid Johar
- Department of Zoology, BMTC and Human Genetics, School of Sciences, Gujarat University, Ahmedabad, India
| | - Rahul Gelat
- Institute of Teaching and Research in Ayurveda (ITRA), Gujarat Ayurved University, Jamnagar, India
| |
Collapse
|
29
|
Cao Y, Lin JH, Hammes HP, Zhang C. Cellular phenotypic transitions in diabetic nephropathy: An update. Front Pharmacol 2022; 13:1038073. [PMID: 36408221 PMCID: PMC9666367 DOI: 10.3389/fphar.2022.1038073] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/17/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic nephropathy (DN) is a major cause of morbidity and mortality in diabetes and is the most common cause of end stage renal disease (ESRD). Renal fibrosis is the final pathological change in DN. It is widely believed that cellular phenotypic switching is the cause of renal fibrosis in diabetic nephropathy. Several types of kidney cells undergo activation and differentiation and become reprogrammed to express markers of mesenchymal cells or podocyte-like cells. However, the development of targeted therapy for DN has not yet been identified. Here, we discussed the pathophysiologic changes of DN and delineated the possible origins that contribute to myofibroblasts and podocytes through phenotypic transitions. We also highlight the molecular signaling pathways involved in the phenotypic transition, which would provide valuable information for the activation of phenotypic switching and designing effective therapies for DN.
Collapse
Affiliation(s)
- Yiling Cao
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ji-Hong Lin
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Hans-Peter Hammes
- 5th Medical Department, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Lausecker F, Lennon R, Randles MJ. The kidney matrisome in health, aging, and disease. Kidney Int 2022; 102:1000-1012. [PMID: 35870643 DOI: 10.1016/j.kint.2022.06.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 02/06/2023]
Abstract
Dysregulated extracellular matrix is the hallmark of fibrosis, and it has a profound impact on kidney function in disease. Furthermore, perturbation of matrix homeostasis is a feature of aging and is associated with declining kidney function. Understanding these dynamic processes, in the hope of developing therapies to combat matrix dysregulation, requires the integration of data acquired by both well-established and novel technologies. Owing to its complexity, the extracellular proteome, or matrisome, still holds many secrets and has great potential for the identification of clinical biomarkers and drug targets. The molecular resolution of matrix composition during aging and disease has been illuminated by cutting-edge mass spectrometry-based proteomics in recent years, but there remain key questions about the mechanisms that drive altered matrix composition. Basement membrane components are particularly important in the context of kidney function; and data from proteomic studies suggest that switches between basement membrane and interstitial matrix proteins are likely to contribute to organ dysfunction during aging and disease. Understanding the impact of such changes on physical properties of the matrix, and the subsequent cellular response to altered stiffness and viscoelasticity, is of critical importance. Likewise, the comparison of proteomic data sets from multiple organs is required to identify common matrix biomarkers and shared pathways for therapeutic intervention. Coupled with single-cell transcriptomics, there is the potential to identify the cellular origin of matrix changes, which could enable cell-targeted therapy. This review provides a contemporary perspective of the complex kidney matrisome and draws comparison to altered matrix in heart and liver disease.
Collapse
Affiliation(s)
- Franziska Lausecker
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Rachel Lennon
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK; Department of Paediatric Nephrology, Royal Manchester Children's Hospital, Manchester University Hospitals National Health Service (NHS) Foundation Trust, Manchester Academic Health Science Centre, Manchester, UK
| | - Michael J Randles
- Chester Medical School, Faculty of Medicine and Life Sciences, University of Chester, Chester, UK.
| |
Collapse
|
31
|
Martínez-Cuazitl A, Gómez-García MDC, Hidalgo-Alegria O, Flores OM, Núñez-Gastélum JA, Martínez ESM, Ríos-Cortés AM, Garcia-Solis M, Pérez-Ishiwara DG. Characterization of Polyphenolic Compounds from Bacopa procumbens and Their Effects on Wound-Healing Process. Molecules 2022; 27:molecules27196521. [PMID: 36235058 PMCID: PMC9571823 DOI: 10.3390/molecules27196521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/26/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
Wounds represent a medical problem that contributes importantly to patient morbidity and to healthcare costs in several pathologies. In Hidalgo, Mexico, the Bacopa procumbens plant has been traditionally used for wound-healing care for several generations; in vitro and in vivo experiments were designed to evaluate the effects of bioactive compounds obtained from a B. procumbens aqueous fraction and to determine the key pathways involved in wound regeneration. Bioactive compounds were characterized by HPLC/QTOF-MS, and proliferation, migration, adhesion, and differentiation studies were conducted on NIH/3T3 fibroblasts. Polyphenolic compounds from Bacopa procumbens (PB) regulated proliferation and cell adhesion; enhanced migration, reducing the artificial scratch area; and modulated cell differentiation. PB compounds were included in a hydrogel for topical administration in a rat excision wound model. Histological, histochemical, and mechanical analyses showed that PB treatment accelerates wound closure in at least 48 h and reduces inflammation, increasing cell proliferation and deposition and organization of collagen at earlier times. These changes resulted in the formation of a scar with better tensile properties. Immunohistochemistry and RT-PCR molecular analyses demonstrated that treatment induces (i) overexpression of transforming growth factor beta (TGF-β) and (ii) the phosphorylation of Smad2/3 and ERK1/2, suggesting the central role of some PB compounds to enhance wound healing, modulating TGF-β activation.
Collapse
Affiliation(s)
- Adriana Martínez-Cuazitl
- Laboratorio de Biomedicina Molecular, ENMyH, Instituto Politécnico Nacional, Mexico City 07320, Mexico
- Escuela Militar de Medicina, Centro Militar de Ciencias de la Salud, UDEFA-SEDENA, Mexico City 11200, Mexico
| | | | - Oriana Hidalgo-Alegria
- Laboratorio de Biomedicina Molecular, ENMyH, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - Olivia Medel Flores
- Laboratorio de Biomedicina Molecular, ENMyH, Instituto Politécnico Nacional, Mexico City 07320, Mexico
| | - José Alberto Núñez-Gastélum
- Departamento de Ciencias Químico Biológicas, Instituto de Ciencias Biomédicas, Universidad Autónoma de Ciudad Juárez, Ciudad Juárez 32310, Mexico
| | - Eduardo San Martín Martínez
- Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada-Unidad Legaria, Instituto Politécnico Nacional, Mexico City 11500, Mexico
| | - Ada María Ríos-Cortés
- Centro de Investigación en Biotecnología Aplicada, Instituto Politécnico Nacional, Tlaxcala de Xicohténcatl 90700, Mexico
| | - Mario Garcia-Solis
- Departamento de Patología, Hospital General de Tláhuac, Mexico City 13250, Mexico
| | - David Guillermo Pérez-Ishiwara
- Laboratorio de Biomedicina Molecular, ENMyH, Instituto Politécnico Nacional, Mexico City 07320, Mexico
- Correspondence: ; Tel.: +01-55-5538993877 (ext. 07320)
| |
Collapse
|
32
|
Chen C, Wang W, Poklis JL, Lichtman AH, Ritter JK, Hu G, Xie D, Li N. Inactivation of fatty acid amide hydrolase protects against ischemic reperfusion injury-induced renal fibrogenesis. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166456. [PMID: 35710061 PMCID: PMC10215004 DOI: 10.1016/j.bbadis.2022.166456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/26/2022] [Accepted: 06/06/2022] [Indexed: 11/26/2022]
Abstract
Although cannabinoid receptors (CB) are recognized as targets for renal fibrosis, the roles of endogenous cannabinoid anandamide (AEA) and its primary hydrolytic enzyme, fatty acid amide hydrolase (FAAH), in renal fibrogenesis remain unclear. The present study used a mouse model of post-ischemia-reperfusion renal injury (PIR) to test the hypothesis that FAAH participates in the renal fibrogenesis. Our results demonstrated that PIR showed upregulated expression of FAAH in renal proximal tubules, accompanied with decreased AEA levels in kidneys. Faah knockout mice recovered the reduced AEA levels and ameliorated PIR-triggered increases in blood urea nitrogen, plasma creatinine as well as renal profibrogenic markers and injuries. Correspondingly, a selective FAAH inhibitor, PF-04457845, inhibited the transforming growth factor-beta 1 (TGF-β1)-induced profibrogenic markers in human proximal tubular cell line (HK-2 cells) and mouse primary cultured tubular cells. Knockdown of FAAH by siRNA in HK-2 cells had similar effects as PF-04457845. Tubular cells isolated from Faah-/- mice further validated the protection against TGF-β1-induced damages. The CB 1 or CB2 receptor antagonist and exogenous FAAH metabolite arachidonic acid failed to reverse the protective effects of FAAH inactivation in HK-2 cells. However, a substrate-selective inhibitor of AEA-cyclooxygenase-2 (COX-2) pathway significantly suppressed the anti-profibrogenic actions of FAAH inhibition. Further, the AEA-COX-2 metabolite, prostamide E2 exerted anti-fibrogenesis effect. These findings suggest that FAAH activation and the consequent reduction of AEA contribute to the renal fibrogenesis, and that FAAH inhibition protects against fibrogenesis in renal cells independently of CB receptors via the AEA-COX-2 pathway by the recovery of reduced AEA.
Collapse
Affiliation(s)
- Chaoling Chen
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Weili Wang
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Justin L Poklis
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Aron H Lichtman
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Joseph K Ritter
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Gaizun Hu
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Dengpiao Xie
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Ningjun Li
- Department of Pharmacology & Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
33
|
Lu Y, Wang L, Zhang M, Chen Z. Mesenchymal Stem Cell-Derived Small Extracellular Vesicles: A Novel Approach for Kidney Disease Treatment. Int J Nanomedicine 2022; 17:3603-3618. [PMID: 35990308 PMCID: PMC9386173 DOI: 10.2147/ijn.s372254] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/04/2022] [Indexed: 12/24/2022] Open
Abstract
Globally, kidney disease has become a serious health challenge, with approximately 10% of adults suffering with the disease, and increasing incidence and mortality rates every year. Small extracellular vesicles (sEVs) are 30 nm-100 nm sized nanovesicles released by cells into the extracellular matrix (ECM), which serve as mediators of intercellular communication. Depending on the cell origin, sEVs have different roles which depend on internal cargoes including, nucleic acids, proteins, and lipids. Mesenchymal stem cell (MSCs) exert anti-inflammatory, anti-aging, and wound healing functions mainly via sEVs in a stable and safe manner. MSC-derived sEVs (MSC-sEVs) exert roles in several kidney diseases by transporting renoprotective cargoes to reduce oxidative stress, inhibit renal cell apoptosis, suppress inflammation, and mediate anti-fibrosis mechanisms. Additionally, because MSC-sEVs efficiently target damaged kidneys, they have the potential to become the next generation cell-free therapies for kidney disease. Herein, we review recent research data on how MSC-sEVs could be used to treat kidney disease.
Collapse
Affiliation(s)
- Yukang Lu
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Mengting Zhang
- First Clinical Medical College, Gannan Medical University, Ganzhou, People's Republic of China.,Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| | - Zhiping Chen
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, People's Republic of China
| |
Collapse
|
34
|
Chae SA, Pyo MC, Yoo HJ, Lee KW. Ochratoxin a induces hepatic fibrosis through TGF-β receptor I/Smad2/3 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2022; 37:2084-2095. [PMID: 35543154 DOI: 10.1002/tox.23552] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 04/20/2022] [Accepted: 04/23/2022] [Indexed: 06/14/2023]
Abstract
Ochratoxin A (OTA) is a mycotoxin generated by Penicillium and Aspergillus species. It is often found in cereals. We hypothesized that OTA exposure induces epithelial-mesenchymal transition (EMT), leading to liver fibrosis. In this research, we explored whether the TGF-β receptor I (TGF-β RI)/Smad2/3 signaling pathway is related to EMT-induced hepatic fibrosis. In vitro and in vivo experiments, mRNA and protein expression of liver fibrosis-related markers such as fibronectin, α-smooth muscle actin (α-SMA) and E-cadherin were assessed. The levels of alkaline phosphatase, alanine transaminase, aspartate aminotransferase, and total bilirubin, which are used to assess damage, increased. We also confirmed the increase in mRNA and protein expression of TGF-β RI, Smad2, and Smad3. The expression of liver fibrosis-related markers was decreased by siRNA-mediated silencing of Smad2/3, as well as TGF-RI suppression. Liver cells exposed to OTA showed enhanced TGF-β RI expression on the cell membrane. These results demonstrated that OTA induces hepatic fibrosis through TGF-β RI and Smad2/3 pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Seung A Chae
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Min Cheol Pyo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Hee Joon Yoo
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| | - Kwang-Won Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
- Department of Food Bioscience and Technology, College of Life Sciences and Biotechnology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
35
|
Mechanism of Cornus Officinalis in Treating Diabetic Kidney Disease Based on Network Pharmacology. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1799106. [PMID: 35855831 PMCID: PMC9288281 DOI: 10.1155/2022/1799106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 11/28/2022]
Abstract
Diabetic kidney disease (DKD), one of the most important diabetic complications, is a great clinical challenge. It still lacks proper therapeutic strategies without side effects due to the complex pathological mechanisms. Cornus officinalis (CO) is a common traditional Chinese medicine, which has been used in the treatment of DKD and takes beneficial effects in therapy. However, the mechanism of CO in treating DKD is not clear yet. In this study, network pharmacology was applied to illustrate the potential mechanism of CO and the interaction between targets of CO and targets of disease. First, the active ingredients of CO and related targets were screened from the online database. Second, the intersection network between CO and disease was constructed, and protein–protein interaction analysis was done. Third, GO and KEGG analysis were employed to figure out the key targets of CO. Finally, molecular docking was carried out in the software SYBYL to verify the effectiveness of the ingredients and targets selected. According to GO and KEGG analysis, drug metabolism-cytochrome P450, sphingolipid signaling pathway, HIF-1 signaling pathway, TGF-beta signaling pathway, cGMP-PKG signaling pathway, estrogen signaling pathway, and TNF signaling pathway were most closely related to the pathogenesis of DKD. Moreover, NOS3, TNF, ROCK1, PPARG, KDR, and HIF1A were identified as key targets in regulating the occurrence and development of the disease. This study provides evidence to elucidate the mechanism of CO comprehensively and systematically and lays the foundation for further research on CO.
Collapse
|
36
|
ShenKang Injection Attenuates Renal Fibrosis by Inhibiting EMT and Regulating the Wnt/β-Catenin Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:9705948. [PMID: 35800011 PMCID: PMC9256403 DOI: 10.1155/2022/9705948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
Abstract
Shenkang Injection (SKI) is a traditional Chinese medicine injection commonly used in the clinical treatment of chronic kidney disease. Although it has been confirmed that SKI has anti-kidney fibrosis effects, the underlying mechanism remains unclear. To investigate the effects of SKI on epithelial-mesenchymal transition (EMT) and Wnt/β-catenin pathway and explore its potential anti-fibrosis mechanism. A unilateral ureteral obstruction (UUO) model was induced by ligating the left ureter of male SD rats. A total of 24 rats were randomly divided into the following four groups: sham group, model group, SKI group, and benazepril group. The rats in each group were treated for 28 days, and renal function was evaluated by blood urea nitrogen (BUN) and serum creatinine (Scr). The degree of renal fibrosis was assessed by hematoxylin and eosin (HE) and Masson staining. Extracellular matrix (ECM) deposition was evaluated by immunohistochemistry. Real-time fluorescent quantitative PCR (RT-qPCR) and western blotting were used to detect the expression of genes and proteins in the Wnt/β-catenin signaling pathway. Further studies were performed in vitro using HK-2 cells treated with TGF-β1. At 28 days postoperation, the levels of BUN and Scr expression were significantly increased in the UUO group. SKI and benazepril reduced the levels of BUN and Scr, which displayed protective renal effects. Pathological staining showed that compared with the sham operation group, the renal parenchymal structure was severely damaged, the number of glomeruli was reduced, and a large amount of collagen was deposited in the kidney tissue of the UUO group. SKI treatment reduced morphological changes. Immunohistochemistry showed that compared with the sham operation group, the content of collagen I and FN in the kidney tissue of the UUO group were significantly increased, whereas the SKI content was decreased. In addition, compared with the UUO group, the levels of Wnt1, active β-catenin, Snail1, and PAI-1 expression were reduced in the SKI group, suggesting that SKI may reduce renal fibrosis by mediating the Wnt/β-catenin pathway. Further in vitro studies showed that collagen I, FN, and α-SMA levels in HK-2 cells were significantly increased following stimulation with TGF-β1. SKI could significantly reduce the expression of collagen I, FN, and α-SMA. A scratch test showed that SKI could reduce HK-2 migration. In addition, by stimulating TGF-β1, the levels of Wnt1, active β-catenin, snail1, and PAI-1 were significantly upregulated. SKI treatment could inhibit the activity of the Wnt/β-catenin signaling pathway in HK-2 cells. SKI improves kidney function by inhibiting renal fibrosis. The anti-fibrotic effects may be mediated by regulation of the Wnt/β-catenin pathway and EMT inhibition.
Collapse
|
37
|
Yuan R, Zhong Y, Zeng Y, Zhang J. Correlation between Tubulointerstitial Lesion and Blood Pressure in Lupus Nephritis Patients: A Pathological, Retrospective Study. Kidney Blood Press Res 2022; 47:391-398. [PMID: 35259751 DOI: 10.1159/000523793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 02/23/2022] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The objective was to study the influence of pathological factors of glomerular lesion (GL), tubulointerstitial lesion (TIL), and arteriosclerotic lesion on the blood pressure (BP) of lupus nephritis (LN). METHODS The pathological data and clinical characteristics of 69 LN patients who underwent their first renal biopsy in Chengdu Second People's Hospital from 2012 to 2018 were retrospectively analyzed. The revised 2018 ISN/RPS classification criteria of LN were used to assess the GL and TIL. The lesion index of interlobar/arcuate artery and arteriolar was calculated. Multiple linear regressions were used to analyze the effects of GL, TIL, and vascular lesion (VL) on estimated glomerular filtration rate, systolic BP (SBP), and proteinuria. RESULTS TIL and VL scores were different between the various grades of BP (p = 0.009, 0.019). After adjusting for gender and age, multiple linear regression showed that only TIL was linearly correlated with SBP (p = 0.022). CONCLUSION After adjusting for gender and age, TIL is related to SBP and has a linear relationship with them.
Collapse
Affiliation(s)
- Ruili Yuan
- Department of Hematology and Rheumatology Chengdu Second People's Hospital, Chengdu, China
| | - Yu Zhong
- Department of the Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Zeng
- Department of Hematology and Rheumatology Chengdu Second People's Hospital, Chengdu, China
| | - Jing Zhang
- Department of Hematology and Rheumatology Chengdu Second People's Hospital, Chengdu, China
| |
Collapse
|
38
|
Gabbin B, Meraviglia V, Mummery CL, Rabelink TJ, van Meer BJ, van den Berg CW, Bellin M. Toward Human Models of Cardiorenal Syndrome in vitro. Front Cardiovasc Med 2022; 9:889553. [PMID: 35694669 PMCID: PMC9177996 DOI: 10.3389/fcvm.2022.889553] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 05/09/2022] [Indexed: 11/13/2022] Open
Abstract
Heart and kidney diseases cause high morbidity and mortality. Heart and kidneys have vital functions in the human body and, interestingly, reciprocally influence each other’s behavior: pathological changes in one organ can damage the other. Cardiorenal syndrome (CRS) is a group of disorders in which there is combined dysfunction of both heart and kidney, but its underlying biological mechanisms are not fully understood. This is because complex, multifactorial, and dynamic mechanisms are likely involved. Effective treatments are currently unavailable, but this may be resolved if more was known about how the disease develops and progresses. To date, CRS has actually only been modeled in mice and rats in vivo. Even though these models can capture cardiorenal interaction, they are difficult to manipulate and control. Moreover, interspecies differences may limit extrapolation to patients. The questions we address here are what would it take to model CRS in vitro and how far are we? There are already multiple independent in vitro (human) models of heart and kidney, but none have so far captured their dynamic organ-organ crosstalk. Advanced in vitro human models can provide an insight in disease mechanisms and offer a platform for therapy development. CRS represents an exemplary disease illustrating the need to develop more complex models to study organ-organ interaction in-a-dish. Human induced pluripotent stem cells in combination with microfluidic chips are one powerful tool with potential to recapitulate the characteristics of CRS in vitro. In this review, we provide an overview of the existing in vivo and in vitro models to study CRS, their limitations and new perspectives on how heart-kidney physiological and pathological interaction could be investigated in vitro for future applications.
Collapse
Affiliation(s)
- Beatrice Gabbin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Viviana Meraviglia
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Christine L. Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Applied Stem Cell Technologies, University of Twente, Enschede, Netherlands
| | - Ton J. Rabelink
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Berend J. van Meer
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
| | - Cathelijne W. van den Berg
- Department of Internal Medicine-Nephrology, Leiden University Medical Center, Leiden, Netherlands
- Einthoven Laboratory of Vascular and Regenerative Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Milena Bellin
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, Netherlands
- Department of Biology, University of Padua, Padua, Italy
- Veneto Institute of Molecular Medicine, Padua, Italy
- *Correspondence: Milena Bellin, ,
| |
Collapse
|
39
|
Nemours S, Castro L, Ribatallada-Soriano D, Semidey ME, Aranda M, Ferrer M, Sanchez A, Morote J, Cantero-Recasens G, Meseguer A. Temporal and sex-dependent gene expression patterns in a renal ischemia-reperfusion injury and recovery pig model. Sci Rep 2022; 12:6926. [PMID: 35484379 PMCID: PMC9051203 DOI: 10.1038/s41598-022-10352-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/30/2022] [Indexed: 12/30/2022] Open
Abstract
Men are more prone to acute kidney injury (AKI) and chronic kidney disease (CKD), progressing to end-stage renal disease (ESRD) than women. Severity and capacity to regenerate after AKI are important determinants of CKD progression, and of patient morbidity and mortality in the hospital setting. To determine sex differences during injury and recovery we have generated a female and male renal ischemia/reperfusion injury (IRI) pig model, which represents a major cause of AKI. Although no differences were found in blood urea nitrogen (BUN) and serum creatinine (SCr) levels between both sexes, females exhibited higher mononuclear infiltrates at basal and recovery, while males showed more tubular damage at injury. Global transcriptomic analyses of kidney biopsies from our IRI pig model revealed a sexual dimorphism in the temporal regulation of genes and pathways relevant for kidney injury and repair, which was also detected in human samples. Enrichment analysis of gene sets revealed five temporal and four sexual patterns governing renal IRI and recovery. Overall, this study constitutes an extensive characterization of the time and sex differences occurring during renal IRI and recovery at gene expression level and offers a template of translational value for further study of sexual dimorphism in kidney diseases.
Collapse
Affiliation(s)
- Stéphane Nemours
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Luis Castro
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Didac Ribatallada-Soriano
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Maria E Semidey
- Department of Pathology, Hospital Vall d'Hebron, Barcelona, Spain
| | - Miguel Aranda
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Marina Ferrer
- Rodent Platform, Vall d'Hebron Research Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Alex Sanchez
- Unitat d'Estadística I Bioinformàtica, (UEB), Vall d'Hebron Research Institute, Barcelona, Spain
- Department of Genetics, Microbiology and Statistics, Universitat de Barcelona, Barcelona, Spain
| | - Joan Morote
- Biomedical Research in Urology Group, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Gerard Cantero-Recasens
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain
| | - Anna Meseguer
- Renal Physiopathology Group, Vall d'Hebron Research Institute, Passeig Vall d'Hebron 119-129, 08035, Barcelona, Spain.
- Departament de Bioquímica I Biologia Molecular, Unitat de Bioquímica de Medicina, Universitat Autònoma de Barcelona, Bellaterra, Spain.
- Red de Investigación Renal (REDINREN), Instituto Carlos III-FEDER, Madrid, Spain.
| |
Collapse
|
40
|
Abstract
INTRODUCTION Tubulo-interstitial injury is a poor prognostic factor for lupus nephritis (LN). Here, we tested whether iguratimod could inhibit tubulo-interstitial injury in LN. METHODS MRL/lpr mice, an animal model of lupus, were treated with iguratimod or vehicle solution. Pathological changes of kidney were evaluated blindly by the same pathologist. Renal type I collagen (COL-I), IgG, E-cadherin, fibroblast-specific protein 1 (FSP-1) were detected by immunofluorescence, immunohistochemical staining or quantitative real-time PCR. After treated with transforming growth factor β1 (TGF-β1) and iguratimod, E-cadherin, fibronectin, Smad2/3, p38 MAPK, p-Smad2/3, and p-p38 MAPK, β-catenin and TGF-β type II receptor (TGFβRII) in HK2 cells were measured by western blotting, quantitative real-time PCR or immunofluorescence. RESULTS Iguratimod reduced immune deposition along the tubular basement membrane, inhibited the tubulo-interstitial infiltration of inflammatory cells, and alleviated tubular injury in MRL/lpr mice. Moreover, Iguratimod eased the tubulo-interstitial deposition of collagen fibers, which was confirmed by decreased expression of COL-I. Furthermore, iguratimod suppressed the expression of FSP-1 and increased that of E-cadherin in renal tubular epithelial cells. In HK2 cells cultured with TGF-β1, iguratimod treatment not only reversed cellular morphological changes, but also prevented E-cadherin downregulation and fibronectin upregulation. In addition, iguratimod inhibited phosphorylation of TGFβRII, Smad2/3 and p38 MAPK in HK2 cells treated with TGF-β1, and also blocked nuclear translocation of β-catenin. CONCLUSION Iguratimod eased tubulo-interstitial lesions in LN, especially tubulo-interstitial fibrosis, and might have potential as a drug for inhibiting the progression of tubulo-interstitial fibrosis in LN.
Collapse
Affiliation(s)
- Leixi Xue
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiajun Xu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wentian Lu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinxiang Fu
- Department of Hematology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhichun Liu
- Department of Rheumatology and Immunology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
41
|
Stecco A, Cowman M, Pirri N, Raghavan P, Pirri C. Densification: Hyaluronan Aggregation in Different Human Organs. Bioengineering (Basel) 2022; 9:159. [PMID: 35447719 PMCID: PMC9028708 DOI: 10.3390/bioengineering9040159] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/28/2022] [Accepted: 04/04/2022] [Indexed: 11/16/2022] Open
Abstract
Hyaluronan (HA) has complex biological roles that have catalyzed clinical interest in several fields of medicine. In this narrative review, we provide an overview of HA aggregation, also called densification, in human organs. The literature suggests that HA aggregation can occur in the liver, eye, lung, kidney, blood vessel, muscle, fascia, skin, pancreatic cancer and malignant melanoma. In all these organs, aggregation of HA leads to an increase in extracellular matrix viscosity, causing stiffness and organ dysfunction. Fibrosis, in some of these organs, may also occur as a direct consequence of densification in the long term. Specific imaging evaluation, such dynamic ultrasonography, elasto-sonography, elasto-MRI and T1ρ MRI can permit early diagnosis to enable the clinician to organize the treatment plan and avoid further progression of the pathology and dysfunction.
Collapse
Affiliation(s)
- Antonio Stecco
- Rusk Rehabilitation, New York University School of Medicine, New York, NY 10016, USA;
| | - Mary Cowman
- Department of Biomedical Engineering, New York University Tandon School of Engineering, New York, NY 10016, USA;
| | - Nina Pirri
- Department of Medicine—DIMED, School of Radiology, Radiology Institute, University of Padua, 35122 Padova, Italy;
| | - Preeti Raghavan
- Department of Physical Medicine and Rehabilitation and Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| | - Carmelo Pirri
- Department of Neurosciences, Institute of Human Anatomy, University of Padova, 35121 Padova, Italy
| |
Collapse
|
42
|
Shaker AM, Rakha NK, El-Shehaby AMR, Ramzy T, Hamza WM, Elkhatib MM. The significance of fibroblast growth factor-2 and kidney injury molecule-1 as biomarker of interstitial renal fibrosis in glomerulonephritis. THE EGYPTIAN JOURNAL OF INTERNAL MEDICINE 2022. [DOI: 10.1186/s43162-022-00112-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Renal tubulointerstitial fibrosis is a structural marker and prominent pathological characteristic of chronic progressive kidney disease, fibroblast growth factor-2 (FGF2) is a key fibrogenic cytokine that is likely to be involved in the pathogenesis of renal fibrosis, kidney injury molecule-1 (KIM-1) is one of the most promising, early biomarkers of renal disease, either acute or chronic, due to its translatability between preclinical and clinical trials. It is believed that this molecule participates in the process of both kidney injury and healing.
Methods
We prospectively enrolled a cohort study of eighty adult patients who had glomerular diseases (with glomerular filtration rate (GFR) > 30 ml/min/m2); serum level of FGF-2 and KIM-1 was measured at the same time of renal biopsy and was correlated with the degree of interstitial renal fibrosis.
Results
We found a significant positive correlation between FGF-2 and KIM-1 and the degree of interstitial renal fibrosis, albumin, and creatinine (P≤ 0.001), and a negative significant correlation with GFR and proteinuria. there is a positive significant correlation between serum KIM-1 and FGF-2 and hypertension with a significant P value (<0.001) that serum KIM-1 has sensitivity 90% and specificity of 95% and serum FGF-2 has sensitivity 95% and specificity 95% for detection of interstitial renal fibrosis.
Conclusions
Serum FGF-2 and KIM-1 seem to be a non-invasive novel biomarker of interstitial renal fibrosis in glomerulonephritis patients. It may become a useful biomarker without the need for the invasive maneuver of the renal biopsy. FGF-2 and KIM-1 are expected to be therapeutic targets for kidney injury.
Collapse
|
43
|
Liu G, Liu X, Yang Y. Comparative transcriptome analysis of miRNA in hydronephrosis male children caused by ureteropelvic junction obstruction with or without renal functional injury. PeerJ 2022; 10:e12962. [PMID: 35237468 PMCID: PMC8884061 DOI: 10.7717/peerj.12962] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 01/27/2022] [Indexed: 01/11/2023] Open
Abstract
MicroRNAs (miRNAs or miRs) are non-coding RNAs that contribute to pathological processes of various kidney diseases. Renal function injury represents a final common outcome of congenital obstructive nephropathy and has attracted a great deal of attention. However the molecular mechanisms are still not fully established. In this study, we compared transcriptome sequencing data of miRNAs of renal tissues from congenital hydronephrosis children with or without renal functional injury, in order to better understand whether microRNAs could play important roles in renal functional injury after ureteropelvic junction obstruction. A total of 22 microRNAs with significant changes in their expression were identified. Five microRNAs were up-regulated and 17 microRNAs were down-regulated in the renal tissues of the hydronephrosis patients with renal function injury compared with those without renal function injury. MicroRNA target genes were predicted by three major online miRNA target prediction algorithms, and all these mRNAs were used to perform the gene ontology analysis and Kyoto Encyclopedia of Gene and Genomes pathway analysis. Then, twelve candidate human and rat homologous miRNAs were selected for validation using RT-qPCR in vitro and in vivo; only miR-187-3p had a trend identical to that detected by the sequencing results among the human tissues, in vivo and in vitro experimental models. In addition, we found that the change of miR-187-3p in vivo was consistent with results in vitro models and showed a decrease trend in time dependence. These results provided a detailed catalog of candidate miRNAs to investigate their regulatory role in renal injury of congenital hydronephrosis, indicating that they may serve as candidate biomarkers or therapeutic targets in the future.
Collapse
Affiliation(s)
- Ge Liu
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Xin Liu
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| | - Yi Yang
- Urology Division, Pediatric Surgery Department, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People’s Republic of China
| |
Collapse
|
44
|
Chemokine (C-C Motif) Ligand 8 and Tubulo-Interstitial Injury in Chronic Kidney Disease. Cells 2022; 11:cells11040658. [PMID: 35203308 PMCID: PMC8869891 DOI: 10.3390/cells11040658] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/19/2022] [Accepted: 02/10/2022] [Indexed: 02/07/2023] Open
Abstract
Kidney fibrosis has been accepted to be a common pathological outcome of chronic kidney disease (CKD). We aimed to examine serum levels and tissue expression of chemokine (C-C motif) ligand 8 (CCL8) in patients with CKD and to investigate their association with kidney fibrosis in CKD model. Serum levels and tissue expression of CCL8 significantly increased with advancing CKD stage, proteinuria level, and pathologic deterioration. In Western blot analysis of primary cultured human tubular epithelial cells after induction of fibrosis with rTGF-β, CCL8 was upregulated by rTGF-β treatment and the simultaneous treatment with anti-CCL8 mAb mitigated the rTGF-β-induced an increase in fibronectin and a decrease E-cadherin and BCL-2 protein levels. The antiapoptotic effect of the anti-CCL8 mAb was also demonstrated by Annexin V/propidium iodide staining assay. In qRT-PCR analysis, mRNA expression levels of the markers for fibrosis and apoptosis showed similar expression patterns to those observed by western blotting. The immunohistochemical analysis revealed CCL8 and fibrosis- and apoptosis-related markers significantly increased in the unilateral ureteral obstruction model, which agrees with our in vitro findings. In conclusion, CCL8 pathway is associated with increased risk of kidney fibrosis and that CCL8 blockade can ameliorate kidney fibrosis and apoptosis.
Collapse
|
45
|
Mitochondrial Pathophysiology on Chronic Kidney Disease. Int J Mol Sci 2022; 23:ijms23031776. [PMID: 35163697 PMCID: PMC8836100 DOI: 10.3390/ijms23031776] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/01/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
In healthy kidneys, interstitial fibroblasts are responsible for the maintenance of renal architecture. Progressive interstitial fibrosis is thought to be a common pathway for chronic kidney diseases (CKD). Diabetes is one of the boosters of CKD. There is no effective treatment to improve kidney function in CKD patients. The kidney is a highly demanding organ, rich in redox reactions occurring in mitochondria, making it particularly vulnerable to oxidative stress (OS). A dysregulation in OS leads to an impairment of the Electron transport chain (ETC). Gene deficiencies in the ETC are closely related to the development of kidney disease, providing evidence that mitochondria integrity is a key player in the early detection of CKD. The development of novel CKD therapies is needed since current methods of treatment are ineffective. Antioxidant targeted therapies and metabolic approaches revealed promising results to delay the progression of some markers associated with kidney disease. Herein, we discuss the role and possible origin of fibroblasts and the possible potentiators of CKD. We will focus on the important features of mitochondria in renal cell function and discuss their role in kidney disease progression. We also discuss the potential of antioxidants and pharmacologic agents to delay kidney disease progression.
Collapse
|
46
|
Zhang Y, Jin D, Duan Y, Zhang Y, Duan L, Lian F, Tong X. Bibliometric Analysis of Renal Fibrosis in Diabetic Kidney Disease From 1985 to 2020. Front Public Health 2022; 10:767591. [PMID: 35186833 PMCID: PMC8855938 DOI: 10.3389/fpubh.2022.767591] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/10/2022] [Indexed: 12/11/2022] Open
Abstract
BackgroundDiabetic renal fibrosis (DRF) is an irreversible renal pathological change in the end-stage of diabetic kidney disease (DKD), which plays a significant role in the development and deterioration of the disease. However, data for bibliometric analysis of renal fibrosis in DKD is currently missing. This study aimed to provide a comprehensive and visualized view of DRF research and lay the foundation for further studies.Materials and MethodsFirstly, the data was collected from the Web of Science Core Collection (WoSCC) database. Secondly, the Web of Science analytic tool was performed to analyze publication years, authors, countries/regions, organizations, and citation frequency. Finally, CiteSpace was employed to construct a visualization bibliometric network to reveal the emerging trends and hotspots of DRF.ResultsA total of 3,821 publications from 1985 to 2020 were included in this study. The number of publications has maintained a growth trend since 2003. Cooper is the most prolific author in this field, and the American Journal of Physiology-Renal Physiology ranking as first place compared with other journals. In terms of the number of publications, China contributed the most to DRF. Monash University is the organization that published the most papers. The top 5 clusters of keyword co-appearance are “chronic kidney disease”, “primary biliary cirrhosis”, “receptor”, “TGF-beta”, “renal tubulointerstitium”. The top 5 clusters of reference co-citation are “microRNAs”, “bone morphogenetic protein”, “hypertrophy”, “glomerulosclerosis”, “diabetic kidney disease”. The strongest citation burst of keyword is “diabetic kidney disease” and the strongest burst of cited reference is “Meng, 2016”.ConclusionsThe present study analyzed the research hotspots, Frontiers, and development trend of DRF and have important implications for future research.
Collapse
Affiliation(s)
- Yuqing Zhang
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - De Jin
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yingying Duan
- Endocrinology Department, Guang'anmen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuehong Zhang
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Liyun Duan
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengmei Lian
- Endocrinology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Fengmei Lian
| | - Xiaolin Tong
- Endocrinology Department, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
- Xiaolin Tong
| |
Collapse
|
47
|
Extracellular Vesicles Derived from Human Liver Stem Cells Attenuate Chronic Kidney Disease Development in an In Vivo Experimental Model of Renal Ischemia and Reperfusion Injury. Int J Mol Sci 2022; 23:ijms23031485. [PMID: 35163409 PMCID: PMC8835844 DOI: 10.3390/ijms23031485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/14/2022] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
The potential therapeutic effect of extracellular vesicles (EVs) that are derived from human liver stem cells (HLSCs) has been tested in an in vivo model of renal ischemia and reperfusion injury (IRI), that induce the development of chronic kidney disease (CKD). EVs were administered intravenously immediately after the IRI and three days later, then their effect was tested at different time points to evaluate how EV-treatment might interfere with fibrosis development. In IRI-mice that were sacrificed two months after the injury, EV- treatment decreased the development of interstitial fibrosis at the histological and molecular levels. Furthermore, the expression levels of pro-inflammatory genes and of epithelial-mesenchymal transition (EMT) genes were significantly reverted by EV-treatment. In IRI-mice that were sacrificed at early time points (two and three days after the injury), functional and histological analyses showed that EV-treatment induced an amelioration of the acute kidney injury (AKI) that was induced by IRI. Interestingly, at the molecular level, a reduction of pro-fibrotic and EMT-genes in sacrificed IRI-mice was observed at days two and three after the injury. These data indicate that in renal IRI, treatment with HLSC-derived EVs improves AKI and interferes with the development of subsequent CKD by modulating the genes that are involved in fibrosis and EMT.
Collapse
|
48
|
Li L, He D, Cai Q, Wang DY. Blockage of TGF-β1-induced epithelial-to- mesenchymal transition by oxymatrine prevents renal interstitial fibrosis. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902020000118738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Affiliation(s)
- Li Li
- University of Chinese Medicine, China
| | | | - Qin Cai
- University of Chinese Medicine, China
| | | |
Collapse
|
49
|
Mariano VS, Boer PA, Gontijo JAR. Fetal Undernutrition Programming, Sympathetic Nerve Activity, and Arterial Hypertension Development. Front Physiol 2021; 12:704819. [PMID: 34867434 PMCID: PMC8635863 DOI: 10.3389/fphys.2021.704819] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 10/14/2021] [Indexed: 12/11/2022] Open
Abstract
A wealth of evidence showed that low birth weight is associated with environmental disruption during gestation, triggering embryotic or fetal adaptations and increasing the susceptibility of progeny to non-communicable diseases, including metabolic and cardiovascular diseases, obesity, and arterial hypertension. In addition, dietary disturbance during pregnancy in animal models has highlighted mechanisms that involve the genesis of arterial hypertension, particularly severe maternal low-protein intake (LP). Functional studies demonstrated that maternal low-protein intake leads to the renal decrease of sodium excretion and the dysfunction of the renin-angiotensin-aldosterone system signaling of LP offspring. The antinatriuretic effect is accentuated by a reduced number of nephron units and glomerulosclerosis, which are critical in establishing arterial hypertension phenotype. Also, in this way, studies have shown that the overactivity of the central and peripheral sympathetic nervous system occurs due to reduced sensory (afferent) renal nerve activity. As a result of this reciprocal and abnormal renorenal reflex, there is an enhanced tubule sodium proximal sodium reabsorption, which, at least in part, contributes directly to arterial hypertension development in some of the programmed models. A recent study has observed that significant changes in adrenal medulla secretion could be involved in the pathophysiological process of increasing blood pressure. Thus, this review aims to compile studies that link the central and peripheral sympathetic system activity mechanisms on water and salt handle and blood pressure control in the maternal protein-restricted offspring. Besides, these pathophysiological mechanisms mainly may involve the modulation of neurokinins and catecholamines pathways.
Collapse
Affiliation(s)
- Vinícius Schiavinatto Mariano
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, São Paulo, Brazil
| | - Patrícia Aline Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, São Paulo, Brazil
| | - José Antônio Rocha Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, Faculty of Medical Sciences, State University of Campinas, São Paulo, Brazil
| |
Collapse
|
50
|
Zhao K, Mao Y, Ye X, Ma J, Sun L, Li P, Li Y. MicroRNA-210-5p alleviates cardiac fibrosis via targeting transforming growth factor-beta type I receptor in rats on high sodium chloride (NaCl)-based diet. Eur J Pharmacol 2021; 912:174587. [PMID: 34678242 DOI: 10.1016/j.ejphar.2021.174587] [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: 07/30/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
The present study was designed to explore whether high sodium chloride (NaCl)-based diet (HSD) caused cardiac fibrosis regardless of blood pressure in Sprague-Dawley (SD) rats, and to further determine the effects and the underlying mechanisms of microRNA (miR)-210-5p on HSD-induced cardiac fibrosis in rats or NaCl-induced cardiac fibroblast activation in neonatal rat cardiac fibroblasts (NRCFs). The SD rats received 8% HSD, and NRCFs were treated with NaCl. The levels of collagen I, alpha-smooth muscle actin (α-SMA) and transforming growth factor-beta 1 (TGF-β1) were increased in the heart of hypertension (HTN), hypertension-prone (HP) and hypertension-resistant (HR) rats on HSD in vivo. NaCl increased the levels of collagen I, α-SMA and TGF-β1 in NRCFs in vitro. The level of miR-210-5p was reduced in both NBD-induced rats' hearts and NaCl-treated NRCFs, which was consistent with the results of miR high-throughput sequencing in NRCFs. The HSD or NaCl-induced increases of collagen I, α-SMA and TGF-β1 were inhibited by miR-210-5p agomiR in vitro and in vivo, respectively. miR-210-5p antagomiR could mimic the pathological effects of NaCl in NRCFS. Bioinformatics analysis and luciferase reporter assays demonstrated that TGF-β type I receptor (TGFBR1) was a direct target gene of miR-210-5p. These results indicated that HSD resulted in cardiac fibrosis regardless of blood pressure. The upregulation of miR-210-5p could attenuate cardiac fibroblast activation in NRCFS via targeting TGFBR1. Thus, upregulating miR-210-5p might be a strategy for the treatment of cardiac fibrosis.
Collapse
Affiliation(s)
- Kun Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yukang Mao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaoman Ye
- Intensive Care Unit, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jiazheng Ma
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Litao Sun
- SEU-FEI Nano-Pico Center, Key Laboratory of MEMS of Ministry of Education, Southeast University, Nanjing, 210096, China
| | - Peng Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yong Li
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|